1
|
Carroll SH, Schafer S, Dalessandro E, Ho TV, Chai Y, Liao EC. Neural crest and periderm-specific requirements of Irf6 during neural tube and craniofacial development. Dev Biol 2025; 522:106-115. [PMID: 40113028 PMCID: PMC12065081 DOI: 10.1016/j.ydbio.2025.03.006] [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: 05/10/2024] [Revised: 11/27/2024] [Accepted: 03/10/2025] [Indexed: 03/22/2025]
Abstract
IRF6 is a key genetic determinant of cleft lip and palate. The ability to interrogate post-embryonic requirements of Irf6 has been hindered, as global Irf6 ablation in the mouse causes neonatal lethality. Prior work analyzing Irf6 in mice defined its role in the embryonic surface epithelium and periderm, where it regulates cell proliferation and differentiation. Several reports have also described Irf6 expression in other cell types, such as muscle, and neuroectoderm. However, analysis of a functional role in non-epithelial cells has been incomplete due to the severity and lethality of the Irf6 knockout model and the paucity of work with a conditional Irf6 allele. Here we describe the generation and characterization of a new Irf6 floxed mouse model and analysis of Irf6 ablation in periderm and neural crest lineages. This work found that loss of Irf6 in periderm recapitulates a mild Irf6 null phenotype, suggesting that Irf6-mediated signaling in periderm plays a crucial role in regulating embryonic development. Further, conditional ablation of Irf6 in neural crest cells resulted in an anterior neural tube defect of variable penetrance. The generation of this conditional Irf6 allele allows for new insights into craniofacial development and new exploration into the post-natal role of Irf6.
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
| | - Sogand Schafer
- Center for Craniofacial Innovation, Children's Hospital of Philadelphia Research Institute, Children's Hospital of Philadelphia, PA, 19104, USA
| | - Eileen Dalessandro
- Center for Craniofacial Innovation, Children's Hospital of Philadelphia Research Institute, Children's Hospital of Philadelphia, PA, 19104, USA
| | - Thach-Vu Ho
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA, USA
| | - Yang Chai
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA, 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
|
2
|
Robinson K, Singh SK, Walkup RB, Fawwal DV, Adeyemo WL, Beaty TH, Butali A, Buxó CJ, Chung WK, Cutler DJ, Epstein MP, Fashina A, Gasser B, Gowans LJJ, Hecht JT, Uribe LM, Scott DA, Shaw GM, Thomas MA, Weinberg SM, Brand H, Marazita ML, Lipinski RJ, Murray JC, Cornell RA, Leslie-Clarkson EJ. Rare variants in PRKCI cause Van der Woude syndrome and other features of peridermopathy. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.01.17.25320742. [PMID: 39867391 PMCID: PMC11759255 DOI: 10.1101/2025.01.17.25320742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Van der Woude syndrome (VWS) is an autosomal dominant disorder characterized by lower lip pits and orofacial clefts (OFCs). With a prevalence of approximately 1 in 35,000 live births, it is the most common form of syndromic clefting and may account for ~2% of all OFCs. The majority of VWS is attributed to genetic variants in IRF6 (~70%) or GRHL3 (~5%), leaving up to 25% of individuals with VWS without a molecular diagnosis. Both IRF6 and GRHL3 function in a transcriptional regulatory network governing differentiation of periderm, a single layer of epithelial cells that prevents pathological adhesions during palatogenesis. Disruption of this layer results in a spectrum of phenotypes ranging from lip pits and OFCs to severe pterygia and other congenital anomalies that can be incompatible with life. Understanding the mechanisms of peridermopathies is vital in improving health outcomes for affected individuals. We reasoned that genes encoding additional members of the periderm gene regulatory network, including kinases acting upstream of IRF6 (i.e., atypical protein kinase C family members, RIPK4, and CHUK), are candidates to harbor variants resulting in VWS. Consistent with this prediction, we identified 6 de novo variants (DNs) and 11 rare variants in PRKCI, an atypical protein kinase C, in 17 individuals with clinical features consistent with syndromic OFCs and peridermopathies. Of the identified DNs, 4 were identical p.(Asn383Ser) variants in unrelated individuals with syndromic OFCs, indicating a likely hotspot mutation. We also performed functional validation of 12 variants using the enveloping layer in zebrafish embryos, a structure analogous to the periderm. Three patient-specific alleles (p.Arg130His, p.(Asn383Ser), and p.Leu385Phe) were confirmed to be loss-of-function variants. In summary, we identified PRKCI as a novel causal gene for VWS and syndromic OFC with other features of peridermopathies.
Collapse
Affiliation(s)
- Kelsey Robinson
- Department of Human Genetics, Emory University, Atlanta, GA, USA
| | - Sunil K. Singh
- Department of Oral Health Sciences, University of Washington, Seattle, WA, USA
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Rachel B Walkup
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | | | - Wasiu Lanre Adeyemo
- Department of Oral and Maxillofacial Surgery, College of Medicine, University of Lagos, Lagos, Nigeria
| | - Terri H. Beaty
- Department of Epidemiology, Johns Hopkins University, Baltimore, MD, USA
| | - Azeez Butali
- Department of Oral Biology, Radiology, and Medicine, University of Iowa, Iowa City, IA, USA
| | - Carmen J. Buxó
- School of Dental Medicine, University of Puerto Rico, San Juan, Puerto Rico, USA
| | - Wendy K. Chung
- Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - David J. Cutler
- Department of Human Genetics, Emory University, Atlanta, GA, USA
| | | | - Azeez Fashina
- Department of Oral Health Sciences, University of Washington, Seattle, WA, USA
| | | | - Lord JJ Gowans
- Department of Biochemistry and Biotechnology, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Jacqueline T. Hecht
- Department of Pediatrics, McGovern Medical School University of Texas Health at Houston, Houston, TX, USA
| | - Lina Moreno Uribe
- Department of Orthodontics & The Iowa Institute for Oral Health Research, University of Iowa, Iowa City, IA, USA
| | - Daryl A. Scott
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Gary M. Shaw
- Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Mary Ann Thomas
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Seth M. Weinberg
- Center for Craniofacial and Dental Genetics, Department of Oral and Craniofacial Sciences, School of Dental Medicine, and Department of Human Genetics, School of Public Health, University of Pittsburgh, PA, USA
| | - Harrison Brand
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Mary L. Marazita
- Center for Craniofacial and Dental Genetics, Department of Oral and Craniofacial Sciences, School of Dental Medicine, and Department of Human Genetics, School of Public Health, University of Pittsburgh, PA, USA
| | - Robert J Lipinski
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | | | - Robert A. Cornell
- Department of Oral Health Sciences, University of Washington, Seattle, WA, USA
| | | |
Collapse
|
3
|
Murali H, Wang P, Liao EC, Wang K. Genetic variant classification by predicted protein structure: A case study on IRF6. Comput Struct Biotechnol J 2024; 23:892-904. [PMID: 38370976 PMCID: PMC10869248 DOI: 10.1016/j.csbj.2024.01.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Revised: 01/24/2024] [Accepted: 01/25/2024] [Indexed: 02/20/2024] Open
Abstract
Next-generation genome sequencing has revolutionized genetic testing, identifying numerous rare disease-associated gene variants. However, to impute pathogenicity, computational approaches remain inadequate and functional testing of gene variant is required to provide the highest level of evidence. The emergence of AlphaFold2 has transformed the field of protein structure determination, and here we outline a strategy that leverages predicted protein structure to enhance genetic variant classification. We used the gene IRF6 as a case study due to its clinical relevance, its critical role in cleft lip/palate malformation, and the availability of experimental data on the pathogenicity of IRF6 gene variants through phenotype rescue experiments in irf6-/- zebrafish. We compared results from over 30 pathogenicity prediction tools on 37 IRF6 missense variants. IRF6 lacks an experimentally derived structure, so we used predicted structures to explore associations between mutational clustering and pathogenicity. We found that among these variants, 19 of 37 were unanimously predicted as deleterious by computational tools. Comparing in silico predictions with experimental findings, 12 variants predicted as pathogenic were experimentally determined as benign. Even with the recently published AlphaMissense model, 15/18 (83%) of the predicted pathogenic variants were experimentally determined as benign. In comparison, mapping variants to the protein revealed deleterious mutation clusters around the protein binding domain, whereas N-terminal variants tend to be benign, suggesting the importance of structural information in determining pathogenicity of mutations in this gene. In conclusion, incorporating gene-specific structural features of known pathogenic/benign mutations may provide meaningful insights into pathogenicity predictions in a gene-specific manner and facilitate the interpretation of variant pathogenicity.
Collapse
Affiliation(s)
- Hemma Murali
- Graduate Program in Biochemistry and Molecular Biophysics, University of Pennsylvania, Philadelphia, PA 19104, United States
- Center for Cellular and Molecular Therapeutics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, United States
| | - Peng Wang
- Center for Cellular and Molecular Therapeutics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, United States
- Master of Biotechnology Program, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Eric C. Liao
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
- Center for Craniofacial Innovation, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, United States
| | - Kai Wang
- Center for Cellular and Molecular Therapeutics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, United States
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| |
Collapse
|
4
|
Caetano da Silva C, Macias Trevino C, Mitchell J, Murali H, Tsimbal C, Dalessandro E, Carroll SH, Kochhar S, Curtis SW, Cheng CHE, Wang F, Kutschera E, Carstens RP, Xing Y, Wang K, Leslie EJ, Liao EC. Functional analysis of ESRP1/2 gene variants and CTNND1 isoforms in orofacial cleft pathogenesis. Commun Biol 2024; 7:1040. [PMID: 39179789 PMCID: PMC11344038 DOI: 10.1038/s42003-024-06715-3] [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: 02/15/2024] [Accepted: 08/09/2024] [Indexed: 08/26/2024] Open
Abstract
Orofacial cleft (OFC) is a common human congenital anomaly. Epithelial-specific RNA splicing regulators ESRP1 and ESRP2 regulate craniofacial morphogenesis and their disruption result in OFC in zebrafish, mouse and humans. Using esrp1/2 mutant zebrafish and murine Py2T cell line models, we functionally tested the pathogenicity of human ESRP1/2 gene variants. We found that many variants predicted by in silico methods to be pathogenic were functionally benign. Esrp1 also regulates the alternative splicing of Ctnnd1 and these genes are co-expressed in the embryonic and oral epithelium. In fact, over-expression of ctnnd1 is sufficient to rescue morphogenesis of epithelial-derived structures in esrp1/2 zebrafish mutants. Additionally, we identified 13 CTNND1 variants from genome sequencing of OFC cohorts, confirming CTNND1 as a key gene in human OFC. This work highlights the importance of functional assessment of human gene variants and demonstrates the critical requirement of Esrp-Ctnnd1 acting in the embryonic epithelium to regulate palatogenesis.
Collapse
Affiliation(s)
- Caroline Caetano da Silva
- Center for Craniofacial Innovation, Division of Plastic and Reconstructive Surgery, Department of Surgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | | | | | - Hemma Murali
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Casey Tsimbal
- Center for Craniofacial Innovation, Division of Plastic and Reconstructive Surgery, Department of Surgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Shriners Hospital for Children, Tampa, FL, USA
| | - Eileen Dalessandro
- Center for Craniofacial Innovation, Division of Plastic and Reconstructive Surgery, Department of Surgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Shannon H Carroll
- Center for Craniofacial Innovation, Division of Plastic and Reconstructive Surgery, Department of Surgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Shriners Hospital for Children, Tampa, FL, USA
| | - Simren Kochhar
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Sarah W Curtis
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Ching Hsun Eric Cheng
- Center for Craniofacial Innovation, Division of Plastic and Reconstructive Surgery, Department of Surgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Feng Wang
- Center for Genomic Medicine, Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Eric Kutschera
- Center for Genomic Medicine, Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Russ P Carstens
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Yi Xing
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Center for Genomic Medicine, Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Kai Wang
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Elizabeth J Leslie
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Eric C Liao
- Center for Craniofacial Innovation, Division of Plastic and Reconstructive Surgery, Department of Surgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Harvard Medical School, Boston, MA, USA.
- Shriners Hospital for Children, Tampa, FL, USA.
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
5
|
da Silva CC, Trevino CM, Mitchell J, Murali H, Tsimbal C, Dalessandro E, Carroll SH, Kochhar S, Curtis SW, Cheng CHE, Wang F, Kutschera E, Carstens RP, Xing Y, Wang K, Leslie EJ, Liao EC. Functional analysis of ESRP1/2 gene variants and CTNND1 isoforms in orofacial cleft pathogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.02.601574. [PMID: 39005284 PMCID: PMC11245018 DOI: 10.1101/2024.07.02.601574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Orofacial cleft (OFC) is a common human congenital anomaly. Epithelial-specific RNA splicing regulators ESRP1 and ESRP2 regulate craniofacial morphogenesis and their disruption result in OFC in zebrafish, mouse and humans. Using esrp1/2 mutant zebrafish and murine Py2T cell line models, we functionally tested the pathogenicity of human ESRP1/2 gene variants. We found that many variants predicted by in silico methods to be pathogenic were functionally benign. Esrp1 also regulates the alternative splicing of Ctnnd1 and these genes are co-expressed in the embryonic and oral epithelium. In fact, over-expression of ctnnd1 is sufficient to rescue morphogenesis of epithelial-derived structures in esrp1/2 zebrafish mutants. Additionally, we identified 13 CTNND1 variants from genome sequencing of OFC cohorts, confirming CTNND1 as a key gene in human OFC. This work highlights the importance of functional assessment of human gene variants and demonstrates the critical requirement of Esrp-Ctnnd1 acting in the embryonic epithelium to regulate palatogenesis.
Collapse
Affiliation(s)
- Caroline Caetano da Silva
- Center for Craniofacial Innovation, Division of Plastic and Reconstructive Surgery, Department of Surgery, Children’s Hospital of Philadelphia, PA, USA
| | | | | | - Hemma Murali
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Casey Tsimbal
- Center for Craniofacial Innovation, Division of Plastic and Reconstructive Surgery, Department of Surgery, Children’s Hospital of Philadelphia, PA, USA
- Shriners Hospital for Children, Tampa, FL, USA
| | - Eileen Dalessandro
- Center for Craniofacial Innovation, Division of Plastic and Reconstructive Surgery, Department of Surgery, Children’s Hospital of Philadelphia, PA, USA
| | - Shannon H. Carroll
- Center for Craniofacial Innovation, Division of Plastic and Reconstructive Surgery, Department of Surgery, Children’s Hospital of Philadelphia, PA, USA
- Shriners Hospital for Children, Tampa, FL, USA
| | - Simren Kochhar
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Sarah W. Curtis
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Ching Hsun Eric Cheng
- Center for Craniofacial Innovation, Division of Plastic and Reconstructive Surgery, Department of Surgery, Children’s Hospital of Philadelphia, PA, USA
| | - Feng Wang
- Center for Genomic Medicine, Department of Biomedical and Health Informatics, Children’s Hospital of Philadelphia, PA, USA
| | - Eric Kutschera
- Center for Genomic Medicine, Department of Biomedical and Health Informatics, Children’s Hospital of Philadelphia, PA, USA
| | - Russ P. Carstens
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Yi Xing
- Center for Genomic Medicine, Department of Biomedical and Health Informatics, Children’s Hospital of Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Kai Wang
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Elizabeth J. Leslie
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Eric C. Liao
- Center for Craniofacial Innovation, Division of Plastic and Reconstructive Surgery, Department of Surgery, Children’s Hospital of Philadelphia, PA, USA
- Harvard Medical School, Boston, MA, USA
- Shriners Hospital for Children, Tampa, FL, USA
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
6
|
Yang CW, Yin B, Shi JY, Shi B, Jia ZL. Causal Variations at IRF6 Gene Identified in Van der Woude Syndrome Pedigrees. Cleft Palate Craniofac J 2024; 61:1134-1142. [PMID: 36866619 DOI: 10.1177/10556656231157575] [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: 03/04/2023] Open
Abstract
The purpose of this study is to analyze the clinical characteristics of patients with Van der Woude syndrome (VWS) and to detect variations in each patient. Finally, the combination of genotype and phenotype can make a clear diagnosis of VWS patients with different phenotype penetrance. Five Chinese VWS pedigree were enrolled. Whole exome sequencing of the proband was performed, and the potential pathogenic variation was further verified by Sanger sequencing in the patient and their parents. The human mutant IRF6 coding sequence was generated from the human full-length IRF6 plasmid by site-directed mutagenesis and cloned into the GV658 vector, RT-qPCR and Western blot were used to detect the expression of IRF6. We found one de novo nonsense variation (p. Gln118Ter) and three novel missense variations (p. Gly301Glu, p. Gly267Ala, and p. Glu404Gly) co-segregated with VWS. RT-qPCR analysis revealed that p. Glu404Gly significantly reduced the expression level of IRF6 mRNA. Western blot of cell lysates confirmed that IRF6 p. Glu404Gly abundance levels were lower than those for IRF6 wild type. This discovery of the novel variation (IRF6 p. Glu404Gly) expands the spectrum of known variations in VWS in Chinese humans. Genetic results combined with clinical phenotypes and differential diagnosis points from other diseases can make a definitive diagnosis and provide genetic counseling for families.
Collapse
Affiliation(s)
- Cheng-Wei Yang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of cleft lip and palate, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Bin Yin
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of cleft lip and palate, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jia-Yu Shi
- Division of Growth and Development and Section of Orthodontics, School of Dentistry, University of California, Los Angeles, USA
| | - Bing Shi
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of cleft lip and palate, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Zhong-Lin Jia
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of cleft lip and palate, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
7
|
Carroll SH, Schafer S, Dalessandro E, Ho TV, Chai Y, Liao EC. Neural crest and periderm-specific requirements of Irf6 during neural tube and craniofacial development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.11.598425. [PMID: 38915513 PMCID: PMC11195129 DOI: 10.1101/2024.06.11.598425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
IRF6 is a key genetic determinant of syndromic and non-syndromic cleft lip and palate. The ability to interrogate post-embryonic requirements of Irf6 has been hindered, as global Irf6 ablation in the mouse causes neonatal lethality. Prior work analyzing Irf6 in mouse models defined its role in the embryonic surface epithelium and periderm where it is required to regulate cell proliferation and differentiation. Several reports have also described Irf6 gene expression in other cell types, such as muscle, and neuroectoderm. However, analysis of a functional role in non-epithelial cell lineages has been incomplete due to the severity and lethality of the Irf6 knockout model and the paucity of work with a conditional Irf6 allele. Here we describe the generation and characterization of a new Irf6 floxed mouse model and analysis of Irf6 ablation in periderm and neural crest lineages. This work found that loss of Irf6 in periderm recapitulates a mild Irf6 null phenotype, suggesting that Irf6-mediated signaling in periderm plays a crucial role in regulating embryonic development. Further, conditional ablation of Irf6 in neural crest cells resulted in an anterior neural tube defect of variable penetrance. The generation of this conditional Irf6 allele allows for new insights into craniofacial development and new exploration into the post-natal role of Irf6.
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
| | - Sogand Schafer
- Center for Craniofacial Innovation, Children's Hospital of Philadelphia Research Institute, Children's Hospital of Philadelphia, PA 19104, USA
| | - Eileen Dalessandro
- Center for Craniofacial Innovation, Children's Hospital of Philadelphia Research Institute, Children's Hospital of Philadelphia, PA 19104, USA
| | - Thach-Vu Ho
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA USA
| | - Yang Chai
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA 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
|
8
|
Alhazmi N, Alamoud KA, Albalawi F, Alalola B, Farook FF. The application of zebrafish model in the study of cleft lip and palate development: A systematic review. Heliyon 2024; 10:e28322. [PMID: 38533046 PMCID: PMC10963633 DOI: 10.1016/j.heliyon.2024.e28322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 03/15/2024] [Accepted: 03/15/2024] [Indexed: 03/28/2024] Open
Abstract
Objective Craniofacial growth and development are more than a scientific curiosity; it is of tremendous interest to clinicians. Insights into the genetic etiology of cleft lip and palate development are essential for improving diagnosis and treatment planning. The purpose of this systematic review was to utilize a zebrafish model to highlight the role of the IRF6 gene in cleft lip and palate development in humans. Data This review adhered to the guidelines outlined in the PRISMA statement. Nine studies were included in the analysis. Sources This study used major scientific databases such as MEDLINE, EMBASE, Web of Science, and the Zebrafish Information Network and yielded 1275 articles. Two reviewers performed the screening using COVIDENCE™ independently, and a third reviewer resolved any conflicts. Study selection After applying the inclusion and exclusion criteria and screening, nine studies were included in the analysis. The Systematic Review Center for Laboratory Animal Experimentation's (SYRCLE's) risk-of-bias tool was used to assess the quality of the included studies. Results The main outcome supports the role of the IRF6 gene in zebrafish periderm development and embryogenesis, and IRF6 variations result in cleft lip and palate development. The overall SYRCLE risk of bias was low-medium. Conclusion In conclusion, this review indicated the critical role of the IRF6 gene and its downstream genes (GRHL3, KLF17, and ESRP1/2) in the development of cleft lip and palate in zebrafish models. Genetic mutation zebrafish models provide a high level of insights into zebrafish craniofacial development. Clinical relevance this review provides a productive avenue for understanding the powerful and conserved zebrafish model for investigating the pathogenesis of human cleft lip and palate.
Collapse
Affiliation(s)
- Nora Alhazmi
- Preventive Dental Science Department, College of Dentistry, King Saud bin Abdulaziz University for Health Sciences, Riyadh, 14611, Saudi Arabia
- King Abdullah International Medical Research Center, Riyadh, 11481, Saudi Arabia
- Ministry of the National Guard Health Affairs, Riyadh, 11426, Saudi Arabia
| | - Khalid A. Alamoud
- Preventive Dental Science Department, College of Dentistry, King Saud bin Abdulaziz University for Health Sciences, Riyadh, 14611, Saudi Arabia
- King Abdullah International Medical Research Center, Riyadh, 11481, Saudi Arabia
- Ministry of the National Guard Health Affairs, Riyadh, 11426, Saudi Arabia
| | - Farraj Albalawi
- Preventive Dental Science Department, College of Dentistry, King Saud bin Abdulaziz University for Health Sciences, Riyadh, 14611, Saudi Arabia
- King Abdullah International Medical Research Center, Riyadh, 11481, Saudi Arabia
- Ministry of the National Guard Health Affairs, Riyadh, 11426, Saudi Arabia
| | - Bassam Alalola
- Preventive Dental Science Department, College of Dentistry, King Saud bin Abdulaziz University for Health Sciences, Riyadh, 14611, Saudi Arabia
- King Abdullah International Medical Research Center, Riyadh, 11481, Saudi Arabia
- Ministry of the National Guard Health Affairs, Riyadh, 11426, Saudi Arabia
| | - Fathima F. Farook
- Preventive Dental Science Department, College of Dentistry, King Saud bin Abdulaziz University for Health Sciences, Riyadh, 14611, Saudi Arabia
- King Abdullah International Medical Research Center, Riyadh, 11481, Saudi Arabia
- Ministry of the National Guard Health Affairs, Riyadh, 11426, Saudi Arabia
| |
Collapse
|
9
|
Li MJ, Kumari P, Lin YS, Yao ML, Zhang BH, Yin B, Duan SJ, Cornell R, Marazita M, Shi B, Jia ZL. A Variant in the IRF6 Promoter Associated with the Risk for Orofacial Clefting. J Dent Res 2023; 102:806-813. [PMID: 37161310 PMCID: PMC10399074 DOI: 10.1177/00220345231165210] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/11/2023] Open
Abstract
The single-nucleotide polymorphism (SNP) rs2235371 (IRF6 V274I) is associated with nonsyndromic cleft lip with or without cleft palate (NSCL/P) in Han Chinese and other populations but appears to be without a functional effect. To find the common etiologic variant or variants within the haplotype tagged by rs2235371, we carried out targeted sequencing of an interval containing IRF6 in 159 Han Chinese with NSCL/P. This study revealed that the SNP rs12403599, within the IRF6 promoter, is associated with all phenotypes of NSCL/P, especially nonsyndromic cleft lip (NSCLO) and a subphenotype of it, microform cleft lip (MCL). This association was replicated in 2 additional much larger cohorts of cases and controls from the Han Chinese. Conditional logistic analysis indicated that association of rs2235371 with NSCL/P was lost if rs12403599 was excluded. rs12403599 contributes the most risk to MCL: its G allele is responsible for 38.47% of the genetic contribution to MCL, and the odds ratios of G/C and G/G genotypes were 2.91 and 6.58, respectively, for MCL. To test if rs12403599 is functional, we carried out reporter assays in a fetal oral epithelium cells (GMSM-K). Unexpectedly, the risk allele G yielded higher promoter activity in GMSM-K. Consistent with the reporter studies, expression of IRF6 in lip tissues from NSCLO and MCL patients with the G/G phenotype was higher than in those from patients with the C/C phenotype. These results indicate that rs12403599 is tagging the risk haplotype for NSCL/P better than rs2235371 in Han Chinese and supports investigation of the mechanisms by which the allele of rs12403599 affects IRF6 expression and tests of this association in different populations.
Collapse
Affiliation(s)
- M.-J. Li
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Cleft Lip and Palate, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - P. Kumari
- Department of Oral Health Sciences, University of Washington, Seattle, WA, USA
| | - Y.-S. Lin
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Cleft Lip and Palate, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - M.-L. Yao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Cleft Lip and Palate, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - B.-H. Zhang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Cleft Lip and Palate, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - B. Yin
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Cleft Lip and Palate, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - S.-J. Duan
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Cleft Lip and Palate, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - R.A. Cornell
- Department of Oral Health Sciences, University of Washington, Seattle, WA, USA
| | - M.L. Marazita
- Centre for Craniofacial and Dental Genetics, Department of Oral Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - B. Shi
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Cleft Lip and Palate, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Z.-l. Jia
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Cleft Lip and Palate, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
10
|
Ochenkowska K, Herold A, Samarut É. Zebrafish Is a Powerful Tool for Precision Medicine Approaches to Neurological Disorders. Front Mol Neurosci 2022; 15:944693. [PMID: 35875659 PMCID: PMC9298522 DOI: 10.3389/fnmol.2022.944693] [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: 05/15/2022] [Accepted: 06/17/2022] [Indexed: 12/17/2022] Open
Abstract
Personalized medicine is currently one of the most promising tools which give hope to patients with no suitable or no available treatment. Patient-specific approaches are particularly needed for common diseases with a broad phenotypic spectrum as well as for rare and yet-undiagnosed disorders. In both cases, there is a need to understand the underlying mechanisms and how to counteract them. Even though, during recent years, we have been observing the blossom of novel therapeutic techniques, there is still a gap to fill between bench and bedside in a patient-specific fashion. In particular, the complexity of genotype-to-phenotype correlations in the context of neurological disorders has dampened the development of successful disease-modifying therapeutics. Animal modeling of human diseases is instrumental in the development of therapies. Currently, zebrafish has emerged as a powerful and convenient model organism for modeling and investigating various neurological disorders. This model has been broadly described as a valuable tool for understanding developmental processes and disease mechanisms, behavioral studies, toxicity, and drug screening. The translatability of findings obtained from zebrafish studies and the broad prospect of human disease modeling paves the way for developing tailored therapeutic strategies. In this review, we will discuss the predictive power of zebrafish in the discovery of novel, precise therapeutic approaches in neurosciences. We will shed light on the advantages and abilities of this in vivo model to develop tailored medicinal strategies. We will also investigate the newest accomplishments and current challenges in the field and future perspectives.
Collapse
Affiliation(s)
- Katarzyna Ochenkowska
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montreal, QC, Canada.,Department of Neuroscience, Université de Montréal, Montreal, QC, Canada
| | - Aveeva Herold
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montreal, QC, Canada.,Department of Neuroscience, Université de Montréal, Montreal, QC, Canada
| | - Éric Samarut
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montreal, QC, Canada.,Department of Neuroscience, Université de Montréal, Montreal, QC, Canada.,Modelis Inc., Montreal, QC, Canada
| |
Collapse
|
11
|
Huang W, He Q, Li M, Ding Y, Liang W, Li W, Lin J, Zhao H, Chen F. Two rare variants reveal the significance of Grainyhead‐like 3 Arginine 391 underlying non‐syndromic cleft palate only. Oral Dis 2022; 29:1632-1643. [PMID: 35189007 DOI: 10.1111/odi.14164] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 02/07/2022] [Accepted: 02/12/2022] [Indexed: 11/27/2022]
Abstract
OBJECTIVES Non-syndromic cleft palate only (NSCPO) is one of the most common craniofacial birth defects with largely undetermined genetic etiology. It has been established that Grainyhead-like 3 (GRHL3) plays an essential role in the pathogenesis of NSCPO. This study aimed to identify and verify the first-reported GRHL3 variant underlying NSCPO among the Chinese cohort. METHODS We performed whole-exome sequencing (WES) on a Chinese NSCPO patient and identified a rare variant of GRHL3 (p.Arg391His). A validated deleterious variant p.Arg391Cys was introduced as a positive control. Zebrafish embryos injection, reporter assays, live-cell imaging, and RNA sequencing were conducted to test the pathogenicity of the variants. RESULTS Zebrafish embryos microinjection demonstrated that overexpression of the variants could disrupt the normal development of zebrafish embryos. Reporter assays showed that Arg391His disturbed transcriptional activity of GRHL3 and exerted a dominant-negative effect. Interestingly, Arg391His and Arg391Cys displayed distinct nuclear localization patterns from that of wild-type GRHL3 in live-cell imaging. Bulk RNA sequencing suggested that the two variants changed the pattern of gene expression. CONCLUSIONS In aggregate, this study identified and characterized a rare GRHL3 variant in NSCPO, revealing the critical role of Arginine 391 in GRHL3. Our findings will help facilitate understanding and genetic counseling of NSCPO.
Collapse
Affiliation(s)
- Wenbin Huang
- Department of Orthodontics Peking University School and Hospital of Stomatology 100081 Beijing China
- National Center of Stomatology National Clinical Research Center for Oral Diseases National Engineering Laboratory for Digital and Material Technology of Stomatology Beijing Key Laboratory for Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials 100081 Beijing China
| | - Qing He
- Department of Physiology and Pathophysiology School of Basic Medical Sciences Xi’an Jiaotong University Health Science Center 710061 Xi’an, Shaanxi China
| | - Mingzhao Li
- Department of Orthodontics Peking University School and Hospital of Stomatology 100081 Beijing China
- National Center of Stomatology National Clinical Research Center for Oral Diseases National Engineering Laboratory for Digital and Material Technology of Stomatology Beijing Key Laboratory for Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials 100081 Beijing China
| | - Yi Ding
- Department of Physiology and Pathophysiology School of Basic Medical Sciences Xi’an Jiaotong University Health Science Center 710061 Xi’an, Shaanxi China
| | - Wei Liang
- Department of Orthodontics Peking University School and Hospital of Stomatology 100081 Beijing China
- National Center of Stomatology National Clinical Research Center for Oral Diseases National Engineering Laboratory for Digital and Material Technology of Stomatology Beijing Key Laboratory for Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials 100081 Beijing China
| | - Weiran Li
- Department of Orthodontics Peking University School and Hospital of Stomatology 100081 Beijing China
- National Center of Stomatology National Clinical Research Center for Oral Diseases National Engineering Laboratory for Digital and Material Technology of Stomatology Beijing Key Laboratory for Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials 100081 Beijing China
| | - Jiuxiang Lin
- Department of Orthodontics Peking University School and Hospital of Stomatology 100081 Beijing China
- National Center of Stomatology National Clinical Research Center for Oral Diseases National Engineering Laboratory for Digital and Material Technology of Stomatology Beijing Key Laboratory for Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials 100081 Beijing China
| | - Huaxiang Zhao
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research College of Stomatology Xi’an Jiaotong University 710004 Xi'an, Shaanxi China
- Department of Orthodontics College of Stomatology Xi’an Jiaotong University 710004 Xi’an, Shaanxi China
| | - Feng Chen
- Central laboratory Peking University School and Hospital of Stomatology 100081 Beijing China
- National Center of Stomatology National Clinical Research Center for Oral Diseases National Engineering Laboratory for Digital and Material Technology of Stomatology Beijing Key Laboratory for Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials 100081 Beijing China
| |
Collapse
|
12
|
Yin B, Shi JY, Shi B, Zheng Q, Jia ZL. Association Between SNPs in 1q32.2 and NSCL ± P in Han Chinese Population. Cleft Palate Craniofac J 2021; 59:1520-1526. [PMID: 34730443 DOI: 10.1177/10556656211052837] [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: 02/05/2023] Open
Abstract
OBJECTIVES Non-syndromic cleft lip with or without cleft palate (NSCL ± P) is one of the most common birth malformations. Currently, numerous susceptibility SNPs have been reported by GWA studies, however, the replications of them among NSCL ± P from Han Chinese were very limited. DESIGN In this study, we selected 16 SNPs around 1q32.2 based on the published GWA studies and replicated them among 302 trios with NSCL ± P from Han Chinese Population. The genotypic data was analyzed with FBAT, PLINK and R package. SETTING The study was conducted in a tertiary medical center. PATIENTS, PARTICIPANTS 302 patients with CL ± P and their parents. MAIN OUTCOME MEASURES To ascertain the genetic variants in 1q32.2 in patients with CL ± P in Han Chinese Population. INTERVENTIONS Blood samples were collected. RESULTS We found T allele (Z = 4.26, p = 0.00002) and T/T homozygotes (Z = 4.4, p = 0.000011) at rs12063989 was significantly over-transmitted among non-syndromic cleft lip with or without cleft palate (NSCL ± P). CONCLUSIONS We found rs12063989 exhibited significant association with the occurrence of NSCL ± P, which would provide new evidence for the future study in the etiology of NSCL ± P.
Collapse
Affiliation(s)
- Bin Yin
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of cleft lip and palate, West China Hospital of Stomatology, 12530Sichuan University, Chengdu, China
| | - Jia-Yu Shi
- Division of Growth and Development and Section of Orthodontics, School of Dentistry, University of California, Los Angeles, USA
| | - Bing Shi
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of cleft lip and palate, West China Hospital of Stomatology, 12530Sichuan University, Chengdu, China
| | - Qian Zheng
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of cleft lip and palate, West China Hospital of Stomatology, 12530Sichuan University, Chengdu, China
| | - Zhong-Lin Jia
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of cleft lip and palate, West China Hospital of Stomatology, 12530Sichuan University, Chengdu, China
| |
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
|
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
|
15
|
Alade AA, Buxo‐Martinez CJ, Mossey PA, Gowans LJ, Eshete MA, Adeyemo WL, Naicker T, Awotoye WA, Adeleke C, Busch T, Toraño AM, Bello CA, Soto M, Soto M, Ledesma R, Marquez M, Cordero JF, Lopez‐Del Valle LM, Salcedo MI, Debs N, Li M, Petrin A, Olotu J, Aldous C, Olutayo J, Ogunlewe MO, Abate F, Hailu T, Muhammed I, Gravem P, Deribew M, Gesses M, Hassan M, Pape J, Adeniyan OA, Obiri‐Yeboah S, Arthur FK, Oti AA, Olatosi O, Miller SE, Donkor P, Dunnwald MM, Marazita ML, Adeyemo AA, Murray JC, Butali A. Non-random distribution of deleterious mutations in the DNA and protein-binding domains of IRF6 are associated with Van Der Woude syndrome. Mol Genet Genomic Med 2020; 8:e1355. [PMID: 32558391 PMCID: PMC7434609 DOI: 10.1002/mgg3.1355] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 05/03/2020] [Accepted: 05/19/2020] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND The development of the face occurs during the early days of intrauterine life by the formation of facial processes from the first Pharyngeal arch. Derangement in these well-organized fusion events results in Orofacial clefts (OFC). Van der Woude syndrome (VWS) is one of the most common causes of syndromic cleft lip and/or palate accounting for 2% of all cases. Mutations in the IRF6 gene account for 70% of cases with the majority of these mutations located in the DNA-binding (exon 3, 4) or protein-binding domains (exon 7-9). The current study was designed to update the list of IRF6 variants reported for VWS by compiling all the published mutations from 2013 to date as well as including the previously unreported VWS cases from Africa and Puerto Rico. METHODS We used PubMed with the search terms; "Van der Woude syndrome," "Popliteal pterygium syndrome," "IRF6," and "Orofacial cleft" to identify eligible studies. We compiled the CADD score for all the mutations to determine the percentage of deleterious variants. RESULTS Twenty-one new mutations were identified from nine papers. The majority of these mutations were in exon 4. Mutations in exon 3 and 4 had CADD scores between 20 and 30 and mutations in exon 7-9 had CADD scores between 30 and 40. The presence of higher CADD scores in the protein-binding domain (exon 7-9) further confirms the crucial role played by this domain in the function of IRF6. In the new cases, we identified five IRF6 mutations, three novel missense mutations (p.Phe36Tyr, p.Lys109Thr, and p.Gln438Leu), and two previously reported nonsense mutations (p.Ser424*and p.Arg250*). CONCLUSION Mutations in the protein and DNA-binding domains of IRF6 ranked among the top 0.1% and 1% most deleterious genetic mutations, respectively. Overall, these findings expand the range of VWS mutations and are important for diagnostic and counseling purposes.
Collapse
Affiliation(s)
- Azeez A. Alade
- Department of Oral Pathology, Radiology and MedicineCollege of DentistryUniversity of IowaIowa CityIAUSA
- Iowa Institute of Oral Health ResearchUniversity of IowaIowa CityIAUSA
- Department of EpidemiologyCollege of Public HealthUniversity of IowaIowa CityIAUSA
| | - Carmen J. Buxo‐Martinez
- Dental and Craniofacial Genomics CoreUniversity of Puerto Rico School of Dental MedicineSan JuanPuerto Rico
| | - Peter A. Mossey
- Department of OrthodonticsUniversity of DundeeDundeeUnited Kingdom
| | - Lord J.J. Gowans
- Komfo Anokye Teaching Hospital and Kwame Nkrumah University of Science and TechnologyKumasiGhana
| | | | - Wasiu L. Adeyemo
- Department of Oral and Maxillofacial SurgeryUniversity of LagosLagosNigeria
| | - Thirona Naicker
- School of clinical medicineKwaZulu‐Natal UniversityDurbanSouth Africa
| | - Waheed A. Awotoye
- Department of Oral Pathology, Radiology and MedicineCollege of DentistryUniversity of IowaIowa CityIAUSA
- Iowa Institute of Oral Health ResearchUniversity of IowaIowa CityIAUSA
| | - Chinyere Adeleke
- Department of Oral Pathology, Radiology and MedicineCollege of DentistryUniversity of IowaIowa CityIAUSA
| | - Tamara Busch
- Department of Oral Pathology, Radiology and MedicineCollege of DentistryUniversity of IowaIowa CityIAUSA
- Iowa Institute of Oral Health ResearchUniversity of IowaIowa CityIAUSA
| | - Ada M. Toraño
- Dental and Craniofacial Genomics CoreUniversity of Puerto Rico School of Dental MedicineSan JuanPuerto Rico
| | - Carolina A. Bello
- Dental and Craniofacial Genomics CoreUniversity of Puerto Rico School of Dental MedicineSan JuanPuerto Rico
| | - Mairim Soto
- Dental and Craniofacial Genomics CoreUniversity of Puerto Rico School of Dental MedicineSan JuanPuerto Rico
| | - Marilyn Soto
- Dental and Craniofacial Genomics CoreUniversity of Puerto Rico School of Dental MedicineSan JuanPuerto Rico
| | - Ricardo Ledesma
- Dental and Craniofacial Genomics CoreUniversity of Puerto Rico School of Dental MedicineSan JuanPuerto Rico
| | - Myrellis Marquez
- Dental and Craniofacial Genomics CoreUniversity of Puerto Rico School of Dental MedicineSan JuanPuerto Rico
| | - Jose F. Cordero
- Dental and Craniofacial Genomics CoreUniversity of Puerto Rico School of Dental MedicineSan JuanPuerto Rico
| | - Lydia M. Lopez‐Del Valle
- Dental and Craniofacial Genomics CoreUniversity of Puerto Rico School of Dental MedicineSan JuanPuerto Rico
| | - Maria I. Salcedo
- Dental and Craniofacial Genomics CoreUniversity of Puerto Rico School of Dental MedicineSan JuanPuerto Rico
| | - Natalio Debs
- Dental and Craniofacial Genomics CoreUniversity of Puerto Rico School of Dental MedicineSan JuanPuerto Rico
| | - Mary Li
- Department of Oral Pathology, Radiology and MedicineCollege of DentistryUniversity of IowaIowa CityIAUSA
- Iowa Institute of Oral Health ResearchUniversity of IowaIowa CityIAUSA
| | - Aline Petrin
- Iowa Institute of Oral Health ResearchUniversity of IowaIowa CityIAUSA
| | - Joy Olotu
- Department of AnatomyUniversity of Port HarcourtPort HarcourtNigeria
| | - Colleen Aldous
- School of clinical medicineKwaZulu‐Natal UniversityDurbanSouth Africa
| | - James Olutayo
- Department of Oral and Maxillofacial SurgeryUniversity of LagosLagosNigeria
| | - Modupe O. Ogunlewe
- Department of Oral and Maxillofacial SurgeryUniversity of LagosLagosNigeria
| | - Fekir Abate
- School of Public HealthAddis Ababa UniversityAddis AbabaEthiopia
| | - Taye Hailu
- School of Public HealthAddis Ababa UniversityAddis AbabaEthiopia
| | - Ibrahim Muhammed
- School of Public HealthAddis Ababa UniversityAddis AbabaEthiopia
| | - Paul Gravem
- School of Public HealthAddis Ababa UniversityAddis AbabaEthiopia
| | - Milliard Deribew
- School of Public HealthAddis Ababa UniversityAddis AbabaEthiopia
| | - Mulualem Gesses
- School of Public HealthAddis Ababa UniversityAddis AbabaEthiopia
| | - Mohaned Hassan
- Department of Oral Pathology, Radiology and MedicineCollege of DentistryUniversity of IowaIowa CityIAUSA
- Iowa Institute of Oral Health ResearchUniversity of IowaIowa CityIAUSA
| | - John Pape
- Department of Oral Pathology, Radiology and MedicineCollege of DentistryUniversity of IowaIowa CityIAUSA
| | - Oluwole A. Adeniyan
- NHS Foundation Trust, (Queens Hospital, Burton‐On‐Trent)StaffordshireUnited Kingdom
| | - Solomon Obiri‐Yeboah
- Komfo Anokye Teaching Hospital and Kwame Nkrumah University of Science and TechnologyKumasiGhana
| | - Fareed K.N. Arthur
- Komfo Anokye Teaching Hospital and Kwame Nkrumah University of Science and TechnologyKumasiGhana
| | - Alexander A. Oti
- Komfo Anokye Teaching Hospital and Kwame Nkrumah University of Science and TechnologyKumasiGhana
| | | | - Sara E. Miller
- Iowa Institute of Oral Health ResearchUniversity of IowaIowa CityIAUSA
| | - Peter Donkor
- Komfo Anokye Teaching Hospital and Kwame Nkrumah University of Science and TechnologyKumasiGhana
| | | | - Mary L. Marazita
- Oral BiologyHuman GeneticsUniversity of PittsburghPittsburghPAUSA
| | | | | | - Azeez Butali
- Department of Oral Pathology, Radiology and MedicineCollege of DentistryUniversity of IowaIowa CityIAUSA
- Iowa Institute of Oral Health ResearchUniversity of IowaIowa CityIAUSA
| |
Collapse
|
16
|
Zhang M, Zhang J, Zhao H, Ievlev V, Zhong W, Huang W, Cornell RA, Lin J, Chen F. Functional Characterization of a Novel IRF6 Frameshift Mutation From a Van Der Woude Syndrome Family. Front Genet 2020; 11:562. [PMID: 32582293 PMCID: PMC7289175 DOI: 10.3389/fgene.2020.00562] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Accepted: 05/11/2020] [Indexed: 12/14/2022] Open
Abstract
Background Loss-of-function mutations in interferon regulatory factor-6 (IRF6) are responsible for about 70% of cases of Van Der Woude Syndrome (VWS), an autosomal dominant developmental disorder characterized by pits and/or sinuses of the lower lip and cleft lip, cleft palate, or both. Methods We collected a Chinese Han VWS pedigree, performed sequencing and screening for the causal gene mutant. Initially, species conservation analysis and homology protein modeling were used to predict the potential pathogenicity of mutations. To test whether a VWS family-derived mutant variant of IRF6 retained function, we carried out rescue assays in irf6 maternal-null mutant zebrafish embryos. To assess protein stability, we overexpressed reference and family-variants of IRF6 in vitro. Results We focused on a VWS family that includes a son with bilateral lip pits, uvula fissa and his father with bilateral cleft lip and palate. After sequencing and screening, a frameshift mutation of IRF6 was identified as the potential causal variant (NM.006147.3, c.1088-1091delTCTA; p.Ile363ArgfsTer33). The residues in this position are strongly conserved among species and homology modeling suggests the variant alters the protein structure. In irf6 maternal-null mutant zebrafish embryos the periderm differentiates abnormally and the embryos rupture and die during gastrulation. Injection of mRNA encoding the reference variant of human IRF6, but not of the frame-shift variant, rescued such embryos through gastrulation. Upon overexpression in HEK293FT cells, the IRF6 frame-shift mutant was relatively unstable and was preferentially targeted to the proteasome in comparison to the reference variant. Conclusion In this VWS pedigree, a novel frameshift of IRF6 was identified as the likely causative gene variant. It is a lost function mutation which could not rescue abnormal periderm phenotype in irf6 maternal-null zebrafish and which causes the protein be unstable through proteasome-dependent degradation.
Collapse
Affiliation(s)
- Mengqi Zhang
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
| | - Jieni Zhang
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
| | - Huaxiang Zhao
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
| | - Vitaly Ievlev
- Department of Anatomy and Cell Biology Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Wenjie Zhong
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
| | - Wenbin Huang
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
| | - Robert A Cornell
- Department of Anatomy and Cell Biology Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Jiuxiang Lin
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
| | - Feng Chen
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, China
| |
Collapse
|
17
|
Drug-selective Anesthetic Insensitivity of Zebrafish Lacking γ-Aminobutyric Acid Type A Receptor β3 Subunits. Anesthesiology 2020; 131:1276-1291. [PMID: 31567362 DOI: 10.1097/aln.0000000000002963] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Transgenic mouse studies suggest that γ-aminobutyric acid type A (GABAA) receptors containing β3 subunits mediate important effects of etomidate, propofol, and pentobarbital. Zebrafish, recently introduced for rapid discovery and characterization of sedative-hypnotics, could also accelerate pharmacogenetic studies if their transgenic phenotypes reflect those of mammals. The authors hypothesized that, relative to wild-type, GABAA-β3 functional knock-out (β3) zebrafish would show anesthetic sensitivity changes similar to those of β3 mice. METHODS Clustered regularly interspaced short palindromic repeats (CRISPR)-Cas9 mutagenesis was used to create a β3 zebrafish line. Wild-type and β3 zebrafish were compared for fertility, growth, and craniofacial development. Sedative and hypnotic effects of etomidate, propofol, pentobarbital, alphaxalone, ketamine, tricaine, dexmedetomidine, butanol, and ethanol, along with overall activity and thigmotaxis were quantified in 7-day postfertilization larvae using video motion analysis of up to 96 animals simultaneously. RESULTS Xenopus oocyte electrophysiology showed that the wild-type zebrafish β3 gene encodes ion channels activated by propofol and etomidate, while the β3 zebrafish transgene does not. Compared to wild-type, β3 zebrafish showed similar morphology and growth, but more rapid swimming. Hypnotic EC50s (mean [95% CI]) were significantly higher for β3 versus wild-type larvae with etomidate (1.3 [1.0 to 1.6] vs. 0.6 [0.5 to 0.7] µM; P < 0.0001), propofol (1.1 [1.0 to 1.4] vs. 0.7 [0.6 to 0.8] µM; P = 0.0005), and pentobarbital (220 [190 to 240] vs. 130 [94 to 179] μM; P = 0.0009), but lower with ethanol (150 [106 to 213] vs. 380 [340 to 420] mM; P < 0.0001) and equivalent with other tested drugs. Comparing β3 versus wild-type sedative EC50s revealed a pattern similar to hypnosis. CONCLUSIONS Global β3 zebrafish are selectively insensitive to the same few sedative-hypnotics previously reported in β3 transgenic mice, indicating phylogenetic conservation of β3-containing GABAA receptors as anesthetic targets. Transgenic zebrafish are potentially valuable models for sedative-hypnotic mechanisms research.
Collapse
|
18
|
Liu H, Duncan K, Helverson A, Kumari P, Mumm C, Xiao Y, Carlson JC, Darbellay F, Visel A, Leslie E, Breheny P, Erives AJ, Cornell RA. Analysis of zebrafish periderm enhancers facilitates identification of a regulatory variant near human KRT8/18. eLife 2020; 9:e51325. [PMID: 32031521 PMCID: PMC7039683 DOI: 10.7554/elife.51325] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Accepted: 02/06/2020] [Indexed: 12/18/2022] Open
Abstract
Genome-wide association studies for non-syndromic orofacial clefting (OFC) have identified single nucleotide polymorphisms (SNPs) at loci where the presumed risk-relevant gene is expressed in oral periderm. The functional subsets of such SNPs are difficult to predict because the sequence underpinnings of periderm enhancers are unknown. We applied ATAC-seq to models of human palate periderm, including zebrafish periderm, mouse embryonic palate epithelia, and a human oral epithelium cell line, and to complementary mesenchymal cell types. We identified sets of enhancers specific to the epithelial cells and trained gapped-kmer support-vector-machine classifiers on these sets. We used the classifiers to predict the effects of 14 OFC-associated SNPs at 12q13 near KRT18. All the classifiers picked the same SNP as having the strongest effect, but the significance was highest with the classifier trained on zebrafish periderm. Reporter and deletion analyses support this SNP as lying within a periderm enhancer regulating KRT18/KRT8 expression.
Collapse
Affiliation(s)
- Huan Liu
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory for Oral Biomedicine of Ministry of Education (KLOBM), School and Hospital of Stomatology, Wuhan UniversityWuhanChina
- Department of Anatomy and Cell Biology, University of IowaIowa CityUnited States
- Department of Periodontology, School of Stomatology, Wuhan UniversityWuhanChina
| | - Kaylia Duncan
- Interdisciplinary Program in Molecular Medicine, University of IowaIowa CityUnited States
| | - Annika Helverson
- Department of Anatomy and Cell Biology, University of IowaIowa CityUnited States
| | - Priyanka Kumari
- Department of Anatomy and Cell Biology, University of IowaIowa CityUnited States
| | - Camille Mumm
- Department of Anatomy and Cell Biology, University of IowaIowa CityUnited States
| | - Yao Xiao
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory for Oral Biomedicine of Ministry of Education (KLOBM), School and Hospital of Stomatology, Wuhan UniversityWuhanChina
| | | | - Fabrice Darbellay
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley LaboratoriesBerkeleyUnited States
| | - Axel Visel
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley LaboratoriesBerkeleyUnited States
- U.S. Department of Energy Joint Genome Institute, Lawrence Berkeley LaboratoriesBerkeleyUnited States
- University of California, MercedMercedUnited States
| | - Elizabeth Leslie
- Department of Human Genetics, Emory University School of MedicineAtlantaGeorgia
| | - Patrick Breheny
- Department of Biostatistics, University of IowaIowa CityUnited States
| | - Albert J Erives
- Department of Biology, University of IowaIowa CityUnited States
| | - Robert A Cornell
- Department of Anatomy and Cell Biology, University of IowaIowa CityUnited States
- Interdisciplinary Program in Molecular Medicine, University of IowaIowa CityUnited States
| |
Collapse
|
19
|
Ma J, Yin B, Shi JY, Lin YS, Duan SJ, Shi B, Jia ZL. Exon sequencing reveals that missense mutation of PBX1 gene may increase the risk of non-syndromic cleft lip/palate. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2019; 12:2691-2698. [PMID: 31934099 PMCID: PMC6949560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 05/24/2019] [Indexed: 06/10/2023]
Abstract
OBJECTIVE Non-syndromic oral cleft (NSOC) is one of the most common multifactorial birth defects. A previous animal study showed PBX1 gene knockout mice consequently exhibited complete cleft lip/palate (CL/P). However, little is known about the association between PBX1 and NSOC in humans. This study investigated the role of the PBX1 gene in NSOC in the Han Chinese population. METHODS In all, 287 NSOCs were recruited for this study. First, exons in the PBX1 gene were sequenced among 50 non-syndromic cleft lip and palate cases to screen for variations by the Sanger sequencing method. Then, we selected four SNPs to replicate among 237 NSOC trios and analyzed the data by using TDT and parent of origin effect methods. RESULTS Exon sequencing identified six variants of the PBX1 gene. Among them, four variants were common variants. TDT analysis revealed allele G at rs2275558 and allele T at rs3835581 were over-transmitted in NSCL/P (P=0.039 and 0.038, respectively), which could increase the risk for NSCL/P. Parent of origin effect analysis indicated that allele G at rs2275558 was paternally over-transmitted for NSCL/P (P=0.0091). CONCLUSION This is the first report that the PBX1 gene is associated with NSCL/P, which indicates that it is a promising candidate gene for NSCL/P.
Collapse
Affiliation(s)
- Jian Ma
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Development of Cleft Lip and Palate, West China Hospital of Stomatology, Sichuan UniversityP.R. China
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, The General Hospital of Ningxia Medical UniversityYinchuan, 750004, Ningxia Province, P.R. China
| | - Bin Yin
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Development of Cleft Lip and Palate, West China Hospital of Stomatology, Sichuan UniversityP.R. China
| | - Jia-Yu Shi
- Division of Growth and Development and Section of Orthodontics, School of Dentistry, University of CaliforniaLos Angeles, USA
| | - Yan-Song Lin
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Development of Cleft Lip and Palate, West China Hospital of Stomatology, Sichuan UniversityP.R. China
| | - Shi-Jun Duan
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Development of Cleft Lip and Palate, West China Hospital of Stomatology, Sichuan UniversityP.R. China
| | - Bing Shi
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Development of Cleft Lip and Palate, West China Hospital of Stomatology, Sichuan UniversityP.R. China
| | - Zhong-Lin Jia
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Development of Cleft Lip and Palate, West China Hospital of Stomatology, Sichuan UniversityP.R. China
| |
Collapse
|
20
|
Kousa YA, Zhu H, Fakhouri WD, Lei Y, Kinoshita A, Roushangar RR, Patel NK, Agopian AJ, Yang W, Leslie EJ, Busch TD, Mansour TA, Li X, Smith AL, Li EB, Sharma DB, Williams TJ, Chai Y, Amendt BA, Liao EC, Mitchell LE, Bassuk AG, Gregory S, Ashley-Koch A, Shaw GM, Finnell RH, Schutte BC. The TFAP2A-IRF6-GRHL3 genetic pathway is conserved in neurulation. Hum Mol Genet 2019; 28:1726-1737. [PMID: 30689861 PMCID: PMC6494790 DOI: 10.1093/hmg/ddz010] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 11/26/2018] [Accepted: 12/31/2018] [Indexed: 02/06/2023] Open
Abstract
Mutations in IRF6, TFAP2A and GRHL3 cause orofacial clefting syndromes in humans. However, Tfap2a and Grhl3 are also required for neurulation in mice. Here, we found that homeostasis of Irf6 is also required for development of the neural tube and associated structures. Over-expression of Irf6 caused exencephaly, a rostral neural tube defect, through suppression of Tfap2a and Grhl3 expression. Conversely, loss of Irf6 function caused a curly tail and coincided with a reduction of Tfap2a and Grhl3 expression in tail tissues. To test whether Irf6 function in neurulation was conserved, we sequenced samples obtained from human cases of spina bifida and anencephaly. We found two likely disease-causing variants in two samples from patients with spina bifida. Overall, these data suggest that the Tfap2a-Irf6-Grhl3 genetic pathway is shared by two embryologically distinct morphogenetic events that previously were considered independent during mammalian development. In addition, these data suggest new candidates to delineate the genetic architecture of neural tube defects and new therapeutic targets to prevent this common birth defect.
Collapse
Affiliation(s)
- Youssef A Kousa
- Departments of Biochemistry and Molecular Biology
- Division of Neurology, Childrens National Health System
- Center for Neuroscience Research, The Childrens Research Institute, Washington, DC, USA
| | - Huiping Zhu
- Dell Pediatric Research Institute, Department of Nutritional Sciences, University of Texas at Austin, Austin, TX, USA
| | - Walid D Fakhouri
- Department of Diagnostic & Biomedical Sciences, School of Dentistry, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Yunping Lei
- Dell Pediatric Research Institute, Department of Nutritional Sciences, University of Texas at Austin, Austin, TX, USA
| | - Akira Kinoshita
- Department of Human Genetics, Nagasaki University, Nagasaki, Japan
| | | | | | - A J Agopian
- Human Genetics Center, Division of Epidemiology, Human Genetics and Environmental Sciences, University of Texas School of Public Health, Houston, TX, USA
| | - Wei Yang
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Elizabeth J Leslie
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | | | - Tamer A Mansour
- Genetics PhD Program
- Department of Clinical Pathology, School of Medicine, University of Mansoura, Mansoura, Egypt
- Department of Population Health and Reproduction, School of Veterinary Medicine, University of California, Davis, CA, USA
| | - Xiao Li
- Anatomy and Cell Biology, University of Iowa, Iowa City, IA, USA
| | | | - Edward B Li
- Center for Regenerative Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Dhruv B Sharma
- Center for Statistical Training & Consulting, Michigan State University, East Lansing, MI, USA
| | - Trevor J Williams
- Department of Craniofacial Biology, University of Colorado Denver at Anschutz Medical Campus, Aurora, CO, USA
| | - Yang Chai
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, USA
| | - Brad A Amendt
- Anatomy and Cell Biology, University of Iowa, Iowa City, IA, USA
| | - Eric C Liao
- Center for Regenerative Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Laura E Mitchell
- Human Genetics Center, Division of Epidemiology, Human Genetics and Environmental Sciences, University of Texas School of Public Health, Houston, TX, USA
| | | | - Simon Gregory
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, USA
| | - Allison Ashley-Koch
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, USA
| | - Gary M Shaw
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Richard H Finnell
- Dell Pediatric Research Institute, Department of Nutritional Sciences, University of Texas at Austin, Austin, TX, USA
| | - Brian C Schutte
- Departments of Biochemistry and Molecular Biology
- Microbiology and Molecular Genetics
- Genetics PhD Program
- Pediatrics and Human Development
| |
Collapse
|
21
|
Model systems inform rare disease diagnosis, therapeutic discovery and pre-clinical efficacy. Emerg Top Life Sci 2019; 3:1-10. [DOI: 10.1042/etls20180057] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 02/11/2019] [Accepted: 02/15/2019] [Indexed: 01/12/2023]
Abstract
Abstract
Model systems have played a large role in understanding human diseases and are instrumental in taking basic research findings to the clinic; however, for rare diseases, model systems play an even larger role. Here, we outline how model organisms are crucial for confirming causal associations, understanding functional mechanisms and developing therapies for disease. As diseases that have been studied extensively through genetics and molecular biology, cystic fibrosis and Rett syndrome are portrayed as primary examples of how genetic diagnosis, model organism development and therapies have led to improved patient health. Considering which model to use, yeast, worms, flies, fish, mice or larger animals requires a careful evaluation of experimental genetic tools and gene pathway conservation. Recent advances in genome editing will aid in confirming diagnoses and developing model systems for rare disease. Genetic or chemical screening for disease suppression may reveal functional pathway members and provide candidate entry points for developing therapies. Model organisms may also be used in drug discovery and as preclinical models as a prelude to testing treatments in patient populations. Now, model organisms will increasingly be used as platforms for understanding variation in rare disease severity and onset, thereby informing therapeutic intervention.
Collapse
|
22
|
Loss-of-function of sox3 causes follicle development retardation and reduces fecundity in zebrafish. Protein Cell 2018; 10:347-364. [PMID: 30588557 PMCID: PMC6468042 DOI: 10.1007/s13238-018-0603-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 11/21/2018] [Indexed: 12/02/2022] Open
Abstract
Folliculogenesis is essential for production of female gametes in vertebrates. However, the molecular mechanisms underlying follicle development, particularly apoptosis regulation in ovary, remain elusive. Here, we generated sox3 knockout zebrafish lines using CRISPR/Cas9. sox3 knockout led to follicle development retardation and a reduced fecundity in females. Comparative analysis of transcriptome between sox3−/− and wild-type ovaries revealed that Sox3 was involved in pathways of ovarian steroidogenesis and apoptosis. Knockout of sox3 promoted follicle apoptosis and obvious apoptosis signals were detected in somatic cells of stages III and IV follicles of sox3−/− ovaries. Moreover, Sox3 can bind to and activate the promoter of cyp19a1a. Up-regulation of Cyp19a1a expression promoted 17β-estradiol synthesis, which inhibited apoptosis in follicle development. Thus, Sox3 functions as a regulator of Cyp19a1a expression, via 17β-E2 linking apoptosis suppression, which is implicated in improving female fecundity.
Collapse
|
23
|
Neves LT, Dionísio TJ, Garbieri TF, Parisi VA, Oliveira FV, Oliveira TM, Santos CF. Novel rare variations in
IRF6
in subjects with non‐syndromic cleft lip and palate and dental agenesis. Oral Dis 2018; 25:223-233. [DOI: 10.1111/odi.12975] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 08/30/2018] [Accepted: 09/03/2018] [Indexed: 12/11/2022]
Affiliation(s)
- Lucimara T. Neves
- Department of Biological Sciences, Bauru School of Dentistry University of São Paulo Bauru Brazil
- Post‐Graduation Program in Rehabilitation Sciences, Hospital for Rehabilitation of Craniofacial Anomalies University of São Paulo Bauru Brazil
| | - Thiago J. Dionísio
- Department of Biological Sciences, Bauru School of Dentistry University of São Paulo Bauru Brazil
| | - Thais F. Garbieri
- Department of Biological Sciences, Bauru School of Dentistry University of São Paulo Bauru Brazil
| | - Viviane A. Parisi
- Department of Biological Sciences, Bauru School of Dentistry University of São Paulo Bauru Brazil
| | - Fernanda V. Oliveira
- Department of Pediatric Dentistry, Orthodontics and Community Health, Bauru School of Dentistry University of São Paulo Bauru Brazil
| | - Thais M. Oliveira
- Post‐Graduation Program in Rehabilitation Sciences, Hospital for Rehabilitation of Craniofacial Anomalies University of São Paulo Bauru Brazil
- Department of Pediatric Dentistry, Orthodontics and Community Health, Bauru School of Dentistry University of São Paulo Bauru Brazil
| | - Carlos F. Santos
- Department of Biological Sciences, Bauru School of Dentistry University of São Paulo Bauru Brazil
- Post‐Graduation Program in Rehabilitation Sciences, Hospital for Rehabilitation of Craniofacial Anomalies University of São Paulo Bauru Brazil
| |
Collapse
|
24
|
Chakravorty S, Hegde M. Inferring the effect of genomic variation in the new era of genomics. Hum Mutat 2018; 39:756-773. [PMID: 29633501 DOI: 10.1002/humu.23427] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 03/20/2018] [Accepted: 03/28/2018] [Indexed: 12/11/2022]
Abstract
Accurate and detailed understanding of the effects of variants in the coding and noncoding regions of the genome is the next big challenge in the new genomic era of personalized medicine, especially to tackle newer findings of genetic and phenotypic heterogeneity of diseases. This is necessary to resolve the gene-variant-disease relationship, the pathogenic variant spectrum of genes, pathogenic variants with variable clinical consequences, and multiloci diseases. In turn, this will facilitate patient recruitment for relevant clinical trials. In this review, we describe the trends in research at the intersection of basic and clinical genomics aiming to (a) overcome molecular diagnostic challenges and increase the clinical utility of next-generation sequencing (NGS) platforms, (b) elucidate variants associated with disease, (c) determine overall genomic complexity including epistasis, complex inheritance patterns such as "synergistic heterozygosity," digenic/multigenic inheritance, modifier effect, and rare variant load. We describe the newly emerging field of integrated functional genomics, in vivo or in vitro large-scale functional approaches, statistical bioinformatics algorithms that support NGS genomics data to interpret variants for timely clinical diagnostics and disease management. Thus, facilitating the discovery of new therapeutic or biomarker options, and their roles in the future of personalized medicine.
Collapse
Affiliation(s)
- Samya Chakravorty
- Department of Human Genetics, Emory University School of Medicine, Whitehead Biomedical Research Building Suite 301, Atlanta, Georgia
| | - Madhuri Hegde
- Department of Human Genetics, Emory University School of Medicine, Whitehead Biomedical Research Building Suite 301, Atlanta, Georgia
| |
Collapse
|
25
|
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
|