1
|
Razmara P, Son HW, Ali DW. Cannabidiol exerts teratogenic effects on developing zebrafish through the sonic hedgehog signaling pathway. Sci Rep 2025; 15:14533. [PMID: 40281058 PMCID: PMC12032106 DOI: 10.1038/s41598-025-99194-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Accepted: 04/17/2025] [Indexed: 04/29/2025] Open
Abstract
The increasing legalization and decriminalization of cannabis for therapeutic and recreational purposes in various regions have influenced public perceptions and attitudes toward cannabidiol (CBD)-containing products, including their use during pregnancy. However, there is a knowledge gap regarding the effect of CBD on fetal development, and the mechanisms by which CBD induces developmental deficits have not been well studied. In this study, we investigated whether the teratogenic effects of CBD on developing zebrafish are mediated through Sonic hedgehog signaling (Shh), a critical pathway in development. Embryonic exposure to CBD reduced hatching and survival rates and suppressed Shh pathway activity, leading to decreased ptch2 expression-a regulatory receptor expressed in response to Shh signaling. Larval swimming activity was impaired by CBD exposure. However, coexposure to CBD and a synthetic Shh pathway activator significantly improved developmental outcomes, including decreased mortality, increased hatching rates, elevated ptch2 expression, and increased locomotor activity. These findings underscore the developmental risks associated with CBD use during pregnancy and highlight the involvement of the Shh signaling pathway in driving these effects. The results of this study can inform regulations for cannabis use during pregnancy and emphasize the need to develop therapeutic guidelines for the safe use of CBD-based treatments.
Collapse
Affiliation(s)
- Parastoo Razmara
- Department of Biological Sciences, University of Alberta, CW- 405, Biological Sciences Building, Edmonton, AB, T6G 2E9, Canada.
| | - Hae-Won Son
- Department of Biological Sciences, University of Alberta, CW- 405, Biological Sciences Building, Edmonton, AB, T6G 2E9, Canada
| | - Declan William Ali
- Department of Biological Sciences, University of Alberta, CW- 405, Biological Sciences Building, Edmonton, AB, T6G 2E9, Canada
| |
Collapse
|
2
|
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
|
3
|
VanWinkle PE, Lee E, Wynn B, Nawara TJ, Thomas H, Parant J, Alvarez C, Serra R, Sztul E. Disruption of the creb3l1 gene causes defects in caudal fin regeneration and patterning in zebrafish Danio rerio. Dev Dyn 2024; 253:1106-1129. [PMID: 39003620 PMCID: PMC11609917 DOI: 10.1002/dvdy.726] [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: 10/02/2023] [Revised: 06/12/2024] [Accepted: 06/22/2024] [Indexed: 07/15/2024] Open
Abstract
BACKGROUND The gene cAMP-Responsive Element Binding protein 3-like-1 (CREB3L1) has been implicated in bone development in mice, with CREB3L1 knock-out mice exhibiting fragile bones, and in humans, with CREB3L1 mutations linked to osteogenesis imperfecta. However, the mechanism through which Creb3l1 regulates bone development is not fully understood. RESULTS To probe the role of Creb3l1 in organismal physiology, we used CRISPR-Cas9 genome editing to generate a Danio rerio (zebrafish) model of Creb3l1 deficiency. In contrast to mammalian phenotypes, the Creb3l1 deficient fish do not display abnormalities in osteogenesis, except for a decrease in the bifurcation pattern of caudal fin. Both, skeletal morphology and overall bone density appear normal in the mutant fish. However, the regeneration of caudal fin postamputation is significantly affected, with decreased overall regenerate and mineralized bone area. Moreover, the mutant fish exhibit a severe patterning defect during regeneration, with a significant decrease in bifurcation complexity of the fin rays and distalization of the bifurcation sites. Analysis of genes implicated in bone development showed aberrant patterning of shha and ptch2 in Creb3l1 deficient fish, linking Creb3l1 with Sonic Hedgehog signaling during fin regeneration. CONCLUSIONS Our results uncover a novel role for Creb3l1 in regulating tissue growth and patterning during regeneration.
Collapse
Affiliation(s)
- Peyton E. VanWinkle
- Department of Cell, Developmental and Integrative BiologyUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Eunjoo Lee
- Department of Cell, Developmental and Integrative BiologyUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Bridge Wynn
- Department of Cell, Developmental and Integrative BiologyUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Tomasz J. Nawara
- Department of Cell, Developmental and Integrative BiologyUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Holly Thomas
- Department of Pharmacology and ToxicologyUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - John Parant
- Department of Pharmacology and ToxicologyUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Cecilia Alvarez
- CIBICI‐CONICET, Facultad de Ciencias QuímicasUniversidad Nacional de CórdobaCórdobaArgentina
| | - Rosa Serra
- Department of Cell, Developmental and Integrative BiologyUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Elizabeth Sztul
- Department of Cell, Developmental and Integrative BiologyUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| |
Collapse
|
4
|
Lewis VM, Fernandez RA, Horst SG, Stankunas K. Early exercise disrupts a pro-repair extracellular matrix program during zebrafish fin regeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.15.623835. [PMID: 39605604 PMCID: PMC11601382 DOI: 10.1101/2024.11.15.623835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Understanding how mechanical stimulation from exercise influences cellular responses during tissue repair could enhance therapeutic strategies. We explored zebrafish caudal fin regeneration to study exercise impacts on a robust model of tissue regeneration. We used a swim tunnel to determine that exercise initiated during but not after blastema establishment impaired fin regeneration, including of the bony ray skeleton. Long-term tracking of fluorescently labeled cell lineages showed exercise disrupted blastemal mesenchyme formation. Transcriptomic profiling and section staining indicated exercise reduced an extracellular matrix (ECM) gene expression program, including for hyaluronic acid (HA) synthesis. Like exercise, HA synthesis inhibition or blastemal HA depletion disrupted blastema formation. We considered if injury-upregulated HA establishes a pro-regenerative environment facilitating mechanotransduction. HA density across the blastema correlated with nuclear localization of the mechanotransducer Yes-associated protein (Yap). Further, exercise loading or reducing HA decreased nuclear Yap and cell proliferation. We conclude early exercise during fin regeneration disrupts expression of an HA-rich ECM supporting blastema expansion. These results highlight the interface between mechanotransduction and ECM as consideration for timing exercise interventions and developing regenerative therapies. Significance Statement Controlled exercise promotes healing and recovery from severe skeletal injuries. However, properly timed interventions are essential to promote recovery and prevent further damage. We use zebrafish caudal fin regeneration to mechanistically study exercise impacts on a naturally robust and experimentally accessible model of tissue repair. We link detrimental early exercise effects during fin regeneration to impaired ECM synthesis, mechanotransduction, and cell proliferation. These insights could explain the value of delaying the onset of physical therapy and suggest pursuing therapies that maintain ECM integrity for regenerative rehabilitation.
Collapse
|
5
|
Lukowicz-Bedford RM, Eisen JS, Miller AC. Gap-junction-mediated bioelectric signaling required for slow muscle development and function in zebrafish. Curr Biol 2024; 34:3116-3132.e5. [PMID: 38936363 PMCID: PMC11265983 DOI: 10.1016/j.cub.2024.06.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 04/11/2024] [Accepted: 06/04/2024] [Indexed: 06/29/2024]
Abstract
Bioelectric signaling, intercellular communication facilitated by membrane potential and electrochemical coupling, is emerging as a key regulator of animal development. Gap junction (GJ) channels can mediate bioelectric signaling by creating a fast, direct pathway between cells for the movement of ions and other small molecules. In vertebrates, GJ channels are formed by a highly conserved transmembrane protein family called the connexins. The connexin gene family is large and complex, creating challenges in identifying specific connexins that create channels within developing and mature tissues. Using the embryonic zebrafish neuromuscular system as a model, we identify a connexin conserved across vertebrate lineages, gjd4, which encodes the Cx46.8 protein, that mediates bioelectric signaling required for slow muscle development and function. Through mutant analysis and in vivo imaging, we show that gjd4/Cx46.8 creates GJ channels specifically in developing slow muscle cells. Using genetics, pharmacology, and calcium imaging, we find that spinal-cord-generated neural activity is transmitted to developing slow muscle cells, and synchronized activity spreads via gjd4/Cx46.8 GJ channels. Finally, we show that bioelectrical signal propagation within the developing neuromuscular system is required for appropriate myofiber organization and that disruption leads to defects in behavior. Our work reveals a molecular basis for GJ communication among developing muscle cells and reveals how perturbations to bioelectric signaling in the neuromuscular system may contribute to developmental myopathies. Moreover, this work underscores a critical motif of signal propagation between organ systems and highlights the pivotal role of GJ communication in coordinating bioelectric signaling during development.
Collapse
Affiliation(s)
| | - Judith S Eisen
- University of Oregon, Institute of Neuroscience, Eugene, OR 97405, USA
| | - Adam C Miller
- University of Oregon, Institute of Neuroscience, Eugene, OR 97405, USA.
| |
Collapse
|
6
|
Surette E, Donahue J, Robinson S, McKenna D, Martinez CS, Fitzgerald B, Karlstrom RO, Cumplido N, McMenamin SK. Adult caudal fin shape is imprinted in the embryonic fin fold. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.16.603744. [PMID: 39071346 PMCID: PMC11275767 DOI: 10.1101/2024.07.16.603744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Appendage shape is formed during development (and re-formed during regeneration) according to spatial and temporal cues that orchestrate local cellular morphogenesis. The caudal fin is the primary appendage used for propulsion in most fish species, and exhibits a range of distinct morphologies adapted for different swimming strategies, however the molecular mechanisms responsible for generating these diverse shapes remain mostly unknown. In zebrafish, caudal fins display a forked shape, with longer supportive bony rays at the periphery and shortest rays at the center. Here, we show that a premature, transient pulse of sonic hedgehog a (shha) overexpression during late embryonic development results in excess proliferation and growth of the central rays, causing the adult caudal fin to grow into a triangular, truncate shape. Both global and regional ectopic shha overexpression are sufficient to alter fin shape, and forked shape may be rescued by subsequent treatment with an antagonist of the canonical Shh pathway. The induced truncate fins show a decreased fin ray number and fail to form the hypural diastema that normally separates the dorsal and ventral fin lobes. While forked fins regenerate their original forked morphology, truncate fins regenerate truncate, suggesting that positional memory of the fin rays can be permanently altered by a transient treatment during embryogenesis. Ray finned fish have evolved a wide spectrum of caudal fin morphologies, ranging from truncate to forked, and the current work offers insights into the developmental mechanisms that may underlie this shape diversity.
Collapse
|
7
|
VanWinkle PE, Wynn B, Lee E, Nawara TJ, Thomas H, Parant JM, Alvarez C, Serra R, Sztul E. Lack of Nuclear Localization of the Creb3l1 Transcription Factor Causes Defects in Caudal Fin Bifurcation in Zebrafish Danio rerio. Cells Tissues Organs 2024; 214:77-95. [PMID: 38964305 PMCID: PMC11739433 DOI: 10.1159/000540103] [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: 11/21/2023] [Accepted: 06/27/2024] [Indexed: 07/06/2024] Open
Abstract
INTRODUCTION The formation of normal bone and bone healing requires the cAMP-responsive element binding protein 3-like-1 (Creb3l1) transmembrane transcription factor, as deletion of the murine CREB3L1 results in osteopenic animals with limited capacity to repair bone after a fracture. Creb3l1 undergoes regulated intramembrane proteolysis (RIP) to release the N-terminal transcription activating (TA) fragment that enters the nucleus and regulates the expression of target genes. METHODS To expand our understanding of Creb3l1's role in skeletal development and skeletal patterning, we aimed to generate animals expressing only the TA fragment of Creb3l1 lacking the transmembrane domain and thereby not regulated through RIP. However, the CRISPR/Cas9-mediated genome editing in zebrafish Danio rerio caused a frameshift mutation that added 56 random amino acids at the C-terminus of the TA fragment (TA+), making it unable to enter the nucleus. Thus, TA+ does not regulate transcription, and the creb3l1TA+/TA+ fish do not mediate creb3l1-dependent transcription. RESULTS We document that the creb3l1TA+/TA+ fish exhibit defects in the patterning of caudal fin lepidotrichia, with significantly distalized points of proximal bifurcation and decreased secondary bifurcations. Moreover, using the caudal fin amputation model, we show that creb3l1TA+/TA+ fish have decreased regeneration and that their regenerates replicate the distalization and bifurcation defects observed in intact fins of creb3l1TA+/TA+ animals. These defects correlate with altered expression of the shha and ptch2 components of the Sonic Hedgehog signaling pathway in creb3l1TA+/TA+ regenerates. CONCLUSION Together, our results uncover a previously unknown intersection between Creb3l1 and the Sonic Hedgehog pathway and document a novel role of Creb3l1 in tissue patterning. INTRODUCTION The formation of normal bone and bone healing requires the cAMP-responsive element binding protein 3-like-1 (Creb3l1) transmembrane transcription factor, as deletion of the murine CREB3L1 results in osteopenic animals with limited capacity to repair bone after a fracture. Creb3l1 undergoes regulated intramembrane proteolysis (RIP) to release the N-terminal transcription activating (TA) fragment that enters the nucleus and regulates the expression of target genes. METHODS To expand our understanding of Creb3l1's role in skeletal development and skeletal patterning, we aimed to generate animals expressing only the TA fragment of Creb3l1 lacking the transmembrane domain and thereby not regulated through RIP. However, the CRISPR/Cas9-mediated genome editing in zebrafish Danio rerio caused a frameshift mutation that added 56 random amino acids at the C-terminus of the TA fragment (TA+), making it unable to enter the nucleus. Thus, TA+ does not regulate transcription, and the creb3l1TA+/TA+ fish do not mediate creb3l1-dependent transcription. RESULTS We document that the creb3l1TA+/TA+ fish exhibit defects in the patterning of caudal fin lepidotrichia, with significantly distalized points of proximal bifurcation and decreased secondary bifurcations. Moreover, using the caudal fin amputation model, we show that creb3l1TA+/TA+ fish have decreased regeneration and that their regenerates replicate the distalization and bifurcation defects observed in intact fins of creb3l1TA+/TA+ animals. These defects correlate with altered expression of the shha and ptch2 components of the Sonic Hedgehog signaling pathway in creb3l1TA+/TA+ regenerates. CONCLUSION Together, our results uncover a previously unknown intersection between Creb3l1 and the Sonic Hedgehog pathway and document a novel role of Creb3l1 in tissue patterning.
Collapse
Affiliation(s)
- Peyton E. VanWinkle
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Bridge Wynn
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Eunjoo Lee
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Tomasz J. Nawara
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Holly Thomas
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - John M. Parant
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Cecilia Alvarez
- CIBICI-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Rosa Serra
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Elizabeth Sztul
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
8
|
Duong P, Rodriguez-Parks A, Kang J, Murphy PJ. CUT&Tag Applied to Zebrafish Adult Tail Fins Reveals a Return of Embryonic H3K4me3 Patterns During Regeneration. RESEARCH SQUARE 2024:rs.3.rs-4189493. [PMID: 38645155 PMCID: PMC11030498 DOI: 10.21203/rs.3.rs-4189493/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Regenerative potential is governed by a complex process of transcriptional reprogramming, involving chromatin reorganization and dynamics in transcription factor binding patterns throughout the genome. The degree to which chromatin and epigenetic changes contribute to this process remains partially understood. Here we provide a modified CUT&Tag protocol suitable for improved characterization and interrogation of epigenetic changes during adult fin regeneration in zebrafish. Our protocol generates data that recapitulates results from previously published ChIP-Seq methods, requires far fewer cells as input, and significantly improves signal to noise ratios. We deliver high-resolution enrichment maps for H3K4me3 of uninjured and regenerating fin tissues. During regeneration, we find that H3K4me3 levels increase over gene promoters which become transcriptionally active and genes which lose H3K4me3 become silenced. Interestingly, these epigenetic reprogramming events recapitulate the H3K4me3 patterns observed in developing fin folds of 24-hour old zebrafish embryos. Our results indicate that changes in genomic H3K4me3 patterns during fin regeneration occur in a manner consistent with reactivation of developmental programs, demonstrating CUT&Tag to be an effective tool for profiling chromatin landscapes in regenerating tissues.
Collapse
|
9
|
Cadiz L, Reed M, Monis S, Akimenko MA, Jonz MG. Identification of signalling pathways involved in gill regeneration in zebrafish. J Exp Biol 2024; 227:jeb246290. [PMID: 38099598 PMCID: PMC10906665 DOI: 10.1242/jeb.246290] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 12/04/2023] [Indexed: 01/31/2024]
Abstract
The occurrence of regeneration of the organs involved in respiratory gas exchange amongst vertebrates is heterogeneous. In some species of amphibians and fishes, the gills regenerate completely following resection or amputation, whereas in mammals, only partial, facultative regeneration of lung tissue occurs following injury. Given the homology between gills and lungs, the capacity of gill regeneration in aquatic species is of major interest in determining the underlying molecular or signalling pathways involved in respiratory organ regeneration. In the present study, we used adult zebrafish (Danio rerio) to characterize signalling pathways involved in the early stages of gill regeneration. Regeneration of the gills was induced by resection of gill filaments and observed over a period of up to 10 days. We screened for the effects on regeneration of the drugs SU5402, dorsomorphin and LY411575, which inhibit FGF, BMP or Notch signalling pathways, respectively. Exposure to each drug for 5 days significantly reduced regrowth of filament tips in regenerating tissue, compared with unresected controls. In separate experiments under normal conditions of regeneration, we used reverse transcription quantitative PCR and observed an increased expression of genes encoding for the bone morphogenetic factor, Bmp2b, fibroblast growth factor, Fgf8a, a transcriptional regulator (Her6) involved in Notch signalling, and Sonic Hedgehog (Shha), in regenerating gills at 10 day post-resection, compared with unresected controls. In situ hybridization confirmed that all four genes were expressed in regenerating gill tissue. This study implicates BMP, FGF, Notch and Shh signalling in gill regeneration in zebrafish.
Collapse
Affiliation(s)
- Laura Cadiz
- Department of Biology, University of Ottawa, Ottawa, ON, Canada, K1N 6N5
| | - Maddison Reed
- Department of Biology, University of Ottawa, Ottawa, ON, Canada, K1N 6N5
| | - Simon Monis
- Department of Biology, University of Ottawa, Ottawa, ON, Canada, K1N 6N5
| | | | - Michael G. Jonz
- Department of Biology, University of Ottawa, Ottawa, ON, Canada, K1N 6N5
| |
Collapse
|
10
|
Stewart S, Stankunas K. Section Immunostaining for Protein Expression and Cell Proliferation Studies of Regenerating Fins. Methods Mol Biol 2024; 2707:235-254. [PMID: 37668917 DOI: 10.1007/978-1-0716-3401-1_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/06/2023]
Abstract
Adult zebrafish fins fully regenerate after resection, providing a highly accessible and remarkable vertebrate model of organ regeneration. Fin injury triggers wound epidermis formation and the dedifferentiation of injury-adjacent mature cells to establish an organized blastema of progenitor cells. Balanced cell proliferation and redifferentiation along with cell movements then progressively reestablish patterned tissues and restore the fin to its original size and shape. A mechanistic understanding of these coordinated cell behaviors and transitions requires direct knowledge of proteins in their physiological context, including expression, subcellular localization, and activity. Antibody-based staining of sectioned fins facilitates such high-resolution analyses of specific, native proteins. Therefore, such methods are mainstays of comprehensive, hypothesis-driven fin regeneration studies. However, section immunostaining requires labor-intensive, empirical optimization. Here, we present detailed, multistep procedures for antibody staining and co-detecting proliferating cells using paraffin and frozen fin sections. We include suggestions to avoid common pitfalls and to streamline the development of optimized, validated protocols for new and challenging antibodies.
Collapse
Affiliation(s)
- Scott Stewart
- Institute of Molecular Biology, University of Oregon, Eugene, OR, USA.
| | - Kryn Stankunas
- Institute of Molecular Biology, University of Oregon, Eugene, OR, USA.
- Department of Biology, University of Oregon, Eugene, OR, USA.
| |
Collapse
|
11
|
Lukowicz-Bedford RM, Eisen JS, Miller AC. Gap junction mediated bioelectric coordination is required for slow muscle development, organization, and function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.20.572619. [PMID: 38187655 PMCID: PMC10769300 DOI: 10.1101/2023.12.20.572619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Bioelectrical signaling, intercellular communication facilitated by membrane potential and electrochemical coupling, is emerging as a key regulator of animal development. Gap junction (GJ) channels can mediate bioelectric signaling by creating a fast, direct pathway between cells for the movement of ions and other small molecules. In vertebrates, GJ channels are formed by a highly conserved transmembrane protein family called the Connexins. The connexin gene family is large and complex, presenting a challenge in identifying the specific Connexins that create channels within developing and mature tissues. Using the embryonic zebrafish neuromuscular system as a model, we identify a connexin conserved across vertebrate lineages, gjd4, which encodes the Cx46.8 protein, that mediates bioelectric signaling required for appropriate slow muscle development and function. Through a combination of mutant analysis and in vivo imaging we show that gjd4/Cx46.8 creates GJ channels specifically in developing slow muscle cells. Using genetics, pharmacology, and calcium imaging we find that spinal cord generated neural activity is transmitted to developing slow muscle cells and synchronized activity spreads via gjd4/Cx46.8 GJ channels. Finally, we show that bioelectrical signal propagation within the developing neuromuscular system is required for appropriate myofiber organization, and that disruption leads to defects in behavior. Our work reveals the molecular basis for GJ communication among developing muscle cells and reveals how perturbations to bioelectric signaling in the neuromuscular system_may contribute to developmental myopathies. Moreover, this work underscores a critical motif of signal propagation between organ systems and highlights the pivotal role played by GJ communication in coordinating bioelectric signaling during development.
Collapse
|
12
|
Rajan AM, Rosin NL, Labit E, Biernaskie J, Liao S, Huang P. Single-cell analysis reveals distinct fibroblast plasticity during tenocyte regeneration in zebrafish. SCIENCE ADVANCES 2023; 9:eadi5771. [PMID: 37967180 PMCID: PMC10651129 DOI: 10.1126/sciadv.adi5771] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Accepted: 10/16/2023] [Indexed: 11/17/2023]
Abstract
Despite their importance in tissue maintenance and repair, fibroblast diversity and plasticity remain poorly understood. Using single-cell RNA sequencing, we uncover distinct sclerotome-derived fibroblast populations in zebrafish, including progenitor-like perivascular/interstitial fibroblasts, and specialized fibroblasts such as tenocytes. To determine fibroblast plasticity in vivo, we develop a laser-induced tendon ablation and regeneration model. Lineage tracing reveals that laser-ablated tenocytes are quickly regenerated by preexisting fibroblasts. By combining single-cell clonal analysis and live imaging, we demonstrate that perivascular/interstitial fibroblasts actively migrate to the injury site, where they proliferate and give rise to new tenocytes. By contrast, perivascular fibroblast-derived pericytes or specialized fibroblasts, including tenocytes, exhibit no regenerative plasticity. Active Hedgehog (Hh) signaling is required for the proliferation of activated fibroblasts to ensure efficient tenocyte regeneration. Together, our work highlights the functional diversity of fibroblasts and establishes perivascular/interstitial fibroblasts as tenocyte progenitors that promote tendon regeneration in a Hh signaling-dependent manner.
Collapse
Affiliation(s)
- Arsheen M. Rajan
- Department of Biochemistry and Molecular Biology, Alberta Children’s Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Nicole L. Rosin
- Faculty of Veterinary Medicine, Alberta Children’s Hospital Research Institute, Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Elodie Labit
- Faculty of Veterinary Medicine, Alberta Children’s Hospital Research Institute, Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Jeff Biernaskie
- Faculty of Veterinary Medicine, Alberta Children’s Hospital Research Institute, Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Shan Liao
- Inflammation Research Network, Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Peng Huang
- Department of Biochemistry and Molecular Biology, Alberta Children’s Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| |
Collapse
|
13
|
Jing J, Wu Z, Wang J, Luo G, Lin H, Fan Y, Zhou C. Hedgehog signaling in tissue homeostasis, cancers, and targeted therapies. Signal Transduct Target Ther 2023; 8:315. [PMID: 37596267 PMCID: PMC10439210 DOI: 10.1038/s41392-023-01559-5] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 07/05/2023] [Indexed: 08/20/2023] Open
Abstract
The past decade has seen significant advances in our understanding of Hedgehog (HH) signaling pathway in various biological events. HH signaling pathway exerts its biological effects through a complex signaling cascade involved with primary cilium. HH signaling pathway has important functions in embryonic development and tissue homeostasis. It plays a central role in the regulation of the proliferation and differentiation of adult stem cells. Importantly, it has become increasingly clear that HH signaling pathway is associated with increased cancer prevalence, malignant progression, poor prognosis and even increased mortality. Understanding the integrative nature of HH signaling pathway has opened up the potential for new therapeutic targets for cancer. A variety of drugs have been developed, including small molecule inhibitors, natural compounds, and long non-coding RNA (LncRNA), some of which are approved for clinical use. This review outlines recent discoveries of HH signaling in tissue homeostasis and cancer and discusses how these advances are paving the way for the development of new biologically based therapies for cancer. Furthermore, we address status quo and limitations of targeted therapies of HH signaling pathway. Insights from this review will help readers understand the function of HH signaling in homeostasis and cancer, as well as opportunities and challenges of therapeutic targets for cancer.
Collapse
Affiliation(s)
- Junjun Jing
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Zhuoxuan Wu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
- Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Jiahe Wang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
- Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Guowen Luo
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
- Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Hengyi Lin
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
- Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Yi Fan
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - Chenchen Zhou
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
- Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
14
|
Robbins AE, Horst SG, Lewis VM, Stewart S, Stankunas K. The Fraser complex interconnects tissue layers to support basal epidermis and osteoblast integrated morphogenesis underlying fin skeletal patterning. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.08.548238. [PMID: 37461516 PMCID: PMC10350090 DOI: 10.1101/2023.07.08.548238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Fraser Syndrome is a rare, multisystemic autosomal recessive disorder characterized by disrupted epithelial-mesenchymal associations upon loss of Fraser Complex genes. Disease manifestation and affected organs are highly variable. Digit malformations such as syndactyly are common but of unclear developmental origins. We explored if zebrafish fraser extracellular matrix complex subunit 1 (fras1) mutants model Fraser Syndrome-associated appendicular skeleton patterning defects. Approximately 10% of fras1 mutants survive to adulthood, displaying striking and varied fin abnormalities, including endochondral bone fusions, ectopic cartilage, and disrupted caudal fin symmetry. The fins of surviving fras1 mutants frequently have fewer and unbranched bony rays. fras1 mutant fins regenerate to their original size but with exacerbated ray branching and fin symmetry defects. Single cell RNA-Seq analysis, in situ hybridizations, and antibody staining show specific Fraser complex expression in the basal epidermis during regenerative outgrowth. Fras1 and Fraser Complex component Frem2 accumulate along the basal side of distal-most basal epidermal cells. Greatly reduced and mislocalized Frem2 accompanies loss of Fras1 in fras1 mutants. The Sonic hedgehog signaling between distal basal epidermis and adjacent mesenchymal pre-osteoblasts that promotes ray branching persists upon Fraser Complex loss. However, fras1 mutant regenerating fins exhibit extensive sub-epidermal blistering associated with a disorganized basal epidermis and adjacent pre-osteoblasts. We propose Fraser Complex-supported tissue layer adhesion enables robust integrated tissue morphogenesis involving the basal epidermis and osteoblasts. Further, we establish zebrafish fin development and regeneration as an accessible model to explore mechanisms of Fraser Syndrome-associated digit defects and Fraser Complex function at epithelial-mesenchymal interfaces.
Collapse
|
15
|
Harper M, Hu Y, Donahue J, Acosta B, Dievenich Braes F, Nguyen S, Zeng J, Barbaro J, Lee H, Bui H, McMenamin SK. Thyroid hormone regulates proximodistal patterning in fin rays. Proc Natl Acad Sci U S A 2023; 120:e2219770120. [PMID: 37186843 PMCID: PMC10214145 DOI: 10.1073/pnas.2219770120] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 03/26/2023] [Indexed: 05/17/2023] Open
Abstract
Processes that regulate size and patterning along an axis must be highly integrated to generate robust shapes; relative changes in these processes underlie both congenital disease and evolutionary change. Fin length mutants in zebrafish have provided considerable insight into the pathways regulating fin size, yet signals underlying patterning have remained less clear. The bony rays of the fins possess distinct patterning along the proximodistal axis, reflected in the location of ray bifurcations and the lengths of ray segments, which show progressive shortening along the axis. Here, we show that thyroid hormone (TH) regulates aspects of proximodistal patterning of the caudal fin rays, regardless of fin size. TH promotes distal gene expression patterns, coordinating ray bifurcations and segment shortening with skeletal outgrowth along the proximodistal axis. This distalizing role for TH is conserved between development and regeneration, in all fins (paired and medial), and between Danio species as well as distantly related medaka. During regenerative outgrowth, TH acutely induces Shh-mediated skeletal bifurcation. Zebrafish have multiple nuclear TH receptors, and we found that unliganded Thrab-but not Thraa or Thrb-inhibits the formation of distal features. Broadly, these results demonstrate that proximodistal morphology is regulated independently from size-instructive signals. Modulating proximodistal patterning relative to size-either through changes to TH metabolism or other hormone-independent pathways-can shift skeletal patterning in ways that recapitulate aspects of fin ray diversity found in nature.
Collapse
Affiliation(s)
- Melody Harper
- Biology Department, Morrissey College of Arts and Sciences, Boston College, Chestnut Hill, MA02467
| | - Yinan Hu
- Biology Department, Morrissey College of Arts and Sciences, Boston College, Chestnut Hill, MA02467
| | - Joan Donahue
- Biology Department, Morrissey College of Arts and Sciences, Boston College, Chestnut Hill, MA02467
| | - Benjamin Acosta
- Biology Department, Morrissey College of Arts and Sciences, Boston College, Chestnut Hill, MA02467
| | - Flora Dievenich Braes
- Biology Department, Morrissey College of Arts and Sciences, Boston College, Chestnut Hill, MA02467
| | - Stacy Nguyen
- Biology Department, Morrissey College of Arts and Sciences, Boston College, Chestnut Hill, MA02467
| | - Jenny Zeng
- Biology Department, Morrissey College of Arts and Sciences, Boston College, Chestnut Hill, MA02467
| | - Julianna Barbaro
- Biology Department, Morrissey College of Arts and Sciences, Boston College, Chestnut Hill, MA02467
| | - Hyungwoo Lee
- Biology Department, Morrissey College of Arts and Sciences, Boston College, Chestnut Hill, MA02467
| | - Hoa Bui
- Biology Department, Morrissey College of Arts and Sciences, Boston College, Chestnut Hill, MA02467
| | - Sarah K. McMenamin
- Biology Department, Morrissey College of Arts and Sciences, Boston College, Chestnut Hill, MA02467
| |
Collapse
|
16
|
Rees L, König D, Jaźwińska A. Regeneration of the dermal skeleton and wound epidermis formation depend on BMP signaling in the caudal fin of platyfish. Front Cell Dev Biol 2023; 11:1134451. [PMID: 36846592 PMCID: PMC9946992 DOI: 10.3389/fcell.2023.1134451] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 01/24/2023] [Indexed: 02/11/2023] Open
Abstract
Fin regeneration has been extensively studied in zebrafish, a genetic model organism. Little is known about regulators of this process in distant fish taxa, such as the Poeciliidae family, represented by the platyfish. Here, we used this species to investigate the plasticity of ray branching morphogenesis following either straight amputation or excision of ray triplets. This approach revealed that ray branching can be conditionally shifted to a more distal position, suggesting non-autonomous regulation of bone patterning. To gain molecular insights into regeneration of fin-specific dermal skeleton elements, actinotrichia and lepidotrichia, we localized expression of the actinodin genes and bmp2 in the regenerative outgrowth. Blocking of the BMP type-I receptor suppressed phospho-Smad1/5 immunoreactivity, and impaired fin regeneration after blastema formation. The resulting phenotype was characterized by the absence of bone and actinotrichia restoration. In addition, the wound epidermis displayed extensive thickening. This malformation was associated with expanded Tp63 expression from the basal epithelium towards more superficial layers, suggesting abnormal tissue differentiation. Our data add to the increasing evidence for the integrative role of BMP signaling in epidermal and skeletal tissue formation during fin regeneration. This expands our understanding of common mechanisms guiding appendage restoration in diverse clades of teleosts.
Collapse
Affiliation(s)
- Lana Rees
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Désirée König
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | | |
Collapse
|
17
|
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
|
18
|
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
|
19
|
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
|
20
|
Mutation of foxl1 Results in Reduced Cartilage Markers in a Zebrafish Model of Otosclerosis. Genes (Basel) 2022; 13:genes13071107. [PMID: 35885890 PMCID: PMC9319681 DOI: 10.3390/genes13071107] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 06/08/2022] [Accepted: 06/11/2022] [Indexed: 02/05/2023] Open
Abstract
Bone diseases such as otosclerosis (conductive hearing loss) and osteoporosis (low bone mineral density) can result from the abnormal expression of genes that regulate cartilage and bone development. The forkhead box transcription factor FOXL1 has been identified as the causative gene in a family with autosomal dominant otosclerosis and has been reported as a candidate gene in GWAS meta-analyses for osteoporosis. This potentially indicates a novel role for foxl1 in chondrogenesis, osteogenesis, and bone remodelling. We created a foxl1 mutant zebrafish strain as a model for otosclerosis and osteoporosis and examined jaw bones that are homologous to the mammalian middle ear bones, and mineralization of the axial skeleton. We demonstrate that foxl1 regulates the expression of collagen genes such as collagen type 1 alpha 1a and collagen type 11 alpha 2, and results in a delay in jawbone mineralization, while the axial skeleton remains unchanged. foxl1 may also act with other forkhead genes such as foxc1a, as loss of foxl1 in a foxc1a mutant background increases the severity of jaw calcification phenotypes when compared to each mutant alone. Our zebrafish model demonstrates atypical cartilage formation and mineralization in the zebrafish craniofacial skeleton in foxl1 mutants and demonstrates that aberrant collagen expression may underlie the development of otosclerosis.
Collapse
|
21
|
Iwasaki M, Kawakami K, Wada H. Remodeling of the hyomandibular skeleton and facial nerve positioning during embryonic and postembryonic development of teleost fish. Dev Biol 2022; 489:134-145. [PMID: 35750208 DOI: 10.1016/j.ydbio.2022.06.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 06/03/2022] [Accepted: 06/16/2022] [Indexed: 11/30/2022]
Abstract
The vertebrate skeleton changes its shape during development through the activities of chondrocytes, osteoblasts and osteoclasts. Although much is known about the mechanisms for differentiation in these cells, it is less understood how they behave in a region-specific manner to acquire unique bone shapes. To address this question, we investigated the development of the hyomandibular (Hm) system in zebrafish. The Hm originates as cartilage carrying a single foramen (the Hm foramen), through which the facial (VII) nerve passes. We reveal that Schwann cells, which myelinate the VII nerve, regulate rearrangement of the chondrocytes to enlarge the Hm foramen. The Hm cartilage then becomes ossified in the perichondrium, where the marrow chondrocytes are replaced by adipocytes. Then, the bone matrix along the VII nerve is resorbed by osteoclasts, generating a gateway to the bone marrow. Subsequent movement of the VII nerve into the marrow, followed by deposition of new bone matrix, isolates the nerve from the jaw muscle insertion. Genetic ablation of osteoblasts and osteoclasts reveals specific roles of these cells during remodeling processes. Interestingly, the VII nerve relocation does not occur in medaka; instead, bone deposition distinct from those in zebrafish separates the VII nerve from the muscle insertion. Our results define novel mechanisms for skeletal remodeling, by which the bone shapes in a region- and species-specific manner.
Collapse
Affiliation(s)
- Miki Iwasaki
- College of Liberal Arts and Sciences, Kitasato University, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa, 252-0373, Japan
| | - Koichi Kawakami
- National Institute of Genetics; Graduate University for Advanced Studies (SOKENDAI), 1111 Yata, Mishima, Shizuoka 411-8540, Japan
| | - Hironori Wada
- College of Liberal Arts and Sciences, Kitasato University, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa, 252-0373, Japan.
| |
Collapse
|
22
|
Zhang T, Zhang C, Zhang J, Lin J, Song D, Zhang P, Liu Y, Chen L, Zhang L. Cadmium impairs zebrafish swim bladder development via ROS mediated inhibition of the Wnt / Hedgehog pathway. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2022; 247:106180. [PMID: 35490551 DOI: 10.1016/j.aquatox.2022.106180] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 04/15/2022] [Accepted: 04/23/2022] [Indexed: 06/14/2023]
Abstract
The posterior swim bladder is an important organ in teleost fishes, that primarily maintains buoyancy and motility for swimming and survival. In this study, we examined the molecular mechanisms of the toxicity of cadmium (Cd) on the early development of the swim bladder in zebrafish. Embryonic Cd exposure resulted in the non-inflation of the swim bladder when the ambient Cd concentration was greater than or equal to 0.25 mg/L. Cd disturbed surfactant lipid distribution and inhibited the formation of all three tissue layers in the swim bladder. Additionally, excessive Cd down-regulated Wnt (fzd3, nkd1, fzd7 and axin2) and Hedgehog (ihh, shh, ptc1 and ptc2) signaling pathways. Conversely, Wnt signaling activation partially neutralized Cd-induced swim bladder developmental defects. Moreover, ROS scavenger reduced Glutathione (GSH) effectively recovered Cd induced defects in swim bladder and Wnt/Hedgehog signaling. Taken together, our results first revealed that Cd caused swim bladder developmental defects via ROS-mediated inhibition of the Wnt and Hedgehog pathways. These results herein provide important data for future toxicological studies and risk assessments of Cd.
Collapse
Affiliation(s)
- Ting Zhang
- Key Laboratory of Tropical Marine Bio-resourcesand Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China; Southern Marine Science and Engineering Guangdong Laboratory, Guangzhou 511458, China; Institution of South China Sea Ecology and Environmental Engineering, Chinese Academy of Sciences, Guangzhou 510301, China
| | - Canchuan Zhang
- Key Laboratory of Tropical Marine Bio-resourcesand Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China; Southern Marine Science and Engineering Guangdong Laboratory, Guangzhou 511458, China; University of Chinese Academy of Science, Beijing 100049, China
| | - Jin Zhang
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, School of life Sciences, Sun Yat-Sen University, Guangzhou 510275, China
| | - Jiangtian Lin
- Key Laboratory of Tropical Marine Bio-resourcesand Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China; Southern Marine Science and Engineering Guangdong Laboratory, Guangzhou 511458, China; University of Chinese Academy of Science, Beijing 100049, China
| | - Dongdong Song
- Key Laboratory of Tropical Marine Bio-resourcesand Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China; Southern Marine Science and Engineering Guangdong Laboratory, Guangzhou 511458, China; University of Chinese Academy of Science, Beijing 100049, China
| | - Peng Zhang
- Key Laboratory of Tropical Marine Bio-resourcesand Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China; Southern Marine Science and Engineering Guangdong Laboratory, Guangzhou 511458, China; University of Chinese Academy of Science, Beijing 100049, China
| | - Yang Liu
- Key Laboratory of Tropical Marine Bio-resourcesand Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China; Southern Marine Science and Engineering Guangdong Laboratory, Guangzhou 511458, China; University of Chinese Academy of Science, Beijing 100049, China
| | - Lizhao Chen
- Key Laboratory of Tropical Marine Bio-resourcesand Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China; Southern Marine Science and Engineering Guangdong Laboratory, Guangzhou 511458, China; University of Chinese Academy of Science, Beijing 100049, China
| | - Li Zhang
- Key Laboratory of Tropical Marine Bio-resourcesand Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China; Southern Marine Science and Engineering Guangdong Laboratory, Guangzhou 511458, China; Institution of South China Sea Ecology and Environmental Engineering, Chinese Academy of Sciences, Guangzhou 510301, China.
| |
Collapse
|
23
|
Purushothaman S, Lopez Aviña BB, Seifert AW. Sonic hedgehog is Essential for Proximal-Distal Outgrowth of the Limb Bud in Salamanders. Front Cell Dev Biol 2022; 10:797352. [PMID: 35433673 PMCID: PMC9010949 DOI: 10.3389/fcell.2022.797352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 01/24/2022] [Indexed: 11/20/2022] Open
Abstract
The developing forelimb has been a foundational model to understand how specified progenitor cells integrate genetic information to produce the tetrapod limb bauplan. Although the reigning hypothesis is that all tetrapods develop limbs in a similar manner, recent work suggests that urodeles have evolved a derived mode of limb dvelopment. Here, we demonstrate through pharmacological and genetic inactivation of Sonic hedgehog (Shh) signaling in axolotls that Shh directs expansion and survival of limb progenitor cells in addition to patterning the limb across the proximodistal and antero-posterior axis. In contrast to inactivation of Shh in mouse or chick embryos where a humerus, radius, and single digit develop, Shh crispant axolotls completely lack forelimbs. In rescuing limb development by implanting SHH-N protein beads into the nascent limb field of Shh crispants, we show that the limb field is specified in the absence of Shh and that hedgehog pathway activation is required to initiate proximodistal outgrowth. When our results are examined alongside other derived aspects of salamander limb development and placed in a phylogenetic context, a new hypothesis emerges whereby the ability for cells at an amputation plane to activate morphogenesis and regenerate a limb may have evolved uniquely in urodeles.
Collapse
|
24
|
Thauvin M, de Sousa RM, Alves M, Volovitch M, Vriz S, Rampon C. An early Shh-H2O2 reciprocal regulatory interaction controls the regenerative program during zebrafish fin regeneration. J Cell Sci 2022; 135:274206. [PMID: 35107164 DOI: 10.1242/jcs.259664] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 01/24/2022] [Indexed: 11/20/2022] Open
Abstract
Reactive oxygen species (ROS), originally classified as toxic molecules, have attracted increasing interest given their actions in cell signaling. Hydrogen peroxide (H2O2), the major ROS produced by cells, acts as a second messenger to modify redox-sensitive proteins or lipids. After caudal fin amputation, tight spatiotemporal regulation of ROS is required first for wound healing and later to initiate the regenerative program. However, the mechanisms carrying out this sustained ROS production and their integration with signaling pathways are still poorly understood. We focused on the early dialog between H2O2 and Sonic Hedgehog (Shh) during fin regeneration. We demonstrate that H2O2 controls Shh expression and that Shh in turn regulates the H2O2 level via a canonical pathway. Moreover, the means of this tight reciprocal control change during the successive phases of the regenerative program. Dysregulation of the Hedgehog pathway has been implicated in several developmental syndromes, diabetes and cancer. These data support the existence of an early positive crosstalk between Shh and H2O2 that might be more generally involved in various processes paving the way to improve regenerative processes, particularly in vertebrates.
Collapse
Affiliation(s)
- Marion Thauvin
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS, INSERM, PSL Research University, Paris, France.,Sorbonne Université, Paris, France
| | - Rodolphe Matias de Sousa
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS, INSERM, PSL Research University, Paris, France.,Sorbonne Université, Paris, France
| | - Marine Alves
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS, INSERM, PSL Research University, Paris, France.,Université de Paris, Faculty of Sciences, Paris, France
| | - Michel Volovitch
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS, INSERM, PSL Research University, Paris, France.,École Normale Supérieure, PSL Research University, Department of Biology, Paris, France
| | - Sophie Vriz
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS, INSERM, PSL Research University, Paris, France.,Université de Paris, Faculty of Sciences, Paris, France
| | - Christine Rampon
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS, INSERM, PSL Research University, Paris, France.,Université de Paris, Faculty of Sciences, Paris, France
| |
Collapse
|
25
|
Kaliya-Perumal AK, Ingham PW. Musculoskeletal regeneration: A zebrafish perspective. Biochimie 2021; 196:171-181. [PMID: 34715269 DOI: 10.1016/j.biochi.2021.10.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 10/17/2021] [Accepted: 10/22/2021] [Indexed: 12/18/2022]
Abstract
Musculoskeletal injuries are common in humans. The cascade of cellular and molecular events following such injuries results either in healing with functional recovery or scar formation. While fibrotic scar tissue serves to bridge between injured planes, it undermines functional integrity. Hence, faithful regeneration is the most desired outcome; however, the potential to regenerate is limited in humans. In contrast, various non-mammalian vertebrates have fascinating capabilities of regenerating even an entire appendage following amputation. Among them, zebrafish is an important and accessible laboratory model organism, sharing striking similarities with mammalian embryonic musculoskeletal development. Moreover, clinically relevant muscle and skeletal injury zebrafish models recapitulate mammalian regeneration. Upon muscle injury, quiescent stem cells - known as satellite cells - become activated, proliferate, differentiate and fuse to form new myofibres, while bone fracture results in a phased response involving hematoma formation, inflammation, fibrocartilaginous callus formation, bony callus formation and remodelling. These models are well suited to testing gene- or pharmaco-therapy for the benefit of conditions like muscle tears and fractures. Insights from further studies on whole body part regeneration, a hallmark of the zebrafish model, have the potential to complement regenerative strategies to achieve faster and desired healing following injuries without any scar formation and, in the longer run, drive progress towards the realisation of large-scale regeneration in mammals. Here, we provide an overview of the basic mechanisms of musculoskeletal regeneration, highlight the key features of zebrafish as a regenerative model and outline the relevant studies that have contributed to the advancement of this field.
Collapse
Affiliation(s)
- Arun-Kumar Kaliya-Perumal
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 59 Nanyang Drive, Singapore 636921, Singapore.
| | - Philip W Ingham
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 59 Nanyang Drive, Singapore 636921, Singapore.
| |
Collapse
|
26
|
Roan HY, Tseng TL, Chen CH. Whole-body clonal mapping identifies giant dominant clones in zebrafish skin epidermis. Development 2021; 148:272161. [PMID: 34463754 DOI: 10.1242/dev.199669] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 08/20/2021] [Indexed: 12/14/2022]
Abstract
Skin expansion during development is predominantly driven by growth of basal epithelial cell (BEC)-derived clonal populations, which often display varied sizes and shapes. However, little is known about the causes of clonal heterogeneity and the maximum size to which a single clone can grow. Here, we created a zebrafish model, basebow, for capturing clonal growth behavior in the BEC population on a whole-body, centimeter scale. By tracking 222 BECs over the course of a 28-fold expansion of body surface area, we determined that most BECs survive and grow clonal populations with an average size of 0.013 mm2. An extensive survey of 742 sparsely labeled BECs further revealed that giant dominant clones occasionally arise on specific body regions, covering up to 0.6% of the surface area. Additionally, a growth-induced extracellular matrix component, Lamb1a, mediates clonal growth in a cell-autonomous manner. Altogether, our findings demonstrate how clonal heterogeneity and clonal dominance may emerge to enable post-embryonic growth of a vertebrate organ, highlighting key cellular mechanisms that may only become evident when visualizing single cell behavior at the whole-animal level.
Collapse
Affiliation(s)
- Hsiao-Yuh Roan
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 11529, Taiwan
| | - Tzu-Lun Tseng
- 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
|
27
|
Braunstein JA, Robbins AE, Stewart S, Stankunas K. Basal epidermis collective migration and local Sonic hedgehog signaling promote skeletal branching morphogenesis in zebrafish fins. Dev Biol 2021; 477:177-190. [PMID: 34038742 PMCID: PMC10802891 DOI: 10.1016/j.ydbio.2021.04.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 04/29/2021] [Accepted: 04/30/2021] [Indexed: 12/23/2022]
Abstract
Teleost fish fins, like all vertebrate limbs, comprise a series of bones laid out in characteristic pattern. Each fin's distal bony rays typically branch to elaborate skeletal networks providing form and function. Zebrafish caudal fin regeneration studies suggest basal epidermal-expressed Sonic hedgehog (Shh) promotes ray branching by partitioning pools of adjacent pre-osteoblasts. This Shh role is distinct from its well-studied Zone of Polarizing Activity role establishing paired limb positional information. Therefore, we investigated if and how Shh signaling similarly functions during developmental ray branching of both paired and unpaired fins while resolving cellular dynamics of branching by live imaging. We found shha is expressed uniquely by basal epidermal cells overlying pre-osteoblast pools at the distal aspect of outgrowing juvenile fins. Lateral splitting of each shha-expressing epidermal domain followed by the pre-osteoblast pools precedes overt ray branching. We use ptch2:Kaede fish and Kaede photoconversion to identify short stretches of shha+basal epidermis and juxtaposed pre-osteoblasts as the Shh/Smoothened (Smo) active zone. Basal epidermal distal collective movements continuously replenish each shha+domain with individual cells transiently expressing and responding to Shh. In contrast, pre-osteoblasts maintain Shh/Smo activity until differentiating. The Smo inhibitor BMS-833923 prevents branching in all fins, paired and unpaired, with surprisingly minimal effects on caudal fin initial skeletal patterning, ray outgrowth or bone differentiation. Staggered BMS-833923 addition indicates Shh/Smo signaling acts throughout the branching process. We use live cell tracking to find Shh/Smo restrains the distal movement of basal epidermal cells by apparent 'tethering' to pre-osteoblasts. We propose short-range Shh/Smo signaling promotes these heterotypic associations to couple instructive basal epidermal collective movements to pre-osteoblast repositioning as a unique mode of branching morphogenesis.
Collapse
Affiliation(s)
- Joshua A Braunstein
- 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, 1370 Franklin Blvd, Eugene, OR, 97403-1210, 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, 1370 Franklin Blvd, Eugene, OR, 97403-1210, USA
| | - Scott Stewart
- 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, 1370 Franklin Blvd, Eugene, OR, 97403-1210, USA.
| |
Collapse
|
28
|
Dagenais P, Blanchoud S, Pury D, Pfefferli C, Aegerter-Wilmsen T, Aegerter CM, Jaźwińska A. Hydrodynamic stress and phenotypic plasticity of the zebrafish regenerating fin. J Exp Biol 2021; 224:271142. [PMID: 34338301 DOI: 10.1242/jeb.242309] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 06/14/2021] [Indexed: 01/23/2023]
Abstract
Understanding how extrinsic factors modulate genetically encoded information to produce a specific phenotype is of prime scientific interest. In particular, the feedback mechanism between abiotic forces and locomotory organs during morphogenesis to achieve efficient movement is a highly relevant example of such modulation. The study of this developmental process can provide unique insights on the transduction of cues at the interface between physics and biology. Here, we take advantage of the natural ability of adult zebrafish to regenerate their amputated fins to assess its morphogenic plasticity upon external modulations. Using a variety of surgical and chemical treatments, we could induce phenotypic responses to the structure of the fin. Through the ablation of specific rays in regenerating caudal fins, we generated artificially narrowed appendages in which the fin cleft depth and the positioning of rays bifurcations were perturbed compared with normal regenerates. To dissect the role of mechanotransduction in this process, we investigated the patterns of hydrodynamic forces acting on the surface of a zebrafish fin during regeneration by using particle tracking velocimetry on a range of biomimetic hydrofoils. This experimental approach enabled us to quantitatively compare hydrodynamic stress distributions over flapping fins of varying sizes and shapes. As a result, viscous shear stress acting on the distal margin of regenerating fins and the resulting internal tension are proposed as suitable signals for guiding the regulation of ray growth dynamics and branching pattern. Our findings suggest that mechanical forces are involved in the fine-tuning of the locomotory organ during fin morphogenesis.
Collapse
Affiliation(s)
- Paule Dagenais
- Physik-Institut, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Simon Blanchoud
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700 Fribourg, Switzerland
| | - David Pury
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700 Fribourg, Switzerland
| | - Catherine Pfefferli
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700 Fribourg, Switzerland
| | - Tinri Aegerter-Wilmsen
- Department of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Christof M Aegerter
- Physik-Institut, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland.,Department of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Anna Jaźwińska
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700 Fribourg, Switzerland
| |
Collapse
|
29
|
Abstract
Species that can regrow their lost appendages have been studied with the ultimate aim of developing methods to enable human limb regeneration. These examinations highlight that appendage regeneration progresses through shared tissue stages and gene activities, leading to the assumption that appendage regeneration paradigms (e.g. tails and limbs) are the same or similar. However, recent research suggests these paradigms operate differently at the cellular level, despite sharing tissue descriptions and gene expressions. Here, collecting the findings from disparate studies, I argue appendage regeneration is context dependent at the cellular level; nonetheless, it requires (i) signalling centres, (ii) stem/progenitor cell types and (iii) a regeneration-permissive environment, and these three common cellular principles could be more suitable for cross-species/paradigm/age comparisons.
Collapse
Affiliation(s)
- Can Aztekin
- School of Life Sciences, Swiss Federal Institute of Technology Lausanne (EPFL), 1015 Lausanne, Switzerland
| |
Collapse
|
30
|
Daponte V, Tylzanowski P, Forlino A. Appendage Regeneration in Vertebrates: What Makes This Possible? Cells 2021; 10:cells10020242. [PMID: 33513779 PMCID: PMC7911911 DOI: 10.3390/cells10020242] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/18/2021] [Accepted: 01/22/2021] [Indexed: 12/26/2022] Open
Abstract
The ability to regenerate amputated or injured tissues and organs is a fascinating property shared by several invertebrates and, interestingly, some vertebrates. The mechanism of evolutionary loss of regeneration in mammals is not understood, yet from the biomedical and clinical point of view, it would be very beneficial to be able, at least partially, to restore that capability. The current availability of new experimental tools, facilitating the comparative study of models with high regenerative ability, provides a powerful instrument to unveil what is needed for a successful regeneration. The present review provides an updated overview of multiple aspects of appendage regeneration in three vertebrates: lizard, salamander, and zebrafish. The deep investigation of this process points to common mechanisms, including the relevance of Wnt/β-catenin and FGF signaling for the restoration of a functional appendage. We discuss the formation and cellular origin of the blastema and the identification of epigenetic and cellular changes and molecular pathways shared by vertebrates capable of regeneration. Understanding the similarities, being aware of the differences of the processes, during lizard, salamander, and zebrafish regeneration can provide a useful guide for supporting effective regenerative strategies in mammals.
Collapse
Affiliation(s)
- Valentina Daponte
- Biochemistry Unit, Department of Molecular Medicine, University of Pavia, via Taramelli 3/B, 27100 Pavia, Italy;
| | - Przemko Tylzanowski
- Skeletal Biology and Engineering Research Center, Department of Development and Regeneration, University of Leuven, 3000 Leuven, Belgium;
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 20-059 Lublin, Poland
| | - Antonella Forlino
- Biochemistry Unit, Department of Molecular Medicine, University of Pavia, via Taramelli 3/B, 27100 Pavia, Italy;
- Correspondence: ; Tel.: +39-0382-987235
| |
Collapse
|
31
|
Lebedeva L, Zhumabayeva B, Gebauer T, Kisselev I, Aitasheva Z. Zebrafish ( Danio rerio) as a Model for Understanding the Process of Caudal Fin Regeneration. Zebrafish 2020; 17:359-372. [PMID: 33259770 DOI: 10.1089/zeb.2020.1926] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
After its introduction for scientific investigation in the 1950s, the cypriniform zebrafish, Danio rerio, has become a valuable model for the study of regenerative processes and mechanisms. Zebrafish exhibit epimorphic regeneration, in which a nondifferentiated cell mass formed after amputation is able to fully regenerate lost tissue such as limbs, heart muscle, brain, retina, and spinal cord. The process of limb regeneration in zebrafish comprises several stages characterized by the activation of specific signaling pathways and gene expression. We review current research on key factors in limb regeneration using zebrafish as a model.
Collapse
Affiliation(s)
- Lina Lebedeva
- Department of Molecular Biology and Genetics, Faculty of Biology and Biotechnology, al-Farabi Kazakh National University, Almaty, The Republic of Kazakhstan
| | - Beibitgul Zhumabayeva
- Department of Molecular Biology and Genetics, Faculty of Biology and Biotechnology, al-Farabi Kazakh National University, Almaty, The Republic of Kazakhstan
| | - Tatyana Gebauer
- South Bohemian Research Center of Aquaculture and Biodiversity of Hydrocenoses, Institute of Aquaculture and Protection of Waters, Faculty of Fisheries and Protection of Waters, University of South Bohemia in Ceske Budejovice, České Budějovice, Czech Republic
| | - Ilya Kisselev
- Institute of General Genetics and Cytology, Almaty, The Republic of Kazakhstan
| | - Zaure Aitasheva
- Department of Molecular Biology and Genetics, Faculty of Biology and Biotechnology, al-Farabi Kazakh National University, Almaty, The Republic of Kazakhstan
| |
Collapse
|
32
|
Hedgehog Signaling Regulates Neurogenesis in the Larval and Adult Zebrafish Hypothalamus. eNeuro 2020; 7:ENEURO.0226-20.2020. [PMID: 33106384 PMCID: PMC7769882 DOI: 10.1523/eneuro.0226-20.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 09/21/2020] [Accepted: 09/28/2020] [Indexed: 12/15/2022] Open
Abstract
Neurogenesis is now known to play a role in adult hypothalamic function, yet the cell-cell mechanisms regulating this neurogenesis remain poorly understood. Here, we show that Hedgehog (Hh)/Gli signaling positively regulates hypothalamic neurogenesis in both larval and adult zebrafish and is necessary and sufficient for normal hypothalamic proliferation rates. Hh-responsive radial glia represent a relatively highly proliferative precursor population that gives rise to dopaminergic, serotonergic, and GABAergic neurons. In situ and transgenic reporter analyses revealed substantial heterogeneity in cell-cell signaling within the hypothalamic niche, with slow cycling Nestin-expressing cells residing among distinct and overlapping populations of Sonic Hh (Shh)-expressing, Hh-responsive, Notch-responsive, and Wnt-responsive radial glia. This work shows for the first time that Hh/Gli signaling is a key component of the complex cell-cell signaling environment that regulates hypothalamic neurogenesis throughout life.
Collapse
|
33
|
Ohgo S, Sakamoto T, Nakajima W, Matsunaga S, Wada N. Visualization of extracellular vesicles in the regenerating caudal fin blastema of zebrafish using in vivo electroporation. Biochem Biophys Res Commun 2020; 533:1371-1377. [PMID: 33077180 DOI: 10.1016/j.bbrc.2020.10.024] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 10/10/2020] [Indexed: 01/03/2023]
Abstract
Zebrafish have high regenerative ability in several organs including the fin. Although various mechanisms underlying fin regeneration have been revealed, some mechanisms remain to be elucidated. Recently, extracellular vesicles (EVs) have been the focus of research with regard to their role in cell-to-cell communication. It has been suggested that cells in regenerating tissues communicate using EVs. In this study, we examined the involvement of EVs in the caudal fin regeneration of zebrafish using an in vivo electroporation method. The process of regeneration appeared normal after in vivo electroporation, and the transferred plasmid showed mosaic expression in the blastema. We took advantage of this mosaic expression to observe the distribution of exosomal markers in the blastema. We transferred exosomal markers by in vivo electroporation and identified EVs in the regenerating caudal fin. The results suggest that blastemal cells communicate with other cells via EVs during caudal fin regeneration.
Collapse
Affiliation(s)
- Shiro Ohgo
- Department of Applied Biological Science, Tokyo University of Science, Noda, Japan.
| | - Takuya Sakamoto
- Department of Applied Biological Science, Tokyo University of Science, Noda, Japan
| | - Wataru Nakajima
- Department of Applied Biological Science, Tokyo University of Science, Noda, Japan
| | - Sachihiro Matsunaga
- Department of Applied Biological Science, Tokyo University of Science, Noda, Japan
| | - Naoyuki Wada
- Department of Applied Biological Science, Tokyo University of Science, Noda, Japan
| |
Collapse
|
34
|
Rangel-Huerta E, Guzman A, Maldonado E. The dynamics of epidermal stratification during post-larval development in zebrafish. Dev Dyn 2020; 250:175-190. [PMID: 32877571 DOI: 10.1002/dvdy.249] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 08/08/2020] [Accepted: 08/22/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The epidermis, as a defensive barrier, is a consistent trait throughout animal evolution. During post-larval development, the zebrafish epidermis thickens by stratification or addition of new cell layers. Epidermal basal stem cells, expressing the transcription factor p63, are known to be involved in this process. Zebrafish post-larval epidermal stratification is a tractable system to study how stem cells participate in organ growth. METHODS We used immunohistochemistry, in combination with EdU cell proliferation detection, to study zebrafish epidermal stratification. For this procedure, we selected a window of post-larval stages (5-8 mm of standard length or SL, which normalizes age by size). Simultaneously, we used markers for asymmetric cell division and the Notch signaling pathway. RESULTS We found that epidermal stratification is the consequence of several events, including changes in cell shape, active cell proliferation and asymmetrical cell divisions. We identified a subset of highly proliferative epidermal cells with reduced levels of p63, which differed from the basal stem cells with high levels of p63. Additionally, we described different mechanisms that participate in the stratification process, including the phosphorylation of p63, asymmetric cell division regulated by the Par3 and LGN proteins, and expression of Notch genes.
Collapse
Affiliation(s)
- Emma Rangel-Huerta
- EvoDevo Research Group, Unidad de Sistemas Arrecifales, Instituto de Ciencias del Mar y Limnología, Universidad Nacional Autónoma de México (UNAM), Puerto Morelos, Quintana Roo, Mexico.,Posgrado en Ciencias Biomédicas, Universidad Nacional Autónoma de México, UNAM, Puerto Morelos, Quintana Roo, Mexico
| | - Aida Guzman
- EvoDevo Research Group, Unidad de Sistemas Arrecifales, Instituto de Ciencias del Mar y Limnología, Universidad Nacional Autónoma de México (UNAM), Puerto Morelos, Quintana Roo, Mexico.,Estudio Técnico Especializado en Histopatología, Escuela Nacional Preparatoria, ENP, Universidad Nacional Autónoma de México, UNAM, Ciudad de México, Mexico
| | - Ernesto Maldonado
- EvoDevo Research Group, Unidad de Sistemas Arrecifales, Instituto de Ciencias del Mar y Limnología, Universidad Nacional Autónoma de México (UNAM), Puerto Morelos, Quintana Roo, Mexico
| |
Collapse
|
35
|
Nasoori A. Formation, structure, and function of extra-skeletal bones in mammals. Biol Rev Camb Philos Soc 2020; 95:986-1019. [PMID: 32338826 DOI: 10.1111/brv.12597] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 03/07/2020] [Accepted: 03/17/2020] [Indexed: 12/12/2022]
Abstract
This review describes the formation, structure, and function of bony compartments in antlers, horns, ossicones, osteoderm and the os penis/os clitoris (collectively referred to herein as AHOOO structures) in extant mammals. AHOOOs are extra-skeletal bones that originate from subcutaneous (dermal) tissues in a wide variety of mammals, and this review elaborates on the co-development of the bone and skin in these structures. During foetal stages, primordial cells for the bony compartments arise in subcutaneous tissues. The epithelial-mesenchymal transition is assumed to play a key role in the differentiation of bone, cartilage, skin and other tissues in AHOOO structures. AHOOO ossification takes place after skeletal bone formation, and may depend on sexual maturity. Skin keratinization occurs in tandem with ossification and may be under the control of androgens. Both endochondral and intramembranous ossification participate in bony compartment formation. There is variation in gradients of density in different AHOOO structures. These gradients, which vary according to function and species, primarily reduce mechanical stress. Anchorage of AHOOOs to their surrounding tissues fortifies these structures and is accomplished by bone-bone fusion and Sharpey fibres. The presence of the integument is essential for the protection and function of the bony compartments. Three major functions can be attributed to AHOOOs: mechanical, visual, and thermoregulatory. This review provides the first extensive comparative description of the skeletal and integumentary systems of AHOOOs in a variety of mammals.
Collapse
Affiliation(s)
- Alireza Nasoori
- School of Veterinary Medicine, Hokkaido University, Kita 18, Nishi 9, Kita-ku, Sapporo, Hokkaido, 060-0818, Japan
| |
Collapse
|
36
|
Lézot F, Corre I, Morice S, Rédini F, Verrecchia F. SHH Signaling Pathway Drives Pediatric Bone Sarcoma Progression. Cells 2020; 9:cells9030536. [PMID: 32110934 PMCID: PMC7140443 DOI: 10.3390/cells9030536] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 02/19/2020] [Accepted: 02/23/2020] [Indexed: 02/07/2023] Open
Abstract
Primary bone tumors can be divided into two classes, benign and malignant. Among the latter group, osteosarcoma and Ewing sarcoma are the most prevalent malignant primary bone tumors in children and adolescents. Despite intensive efforts to improve treatments, almost 40% of patients succumb to the disease. Specifically, the clinical outcome for metastatic osteosarcoma or Ewing sarcoma remains poor; less than 30% of patients who present metastases will survive 5 years after initial diagnosis. One common and specific point of these bone tumors is their ability to deregulate bone homeostasis and remodeling and divert them to their benefit. Over the past years, considerable interest in the Sonic Hedgehog (SHH) pathway has taken place within the cancer research community. The activation of this SHH cascade can be done through different ways and, schematically, two pathways can be described, the canonical and the non-canonical. This review discusses the current knowledge about the involvement of the SHH signaling pathway in skeletal development, pediatric bone sarcoma progression and the related therapeutic options that may be possible for these tumors.
Collapse
|
37
|
Wu Y, Li W, Yuan M, Liu X. The synthetic pyrethroid deltamethrin impairs zebrafish (Danio rerio) swim bladder development. THE SCIENCE OF THE TOTAL ENVIRONMENT 2020; 701:134870. [PMID: 31726413 DOI: 10.1016/j.scitotenv.2019.134870] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Revised: 09/26/2019] [Accepted: 10/05/2019] [Indexed: 06/10/2023]
Abstract
Deltamethrin (DM) is a widely used insecticide and reveals neural, cardiovascular and reproductive toxicity to various aquatic organisms. It has been known that DM negatively affects motion of zebrafish (Danio rerio). However, little is known in relation to the impacts of DM on development of swim bladder, which is a key organ for motion. In the present study, zebrafish embryos were exposed to 20 and 40 µg/L DM. The changes of swim bladder morphology were observed and transcription levels of key genes were compared between DM treatments and the control. The results showed that DM treatments significantly blocked the formation of progenitor and tissue layers in swim bladder of zebrafish embryos, leading to failed inflation of swim bladder. Compared with the control, the key genes (pbx1, foxA3, mnx1, has2, anxa5b, hprt1l and elovl1a) responsible for swim bladder development also showed decreased levels in response to DM treatments, suggesting that DM might specifically affect swim bladder development. Moreover, transcription levels of genes in the Wnt (wnt5b, tcf3a, wnt1, wnt9b, fzd1, fzd3 and fzd5) and Hedgehog (ihhb, ptc1 and ptc2) signaling pathways all decreased significantly in response to DM treatments, compared with the control. Considering the importance of Wnt and Hedgehog pathways in development of swim bladder, these results suggested that DM might affect swim bladder development through inhibiting the Wnt and Hedgehog pathways. Overall, the present study reported that swim bladder might be a potential target organ of DM toxicity in zebrafish, which contributed more information to the evaluation of DM's environmental risks.
Collapse
Affiliation(s)
- Yaqin Wu
- Key Laboratory of Xiamen Marine and Gene Drugs, School of Biomedical Sciences, Huaqiao University, Xiamen 361021, China
| | - Wenhua Li
- Key Laboratory of Xiamen Marine and Gene Drugs, School of Biomedical Sciences, Huaqiao University, Xiamen 361021, China.
| | - Mingrui Yuan
- Key Laboratory of Xiamen Marine and Gene Drugs, School of Biomedical Sciences, Huaqiao University, Xiamen 361021, China
| | - Xuan Liu
- Aquatic EcoHealth Group, Key Laboratory of Urban Environment and Health, Fujian Provincial Key Laboratory of Watershed Ecology, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, China
| |
Collapse
|
38
|
Genetic Reprogramming of Positional Memory in a Regenerating Appendage. Curr Biol 2019; 29:4193-4207.e4. [PMID: 31786062 DOI: 10.1016/j.cub.2019.10.038] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 09/01/2019] [Accepted: 10/21/2019] [Indexed: 12/31/2022]
Abstract
Certain vertebrates such as salamanders and zebrafish are able to regenerate complex tissues (e.g., limbs and fins) with remarkable fidelity. However, how positional information of the missing structure is recalled by appendage stump cells has puzzled researchers for centuries. Here, we report that sizing information for adult zebrafish tailfins is encoded within proliferating blastema cells during a critical period of regeneration. Using a chemical mutagenesis screen, we identified a temperature-sensitive allele of the gene encoding DNA polymerase alpha subunit 2 (pola2) that disrupts fin regeneration in zebrafish. Temperature shift assays revealed a 48-h window of regeneration, during which positional identities could be disrupted in pola2 mutants, leading to regeneration of miniaturized appendages. These fins retained memory of the new size in subsequent rounds of amputation and regeneration. Similar effects were observed upon transient genetic or pharmacological disruption of progenitor cell proliferation after plucking of zebrafish scales or head or tail amputation in amphioxus and annelids. Our results provide evidence that positional information in regenerating tissues is not hardwired but malleable, based on regulatory mechanisms that appear to be evolutionarily conserved across distantly related phyla.
Collapse
|
39
|
Recent advancements in understanding fin regeneration in zebrafish. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2019; 9:e367. [DOI: 10.1002/wdev.367] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 10/07/2019] [Accepted: 10/23/2019] [Indexed: 11/07/2022]
|
40
|
Li L, Zhang J, Akimenko MA. Inhibition of mmp13a during zebrafish fin regeneration disrupts fin growth, osteoblasts differentiation, and Laminin organization. Dev Dyn 2019; 249:187-198. [PMID: 31487071 DOI: 10.1002/dvdy.112] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 08/29/2019] [Accepted: 08/31/2019] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Matrix metalloproteinases 13 (MMP13) is a potent endopeptidase that regulate cell growth, migration, and extracellular matrix remodeling. However, its role in fin regeneration remains unclear. RESULTS mmp13a expression is strongly upregulated during blastema formation and persists in the distal blastema. mmp13a knockdown via morpholino electroporation impairs regenerative outgrowth by decreasing cell proliferation, which correlates with a downregulation of fgf10a and sall4 expression in the blastema. Laminin distribution in the basement membrane is also affected in mmp13a MO-injected rays. Another impact of mmp13a knockdown is observed in the skeletal elements of the fin rays. Expression of two main components of actinotrichia, Collagen II and Actinodin 1 is highly reduced in mmp13a MO-injected rays leading to highly disorganized actinotrichia pattern. Inhibition of mmp13a strongly affects bone formation as shown by a reduction of Zns5 and sp7 expression and of bone matrix mineralization in rays. These defects are accompanied by a significant increase in apoptosis in mmp13a MO-injected fin regenerates. CONCLUSION Defects of expression of this multifunctional proteinase drastically affects osteoblast differentiation, bone and actinotrichia formation as well as Laminin distribution in the basement membrane of the fin regenerate, suggesting the important role of Mmp13 during the regenerative process.
Collapse
Affiliation(s)
- Li Li
- College of Life Science, Henan Normal University, Xinxiang, Henan, China.,CAREG, University of Ottawa, Ottawa, Ontario, Canada.,Department of Biology, University of Ottawa, Ottawa, Ontario, Canada
| | - Jing Zhang
- CAREG, University of Ottawa, Ottawa, Ontario, Canada.,Department of Biology, University of Ottawa, Ottawa, Ontario, Canada
| | - Marie-Andrée Akimenko
- CAREG, University of Ottawa, Ottawa, Ontario, Canada.,Department of Biology, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
41
|
Cortes JE, Gutzmer R, Kieran MW, Solomon JA. Hedgehog signaling inhibitors in solid and hematological cancers. Cancer Treat Rev 2019; 76:41-50. [PMID: 31125907 DOI: 10.1016/j.ctrv.2019.04.005] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 04/25/2019] [Accepted: 04/26/2019] [Indexed: 12/21/2022]
Abstract
BACKGROUND The hedgehog signaling pathway is normally tightly regulated. Mutations in hedgehog pathway components may lead to abnormal activation. Aberrantly activated hedgehog signaling plays a major role in the development of solid and hematological cancer. In recent years, inhibitors have been developed that attenuate hedgehog signaling; 2 have been approved for use in basal cell carcinoma (BCC), while others are under development or in clinical trials. The aim of this review is to provide an overview of known hedgehog inhibitors (HHIs) and their potential for the treatment of hematological cancers and solid tumors beyond BCC. DESIGN Published literature was searched to identify articles relating to HHIs in noncutaneous cancer. Both preclinical and clinical research articles were included. In addition, relevant clinical trial results were identified from www.clinicaltrials.gov. Information on the pharmacology of HHIs is also included. RESULTS HHIs show activity in a variety of solid and hematological cancers. In preclinical studies, HHIs demonstrated efficacy in pancreatic cancer, rhabdomyosarcoma, breast cancer, and acute myeloid leukemia (AML). In clinical studies, HHIs showed activity in medulloblastoma, as well as prostate, pancreatic, and hematological cancers. Current clinical trials testing the efficacy of HHIs are underway for prostate, pancreatic, and breast cancers, as well as multiple myeloma and AML. CONCLUSIONS As clinical trial results become available, it will be possible to discern which additional tumor types are suited to HHI mono- or combination therapy with other anticancer agents. The latter strategy may be useful for delaying or overcoming drug resistance.
Collapse
Affiliation(s)
- Jorge E Cortes
- Department of Leukemia, MD Anderson Cancer Center, 1515 Holcombe Blvd. #428, Houston, TX 77030, USA.
| | - Ralf Gutzmer
- Skin Cancer Center Hannover, Department of Dermatology, Hannover Medical School, Carl-Neuberg Str 1, D-30625 Hannover, Germany.
| | - Mark W Kieran
- Dana-Farber Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA.
| | - James A Solomon
- Ameriderm Research, 725 W Granada Blvd Ste 44, Ormond Beach, FL 32174, USA; University of Central Florida, Orlando, FL, USA.
| |
Collapse
|
42
|
Phan HE, Northorp M, Lalonde RL, Ngo D, Akimenko MA. Differential actinodin1 regulation in embryonic development and adult fin regeneration in Danio rerio. PLoS One 2019; 14:e0216370. [PMID: 31048899 PMCID: PMC6497306 DOI: 10.1371/journal.pone.0216370] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 04/18/2019] [Indexed: 12/22/2022] Open
Abstract
Actinotrichia are the first exoskeletal elements formed during zebrafish fin development. These rigid fibrils serve as skeletal support for the fin fold and as substrates for mesenchymal cell migration. In the adult intact fins, actinotrichia are restricted to the distal domain of the fin. Following fin amputation, actinotrichia also reform during regeneration. The actinodin gene family codes for structural proteins of actinotrichia. We have previously identified cis-acting regulatory elements in a 2kb genomic region upstream of the first exon of actinodin1, termed 2P, required for tissue-specific expression in the fin fold ectoderm and mesenchyme during embryonic development. Indeed, 2P contains an ectodermal enhancer in a 150bp region named epi. Deletion of epi from 2P results in loss of ectodermal-specific activity. In the present study, we sought to further characterize the activity of these regulatory sequences throughout fin development and during adult fin regeneration. Using a reporter transgenic approach, we show that a site within the epi region, termed epi3, contains an early mesenchymal-specific repressor. We also show that the larval fin fold ectodermal enhancer within epi3 remains functional in the basal epithelial layer during fin regeneration. We show that the first non-coding exon and first intron of actinodin1 contains a transcriptional enhancer and an alternative promoter that are necessary for the persistence of reporter expression reminiscent of actinodin1 expression during adulthood. Altogether, we have identified cis-acting regulatory elements that are required for tissue-specific expression as well as full recapitulation of actinodin1 expression during adulthood. Furthermore, the characterization of these elements provides us with useful molecular tools for the enhancement of transgene expression in adulthood.
Collapse
Affiliation(s)
- Hue-Eileen Phan
- Department of Biology, University of Ottawa, Ottawa, Ontario, Canada
| | - Marissa Northorp
- Department of Biology, University of Ottawa, Ottawa, Ontario, Canada
| | - Robert L. Lalonde
- Department of Biology, University of Ottawa, Ottawa, Ontario, Canada
| | - Dung Ngo
- Department of Biology, University of Ottawa, Ottawa, Ontario, Canada
| | | |
Collapse
|
43
|
Hu Z, Chen B, Zhao Q. Hedgehog signaling regulates osteoblast differentiation in zebrafish larvae through modulation of autophagy. Biol Open 2019; 8:bio.040840. [PMID: 30992325 PMCID: PMC6550075 DOI: 10.1242/bio.040840] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Impaired osteoblast differentiation may result in bone metabolic diseases such as osteoporosis. It was reported recently that hedgehog (Hh) signaling and autophagy are two important regulators of bone differentiation. In order to further dissect their relationship in bone development, we used a zebrafish larvae model to investigate how disruption of one of these signals affects the function of the other and impacts osteoblast differentiation. Our results showed that activation of Hh signaling negatively regulated autophagy. However, suppression of autophagy by knocking down atg5 expression did not alter Hh signaling, but dramatically upregulated the expression of osteoblast-related genes and increased bone mineralization, especially in the den region. On the contrary, inhibition of the Hh signaling pathway by cyclopamine treatment suppressed the expression of osteoblast-related genes and decreased bone mineralization. In agreement with these findings, blocking Hh signaling through knockdown SHH and Gli2 genes led to defective osteoblast differentiation, while promoting Hh signaling by knockdown Ptch1 was beneficial to osteoblast differentiation. Our results thus support that activation of the Hh signaling pathway negatively regulates autophagy and consequentially promotes osteoblast differentiation. On the contrary, induction of autophagy inhibits osteoblast differentiation. Our work reveals the mechanism underlying Hh signaling pathway regulation of bone development. Summary: Our report of an essential regulation role of hedgehog signaling and autophagy on osteoblast differentiation may contribute to research on bone development biology, hedgehog signaling and the autophagy pathway.
Collapse
Affiliation(s)
- Zhanying Hu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Bo Chen
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Qiong Zhao
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
44
|
Deng W, Shao F, He Q, Wang Q, Shi W, Yu Q, Cao X, Feng C, Bi S, Chen J, Ma P, Li Y, Gong A, Tong S, Yu J, Spector M, Xu X, Zhang Z. EMSCs Build an All-in-One Niche via Cell-Cell Lipid Raft Assembly for Promoted Neuronal but Suppressed Astroglial Differentiation of Neural Stem Cells. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1806861. [PMID: 30633831 DOI: 10.1002/adma.201806861] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 12/29/2018] [Indexed: 05/11/2023]
Abstract
The therapeutic efficiency of allogenic/intrinsic neural stem cells (NSCs) after spinal cord injury is severely compromised because the hostile niche at the lesion site incurs massive astroglial but not neuronal differentiation of NSCs. Although many attempts are made to reconstruct a permissive niche for nerve regeneration, solely using a living cell material to build an all-in-one, multifunctional, permissive niche for promoting neuronal while inhibiting astroglial differentiation of NSCs is not reported. Here, ectomesenchymal stem cells (EMSCs) are reported to serve as a living, smart material that creates a permissive, all-in-one niche which provides neurotrophic factors, extracellular matrix molecules, cell-cell contact, and favorable substrate stiffness for directing NSC differentiation. Interestingly, in this all-in-one niche, a corresponding all-in-one signal-sensing platform is assembled through recruiting various niche signaling molecules into lipid rafts for promoting neuronal differentiation of NSCs, and meanwhile, inhibiting astrocyte overproliferation through the connexin43/YAP/14-3-3θ pathway. In vivo studies confirm that EMSCs can promote intrinsic NSC neuronal differentiation and domesticating astrocyte behaviors for nerve regeneration. Collectively, this study represents an all-in-one niche created by a single-cell material-EMSCs for directing NSC differentiation.
Collapse
Affiliation(s)
- Wenwen Deng
- School of Pharmacy, Jiangsu University, Laboratory of Drug Delivery and Tissue Regeneration and Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, 212001, P. R. China
| | - Fengxia Shao
- School of Pharmacy, Jiangsu University, Laboratory of Drug Delivery and Tissue Regeneration and Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, 212001, P. R. China
| | - Qinghua He
- School of Pharmacy, Jiangsu University, Laboratory of Drug Delivery and Tissue Regeneration and Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, 212001, P. R. China
| | - Qiang Wang
- School of Pharmacy, Jiangsu University, Laboratory of Drug Delivery and Tissue Regeneration and Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, 212001, P. R. China
| | - Wentao Shi
- School of Medical Science and Laboratory Medicine, Jiangsu University, Zhenjiang, 212001, P. R. China
| | - Qingtong Yu
- School of Pharmacy, Jiangsu University, Laboratory of Drug Delivery and Tissue Regeneration and Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, 212001, P. R. China
| | - Xia Cao
- School of Pharmacy, Jiangsu University, Laboratory of Drug Delivery and Tissue Regeneration and Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, 212001, P. R. China
| | - Chunlai Feng
- School of Pharmacy, Jiangsu University, Laboratory of Drug Delivery and Tissue Regeneration and Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, 212001, P. R. China
| | - Shiqi Bi
- School of Medical Science and Laboratory Medicine, Jiangsu University, Zhenjiang, 212001, P. R. China
| | - Jiaxin Chen
- School of Pharmacy, Jiangsu University, Laboratory of Drug Delivery and Tissue Regeneration and Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, 212001, P. R. China
| | - Ping Ma
- School of Pharmacy, Jiangsu University, Laboratory of Drug Delivery and Tissue Regeneration and Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, 212001, P. R. China
| | - Yang Li
- School of Pharmacy, Jiangsu University, Laboratory of Drug Delivery and Tissue Regeneration and Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, 212001, P. R. China
| | - Aihua Gong
- School of Medical Science and Laboratory Medicine, Jiangsu University, Zhenjiang, 212001, P. R. China
| | - Shanshan Tong
- School of Pharmacy, Jiangsu University, Laboratory of Drug Delivery and Tissue Regeneration and Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, 212001, P. R. China
| | - Jiangnan Yu
- School of Pharmacy, Jiangsu University, Laboratory of Drug Delivery and Tissue Regeneration and Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, 212001, P. R. China
| | - Myron Spector
- Department of Orthopedic Surgery, Harvard Medical School, Brigham and Women's Hospital, 75 Francis St, Boston, MA, 02115, USA
| | - Ximing Xu
- School of Pharmacy, Jiangsu University, Laboratory of Drug Delivery and Tissue Regeneration and Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, 212001, P. R. China
| | - Zhijian Zhang
- School of Medical Science and Laboratory Medicine, Jiangsu University, Zhenjiang, 212001, P. R. China
| |
Collapse
|
45
|
Bergen DJM, Kague E, Hammond CL. Zebrafish as an Emerging Model for Osteoporosis: A Primary Testing Platform for Screening New Osteo-Active Compounds. Front Endocrinol (Lausanne) 2019; 10:6. [PMID: 30761080 PMCID: PMC6361756 DOI: 10.3389/fendo.2019.00006] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 01/09/2019] [Indexed: 12/16/2022] Open
Abstract
Osteoporosis is metabolic bone disease caused by an altered balance between bone anabolism and catabolism. This dysregulated balance is responsible for fragile bones that fracture easily after minor falls. With an aging population, the incidence is rising and as yet pharmaceutical options to restore this imbalance is limited, especially stimulating osteoblast bone-building activity. Excitingly, output from large genetic studies on people with high bone mass (HBM) cases and genome wide association studies (GWAS) on the population, yielded new insights into pathways containing osteo-anabolic players that have potential for drug target development. However, a bottleneck in development of new treatments targeting these putative osteo-anabolic genes is the lack of animal models for rapid and affordable testing to generate functional data and that simultaneously can be used as a compound testing platform. Zebrafish, a small teleost fish, are increasingly used in functional genomics and drug screening assays which resulted in new treatments in the clinic for other diseases. In this review we outline the zebrafish as a powerful model for osteoporosis research to validate potential therapeutic candidates, describe the tools and assays that can be used to study bone homeostasis, and affordable (semi-)high-throughput compound testing.
Collapse
Affiliation(s)
- Dylan J. M. Bergen
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building, University of Bristol, Bristol, United Kingdom
- Musculoskeletal Research Unit, Translational Health Sciences, Bristol Medical School, Southmead Hospital, University of Bristol, Bristol, United Kingdom
| | - Erika Kague
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building, University of Bristol, Bristol, United Kingdom
| | - Chrissy L. Hammond
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
46
|
Gordon HB, Lusk S, Carney KR, Wirick EO, Murray BF, Kwan KM. Hedgehog signaling regulates cell motility and optic fissure and stalk formation during vertebrate eye morphogenesis. Development 2018; 145:dev.165068. [PMID: 30333214 PMCID: PMC6262791 DOI: 10.1242/dev.165068] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 10/08/2018] [Indexed: 12/18/2022]
Abstract
Establishment of precise three-dimensional tissue structure is vital for organ function. In the visual system, optic fissure and stalk morphogenesis is a crucial yet poorly understood process, disruptions of which can lead to coloboma, a birth defect causing visual impairment. Here, we use four-dimensional imaging, cell tracking, and molecular genetics in zebrafish to define the cell movements underlying normal optic fissure and stalk formation. We determine how these events are disrupted in a coloboma model in which the Hedgehog (Hh) receptor ptch2 is lost, resulting in overactive Hh signaling. In the ptch2 mutant, cells exhibit defective motile behaviors and morphology. Cells that should contribute to the fissure do not arrive at their correct position, and instead contribute to an ectopically large optic stalk. Our results suggest that overactive Hh signaling, through overexpression of downstream transcriptional targets, impairs cell motility underlying optic fissure and stalk formation, via non-cell-autonomous and cell-autonomous mechanisms. More broadly, our cell motility and morphology analyses provide a new framework for studying other coloboma-causing mutations that disrupt optic fissure or stalk formation. Summary: Multidimensional imaging of ptch2 mutant zebrafish uncovers a role for the Hh signaling pathway in regulating the cell and tissue dynamics underlying early eye morphogenesis.
Collapse
Affiliation(s)
- Hannah B Gordon
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Sarah Lusk
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Keith R Carney
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Emily O Wirick
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | | | - Kristen M Kwan
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
47
|
Aman AJ, Fulbright AN, Parichy DM. Wnt/β-catenin regulates an ancient signaling network during zebrafish scale development. eLife 2018; 7:37001. [PMID: 30014845 PMCID: PMC6072442 DOI: 10.7554/elife.37001] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 06/21/2018] [Indexed: 12/24/2022] Open
Abstract
Understanding how patterning influences cell behaviors to generate three dimensional morphologies is a central goal of developmental biology. Additionally, comparing these regulatory mechanisms among morphologically diverse tissues allows for rigorous testing of evolutionary hypotheses. Zebrafish skin is endowed with a coat of precisely patterned bony scales. We use in-toto live imaging during scale development and manipulations of cell signaling activity to elucidate core features of scale patterning and morphogenesis. These analyses show that scale development requires the concerted activity of Wnt/β-catenin, Ectodysplasin (Eda) and Fibroblast growth factor (Fgf) signaling. This regulatory module coordinates Hedgehog (HH) dependent collective cell migration during epidermal invagination, a cell behavior not previously implicated in skin appendage morphogenesis. Our analyses demonstrate the utility of zebrafish scale development as a tractable system in which to elucidate mechanisms of developmental patterning and morphogenesis, and suggest a single, ancient origin of skin appendage patterning mechanisms in vertebrates.
Collapse
Affiliation(s)
- Andrew J Aman
- Department of Biology and Department of Cell Biology, University of Virginia, Charlottesville, United States
| | - Alexis N Fulbright
- Department of Biology and Department of Cell Biology, University of Virginia, Charlottesville, United States
| | - David M Parichy
- Department of Biology and Department of Cell Biology, University of Virginia, Charlottesville, United States
| |
Collapse
|
48
|
McMillan SC, Zhang J, Phan HE, Jeradi S, Probst L, Hammerschmidt M, Akimenko MA. A regulatory pathway involving retinoic acid and calcineurin demarcates and maintains joint cells and osteoblasts in regenerating fin. Development 2018; 145:dev.161158. [PMID: 29752384 DOI: 10.1242/dev.161158] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 05/01/2018] [Indexed: 12/21/2022]
Abstract
During zebrafish fin regeneration, blastema cells lining the epidermis differentiate into osteoblasts and joint cells to reconstruct the segmented bony rays. We show that osteoblasts and joint cells originate from a common cell lineage, but are committed to different cell fates. Pre-osteoblasts expressing runx2a/b commit to the osteoblast lineage upon expressing sp7, whereas the strong upregulation of hoxa13a correlates with a commitment to a joint cell type. In the distal regenerate, hoxa13a, evx1 and pthlha are sequentially upregulated at regular intervals to define the newly identified presumptive joint cells. Presumptive joint cells mature into joint-forming cells, a distinct cell cluster that maintains the expression of these factors. Analysis of evx1 null mutants reveals that evx1 is acting upstream of pthlha and downstream of or in parallel with hoxa13a Calcineurin activity, potentially through the inhibition of retinoic acid signaling, regulates evx1, pthlha and hoxa13a expression during joint formation. Furthermore, retinoic acid treatment induces osteoblast differentiation in mature joint cells, leading to ectopic bone deposition in joint regions. Overall, our data reveal a novel regulatory pathway essential for joint formation in the regenerating fin.
Collapse
Affiliation(s)
- Stephanie C McMillan
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada K1N 6N5.,CAREG, 30 Marie Curie, University of Ottawa, Ottawa, ON, Canada K1N 6N5
| | - Jing Zhang
- CAREG, 30 Marie Curie, University of Ottawa, Ottawa, ON, Canada K1N 6N5.,Department of Biology, 30 Marie Curie, University of Ottawa, Ottawa, ON, Canada K1N 6N5
| | - Hue-Eileen Phan
- CAREG, 30 Marie Curie, University of Ottawa, Ottawa, ON, Canada K1N 6N5.,Department of Biology, 30 Marie Curie, University of Ottawa, Ottawa, ON, Canada K1N 6N5
| | - Shirine Jeradi
- Institute for Developmental Biology, Cologne University, Cologne 50674, Germany.,Institut Polytechnique Privé, Université Libre de Tunis, Tunis 1003, Tunisia
| | - Leona Probst
- CAREG, 30 Marie Curie, University of Ottawa, Ottawa, ON, Canada K1N 6N5.,Department of Biology, 30 Marie Curie, University of Ottawa, Ottawa, ON, Canada K1N 6N5
| | | | - Marie-Andrée Akimenko
- CAREG, 30 Marie Curie, University of Ottawa, Ottawa, ON, Canada K1N 6N5 .,Department of Biology, 30 Marie Curie, University of Ottawa, Ottawa, ON, Canada K1N 6N5
| |
Collapse
|
49
|
Epidermal regulation of bone morphogenesis through the development and regeneration of osteoblasts in the zebrafish scale. Dev Biol 2018. [DOI: 10.1016/j.ydbio.2018.03.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
50
|
Murciano C, Cazorla-Vázquez S, Gutiérrez J, Hijano JA, Ruiz-Sánchez J, Mesa-Almagro L, Martín-Reyes F, Fernández TD, Marí-Beffa M. Widening control of fin inter-rays in zebrafish and inferences about actinopterygian fins. J Anat 2018; 232:783-805. [PMID: 29441573 DOI: 10.1111/joa.12785] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/05/2018] [Indexed: 01/03/2023] Open
Abstract
The amputation of a teleost fin rapidly triggers an intricate maze of hierarchically regulated signalling processes which ultimately reconstruct the diverse tissues of the appendage. Whereas the generation of the fin pattern along the proximodistal axis brings with it several well-known developmental regulators, the mechanisms by which the fin widens along its dorsoventral axis remain poorly understood. Utilizing the zebrafish as an experimental model of fin regeneration and studying more than 1000 actinopterygian species, we hypothesized a connection between specific inter-ray regulatory mechanisms and the morphological variability of inter-ray membranes found in nature. To tackle these issues, both cellular and molecular approaches have been adopted and our results suggest the existence of two distinguishable inter-ray areas in the zebrafish caudal fin, a marginal and a central region. The present work associates the activity of the cell membrane potassium channel kcnk5b, the fibroblast growth factor receptor 1 and the sonic hedgehog pathway to the control of several cell functions involved in inter-ray wound healing or dorsoventral regeneration of the zebrafish caudal fin. This ray-dependent regulation controls cell migration, cell-type patterning and gene expression. The possibility that modifications of these mechanisms are responsible for phenotypic variations found in euteleostean species, is discussed.
Collapse
Affiliation(s)
- Carmen Murciano
- Department of Cell Biology, Genetics and Physiology, Biomedical Research Institute of Málaga (IBIMA), Faculty of Science, University of Málaga, Málaga, Spain
| | - Salvador Cazorla-Vázquez
- Department of Cell Biology, Genetics and Physiology, Biomedical Research Institute of Málaga (IBIMA), Faculty of Science, University of Málaga, Málaga, Spain
| | - Javier Gutiérrez
- Department of Cell Biology, Genetics and Physiology, Biomedical Research Institute of Málaga (IBIMA), Faculty of Science, University of Málaga, Málaga, Spain
| | - Juan Antonio Hijano
- Department of Cell Biology, Genetics and Physiology, Biomedical Research Institute of Málaga (IBIMA), Faculty of Science, University of Málaga, Málaga, Spain
| | - Josefa Ruiz-Sánchez
- Department of Cell Biology, Genetics and Physiology, Biomedical Research Institute of Málaga (IBIMA), Faculty of Science, University of Málaga, Málaga, Spain
| | - Laura Mesa-Almagro
- Department of Cell Biology, Genetics and Physiology, Biomedical Research Institute of Málaga (IBIMA), Faculty of Science, University of Málaga, Málaga, Spain
| | - Flores Martín-Reyes
- Department of Cell Biology, Genetics and Physiology, Biomedical Research Institute of Málaga (IBIMA), Faculty of Science, University of Málaga, Málaga, Spain
| | | | - Manuel Marí-Beffa
- Department of Cell Biology, Genetics and Physiology, Biomedical Research Institute of Málaga (IBIMA), Faculty of Science, University of Málaga, Málaga, Spain.,Networking Research Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Málaga, Spain
| |
Collapse
|