1
|
Wang H, Ma C, Liu C, Sun L, Wang Y, Xue J, Zhao B, Dong W. The c-Fos/AP-1 inhibitor inhibits sulfur mustard-induced chondrogenesis impairment in zebrafish larvae. CHEMOSPHERE 2024; 359:142299. [PMID: 38761826 DOI: 10.1016/j.chemosphere.2024.142299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 05/07/2024] [Accepted: 05/08/2024] [Indexed: 05/20/2024]
Abstract
Sulfur mustard (SM, dichlorodiethyl sulfide) is a potent erosive chemical poison that can cause pulmonary lung, skin and eye disease complications in humans. Currently, there is no designated remedy for SM, and its operation's toxicological process remains unidentified. This work employed zebrafish as a model organism to investigate the toxic manifestations and mechanisms of exposure to SM, aiming to offer novel insights for preventing and treating this condition. The results showed that SM caused a decrease in the survival rate of the zebrafish larvae (LC50 = 2.47 mg/L), a reduction in the hatching rate, an increase in the pericardial area, and small head syndrome. However, T-5224 (a selective inhibitor of c-Fos/activator protein) attenuated the reduction in mortality (LC50 = 2.79 mg/L), the reduction in hatching rate, and the worsening of morphological changes. We discovered that SM causes cartilage developmental disorders in zebrafish larvae. The reverse transcription-quantitative polymerase chain reaction found that SM increased the expression of inflammation-related genes (IL-1β, IL-6, and TNF-α) and significantly increased cartilage development-related gene expression (fosab, mmp9, and atf3). However, the expression of sox9a, sox9b, and Col2a1a was reduced. The protein level detection also found an increase in c-fos protein expression and a significant decrease in COL2A1 expression. However, T-5224,also and mitigated the changes in gene expression, and protein levels caused by SM exposure. The results of this study indicate that SM-induced cartilage development disorders are closely related to the c-Fos/AP-1 pathway in zebrafish.
Collapse
Affiliation(s)
- Huan Wang
- Inner Mongolia Key Laboratory of Toxicant Monitoring and Toxicology, College of Animal Science and Technology, Inner Mongolia Minzu University, Tongliao, 028000, China; State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, Beijing, 100850, China
| | - Chenglong Ma
- Inner Mongolia Key Laboratory of Toxicant Monitoring and Toxicology, College of Animal Science and Technology, Inner Mongolia Minzu University, Tongliao, 028000, China; State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, Beijing, 100850, China
| | - Chunyu Liu
- Inner Mongolia Key Laboratory of Toxicant Monitoring and Toxicology, College of Animal Science and Technology, Inner Mongolia Minzu University, Tongliao, 028000, China
| | - Lan Sun
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, Beijing, 100850, China
| | - Yongan Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, Beijing, 100850, China
| | - Jiangdong Xue
- Inner Mongolia Key Laboratory of Toxicant Monitoring and Toxicology, College of Animal Science and Technology, Inner Mongolia Minzu University, Tongliao, 028000, China.
| | - Baoquan Zhao
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, Beijing, 100850, China.
| | - Wu Dong
- Inner Mongolia Key Laboratory of Toxicant Monitoring and Toxicology, College of Animal Science and Technology, Inner Mongolia Minzu University, Tongliao, 028000, China.
| |
Collapse
|
2
|
Popsuj S, Di Gregorio A, Swalla BJ, Stolfi A. Loss of collagen gene expression in the notochord of the tailless tunicate Molgula occulta. Integr Comp Biol 2023; 63:990-998. [PMID: 37403333 PMCID: PMC10714901 DOI: 10.1093/icb/icad071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 05/22/2023] [Accepted: 06/11/2023] [Indexed: 07/06/2023] Open
Abstract
In tunicates, several species in the Molgulidae family have convergently lost the tailed, swimming larval body plan, including the morphogenesis of the notochord, a major chordate-defining trait. Through the comparison of tailless M. occulta and a close relative, the tailed species M. oculata, we show that notochord-specific expression of the Collagen Type I/II Alpha (Col1/2a) gene appears to have been lost specifically in the tailless species. Using CRISPR/Cas9-mediated mutagenesis in the tailed laboratory model tunicate Ciona robusta, we demonstrate that Col1/2a plays a crucial role in the convergent extension of notochord cells during tail elongation. Our results suggest that the expression of Col1/2a in the notochord, although necessary for its morphogenesis in tailed species, is dispensable for tailless species. This loss is likely a result of the accumulation of cis-regulatory mutations in the absence of purifying selective pressure. More importantly, the gene itself is not lost, likely due to its roles in other developmental processes, including during the adult stage. Our study further confirms the Molgulidae as an interesting family in which to study the evolutionary loss of tissue-specific expression of indispensable genes.
Collapse
Affiliation(s)
- Sydney Popsuj
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Anna Di Gregorio
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA
| | - Billie J Swalla
- Department of Biology, University of Washington, Seattle, WA 98195, USA
| | - Alberto Stolfi
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA 30332, USA
| |
Collapse
|
3
|
Truong BT, Shull LC, Lencer E, Bend EG, Field M, Blue EE, Bamshad MJ, Skinner C, Everman D, Schwartz CE, Flanagan-Steet H, Artinger KB. PRDM1 DNA-binding zinc finger domain is required for normal limb development and is disrupted in split hand/foot malformation. Dis Model Mech 2023; 16:dmm049977. [PMID: 37083955 PMCID: PMC10151829 DOI: 10.1242/dmm.049977] [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: 11/10/2022] [Accepted: 03/09/2023] [Indexed: 04/22/2023] Open
Abstract
Split hand/foot malformation (SHFM) is a rare limb abnormality with clefting of the fingers and/or toes. For many individuals, the genetic etiology is unknown. Through whole-exome and targeted sequencing, we detected three novel variants in a gene encoding a transcription factor, PRDM1, that arose de novo in families with SHFM or segregated with the phenotype. PRDM1 is required for limb development; however, its role is not well understood and it is unclear how the PRDM1 variants affect protein function. Using transient and stable overexpression rescue experiments in zebrafish, we show that the variants disrupt the proline/serine-rich and DNA-binding zinc finger domains, resulting in a dominant-negative effect. Through gene expression assays, RNA sequencing, and CUT&RUN in isolated pectoral fin cells, we demonstrate that Prdm1a directly binds to and regulates genes required for fin induction, outgrowth and anterior/posterior patterning, such as fgfr1a, dlx5a, dlx6a and smo. Taken together, these results improve our understanding of the role of PRDM1 in the limb gene regulatory network and identified novel PRDM1 variants that link to SHFM in humans.
Collapse
Affiliation(s)
- Brittany T. Truong
- Human Medical Genetics & Genomics Graduate Program, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Craniofacial Biology, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Lomeli C. Shull
- Department of Craniofacial Biology, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Ezra Lencer
- Biology Department, Lafayette College, Easton, PA 18042, USA
| | - Eric G. Bend
- Greenwood Genetics Center, Greenwood, SC 29646, USA
| | - Michael Field
- Genetics of Learning Disability Service, Hunter Genetics, Waratah, NSW 2298, AUS
| | - Elizabeth E. Blue
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, WA 98195, USA
- Brotman-Baty Institute for Precision Medicine, Seattle, WA 98195, USA
| | - Michael J. Bamshad
- Brotman-Baty Institute for Precision Medicine, Seattle, WA 98195, USA
- Division of Genetic Medicine, Department of Pediatrics, University of Washington, Seattle, WA 98195, USA
| | | | | | | | | | - Kristin B. Artinger
- Department of Craniofacial Biology, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
4
|
Ritter DJ, Choudhary D, Unlu G, Knapik EW. Rgp1 contributes to craniofacial cartilage development and Rab8a-mediated collagen II secretion. Front Endocrinol (Lausanne) 2023; 14:1120420. [PMID: 36843607 PMCID: PMC9947155 DOI: 10.3389/fendo.2023.1120420] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 01/24/2023] [Indexed: 02/11/2023] Open
Abstract
Rgp1 was previously identified as a component of a guanine nucleotide exchange factor (GEF) complex to activate Rab6a-mediated trafficking events in and around the Golgi. While the role of Rgp1 in protein trafficking has been examined in vitro and in yeast, the role of Rgp1 during vertebrate embryogenesis and protein trafficking in vivo is unknown. Using genetic, CRISPR-induced zebrafish mutants for Rgp1 loss-of-function, we found that Rgp1 is required for craniofacial cartilage development. Within live rgp1-/- craniofacial chondrocytes, we observed altered movements of Rab6a+ vesicular compartments, consistent with a conserved mechanism described in vitro. Using transmission electron microscopy (TEM) and immunofluorescence analyses, we show that Rgp1 plays a role in the secretion of collagen II, the most abundant protein in cartilage. Our overexpression experiments revealed that Rab8a is a part of the post-Golgi collagen II trafficking pathway. Following loss of Rgp1, chondrocytes activate an Arf4b-mediated stress response and subsequently respond with nuclear DNA fragmentation and cell death. We propose that an Rgp1-regulated Rab6a-Rab8a pathway directs secretion of ECM cargoes such as collagen II, a pathway that may also be utilized in other tissues where coordinated trafficking and secretion of collagens and other large cargoes is required for normal development and tissue function.
Collapse
Affiliation(s)
- Dylan J. Ritter
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, United States
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Dharmendra Choudhary
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Gokhan Unlu
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, United States
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Ela W. Knapik
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, United States
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
5
|
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: 1.0] [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
|
6
|
Tonelotto V, Consorti C, Facchinello N, Trapani V, Sabatelli P, Giraudo C, Spizzotin M, Cescon M, Bertolucci C, Bonaldo P. Collagen VI ablation in zebrafish causes neuromuscular defects during developmental and adult stages. Matrix Biol 2022; 112:39-61. [PMID: 35961424 DOI: 10.1016/j.matbio.2022.08.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 07/21/2022] [Accepted: 08/08/2022] [Indexed: 10/15/2022]
Abstract
Collagen VI (COL6) is an extracellular matrix protein exerting multiple functions in different tissues. In humans, mutations of COL6 genes cause rare inherited congenital disorders, primarily affecting skeletal muscles and collectively known as COL6-related myopathies, for which no cure is available yet. In order to get insights into the pathogenic mechanisms underlying COL6-related diseases, diverse animal models were produced. However, the roles exerted by COL6 during embryogenesis remain largely unknown. Here, we generated the first zebrafish COL6 knockout line through CRISPR/Cas9 site-specific mutagenesis of the col6a1 gene. Phenotypic characterization during embryonic and larval development revealed that lack of COL6 leads to neuromuscular defects and motor dysfunctions, together with distinctive alterations in the three-dimensional architecture of craniofacial cartilages. These phenotypic features were maintained in adult col6a1 null fish, which displayed defective muscle organization and impaired swimming capabilities. Moreover, col6a1 null fish showed autophagy defects and organelle abnormalities at both embryonic and adult stages, thus recapitulating the main features of patients affected by COL6-related myopathies. Mechanistically, lack of COL6 led to increased BMP signaling, and direct inhibition of BMP activity ameliorated the locomotor col6a1 null embryos. Finally performance of, treatment with salbutamol, a β2-adrenergic receptor agonist, elicited a significant amelioration of the neuromuscular and motility defects of col6a1 null fish embryos. Altogether, these findings indicate that this newly generated zebrafish col6a1 null line is a valuable in vivo tool to model COL6-related myopathies and suitable for drug screenings aimed at addressing the quest for effective therapeutic strategies for these disorders.
Collapse
Affiliation(s)
| | - Chiara Consorti
- Department of Molecular Medicine, University of Padova, 35131 Padova, Italy
| | - Nicola Facchinello
- Department of Molecular Medicine, University of Padova, 35131 Padova, Italy
| | - Valeria Trapani
- Department of Molecular Medicine, University of Padova, 35131 Padova, Italy
| | - Patrizia Sabatelli
- CNR - Institute of Molecular Genetics "Luigi Luca Cavalli-Sforza", Unit of Bologna, 40136, Bologna, Italy; IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Chiara Giraudo
- Department of Medicine, Unit of Advanced Clinical and Translational Imaging, University of Padova, 35128 Padova, Italy
| | - Marianna Spizzotin
- Department of Molecular Medicine, University of Padova, 35131 Padova, Italy
| | - Matilde Cescon
- Department of Molecular Medicine, University of Padova, 35131 Padova, Italy
| | - Cristiano Bertolucci
- Department of Life Sciences and Biotechnology, University of Ferrara, 44121 Ferrara, Italy
| | - Paolo Bonaldo
- Department of Molecular Medicine, University of Padova, 35131 Padova, Italy; CRIBI Biotechnology Center, University of Padova, 35131 Padova, Italy.
| |
Collapse
|
7
|
Effect of monosultap on notochord development in zebrafish (Danio rerio) embryos. Toxicology 2022; 477:153276. [PMID: 35933024 DOI: 10.1016/j.tox.2022.153276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 07/21/2022] [Accepted: 07/28/2022] [Indexed: 11/22/2022]
Abstract
Monosultap (Mon) is a broad-spectrum insecticide used in agricultural production to control stem borers in rice fields. Currently, little evidence shows how Mon affects notochord development in zebrafish (Danio rerio). In our study, zebrafish embryos were exposed to 0.25, 0.5, and 0.75 mg/L Mon to determine the effects of different concentrations of Mon on notochord development. Mon exposure reduced the body length, decreased the heart rate and hatchability, and induced notochord deformity in zebrafish. The effects of Mon exposure on the internal organization of the notochord and the structural abnormalities were determined based on histological staining of paraffinized tissue sections. Quantitative polymerase chain reaction (qPCR) and in situ hybridization findings revealed that the expression levels of genes related to notochord development (shha, col2a, and ptch2) showed an increasing trend in a concentration-dependent manner. An abnormal increase of apoptosis and cell proliferation in some parts of the notochord suggested that Mon exposure could cause developmental abnormality of the notochord. This study revealed the toxicity of Mon in notochord development. Our findings provide information in assessing the risk of Mon to the ecological environment and human health.
Collapse
|
8
|
Yoon B, Yeung P, Santistevan N, Bluhm LE, Kawasaki K, Kueper J, Dubielzig R, VanOudenhove J, Cotney J, Liao EC, Grinblat Y. Zebrafish models of alx-linked frontonasal dysplasia reveal a role for Alx1 and Alx3 in the anterior segment and vasculature of the developing eye. Biol Open 2022; 11:bio059189. [PMID: 35142342 PMCID: PMC9167625 DOI: 10.1242/bio.059189] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 01/28/2022] [Indexed: 11/18/2022] Open
Abstract
The cellular and genetic mechanisms that coordinate formation of facial sensory structures with surrounding skeletal and soft tissue elements remain poorly understood. Alx1, a homeobox transcription factor, is a key regulator of midfacial morphogenesis. ALX1 mutations in humans are linked to severe congenital anomalies of the facial skeleton (frontonasal dysplasia, FND) with malformation or absence of eyes and orbital contents (micro- and anophthalmia). Zebrafish with loss-of-function alx1 mutations develop with craniofacial and ocular defects of variable penetrance, likely due to compensatory upregulation in expression of a paralogous gene, alx3. Here we show that zebrafish alx1;alx3 mutants develop with highly penetrant cranial and ocular defects that resemble human ALX1-linked FND. alx1 and alx3 are expressed in anterior cranial neural crest (aCNC), which gives rise to the anterior neurocranium (ANC), anterior segment structures of the eye and vascular pericytes. Consistent with a functional requirement for alx genes in aCNC, alx1; alx3 mutants develop with nearly absent ANC and grossly aberrant hyaloid vasculature and ocular anterior segment, but normal retina. In vivo lineage labeling identified a requirement for alx1 and alx3 during aCNC migration, and transcriptomic analysis suggested oxidative stress response as a key target mechanism of this function. Oxidative stress is a hallmark of fetal alcohol toxicity, and we found increased penetrance of facial and ocular malformations in alx1 mutants exposed to ethanol, consistent with a protective role for alx1 against ethanol toxicity. Collectively, these data demonstrate a conserved role for zebrafish alx genes in controlling ocular and facial development, and a novel role in protecting these key midfacial structures from ethanol toxicity during embryogenesis. These data also reveal novel roles for alx genes in ocular anterior segment formation and vascular development and suggest that retinal deficits in alx mutants may be secondary to aberrant ocular vascularization and anterior segment defects. This study establishes robust zebrafish models for interrogating conserved genetic mechanisms that coordinate facial and ocular development, and for exploring gene--environment interactions relevant to fetal alcohol syndrome.
Collapse
Affiliation(s)
- Baul Yoon
- Departments of Integrative Biology and Neuroscience, University of Wisconsin, Madison, WI 53706, USA
- Genetics Ph.D. Training Program, University of Wisconsin, Madison, WI 53706, USA
| | - Pan Yeung
- Center for Regenerative Medicine, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, and Shriners Hospital for Children, Boston, 02114, USA
| | - Nicholas Santistevan
- Departments of Integrative Biology and Neuroscience, University of Wisconsin, Madison, WI 53706, USA
- Genetics Ph.D. Training Program, University of Wisconsin, Madison, WI 53706, USA
| | - Lauren E. Bluhm
- Departments of Integrative Biology and Neuroscience, University of Wisconsin, Madison, WI 53706, USA
| | - Kenta Kawasaki
- Center for Regenerative Medicine, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, and Shriners Hospital for Children, Boston, 02114, USA
| | - Janina Kueper
- Center for Regenerative Medicine, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, and Shriners Hospital for Children, Boston, 02114, USA
- Institute of Human Genetics, University of Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Richard Dubielzig
- Comparative Ocular Pathology Laboratory of Wisconsin (COPLOW), University of Wisconsin, Madison, WI 53706, USA
| | - Jennifer VanOudenhove
- University of Connecticut School of Medicine, Department of Genetics and Genome Sciences, Farmington, CT 06030, USA
| | - Justin Cotney
- University of Connecticut School of Medicine, Department of Genetics and Genome Sciences, Farmington, CT 06030, USA
| | - Eric C. Liao
- Center for Regenerative Medicine, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, and Shriners Hospital for Children, Boston, 02114, USA
| | - Yevgenya Grinblat
- Departments of Integrative Biology and Neuroscience, University of Wisconsin, Madison, WI 53706, USA
- Genetics Ph.D. Training Program, University of Wisconsin, Madison, WI 53706, USA
| |
Collapse
|
9
|
Metikala S, Casie Chetty S, Sumanas S. Single-cell transcriptome analysis of the zebrafish embryonic trunk. PLoS One 2021; 16:e0254024. [PMID: 34234366 PMCID: PMC8263256 DOI: 10.1371/journal.pone.0254024] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 06/17/2021] [Indexed: 11/27/2022] Open
Abstract
During embryonic development, cells differentiate into a variety of distinct cell types and subtypes with diverse transcriptional profiles. To date, transcriptomic signatures of different cell lineages that arise during development have been only partially characterized. Here we used single-cell RNA-seq to perform transcriptomic analysis of over 20,000 cells disaggregated from the trunk region of zebrafish embryos at the 30 hpf stage. Transcriptional signatures of 27 different cell types and subtypes were identified and annotated during this analysis. This dataset will be a useful resource for many researchers in the fields of developmental and cellular biology and facilitate the understanding of molecular mechanisms that regulate cell lineage choices during development.
Collapse
Affiliation(s)
- Sanjeeva Metikala
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States of America
- Department of Pathology and Cell Biology, USF Health Heart Institute, University of South Florida, Tampa, FL, United States of America
| | - Satish Casie Chetty
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States of America
- Molecular and Developmental Biology Graduate Program, University of Cincinnati, Cincinnati, OH, United States of America
| | - Saulius Sumanas
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States of America
- Department of Pathology and Cell Biology, USF Health Heart Institute, University of South Florida, Tampa, FL, United States of America
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States of America
- * E-mail:
| |
Collapse
|
10
|
Conservation of Zebrafish MicroRNA-145 and Its Role during Neural Crest Cell Development. Genes (Basel) 2021; 12:genes12071023. [PMID: 34209401 PMCID: PMC8306979 DOI: 10.3390/genes12071023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 06/24/2021] [Accepted: 06/28/2021] [Indexed: 02/06/2023] Open
Abstract
The neural crest is a multipotent cell population that develops from the dorsal neural fold of vertebrate embryos in order to migrate extensively and differentiate into a variety of tissues. A number of gene regulatory networks coordinating neural crest cell specification and differentiation have been extensively studied to date. Although several publications suggest a common role for microRNA-145 (miR-145) in molecular reprogramming for cell cycle regulation and/or cellular differentiation, little is known about its role during in vivo cranial neural crest development. By modifying miR-145 levels in zebrafish embryos, abnormal craniofacial development and aberrant pigmentation phenotypes were detected. By whole-mount in situ hybridization, changes in expression patterns of col2a1a and Sry-related HMG box (Sox) transcription factors sox9a and sox9b were observed in overexpressed miR-145 embryos. In agreement, zebrafish sox9b expression was downregulated by miR-145 overexpression. In silico and in vivo analysis of the sox9b 3′UTR revealed a conserved potential miR-145 binding site likely involved in its post-transcriptional regulation. Based on these findings, we speculate that miR-145 participates in the gene regulatory network governing zebrafish chondrocyte differentiation by controlling sox9b expression.
Collapse
|
11
|
Oonuma K, Yamamoto M, Moritsugu N, Okawa N, Mukai M, Sotani M, Tsunemi S, Sugimoto H, Nakagome E, Hasegawa Y, Shimai K, Horie T, Kusakabe TG. Evolution of Developmental Programs for the Midline Structures in Chordates: Insights From Gene Regulation in the Floor Plate and Hypochord Homologues of Ciona Embryos. Front Cell Dev Biol 2021; 9:704367. [PMID: 34235159 PMCID: PMC8256262 DOI: 10.3389/fcell.2021.704367] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Accepted: 05/25/2021] [Indexed: 11/24/2022] Open
Abstract
In vertebrate embryos, dorsal midline tissues, including the notochord, the prechordal plate, and the floor plate, play important roles in patterning of the central nervous system, somites, and endodermal tissues by producing extracellular signaling molecules, such as Sonic hedgehog (Shh). In Ciona, hedgehog.b, one of the two hedgehog genes, is expressed in the floor plate of the embryonic neural tube, while none of the hedgehog genes are expressed in the notochord. We have identified a cis-regulatory region of hedgehog.b that was sufficient to drive a reporter gene expression in the floor plate. The hedgehog.b cis-regulatory region also drove ectopic expression of the reporter gene in the endodermal strand, suggesting that the floor plate and the endodermal strand share a part of their gene regulatory programs. The endodermal strand occupies the same topographic position of the embryo as does the vertebrate hypochord, which consists of a row of single cells lined up immediately ventral to the notochord. The hypochord shares expression of several genes with the floor plate, including Shh and FoxA, and play a role in dorsal aorta development. Whole-embryo single-cell transcriptome analysis identified a number of genes specifically expressed in both the floor plate and the endodermal strand in Ciona tailbud embryos. A Ciona FoxA ortholog FoxA.a is shown to be a candidate transcriptional activator for the midline gene battery. The present findings suggest an ancient evolutionary origin of a common developmental program for the midline structures in Olfactores.
Collapse
Affiliation(s)
- Kouhei Oonuma
- Department of Biology, Faculty of Science and Engineering, Konan University, Kobe, Japan.,Institute for Integrative Neurobiology, Graduate School of Natural Science, Konan University, Kobe, Japan
| | - Maho Yamamoto
- Department of Biology, Faculty of Science and Engineering, Konan University, Kobe, Japan
| | - Naho Moritsugu
- Department of Biology, Faculty of Science and Engineering, Konan University, Kobe, Japan
| | - Nanako Okawa
- Department of Biology, Faculty of Science and Engineering, Konan University, Kobe, Japan.,Institute for Integrative Neurobiology, Graduate School of Natural Science, Konan University, Kobe, Japan
| | - Megumi Mukai
- Department of Biology, Faculty of Science and Engineering, Konan University, Kobe, Japan.,Institute for Integrative Neurobiology, Graduate School of Natural Science, Konan University, Kobe, Japan
| | - Miku Sotani
- Department of Biology, Faculty of Science and Engineering, Konan University, Kobe, Japan.,Institute for Integrative Neurobiology, Graduate School of Natural Science, Konan University, Kobe, Japan
| | - Shuto Tsunemi
- Department of Biology, Faculty of Science and Engineering, Konan University, Kobe, Japan.,Institute for Integrative Neurobiology, Graduate School of Natural Science, Konan University, Kobe, Japan
| | - Haruka Sugimoto
- Department of Biology, Faculty of Science and Engineering, Konan University, Kobe, Japan
| | - Eri Nakagome
- Department of Biology, Faculty of Science and Engineering, Konan University, Kobe, Japan
| | - Yuichi Hasegawa
- Department of Biology, Faculty of Science and Engineering, Konan University, Kobe, Japan.,Institute for Integrative Neurobiology, Graduate School of Natural Science, Konan University, Kobe, Japan
| | - Kotaro Shimai
- Department of Biology, Faculty of Science and Engineering, Konan University, Kobe, Japan.,Institute for Integrative Neurobiology, Graduate School of Natural Science, Konan University, Kobe, Japan
| | - Takeo Horie
- Shimoda Marine Research Center, University of Tsukuba, Shimoda, Japan
| | - Takehiro G Kusakabe
- Department of Biology, Faculty of Science and Engineering, Konan University, Kobe, Japan.,Institute for Integrative Neurobiology, Graduate School of Natural Science, Konan University, Kobe, Japan
| |
Collapse
|
12
|
Park H, You HH, Song G. Multiple toxicity of propineb in developing zebrafish embryos: Neurotoxicity, vascular toxicity, and notochord defects in normal vertebrate development. Comp Biochem Physiol C Toxicol Pharmacol 2021; 243:108993. [PMID: 33529709 DOI: 10.1016/j.cbpc.2021.108993] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 01/06/2021] [Accepted: 01/24/2021] [Indexed: 12/15/2022]
Abstract
A dithiocarbamate (DTC) fungicide, propineb, affects thyroid function and exerts immunotoxicity, cytotoxicity, and neurotoxicity in humans. Long-term exposure to propineb is associated with carcinogenicity, teratogenicity, malfunction of the reproductive system, and abnormalities in vital signs during organ development. However, there is no evidence of acute toxicity attributable to propineb in zebrafish. Therefore, in the present study, we assessed the toxicity of propineb in zebrafish by studying its adverse effects on embryo development, angiogenesis, and notochord development. Embryos with propineb exposure developed morphological and physiological defects and in larvae, apoptosis and notochord defects were induced in the early development stage. Transgenic fli1:eGFP zebrafish exposed to propineb showed abnormal larval development with defects in angiogenesis and deformed vasculature. Propineb induced irreversible damage to the neural development of embryos and neurogenic defects in developing zebrafish in transgenic olig2:dsRED zebrafish. These results show that exposure to propineb triggers abnormalities in different organ systems of zebrafish and suggests the physiological complexity of the response to propineb.
Collapse
Affiliation(s)
- Hahyun Park
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Hyekyoung Hannah You
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Gwonhwa Song
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea.
| |
Collapse
|
13
|
Heubel BP, Bredesen CA, Schilling TF, Le Pabic P. Endochondral growth zone pattern and activity in the zebrafish pharyngeal skeleton. Dev Dyn 2020; 250:74-87. [PMID: 32852849 DOI: 10.1002/dvdy.241] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 08/19/2020] [Accepted: 08/22/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Endochondral ossification is a major bone forming mechanism in vertebrates, defects in which can result in skeletal dysplasia or craniofacial anomalies in humans. The zebrafish holds great potential to advance our understanding of endochondral growth zone development and genetics, yet several important aspects of its biology remain unexplored. Here we provide a comprehensive description of endochondral growth zones in the pharyngeal skeleton, including their developmental progression, cellular activity, and adult fates. RESULTS Postembryonic growth of the pharyngeal skeleton is supported by endochondral growth zones located either at skeletal epiphyses or synchondroses. Col2a1a and col10a1a in situ hybridization and anti-PCNA immunostaining identify resting-, hypertrophic- and proliferative zones, respectively, in pharyngeal synchondroses. Cellular hypertrophy and matrix deposition contribute little, if at all, to axial growth in most skeletal elements. Zebrafish endochondral growth zones develop during metamorphosis and arrest in adults. CONCLUSIONS Two endochondral growth zone configurations in the zebrafish pharyngeal skeleton produce either unidirectional (epiphyses) or bidirectional (synchondroses) growth. Cell proliferation drives endochondral growth and its modulation, in contrast to mammalian long bones in which bone length depends more on cell enlargement during hypertrophy and intramembranous ossification is the default mechanism of bone growth in zebrafish adults.
Collapse
Affiliation(s)
- Brian P Heubel
- Department of Biological Sciences, University of Delaware, Newark, Delaware, USA
| | - Carson A Bredesen
- Department of Biology and Marine Biology, University of North Carolina Wilmington, Wilmington, North Carolina, USA
| | - Thomas F Schilling
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, California, USA
| | - Pierre Le Pabic
- Department of Biology and Marine Biology, University of North Carolina Wilmington, Wilmington, North Carolina, USA
| |
Collapse
|
14
|
Chen JW, Niu X, King MJ, Noedl MT, Tabin CJ, Galloway JL. The mevalonate pathway is a crucial regulator of tendon cell specification. Development 2020; 147:dev.185389. [PMID: 32467241 DOI: 10.1242/dev.185389] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 05/04/2020] [Indexed: 12/20/2022]
Abstract
Tendons and ligaments are crucial components of the musculoskeletal system, yet the pathways specifying these fates remain poorly defined. Through a screen of known bioactive chemicals in zebrafish, we identified a new pathway regulating tendon cell induction. We established that statin, through inhibition of the mevalonate pathway, causes an expansion of the tendon progenitor population. Co-expression and live imaging studies indicate that the expansion does not involve an increase in cell proliferation, but rather results from re-specification of cells from the neural crest-derived sox9a+/sox10+ skeletal lineage. The effect on tendon cell expansion is specific to the geranylgeranylation branch of the mevalonate pathway and is mediated by inhibition of Rac activity. This work establishes a novel role for the mevalonate pathway and Rac activity in regulating specification of the tendon lineage.
Collapse
Affiliation(s)
- Jessica W Chen
- Center for Regenerative Medicine, Harvard Stem Cell Institute, Department of Orthopedic Surgery, Massachusetts General Hospital, Harvard Medical School, 185 Cambridge Street, Boston, MA 02114, USA.,Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Xubo Niu
- Center for Regenerative Medicine, Harvard Stem Cell Institute, Department of Orthopedic Surgery, Massachusetts General Hospital, Harvard Medical School, 185 Cambridge Street, Boston, MA 02114, USA
| | - Matthew J King
- Center for Regenerative Medicine, Harvard Stem Cell Institute, Department of Orthopedic Surgery, Massachusetts General Hospital, Harvard Medical School, 185 Cambridge Street, Boston, MA 02114, USA
| | - Marie-Therese Noedl
- Center for Regenerative Medicine, Harvard Stem Cell Institute, Department of Orthopedic Surgery, Massachusetts General Hospital, Harvard Medical School, 185 Cambridge Street, Boston, MA 02114, USA
| | - Clifford J Tabin
- Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Jenna L Galloway
- Center for Regenerative Medicine, Harvard Stem Cell Institute, Department of Orthopedic Surgery, Massachusetts General Hospital, Harvard Medical School, 185 Cambridge Street, Boston, MA 02114, USA
| |
Collapse
|
15
|
Yasuoka Y. Morphogenetic mechanisms forming the notochord rod: The turgor pressure-sheath strength model. Dev Growth Differ 2020; 62:379-390. [PMID: 32275068 DOI: 10.1111/dgd.12665] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 03/30/2020] [Accepted: 03/31/2020] [Indexed: 12/14/2022]
Abstract
The notochord is a defining feature of chordates. During notochord formation in vertebrates and tunicates, notochord cells display dynamic morphogenetic movement, called convergent extension, in which cells intercalate and align at the dorsal midline. However, in cephalochordates, the most basal group of chordates, the notochord is formed without convergent extension. It is simply developed from mesodermal cells at the dorsal midline. This suggests that convergent extension movement of notochord cells is a secondarily acquired developmental attribute in the common ancestor of olfactores (vertebrates + tunicates), and that the chordate ancestor innovated the notochord upon a foundation of morphogenetic mechanisms independent of cell movement. Therefore, this review focuses on biological features specific to notochord cells, which have been well studied using clawed frogs, zebrafish, and tunicates. Attributes of notochord cells, such as vacuolation, membrane trafficking, extracellular matrix formation, and apoptosis, can be understood in terms of two properties: turgor pressure of vacuoles and strength of the notochord sheath. To maintain the straight rod-like structure of the notochord, these parameters must be counterbalanced. In the future, the turgor pressure-sheath strength model, proposed in this review, will be examined in light of quantitative molecular data and mathematical simulations, illuminating the evolutionary origin of the notochord.
Collapse
Affiliation(s)
- Yuuri Yasuoka
- Marine Genomics Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan.,Laboratory for Comprehensive Genomic Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| |
Collapse
|
16
|
Wood AJ, Cohen N, Joshi V, Li M, Costin A, Hersey L, McKaige EA, Manneken JD, Sonntag C, Miles LB, Siegel A, Currie PD. RGD inhibition of itgb1 ameliorates laminin-α2-deficient zebrafish fibre pathology. Hum Mol Genet 2020; 28:1403-1413. [PMID: 30566586 DOI: 10.1093/hmg/ddy426] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 11/15/2018] [Accepted: 11/22/2018] [Indexed: 01/27/2023] Open
Abstract
Deficiency of muscle basement membrane (MBM) component laminin-α2 leads to muscular dystrophy congenital type 1A (MDC1A), a currently untreatable myopathy. Laminin--α2 has two main binding partners within the MBM, dystroglycan and integrin. Integrins coordinate both cell adhesion and signalling; however, there is little mechanistic insight into integrin's function at the MBM. In order to study integrin's role in basement membrane development and how this relates to the MBM's capacity to handle force, an itgβ1.b-/- zebrafish line was created. Histological examination revealed increased extracellular matrix (ECM) deposition at the MBM in the itgβ1.b-/- fish when compared with controls. Surprisingly, both laminin and collagen proteins were found to be increased in expression at the MBM of the itgβ1.b-/- larvae when compared with controls. This increase in ECM components resulted in a decrease in myotomal elasticity as determined by novel passive force analyses. To determine if it was possible to control ECM deposition at the MBM by manipulating integrin activity, RGD peptide, a potent inhibitor of integrin-β1, was injected into a zebrafish model of MDC1A. As postulated an increase in laminin and collagen was observed in the lama2-/- mutant MBM. Importantly, there was also an improvement in fibre stability at the MBM, judged by a reduction in fibre pathology. These results therefore show that blocking ITGβ1 signalling increases ECM deposition at the MBM, a process that could be potentially exploited for treatment of MDC1A.
Collapse
Affiliation(s)
- Alasdair J Wood
- Australian Regenerative Medicine Institute, Monash University, Innovation Walk, Clayton Campus, Wellington Road, Clayton, VIC, Australia
| | - Naomi Cohen
- Australian Regenerative Medicine Institute, Monash University, Innovation Walk, Clayton Campus, Wellington Road, Clayton, VIC, Australia
| | - Veronica Joshi
- Australian Regenerative Medicine Institute, Monash University, Innovation Walk, Clayton Campus, Wellington Road, Clayton, VIC, Australia
| | - Mei Li
- Australian Regenerative Medicine Institute, Monash University, Innovation Walk, Clayton Campus, Wellington Road, Clayton, VIC, Australia
| | - Adam Costin
- Ramaciotti Centre for Electron Microscopy, Monash University, Innovation Walk, Clayton Campus, Wellington Road, Clayton, VIC, Australia
| | - Lucy Hersey
- Australian Regenerative Medicine Institute, Monash University, Innovation Walk, Clayton Campus, Wellington Road, Clayton, VIC, Australia
| | - Emily A McKaige
- Australian Regenerative Medicine Institute, Monash University, Innovation Walk, Clayton Campus, Wellington Road, Clayton, VIC, Australia
| | - Jessica D Manneken
- Australian Regenerative Medicine Institute, Monash University, Innovation Walk, Clayton Campus, Wellington Road, Clayton, VIC, Australia
| | - Carmen Sonntag
- Australian Regenerative Medicine Institute, Monash University, Innovation Walk, Clayton Campus, Wellington Road, Clayton, VIC, Australia
| | - Lee B Miles
- Department of Physiology, Anatomy and Microbiology, Latrobe University, Melbourne (Bundoora), VIC, Australia
| | - Ashley Siegel
- Australian Regenerative Medicine Institute, Monash University, Innovation Walk, Clayton Campus, Wellington Road, Clayton, VIC, Australia
| | - Peter D Currie
- Australian Regenerative Medicine Institute, Monash University, Innovation Walk, Clayton Campus, Wellington Road, Clayton, VIC, Australia.,Victorian Node, EMBL Australia, Clayton, VIC, Australia
| |
Collapse
|
17
|
Dicks A, Wu CL, Steward N, Adkar SS, Gersbach CA, Guilak F. Prospective isolation of chondroprogenitors from human iPSCs based on cell surface markers identified using a CRISPR-Cas9-generated reporter. Stem Cell Res Ther 2020; 11:66. [PMID: 32070421 PMCID: PMC7026983 DOI: 10.1186/s13287-020-01597-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 02/05/2020] [Accepted: 02/11/2020] [Indexed: 12/29/2022] Open
Abstract
Background Articular cartilage shows little or no capacity for intrinsic repair, generating a critical need of regenerative therapies for joint injuries and diseases such as osteoarthritis. Human-induced pluripotent stem cells (hiPSCs) offer a promising cell source for cartilage tissue engineering and in vitro human disease modeling; however, off-target differentiation remains a challenge during hiPSC chondrogenesis. Therefore, the objective of this study was to identify cell surface markers that define the true chondroprogenitor population and use these markers to purify iPSCs as a means of improving the homogeneity and efficiency of hiPSC chondrogenic differentiation. Methods We used a CRISPR-Cas9-edited COL2A1-GFP knock-in reporter hiPSC line, coupled with a surface marker screen, to identify a novel chondroprogenitor population. Single-cell RNA sequencing was then used to analyze the distinct clusters within the population. An unpaired t test with Welch’s correction or an unpaired Kolmogorov-Smirnov test was performed with significance reported at a 95% confidence interval. Results Chondroprogenitors expressing CD146, CD166, and PDGFRβ, but not CD45, made up an average of 16.8% of the total population. Under chondrogenic culture conditions, these triple-positive chondroprogenitor cells demonstrated decreased heterogeneity as measured by single-cell RNA sequencing with fewer clusters (9 clusters in unsorted vs. 6 in sorted populations) closer together. Additionally, there was more robust and homogenous matrix production (unsorted: 1.5 ng/ng vs. sorted: 19.9 ng/ng sGAG/DNA; p < 0.001) with significantly higher chondrogenic gene expression (i.e., SOX9, COL2A1, ACAN; p < 0.05). Conclusions Overall, this study has identified a unique hiPSC-derived subpopulation of chondroprogenitors that are CD146+/CD166+/PDGFRβ+/CD45− and exhibit high chondrogenic potential, providing a purified cell source for cartilage tissue engineering or disease modeling studies.
Collapse
Affiliation(s)
- Amanda Dicks
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO, 63110, USA.,Shriners Hospitals for Children - St. Louis, St. Louis, MO, 63110, USA.,Department of Biomedical Engineering, Washington University, St. Louis, MO, 63110, USA.,Center of Regenerative Medicine, Washington University, St. Louis, MO, 63110, USA
| | - Chia-Lung Wu
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO, 63110, USA.,Shriners Hospitals for Children - St. Louis, St. Louis, MO, 63110, USA.,Center of Regenerative Medicine, Washington University, St. Louis, MO, 63110, USA
| | - Nancy Steward
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO, 63110, USA.,Shriners Hospitals for Children - St. Louis, St. Louis, MO, 63110, USA.,Center of Regenerative Medicine, Washington University, St. Louis, MO, 63110, USA
| | - Shaunak S Adkar
- Department of Cell Biology, Duke University Medical Center, Durham, NC, 27710, USA
| | - Charles A Gersbach
- Department of Biomedical Engineering, Duke University, Durham, NC, 27710, USA
| | - Farshid Guilak
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO, 63110, USA. .,Shriners Hospitals for Children - St. Louis, St. Louis, MO, 63110, USA. .,Department of Biomedical Engineering, Washington University, St. Louis, MO, 63110, USA. .,Center of Regenerative Medicine, Washington University, St. Louis, MO, 63110, USA.
| |
Collapse
|
18
|
Spatio-temporal expression and distribution of collagen VI during zebrafish development. Sci Rep 2019; 9:19851. [PMID: 31882701 PMCID: PMC6934817 DOI: 10.1038/s41598-019-56445-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 12/10/2019] [Indexed: 01/27/2023] Open
Abstract
Collagen VI (ColVI) is an extracellular matrix (ECM) protein involved in a range of physiological and pathological conditions. Zebrafish (Danio rerio) is a powerful model organism for studying vertebrate development and for in vivo analysis of tissue patterning. Here, we performed a thorough characterization of ColVI gene and protein expression in zebrafish during development and adult life. Bioinformatics analyses confirmed that zebrafish genome contains single genes encoding for α1(VI), α2(VI) and α3(VI) ColVI chains and duplicated genes encoding for α4(VI) chains. At 1 day post-fertilization (dpf) ColVI transcripts are expressed in myotomes, pectoral fin buds and developing epidermis, while from 2 dpf abundant transcript levels are present in myosepta, pectoral fins, axial vasculature, gut and craniofacial cartilage elements. Using newly generated polyclonal antibodies against zebrafish α1(VI) protein, we found that ColVI deposition in adult fish delineates distinct domains in the ECM of several organs, including cartilage, eye, skin, spleen and skeletal muscle. Altogether, these data provide the first detailed characterization of ColVI expression and ECM deposition in zebrafish, thus paving the way for further functional studies in this species.
Collapse
|
19
|
Watt KEN, Neben CL, Hall S, Merrill AE, Trainor PA. tp53-dependent and independent signaling underlies the pathogenesis and possible prevention of Acrofacial Dysostosis-Cincinnati type. Hum Mol Genet 2019; 27:2628-2643. [PMID: 29750247 DOI: 10.1093/hmg/ddy172] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 04/11/2018] [Accepted: 05/02/2018] [Indexed: 01/05/2023] Open
Abstract
Ribosome biogenesis is a global process required for growth and proliferation in all cells, but disruptions in this process surprisingly lead to tissue-specific phenotypic disorders termed ribosomopathies. Pathogenic variants in the RNA Polymerase (Pol) I subunit POLR1A cause Acrofacial Dysostosis-Cincinnati type, which is characterized by craniofacial and limb anomalies. In a zebrafish model of Acrofacial Dysostosis-Cincinnati type, we demonstrate that polr1a-/- mutants exhibit deficient 47S rRNA transcription, reduced monosomes and polysomes and, consequently, defects in protein translation. This results in Tp53-dependent neuroepithelial apoptosis, diminished neural crest cell proliferation and cranioskeletal anomalies. This indicates that POLR1A is critical for rRNA transcription, which is considered a rate limiting step in ribosome biogenesis, underpinning its requirement for neuroepithelial cell and neural crest cell proliferation and survival. To understand the contribution of the Tp53 pathway to the pathogenesis of Acrofacial Dysostosis-Cincinnati type, we genetically inhibited tp53 in polr1a-/- mutant embryos. Tp53 inhibition suppresses neuroepithelial apoptosis and partially ameliorates the polr1a mutant phenotype. However, complete rescue of cartilage development is not observed due to the failure to improve rDNA transcription and neural crest cell proliferation. Altogether, these data reveal specific functions for both Tp53-dependent and independent signaling downstream of polr1a in ribosome biogenesis during neural crest cell and craniofacial development, in the pathogenesis of Acrofacial Dysostosis-Cincinnati type. Furthermore, our work sets the stage for identifying Tp53-independent therapies to potentially prevent Acrofacial dysostosis-Cincinnati type and other similar ribosomopathies.
Collapse
Affiliation(s)
- Kristin E N Watt
- Stowers Institute for Medical Research, Kansas City, MO, USA.,Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Cynthia L Neben
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, USA.,Department of Biochemistry and Molecular Biology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Shawn Hall
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Amy E Merrill
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, USA.,Department of Biochemistry and Molecular Biology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Paul A Trainor
- Stowers Institute for Medical Research, Kansas City, MO, USA.,Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
20
|
Rocha M, Singh N, Ahsan K, Beiriger A, Prince VE. Neural crest development: insights from the zebrafish. Dev Dyn 2019; 249:88-111. [PMID: 31591788 DOI: 10.1002/dvdy.122] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 09/21/2019] [Accepted: 09/22/2019] [Indexed: 12/12/2022] Open
Abstract
Our understanding of the neural crest, a key vertebrate innovation, is built upon studies of multiple model organisms. Early research on neural crest cells (NCCs) was dominated by analyses of accessible amphibian and avian embryos, with mouse genetics providing complementary insights in more recent years. The zebrafish model is a relative newcomer to the field, yet it offers unparalleled advantages for the study of NCCs. Specifically, zebrafish provide powerful genetic and transgenic tools, coupled with rapidly developing transparent embryos that are ideal for high-resolution real-time imaging of the dynamic process of neural crest development. While the broad principles of neural crest development are largely conserved across vertebrate species, there are critical differences in anatomy, morphogenesis, and genetics that must be considered before information from one model is extrapolated to another. Here, our goal is to provide the reader with a helpful primer specific to neural crest development in the zebrafish model. We focus largely on the earliest events-specification, delamination, and migration-discussing what is known about zebrafish NCC development and how it differs from NCC development in non-teleost species, as well as highlighting current gaps in knowledge.
Collapse
Affiliation(s)
- Manuel Rocha
- Committee on Development, Regeneration and Stem Cell Biology, The University of Chicago, Chicago, Illinois
| | - Noor Singh
- Department of Organismal Biology and Anatomy, The University of Chicago, Chicago, Illinois
| | - Kamil Ahsan
- Committee on Development, Regeneration and Stem Cell Biology, The University of Chicago, Chicago, Illinois
| | - Anastasia Beiriger
- Committee on Development, Regeneration and Stem Cell Biology, The University of Chicago, Chicago, Illinois
| | - Victoria E Prince
- Committee on Development, Regeneration and Stem Cell Biology, The University of Chicago, Chicago, Illinois.,Department of Organismal Biology and Anatomy, The University of Chicago, Chicago, Illinois
| |
Collapse
|
21
|
Gebuijs IGE, Raterman ST, Metz JR, Swanenberg L, Zethof J, Van den Bos R, Carels CEL, Wagener FADTG, Von den Hoff JW. Fgf8a mutation affects craniofacial development and skeletal gene expression in zebrafish larvae. Biol Open 2019; 8:bio.039834. [PMID: 31471293 PMCID: PMC6777363 DOI: 10.1242/bio.039834] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Craniofacial development is tightly regulated and therefore highly vulnerable to disturbance by genetic and environmental factors. Fibroblast growth factors (FGFs) direct migration, proliferation and survival of cranial neural crest cells (CNCCs) forming the human face. In this study, we analyzed bone and cartilage formation in the head of five dpf fgf8ati282 zebrafish larvae and assessed gene expression levels for 11 genes involved in these processes. In addition, in situ hybridization was performed on 8 and 24 hours post fertilization (hpf) larvae (fgf8a, dlx2a, runx2a, col2a1a). A significant size reduction of eight out of nine craniofacial cartilage structures was found in homozygous mutant (6–36%, P<0.01) and heterozygous (7–24%, P<0.01) larvae. Also, nine mineralized structures were not observed in all or part of the homozygous (0–71%, P<0.0001) and heterozygous (33–100%, P<0.0001) larvae. In homozygote mutants, runx2a and sp7 expression was upregulated compared to wild type, presumably to compensate for the reduced bone formation. Decreased col9a1b expression may compromise cartilage formation. Upregulated dlx2a in homozygotes indicates impaired CNCC function. Dlx2a expression was reduced in the first and second stream of CNCCs in homozygous mutants at 24 hpf, as shown by in situ hybridization. This indicates an impairment of CNCC migration and survival by fgf8 mutation. Summary: A function-blocking mutation in fgf8a causes craniofacial malformations in zebrafish larvae due to impaired cranial neural crest cell migration and survival.
Collapse
Affiliation(s)
- I G E Gebuijs
- Department of Orthodontics and Craniofacial Biology, Radboudumc, Nijmegen, The Netherlands.,Department of Orthodontics and Craniofacial Biology, Radboud Institute of Molecular Life Sciences, Nijmegen, The Netherlands.,Department of Animal Ecology and Physiology, Radboud University, Nijmegen, The Netherlands
| | - S T Raterman
- Department of Orthodontics and Craniofacial Biology, Radboudumc, Nijmegen, The Netherlands.,Department of Orthodontics and Craniofacial Biology, Radboud Institute of Molecular Life Sciences, Nijmegen, The Netherlands.,Department of Animal Ecology and Physiology, Radboud University, Nijmegen, The Netherlands
| | - J R Metz
- Department of Animal Ecology and Physiology, Radboud University, Nijmegen, The Netherlands
| | - L Swanenberg
- Department of Orthodontics and Craniofacial Biology, Radboudumc, Nijmegen, The Netherlands.,Department of Animal Ecology and Physiology, Radboud University, Nijmegen, The Netherlands
| | - J Zethof
- Department of Animal Ecology and Physiology, Radboud University, Nijmegen, The Netherlands
| | - R Van den Bos
- Department of Animal Ecology and Physiology, Radboud University, Nijmegen, The Netherlands
| | - C E L Carels
- Department of Orthodontics and Craniofacial Biology, Radboud Institute of Molecular Life Sciences, Nijmegen, The Netherlands.,Department of Human Genetics, Radboudumc, Nijmegen, The Netherlands.,Department of Oral Health Sciences and Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - F A D T G Wagener
- Department of Orthodontics and Craniofacial Biology, Radboudumc, Nijmegen, The Netherlands.,Department of Orthodontics and Craniofacial Biology, Radboud Institute of Molecular Life Sciences, Nijmegen, The Netherlands
| | - J W Von den Hoff
- Department of Orthodontics and Craniofacial Biology, Radboudumc, Nijmegen, The Netherlands .,Department of Orthodontics and Craniofacial Biology, Radboud Institute of Molecular Life Sciences, Nijmegen, The Netherlands
| |
Collapse
|
22
|
Nucleoporin 62-Like Protein is Required for the Development of Pharyngeal Arches through Regulation of Wnt/β-Catenin Signaling and Apoptotic Homeostasis in Zebrafish. Cells 2019; 8:cells8091038. [PMID: 31492028 PMCID: PMC6770318 DOI: 10.3390/cells8091038] [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: 07/24/2019] [Revised: 09/04/2019] [Accepted: 09/04/2019] [Indexed: 12/12/2022] Open
Abstract
We have previously observed the predominant expression of nucleoporin 62-like (Nup62l) mRNA in the pharyngeal region of zebrafish, which raises the question whether Nup62l has important implications in governing the morphogenesis of pharyngeal arches (PA) in zebrafish. Herein, we explored the functions of Nup62l in PA development. The disruption of Nup62l with a CRISPR/Cas9-dependent gene knockout approach led to defective PA, which was characterized by a thinned and shortened pharyngeal region and a significant loss of pharyngeal cartilages. During pharyngeal cartilage formation, prechondrogenic condensation and chondrogenic differentiation were disrupted in homozygous nup62l-mutants, while the specification and migration of cranial neural crest cells (CNCCs) were unaffected. Mechanistically, the impaired PA region of nup62l-mutants underwent extensive apoptosis, which was mainly dependent on activation of p53-dependent apoptotic pathway. Moreover, aberrant activation of a series of apoptotic pathways in nup62l-mutants is closely associated with the inactivation of Wnt/β-catenin signaling. Thus, these findings suggest that the regulation of Wnt/β-catenin activity by Nup62l is crucial for PA formation in zebrafish.
Collapse
|
23
|
Kindermann B, Valkova C, Krämer A, Perner B, Engelmann C, Behrendt L, Kritsch D, Jungnickel B, Kehlenbach RH, Oswald F, Englert C, Kaether C. The nuclear pore proteins Nup88/214 and T-cell acute lymphatic leukemia-associated NUP214 fusion proteins regulate Notch signaling. J Biol Chem 2019; 294:11741-11750. [PMID: 31186352 DOI: 10.1074/jbc.ra118.006357] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 05/20/2019] [Indexed: 01/14/2023] Open
Abstract
The Notch receptor is a key mediator of developmental programs and cell-fate decisions. Imbalanced Notch signaling leads to developmental disorders and cancer. To fully characterize the Notch signaling pathway and exploit it in novel therapeutic interventions, a comprehensive view on the regulation and requirements of Notch signaling is needed. Notch is regulated at different levels, ranging from ligand binding, stability to endocytosis. Using an array of different techniques, including reporter gene assays, immunocytochemistry, and ChIP-qPCR we show here, to the best of our knowledge for the first time, regulation of Notch signaling at the level of the nuclear pore. We found that the nuclear pore protein Nup214 (nucleoporin 214) and its interaction partner Nup88 negatively regulate Notch signaling in vitro and in vivo in zebrafish. In mammalian cells, loss of Nup88/214 inhibited nuclear export of recombination signal-binding protein for immunoglobulin κJ region (RBP-J), the DNA-binding component of the Notch pathway. This inhibition increased binding of RBP-J to its cognate promoter regions, resulting in increased downstream Notch signaling. Interestingly, we also found that NUP214 fusion proteins, causative for certain cases of T-cell acute lymphatic leukemia, potentially contribute to tumorigenesis via a Notch-dependent mechanism. In summary, the nuclear pore components Nup88/214 suppress Notch signaling in vitro, and in zebrafish, nuclear RBP-J levels are rate-limiting factors for Notch signaling in mammalian cells, and regulation of nucleocytoplasmic transport of RBP-J may contribute to fine-tuning Notch activity in cells.
Collapse
Affiliation(s)
- Bastian Kindermann
- Leibniz Institut für Alternsforschung-Fritz Lipmann Institut, 07745 Jena, Germany
| | - Christina Valkova
- Leibniz Institut für Alternsforschung-Fritz Lipmann Institut, 07745 Jena, Germany
| | - Andreas Krämer
- Leibniz Institut für Alternsforschung-Fritz Lipmann Institut, 07745 Jena, Germany
| | - Birgit Perner
- Leibniz Institut für Alternsforschung-Fritz Lipmann Institut, 07745 Jena, Germany
| | - Christian Engelmann
- Leibniz Institut für Alternsforschung-Fritz Lipmann Institut, 07745 Jena, Germany
| | - Laura Behrendt
- Leibniz Institut für Alternsforschung-Fritz Lipmann Institut, 07745 Jena, Germany
| | - Daniel Kritsch
- Institut für Biochemie und Biophysik, Friedrich Schiller Universität Jena, 07745 Jena, Germany
| | - Berit Jungnickel
- Institut für Biochemie und Biophysik, Friedrich Schiller Universität Jena, 07745 Jena, Germany
| | - Ralph H Kehlenbach
- Department of Molecular Biology, Universitätsmedizin Göttingen, 37073 Göttingen, Germany
| | - Franz Oswald
- Universitätsklinikum Ulm, Zentrum für Innere Medizin, Abteilung für Innere Medizin I, 89081 Ulm, Germany
| | - Christoph Englert
- Leibniz Institut für Alternsforschung-Fritz Lipmann Institut, 07745 Jena, Germany.,Institut für Biochemie und Biophysik, Friedrich Schiller Universität Jena, 07745 Jena, Germany
| | - Christoph Kaether
- Leibniz Institut für Alternsforschung-Fritz Lipmann Institut, 07745 Jena, Germany
| |
Collapse
|
24
|
Bretaud S, Nauroy P, Malbouyres M, Ruggiero F. Fishing for collagen function: About development, regeneration and disease. Semin Cell Dev Biol 2019; 89:100-108. [DOI: 10.1016/j.semcdb.2018.10.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2018] [Revised: 09/06/2018] [Accepted: 10/08/2018] [Indexed: 02/07/2023]
|
25
|
Mochida K, Ito K, Ito M, Hano T, Ohkubo N. Toxicity of the biocide polycarbamate, used for aquaculture nets, to some marine fish species. Comp Biochem Physiol C Toxicol Pharmacol 2018; 214:61-67. [PMID: 30201584 DOI: 10.1016/j.cbpc.2018.09.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 08/29/2018] [Accepted: 09/04/2018] [Indexed: 01/05/2023]
Abstract
We investigated toxic effects of the antifouling biocide polycarbamate (PC) on marine fish by conducting acute, early-life stage toxicity (ELS), and embryo toxicity tests. Mummichog (Fundulus heteroclitus) 96-h LC50 values for hatched larvae (body weight about 2.0 mg) and juveniles (660 ± 36 mg) were about 12 and 630 μg/L, respectively. The ELS test using mummichog embryos yielded a lowest-observed-effect concentration of 3.9 μg/L and a no-observed-effect concentration of 2.1 μg/L with growth as the most sensitive endpoint. The embryo toxicity test for spotted halibut (Verasper variegatus) revealed a 10-d EC50 of 8.1 μg/L with abnormality as an endpoint. During the ELS and embryo toxicity tests, morphological abnormalities (notochord undulation) were induced in the embryos. Biochemical and gene-expression analysis suggest that PC-induced morphological abnormalities involve disruption of lysyl oxidase-mediated collagen fiber organization, essential for notochord formation, and inhibition of gene expression related to notochord formation.
Collapse
Affiliation(s)
- Kazuhiko Mochida
- National Research Institute of Fisheries and Environment of Inland Sea, Fisheries Research and Education Agency, 2-17-5 Maruishi, Hatsukaichi, Hiroshima 739-0452, Japan.
| | - Katsutoshi Ito
- National Research Institute of Fisheries and Environment of Inland Sea, Fisheries Research and Education Agency, 2-17-5 Maruishi, Hatsukaichi, Hiroshima 739-0452, Japan
| | - Mana Ito
- National Research Institute of Fisheries and Environment of Inland Sea, Fisheries Research and Education Agency, 2-17-5 Maruishi, Hatsukaichi, Hiroshima 739-0452, Japan
| | - Takeshi Hano
- National Research Institute of Fisheries and Environment of Inland Sea, Fisheries Research and Education Agency, 2-17-5 Maruishi, Hatsukaichi, Hiroshima 739-0452, Japan
| | - Nobuyuki Ohkubo
- National Research Institute of Fisheries and Environment of Inland Sea, Fisheries Research and Education Agency, 2-17-5 Maruishi, Hatsukaichi, Hiroshima 739-0452, Japan
| |
Collapse
|
26
|
Kindt LM, Coughlin AR, Perosino TR, Ersfeld HN, Hampton M, Liang JO. Identification of transcripts potentially involved in neural tube closure using RNA sequencing. Genesis 2018; 56:e23096. [PMID: 29488319 DOI: 10.1002/dvg.23096] [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: 05/13/2017] [Revised: 02/02/2018] [Accepted: 02/19/2018] [Indexed: 01/08/2023]
Abstract
Anencephaly is a fatal human neural tube defect (NTD) in which the anterior neural tube remains open. Zebrafish embryos with reduced Nodal signaling display an open anterior neural tube phenotype that is analogous to anencephaly. Previous work from our laboratory suggests that Nodal signaling acts through induction of the head mesendoderm and mesoderm. Head mesendoderm/mesoderm then, through an unknown mechanism, promotes formation of the polarized neuroepithelium that is capable of undergoing the movements required for closure. We compared the transcriptome of embryos treated with a Nodal signaling inhibitor at sphere stage, which causes NTDs, to embryos treated at 30% epiboly, which does not cause NTDs. This screen identified over 3,000 transcripts with potential roles in anterior neurulation. Expression of several genes encoding components of tight and adherens junctions was significantly reduced, supporting the model that Nodal signaling regulates formation of the neuroepithelium. mRNAs involved in Wnt, FGF, and BMP signaling were also differentially expressed, suggesting these pathways might regulate anterior neurulation. In support of this, we found that pharmacological inhibition of FGF-receptor function causes an open anterior NTD as well as loss of mesodermal derivatives. This suggests that Nodal and FGF signaling both promote anterior neurulation through induction of head mesoderm.
Collapse
Affiliation(s)
- Lexy M Kindt
- Department of Biology, University of Minnesota Duluth, Duluth.,Integrated Biosciences Graduate Program, University of Minnesota, Duluth
| | - Alicia R Coughlin
- Department of Biology, University of Minnesota Duluth, Duluth.,Integrated Biosciences Graduate Program, University of Minnesota, Duluth
| | | | - Haley N Ersfeld
- Department of Biology, University of Minnesota Duluth, Duluth
| | - Marshall Hampton
- Integrated Biosciences Graduate Program, University of Minnesota, Duluth.,Department of Mathematics and Statistics, University of Minnesota Duluth, Duluth
| | - Jennifer O Liang
- Department of Biology, University of Minnesota Duluth, Duluth.,Integrated Biosciences Graduate Program, University of Minnesota, Duluth
| |
Collapse
|
27
|
Trapani V, Bonaldo P, Corallo D. Role of the ECM in notochord formation, function and disease. J Cell Sci 2017; 130:3203-3211. [PMID: 28883093 DOI: 10.1242/jcs.175950] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The notochord is a midline structure common to all chordate animals; it provides mechanical and signaling cues for the developing embryo. In vertebrates, the notochord plays key functions during embryogenesis, being a source of developmental signals that pattern the surrounding tissues. It is composed of a core of vacuolated cells surrounded by an epithelial-like sheath of cells that secrete a thick peri-notochordal basement membrane made of different extracellular matrix (ECM) proteins. The correct deposition and organization of the ECM is essential for proper notochord morphogenesis and function. Work carried out in the past two decades has allowed researchers to dissect the contribution of different ECM components to this embryonic tissue. Here, we will provide an overview of these genetic and mechanistic studies. In particular, we highlight the specific functions of distinct matrix molecules in regulating notochord development and notochord-derived signals. Moreover, we also discuss the involvement of ECM synthesis and its remodeling in the pathogenesis of chordoma, a malignant bone cancer that originates from remnants of notochord remaining after embryogenesis.
Collapse
Affiliation(s)
- Valeria Trapani
- Department of Molecular Medicine, University of Padova, 35131 Padova, Italy
| | - Paolo Bonaldo
- Department of Molecular Medicine, University of Padova, 35131 Padova, Italy .,CRIBI Biotechnology Center, University of Padova, Padova, 35131, Italy
| | - Diana Corallo
- Department of Molecular Medicine, University of Padova, 35131 Padova, Italy .,Pediatric Research Institute, Città della Speranza, 35127 Padova, Italy
| |
Collapse
|
28
|
Kara N, Wei C, Commanday AC, Patton JG. miR-27 regulates chondrogenesis by suppressing focal adhesion kinase during pharyngeal arch development. Dev Biol 2017. [PMID: 28625871 PMCID: PMC5582384 DOI: 10.1016/j.ydbio.2017.06.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Cranial neural crest cells are a multipotent cell population that generate all the elements of the pharyngeal cartilage with differentiation into chondrocytes tightly regulated by temporal intracellular and extracellular cues. Here, we demonstrate a novel role for miR-27, a highly enriched microRNA in the pharyngeal arches, as a positive regulator of chondrogenesis. Knock down of miR-27 led to nearly complete loss of pharyngeal cartilage by attenuating proliferation and blocking differentiation of pre-chondrogenic cells. Focal adhesion kinase (FAK) is a key regulator in integrin-mediated extracellular matrix (ECM) adhesion and has been proposed to function as a negative regulator of chondrogenesis. We show that FAK is downregulated in the pharyngeal arches during chondrogenesis and is a direct target of miR-27. Suppressing the accumulation of FAK in miR-27 morphants partially rescued the severe pharyngeal cartilage defects observed upon knock down of miR-27. These data support a crucial role for miR-27 in promoting chondrogenic differentiation in the pharyngeal arches through regulation of FAK.
Collapse
Affiliation(s)
- Nergis Kara
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, United States
| | - Chunyao Wei
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, United States
| | - Alexander C Commanday
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, United States
| | - James G Patton
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, United States.
| |
Collapse
|
29
|
Development Aspects of Zebrafish Myotendinous Junction: a Model System for Understanding Muscle Basement Membrane Formation and Failure. CURRENT PATHOBIOLOGY REPORTS 2017. [DOI: 10.1007/s40139-017-0140-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
30
|
Signore IA, Jerez C, Figueroa D, Suazo J, Marcelain K, Cerda O, Colombo Flores A. Inhibition of the 3-hydroxy-3-methyl-glutaryl-CoA reductase induces orofacial defects in zebrafish. ACTA ACUST UNITED AC 2016; 106:814-830. [PMID: 27488927 DOI: 10.1002/bdra.23546] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 06/13/2016] [Accepted: 06/22/2016] [Indexed: 12/20/2022]
Abstract
BACKGROUND Orofacial clefts (OFCs) are common birth defects, which include a range of disorders with a complex etiology affecting formation of craniofacial structures. Some forms of syndromic OFCs are produced by defects in the cholesterol pathway. The principal enzyme of the cholesterol pathway is the 3-hydroxy-3-methyl-glutaryl-CoA reductase (HMGCR). Our aim is to study whether defects of HMGCR function would produce orofacial malformation similar to those found in disorders of cholesterol synthesis. METHODS We used zebrafish hmgcrb mutants and HMGCR inhibition assay using atorvastatin during early and late stages of orofacial morphogenesis in zebrafish. To describe craniofacial phenotypes, we stained cartilage and bone and performed in situ hybridization using known craniofacial markers. Also, we visualized neural crest cell migration in a transgenic fish. RESULTS Our results showed that mutants displayed loss of cartilage and diminished orofacial outgrowth, and in some cases palatal cleft. Late treatments with statin show a similar phenotype. Affected-siblings displayed a moderate phenotype, whereas early-treated embryos had a minor cleft. We found reduced expression of the downstream component of Sonic Hedgehog-signaling gli1 in ventral brain, oral ectoderm, and pharyngeal endoderm in mutants and in late atorvastatin-treated embryos. CONCLUSION Our results suggest that HMGCR loss-of-function primarily affects postmigratory cranial neural crest cells through abnormal Sonic Hedgehog signaling, probably induced by reduction in metabolites of the cholesterol pathway. Malformation severity correlates with the grade of HMGCR inhibition, developmental stage of its disruption, and probably with availability of maternal lipids. Together, our results might help to understand the spectrum of orofacial phenotypes found in cholesterol synthesis disorders. Birth Defects Research (Part A) 106:814-830, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Iskra A Signore
- Programa de Anatomía y Biología del Desarrollo, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, Chile.,Instituto de Filosofía y Ciencias de la Complejidad (IFICC), Santiago, Chile
| | - Carolina Jerez
- Programa de Anatomía y Biología del Desarrollo, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Diego Figueroa
- Programa de Anatomía y Biología del Desarrollo, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - José Suazo
- Institute for Research in Dental Sciences, Facultad de Odontología, Universidad de Chile, Santiago, Chile
| | - Katherine Marcelain
- Programa de Genética Humana, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Oscar Cerda
- Programa de Biología Celular y Molecular, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Alicia Colombo Flores
- Programa de Anatomía y Biología del Desarrollo, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, Chile. .,Servicio de Anatomía Patológica, Hospital Clínico de la Universidad de Chile, Santiago, Chile.
| |
Collapse
|
31
|
Talbot JC, Nichols JT, Yan YL, Leonard IF, BreMiller RA, Amacher SL, Postlethwait JH, Kimmel CB. Pharyngeal morphogenesis requires fras1-itga8-dependent epithelial-mesenchymal interaction. Dev Biol 2016; 416:136-148. [PMID: 27265864 PMCID: PMC4967372 DOI: 10.1016/j.ydbio.2016.05.035] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 05/31/2016] [Indexed: 01/08/2023]
Abstract
Both Fras1 and Itga8 connect mesenchymal cells to epithelia by way of an extracellular 'Fraser protein complex' that functions in signaling and adhesion; these proteins are vital to the development of several vertebrate organs. We previously found that zebrafish fras1 mutants have craniofacial defects, specifically, shortened symplectic cartilages and cartilage fusions that spare joint elements. During a forward mutagenesis screen, we identified a new zebrafish mutation, b1161, that we show here disrupts itga8, as confirmed using CRISPR-generated itga8 alleles. fras1 and itga8 single mutants and double mutants have similar craniofacial phenotypes, a result expected if loss of either gene disrupts function of the Fraser protein complex. Unlike fras1 mutants or other Fraser-related mutants, itga8 mutants do not show blistered tail fins. Thus, the function of the Fraser complex differs in the craniofacial skeleton and the tail fin. Focusing on the face, we find that itga8 mutants consistently show defective outpocketing of a late-forming portion of the first pharyngeal pouch, and variably express skeletal defects, matching previously characterized fras1 mutant phenotypes. In itga8 and fras1 mutants, skeletal severity varies markedly between sides, indicating that both mutants have increased developmental instability. Whereas fras1 is expressed in epithelia, we show that itga8 is expressed complementarily in facial mesenchyme. Paired with the observed phenotypic similarity, this expression indicates that the genes function in epithelial-mesenchymal interactions. Similar interactions between Fras1 and Itga8 have previously been found in mouse kidney, where these genes both regulate Nephronectin (Npnt) protein abundance. We find that zebrafish facial tissues express both npnt and the Fraser gene fibrillin2b (fbn2b), but their transcript levels do not depend on fras1 or itga8 function. Using a revertible fras1 allele, we find that the critical window for fras1 function in the craniofacial skeleton is between 1.5 and 3 days post fertilization, which coincides with the onset of fras1-dependent and itga8-dependent morphogenesis. We propose a model wherein Fras1 and Itga8 interact during late pharyngeal pouch morphogenesis to sculpt pharyngeal arches through epithelial-mesenchymal interactions, thereby stabilizing the developing craniofacial skeleton.
Collapse
Affiliation(s)
- Jared Coffin Talbot
- Institute of Neuroscience, 1254 University of Oregon, Eugene, OR 97403, USA; Departments of Molecular Genetics and Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH 43210, USA.
| | - James T Nichols
- Institute of Neuroscience, 1254 University of Oregon, Eugene, OR 97403, USA
| | - Yi-Lin Yan
- Institute of Neuroscience, 1254 University of Oregon, Eugene, OR 97403, USA
| | - Isaac F Leonard
- Institute of Neuroscience, 1254 University of Oregon, Eugene, OR 97403, USA
| | - Ruth A BreMiller
- Institute of Neuroscience, 1254 University of Oregon, Eugene, OR 97403, USA
| | - Sharon L Amacher
- Departments of Molecular Genetics and Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH 43210, USA
| | | | - Charles B Kimmel
- Institute of Neuroscience, 1254 University of Oregon, Eugene, OR 97403, USA.
| |
Collapse
|
32
|
McCarthy N, Sidik A, Bertrand JY, Eberhart JK. An Fgf-Shh signaling hierarchy regulates early specification of the zebrafish skull. Dev Biol 2016; 415:261-277. [PMID: 27060628 PMCID: PMC4967541 DOI: 10.1016/j.ydbio.2016.04.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Revised: 03/30/2016] [Accepted: 04/05/2016] [Indexed: 02/03/2023]
Abstract
The neurocranium generates most of the craniofacial skeleton and consists of prechordal and postchordal regions. Although development of the prechordal is well studied, little is known of the postchordal region. Here we characterize a signaling hierarchy necessary for postchordal neurocranial development involving Fibroblast growth factor (Fgf) signaling for early specification of mesodermally-derived progenitor cells. The expression of hyaluron synthetase 2 (has2) in the cephalic mesoderm requires Fgf signaling and Has2 function, in turn, is required for postchordal neurocranial development. While Hedgehog (Hh)-deficient embryos also lack a postchordal neurocranium, this appears primarily due to a later defect in chondrocyte differentiation. Inhibitor studies demonstrate that postchordal neurocranial development requires early Fgf and later Hh signaling. Collectively, our results provide a mechanistic understanding of early postchordal neurocranial development and demonstrate a hierarchy of signaling between Fgf and Hh in the development of this structure.
Collapse
Affiliation(s)
- Neil McCarthy
- Department of Molecular Biosciences; Institute of Cell and Molecular Biology, Waggoner Center for Alcohol and Alcohol Addiction Research, University of Texas, Austin, TX, United States
| | - Alfire Sidik
- Department of Molecular Biosciences; Institute of Cell and Molecular Biology, Waggoner Center for Alcohol and Alcohol Addiction Research, University of Texas, Austin, TX, United States
| | - Julien Y Bertrand
- Department of Pathology and Immunology, University of Geneva Medical School, Geneva, Switzerland
| | - Johann K Eberhart
- Department of Molecular Biosciences; Institute of Cell and Molecular Biology, Waggoner Center for Alcohol and Alcohol Addiction Research, University of Texas, Austin, TX, United States; Department of Molecular Biosciences; Institute of Neurobiology, University of Texas, Austin, TX, United States.
| |
Collapse
|
33
|
Li L, Shi H, Zhang M, Guo X, Tong F, Zhang W, Zhou J, Wang H, Yang S. Upregulation of metastasis-associated PRL-3 initiates chordoma in zebrafish. Int J Oncol 2016; 48:1541-52. [PMID: 26846972 DOI: 10.3892/ijo.2016.3363] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Accepted: 01/15/2016] [Indexed: 11/06/2022] Open
Abstract
The metastasis-associated phosphatase of regenerating liver-3 (PRL-3) plays multiple roles in progression of various human cancers; however, significance of its role during development has not been addressed. Here we cloned and characterized the expression pattern of zebrafish prl-3 transcript and showed that it is ubiquitiously expressed in the first 24 h of development with both maternal and zygotic expressions. The transcripts become progressively restricted to the notochord, vessels and the intestine by 96 h post-fertilization. Notably, overexpression of zebrafish Prl-3 (zPrl-3) and human PRL-3 induces notochord malformation in zebrafish. This phenotype resembles chordoma and is confirmed by associated misexpression of notochord-specific markers. Clinical significance of the PRL-3 in chordoma is strongly suggested by detection of PRL-3 antigen in clinical chordoma specimens. Collectively, our results uncovered that aberrant overexpression of PRL-3 could initiate chordoma in early development and suggest the use of PRL-3 could be used as a predictor and a therapeutic target for chordoma.
Collapse
Affiliation(s)
- Li Li
- Translational Medicine Centre, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong CPZN 510080, P.R. China
| | - Hongshun Shi
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong CPZN 510080, P.R. China
| | - Mingming Zhang
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong CPZN 510080, P.R. China
| | - Xiaoling Guo
- Translational Medicine Centre, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong CPZN 510080, P.R. China
| | - Fang Tong
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong CPZN 510080, P.R. China
| | - Wenliang Zhang
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong CPZN 510080, P.R. China
| | - Junyi Zhou
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong CPZN 510080, P.R. China
| | - Haihe Wang
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong CPZN 510080, P.R. China
| | - Shulan Yang
- Translational Medicine Centre, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong CPZN 510080, P.R. China
| |
Collapse
|
34
|
Gonsar N, Coughlin A, Clay-Wright JA, Borg BR, Kindt LM, Liang JO. Temporal and spatial requirements for Nodal-induced anterior mesendoderm and mesoderm in anterior neurulation. Genesis 2016; 54:3-18. [PMID: 26528772 DOI: 10.1002/dvg.22908] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Revised: 10/29/2015] [Accepted: 10/30/2015] [Indexed: 01/28/2023]
Abstract
Zebrafish with defective Nodal signaling have a phenotype analogous to the fatal human birth defect anencephaly, which is caused by an open anterior neural tube. Previous work in our laboratory found that anterior open neural tube phenotypes in Nodal signaling mutants were caused by lack of mesendodermal/mesodermal tissues. Defects in these mutants are already apparent at neural plate stage, before the neuroepithelium starts to fold into a tube. Consistent with this, we found that the requirement for Nodal signaling maps to mid-late blastula stages. This timing correlates with the timing of prechordal plate mesendoderm and anterior mesoderm induction, suggesting these tissues act to promote neurulation. To further identify tissues important for neurulation, we took advantage of the variable phenotypes in Nodal signaling-deficient sqt mutant and Lefty1-overexpressing embryos. Statistical analysis indicated a strong, positive correlation between a closed neural tube and presence of several mesendoderm/mesoderm-derived tissues (hatching glands, cephalic paraxial mesoderm, notochord, and head muscles). However, the neural tube was closed in a subset of embryos that lacked any one of these tissues. This suggests that several types of Nodal-induced mesendodermal/mesodermal precursors are competent to promote neurulation.
Collapse
Affiliation(s)
- Ngawang Gonsar
- Department of Biology, University of Minnesota Duluth, Duluth, MN.,Integrated Biosciences Graduate Program, University of Minnesota, Duluth, MN
| | - Alicia Coughlin
- Department of Biology, University of Minnesota Duluth, Duluth, MN.,Integrated Biosciences Graduate Program, University of Minnesota, Duluth, MN
| | | | - Bethanie R Borg
- Department of Biology, University of Minnesota Duluth, Duluth, MN
| | - Lexy M Kindt
- Department of Biology, University of Minnesota Duluth, Duluth, MN.,Integrated Biosciences Graduate Program, University of Minnesota, Duluth, MN
| | - Jennifer O Liang
- Department of Biology, University of Minnesota Duluth, Duluth, MN.,Integrated Biosciences Graduate Program, University of Minnesota, Duluth, MN
| |
Collapse
|
35
|
Row RH, Tsotras SR, Goto H, Martin BL. The zebrafish tailbud contains two independent populations of midline progenitor cells that maintain long-term germ layer plasticity and differentiate in response to local signaling cues. Development 2015; 143:244-54. [PMID: 26674311 DOI: 10.1242/dev.129015] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 12/09/2015] [Indexed: 12/25/2022]
Abstract
Vertebrate body axis formation depends on a population of bipotential neuromesodermal cells along the posterior wall of the tailbud that make a germ layer decision after gastrulation to form spinal cord and mesoderm. Despite exhibiting germ layer plasticity, these cells never give rise to midline tissues of the notochord, floor plate and dorsal endoderm, raising the question of whether midline tissues also arise from basal posterior progenitors after gastrulation. We show in zebrafish that local posterior signals specify germ layer fate in two basal tailbud midline progenitor populations. Wnt signaling induces notochord within a population of notochord/floor plate bipotential cells through negative transcriptional regulation of sox2. Notch signaling, required for hypochord induction during gastrulation, continues to act in the tailbud to specify hypochord from a notochord/hypochord bipotential cell population. Our results lend strong support to a continuous allocation model of midline tissue formation in zebrafish, and provide an embryological basis for zebrafish and mouse bifurcated notochord phenotypes as well as the rare human congenital split notochord syndrome. We demonstrate developmental equivalency between the tailbud progenitor cell populations. Midline progenitors can be transfated from notochord to somite fate after gastrulation by ectopic expression of msgn1, a master regulator of paraxial mesoderm fate, or if transplanted into the bipotential progenitors that normally give rise to somites. Our results indicate that the entire non-epidermal posterior body is derived from discrete, basal tailbud cell populations. These cells remain receptive to extracellular cues after gastrulation and continue to make basic germ layer decisions.
Collapse
Affiliation(s)
- Richard H Row
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794-5215, USA
| | - Steve R Tsotras
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794-5215, USA
| | - Hana Goto
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794-5215, USA
| | - Benjamin L Martin
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794-5215, USA
| |
Collapse
|
36
|
Corallo D, Trapani V, Bonaldo P. The notochord: structure and functions. Cell Mol Life Sci 2015; 72:2989-3008. [PMID: 25833128 PMCID: PMC11114051 DOI: 10.1007/s00018-015-1897-z] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Revised: 03/23/2015] [Accepted: 03/26/2015] [Indexed: 01/08/2023]
Abstract
The notochord is an embryonic midline structure common to all members of the phylum Chordata, providing both mechanical and signaling cues to the developing embryo. In vertebrates, the notochord arises from the dorsal organizer and it is critical for proper vertebrate development. This evolutionary conserved structure located at the developing midline defines the primitive axis of embryos and represents the structural element essential for locomotion. Besides its primary structural function, the notochord is also a source of developmental signals that patterns surrounding tissues. Among the signals secreted by the notochord, Hedgehog proteins play key roles during embryogenesis. The Hedgehog signaling pathway is a central regulator of embryonic development, controlling the patterning and proliferation of a wide variety of organs. In this review, we summarize the current knowledge on notochord structure and functions, with a particular emphasis on the key developmental events that take place in vertebrates. Moreover, we discuss some genetic studies highlighting the phenotypic consequences of impaired notochord development, which enabled to understand the molecular basis of different human congenital defects and diseases.
Collapse
Affiliation(s)
- Diana Corallo
- Department of Molecular Medicine, University of Padova, Viale G. Colombo 3, 35131 Padua, Italy
| | - Valeria Trapani
- Department of Molecular Medicine, University of Padova, Viale G. Colombo 3, 35131 Padua, Italy
| | - Paolo Bonaldo
- Department of Molecular Medicine, University of Padova, Viale G. Colombo 3, 35131 Padua, Italy
| |
Collapse
|
37
|
Ferrari L, Pistocchi A, Libera L, Boari N, Mortini P, Bellipanni G, Giordano A, Cotelli F, Riva P. FAS/FASL are dysregulated in chordoma and their loss-of-function impairs zebrafish notochord formation. Oncotarget 2015; 5:5712-24. [PMID: 25071022 PMCID: PMC4170636 DOI: 10.18632/oncotarget.2145] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Chordoma is a rare malignant tumor that recapitulates the notochord phenotype and is thought to derive from notochord remnants not correctly regressed during development. Apoptosis is necessary for the proper notochord development in vertebrates, and the apoptotic pathway mediated by Fas and Fasl has been demonstrated to be involved in notochord cells regression. This study was conducted to investigate the expression of FAS/FASL pathway in a cohort of skull base chordomas and to analyze the role of fas/fasl homologs in zebrafish notochord formation. FAS/FASL expression was found to be dysregulated in chordoma leading to inactivation of the downstream Caspases in the samples analyzed. Both fas and fasl were specifically expressed in zebrafish notochord sorted cells. fas and fasl loss-of-function mainly resulted in larvae with notochord multi-cell-layer jumps organization, larger vacuolated notochord cells, defects in the peri-notochordal sheath structure and in vertebral mineralization. Interestingly, we observed the persistent expression of ntla and col2a1a, the zebrafish homologs of the human T gene and COL2A1 respectively, which are specifically up-regulated in chordoma. These results demonstrate for the first time the dysregulation of FAS/FASL in chordoma and their role in notochord formation in the zebrafish model, suggesting their possible implication in chordoma onset.
Collapse
Affiliation(s)
- Luca Ferrari
- Dipartimento di Biotecnologie Mediche e Medicina Traslazionale, Università Degli Studi di Milano, Via Viotti 3/5 20133 Milan, Italy; These authors contribute equally in this study
| | - Anna Pistocchi
- Dipartimento di Biotecnologie Mediche e Medicina Traslazionale, Università Degli Studi di Milano, Via Viotti 3/5 20133 Milan, Italy; Dipartimento di Bioscienze, Università Degli Studi di Milano, Via Celoria 26 20133 Milan, Italy; These authors contribute equally in this study
| | - Laura Libera
- Dipartimento di Biotecnologie Mediche e Medicina Traslazionale, Università Degli Studi di Milano, Via Viotti 3/5 20133 Milan, Italy
| | - Nicola Boari
- Dipartimento di Neurochirurgia, Università Vita-Salute IRCCS Ospedale San Raffaele, Via Olgettina 60, 20132 Milan, Italy
| | - Pietro Mortini
- Dipartimento di Neurochirurgia, Università Vita-Salute IRCCS Ospedale San Raffaele, Via Olgettina 60, 20132 Milan, Italy
| | - Gianfranco Bellipanni
- Department of Biology, College of Science and Technology, Temple University, Philadelphia, Pennsylvania 19122, USA; Sbarro Institute for Cancer Research and Molecular Medicine, College of Science and Technology, Temple University, Philadelphia, Pennsylvania 19122, USA
| | - Antonio Giordano
- Department of Biology, College of Science and Technology, Temple University, Philadelphia, Pennsylvania 19122, USA; Sbarro Institute for Cancer Research and Molecular Medicine, College of Science and Technology, Temple University, Philadelphia, Pennsylvania 19122, USA
| | - Franco Cotelli
- Dipartimento di Bioscienze, Università Degli Studi di Milano, Via Celoria 26 20133 Milan, Italy
| | - Paola Riva
- Dipartimento di Biotecnologie Mediche e Medicina Traslazionale, Università Degli Studi di Milano, Via Viotti 3/5 20133 Milan, Italy
| |
Collapse
|
38
|
Moreno-Ayala R, Schnabel D, Salas-Vidal E, Lomelí H. PIAS-like protein Zimp7 is required for the restriction of the zebrafish organizer and mesoderm development. Dev Biol 2015; 403:89-100. [PMID: 25912688 DOI: 10.1016/j.ydbio.2015.04.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Revised: 03/30/2015] [Accepted: 04/03/2015] [Indexed: 12/16/2022]
Abstract
The Zmiz2 (Zimp7) protein and its homolog Zmiz1 (Zimp10) were initially identified in humans as androgen receptor co-activators. Sequence analysis revealed the presence of an SP-RING/Miz domain, which is highly conserved in members of the PIAS family and confers SUMO-conjugating activity. Zimp7 has been shown to interact with components of the Wnt/β-Catenin signaling pathway and with Brg1 and BAF57, components of the ATP-dependent mammalian SWI/SNF-like BAF chromatin-remodeling complexes. In this work, we analyze the role of zygotic Zimp7 in zebrafish development. We describe evidence indicating that Zimp7 is required for mesoderm development and dorsoventral patterning. Morpholino-mediated reduction of zygotic Zimp7 produced axial mesodermal defects that were preceded by up-regulation of organizer genes such as bozozok, goosecoid and floating head at the onset of gastrulation and by down-regulation of the ventral markers vox, vent and eve1 indicating loss of the ventrolateral mesoderm. Consistently, embryos overexpressing zimp7 RNA exhibited midline defects such as loss of forebrain and cyclopia accompanied by transcriptional changes directly opposite of those found in the morphants. In addition, the patterning of ventralized embryos produced by the overexpression of vox and vent was restored by a reduction of Zimp7 activity. Altogether, our findings indicate that Zimp7 is involved in transcriptional regulation of factors that are essential for patterning in the dorsoventral axis.
Collapse
Affiliation(s)
- Roberto Moreno-Ayala
- Departamento de Genética del Desarrollo y Fisiología Molecular Instituto de Biotecnología, Universidad Nacional Autónoma de México, Mexico
| | - Denhí Schnabel
- Departamento de Genética del Desarrollo y Fisiología Molecular Instituto de Biotecnología, Universidad Nacional Autónoma de México, Mexico
| | - Enrique Salas-Vidal
- Departamento de Genética del Desarrollo y Fisiología Molecular Instituto de Biotecnología, Universidad Nacional Autónoma de México, Mexico
| | - Hilda Lomelí
- Departamento de Genética del Desarrollo y Fisiología Molecular Instituto de Biotecnología, Universidad Nacional Autónoma de México, Mexico.
| |
Collapse
|
39
|
Duran I, Ruiz-Sánchez J, Santamaría JA, Marí-Beffa M. Holmgren's principle of delamination during fin skeletogenesis. Mech Dev 2014; 135:16-30. [PMID: 25460362 DOI: 10.1016/j.mod.2014.11.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Revised: 11/12/2014] [Accepted: 11/14/2014] [Indexed: 10/24/2022]
Abstract
During fin morphogenesis, several mesenchyme condensations occur to give rise to the dermal skeleton. Although each of them seems to create distinctive and unique structures, they all follow the premises of the same morphogenetic principle. Holmgren's principle of delamination was first proposed to describe the morphogenesis of skeletal elements of the cranium, but Jarvik extended it to the development of the fin exoskeleton. Since then, some cellular or molecular explanations, such as the "flypaper" model (Thorogood et al.), or the evolutionary description by Moss, have tried to clarify this topic. In this article, we review new data from zebrafish studies to meet these criteria described by Holmgren and other authors. The variety of cell lineages involved in these skeletogenic condensations sheds light on an open discussion of the contributions of mesoderm- versus neural crest-derived cell lineages to the development of the head and trunk skeleton. Moreover, we discuss emerging molecular studies that are disclosing conserved regulatory mechanisms for dermal skeletogenesis and similarities during fin development and regeneration, which may have important implications in the potential use of the zebrafish fin as a model for regenerative medicine.
Collapse
Affiliation(s)
- I Duran
- Laboratory of Bioengineering and Tissue Regeneration (LABRET), Department of Cell Biology, Genetics and Physiology, Biomedical Research Institute of Málaga (IBIMA), Faculty of Sciences, University of Málaga, 29071 Málaga, Spain; Department of Orthopedic Surgery, University of California, Los Angeles, CA 90095, USA; Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 29071 Málaga, Spain.
| | - J Ruiz-Sánchez
- Laboratory of Bioengineering and Tissue Regeneration (LABRET), Department of Cell Biology, Genetics and Physiology, Biomedical Research Institute of Málaga (IBIMA), Faculty of Sciences, University of Málaga, 29071 Málaga, Spain; Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 29071 Málaga, Spain
| | - J A Santamaría
- Laboratory of Bioengineering and Tissue Regeneration (LABRET), Department of Cell Biology, Genetics and Physiology, Biomedical Research Institute of Málaga (IBIMA), Faculty of Sciences, University of Málaga, 29071 Málaga, Spain; Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 29071 Málaga, Spain
| | - M Marí-Beffa
- Laboratory of Bioengineering and Tissue Regeneration (LABRET), Department of Cell Biology, Genetics and Physiology, Biomedical Research Institute of Málaga (IBIMA), Faculty of Sciences, University of Málaga, 29071 Málaga, Spain; Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 29071 Málaga, Spain.
| |
Collapse
|
40
|
Le Pabic P, Ng C, Schilling TF. Fat-Dachsous signaling coordinates cartilage differentiation and polarity during craniofacial development. PLoS Genet 2014; 10:e1004726. [PMID: 25340762 PMCID: PMC4207671 DOI: 10.1371/journal.pgen.1004726] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Accepted: 09/02/2014] [Indexed: 11/19/2022] Open
Abstract
Organogenesis requires coordinated regulation of cellular differentiation and morphogenesis. Cartilage cells in the vertebrate skeleton form polarized stacks, which drive the elongation and shaping of skeletal primordia. Here we show that an atypical cadherin, Fat3, and its partner Dachsous-2 (Dchs2), control polarized cell-cell intercalation of cartilage precursors during craniofacial development. In zebrafish embryos deficient in Fat3 or Dchs2, chondrocytes fail to stack and misregulate expression of sox9a. Similar morphogenetic defects occur in rerea/atr2a−/− mutants, and Fat3 binds REREa, consistent with a model in which Fat3, Dchs2 and REREa interact to control polarized cell-cell intercalation and simultaneously control differentiation through Sox9. Chimaeric analyses support such a model, and reveal long-range influences of all three factors, consistent with the activation of a secondary signal that regulates polarized cell-cell intercalation. This coordinates the spatial and temporal morphogenesis of chondrocytes to shape skeletal primordia and defects in these processes underlie human skeletal malformations. Similar links between cell polarity and differentiation mechanisms are also likely to control organ formation in other contexts. Little is known about the mechanisms of cell-cell communication necessary to assemble skeletal elements of appropriate size and shape. In this study, we investigate the roles of genetic factors belonging to a developmental pathway that affects skeletal progenitor behavior: the atypical cadherins Fat3 and Dachsous2 (Dchs2), and REREa/Atr2a. We show that cartilage precursors fail to rearrange into linear stacks and at the same time misregulate expression of sox9a, a key regulator of cartilage differentiation, in zebrafish embryos deficient in Fat3 or its partner Dchs2. Similar cartilage defects are observed in rerea−/− mutants, and Fat3 interacts physically and genetically with REREa. Our results suggest that Fat3, Dchs2 and REREa interact to control polarized cell-cell intercalation and simultaneously control skeletal differentiation through Sox9. By transplanting cartilage precursors between wild-type and Fat3, Dchs2 or REREa deficient embryos we demonstrate that all three factors exert long-range influences on neighboring cells, most likely mediated by another polarizing signal. We propose a model in which this coordinates the polarity and differentiation of chondrocytes to shape skeletal primordia, and that defects in these processes underlie human skeletal malformations.
Collapse
Affiliation(s)
- Pierre Le Pabic
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, California, United States of America
| | - Carrie Ng
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, California, United States of America
| | - Thomas F. Schilling
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, California, United States of America
- * E-mail:
| |
Collapse
|
41
|
Hemichordate neurulation and the origin of the neural tube. Nat Commun 2014; 4:2713. [PMID: 24177053 DOI: 10.1038/ncomms3713] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Accepted: 10/04/2013] [Indexed: 11/08/2022] Open
Abstract
The origin of the body plan of our own phylum, Chordata, is one of the most fascinating questions in evolutionary biology. Yet, after more than a century of debate, the evolutionary origins of the neural tube and notochord remain unclear. Here we examine the development of the collar nerve cord in the hemichordate Balanoglossus simodensis and find shared gene expression patterns between hemichordate and chordate neurulation. Moreover, we show that the dorsal endoderm of the buccal tube and the stomochord expresses Hedgehog RNA, and it seems likely that collar cord cells can receive the signal. Our data suggest that the endoderm functions as an organizer to pattern the overlying collar cord, similar to the relationship between the notochord and neural tube in chordates. We propose that the origin of the core genetic mechanisms for the development of the notochord and the neural tube date back to the last common deuterostome ancestor.
Collapse
|
42
|
Abstract
Despite the importance of tendons and ligaments for transmitting movement and providing stability to the musculoskeletal system, their development is considerably less well understood than that of the tissues they serve to connect. Zebrafish have been widely used to address questions in muscle and skeletal development, yet few studies describe their tendon and ligament tissues. We have analyzed in zebrafish the expression of several genes known to be enriched in mammalian tendons and ligaments, including scleraxis (scx), collagen 1a2 (col1a2) and tenomodulin (tnmd), or in the tendon-like myosepta of the zebrafish (xirp2a). Co-expression studies with muscle and cartilage markers demonstrate the presence of scxa, col1a2 and tnmd at sites between the developing muscle and cartilage, and xirp2a at the myotendinous junctions. We determined that the zebrafish craniofacial tendon and ligament progenitors are neural crest derived, as in mammals. Cranial and fin tendon progenitors can be induced in the absence of differentiated muscle or cartilage, although neighboring muscle and cartilage are required for tendon cell maintenance and organization, respectively. By contrast, myoseptal scxa expression requires muscle for its initiation. Together, these data suggest a conserved role for muscle in tendon development. Based on the similarities in gene expression, morphology, collagen ultrastructural arrangement and developmental regulation with that of mammalian tendons, we conclude that the zebrafish tendon populations are homologous to their force-transmitting counterparts in higher vertebrates. Within this context, the zebrafish model can be used to provide new avenues for studying tendon biology in a vertebrate genetic system.
Collapse
Affiliation(s)
- Jessica W Chen
- Center for Regenerative Medicine, Harvard Stem Cell Institute, Department of Orthopaedic Surgery, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA
| | | |
Collapse
|
43
|
Wang S, Furmanek T, Kryvi H, Krossøy C, Totland GK, Grotmol S, Wargelius A. Transcriptome sequencing of Atlantic salmon (Salmo salar L.) notochord prior to development of the vertebrae provides clues to regulation of positional fate, chordoblast lineage and mineralisation. BMC Genomics 2014; 15:141. [PMID: 24548379 PMCID: PMC3943441 DOI: 10.1186/1471-2164-15-141] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2013] [Accepted: 02/13/2014] [Indexed: 11/26/2022] Open
Abstract
Background In teleosts such as Atlantic salmon (Salmo salar L.), segmentation and subsequent mineralisation of the notochord during embryonic stages are essential for normal vertebrae formation. However, the molecular mechanisms leading to segmentation and mineralisation of the notochord are poorly understood. The aim of this study was to identify genes/pathways acting in gradients over time and along the anterior-posterior axis during notochord segmentation and immediately prior to mineralisation of the vertebral bodies in Atlantic salmon. Results Notochord samples were collected from unsegmented, pre-segmented and segmented developmental stages. In each stage, the cellular core of the notochord was cut into three pieces along the longitudinal axis (anterior, mid, posterior). RNA was sequenced (22 million pair-end 100 bp/ library) and mapped to the salmon genome. 66569 transcripts were predicted and 55775 were annotated. In order to identify possible gradients leading to segmentation of the notochord, all 71 notochord-expressed hox genes were investigated, most of them displaying a typical anterior-posterior expression pattern along the notochord axis. The clustering of hox genes revealed a pattern that could be related to notochord segmentation. We further investigated how mineralisation is initiated in the notochord, and several factors related to chondrogenic lineage were identified (sox9, sox5, sox6, tgfb3, ihhb and col2a1), suggesting a cartilage-like character of the notochord. KEGG analysis of differentially expressed genes between stages revealed down-regulation of pathways associated with ECM, cell division, metabolism and development at onset of notochord segmentation. This implies that inhibitory signals produce segmentation of the notochord. One such potential inhibitory signal was identified, col11a2, which was detected in segments of non-mineralising notochord. Conclusions An incomplete salmon genome was successfully used to analyse RNA-seq data from the cellular core of the Atlantic salmon notochord. In transcriptome we found; hox gene patterns possibly linked to segmentation; down-regulation of pathways in the notochord at onset of segmentation; segmented expression of col11a2 in non-mineralised segments of the notochord; and a chondroblast-like footprint in the notochord.
Collapse
|
44
|
Neacsu CD, Ko YP, Tagariello A, Røkenes Karlsen K, Neiss WF, Paulsson M, Wagener R. Matrilin-1 is essential for zebrafish development by facilitating collagen II secretion. J Biol Chem 2013; 289:1505-18. [PMID: 24293366 DOI: 10.1074/jbc.m113.529933] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Matrilin-1 is the prototypical member of the matrilin protein family and is highly expressed in cartilage. However, gene targeting of matrilin-1 in mouse did not lead to pronounced phenotypes. Here we used the zebrafish as an alternative model to study matrilin function in vivo. Matrilin-1 displays a multiphasic expression during zebrafish development. In an early phase, with peak expression at about 15 h post-fertilization, matrilin-1 is present throughout the zebrafish embryo with exception of the notochord. Later, when the skeleton develops, matrilin-1 is expressed mainly in cartilage. Morpholino knockdown of matrilin-1 results both in overall growth defects and in disturbances in the formation of the craniofacial cartilage, most prominently loss of collagen II deposition. In fish with mild phenotypes, certain cartilage extracellular matrix components were present, but the tissue did not show features characteristic for cartilage. The cells showed endoplasmic reticulum aberrations but no activation of XBP-1, a marker for endoplasmic reticulum stress. In severe phenotypes nearly all chondrocytes died. During the early expression phase the matrilin-1 knockdown had no effects on cell morphology, but increased cell death was observed. In addition, the broad deposition of collagen II was largely abolished. Interestingly, the early phenotype could be rescued by the co-injection of mRNA coding for the von Willebrand factor C domain of collagen IIα1a, indicating that the functional loss of this domain occurs as a consequence of matrilin-1 deficiency. The results show that matrilin-1 is indispensible for zebrafish cartilage formation and plays a role in the early collagen II-dependent developmental events.
Collapse
|
45
|
Corallo D, Schiavinato A, Trapani V, Moro E, Argenton F, Bonaldo P. Emilin3 is required for notochord sheath integrity and interacts with Scube2 to regulate notochord-derived Hedgehog signals. Development 2013; 140:4594-601. [PMID: 24131633 DOI: 10.1242/dev.094078] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The notochord is a transient and essential structure that provides both mechanical and signaling cues to the developing vertebrate embryo. In teleosts, the notochord is composed of a core of large vacuolated cells and an outer layer of cells that secrete the notochord sheath. In this work, we have identified the extracellular matrix glycoprotein Emilin3 as a novel essential component of the zebrafish notochord sheath. The development of the notochord sheath is impaired in Emilin3 knockdown embryos. The patterning activity of the notochord is also affected by Emilin3, as revealed by the increase of Hedgehog (Hh) signaling in Emilin3-depleted embryos and the decreased Hh signaling in embryos overexpressing Emilin3 in the notochord. In vitro and in vivo experiments indicate that Emilin3 modulates the availability of Hh ligands by interacting with the permissive factor Scube2 in the notochord sheath. Overall, this study reveals a new role for an EMILIN protein and reinforces the concept that structure and function of the notochord are strictly linked.
Collapse
Affiliation(s)
- Diana Corallo
- Department of Biomedical Sciences, University of Padova, I-35121 Padova, Italy
| | | | | | | | | | | |
Collapse
|
46
|
Tsukamoto H, Yokoyama Y, Suzuki T, Mizuta S, Yoshinaka R, Akahane Y. Cloning and expression of cDNA encoding lysyl hydroxylase 1, 2 and 3 in tiger puffer Takifugu rubripes. Comp Biochem Physiol B Biochem Mol Biol 2013; 166:123-32. [DOI: 10.1016/j.cbpb.2013.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Revised: 08/05/2013] [Accepted: 08/06/2013] [Indexed: 11/26/2022]
|
47
|
The polycomb group protein ring1b/rnf2 is specifically required for craniofacial development. PLoS One 2013; 8:e73997. [PMID: 24040141 PMCID: PMC3770662 DOI: 10.1371/journal.pone.0073997] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Accepted: 07/29/2013] [Indexed: 01/24/2023] Open
Abstract
Polycomb group (PcG) genes are chromatin modifiers that mediate epigenetic silencing of target genes. PcG-mediated epigenetic silencing is implicated in embryonic development, stem cell plasticity, cell fate maintenance, cellular differentiation and cancer. However, analysis of the roles of PcG proteins in maintaining differentiation programs during vertebrate embryogenesis has been hampered due to the early embryonic lethality of several PcG knock-outs in the mouse. Here, we show that zebrafish Ring1b/Rnf2, the single E3 ubiquitin ligase in the Polycomb Repressive Complex 1, critically regulates the developmental program of craniofacial cell lineages. Zebrafish ring1b mutants display a severe craniofacial phenotype, which includes an almost complete absence of all cranial cartilage, bone and musculature. We show that Cranial Neural Crest (CNC)-derived cartilage precursors migrate correctly into the pharyngeal arches, but fail to differentiate into chondrocytes. This phenotype is specific for cartilage precursors, since other neural crest-derived cell lineages, including glia, neurons and chromatophores, are formed normally in ring1b mutants. Our results therefore reveal a critical and specific role for Ring1b in promoting the differentiation of cranial neural crest cells into chondrocytes. The molecular mechanisms underlying the pathogenesis of craniofacial abnormalities, which are among the most common genetic birth defects in humans, remain poorly understood. The zebrafish ring1b mutant provides a molecular model for investigating these mechanisms and may lead to the discovery of new treatments or preventions of craniofacial abnormalities.
Collapse
|
48
|
Lien HW, Yang CH, Cheng CH, Hung CC, Liao WH, Hwang PP, Han YS, Huang CJ. A novel zinc finger protein 219-like (ZNF219L) is involved in the regulation of collagen type 2 alpha 1a (col2a1a) gene expression in zebrafish notochord. Int J Biol Sci 2013; 9:872-86. [PMID: 24155663 PMCID: PMC3805895 DOI: 10.7150/ijbs.7126] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Accepted: 08/24/2013] [Indexed: 12/01/2022] Open
Abstract
The notochord is required for body plan patterning in vertebrates, and defects in notochord development during embryogenesis can lead to diseases affecting the adult. It is therefore important to elucidate the gene regulatory mechanism underlying notochord formation. In this study, we cloned the zebrafish zinc finger 219-like (ZNF219L) based on mammalian ZNF219, which contains nine C2H2-type zinc finger domains. Through whole-mount in situ hybridization, we found that znf219L mRNA is mainly expressed in the zebrafish midbrain-hindbrain boundary, hindbrain, and notochord during development. The znf219L morpholino knockdown caused partial abnormal notochord phenotype and reduced expression of endogenous col2a1a in the notochord specifically. In addition, ZNF219L could recognize binding sites with GGGGG motifs and trigger augmented activity of the col2a1a promoter in a luciferase assay. Furthermore, in vitro binding experiments revealed that ZNF219L recognizes the GGGGG motifs in the promoter region of the zebrafish col2a1a gene through its sixth and ninth zinc finger domains. Taken together, our results reveal that ZNF219L is involved in regulating the expression of col2a1a in zebrafish notochord specifically.
Collapse
Affiliation(s)
- Huang-Wei Lien
- 1. Institute of Fisheries Sciences, National Taiwan University, Taipei 106, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Ding V, Lew QJ, Chu KL, Natarajan S, Rajasegaran V, Gurumurthy M, Choo ABH, Chao SH. HEXIM1 induces differentiation of human pluripotent stem cells. PLoS One 2013; 8:e72823. [PMID: 23977357 PMCID: PMC3748041 DOI: 10.1371/journal.pone.0072823] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Accepted: 07/19/2013] [Indexed: 02/07/2023] Open
Abstract
Hexamethylene bisacetamide inducible protein 1 (HEXIM1) is best known as the inhibitor of positive transcription elongation factor b (P-TEFb), which is composed of cyclin-dependent kinase 9 (CDK9)/cyclin T1. P-TEFb is an essential regulator for the transcriptional elongation by RNA polymerase II. A genome-wide study using human embryonic stem cells shows that most mRNA synthesis is regulated at the stage of transcription elongation, suggesting a possible role for P-TEFb/HEXIM1 in the gene regulation of stem cells. In this report, we detected a marked increase in HEXIM1 protein levels in the differentiated human pluripotent stem cells (hPSCs) induced by LY294002 treatment. Since no changes in CDK9 and cyclin T1 were observed in the LY294002-treated cells, increased levels of HEXIM1 might lead to inhibition of P-TEFb activity. However, treatment with a potent P-TEFb inhibiting compound, flavopiridol, failed to induce hPSC differentiation, ruling out the possible requirement for P-TEFb kinase activity in hPSC differentiation. Conversely, differentiation was observed when hPSCs were incubated with hexamethylene bisacetamide, a HEXIM1 inducing reagent. The involvement of HEXIM1 in the regulation of hPSCs was further supported when overexpression of HEXIM1 concomitantly induced hPSC differentiation. Collectively, our study demonstrates a novel role of HEXIM1 in regulating hPSC fate through a P-TEFb-independent pathway.
Collapse
Affiliation(s)
- Vanessa Ding
- Stem Cell Group, Bioprocessing Technology Institute, A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
| | - Qiao Jing Lew
- Expression Engineering Group, Bioprocessing Technology Institute, A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
| | - Kai Ling Chu
- Expression Engineering Group, Bioprocessing Technology Institute, A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
| | - Subaashini Natarajan
- Stem Cell Group, Bioprocessing Technology Institute, A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
| | - Vikneswari Rajasegaran
- Expression Engineering Group, Bioprocessing Technology Institute, A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
| | - Meera Gurumurthy
- Expression Engineering Group, Bioprocessing Technology Institute, A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
| | - Andre B. H. Choo
- Stem Cell Group, Bioprocessing Technology Institute, A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
- Department of Bioengineering, Faculty of Engineering, National University of Singapore, Singapore, Singapore
| | - Sheng-Hao Chao
- Expression Engineering Group, Bioprocessing Technology Institute, A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
- Department of Microbiology, National University of Singapore, Singapore, Singapore
- * E-mail:
| |
Collapse
|
50
|
Teslaa JJ, Keller AN, Nyholm MK, Grinblat Y. Zebrafish Zic2a and Zic2b regulate neural crest and craniofacial development. Dev Biol 2013; 380:73-86. [PMID: 23665173 DOI: 10.1016/j.ydbio.2013.04.033] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2012] [Revised: 04/25/2013] [Accepted: 04/29/2013] [Indexed: 11/25/2022]
Abstract
Holoprosencephaly (HPE), the most common malformation of the human forebrain, is associated with defects of the craniofacial skeleton. ZIC2, a zinc-finger transcription factor, is strongly linked to HPE and to a characteristic set of dysmorphic facial features in humans. We have previously identified important functions for zebrafish Zic2 in the developing forebrain. Here, we demonstrate that ZIC2 orthologs zic2a and zic2b also regulate the forming zebrafish craniofacial skeleton, including the jaw and neurocranial cartilages, and use the zebrafish to study Zic2-regulated processes that may contribute to the complex etiology of HPE. Using temporally controlled Zic2a overexpression, we show that the developing craniofacial cartilages are sensitive to Zic2 elevation prior to 24hpf. This window of sensitivity overlaps the critical expansion and migration of the neural crest (NC) cells, which migrate from the developing neural tube to populate vertebrate craniofacial structures. We demonstrate that zic2b influences the induction of NC at the neural plate border, while both zic2a and zic2b regulate NC migratory onset and strongly contribute to chromatophore development. Both Zic2 depletion and early ectopic Zic2 expression cause moderate, incompletely penetrant mispatterning of the NC-derived jaw precursors at 24hpf, yet by 2dpf these changes in Zic2 expression result in profoundly mispatterned chondrogenic condensations. We attribute this discrepancy to an additional role for Zic2a and Zic2b in patterning the forebrain primordium, an important signaling source during craniofacial development. This hypothesis is supported by evidence that transplanted Zic2-deficient cells can contribute to craniofacial cartilages in a wild-type background. Collectively, these data suggest that zebrafish Zic2 plays a dual role during craniofacial development, contributing to two disparate aspects of craniofacial morphogenesis: (1) neural crest induction and migration, and (2) early patterning of tissues adjacent to craniofacial chondrogenic condensations.
Collapse
Affiliation(s)
- Jessica J Teslaa
- Department of Zoology, University of Wisconsin, Madison, WI 53706, USA
| | | | | | | |
Collapse
|