1
|
Ford C, de Sena-Tomás C, Wun TTR, Aleman AG, Rangaswamy U, Leyhr J, Nuñez MI, Gao CZ, Nim HT, See M, Coppola U, Waxman JS, Ramialison M, Haitina T, Smeeton J, Sanges R, Targoff KL. Nkx2.7 is a conserved regulator of craniofacial development. Nat Commun 2025; 16:3802. [PMID: 40268889 PMCID: PMC12019251 DOI: 10.1038/s41467-025-58821-3] [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: 12/01/2023] [Accepted: 04/02/2025] [Indexed: 04/25/2025] Open
Abstract
Craniofacial malformations arise from developmental defects in the head, face, and neck with phenotypes such as 22q11.2 deletion syndrome illustrating a developmental link between cardiovascular and craniofacial morphogenesis. NKX2-5 is a key cardiac transcription factor associated with congenital heart disease and mouse models of Nkx2-5 deficiency highlight roles in cardiac development. In zebrafish, nkx2.5 and nkx2.7 are paralogues in the NK4 family expressed in cardiomyocytes and pharyngeal arches. Despite shared cellular origins of cardiac and craniofacial tissues, the function of NK4 factors in head and neck patterning has not been elucidated. Molecular evolutionary analysis of NK4 genes shows that nkx2.5 and nkx2.7 are ohnologs resulting from whole genome duplication events. Nkx2.7 serves as a previously unappreciated regulator of branchiomeric muscle and cartilage formation for which nkx2.5 cannot fully compensate. Mechanistically, our results highlight that Nkx2.7 patterns the cranial neural crest and functions upstream of Endothelin1 to inhibit Notch signals. Together, our studies shed light on an evolutionarily conserved Nkx transcription factor with unique functions in vertebrate craniofacial development, advancing our understanding of congenital head and neck deformities.
Collapse
Affiliation(s)
- Caitlin Ford
- Department of Genetics & Development, College of Physicians & Surgeons, Columbia University, New York, NY, 10032, USA
- Division of Cardiology, Department of Pediatrics, College of Physicians & Surgeons, Columbia University, New York, NY, 10032, USA
- Columbia Stem Cell Initiative, Columbia University, New York, NY, 10032, USA
| | - Carmen de Sena-Tomás
- Division of Cardiology, Department of Pediatrics, College of Physicians & Surgeons, Columbia University, New York, NY, 10032, USA
- Columbia Stem Cell Initiative, Columbia University, New York, NY, 10032, USA
- Lipid Trafficking and Disease Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer, 08036, Barcelona, Spain
- Department of Genetics, Microbiology and Statistics, University of Barcelona, 08028, Barcelona, Spain
| | - Tint Tha Ra Wun
- Division of Cardiology, Department of Pediatrics, College of Physicians & Surgeons, Columbia University, New York, NY, 10032, USA
- Columbia Stem Cell Initiative, Columbia University, New York, NY, 10032, USA
| | - Angelika G Aleman
- Division of Cardiology, Department of Pediatrics, College of Physicians & Surgeons, Columbia University, New York, NY, 10032, USA
- Columbia Stem Cell Initiative, Columbia University, New York, NY, 10032, USA
- Department of Physiology & Cellular Biophysics, College of Physicians & Surgeons, Columbia University, New York, NY, 10032, USA
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Uday Rangaswamy
- Functional and Structural Genomics, Scuola Internazionale Superiore di Studi Avanzati (SISSA), Via Bonomea 265, 34136, Trieste, Italy
| | - Jake Leyhr
- Department of Organismal Biology, Uppsala University, 75236, Uppsala, Sweden
| | - María I Nuñez
- Division of Cardiology, Department of Pediatrics, College of Physicians & Surgeons, Columbia University, New York, NY, 10032, USA
- Columbia Stem Cell Initiative, Columbia University, New York, NY, 10032, USA
| | - Cynthia Zehui Gao
- Division of Cardiology, Department of Pediatrics, College of Physicians & Surgeons, Columbia University, New York, NY, 10032, USA
- Columbia Stem Cell Initiative, Columbia University, New York, NY, 10032, USA
- Department of Computer Science, Columbia University, New York, NY, 10027, USA
| | - Hieu T Nim
- The Novo Nordisk Foundation Center for Stem Cell Medicine & Stem Cell Biology, Murdoch Children's Research Institute, Parkville, VIC, 3052, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, VIC, 3052, Australia
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, 3800, Australia
- Stem Cell Medicine, Murdoch Children's Research Institute, Parkville, VIC, 3052, Australia
| | - Michael See
- The Novo Nordisk Foundation Center for Stem Cell Medicine & Stem Cell Biology, Murdoch Children's Research Institute, Parkville, VIC, 3052, Australia
- Stem Cell Medicine, Murdoch Children's Research Institute, Parkville, VIC, 3052, Australia
| | - Ugo Coppola
- Molecular Cardiovascular Biology Division and Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Biological Sciences, Florida Gulf Coast University, Fort Myers, FL, 33965, USA
| | - Joshua S Waxman
- Molecular Cardiovascular Biology Division and Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Mirana Ramialison
- The Novo Nordisk Foundation Center for Stem Cell Medicine & Stem Cell Biology, Murdoch Children's Research Institute, Parkville, VIC, 3052, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, VIC, 3052, Australia
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, 3800, Australia
- Stem Cell Medicine, Murdoch Children's Research Institute, Parkville, VIC, 3052, Australia
| | - Tatjana Haitina
- Department of Organismal Biology, Uppsala University, 75236, Uppsala, Sweden
| | - Joanna Smeeton
- Department of Genetics & Development, College of Physicians & Surgeons, Columbia University, New York, NY, 10032, USA
- Columbia Stem Cell Initiative, Columbia University, New York, NY, 10032, USA
- Department of Rehabilitation and Regenerative Medicine, College of Physicians & Surgeons, Columbia University, New York, NY, 10032, USA
| | - Remo Sanges
- Functional and Structural Genomics, Scuola Internazionale Superiore di Studi Avanzati (SISSA), Via Bonomea 265, 34136, Trieste, Italy
- Central RNA Laboratory, Istituto Italiano di Tecnologia (IIT), Via Enrico Melen 83, 16152, Genova, Italy
| | - Kimara L Targoff
- Division of Cardiology, Department of Pediatrics, College of Physicians & Surgeons, Columbia University, New York, NY, 10032, USA.
- Columbia Stem Cell Initiative, Columbia University, New York, NY, 10032, USA.
| |
Collapse
|
2
|
Carroll SH, Schafer S, Kawasaki K, Tsimbal C, Jule AM, Hallett SA, Li E, Liao EC. Genetic requirement of dact1/2 to regulate noncanonical Wnt signaling and calpain 8 during embryonic convergent extension and craniofacial morphogenesis. eLife 2024; 13:RP91648. [PMID: 39570288 PMCID: PMC11581427 DOI: 10.7554/elife.91648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2024] Open
Abstract
Wnt signaling plays crucial roles in embryonic patterning including the regulation of convergent extension (CE) during gastrulation, the establishment of the dorsal axis, and later, craniofacial morphogenesis. Further, Wnt signaling is a crucial regulator of craniofacial morphogenesis. The adapter proteins Dact1 and Dact2 modulate the Wnt signaling pathway through binding to Disheveled. However, the distinct relative functions of Dact1 and Dact2 during embryogenesis remain unclear. We found that dact1 and dact2 genes have dynamic spatiotemporal expression domains that are reciprocal to one another suggesting distinct functions during zebrafish embryogenesis. Both dact1 and dact2 contribute to axis extension, with compound mutants exhibiting a similar CE defect and craniofacial phenotype to the wnt11f2 mutant. Utilizing single-cell RNAseq and an established noncanonical Wnt pathway mutant with a shortened axis (gpc4), we identified dact1/2-specific roles during early development. Comparative whole transcriptome analysis between wildtype and gpc4 and wildtype and dact1/2 compound mutants revealed a novel role for dact1/2 in regulating the mRNA expression of the classical calpain capn8. Overexpression of capn8 phenocopies dact1/2 craniofacial dysmorphology. These results identify a previously unappreciated role of capn8 and calcium-dependent proteolysis during embryogenesis. Taken together, our findings highlight the distinct and overlapping roles of dact1 and dact2 in embryonic craniofacial development, providing new insights into the multifaceted regulation of Wnt signaling.
Collapse
Affiliation(s)
- Shannon H Carroll
- Center for Craniofacial Innovation, Children’s Hospital of Philadelphia Research, Institute, Children’s Hospital of PhiladelphiaPhiladelphiaUnited States
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Children’s Hospital of PhiladelphiaPhiladelphiaUnited States
- Shriners Hospital for ChildrenTampaUnited States
| | - Sogand Schafer
- Center for Craniofacial Innovation, Children’s Hospital of Philadelphia Research, Institute, Children’s Hospital of PhiladelphiaPhiladelphiaUnited States
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Children’s Hospital of PhiladelphiaPhiladelphiaUnited States
| | - Kenta Kawasaki
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Children’s Hospital of PhiladelphiaPhiladelphiaUnited States
- Shriners Hospital for ChildrenTampaUnited States
| | - Casey Tsimbal
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Children’s Hospital of PhiladelphiaPhiladelphiaUnited States
- Shriners Hospital for ChildrenTampaUnited States
| | - Amelie M Jule
- Department of Biostatistics, Harvard T.H. Chan School of Public HealthBostonUnited States
| | - Shawn A Hallett
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Children’s Hospital of PhiladelphiaPhiladelphiaUnited States
- Shriners Hospital for ChildrenTampaUnited States
| | - Edward Li
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Children’s Hospital of PhiladelphiaPhiladelphiaUnited States
| | - Eric C Liao
- Center for Craniofacial Innovation, Children’s Hospital of Philadelphia Research, Institute, Children’s Hospital of PhiladelphiaPhiladelphiaUnited States
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Children’s Hospital of PhiladelphiaPhiladelphiaUnited States
- Shriners Hospital for ChildrenTampaUnited States
| |
Collapse
|
3
|
Carroll SH, Schafer S, Kawasaki K, Tsimbal C, Julé AM, Hallett SA, Li E, Liao EC. Genetic requirement of dact1/2 to regulate noncanonical Wnt signaling and calpain 8 during embryonic convergent extension and craniofacial morphogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.11.07.566024. [PMID: 37986847 PMCID: PMC10659360 DOI: 10.1101/2023.11.07.566024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Wnt signaling plays crucial roles in embryonic patterning including the regulation of convergent extension during gastrulation, the establishment of the dorsal axis, and later, craniofacial morphogenesis. Further, Wnt signaling is a crucial regulator of craniofacial morphogenesis. The adapter proteins Dact1 and Dact2 modulate the Wnt signaling pathway through binding to Disheveled. However, the distinct relative functions of Dact1 and Dact2 during embryogenesis remain unclear. We found that dact1 and dact2 genes have dynamic spatiotemporal expression domains that are reciprocal to one another suggesting distinct functions during zebrafish embryogenesis. Both dact1 and dact2 contribute to axis extension, with compound mutants exhibiting a similar convergent extension defect and craniofacial phenotype to the wnt11f2 mutant. Utilizing single-cell RNAseq and an established noncanonical Wnt pathway mutant with a shortened axis (gpc4), we identified dact1/2 specific roles during early development. Comparative whole transcriptome analysis between wildtype and gpc4 and wildtype and dact1/2 compound mutants revealed a novel role for dact1/2 in regulating the mRNA expression of the classical calpain capn8. Over-expression of capn8 phenocopies dact1/2 craniofacial dysmorphology. These results identify a previously unappreciated role of capn8 and calcium-dependent proteolysis during embryogenesis. Taken together, our findings highlight the distinct and overlapping roles of dact1 and dact2 in embryonic craniofacial development, providing new insights into the multifaceted regulation of Wnt signaling.
Collapse
Affiliation(s)
- Shannon H Carroll
- Center for Craniofacial Innovation, Children's Hospital of Philadelphia Research Institute, Children's Hospital of Philadelphia, PA 19104, USA
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Children's Hospital of Philadelphia, PA 19104, USA
- Shriners Hospital for Children, Tampa, FL 33607, USA
| | - Sogand Schafer
- Center for Craniofacial Innovation, Children's Hospital of Philadelphia Research Institute, Children's Hospital of Philadelphia, PA 19104, USA
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Children's Hospital of Philadelphia, PA 19104, USA
| | - Kenta Kawasaki
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Children's Hospital of Philadelphia, PA 19104, USA
- Shriners Hospital for Children, Tampa, FL 33607, USA
| | - Casey Tsimbal
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Children's Hospital of Philadelphia, PA 19104, USA
- Shriners Hospital for Children, Tampa, FL 33607, USA
| | - Amélie M Julé
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Shawn A Hallett
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Children's Hospital of Philadelphia, PA 19104, USA
- Shriners Hospital for Children, Tampa, FL 33607, USA
| | - Edward Li
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Children's Hospital of Philadelphia, PA 19104, USA
| | - Eric C Liao
- Center for Craniofacial Innovation, Children's Hospital of Philadelphia Research Institute, Children's Hospital of Philadelphia, PA 19104, USA
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Children's Hospital of Philadelphia, PA 19104, USA
- Shriners Hospital for Children, Tampa, FL 33607, USA
| |
Collapse
|
4
|
Incardona JP, Linbo TL, Cameron JR, Scholz NL. Structure-activity relationships for alkyl-phenanthrenes support two independent but interacting synergistic models for PAC mixture potency. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 918:170544. [PMID: 38309367 DOI: 10.1016/j.scitotenv.2024.170544] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 01/19/2024] [Accepted: 01/27/2024] [Indexed: 02/05/2024]
Abstract
Multiple lines of evidence at whole animal, cellular and molecular levels implicate polycyclic aromatic compounds (PACs) with three rings as drivers of crude oil toxicity to developing fish. Phenanthrene (P0) and its alkylated homologs (C1- through C4-phenanthrenes) comprise the most prominent subfraction of tricyclic PACs in crude oils. Among this family, P0 has been studied intensively, with more limited detail available for the C4-phenanthrene 1-methyl-7-isopropyl-phenanthrene (1-M,7-IP, or retene). While both compounds are cardiotoxic, P0 impacts embryonic cardiac function and development through direct blockade of K+ and Ca2+ currents that regulate cardiomyocyte contractions. In contrast, 1-M,7-IP dysregulates aryl hydrocarbon receptor (AHR) activation in developing ventricular cardiomyocytes. Although no other compounds have been assessed in detail across the larger family of alkylated phenanthrenes, increasing alkylation might be expected to shift phenanthrene family member activity from K+/Ca2+ ion current blockade to AHR activation. Using embryos of two distantly related fish species, zebrafish and Atlantic haddock, we tested 14 alkyl-phenanthrenes in both acute and latent developmental cardiotoxicity assays. All compounds were cardiotoxic, and effects were resolved into impacts on multiple, highly specific aspects of heart development or function. Craniofacial defects were clearly linked to developmental cardiotoxicity. Based on these findings, we suggest a novel framework to delineate the developmental toxicity of petrogenic PAC mixtures in fish, which incorporates multi-mechanistic pathways that produce interactive synergism at the organ level. In addition, relationships among measured embryo tissue concentrations, cytochrome P4501A mRNA induction, and cardiotoxic responses suggest a two-compartment toxicokinetic model that independently predicts high potency of PAC mixtures through classical metabolic synergism. These two modes of synergism, specific to the sub-fraction of phenanthrenes, are sufficient to explain the high embryotoxic potency of crude oils, independent of as-yet unmeasured compounds in these complex environmental mixtures.
Collapse
Affiliation(s)
- John P Incardona
- Environmental and Fisheries Science Division, Northwest Fisheries Science Center, National Marine Fisheries Service, NOAA Fisheries, Seattle, WA, USA.
| | - Tiffany L Linbo
- Environmental and Fisheries Science Division, Northwest Fisheries Science Center, National Marine Fisheries Service, NOAA Fisheries, Seattle, WA, USA
| | - James R Cameron
- Saltwater, Inc., Under Contract to Northwest Fisheries Science Center, National Oceanic and Atmospheric Administration, Seattle, WA, USA
| | - Nathaniel L Scholz
- Environmental and Fisheries Science Division, Northwest Fisheries Science Center, National Marine Fisheries Service, NOAA Fisheries, Seattle, WA, USA
| |
Collapse
|
5
|
Truong BT, Shull LC, Zepeda BJ, Lencer E, Artinger KB. Human split hand/foot variants are not as functional as wildtype human PRDM1 in the rescue of craniofacial defects. Birth Defects Res 2024; 116:e2327. [PMID: 38456586 PMCID: PMC10949536 DOI: 10.1002/bdr2.2327] [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: 05/19/2023] [Revised: 01/24/2024] [Accepted: 02/21/2024] [Indexed: 03/09/2024]
Abstract
BACKGROUND Split hand/foot malformation (SHFM) is a congenital limb disorder presenting with limb anomalies, such as missing, hypoplastic, or fused digits, and often craniofacial defects, including a cleft lip/palate, microdontia, micrognathia, or maxillary hypoplasia. We previously identified three novel variants in the transcription factor, PRDM1, that are associated with SHFM phenotypes. One individual also presented with a high arch palate. Studies in vertebrates indicate that PRDM1 is important for development of the skull; however, prior to our study, human variants in PRDM1 had not been associated with craniofacial anomalies. METHODS Using transient mRNA overexpression assays in prdm1a-/- mutant zebrafish, we tested whether the PRDM1 SHFM variants were functional and could lead to a rescue of the craniofacial defects observed in prdm1a-/- mutants. We also mined previously published CUT&RUN and RNA-seq datasets that sorted EGFP-positive cells from a Tg(Mmu:Prx1-EGFP) transgenic line that labels the pectoral fin, pharyngeal arches, and dorsal part of the head to examine Prdm1a binding and the effect of Prdm1a loss on craniofacial genes. RESULTS The prdm1a-/- mutants exhibit craniofacial defects including a hypoplastic neurocranium, a loss of posterior ceratobranchial arches, a shorter palatoquadrate, and an inverted ceratohyal. Injection of wildtype (WT) hPRDM1 in prdm1a-/- mutants partially rescues the palatoquadrate phenotype. However, injection of each of the three SHFM variants fails to rescue this skeletal defect. Loss of prdm1a leads to a decreased expression of important craniofacial genes by RNA-seq, including emilin3a, confirmed by hybridization chain reaction expression. Other genes including dlx5a/dlx6a, hand2, sox9b, col2a1a, and hoxb genes are also reduced. Validation by real-time quantitative PCR in the anterior half of zebrafish embryos failed to confirm the expression changes suggesting that the differences are enriched in prx1 expressing cells. CONCLUSION These data suggest that the three SHFM variants are likely not functional and may be associated with the craniofacial defects observed in the humans. Finally, they demonstrate how Prdm1a can directly bind and regulate genes involved in craniofacial development.
Collapse
Affiliation(s)
- Brittany T Truong
- Human Medical Genetics & Genomics Graduate Program, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado, USA
- Department of Craniofacial Development, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado, USA
| | - Lomeli C Shull
- Department of Craniofacial Development, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado, USA
| | - Bryan J Zepeda
- Department of Diagnostic and Biological Sciences, University of Minnesota School of Dentistry, Minneapolis, Minnesota, USA
| | - Ezra Lencer
- Biology Department, Lafayette College, Easton, Pennsylvania, USA
| | - Kristin B Artinger
- Department of Craniofacial Development, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado, USA
- Department of Diagnostic and Biological Sciences, University of Minnesota School of Dentistry, Minneapolis, Minnesota, USA
| |
Collapse
|
6
|
Liu S, Kawanishi T, Shimada A, Ikeda N, Yamane M, Takeda H, Tasaki J. Identification of an adverse outcome pathway (AOP) for chemical-induced craniofacial anomalies using the transgenic zebrafish model. Toxicol Sci 2023; 196:38-51. [PMID: 37531284 PMCID: PMC10614053 DOI: 10.1093/toxsci/kfad078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/04/2023] Open
Abstract
Craniofacial anomalies are one of the most frequent birth defects worldwide and are often caused by genetic and environmental factors such as pharmaceuticals and chemical agents. Although identifying adverse outcome pathways (AOPs) is a central issue for evaluating the teratogenicity, the AOP causing craniofacial anomalies has not been identified. Recently, zebrafish has gained interest as an emerging model for predicting teratogenicity because of high throughput, cost-effectiveness and availability of various tools for examining teratogenic mechanisms. Here, we established zebrafish sox10-EGFP reporter lines to visualize cranial neural crest cells (CNCCs) and have identified the AOPs for craniofacial anomalies. When we exposed the transgenic embryos to teratogens that were reported to cause craniofacial anomalies in mammals, CNCC migration and subsequent morphogenesis of the first pharyngeal arch were impaired at 24 hours post-fertilization. We also found that cell proliferation and apoptosis of the migratory CNCCs were disturbed, which would be key events of the AOP. From these results, we propose that our sox10-EGFP reporter lines serve as a valuable model for detecting craniofacial skeletal abnormalities, from early to late developmental stages. Given that the developmental process of CNCCs around this stage is highly conserved between zebrafish and mammals, our findings can be extrapolated to mammalian craniofacial development and thus help in predicting craniofacial anomalies in human.
Collapse
Affiliation(s)
- Shujie Liu
- R&D, Safety Science Research, Kao Corporation, Tochigi 321-3497, Japan
| | - Toru Kawanishi
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Tokyo 113-0033, Japan
- School of Life Science and Technology, Tokyo Institute of Technology, Kanagawa 226-8501, Japan
| | - Atsuko Shimada
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Tokyo 113-0033, Japan
| | - Naohiro Ikeda
- R&D, Safety Science Research, Kao Corporation, Kanagawa 210-0821, Japan
| | - Masayuki Yamane
- R&D, Safety Science Research, Kao Corporation, Tochigi 321-3497, Japan
| | - Hiroyuki Takeda
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Tokyo 113-0033, Japan
- Faculty of Life Sciences, Kyoto Sangyo University, Kyoto 603-8555, Japan
| | - Junichi Tasaki
- R&D, Safety Science Research, Kao Corporation, Kanagawa 210-0821, Japan
| |
Collapse
|
7
|
Williams AL, Bohnsack BL. Zebrafish Model of Stickler Syndrome Suggests a Role for Col2a1a in the Neural Crest during Early Eye Development. J Dev Biol 2022; 10:jdb10040042. [PMID: 36278547 PMCID: PMC9589970 DOI: 10.3390/jdb10040042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 09/21/2022] [Accepted: 09/27/2022] [Indexed: 11/16/2022] Open
Abstract
Most cases of Stickler syndrome are due to autosomal-dominant COL2A1 gene mutations leading to abnormal type II collagen. Ocular findings include axial eye lengthening with vitreal degeneration and early-onset glaucoma, which can result in vision loss. Although COL2A1 is a major player in cartilage and bone formation, its specific role in eye development remains elusive. We investigated the role of Col2a1a in neural crest migration and differentiation during early zebrafish eye development. In situ hybridization, immunofluorescence, live imaging, exogenous treatments [10 μM diethylaminobenzaldehyde (DEAB), 100 nM all-trans retinoic acid (RA) and 1-3% ethanol (ETOH)] and morpholino oligonucleotide (MO) injections were used to analyze wildtype Casper (roy-/-;nacre-/-), TgBAC(col2a1a::EGFP), Tg(sox10::EGFP) and Tg(foxd3::EGFP) embryos. Col2a1a colocalized with Foxd3- and Sox10-positive cells in the anterior segment and neural crest-derived jaw. Col2a1a expression was regulated by RA and inhibited by 3% ETOH. Furthermore, MO knockdown of Col2a1a delayed jaw formation and disrupted the ocular anterior segment neural crest migration of Sox10-positive cells. Interestingly, human COL2A1 protein rescued the MO effects. Altogether, these results suggest that Col2a1a is a downstream target of RA in the cranial neural crest and is required for both craniofacial and eye development.
Collapse
Affiliation(s)
- Antionette L. Williams
- Division of Ophthalmology, Ann & Robert H. Lurie Children’s Hospital of Chicago, 225 E. Chicago Ave., Chicago, IL 60611, USA
- Department of Ophthalmology, Northwestern University Feinberg School of Medicine, 645 N. Michigan Ave., Chicago, IL 60611, USA
- Correspondence: (A.L.W.); (B.L.B.); Tel.: +1-312-503-4706 (A.L.W.); +1-312-227-6180 (B.L.B.)
| | - Brenda L. Bohnsack
- Division of Ophthalmology, Ann & Robert H. Lurie Children’s Hospital of Chicago, 225 E. Chicago Ave., Chicago, IL 60611, USA
- Department of Ophthalmology, Northwestern University Feinberg School of Medicine, 645 N. Michigan Ave., Chicago, IL 60611, USA
- Correspondence: (A.L.W.); (B.L.B.); Tel.: +1-312-503-4706 (A.L.W.); +1-312-227-6180 (B.L.B.)
| |
Collapse
|
8
|
Exploring tissue morphodynamics using the photoconvertible Kaede protein in amphioxus embryos. PLoS One 2022; 17:e0275193. [PMID: 36166455 PMCID: PMC9514637 DOI: 10.1371/journal.pone.0275193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 09/12/2022] [Indexed: 12/02/2022] Open
Abstract
Photoconvertible proteins are powerful tools widely used in cellular biology to study cell dynamics and organelles. Over the past decade, photoconvertible proteins have also been used for developmental biology applications to analyze cell lineage and cell fate during embryonic development. One of these photoconvertible proteins called Kaede, from the stony coral Trachyphyllia geoffroyi, undergoes irreversible photoconversion from green to red fluorescence when illuminated with UV light. Undertaking a cell tracing approach using photoconvertible proteins can be challenging when using unconventional animal models. In this protocol, we describe the use of Kaede to track specific cells during embryogenesis of the cephalochordate Branchiostoma lanceolatum. This protocol can be adapted to other unconventional models, especially marine animals.
Collapse
|
9
|
Cui R, Chen D, Li N, Cai M, Wan T, Zhang X, Zhang M, Du S, Ou H, Jiao J, Jiang N, Zhao S, Song H, Song X, Ma D, Zhang J, Li S. PARD3 gene variation as candidate cause of nonsyndromic cleft palate only. J Cell Mol Med 2022; 26:4292-4304. [PMID: 35789100 PMCID: PMC9344820 DOI: 10.1111/jcmm.17452] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 06/01/2022] [Accepted: 06/06/2022] [Indexed: 12/16/2022] Open
Abstract
Nonsyndromic cleft palate only (NSCP) is a common congenital malformation worldwide. In this study, we report a three‐generation pedigree with NSCP following the autosomal‐dominant pattern. Whole‐exome sequencing and Sanger sequencing revealed that only the frameshift variant c.1012dupG [p. E338Gfs*26] in PARD3 cosegregated with the disease. In zebrafish embryos, ethmoid plate patterning defects were observed with PARD3 ortholog disruption or expression of patient‐derived N‐terminal truncating PARD3 (c.1012dupG), which implicated PARD3 in ethmoid plate morphogenesis. PARD3 plays vital roles in determining cellular polarity. Compared with the apical distribution of wild‐type PARD3, PARD3‐p. E338Gfs*26 mainly localized to the basal membrane in 3D‐cultured MCF‐10A epithelial cells. The interaction between PARD3‐p. E338Gfs*26 and endogenous PARD3 was identified by LC–MS/MS and validated by co‐IP. Immunofluorescence analysis showed that PARD3‐p. E338Gfs*26 substantially altered the localization of endogenous PARD3 to the basement membrane in 3D‐cultured MCF‐10A cells. Furthermore, seven variants, including one nonsense variant and six missense variants, were identified in the coding region of PARD3 in sporadic cases with NSCP. Subsequent analysis showed that PARD3‐p. R133*, like the insertion variant of c.1012dupG, also changed the localization of endogenous full‐length PARD3 and that its expression induced abnormal ethmoid plate morphogenesis in zebrafish. Based on these data, we reveal PARD3 gene variation as a novel candidate cause of nonsyndromic cleft palate only.
Collapse
Affiliation(s)
- Renjie Cui
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, Collaborative Innovation Center of Genetics and Development, Institutes of Biomedical Sciences, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Dingli Chen
- Department of Clinical Laboratory, Central Hospital of Handan, Hebei, China
| | - Na Li
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, Collaborative Innovation Center of Genetics and Development, Institutes of Biomedical Sciences, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ming Cai
- Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Teng Wan
- Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Xueqiang Zhang
- Department of Clinical Laboratory, Central Hospital of Handan, Hebei, China.,Oral and Maxillofacial Surgery, Central Hospital of Handan, Hebei, China
| | - Meiqin Zhang
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, Collaborative Innovation Center of Genetics and Development, Institutes of Biomedical Sciences, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Sichen Du
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, Collaborative Innovation Center of Genetics and Development, Institutes of Biomedical Sciences, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Huayuan Ou
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, Collaborative Innovation Center of Genetics and Development, Institutes of Biomedical Sciences, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jianjun Jiao
- Oral and Maxillofacial Surgery, Central Hospital of Handan, Hebei, China
| | - Nan Jiang
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, Collaborative Innovation Center of Genetics and Development, Institutes of Biomedical Sciences, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shuangxia Zhao
- Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Huaidong Song
- Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Xuedong Song
- Department of Clinical Laboratory, Central Hospital of Handan, Hebei, China
| | - Duan Ma
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, Collaborative Innovation Center of Genetics and Development, Institutes of Biomedical Sciences, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China.,Children's Hospital of Fudan University, Shanghai, China
| | - Jin Zhang
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, Collaborative Innovation Center of Genetics and Development, Institutes of Biomedical Sciences, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shouxia Li
- Department of Clinical Laboratory, Central Hospital of Handan, Hebei, China
| |
Collapse
|
10
|
Gao Y, Hu B, Flores R, Xie H, Lin F. Fibronectin and Integrin α5 play overlapping and independent roles in regulating the development of pharyngeal endoderm and cartilage. Dev Biol 2022; 489:122-133. [PMID: 35732225 DOI: 10.1016/j.ydbio.2022.06.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 06/12/2022] [Accepted: 06/16/2022] [Indexed: 11/30/2022]
Abstract
Craniofacial skeletal elements are derived from cranial neural crest cells (CNCCs), which migrate along discrete paths and populate distinct pharyngeal arches, structures that are separated by the neighboring endodermal pouches (EPs). Interactions between the CNCCs and the endoderm are critical for proper craniofacial development. In zebrafish, integrin α5 (Itga5) functions in the endoderm to regulate formation of specifically the first EP (EP1) and the development of the hyoid cartilage. Here we show that fibronectin (Fn), a major component of the extracellular matrix (ECM), is also required for these developmental processes, and that the penetrance of defects in mutants is temperature-dependent. fn1a-/- embryos exhibited defects that are similar to, but much more severe than, those of itga5-/- embryos, and a loss of integrin av (itgav) function enhanced both endoderm and cartilage defects in itga5-/- embryos, suggesting that Itga5 and Itgav cooperate to transmit signals from Fn to regulate the development of endoderm and cartilage. Whereas the endodermal defects in itga5; itga5v-/- double mutant embryos were comparable to those of fn1a-/- mutants, the cartilage defects were much milder. Furthermore, Fn assembly was detected in migrating CNCCs, and the epithelial organization and differentiation of CNCC-derived arches were impaired in fn1a-/- embryos, indicating that Fn1 exerts functions in arch development that are independent of Itga5 and Itgav. Additionally, reduction of itga5 function in fn1a-/- embryos led to profound defects in body axis elongation, as well as in endoderm and cartilage formation, suggesting that other ECM proteins signal through Itga5 to regulate development of the endoderm and cartilage. Thus, our studies reveal that Fn1a and Itga5 have both overlapping and independent functions in regulating development of the pharyngeal endoderm and cartilage.
Collapse
Affiliation(s)
- Yuanyuan Gao
- Department of Anatomy and Cell Biology, Carver College of Medicine, The University of Iowa, Iowa City, IA, 52242, USA
| | - Bo Hu
- Department of Anatomy and Cell Biology, Carver College of Medicine, The University of Iowa, Iowa City, IA, 52242, USA
| | - Rickcardo Flores
- Department of Anatomy and Cell Biology, Carver College of Medicine, The University of Iowa, Iowa City, IA, 52242, USA
| | - Huaping Xie
- Department of Anatomy and Cell Biology, Carver College of Medicine, The University of Iowa, Iowa City, IA, 52242, USA
| | - Fang Lin
- Department of Anatomy and Cell Biology, Carver College of Medicine, The University of Iowa, Iowa City, IA, 52242, USA.
| |
Collapse
|
11
|
Bhagirath AY, Medapati MR, de Jesus VC, Yadav S, Hinton M, Dakshinamurti S, Atukorallaya D. Role of Maternal Infections and Inflammatory Responses on Craniofacial Development. FRONTIERS IN ORAL HEALTH 2021; 2:735634. [PMID: 35048051 PMCID: PMC8757860 DOI: 10.3389/froh.2021.735634] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Accepted: 08/10/2021] [Indexed: 12/12/2022] Open
Abstract
Pregnancy is a tightly regulated immunological state. Mild environmental perturbations can affect the developing fetus significantly. Infections can elicit severe immunological cascades in the mother's body as well as the developing fetus. Maternal infections and resulting inflammatory responses can mediate epigenetic changes in the fetal genome, depending on the developmental stage. The craniofacial development begins at the early stages of embryogenesis. In this review, we will discuss the immunology of pregnancy and its responsive mechanisms on maternal infections. Further, we will also discuss the epigenetic effects of pathogens, their metabolites and resulting inflammatory responses on the fetus with a special focus on craniofacial development. Understanding the pathophysiological mechanisms of infections and dysregulated inflammatory responses during prenatal development could provide better insights into the origins of craniofacial birth defects.
Collapse
Affiliation(s)
- Anjali Y. Bhagirath
- Department of Pediatrics and Physiology, University of Manitoba, Winnipeg, MB, Canada
- Biology of Breathing, Children's Hospital Research Institute of Manitoba (CHRIM), Winnipeg, MB, Canada
- Department of Oral Biology, Dr. Gerald Niznick College of Dentistry, University of Manitoba, Winnipeg, MB, Canada
| | - Manoj Reddy Medapati
- Biology of Breathing, Children's Hospital Research Institute of Manitoba (CHRIM), Winnipeg, MB, Canada
- Department of Oral Biology, Dr. Gerald Niznick College of Dentistry, University of Manitoba, Winnipeg, MB, Canada
| | - Vivianne Cruz de Jesus
- Biology of Breathing, Children's Hospital Research Institute of Manitoba (CHRIM), Winnipeg, MB, Canada
- Department of Oral Biology, Dr. Gerald Niznick College of Dentistry, University of Manitoba, Winnipeg, MB, Canada
| | - Sneha Yadav
- Mahatma Gandhi Institute of Medical Sciences, Wardha, India
| | - Martha Hinton
- Department of Pediatrics and Physiology, University of Manitoba, Winnipeg, MB, Canada
- Biology of Breathing, Children's Hospital Research Institute of Manitoba (CHRIM), Winnipeg, MB, Canada
| | - Shyamala Dakshinamurti
- Department of Pediatrics and Physiology, University of Manitoba, Winnipeg, MB, Canada
- Biology of Breathing, Children's Hospital Research Institute of Manitoba (CHRIM), Winnipeg, MB, Canada
| | - Devi Atukorallaya
- Biology of Breathing, Children's Hospital Research Institute of Manitoba (CHRIM), Winnipeg, MB, Canada
- Department of Oral Biology, Dr. Gerald Niznick College of Dentistry, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
12
|
Kamenev D, Sunadome K, Shirokov M, Chagin AS, Singh A, Irion U, Adameyko I, Fried K, Dyachuk V. Schwann cell precursors generate sympathoadrenal system during zebrafish development. J Neurosci Res 2021; 99:2540-2557. [PMID: 34184294 DOI: 10.1002/jnr.24909] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 05/24/2021] [Accepted: 06/01/2021] [Indexed: 11/07/2022]
Abstract
The autonomic portion of the peripheral nervous system orchestrates tissue homeostasis through direct innervation of internal organs, and via release of adrenalin and noradrenalin into the blood flow. The developmental mechanisms behind the formation of autonomic neurons and chromaffin cells are not fully understood. Using genetic tracing, we discovered that a significant proportion of sympathetic neurons in zebrafish originates from Schwann cell precursors (SCPs) during a defined period of embryonic development. Moreover, SCPs give rise to the main portion of the chromaffin cells, as well as to a significant proportion of enteric and other autonomic neurons associated with internal organs. The conversion of SCPs into neuronal and chromaffin cells is ErbB receptor dependent, as the pharmacological inhibition of the ErbB pathway effectively perturbed this transition. Finally, using genetic ablations, we revealed that SCPs producing neurons and chromaffin cells migrate along spinal motor axons to reach appropriate target locations. This study reveals the evolutionary conservation of SCP-to-neuron and SCP-to-chromaffin cell transitions over significant growth periods in fish and highlights relevant cellular-genetic mechanisms. Based on this, we anticipate that multipotent SCPs might be present in postnatal vertebrate tissues, retaining the capacity to regenerate autonomic neurons and chromaffin cells.
Collapse
Affiliation(s)
- Dmitrii Kamenev
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Kazunori Sunadome
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Maxim Shirokov
- National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, Vladivostok, Russia
| | - Andrey S Chagin
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.,Institute for Regenerative Medicine, Sechenov University, Moscow, Russia
| | - Ajeet Singh
- Max-Planck-Institut für Entwicklungsbiologie, Tübingen, Germany
| | - Uwe Irion
- Max-Planck-Institut für Entwicklungsbiologie, Tübingen, Germany
| | - Igor Adameyko
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.,Center for Brain Research, Medical University Vienna, Vienna, Austria
| | - Kaj Fried
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Vyacheslav Dyachuk
- National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, Vladivostok, Russia
| |
Collapse
|
13
|
Carroll SH, Macias Trevino C, Li EB, Kawasaki K, Myers N, Hallett SA, Alhazmi N, Cotney J, Carstens RP, Liao EC. An Irf6- Esrp1/2 regulatory axis controls midface morphogenesis in vertebrates. Development 2020; 147:dev194498. [PMID: 33234718 PMCID: PMC7774891 DOI: 10.1242/dev.194498] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 11/02/2020] [Indexed: 12/25/2022]
Abstract
Irf6 and Esrp1 are important for palate development across vertebrates. In zebrafish, we found that irf6 regulates the expression of esrp1 We detailed overlapping Irf6 and Esrp1/2 expression in mouse orofacial epithelium. In zebrafish, irf6 and esrp1/2 share expression in periderm, frontonasal ectoderm and oral epithelium. Genetic disruption of irf6 and esrp1/2 in zebrafish resulted in cleft of the anterior neurocranium. The esrp1/2 mutant also developed cleft of the mouth opening. Lineage tracing of cranial neural crest cells revealed that the cleft resulted not from migration defect, but from impaired chondrogenesis. Analysis of aberrant cells within the cleft revealed expression of sox10, col1a1 and irf6, and these cells were adjacent to krt4+ and krt5+ cells. Breeding of mouse Irf6; Esrp1; Esrp2 compound mutants suggested genetic interaction, as the triple homozygote and the Irf6; Esrp1 double homozygote were not observed. Further, Irf6 heterozygosity reduced Esrp1/2 cleft severity. These studies highlight the complementary analysis of Irf6 and Esrp1/2 in mouse and zebrafish, and identify a unique aberrant cell population in zebrafish expressing sox10, col1a1 and irf6 Future work characterizing this cell population will yield additional insight into cleft pathogenesis.
Collapse
Affiliation(s)
- Shannon H. Carroll
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Shriners Hospital for Children, Boston, MA 02114, USA
| | - Claudio Macias Trevino
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Harvard Medical School, Boston, MA 02115, USA
| | | | - Kenta Kawasaki
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Shriners Hospital for Children, Boston, MA 02114, USA
| | - Nikita Myers
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Shawn A. Hallett
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Nora Alhazmi
- Harvard School of Dental Medicine, Boston, MA 02115, USA
| | - Justin Cotney
- Department of Genetics and Genome Sciences, University of Connecticut Health, CT 06030, USA
| | - Russ P. Carstens
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Eric C. Liao
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Shriners Hospital for Children, Boston, MA 02114, USA
- Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
14
|
Raterman ST, Metz JR, Wagener FADTG, Von den Hoff JW. Zebrafish Models of Craniofacial Malformations: Interactions of Environmental Factors. Front Cell Dev Biol 2020; 8:600926. [PMID: 33304906 PMCID: PMC7701217 DOI: 10.3389/fcell.2020.600926] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 10/23/2020] [Indexed: 11/13/2022] Open
Abstract
The zebrafish is an appealing model organism for investigating the genetic (G) and environmental (E) factors, as well as their interactions (GxE), which contribute to craniofacial malformations. Here, we review zebrafish studies on environmental factors involved in the etiology of craniofacial malformations in humans including maternal smoking, alcohol consumption, nutrition and drug use. As an example, we focus on the (cleft) palate, for which the zebrafish ethmoid plate is a good model. This review highlights the importance of investigating ExE interactions and discusses the variable effects of exposure to environmental factors on craniofacial development depending on dosage, exposure time and developmental stage. Zebrafish also promise to be a good tool to study novel craniofacial teratogens and toxin mixtures. Lastly, we discuss the handful of studies on gene–alcohol interactions using mutant sensitivity screens and reverse genetic techniques. We expect that studies addressing complex interactions (ExE and GxE) in craniofacial malformations will increase in the coming years. These are likely to uncover currently unknown mechanisms with implications for the prevention of craniofacial malformations. The zebrafish appears to be an excellent complementary model with high translational value to study these complex interactions.
Collapse
Affiliation(s)
- S T Raterman
- Radboud Institute of Molecular Life Sciences, Nijmegen, Netherlands.,Department of Dentistry-Orthodontics and Craniofacial Biology, Radboud University Medical Center, Nijmegen, Netherlands.,Department of Animal Ecology and Physiology, Institute for Water and Wetland Research, Radboud University, Nijmegen, Netherlands
| | - J R Metz
- Department of Animal Ecology and Physiology, Institute for Water and Wetland Research, Radboud University, Nijmegen, Netherlands
| | - Frank A D T G Wagener
- Radboud Institute of Molecular Life Sciences, Nijmegen, Netherlands.,Department of Dentistry-Orthodontics and Craniofacial Biology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Johannes W Von den Hoff
- Radboud Institute of Molecular Life Sciences, Nijmegen, Netherlands.,Department of Dentistry-Orthodontics and Craniofacial Biology, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
15
|
Weigele J, Bohnsack BL. Genetics Underlying the Interactions between Neural Crest Cells and Eye Development. J Dev Biol 2020; 8:jdb8040026. [PMID: 33182738 PMCID: PMC7712190 DOI: 10.3390/jdb8040026] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 11/03/2020] [Accepted: 11/07/2020] [Indexed: 12/14/2022] Open
Abstract
The neural crest is a unique, transient stem cell population that is critical for craniofacial and ocular development. Understanding the genetics underlying the steps of neural crest development is essential for gaining insight into the pathogenesis of congenital eye diseases. The neural crest cells play an under-appreciated key role in patterning the neural epithelial-derived optic cup. These interactions between neural crest cells within the periocular mesenchyme and the optic cup, while not well-studied, are critical for optic cup morphogenesis and ocular fissure closure. As a result, microphthalmia and coloboma are common phenotypes in human disease and animal models in which neural crest cell specification and early migration are disrupted. In addition, neural crest cells directly contribute to numerous ocular structures including the cornea, iris, sclera, ciliary body, trabecular meshwork, and aqueous outflow tracts. Defects in later neural crest cell migration and differentiation cause a constellation of well-recognized ocular anterior segment anomalies such as Axenfeld–Rieger Syndrome and Peters Anomaly. This review will focus on the genetics of the neural crest cells within the context of how these complex processes specifically affect overall ocular development and can lead to congenital eye diseases.
Collapse
Affiliation(s)
- Jochen Weigele
- Division of Ophthalmology, Ann & Robert H. Lurie Children’s Hospital of Chicago, 225 E. Chicago Ave, Chicago, IL 60611, USA;
- Department of Ophthalmology, Northwestern University Feinberg School of Medicine, 645 N. Michigan Ave, Chicago, IL 60611, USA
| | - Brenda L. Bohnsack
- Division of Ophthalmology, Ann & Robert H. Lurie Children’s Hospital of Chicago, 225 E. Chicago Ave, Chicago, IL 60611, USA;
- Department of Ophthalmology, Northwestern University Feinberg School of Medicine, 645 N. Michigan Ave, Chicago, IL 60611, USA
- Correspondence: ; Tel.: +1-312-227-6180; Fax: +1-312-227-9411
| |
Collapse
|
16
|
Atukorala ADS, Ratnayake RK. Cellular and molecular mechanisms in the development of a cleft lip and/or cleft palate; insights from zebrafish (Danio rerio). Anat Rec (Hoboken) 2020; 304:1650-1660. [PMID: 33099891 DOI: 10.1002/ar.24547] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 08/31/2020] [Accepted: 09/09/2020] [Indexed: 12/18/2022]
Abstract
Human cleft lip and/or palate (CLP) are immediately recognizable congenital abnormalities of the face. Lip and palate develop from facial primordia through the coordinated activities of ectodermal epithelium and neural crest cells (NCCs) derived from ectomesenchyme tissue. Subtle changes in the regulatory mechanisms of NCC or ectodermal epithelial cells can result in CLP. Genetic and environmental contributions or a combination of both play a significant role in the progression of CLP. Model organisms provide us with a wealth of information in understanding the pathophysiology and genetic etiology of this complex disease. Small teleost, zebrafish (Danio rerio) is one of the popular model in craniofacial developmental biology. The short generation time and large number of optically transparent, easily manipulated embryos increase the value of zebrafish to identify novel candidate genes and gene regulatory networks underlying craniofacial development. In addition, it is widely used to identify the mechanisms of environmental teratogens and in therapeutic drug screening. Here, we discuss the value of zebrafish as a model to understand epithelial and NCC induced ectomesenchymal cell activities during early palate morphogenesis and robustness of the zebrafish in modern research on identifying the genetic and environmental etiological factors of CLP.
Collapse
Affiliation(s)
- Atukorallaya Devi Sewvandini Atukorala
- Rady Faculty of Health Sciences, Department of Oral Biology, Dr. Gerald Niznick College of Dentistry, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Ravindra Kumar Ratnayake
- Rady Faculty of Health Sciences, Department of Oral Biology, Dr. Gerald Niznick College of Dentistry, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
17
|
Pini J, Kueper J, Hu YD, Kawasaki K, Yeung P, Tsimbal C, Yoon B, Carmichael N, Maas RL, Cotney J, Grinblat Y, Liao EC. ALX1-related frontonasal dysplasia results from defective neural crest cell development and migration. EMBO Mol Med 2020; 12:e12013. [PMID: 32914578 PMCID: PMC7539331 DOI: 10.15252/emmm.202012013] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 08/12/2020] [Accepted: 08/13/2020] [Indexed: 01/02/2023] Open
Abstract
A pedigree of subjects presented with frontonasal dysplasia (FND). Genome sequencing and analysis identified a p.L165F missense variant in the homeodomain of the transcription factor ALX1 which was imputed to be pathogenic. Induced pluripotent stem cells (iPSC) were derived from the subjects and differentiated to neural crest cells (NCC). NCC derived from ALX1L165F/L165F iPSC were more sensitive to apoptosis, showed an elevated expression of several neural crest progenitor state markers, and exhibited impaired migration compared to wild-type controls. NCC migration was evaluated in vivo using lineage tracing in a zebrafish model, which revealed defective migration of the anterior NCC stream that contributes to the median portion of the anterior neurocranium, phenocopying the clinical presentation. Analysis of human NCC culture media revealed a change in the level of bone morphogenic proteins (BMP), with a low level of BMP2 and a high level of BMP9. Soluble BMP2 and BMP9 antagonist treatments were able to rescue the defective migration phenotype. Taken together, these results demonstrate a mechanistic requirement of ALX1 in NCC development and migration.
Collapse
Affiliation(s)
- Jonathan Pini
- Center for Regenerative MedicineDepartment of SurgeryMassachusetts General HospitalBostonMAUSA
- Shriners Hospital for ChildrenBostonMAUSA
| | - Janina Kueper
- Center for Regenerative MedicineDepartment of SurgeryMassachusetts General HospitalBostonMAUSA
- Shriners Hospital for ChildrenBostonMAUSA
- Life and Brain CenterUniversity of BonnBonnGermany
| | - Yiyuan David Hu
- Center for Regenerative MedicineDepartment of SurgeryMassachusetts General HospitalBostonMAUSA
- Shriners Hospital for ChildrenBostonMAUSA
| | - Kenta Kawasaki
- Center for Regenerative MedicineDepartment of SurgeryMassachusetts General HospitalBostonMAUSA
- Shriners Hospital for ChildrenBostonMAUSA
| | - Pan Yeung
- Center for Regenerative MedicineDepartment of SurgeryMassachusetts General HospitalBostonMAUSA
- Shriners Hospital for ChildrenBostonMAUSA
| | - Casey Tsimbal
- Center for Regenerative MedicineDepartment of SurgeryMassachusetts General HospitalBostonMAUSA
- Shriners Hospital for ChildrenBostonMAUSA
| | - Baul Yoon
- Departments of Integrative Biology, Neuroscience, and Genetics Ph.D. Training ProgramUniversity of Wisconsin‐MadisonMadisonWIUSA
| | - Nikkola Carmichael
- Department of GeneticsBrigham and Women's HospitalHarvard Medical SchoolBostonMAUSA
| | - Richard L Maas
- Department of GeneticsBrigham and Women's HospitalHarvard Medical SchoolBostonMAUSA
| | - Justin Cotney
- Genetics and Genome SciencesUConn HealthFarmingtonCTUSA
| | - Yevgenya Grinblat
- Departments of Integrative Biology, Neuroscience, and Genetics Ph.D. Training ProgramUniversity of Wisconsin‐MadisonMadisonWIUSA
| | - Eric C Liao
- Center for Regenerative MedicineDepartment of SurgeryMassachusetts General HospitalBostonMAUSA
- Shriners Hospital for ChildrenBostonMAUSA
| |
Collapse
|
18
|
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: 1.0] [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
|
19
|
Duan C, Allard J. Gonadotropin-releasing hormone neuron development in vertebrates. Gen Comp Endocrinol 2020; 292:113465. [PMID: 32184073 DOI: 10.1016/j.ygcen.2020.113465] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 02/26/2020] [Accepted: 03/12/2020] [Indexed: 11/21/2022]
Abstract
Gonadotropin-releasing hormone (GnRH) neurons are master regulators of the reproductive axis in vertebrates. During early mammalian embryogenesis, GnRH1 neurons emerge in the nasal/olfactory placode. These neurons undertake a long-distance migration, moving from the nose to the preoptic area and hypothalamus. While significant advances have been made in understanding the functional importance of the GnRH1 neurons in reproduction, where GnRH1 neurons come from and how are they specified during early development is still under debate. In addition to the GnRH1 gene, most vertebrate species including humans have one or two additional GnRH genes. Compared to the GnRH1 neurons, much less is known about the development and regulation of GnRH2 neuron and GnRH3 neurons. The objective of this article is to review what is currently known about GnRH neuron development. We will survey various cell autonomous and non-autonomous factors implicated in the regulation of GnRH neuron development. Finally, we will discuss emerging tools and new approaches to resolve open questions pertaining to GnRH neuron development.
Collapse
Affiliation(s)
- Cunming Duan
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, United States.
| | - John Allard
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, United States
| |
Collapse
|
20
|
Tonelli F, Bek JW, Besio R, De Clercq A, Leoni L, Salmon P, Coucke PJ, Willaert A, Forlino A. Zebrafish: A Resourceful Vertebrate Model to Investigate Skeletal Disorders. Front Endocrinol (Lausanne) 2020; 11:489. [PMID: 32849280 PMCID: PMC7416647 DOI: 10.3389/fendo.2020.00489] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Accepted: 06/22/2020] [Indexed: 12/11/2022] Open
Abstract
Animal models are essential tools for addressing fundamental scientific questions about skeletal diseases and for the development of new therapeutic approaches. Traditionally, mice have been the most common model organism in biomedical research, but their use is hampered by several limitations including complex generation, demanding investigation of early developmental stages, regulatory restrictions on breeding, and high maintenance cost. The zebrafish has been used as an efficient alternative vertebrate model for the study of human skeletal diseases, thanks to its easy genetic manipulation, high fecundity, external fertilization, transparency of rapidly developing embryos, and low maintenance cost. Furthermore, zebrafish share similar skeletal cells and ossification types with mammals. In the last decades, the use of both forward and new reverse genetics techniques has resulted in the generation of many mutant lines carrying skeletal phenotypes associated with human diseases. In addition, transgenic lines expressing fluorescent proteins under bone cell- or pathway- specific promoters enable in vivo imaging of differentiation and signaling at the cellular level. Despite the small size of the zebrafish, many traditional techniques for skeletal phenotyping, such as x-ray and microCT imaging and histological approaches, can be applied using the appropriate equipment and custom protocols. The ability of adult zebrafish to remodel skeletal tissues can be exploited as a unique tool to investigate bone formation and repair. Finally, the permeability of embryos to chemicals dissolved in water, together with the availability of large numbers of small-sized animals makes zebrafish a perfect model for high-throughput bone anabolic drug screening. This review aims to discuss the techniques that make zebrafish a powerful model to investigate the molecular and physiological basis of skeletal disorders.
Collapse
Affiliation(s)
- Francesca Tonelli
- Biochemistry Unit, Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Jan Willem Bek
- Department of Biomolecular Medicine, Center of Medical Genetics, Ghent University-University Hospital, Ghent, Belgium
| | - Roberta Besio
- Biochemistry Unit, Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Adelbert De Clercq
- Department of Biomolecular Medicine, Center of Medical Genetics, Ghent University-University Hospital, Ghent, Belgium
| | - Laura Leoni
- Biochemistry Unit, Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | | | - Paul J. Coucke
- Department of Biomolecular Medicine, Center of Medical Genetics, Ghent University-University Hospital, Ghent, Belgium
| | - Andy Willaert
- Department of Biomolecular Medicine, Center of Medical Genetics, Ghent University-University Hospital, Ghent, Belgium
| | - Antonella Forlino
- Biochemistry Unit, Department of Molecular Medicine, University of Pavia, Pavia, Italy
- *Correspondence: Antonella Forlino
| |
Collapse
|
21
|
Sander V, Salleh L, Naylor RW, Schierding W, Sontam D, O’Sullivan JM, Davidson AJ. Transcriptional profiling of the zebrafish proximal tubule. Am J Physiol Renal Physiol 2019; 317:F478-F488. [DOI: 10.1152/ajprenal.00174.2019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The hepatocyte nuclear factor-1β (Hnf1b) transcription factor is a key regulator of kidney tubule formation and is associated with a syndrome of renal cysts and early onset diabetes. To further our understanding of Hnf1b in the developing zebrafish kidney, we performed RNA sequencing analysis of proximal tubules from hnf1b-deficient larvae. This analysis revealed an enrichment of gene transcripts encoding transporters of the solute carrier (SLC) superfamily, including multiple members of slc2 and slc5 glucose transporters. An investigation of expression of slc2a1a, slc2a2, and slc5a2 as well as a poorly studied glucose/mannose transporter encoded by slc5a9 revealed that these genes undergo dynamic spatiotemporal changes during tubule formation and maturation. A comparative analysis of zebrafish SLC genes with those expressed in mouse proximal tubules showed a substantial overlap at the level of gene families, indicating a high degree of functional conservation between zebrafish and mammalian proximal tubules. Taken together, our findings are consistent with a role for Hnf1b as a critical determinant of proximal tubule transport function by acting upstream of a large number of SLC genes and validate the zebrafish as a physiologically relevant model of the mammalian proximal tubule.
Collapse
Affiliation(s)
- Veronika Sander
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - Liam Salleh
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - Richard W. Naylor
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | | | - Dharani Sontam
- The Liggins Institute, University of Auckland, Auckland, New Zealand
| | | | - Alan J. Davidson
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| |
Collapse
|
22
|
Manocha S, Farokhnia N, Khosropanah S, Bertol JW, Santiago J, Fakhouri WD. Systematic review of hormonal and genetic factors involved in the nonsyndromic disorders of the lower jaw. Dev Dyn 2019; 248:162-172. [PMID: 30576023 DOI: 10.1002/dvdy.8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 11/30/2018] [Accepted: 12/14/2018] [Indexed: 12/14/2022] Open
Abstract
Mandibular disorders are among the most common birth defects in humans, yet the etiological factors are largely unknown. Most of the neonates affected by mandibular abnormalities have a sequence of secondary anomalies, including airway obstruction and feeding problems, that reduce the quality of life. In the event of lacking corrective surgeries, patients with mandibular congenital disorders suffer from additional lifelong problems such as sleep apnea and temporomandibular disorders, among others. The goal of this systematic review is to gather evidence on hormonal and genetic factors that are involved in signaling pathways and interactions that are potentially associated with the nonsyndromic mandibular disorders. We found that members of FGF and BMP pathways, including FGF8/10, FGFR2/3, BMP2/4/7, BMPR1A, ACVR1, and ACVR2A/B, have a prominent number of gene-gene interactions among all identified genes in this review. Gene ontology of the 154 genes showed that the functional gene sets are involved in all aspects of cellular processes and organogenesis. Some of the genes identified by the genome-wide association studies of common mandibular disorders are involved in skeletal formation and growth retardation based on animal models, suggesting a potential direct role as genetic risk factors in the common complex jaw disorders. Developmental Dynamics 248:162-172, 2019. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Srishti Manocha
- Center for Craniofacial Research, Department of Diagnostic and Biomedical Sciences, School of Dentistry, University of Texas Health Science Center at Houston, Houston, Texas
| | - Nadia Farokhnia
- Center for Craniofacial Research, Department of Diagnostic and Biomedical Sciences, School of Dentistry, University of Texas Health Science Center at Houston, Houston, Texas
| | - Sepideh Khosropanah
- Ostrow School of Dentistry, University of Southern California, California, Los Angeles
| | - Jessica W Bertol
- Center for Craniofacial Research, Department of Diagnostic and Biomedical Sciences, School of Dentistry, University of Texas Health Science Center at Houston, Houston, Texas
| | - Joel Santiago
- Pró-Reitoria de Pesquisa e Pós-graduação (PRPPG), Universidade do Sagrado Coração, Jardim Brasil, Bauru, Sao Paulo, Brazil
| | - Walid D Fakhouri
- Center for Craniofacial Research, Department of Diagnostic and Biomedical Sciences, School of Dentistry, University of Texas Health Science Center at Houston, Houston, Texas.,Department of Pediatrics, McGovern Medical School, University of Texas Health Science Center, Houston, Texas
| |
Collapse
|
23
|
Affiliation(s)
- Seth M. Weinberg
- Department of Oral Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Anthropology, University of Pittsburgh, Pittsburgh, Pennsylvania, United Staes of America
- * E-mail:
| | - Robert Cornell
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, Iowa, United States America
| | - Elizabeth J. Leslie
- Department of Human Genetics, Emory University, Atlanta, Georgia, United States of America
| |
Collapse
|
24
|
Genetic Requirement of talin1 for Proliferation of Cranial Neural Crest Cells during Palate Development. PLASTIC AND RECONSTRUCTIVE SURGERY-GLOBAL OPEN 2018; 6:e1633. [PMID: 29707441 PMCID: PMC5908504 DOI: 10.1097/gox.0000000000001633] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 11/16/2017] [Indexed: 01/20/2023]
Abstract
Supplemental Digital Content is available in the text. Background: Craniofacial malformations are among the most common congenital anomalies. Cranial neural crest cells (CNCCs) form craniofacial structures involving multiple cellular processes, perturbations of which contribute to craniofacial malformations. Adhesion of cells to the extracellular matrix mediates bidirectional interactions of the cells with their extracellular environment that plays an important role in craniofacial morphogenesis. Talin (tln) is crucial in cell-matrix adhesion between cells, but its role in craniofacial morphogenesis is poorly understood. Methods: Talin gene expression was determined by whole mount in situ hybridization. Craniofacial cartilage and muscles were analyzed by Alcian blue in Tg(mylz2:mCherry) and by transmission electron microscopy. Pulse-chase photoconversion, 5-ethynyl-2’-deoxyuridine proliferation, migration, and apoptosis assays were performed for functional analysis. Results: Expression of tln1 was observed in the craniofacial cartilage structures, including the palate. The Meckel’s cartilage was hypoplastic, the palate was shortened, and the craniofacial muscles were malformed in tln1 mutants. Pulse-chase and EdU assays during palate morphogenesis revealed defects in CNCC proliferation in mutants. No defects were observed in CNCC migration and apoptosis. Conclusions: The work shows that tln1 is critical for craniofacial morphogenesis in zebrafish. Loss of tln1 leads to a shortened palate and Meckel’s cartilage along with disorganized skeletal muscles. Investigations into the cellular processes show that tln1 is required for CNCC proliferation during palate morphogenesis. The work will lead to a better understanding of the involvement of cytoskeletal proteins in craniofacial morphogenesis.
Collapse
|
25
|
Early life exposure to ethinylestradiol enhances subsequent responses to environmental estrogens measured in a novel transgenic zebrafish. Sci Rep 2018; 8:2699. [PMID: 29426849 PMCID: PMC5807302 DOI: 10.1038/s41598-018-20922-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 01/04/2018] [Indexed: 11/11/2022] Open
Abstract
Estrogen plays fundamental roles in a range of developmental processes and exposure to estrogen mimicking chemicals has been associated with various adverse health effects in both wildlife and human populations. Estrogenic chemicals are found commonly as mixtures in the environment and can have additive effects, however risk analysis is typically conducted for single-chemicals with little, or no, consideration given for an animal’s exposure history. Here we developed a transgenic zebrafish with a photoconvertable fluorophore (Kaede, green to red on UV light exposure) in a skin pigment-free mutant element (ERE)-Kaede-Casper model and applied it to quantify tissue-specific fluorescence biosensor responses for combinations of estrogen exposures during early life using fluorescence microscopy and image analysis. We identify windows of tissue-specific sensitivity to ethinylestradiol (EE2) for exposure during early-life (0–5 dpf) and illustrate that exposure to estrogen (EE2) during 0–48 hpf enhances responsiveness (sensitivity) to different environmental estrogens (EE2, genistein and bisphenol A) for subsequent exposures during development. Our findings illustrate the importance of an organism’s stage of development and estrogen exposure history for assessments on, and possible health risks associated with, estrogen exposure.
Collapse
|
26
|
Duncan KM, Mukherjee K, Cornell RA, Liao EC. Zebrafish models of orofacial clefts. Dev Dyn 2017; 246:897-914. [PMID: 28795449 DOI: 10.1002/dvdy.24566] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 07/06/2017] [Accepted: 07/31/2017] [Indexed: 12/12/2022] Open
Abstract
Zebrafish is a model organism that affords experimental advantages toward investigating the normal function of genes associated with congenital birth defects. Here we summarize zebrafish studies of genes implicated in orofacial cleft (OFC). The most common use of zebrafish in this context has been to explore the normal function an OFC-associated gene product in craniofacial morphogenesis by inhibiting expression of its zebrafish ortholog. The most frequently deployed method has been to inject embryos with antisense morpholino oligonucleotides targeting the desired transcript. However, improvements in targeted mutagenesis strategies have led to widespread adoption of CRISPR/Cas9 technology. A second application of zebrafish has been for functional assays of gene variants found in OFC patients; such in vivo assays are valuable because the success of in silico methods for testing allele severity has been mixed. Finally, zebrafish have been used to test the tissue specificity of enhancers that harbor single nucleotide polymorphisms associated with risk for OFC. We review examples of each of these approaches in the context of genes that are implicated in syndromic and non-syndromic OFC. Developmental Dynamics 246:897-914, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Kaylia M Duncan
- Department of Anatomy and Cell Biology, Molecular and Cell Biology Graduate Program, University of Iowa, Iowa City, Iowa
| | - Kusumika Mukherjee
- Center for Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Robert A Cornell
- Department of Anatomy and Cell Biology, Molecular and Cell Biology Graduate Program, University of Iowa, Iowa City, Iowa
| | - Eric C Liao
- Center for Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
27
|
Ciarlo C, Kaufman CK, Kinikoglu B, Michael J, Yang S, D′Amato C, Blokzijl-Franke S, den Hertog J, Schlaeger TM, Zhou Y, Liao E, Zon LI. A chemical screen in zebrafish embryonic cells establishes that Akt activation is required for neural crest development. eLife 2017; 6:e29145. [PMID: 28832322 PMCID: PMC5599238 DOI: 10.7554/elife.29145] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 08/08/2017] [Indexed: 01/09/2023] Open
Abstract
The neural crest is a dynamic progenitor cell population that arises at the border of neural and non-neural ectoderm. The inductive roles of FGF, Wnt, and BMP at the neural plate border are well established, but the signals required for subsequent neural crest development remain poorly characterized. Here, we conducted a screen in primary zebrafish embryo cultures for chemicals that disrupt neural crest development, as read out by crestin:EGFP expression. We found that the natural product caffeic acid phenethyl ester (CAPE) disrupts neural crest gene expression, migration, and melanocytic differentiation by reducing Sox10 activity. CAPE inhibits FGF-stimulated PI3K/Akt signaling, and neural crest defects in CAPE-treated embryos are suppressed by constitutively active Akt1. Inhibition of Akt activity by constitutively active PTEN similarly decreases crestin expression and Sox10 activity. Our study has identified Akt as a novel intracellular pathway required for neural crest differentiation.
Collapse
Affiliation(s)
- Christie Ciarlo
- Stem Cell Program and Hematology/OncologyChildren’s Hospital Boston, Howard Hughes Medical InstituteBostonUnited States
- Harvard Medical SchoolBostonUnited States
| | - Charles K Kaufman
- Division of Oncology, Department of MedicineWashington University School of MedicineSt. LouisUnited States
- Department of Developmental BiologyWashington University School of MedicineSt. LouisUnited States
| | - Beste Kinikoglu
- Center for Regenerative MedicineMassachusetts General HospitalBostonUnited States
- Division of Plastic and Reconstructive SurgeryMassachusetts General HospitalBostonUnited States
| | - Jonathan Michael
- Stem Cell Program and Hematology/OncologyChildren’s Hospital Boston, Howard Hughes Medical InstituteBostonUnited States
| | - Song Yang
- Stem Cell Program and Hematology/OncologyChildren’s Hospital Boston, Howard Hughes Medical InstituteBostonUnited States
| | - Christopher D′Amato
- Stem Cell Program and Hematology/OncologyChildren’s Hospital Boston, Howard Hughes Medical InstituteBostonUnited States
| | - Sasja Blokzijl-Franke
- Hubrecht Institute, Koninklijke Nederlandse Akademie van WetenschappenUniversity Medical Center UtrechtUtrechtNetherlands
| | - Jeroen den Hertog
- Hubrecht Institute, Koninklijke Nederlandse Akademie van WetenschappenUniversity Medical Center UtrechtUtrechtNetherlands
| | - Thorsten M Schlaeger
- Stem Cell Program and Hematology/OncologyChildren’s Hospital Boston, Howard Hughes Medical InstituteBostonUnited States
| | - Yi Zhou
- Stem Cell Program and Hematology/OncologyChildren’s Hospital Boston, Howard Hughes Medical InstituteBostonUnited States
| | - Eric Liao
- Harvard Medical SchoolBostonUnited States
- Center for Regenerative MedicineMassachusetts General HospitalBostonUnited States
- Division of Plastic and Reconstructive SurgeryMassachusetts General HospitalBostonUnited States
- Harvard Stem Cell InstituteCambridgeUnited States
| | - Leonard I Zon
- Stem Cell Program and Hematology/OncologyChildren’s Hospital Boston, Howard Hughes Medical InstituteBostonUnited States
- Harvard Medical SchoolBostonUnited States
- Harvard Stem Cell InstituteCambridgeUnited States
| |
Collapse
|
28
|
Eason J, Williams AL, Chawla B, Apsey C, Bohnsack BL. Differences in neural crest sensitivity to ethanol account for the infrequency of anterior segment defects in the eye compared with craniofacial anomalies in a zebrafish model of fetal alcohol syndrome. Birth Defects Res 2017; 109:1212-1227. [PMID: 28681995 DOI: 10.1002/bdr2.1069] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 05/21/2017] [Accepted: 05/22/2017] [Indexed: 11/10/2022]
Abstract
BACKGROUND Ethanol (ETOH) exposure during pregnancy is associated with craniofacial and neurologic abnormalities, but infrequently disrupts the anterior segment of the eye. In these studies, we used zebrafish to investigate differences in the teratogenic effect of ETOH on craniofacial, periocular, and ocular neural crest. METHODS Zebrafish eye and neural crest development was analyzed by means of live imaging, TUNEL (terminal deoxynucleotidyl transferase dUTP nick end labeling) assay, immunostaining, detection of reactive oxygen species, and in situ hybridization. RESULTS Our studies demonstrated that foxd3-positive neural crest cells in the periocular mesenchyme and developing eye were less sensitive to ETOH than sox10-positive craniofacial neural crest cells that form the pharyngeal arches and jaw. ETOH increased apoptosis in the retina, but did not affect survival of periocular and ocular neural crest cells. ETOH also did not increase reactive oxygen species within the eye. In contrast, ETOH increased ventral neural crest apoptosis and reactive oxygen species production in the facial mesenchyme. In the eye and craniofacial region, sod2 showed high levels of expression in the anterior segment and in the setting of Sod2 knockdown, low levels of ETOH decreased migration of foxd3-positive neural crest cells into the developing eye. However, ETOH had minimal effect on the periocular and ocular expression of transcription factors (pitx2 and foxc1) that regulate anterior segment development. CONCLUSION Neural crest cells contributing to the anterior segment of the eye exhibit increased ability to withstand ETOH-induced oxidative stress and apoptosis. These studies explain the rarity of anterior segment dysgenesis despite the frequent craniofacial abnormalities in fetal alcohol syndrome. Birth Defects Research 109:1212-1227, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Jessica Eason
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, Michigan
| | - Antionette L Williams
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, Michigan
| | - Bahaar Chawla
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, Michigan
| | - Christian Apsey
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, Michigan
| | - Brenda L Bohnsack
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
29
|
Schoen C, Aschrafi A, Thonissen M, Poelmans G, Von den Hoff JW, Carels CEL. MicroRNAs in Palatogenesis and Cleft Palate. Front Physiol 2017; 8:165. [PMID: 28420997 PMCID: PMC5378724 DOI: 10.3389/fphys.2017.00165] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 03/06/2017] [Indexed: 01/01/2023] Open
Abstract
Palatogenesis requires a precise spatiotemporal regulation of gene expression, which is controlled by an intricate network of transcription factors and their corresponding DNA motifs. Even minor perturbations of this network may cause cleft palate, the most common congenital craniofacial defect in humans. MicroRNAs (miRNAs), a class of small regulatory non-coding RNAs, have elicited strong interest as key regulators of embryological development, and as etiological factors in disease. MiRNAs function as post-transcriptional repressors of gene expression and are therefore able to fine-tune gene regulatory networks. Several miRNAs are already identified to be involved in congenital diseases. Recent evidence from research in zebrafish and mice indicates that miRNAs are key factors in both normal palatogenesis and cleft palate formation. Here, we provide an overview of recently identified molecular mechanisms underlying palatogenesis involving specific miRNAs, and discuss how dysregulation of these miRNAs may result in cleft palate.
Collapse
Affiliation(s)
- Christian Schoen
- Department of Orthodontics and Craniofacial Biology, Radboud University Medical CenterNijmegen, Netherlands
| | - Armaz Aschrafi
- Laboratory of Molecular Biology, Division of Intramural Research Programs, National Institute of Mental Health, National Institutes of HealthBethesda, MD, USA
| | - Michelle Thonissen
- Department of Orthodontics and Craniofacial Biology, Radboud University Medical CenterNijmegen, Netherlands
| | - Geert Poelmans
- Department of Human Genetics, Radboud University Medical CenterNijmegen, Netherlands.,Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical CenterNijmegen, Netherlands.,Department of Molecular Animal Physiology, Donders Institute for Brain, Cognition and Behaviour, Radboud Institute for Molecular Life Sciences, Radboud UniversityNijmegen, Netherlands
| | - Johannes W Von den Hoff
- Department of Orthodontics and Craniofacial Biology, Radboud University Medical CenterNijmegen, Netherlands
| | - Carine E L Carels
- Department of Orthodontics and Craniofacial Biology, Radboud University Medical CenterNijmegen, Netherlands.,Department of Human Genetics, Radboud University Medical CenterNijmegen, Netherlands.,Department of Oral Health Sciences, University Hospitals-KU LeuvenLeuven, Belgium
| |
Collapse
|
30
|
McCarthy N, Liu JS, Richarte AM, Eskiocak B, Lovely CB, Tallquist MD, Eberhart JK. Pdgfra and Pdgfrb genetically interact during craniofacial development. Dev Dyn 2016; 245:641-52. [PMID: 26971580 DOI: 10.1002/dvdy.24403] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 02/15/2016] [Accepted: 02/24/2016] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND One of the most prevalent congenital birth defects is cleft palate. The palatal skeleton is derived from the cranial neural crest and platelet-derived growth factors (Pdgf) are critical in palatogenesis. Of the two Pdgf receptors, pdgfra is required for neural crest migration and palatogenesis. However, the role pdgfrb plays in the neural crest, or whether pdgfra and pdgfrb interact during palatogenesis is unclear. RESULTS We find that pdgfrb is dispensable for craniofacial development in zebrafish. However, the palatal defect in pdgfra;pdgfrb double mutants is significantly more severe than in pdgfra single mutants. Data in mouse suggest this interaction is conserved and that neural crest requires both genes. In zebrafish, pdgfra and pdgfrb are both expressed by neural crest within the pharyngeal arches, and pharmacological analyses demonstrate Pdgf signaling is required at these times. While neither proliferation nor cell death appears affected, time-lapsed confocal analysis of pdgfra;pdgfrb mutants shows a failure of proper neural crest condensation during palatogenesis. CONCLUSIONS We provide data showing that pdgfra and pdgfrb interact during palatogenesis in both zebrafish and mouse. In zebrafish, this interaction affects proper condensation of maxillary neural crest cells, revealing a previously unknown interaction between Pdgfra and Pdgfrb during palate formation. Developmental Dynamics 245:641-652, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Neil McCarthy
- Department of Molecular Biosciences, Institute for Cellular and Molecular Biology, Institute for Neuroscience, Waggoner Center for Alcohol and Addiction Research, University of Texas, Austin, Texas
| | - Jocelyn S Liu
- Center for Cardiovascular Research, University of Hawaii, Honolulu, Hawaii
| | - Alicia M Richarte
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Banu Eskiocak
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - C Ben Lovely
- Department of Molecular Biosciences, Institute for Cellular and Molecular Biology, Institute for Neuroscience, Waggoner Center for Alcohol and Addiction Research, University of Texas, Austin, Texas
| | | | - Johann K Eberhart
- Department of Molecular Biosciences, Institute for Cellular and Molecular Biology, Institute for Neuroscience, Waggoner Center for Alcohol and Addiction Research, University of Texas, Austin, Texas
| |
Collapse
|
31
|
Abstract
The zebrafish model is the only available high-throughput vertebrate assessment system, and it is uniquely suited for studies of in vivo cell biology. A sequenced and annotated genome has revealed a large degree of evolutionary conservation in comparison to the human genome. Due to our shared evolutionary history, the anatomical and physiological features of fish are highly homologous to humans, which facilitates studies relevant to human health. In addition, zebrafish provide a very unique vertebrate data stream that allows researchers to anchor hypotheses at the biochemical, genetic, and cellular levels to observations at the structural, functional, and behavioral level in a high-throughput format. In this review, we will draw heavily from toxicological studies to highlight advances in zebrafish high-throughput systems. Breakthroughs in transgenic/reporter lines and methods for genetic manipulation, such as the CRISPR-Cas9 system, will be comprised of reports across diverse disciplines.
Collapse
Affiliation(s)
- Gloria R Garcia
- Oregon State University, Department of Environmental and Molecular Toxicology, Environmental Health Sciences Center, Corvallis, OR 97331, USA
| | - Pamela D Noyes
- Oregon State University, Department of Environmental and Molecular Toxicology, Environmental Health Sciences Center, Corvallis, OR 97331, USA
| | - Robert L Tanguay
- Oregon State University, Department of Environmental and Molecular Toxicology, Environmental Health Sciences Center, Corvallis, OR 97331, USA.
| |
Collapse
|
32
|
Mukherjee K, Ishii K, Pillalamarri V, Kammin T, Atkin JF, Hickey SE, Xi QJ, Zepeda CJ, Gusella JF, Talkowski ME, Morton CC, Maas RL, Liao EC. Actin capping protein CAPZB regulates cell morphology, differentiation, and neural crest migration in craniofacial morphogenesis†. Hum Mol Genet 2016; 25:1255-70. [PMID: 26758871 DOI: 10.1093/hmg/ddw006] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 01/05/2016] [Indexed: 12/22/2022] Open
Abstract
CAPZB is an actin-capping protein that caps the growing end of F-actin and modulates the cytoskeleton and tethers actin filaments to the Z-line of the sarcomere in muscles. Whole-genome sequencing was performed on a subject with micrognathia, cleft palate and hypotonia that harbored a de novo, balanced chromosomal translocation that disrupts the CAPZB gene. The function of capzb was analyzed in the zebrafish model. capzb(-/-) mutants exhibit both craniofacial and muscle defects that recapitulate the phenotypes observed in the human subject. Loss of capzb affects cell morphology, differentiation and neural crest migration. Differentiation of both myogenic stem cells and neural crest cells requires capzb. During palate morphogenesis, defective cranial neural crest cell migration in capzb(-/-) mutants results in loss of the median cell population, creating a cleft phenotype. capzb is also required for trunk neural crest migration, as evident from melanophores disorganization in capzb(-/-) mutants. In addition, capzb over-expression results in embryonic lethality. Therefore, proper capzb dosage is important during embryogenesis, and regulates both cell behavior and tissue morphogenesis.
Collapse
Affiliation(s)
- Kusumika Mukherjee
- Center for Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Harvard Medical School, Harvard University, Boston, MA 02114, USA
| | - Kana Ishii
- Center for Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Biochemistry and Molecular Biology, Nippon Medical School, Bunkyo, Tokyo 113-0022, Japan, Harvard Medical School, Harvard University, Boston, MA 02114, USA
| | - Vamsee Pillalamarri
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Tammy Kammin
- Department of Obstetrics, Gynecology and Reproductive Biology
| | - Joan F Atkin
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH 43210, USA, Division of Molecular and Human Genetics, Nationwide Children's Hospital, Columbus, OH 43205, USA
| | - Scott E Hickey
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH 43210, USA, Division of Molecular and Human Genetics, Nationwide Children's Hospital, Columbus, OH 43205, USA
| | - Qiongchao J Xi
- Department of Medicine, Division of Genetics, Brigham and Women's Hospital, Boston, MA 02115, USA, Harvard Medical School, Harvard University, Boston, MA 02114, USA
| | | | - James F Gusella
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA 02114, USA, Harvard Medical School, Harvard University, Boston, MA 02114, USA
| | - Michael E Talkowski
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA 02114, USA, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA and Harvard Medical School, Harvard University, Boston, MA 02114, USA
| | - Cynthia C Morton
- Department of Obstetrics, Gynecology and Reproductive Biology, Department of Pathology and Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA and Harvard Medical School, Harvard University, Boston, MA 02114, USA
| | - Richard L Maas
- Department of Medicine, Division of Genetics, Brigham and Women's Hospital, Boston, MA 02115, USA, Harvard Medical School, Harvard University, Boston, MA 02114, USA
| | - Eric C Liao
- Center for Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Harvard Medical School, Harvard University, Boston, MA 02114, USA
| |
Collapse
|
33
|
Rochard LJ, Ling ITC, Kong Y, Liao EC. Visualization of Chondrocyte Intercalation and Directional Proliferation via Zebrabow Clonal Cell Analysis in the Embryonic Meckel's Cartilage. J Vis Exp 2015:e52935. [PMID: 26555721 DOI: 10.3791/52935] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Development of the vertebrate craniofacial structures requires precise coordination of cell migration, proliferation, adhesion and differentiation. Patterning of the Meckel's cartilage, a first pharyngeal arch derivative, involves the migration of cranial neural crest (CNC) cells and the progressive partitioning, proliferation and organization of differentiated chondrocytes. Several studies have described CNC migration during lower jaw morphogenesis, but the details of how the chondrocytes achieve organization in the growth and extension of Meckel's cartilage remains unclear. The sox10 restricted and chemically induced Cre recombinase-mediated recombination generates permutations of distinct fluorescent proteins (RFP, YFP and CFP), thereby creating a multi-spectral labeling of progenitor cells and their progeny, reflecting distinct clonal populations. Using confocal time-lapse photography, it is possible to observe the chondrocytes behavior during the development of the zebrafish Meckel's cartilage. Multispectral cell labeling enables scientists to demonstrate extension of the Meckel's chondrocytes. During extension phase of the Meckel's cartilage, which prefigures the mandible, chondrocytes intercalate to effect extension as they stack in an organized single-cell layered row. Failure of this organized intercalating process to mediate cell extension provides the cellular mechanistic explanation for hypoplastic mandible that we observe in mandibular malformations.
Collapse
Affiliation(s)
- Lucie J Rochard
- Massachusetts General Hospital, Center for Regenerative Medicine, Harvard Medical School
| | - Irving T C Ling
- Massachusetts General Hospital, Center for Regenerative Medicine, Harvard Medical School
| | - Yawei Kong
- Massachusetts General Hospital, Center for Regenerative Medicine, Harvard Medical School
| | - Eric C Liao
- Massachusetts General Hospital, Center for Regenerative Medicine, Harvard Medical School;
| |
Collapse
|
34
|
Abstract
The formation of the face and skull involves a complex series of developmental events mediated by cells derived from the neural crest, endoderm, mesoderm, and ectoderm. Although vertebrates boast an enormous diversity of adult facial morphologies, the fundamental signaling pathways and cellular events that sculpt the nascent craniofacial skeleton in the embryo have proven to be highly conserved from fish to man. The zebrafish Danio rerio, a small freshwater cyprinid fish from eastern India, has served as a popular model of craniofacial development since the 1990s. Unique strengths of the zebrafish model include a simplified skeleton during larval stages, access to rapidly developing embryos for live imaging, and amenability to transgenesis and complex genetics. In this chapter, we describe the anatomy of the zebrafish craniofacial skeleton; its applications as models for the mammalian jaw, middle ear, palate, and cranial sutures; the superior imaging technology available in fish that has provided unprecedented insights into the dynamics of facial morphogenesis; the use of the zebrafish to decipher the genetic underpinnings of craniofacial biology; and finally a glimpse into the most promising future applications of zebrafish craniofacial research.
Collapse
|
35
|
Tavares ALP, Artinger KB, Clouthier DE. Regulating Craniofacial Development at the 3' End: MicroRNAs and Their Function in Facial Morphogenesis. Curr Top Dev Biol 2015; 115:335-75. [PMID: 26589932 DOI: 10.1016/bs.ctdb.2015.08.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Defects in craniofacial development represent a majority of observed human birth defects, occurring at a rate as high as 1:800 live births. These defects often occur due to changes in neural crest cell (NCC) patterning and development and can affect non-NCC-derived structures due to interactions between NCCs and the surrounding cell types. Proper craniofacial development requires an intricate array of gene expression networks that are tightly controlled spatiotemporally by a number of regulatory mechanisms. One of these mechanisms involves the action of microRNAs (miRNAs), a class of noncoding RNAs that repress gene expression by binding to miRNA recognition sequences typically located in the 3' UTR of target mRNAs. Recent evidence illustrates that miRNAs are crucial for vertebrate facial morphogenesis, with changes in miRNA expression leading to facial birth defects, including some in complex human syndromes such as 22q11 (DiGeorge Syndrome). In this review, we highlight the current understanding of miRNA biogenesis, the roles of miRNAs in overall craniofacial development, the impact that loss of miRNAs has on normal development and the requirement for miRNAs in the development of specific craniofacial structures, including teeth. From these studies, it is clear that miRNAs are essential for normal facial development and morphogenesis, and a potential key in establishing new paradigms for repair and regeneration of facial defects.
Collapse
Affiliation(s)
- Andre L P Tavares
- Department of Craniofacial Biology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Kristin B Artinger
- Department of Craniofacial Biology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - David E Clouthier
- Department of Craniofacial Biology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA.
| |
Collapse
|
36
|
Functional analysis of SPECC1L in craniofacial development and oblique facial cleft pathogenesis. Plast Reconstr Surg 2014; 134:748-759. [PMID: 25357034 DOI: 10.1097/prs.0000000000000517] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Oblique facial clefts, also known as Tessier clefts, are severe orofacial clefts, the genetic basis of which is poorly understood. Human genetics studies revealed that disruption in SPECC1L resulted in oblique facial clefts, demonstrating that oblique facial cleft malformation has a genetic basis. An important step toward innovation in treatment of oblique facial clefts would be improved understanding of its genetic pathogenesis. The authors exploit the zebrafish model to elucidate the function of SPECC1L by studying its homolog, specc1lb. METHODS Gene and protein expression analysis was carried out by reverse-transcriptase polymerase chain reaction and immunohistochemistry staining. Morpholino knockdown, mRNA rescue, lineage tracing and terminal deoxynucleotidyl transferase-mediated dUTP nick end-labeling assays were performed for functional analysis. RESULTS Expression of specc1lb was detected in epithelia juxtaposed to chondrocytes. Knockdown of specc1lb resulted in bilateral clefts between median and lateral elements of the ethmoid plate, structures analogous to the frontonasal process and the paired maxillary processes. Lineage tracing analysis revealed that cranial neural crest cells contributing to the frontonasal prominence failed to integrate with the maxillary prominence populations. Cells contributing to lower jaw structures were able to migrate to their destined pharyngeal segment but failed to converge to form mandibular elements. CONCLUSIONS These results demonstrate that specc1lb is required for integration of frontonasal and maxillary elements and convergence of mandibular prominences. The authors confirm the role of SPECC1L in orofacial cleft pathogenesis in the first animal model of Tessier cleft, providing morphogenetic insight into the mechanisms of normal craniofacial development and oblique facial cleft pathogenesis.
Collapse
|
37
|
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
|
38
|
Cohen SP, LaChappelle AR, Walker BS, Lassiter CS. Modulation of estrogen causes disruption of craniofacial chondrogenesis in Danio rerio. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2014; 152:113-120. [PMID: 24747083 DOI: 10.1016/j.aquatox.2014.03.028] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2014] [Revised: 03/30/2014] [Accepted: 03/31/2014] [Indexed: 06/03/2023]
Abstract
Estrogen is a steroid hormone that is ubiquitous in vertebrates, but its role in cartilage formation has not been extensively studied. Abnormalities of craniofacial cartilage and bone account for a large portion of birth defects in the United States. Zebrafish (Danio rerio) have been used as models of human disease, and their transparency in the embryonic period affords additional advantages in studying craniofacial development. In this study, zebrafish embryos were treated with 17-β estradiol (E2) or with an aromatase inhibitor and observed for defects in craniofacial cartilage. Concentrations of E2 greater than 2μM caused major disruptions in cartilage formation. Concentrations below 2μM caused subtle changed in cartilage morphology that were only revealed by measurement. The angles formed by cartilage elements in fish treated with 1.5 and 2μM E2 were increasingly wide, while the length of the primary anterior-posterior cartilage element in these fish decreased significantly from controls. These treatments resulted in fish with shorter, flatter faces as estrogen concentration increased. Inhibition of aromatase activity also resulted in similar craniofacial disruption indicating that careful control of estrogen signaling is required for appropriate development. Further investigation of the phenomena described in this study could lead to a better understanding of the etiology of craniofacial birth defects and endocrine disruption of cartilage formation.
Collapse
Affiliation(s)
- Sarah P Cohen
- Department of Biology, Roanoke College, 221 College Lane, Salem, VA 24153, USA
| | - Adam R LaChappelle
- Department of Biology, Roanoke College, 221 College Lane, Salem, VA 24153, USA
| | - Benjamin S Walker
- Department of Biology, Roanoke College, 221 College Lane, Salem, VA 24153, USA
| | | |
Collapse
|
39
|
Kong Y, Grimaldi M, Curtin E, Dougherty M, Kaufman C, White RM, Zon LI, Liao EC. Neural crest development and craniofacial morphogenesis is coordinated by nitric oxide and histone acetylation. CHEMISTRY & BIOLOGY 2014; 21:488-501. [PMID: 24684905 PMCID: PMC4349424 DOI: 10.1016/j.chembiol.2014.02.013] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Revised: 01/23/2014] [Accepted: 02/10/2014] [Indexed: 11/30/2022]
Abstract
Cranial neural crest (CNC) cells are patterned and coalesce to facial prominences that undergo convergence and extension to generate the craniofacial form. We applied a chemical genetics approach to identify pathways that regulate craniofacial development during embryogenesis. Treatment with the nitric oxide synthase inhibitor 1-(2-[trifluoromethyl] phenyl) imidazole (TRIM) abrogated first pharyngeal arch structures and induced ectopic ceratobranchial formation. TRIM promoted a progenitor CNC fate and inhibited chondrogenic differentiation, which were mediated through impaired nitric oxide (NO) production without appreciable effect on global protein S-nitrosylation. Instead, TRIM perturbed hox gene patterning and caused histone hypoacetylation. Rescue of TRIM phenotype was achieved with overexpression of histone acetyltransferase kat6a, inhibition of histone deacetylase, and complementary NO. These studies demonstrate that NO signaling and histone acetylation are coordinated mechanisms that regulate CNC patterning, differentiation, and convergence during craniofacial morphogenesis.
Collapse
Affiliation(s)
- Yawei Kong
- Center for Regenerative Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Division of Plastic and Reconstructive Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Shriners Hospitals for Children, Boston, MA 02114, USA
| | - Michael Grimaldi
- Center for Regenerative Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Division of Plastic and Reconstructive Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Shriners Hospitals for Children, Boston, MA 02114, USA
| | - Eugene Curtin
- Center for Regenerative Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Division of Plastic and Reconstructive Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Max Dougherty
- Center for Regenerative Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Division of Plastic and Reconstructive Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Charles Kaufman
- Howard Hughes Medical Institute, Children's Hospital Boston, Harvard Medical School, Boston, MA 02115, USA
| | - Richard M White
- Howard Hughes Medical Institute, Children's Hospital Boston, Harvard Medical School, Boston, MA 02115, USA
| | - Leonard I Zon
- Howard Hughes Medical Institute, Children's Hospital Boston, Harvard Medical School, Boston, MA 02115, USA; Harvard Stem Cell Institute, Boston, MA 02114, USA
| | - Eric C Liao
- Center for Regenerative Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Division of Plastic and Reconstructive Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Shriners Hospitals for Children, Boston, MA 02114, USA; Harvard Stem Cell Institute, Boston, MA 02114, USA.
| |
Collapse
|
40
|
Kinikoglu B, Kong Y, Liao EC. Characterization of cultured multipotent zebrafish neural crest cells. Exp Biol Med (Maywood) 2013; 239:159-68. [PMID: 24326414 DOI: 10.1177/1535370213513997] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The neural crest is a unique cell population associated with vertebrate evolution. Neural crest cells (NCCs) are characterized by their multipotent and migratory potentials. While zebrafish is a powerful genetic model organism, the isolation and culture of zebrafish NCCs would provide a useful adjunct to fully interrogate the genetic networks that regulate NCC development. Here we report for the first time the isolation, in vitro culture, and characterization of NCCs from zebrafish embryos. NCCs were isolated from transgenic sox10:egfp embryos using fluorescence activated cell sorting and cultured in complex culture medium without feeder layers. NCC multilineage differentiation was determined by immunocytochemistry and real-time qPCR, cell migration was assessed by wound healing assay, and the proliferation index was calculated by immunostaining against the mitosis marker phospho-histone H3. Cultured NCCs expressed major neural crest lineage markers such as sox10, sox9a, hnk1, p75, dlx2a, and pax3, and the pluripotency markers c-myc and klf4. We showed that the cultured NCCs can be differentiated into multiple neural crest lineages, contributing to neurons, glial cells, smooth muscle cells, melanocytes, and chondrocytes. We applied the NCC in vitro model to study the effect of retinoic acid on NCC development. We showed that retinoic acid had a profound effect on NCC morphology and differentiation, significantly inhibited proliferation and enhanced cell migration. The availability of high numbers of NCCs and reproducible functional assays offers new opportunities for mechanistic studies of neural crest development, in genetic and chemical biology applications.
Collapse
Affiliation(s)
- Beste Kinikoglu
- Center for Regenerative Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | | | | |
Collapse
|
41
|
Gfrerer L, Dougherty M, Liao EC. Visualization of craniofacial development in the sox10: kaede transgenic zebrafish line using time-lapse confocal microscopy. J Vis Exp 2013:e50525. [PMID: 24121214 DOI: 10.3791/50525] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Vertebrate palatogenesis is a highly choreographed and complex developmental process, which involves migration of cranial neural crest (CNC) cells, convergence and extension of facial prominences, and maturation of the craniofacial skeleton. To study the contribution of the cranial neural crest to specific regions of the zebrafish palate a sox10: kaede transgenic zebrafish line was generated. Sox10 provides lineage restriction of the kaede reporter protein to the neural crest, thereby making the cell labeling a more precise process than traditional dye or reporter mRNA injection. Kaede is a photo-convertible protein that turns from green to red after photo activation and makes it possible to follow cells precisely. The sox10: kaede transgenic line was used to perform lineage analysis to delineate CNC cell populations that give rise to maxillary versus mandibular elements and illustrate homology of facial prominences to amniotes. This protocol describes the steps to generate a live time-lapse video of a sox10: kaede zebrafish embryo. Development of the ethmoid plate will serve as a practical example. This protocol can be applied to making a time-lapse confocal recording of any kaede or similar photoconvertible reporter protein in transgenic zebrafish. Furthermore, it can be used to capture not only normal, but also abnormal development of craniofacial structures in the zebrafish mutants.
Collapse
Affiliation(s)
- Lisa Gfrerer
- Center for Regenerative Medicine, Massachusetts General Hospital
| | | | | |
Collapse
|
42
|
Kamel G, Hoyos T, Rochard L, Dougherty M, Kong Y, Tse W, Shubinets V, Grimaldi M, Liao EC. Requirement for frzb and fzd7a in cranial neural crest convergence and extension mechanisms during zebrafish palate and jaw morphogenesis. Dev Biol 2013; 381:423-33. [PMID: 23806211 DOI: 10.1016/j.ydbio.2013.06.012] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2012] [Revised: 06/04/2013] [Accepted: 06/05/2013] [Indexed: 11/29/2022]
Abstract
Regulation of convergence and extension by wnt-frizzled signaling is a common theme in embryogenesis. This study examines the functional requirements of frzb and fzd7a in convergence and extension mechanisms during craniofacial development. Using a morpholino knockdown approach, we found that frzb and fzd7a are dispensable for directed migration of the bilateral trabeculae, but necessary for the convergence and extension of the palatal elements, where the extension process is mediated by chondrocyte proliferation, morphologic change and intercalation. In contrast, frzb and fzd7a are required for convergence of the mandibular prominences, where knockdown of either frzb or fzd7a resulted in complete loss of lower jaw structures. Further, we found that bapx1 was specifically downregulated in the wnt9a/frzb/fzd7a morphants, while general neural crest markers were unaffected. In addition, expression of wnt9a and frzb was also absent in the edn-/- mutant. Notably, over-expression of bapx1 was sufficient to partially rescue mandibular elements in the wnt9a/frzb/fzd7a morphants, demonstrating genetic epistasis of bapx1 acting downstream of edn1 and wnt9a/frzb/fzd7a in lower jaw development. This study underscores the important role of wnt-frizzled signaling in convergence and extension in palate and craniofacial morphogenesis, distinct regulation of upper vs. lower jaw structures, and integration of wnt-frizzled with endothelin signaling to coordinate shaping of the facial form.
Collapse
Affiliation(s)
- George Kamel
- Center for Regenerative Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, United States
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Moon HY, Kim OH, Kim HT, Choi JH, Yeo SY, Kim NS, Park DS, Oh HW, You KH, De Zoysa M, Kim CH. Establishment of a transgenic zebrafish EF1α:Kaede for monitoring cell proliferation during regeneration. FISH & SHELLFISH IMMUNOLOGY 2013; 34:1390-1394. [PMID: 23470815 DOI: 10.1016/j.fsi.2013.02.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2012] [Revised: 02/12/2013] [Accepted: 02/22/2013] [Indexed: 06/01/2023]
Abstract
Zebrafish is considered as a versatile experimental animal for various research models from development to diseases. In this study, we report the development of transgenic zebrafish line named as Tg(EF1α:Kaede) that expresses translation elongation factor 1 subunit alpha (EF1α) promoter linked to a fluorescent protein (FP), Kaede for monitoring proliferating cells in during regeneration. It was revealed that about 1.4 kb 5'-flanking region of the EF1α was sufficient for its promoter activity. Expression of Kaede with a property of photo-conversion from green to red was detected in different embryonic stages as well as various organs such as brain, heart, pancreas, intestine, ovary, and fins of adult fish. Cell proliferation pattern during fin regeneration was monitored after amputation of Tg(EF1α:Kaede) caudal fin and results shown that this system is simple and efficient method for detecting proliferating cells during tissue regeneration. Developed Tg(EF1α:Kaede) line has potential to investigate the cell proliferation, regeneration, wound healing capacities after tissue damage and evaluate the therapeutic power of wound healing drugs.
Collapse
Affiliation(s)
- Hyun-Yi Moon
- Department of Biology, Chungnam National University, Daejeon 305-764, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Lindgren AM, Hoyos T, Talkowski ME, Hanscom C, Blumenthal I, Chiang C, Ernst C, Pereira S, Ordulu Z, Clericuzio C, Drautz JM, Rosenfeld JA, Shaffer LG, Velsher L, Pynn T, Vermeesch J, Harris DJ, Gusella JF, Liao EC, Morton CC. Haploinsufficiency of KDM6A is associated with severe psychomotor retardation, global growth restriction, seizures and cleft palate. Hum Genet 2013; 132:537-52. [PMID: 23354975 PMCID: PMC3627823 DOI: 10.1007/s00439-013-1263-x] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2012] [Accepted: 01/02/2013] [Indexed: 12/17/2022]
Abstract
We describe a female subject (DGAP100) with a 46,X,t(X;5)(p11.3;q35.3)inv(5)(q35.3q35.1)dn, severe psychomotor retardation with hypotonia, global postnatal growth restriction, microcephaly, globally reduced cerebral volume, seizures, facial dysmorphia and cleft palate. Fluorescence in situ hybridization and whole-genome sequencing demonstrated that the X chromosome breakpoint disrupts KDM6A in the second intron. No genes were directly disrupted on chromosome 5. KDM6A is a histone 3 lysine 27 demethylase and a histone 3 lysine 4 methyltransferase. Expression of KDM6A is significantly reduced in DGAP100 lymphoblastoid cells compared to control samples. We identified nine additional cases with neurodevelopmental delay and various other features consistent with the DGAP100 phenotype with copy number variation encompassing KDM6A from microarray databases. We evaluated haploinsufficiency of kdm6a in a zebrafish model. kdm6a is expressed in the pharyngeal arches and ethmoid plate of the developing zebrafish, while a kdm6a morpholino knockdown exhibited craniofacial defects. We conclude KDM6A dosage regulation is associated with severe and diverse structural defects and developmental abnormalities.
Collapse
Affiliation(s)
- Amelia M. Lindgren
- Department of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women’s Hospital, Boston, MA, USA
| | - Tatiana Hoyos
- Department of Plastic and Reconstructive Surgery, Center for Regenerative Medicine, Massachusetts General Hospital, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, USA
| | - Michael E. Talkowski
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA, USA. Departments of Neurology and Genetics, Harvard Medical School, Boston, MA, USA. Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, USA
| | - Carrie Hanscom
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA, USA
| | - Ian Blumenthal
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA, USA
| | - Colby Chiang
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA, USA
| | - Carl Ernst
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA, USA
| | - Shahrin Pereira
- Department of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women’s Hospital, Boston, MA, USA
| | - Zehra Ordulu
- Department of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women’s Hospital, Boston, MA, USA
| | - Carol Clericuzio
- Department of Pediatrics/Genetics, University of New Mexico, Albuquerque, NM, USA
| | - Joanne M. Drautz
- Department of Pediatrics/Genetics, University of New Mexico, Albuquerque, NM, USA
| | | | - Lisa G. Shaffer
- Signature Genomic Laboratories, PerkinElmer Inc., Spokane, WA, USA
| | - Lea Velsher
- Northwestern Ontario Regional Genetics Program, Thunder Bay, ON, Canada
| | - Tania Pynn
- Northwestern Ontario Regional Genetics Program, Thunder Bay, ON, Canada
| | | | - David J. Harris
- Division of Genetics, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - James F. Gusella
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA, USA. Departments of Neurology and Genetics, Harvard Medical School, Boston, MA, USA. Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, USA. Autism Consortium of Boston, Boston, MA, USA
| | - Eric C. Liao
- Department of Plastic and Reconstructive Surgery, Center for Regenerative Medicine, Massachusetts General Hospital, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, USA
| | - Cynthia C. Morton
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, USA. Departments of Obstetrics, Gynecology and Reproductive Biology and Pathology, Brigham and Women’s Hospital and Harvard Medical School, New Research Building, Room 160D, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| |
Collapse
|
45
|
Dougherty M, Kamel G, Grimaldi M, Gfrerer L, Shubinets V, Ethier R, Hickey G, Cornell RA, Liao EC. Distinct requirements for wnt9a and irf6 in extension and integration mechanisms during zebrafish palate morphogenesis. Development 2012; 140:76-81. [PMID: 23154410 DOI: 10.1242/dev.080473] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Development of the palate in vertebrates involves cranial neural crest migration, convergence of facial prominences and extension of the cartilaginous framework. Dysregulation of palatogenesis results in orofacial clefts, which represent the most common structural birth defects. Detailed analysis of zebrafish palatogenesis revealed distinct mechanisms of palatal morphogenesis: extension, proliferation and integration. We show that wnt9a is required for palatal extension, wherein the chondrocytes form a proliferative front, undergo morphological change and intercalate to form the ethmoid plate. Meanwhile, irf6 is required specifically for integration of facial prominences along a V-shaped seam. This work presents a mechanistic analysis of palate morphogenesis in a clinically relevant context.
Collapse
Affiliation(s)
- Max Dougherty
- Center for Regenerative Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | | | | | | | | | | | | | | | | |
Collapse
|