1
|
Carmichael JS, Darland DC, Pedersen DE, Simmons RB. Bones, brains, and bias-neural crest cell contribution to craniofacial structure. Dev Biol 2025; 524:116-122. [PMID: 40349905 DOI: 10.1016/j.ydbio.2025.05.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 05/06/2025] [Accepted: 05/09/2025] [Indexed: 05/14/2025]
Abstract
Implicit bias is a natural part of the human psyche and facial appearance is one of the first aspects that people notice when encountering new individuals. The cranial neural crest is key to formation of the facial structure as it is a developmentally transient and plastic cell population that contributes to the neural, muscular, cartilage and bone structures of the face and head. This remarkable cell population, within the context of genetic potential, can play a substantial role in determining how individuals' faces and heads form as well as contributing to anterior brain development. How humans interpret face and head features can lead to biased perceptions, including perceived cognitive ability associated with differences across craniofacial phenotypes. In this Opinion Article, we offer strategies to introduce students to an overview of cranial neural crest development linked to primary research in model organisms and in humans. We then bridge this knowledge with a follow-up activity to foster awareness of cognitive processes and implicit bias in human perception. We provide explicit Learning Goals and guided learning strategies to achieve clear Learning Outcomes. We have developed critical thinking assignments and self-reflection opportunities to engage students and shed light on misconceptions regarding craniofacial features. By introducing the generalized processes whereby neural crest cells contribute to head and face formation, we provide an opportunity to focus on bones, brains, and bias in development in order to encourage students to consider how implicit bias shapes human interaction and scientific work.
Collapse
Affiliation(s)
| | - Diane C Darland
- Department of Biology, University of North Dakota, Grand Forks, ND, 58202, USA.
| | - Daphne E Pedersen
- Department of Sociology, University of North Dakota, Grand Forks, ND, 58202, USA
| | - Rebecca B Simmons
- Department of Biology, University of North Dakota, Grand Forks, ND, 58202, USA
| |
Collapse
|
2
|
Nakamura M, Sandell LL. Multiple roles for retinoid signaling in craniofacial development. Curr Top Dev Biol 2024; 161:33-57. [PMID: 39870438 DOI: 10.1016/bs.ctdb.2024.09.002] [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: 01/29/2025]
Abstract
Retinoic acid (RA) signaling plays multiple essential roles in development of the head and face. Animal models with mutations in genes involved in RA signaling have enabled understanding of craniofacial morphogenic processes that are regulated by the retinoid pathway. During craniofacial morphogenesis RA signaling is active in spatially restricted domains defined by the expression of genes involved in RA production and RA breakdown. The spatial distribution of RA signaling changes with progressive development, corresponding to a multiplicity of craniofacial developmental processes that are regulated by RA. One important role of RA signaling occurs in the hindbrain. There RA contributes to specification of the anterior-posterior (AP) axis of the developing CNS and to the neural crest cells (NCC) which form the bones and nerves of the face and pharyngeal region. In the optic vesicles and frontonasal process RA orchestrates development of the midface, eyes, and nasal airway. Additional roles for RA in craniofacial development include regulation of submandibular salivary gland development and maintaining patency in the sutures of the cranial vault.
Collapse
Affiliation(s)
- Masahiro Nakamura
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, KY, United States
| | - Lisa L Sandell
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, KY, United States.
| |
Collapse
|
3
|
Johnson HK, Wahl SE, Sesay F, Litovchick L, Dickinson AJ. Dyrk1a is required for craniofacial development in Xenopus laevis. Dev Biol 2024; 511:63-75. [PMID: 38621649 PMCID: PMC12024765 DOI: 10.1016/j.ydbio.2024.04.004] [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: 12/16/2023] [Revised: 03/29/2024] [Accepted: 04/11/2024] [Indexed: 04/17/2024]
Abstract
Loss of function variations in the dual specificity tyrosine-phosphorylation-regulated kinase 1 A (DYRK1A) gene are associated with craniofacial malformations in humans. Here we characterized the effects of deficient DYRK1A in craniofacial development using a developmental model, Xenopus laevis. Dyrk1a mRNA and protein were expressed throughout the developing head and both were enriched in the branchial arches which contribute to the face and jaw. Consistently, reduced Dyrk1a function, using dyrk1a morpholinos and pharmacological inhibitors, resulted in orofacial malformations including hypotelorism, altered mouth shape, slanted eyes, and narrower face accompanied by smaller jaw cartilage and muscle. Inhibition of Dyrk1a function resulted in misexpression of key craniofacial regulators including transcription factors and members of the retinoic acid signaling pathway. Two such regulators, sox9 and pax3 are required for neural crest development and their decreased expression corresponds with smaller neural crest domains within the branchial arches. Finally, we determined that the smaller size of the faces, jaw elements and neural crest domains in embryos deficient in Dyrk1a could be explained by increased cell death and decreased proliferation. This study is the first to provide insight into why craniofacial birth defects might arise in humans with variants of DYRK1A.
Collapse
Affiliation(s)
| | - Stacey E Wahl
- Department of Biology, Virginia Commonwealth University, Richmond, VA, USA
| | - Fatmata Sesay
- Department of Internal Medicine, Division of Hematology, Oncology and Palliative Care, Virginia Commonwealth University, Richmond, VA, USA
| | - Larisa Litovchick
- Department of Internal Medicine, Division of Hematology, Oncology and Palliative Care, Virginia Commonwealth University, Richmond, VA, USA; Massey Comprehensive Cancer Center, Richmond, VA, USA
| | | |
Collapse
|
4
|
Seto Y, Ogihara R, Takizawa K, Eiraku M. In vitro induction of patterned branchial arch-like aggregate from human pluripotent stem cells. Nat Commun 2024; 15:1351. [PMID: 38355589 PMCID: PMC10867012 DOI: 10.1038/s41467-024-45285-0] [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: 05/16/2023] [Accepted: 01/19/2024] [Indexed: 02/16/2024] Open
Abstract
Early patterning of neural crest cells (NCCs) in the craniofacial primordium is important for subsequent development of proper craniofacial structures. However, because of the complexity of the environment of developing tissues, surveying the early specification and patterning of NCCs is difficult. In this study, we develop a simplified in vitro 3D model using human pluripotent stem cells to analyze the early stages of facial development. In this model, cranial NCC-like cells spontaneously differentiate from neural plate border-like cells into maxillary arch-like mesenchyme after a long-term culture. Upon the addition of EDN1 and BMP4, these aggregates are converted into a mandibular arch-like state. Furthermore, temporary treatment with EDN1 and BMP4 induces the formation of spatially separated domains expressing mandibular and maxillary arch markers within a single aggregate. These results suggest that this in vitro model is useful for determining the mechanisms underlying cell fate specification and patterning during early facial development.
Collapse
Affiliation(s)
- Yusuke Seto
- Laboratory of Developmental Systems, Institute for Life and Medical Sciences, Kyoto University, 53 Shogoin Kawaharacho, Sakyo-ku, Kyoto, 606-8507, Japan.
- Department of Polymer Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, 615-8510, Japan.
| | - Ryoma Ogihara
- Department of Polymer Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, 615-8510, Japan
| | - Kaori Takizawa
- Laboratory of Developmental Systems, Institute for Life and Medical Sciences, Kyoto University, 53 Shogoin Kawaharacho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Mototsugu Eiraku
- Laboratory of Developmental Systems, Institute for Life and Medical Sciences, Kyoto University, 53 Shogoin Kawaharacho, Sakyo-ku, Kyoto, 606-8507, Japan.
- Department of Polymer Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, 615-8510, Japan.
- Institute for Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan.
| |
Collapse
|
5
|
Bardhan S, Bhargava N, Dighe S, Vats N, Naganathan SR. Emergence of a left-right symmetric body plan in vertebrate embryos. Curr Top Dev Biol 2024; 159:310-342. [PMID: 38729680 DOI: 10.1016/bs.ctdb.2024.01.003] [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: 05/12/2024]
Abstract
External bilateral symmetry is a prevalent feature in vertebrates, which emerges during early embryonic development. To begin with, vertebrate embryos are largely radially symmetric before transitioning to bilaterally symmetry, after which, morphogenesis of various bilateral tissues (e.g somites, otic vesicle, limb bud), and structures (e.g palate, jaw) ensue. While a significant amount of work has probed the mechanisms behind symmetry breaking in the left-right axis leading to asymmetric positioning of internal organs, little is known about how bilateral tissues emerge at the same time with the same shape and size and at the same position on the two sides of the embryo. By discussing emergence of symmetry in many bilateral tissues and structures across vertebrate model systems, we highlight that understanding symmetry establishment is largely an open field, which will provide deep insights into fundamental problems in developmental biology for decades to come.
Collapse
Affiliation(s)
- Siddhartha Bardhan
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Nandini Bhargava
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Swarali Dighe
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Neha Vats
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Sundar Ram Naganathan
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India.
| |
Collapse
|
6
|
Johnson HK, Wahl SE, Sesay F, Litovchick L, Dickinson AJ. Dyrk1a is required for craniofacial development in Xenopus laevis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.13.575394. [PMID: 38260562 PMCID: PMC10802584 DOI: 10.1101/2024.01.13.575394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Loss of function mutations in the dual specificity tyrosine-phosphorylation-regulated kinase 1A (DYRK1A) gene are associated with craniofacial malformations in humans. Here we characterized the effects of deficient DYRK1A in craniofacial development using a developmental model, Xenopus laevis . Dyrk1a mRNA and protein was expressed throughout the developing head and was enriched in the branchial arches which contribute to the face and jaw. Consistently, reduced Dyrk1a function, using dyrk1a morpholinos and pharmacological inhibitors, resulted in orofacial malformations including hypotelorism, altered mouth shape, slanted eyes, and narrower face accompanied by smaller jaw cartilage and muscle. Inhibition of Dyrk1a function resulted in misexpression of key craniofacial regulators including transcription factors and members of the retinoic acid signaling pathway. Two such regulators, sox9 and pax3 are required for neural crest development and their decreased expression corresponds with smaller neural crest domains within the branchial arches. Finally, we determined that the smaller size of the faces, jaw elements and neural crest domains in embryos deficient in Dyrk1a could be explained by increased cell death and decreased proliferation. This study is the first to provide insight into why craniofacial birth defects might arise in humans with DYRK1A mutations.
Collapse
|
7
|
Shankar P, Villeneuve DL. AOP Report: Aryl Hydrocarbon Receptor Activation Leads to Early-Life Stage Mortality via Sox9 Repression-Induced Craniofacial and Cardiac Malformations. ENVIRONMENTAL TOXICOLOGY AND CHEMISTRY 2023; 42:2063-2077. [PMID: 37341548 PMCID: PMC10772968 DOI: 10.1002/etc.5699] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 06/15/2023] [Indexed: 06/22/2023]
Abstract
The aryl hydrocarbon receptors (Ahrs) are evolutionarily conserved ligand-dependent transcription factors that are activated by structurally diverse endogenous compounds as well as environmental chemicals such as polycyclic aromatic hydrocarbons and halogenated aromatic hydrocarbons. Activation of the Ahr leads to several transcriptional changes that can cause developmental toxicity resulting in mortality. Evidence was assembled and evaluated for two novel adverse outcome pathways (AOPs) which describe how Ahr activation (molecular initiating event) can lead to early-life stage mortality (adverse outcome), via either SOX9-mediated craniofacial malformations (AOP 455) or cardiovascular toxicity (AOP 456). Using a key event relationship (KER)-by-KER approach, we collected evidence using both a narrative search and a systematic review based on detailed search terms. Weight of evidence for each KER was assessed to inform overall confidence of the AOPs. The AOPs link to previous descriptions of Ahr activation and connect them to two novel key events (KEs), increase in slincR expression, a newly characterized long noncoding RNA with regulatory functions, and suppression of SOX9, a critical transcription factor implicated in chondrogenesis and cardiac development. In general, confidence levels for KERs ranged between medium and strong, with few inconsistencies, as well as several opportunities for future research identified. While the majority of KEs have only been demonstrated in zebrafish with 2,3,7,8-tetrachlorodibenzo-p-dioxin as an Ahr activator, evidence suggests that the two AOPs likely apply to most vertebrates and many Ahr-activating chemicals. Addition of the AOPs into the AOP-Wiki (https://aopwiki.org/) helps expand the growing Ahr-related AOP network to 19 individual AOPs, of which six are endorsed or in progress and the remaining 13 relatively underdeveloped. Environ Toxicol Chem 2023;42:2063-2077. © 2023 SETAC. This article has been contributed to by U.S. Government employees and their work is in the public domain in the USA.
Collapse
Affiliation(s)
- Prarthana Shankar
- Great Lakes Toxicology and Ecology Division, US Environmental Protection Agency, Duluth, Minnesota, USA
- University of Wisconsin Madison Sea Grant Fellow at Great Lakes Toxicology and Ecology Division, US Environmental Protection Agency, Duluth, Minnesota, USA
| | - Daniel L. Villeneuve
- Great Lakes Toxicology and Ecology Division, US Environmental Protection Agency, Duluth, Minnesota, USA
| |
Collapse
|
8
|
Dickinson AJG. Jak2 and Jaw Muscles Are Required for Buccopharyngeal Membrane Perforation during Mouth Development. J Dev Biol 2023; 11:24. [PMID: 37367478 DOI: 10.3390/jdb11020024] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/23/2023] [Accepted: 05/28/2023] [Indexed: 06/28/2023] Open
Abstract
The mouth is a central feature of our face, without which we could not eat, breathe, or communicate. A critical and early event in mouth formation is the creation of a "hole" which connects the digestive system and the external environment. This hole, which has also been called the primary or embryonic mouth in vertebrates, is initially covered by a 1-2 cell layer thick structure called the buccopharyngeal membrane. When the buccopharyngeal membrane does not rupture, it impairs early mouth functions and may also lead to further craniofacial malformations. Using a chemical screen in an animal model (Xenopus laevis) and genetic data from humans, we determined that Janus kinase 2 (Jak2) has a role in buccopharyngeal membrane rupture. We have determined that decreased Jak2 function, using antisense morpholinos or a pharmacological antagonist, caused a persistent buccopharyngeal membrane as well as the loss of jaw muscles. Surprisingly, we observed that the jaw muscle compartments were connected to the oral epithelium that is continuous with the buccopharyngeal membrane. Severing such connections resulted in buccopharyngeal membrane buckling and persistence. We also noted puncta accumulation of F-actin, an indicator of tension, in the buccopharyngeal membrane during perforation. Taken together, the data has led us to a hypothesis that muscles are required to exert tension across the buccopharyngeal membrane, and such tension is necessary for its perforation.
Collapse
Affiliation(s)
- Amanda J G Dickinson
- Department of Biology, Virginia Commonwealth University, Richmond, VA 23284, USA
| |
Collapse
|
9
|
Gu R, Zhang S, Saha SK, Ji Y, Reynolds K, McMahon M, Sun B, Islam M, Trainor PA, Chen Y, Xu Y, Chai Y, Burkart-Waco D, Zhou CJ. Single-cell transcriptomic signatures and gene regulatory networks modulated by Wls in mammalian midline facial formation and clefts. Development 2022; 149:dev200533. [PMID: 35781558 PMCID: PMC9382898 DOI: 10.1242/dev.200533] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 06/21/2022] [Indexed: 07/24/2023]
Abstract
Formation of highly unique and complex facial structures is controlled by genetic programs that are responsible for the precise coordination of three-dimensional tissue morphogenesis. However, the underlying mechanisms governing these processes remain poorly understood. We combined mouse genetic and genomic approaches to define the mechanisms underlying normal and defective midfacial morphogenesis. Conditional inactivation of the Wnt secretion protein Wls in Pax3-expressing lineage cells disrupted frontonasal primordial patterning, cell survival and directional outgrowth, resulting in altered facial structures, including midfacial hypoplasia and midline facial clefts. Single-cell RNA sequencing revealed unique transcriptomic atlases of mesenchymal subpopulations in the midfacial primordia, which are disrupted in the conditional Wls mutants. Differentially expressed genes and cis-regulatory sequence analyses uncovered that Wls modulates and integrates a core gene regulatory network, consisting of key midfacial regulatory transcription factors (including Msx1, Pax3 and Pax7) and their downstream targets (including Wnt, Shh, Tgfβ and retinoic acid signaling components), in a mesenchymal subpopulation of the medial nasal prominences that is responsible for midline facial formation and fusion. These results reveal fundamental mechanisms underlying mammalian midfacial morphogenesis and related defects at single-cell resolution.
Collapse
Affiliation(s)
- Ran Gu
- Department of Biochemistry and Molecular Medicine, University of California at Davis, School of Medicine, Sacramento, CA 95817, USA
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children and UC Davis School of Medicine, Sacramento, CA 95817, USA
| | - Shuwen Zhang
- Department of Biochemistry and Molecular Medicine, University of California at Davis, School of Medicine, Sacramento, CA 95817, USA
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children and UC Davis School of Medicine, Sacramento, CA 95817, USA
| | - Subbroto Kumar Saha
- Department of Biochemistry and Molecular Medicine, University of California at Davis, School of Medicine, Sacramento, CA 95817, USA
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children and UC Davis School of Medicine, Sacramento, CA 95817, USA
| | - Yu Ji
- Department of Biochemistry and Molecular Medicine, University of California at Davis, School of Medicine, Sacramento, CA 95817, USA
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children and UC Davis School of Medicine, Sacramento, CA 95817, USA
| | - Kurt Reynolds
- Department of Biochemistry and Molecular Medicine, University of California at Davis, School of Medicine, Sacramento, CA 95817, USA
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children and UC Davis School of Medicine, Sacramento, CA 95817, USA
| | - Moira McMahon
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children and UC Davis School of Medicine, Sacramento, CA 95817, USA
| | - Bo Sun
- Department of Biochemistry and Molecular Medicine, University of California at Davis, School of Medicine, Sacramento, CA 95817, USA
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children and UC Davis School of Medicine, Sacramento, CA 95817, USA
| | - Mohammad Islam
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children and UC Davis School of Medicine, Sacramento, CA 95817, USA
| | - Paul A. Trainor
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - YiPing Chen
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118, USA
| | - Ying Xu
- Can-SU Genomic Resource Center, Medical College of Soochow University, Suzhou 215006, China
| | - Yang Chai
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA 90033, USA
| | - Diana Burkart-Waco
- DNA Technologies and Expression Analysis Core, Genome Center, University of California, Davis, California 95616, USA
| | - Chengji J. Zhou
- Department of Biochemistry and Molecular Medicine, University of California at Davis, School of Medicine, Sacramento, CA 95817, USA
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children and UC Davis School of Medicine, Sacramento, CA 95817, USA
| |
Collapse
|
10
|
Dickinson AJG, Turner SD, Wahl S, Kennedy AE, Wyatt BH, Howton DA. E-liquids and vanillin flavoring disrupts retinoic acid signaling and causes craniofacial defects in Xenopus embryos. Dev Biol 2022; 481:14-29. [PMID: 34543654 PMCID: PMC8665092 DOI: 10.1016/j.ydbio.2021.09.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 09/08/2021] [Accepted: 09/11/2021] [Indexed: 01/03/2023]
Abstract
Environmental teratogens such as smoking are known risk factors for developmental disorders such as cleft palate. While smoking rates have declined, a new type of smoking, called vaping is on the rise. Vaping is the use of e-cigarettes to vaporize and inhale an e-liquid containing nicotine and food-like flavors. There is the potential that, like smoking, vaping could also pose a danger to the developing human. Rather than waiting for epidemiological and mammalian studies, we have turned to an aquatic developmental model, Xenopus laevis, to more quickly assess whether e-liquids contain teratogens that could lead to craniofacial malformations. Xenopus, like zebrafish, has the benefit of being a well-established developmental model and has also been effective in predicting whether a chemical could be a teratogen. We have determined that embryonic exposure to dessert flavored e-liquids can cause craniofacial abnormalities, including an orofacial cleft in Xenopus. To better understand the underlying mechanisms contributing to these defects, transcriptomic analysis of the facial tissues of embryos exposed to a representative dessert flavored e-liquid vapor extract was performed. Analysis of differentially expressed genes in these embryos revealed several genes associated with retinoic acid metabolism or the signaling pathway. Consistently, retinoic acid receptor inhibition phenocopied the craniofacial defects as those embryos exposed to the vapor extract of the e-liquid. Such malformations also correlated with a group of common differentially expressed genes, two of which are associated with midface birth defects in humans. Further, e-liquid exposure sensitized embryos to forming craniofacial malformations when they already had depressed retinoic acid signaling. Moreover, 13-cis-retinoic acid treatment could significantly reduce the e-liquid induced malformation in the midface. Such results suggest the possibility of an interaction between retinoic acid signaling and e-liquid exposure. One of the most popular and concentrated flavoring chemicals in dessert flavored e-liquids is vanillin. Xenopus embryos exposed to this chemical closely resembled embryos exposed to dessert-like e-liquids and a retinoic acid receptor antagonist. In summary, we determined that e-liquid chemicals, in particular vanillin, can cause craniofacial defects potentially by dysregulating retinoic acid signaling. This work warrants the evaluation of vanillin and other such flavoring additives in e-liquids on mammalian development.
Collapse
Affiliation(s)
| | - Stephen D Turner
- Department of Public Health Sciences, University of Virginia School of Medicine, Charlottesville, VA, USA; Signature Science LLC, Charlottesville, VA, USA
| | - Stacey Wahl
- Research and Education Department, Tompkins-McCaw Library for the Health Sciences, Virginia Commonwealth University, Richmond, VA, USA
| | - Allyson E Kennedy
- Directorate for Computer and Information Science and Engineering, National Science Foundation, Alexandria, VA, USA
| | - Brent H Wyatt
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, 27607, USA
| | - Deborah A Howton
- Department of Biology, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
11
|
Neves VCM, Pugh J, Savulescu J. Beyond oral hygiene, are capacity-altering, biologically based interventions within the moral domain of dentistry? Br Dent J 2021; 231:277-280. [PMID: 34508196 DOI: 10.1038/s41415-021-3335-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 03/25/2021] [Indexed: 11/09/2022]
Abstract
Oral diseases such as dental caries (DC) and periodontitis are widely prevalent, and existing approaches to managing these conditions have only a limited effect. Accordingly, there is growing interest in the development of novel biological interventions (including, among others, CRISPR-Cas9) that might, in the future, be used to prevent the development of or cure these conditions. However, in addition to familiar concerns about using biological interventions in children who cannot provide valid consent, it is not clear whether the provision of these interventions would fall within the proper domain of dentistry. In this opinion paper, we defend the view that the provision of reasonably safe and effective novel biological interventions aimed at preventing DC and periodontitis should be understood to fall within the proper domain of dentistry. To do so, we first argue that their use would be consistent with existing practice in dentistry. We then argue that: i) they may substantially increase the recipient's wellbeing and future autonomy; and ii) that their use could constitute a form of indirect preventative medicine by addressing a threat to systemic health.
Collapse
Affiliation(s)
- Vitor C M Neves
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, UK.
| | - Jonathan Pugh
- Oxford Uehiro Centre for Practical Ethics, University of Oxford, UK
| | - Julian Savulescu
- Oxford Uehiro Centre for Practical Ethics, University of Oxford, UK
| |
Collapse
|
12
|
Wyatt BH, Raymond TO, Lansdon LA, Darbro BW, Murray JC, Manak JR, Dickinson AJG. Using an aquatic model, Xenopus laevis, to uncover the role of chromodomain 1 in craniofacial disorders. Genesis 2021; 59:e23394. [PMID: 32918369 PMCID: PMC10701884 DOI: 10.1002/dvg.23394] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 08/04/2020] [Accepted: 08/08/2020] [Indexed: 12/14/2022]
Abstract
The chromodomain family member chromodomain 1 (CHD1) has been shown to have numerous critical molecular functions including transcriptional regulation, splicing, and DNA repair. Complete loss of function of this gene is not compatible with life. On the other hand, missense and copy number variants of CHD1 can result in intellectual disabilities and craniofacial malformations in human patients including cleft palate and Pilarowski-Bjornsson Syndrome. We have used the aquatic developmental model organism Xenopus laevis, to determine a specific role for Chd1 in such cranioafcial disorders. Protein and gene knockdown techniques in Xenopus, including antisense oligos and mosaic Crispr/Cas9-mediated mutagenesis, recapitulated the craniofacial defects observed in humans. Further analysis indicated that embryos deficient in Chd1 had defects in cranial neural crest development and jaw cartilage morphology. Additionally, flow cytometry and immunohistochemistry revealed that decreased Chd1 resulted in increased in apoptosis in the developing head. Together, these experiments demonstrate that Chd1 is critical for fundamental processes and cell survival in craniofacial development. We also presented evidence that Chd1 is regulated by retinoic acid signaling during craniofacial development. Expression levels of chd1 mRNA, specifically in the head, were increased by RAR agonist exposure and decreased upon antagonist treatment. Subphenotypic levels of an RAR antagonist and Chd1 morpholinos synergized to result in orofacial defects. Further, RAR DNA binding sequences (RAREs) were detected in chd1 regulatory regions by bioinformatic analysis. In summary, by combining human genetics and experiments in an aquatic model we now have a better understanding of the role of CHD1 in craniofacial disorders.
Collapse
Affiliation(s)
- Brent H. Wyatt
- Department of Biology, Virginia Commonwealth University, Richmond, Virginia
| | - Thomas O. Raymond
- Department of Biology, Virginia Commonwealth University, Richmond, Virginia
| | - Lisa A. Lansdon
- Department of Biology, University of Iowa, Iowa City, Iowa
- Department of Pathology and Laboratory Medicine, Children’s Mercy Hospital, Kansas City, Missouri
| | | | | | - John Robert Manak
- Department of Biology, University of Iowa, Iowa City, Iowa
- Department of Pediatrics, University of Iowa, Iowa City, Iowa
| | | |
Collapse
|
13
|
McGirr JA, Martin CH. Few Fixed Variants between Trophic Specialist Pupfish Species Reveal Candidate Cis-Regulatory Alleles Underlying Rapid Craniofacial Divergence. Mol Biol Evol 2021; 38:405-423. [PMID: 32877534 PMCID: PMC7826174 DOI: 10.1093/molbev/msaa218] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Investigating closely related species that rapidly evolved divergent feeding morphology is a powerful approach to identify genetic variation underlying variation in complex traits. This can also lead to the discovery of novel candidate genes influencing natural and clinical variation in human craniofacial phenotypes. We combined whole-genome resequencing of 258 individuals with 50 transcriptomes to identify candidate cis-acting genetic variation underlying rapidly evolving craniofacial phenotypes within an adaptive radiation of Cyprinodon pupfishes. This radiation consists of a dietary generalist species and two derived trophic niche specialists-a molluscivore and a scale-eating species. Despite extensive morphological divergence, these species only diverged 10 kya and produce fertile hybrids in the laboratory. Out of 9.3 million genome-wide SNPs and 80,012 structural variants, we found very few alleles fixed between species-only 157 SNPs and 87 deletions. Comparing gene expression across 38 purebred F1 offspring sampled at three early developmental stages, we identified 17 fixed variants within 10 kb of 12 genes that were highly differentially expressed between species. By measuring allele-specific expression in F1 hybrids from multiple crosses, we found that the majority of expression divergence between species was explained by trans-regulatory mechanisms. We also found strong evidence for two cis-regulatory alleles affecting expression divergence of two genes with putative effects on skeletal development (dync2li1 and pycr3). These results suggest that SNPs and structural variants contribute to the evolution of novel traits and highlight the utility of the San Salvador Island pupfish system as an evolutionary model for craniofacial development.
Collapse
Affiliation(s)
- Joseph A McGirr
- Environmental Toxicology Department, University of California, Davis, CA
| | - Christopher H Martin
- Department of Integrative Biology and Museum of Vertebrate Zoology, University of California, Berkeley, CA
| |
Collapse
|
14
|
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
|
15
|
Watanabe M, Zhou CJ. Introduction to the special issue on orofacial clefts. Birth Defects Res 2020; 112:1555-1557. [PMID: 33124169 DOI: 10.1002/bdr2.1833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 10/18/2020] [Indexed: 11/11/2022]
Affiliation(s)
- Michiko Watanabe
- Department of Pediatrics, Case Western Reserve University School of Medicine, Rainbow Babies and Children's Hospital, The Congenital Heart Collaborative, Cleveland, Ohio, USA
| | - Chengji J Zhou
- Department of Biochemistry and Molecular Medicine, The Institute for Pediatric Regenerative Medicine of the Shriners Hospitals for Children-Northern California, University of California at Davis, School of Medicine, Sacramento, California, USA
| |
Collapse
|
16
|
Gupta P, Tripathi T, Singh N, Bhutiani N, Rai P, Gopal R. A review of genetics of nasal development and morphological variation. J Family Med Prim Care 2020; 9:1825-1833. [PMID: 32670926 PMCID: PMC7346930 DOI: 10.4103/jfmpc.jfmpc_1265_19] [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: 01/30/2020] [Revised: 03/06/2020] [Accepted: 03/26/2020] [Indexed: 11/04/2022] Open
Abstract
The nose is central in the determination of facial esthetics. The variations in its structural characteristics greatly influence the ultimate dentoskeletal positioning at the end of an orthodontic therapy. A careful insight into its developmental etiology will greatly aid the health care professional in identifying patient's real concern about the facial appearance. This in turn will aid in the fabrication of a better treatment plan regarding the end placement goals for the teeth and jaws in all the three dimensions of space. However, this important structure is often missed as a part of the diagnostic and treatment planning regime owing to the lack of meticulous understanding of its developmental etiology by the orthodontists. The development of the nose in the embryo occurs in pre skeletal and skeletal phases by a well-coordinated and regulated interaction of multiple signaling cascades with the crucial importance of each factor in the entire mechanism. The five key factors, which control frontonasal development are sonic hedgehog (SHH), fibroblast growth factors (FGF), transforming growth factor β (TGFβ), wingless (WNT) proteins, and bone morphogenetic protein (BMP). The recent evidence suggests the association of various nasal dimensions and their related syndromes with multiple genes. The revelation of nasal genetic makeup in totality will aid in ascertaining the direction of growth, which will govern our orthodontic treatment results and will also act as a harbinger for potential genetic editing and tissue engineering. This article describes at length the morphological and genetic aspect of nasal growth and development in light of the gender and racial variability along with the emphasis on the importance of knowing these nasal features with regard to diagnosis and treatment planning in orthodontics.
Collapse
Affiliation(s)
- Prateek Gupta
- Department of Orthodontics and Dentofacial Orthopaedics, Maulana Azad Institute of Dental Sciences, Bahadur Shah ZafarMarg, New Delhi, India
| | - Tulika Tripathi
- Department of Orthodontics and Dentofacial Orthopaedics, Maulana Azad Institute of Dental Sciences, Bahadur Shah ZafarMarg, New Delhi, India
| | - Navneet Singh
- Department of Orthodontics and Dentofacial Orthopaedics, Maulana Azad Institute of Dental Sciences, Bahadur Shah ZafarMarg, New Delhi, India
| | - Neha Bhutiani
- Department of Orthodontics and Dentofacial Orthopaedics, Maulana Azad Institute of Dental Sciences, Bahadur Shah ZafarMarg, New Delhi, India
| | - Priyank Rai
- Department of Orthodontics and Dentofacial Orthopaedics, Maulana Azad Institute of Dental Sciences, Bahadur Shah ZafarMarg, New Delhi, India
| | - Ram Gopal
- Department of Orthodontics and Dentofacial Orthopaedics, Maulana Azad Institute of Dental Sciences, Bahadur Shah ZafarMarg, New Delhi, India
| |
Collapse
|
17
|
Abramyan J. Hedgehog Signaling and Embryonic Craniofacial Disorders. J Dev Biol 2019; 7:E9. [PMID: 31022843 PMCID: PMC6631594 DOI: 10.3390/jdb7020009] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 04/18/2019] [Accepted: 04/23/2019] [Indexed: 02/06/2023] Open
Abstract
Since its initial discovery in a Drosophila mutagenesis screen, the Hedgehog pathway has been revealed to be instrumental in the proper development of the vertebrate face. Vertebrates possess three hedgehog paralogs: Sonic hedgehog (Shh), Indian hedgehog (Ihh), and Desert hedgehog (Dhh). Of the three, Shh has the broadest range of functions both in the face and elsewhere in the embryo, while Ihh and Dhh play more limited roles. The Hedgehog pathway is instrumental from the period of prechordal plate formation early in the embryo, until the fusion of the lip and secondary palate, which complete the major patterning events of the face. Disruption of Hedgehog signaling results in an array of developmental disorders in the face, ranging from minor alterations in the distance between the eyes to more serious conditions such as severe clefting of the lip and palate. Despite its critical role, Hedgehog signaling seems to be disrupted through a number of mechanisms that may either be direct, as in mutation of a downstream target of the Hedgehog ligand, or indirect, such as mutation in a ciliary protein that is otherwise seemingly unrelated to the Hedgehog pathway. A number of teratogens such as alcohol, statins and steroidal alkaloids also disrupt key aspects of Hedgehog signal transduction, leading to developmental defects that are similar, if not identical, to those of Hedgehog pathway mutations. The aim of this review is to highlight the variety of roles that Hedgehog signaling plays in developmental disorders of the vertebrate face.
Collapse
Affiliation(s)
- John Abramyan
- Department of Natural Sciences, University of Michigan-Dearborn, Dearborn, MI 48128, USA.
| |
Collapse
|
18
|
Sudiwala S, Knox SM. The emerging role of cranial nerves in shaping craniofacial development. Genesis 2019; 57:e23282. [PMID: 30628162 DOI: 10.1002/dvg.23282] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 01/05/2019] [Accepted: 01/07/2019] [Indexed: 12/17/2022]
Abstract
Organs and structures of the vertebrate head perform a plethora of tasks including visualization, digestion, vocalization/communication, auditory functions, and respiration in response to neuronal input. This input is primarily derived from afferent and efferent fibers of the cranial nerves (sensory and motor respectively) and efferent fibers of the cervical sympathetic trunk. Despite their essential contribution to the function and integration of processes necessary for survival, how organ innervation is established remains poorly understood. Furthermore, while it has been appreciated for some time that innervation of organs by cranial nerves is regulated in part by secreted factors and cell surface ligands expressed by those organs, whether nerves also regulate the development of facial organs is only beginning to be elucidated. This review will provide an overview of cranial nerve development in relation to the organs they innervate, and outline their known contributions to craniofacial development, thereby providing insight into how nerves may shape the organs they innervate during development. Throughout, the interaction between different cell and tissue types will be highlighted.
Collapse
Affiliation(s)
- Sonia Sudiwala
- Program in Craniofacial Biology, Department of Cell and Tissue Biology, University of California, San Francisco, California
| | - Sarah M Knox
- Program in Craniofacial Biology, Department of Cell and Tissue Biology, University of California, San Francisco, California
| |
Collapse
|
19
|
|
20
|
Transcriptome analysis of Xenopus orofacial tissues deficient in retinoic acid receptor function. BMC Genomics 2018; 19:795. [PMID: 30390632 PMCID: PMC6215681 DOI: 10.1186/s12864-018-5186-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 10/18/2018] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Development of the face and mouth is orchestrated by a large number of transcription factors, signaling pathways and epigenetic regulators. While we know many of these regulators, our understanding of how they interact with each other and implement changes in gene expression during orofacial development is still in its infancy. Therefore, this study focuses on uncovering potential cooperation between transcriptional regulators and one important signaling pathway, retinoic acid, during development of the midface. RESULTS Transcriptome analyses was performed on facial tissues deficient for retinoic acid receptor function at two time points in development; early (35 hpf) just after the neural crest migrates and facial tissues are specified and later (60 hpf) when the mouth has formed and facial structures begin to differentiate. Functional and network analyses revealed that retinoic acid signaling could cooperate with novel epigenetic factors and calcium-NFAT signaling during early orofacial development. At the later stage, retinoic acid may work with WNT and BMP and regulate homeobox containing transcription factors. Finally, there is an overlap in genes dysregulated in Xenopus embryos with median clefts with human genes associated with similar orofacial defects. CONCLUSIONS This study uncovers novel signaling pathways required for orofacial development as well as pathways that could interact with retinoic acid signaling during the formation of the face. We show that frog faces are an important tool for studying orofacial development and birth defects.
Collapse
|
21
|
Rothstein M, Bhattacharya D, Simoes-Costa M. The molecular basis of neural crest axial identity. Dev Biol 2018; 444 Suppl 1:S170-S180. [PMID: 30071217 DOI: 10.1016/j.ydbio.2018.07.026] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 07/27/2018] [Accepted: 07/27/2018] [Indexed: 10/28/2022]
Abstract
The neural crest is a migratory cell population that contributes to multiple tissues and organs during vertebrate embryonic development. It is remarkable in its ability to differentiate into an array of different cell types, including melanocytes, cartilage, bone, smooth muscle, and peripheral nerves. Although neural crest cells are formed along the entire anterior-posterior axis of the developing embryo, they can be divided into distinct subpopulations based on their axial level of origin. These groups of cells, which include the cranial, vagal, trunk, and sacral neural crest, display varied migratory patterns and contribute to multiple derivatives. While these subpopulations have been shown to be mostly plastic and to differentiate according to environmental cues, differences in their intrinsic potentials have also been identified. For instance, the cranial neural crest is unique in its ability to give rise to cartilage and bone. Here, we examine the molecular features that underlie such developmental restrictions and discuss the hypothesis that distinct gene regulatory networks operate in these subpopulations. We also consider how reconstructing the phylogeny of the trunk and cranial neural crest cells impacts our understanding of vertebrate evolution.
Collapse
Affiliation(s)
- Megan Rothstein
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA
| | | | - Marcos Simoes-Costa
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA.
| |
Collapse
|
22
|
Sakagami N, Ono W, Ono N. Diverse contribution of Col2a1-expressing cells to the craniofacial skeletal cell lineages. Orthod Craniofac Res 2018. [PMID: 28643905 DOI: 10.1111/ocr.12168] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
OBJECTIVES Craniofacial skeletal development requires deliberate coordination of two distinct mechanisms of endochondral and intramembranous ossification. Col2a1-expressing cells encompass growth-associated skeletal progenitors in endochondral bones of the limb. The objective of this study was to determine the contribution of Col2a1-expressing cells to the craniofacial skeletal cell lineages. We hypothesize that Col2a1-expressing progenitors significantly contribute to various modes of ossification associated with the craniofacial development. METHODS Cellular fates of Col2a1-expressing cells were studied based on a cre-loxP system using a Col2a1-cre transgene and an R26R-tdTomato reporter allele. We analysed three distinct locations of the craniofacial skeletal complex representing unique ossification mechanisms: the cranial base, the calvaria and the mandibular condyle. RESULTS Col2a1-cre consistently marked a majority of skeletal cells in the cranial base. Interestingly, Col2a1-cre also marked a large number of osteoblasts and suture mesenchymal cells in the calvaria, in addition to chondrocytes in the underlying transient cartilage. In the mandibular condyle, Col2a1-cre marked chondrocytes and osteoblasts only during the growth phase. CONCLUSIONS Col2a1 is expressed by progenitors of the skeletal lineage in canonical endochondral bone formation occurring in the cranial base. In contrast, other ossification mechanisms of the craniofacial complex utilize Col2a1-expressing cells in a different manner, whereby Col2a1 may be expressed in more differentiated or transient cell types of the skeletal lineage.
Collapse
Affiliation(s)
- N Sakagami
- Department of Orthodontics and Pediatric Dentistry, University of Michigan School of Dentistry, Ann Arbor, MI, USA
| | - W Ono
- Department of Orthodontics and Pediatric Dentistry, University of Michigan School of Dentistry, Ann Arbor, MI, USA
| | - N Ono
- Department of Orthodontics and Pediatric Dentistry, University of Michigan School of Dentistry, Ann Arbor, MI, USA
| |
Collapse
|
23
|
Cha S, Lim JE, Park AY, Do JH, Lee SW, Shin C, Cho NH, Kang JO, Nam JM, Kim JS, Woo KM, Lee SH, Kim JY, Oh B. Identification of five novel genetic loci related to facial morphology by genome-wide association studies. BMC Genomics 2018; 19:481. [PMID: 29921221 PMCID: PMC6008943 DOI: 10.1186/s12864-018-4865-9] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Accepted: 06/12/2018] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Face morphology is strongly determined by genetic factors. However, only a small number of genes related to face morphology have been identified to date. Here, we performed a two-stage genome-wide association study (GWAS) of 85 face morphological traits in 7569 Koreans (5643 in the discovery set and 1926 in the replication set). RESULTS In this study, we analyzed 85 facial traits, including facial angles. After discovery GWAS, 128 single nucleotide polymorphisms (SNPs) showing an association of P < 5 × 10- 6 were selected to determine the replication of the associations, and meta-analysis of discovery GWAS and the replication analysis resulted in five genome-wide significant loci. The OSR1-WDR35 [rs7567283, G allele, beta (se) = -0.536 (0.096), P = 2.75 × 10- 8] locus was associated with the facial frontal contour; the HOXD1-MTX2 [rs970797, A allele, beta (se) = 0.015 (0.003), P = 3.97 × 10- 9] and WDR27 [rs3736712, C allele, beta (se) = 0.293 (0.048), P = 8.44 × 10- 10] loci were associated with eye shape; and the SOX9 [rs2193054, C allele, beta (se) (ln-transformed) = -0.007 (0.001), P = 6.17 × 10- 17] and DHX35 [rs2206437, A allele, beta (se) = -0.283 (0.047), P = 1.61 × 10- 9] loci were associated with nose shape. WDR35 and SOX9 were related to known craniofacial malformations, i.e., cranioectodermal dysplasia 2 and campomelic dysplasia, respectively. In addition, we found three independent association signals in the SOX9 locus, and six known loci for nose size and shape were replicated in this study population. Interestingly, four SNPs within these five face morphology-related loci showed discrepancies in allele frequencies among ethnic groups. CONCLUSIONS We identified five novel face morphology loci that were associated with facial frontal contour, nose shape, and eye shape. Our findings provide useful genetic information for the determination of face morphology.
Collapse
Affiliation(s)
- Seongwon Cha
- Future Medicine Division, Korea Institute of Oriental Medicine, Daejeon, 34054, Republic of Korea
| | - Ji Eun Lim
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Ah Yeon Park
- Mibyeong Research Center, Korea Institute of Oriental Medicine, Daejeon, 34054, Republic of Korea
| | - Jun-Hyeong Do
- Future Medicine Division, Korea Institute of Oriental Medicine, Daejeon, 34054, Republic of Korea
| | - Si Woo Lee
- Future Medicine Division, Korea Institute of Oriental Medicine, Daejeon, 34054, Republic of Korea
| | - Chol Shin
- Division of Pulmonary Sleep and Critical Care Medicine, Department of Internal Medicine, Korea University Ansan Hospital and Institute of Human Genomic Study, Korea University Ansan Hospital, Ansan, 15355, Republic of Korea
| | - Nam Han Cho
- Department of Preventive Medicine, Ajou University School of Medicine, Suwon, 16499, Republic of Korea
| | - Ji-One Kang
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Jeong Min Nam
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Jong-Sik Kim
- DNA Forensic Division, Supreme Prosecutors' Office, Seoul, 06590, Republic of Korea
| | - Kwang-Man Woo
- DNA Forensic Division, Supreme Prosecutors' Office, Seoul, 06590, Republic of Korea
| | - Seung-Hwan Lee
- DNA Forensic Division, Supreme Prosecutors' Office, Seoul, 06590, Republic of Korea
| | - Jong Yeol Kim
- KM Fundamental Research Division, Korea Institute of Oriental Medicine, Daejeon, 34054, Republic of Korea
| | - Bermseok Oh
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea.
| |
Collapse
|
24
|
Hong M, Krauss RS. Modeling the complex etiology of holoprosencephaly in mice. AMERICAN JOURNAL OF MEDICAL GENETICS PART C-SEMINARS IN MEDICAL GENETICS 2018; 178:140-150. [PMID: 29749693 DOI: 10.1002/ajmg.c.31611] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 03/28/2018] [Accepted: 04/02/2018] [Indexed: 12/14/2022]
Abstract
Holoprosencephaly (HPE) is a common developmental defect caused by failure to define the midline of the forebrain and/or midface. HPE is associated with heterozygous mutations in Nodal and Sonic hedgehog (SHH) pathway components, but clinical presentation is highly variable, and many mutation carriers are unaffected. It is therefore thought that such mutations interact with more common modifiers, genetic and/or environmental, to produce severe patterning defects. Modifiers are difficult to identify, as their effects are context-dependent and occur within the complex genetic and environmental landscapes that characterize human populations. This has made a full understanding of HPE etiology challenging. We discuss here the use of mice, a genetically tractable model sensitive to teratogens, as a system to address this challenge. Mice carrying mutations in human HPE genes often display wide variations in phenotypic penetrance and expressivity when placed on different genetic backgrounds, demonstrating the existence of silent HPE modifier genes. Studies with mouse lines carrying SHH pathway mutations on appropriate genetic backgrounds have led to identification of both genetic and environmental modifiers that synergize with the mutations to produce a spectrum of HPE phenotypes. These models favor a scenario in which multiple modifying influences-both genetic and environmental, sensitizing and protective-interact with bona fide HPE mutations to grade phenotypic outcomes. Despite the complex interplay of HPE risk factors, mouse models have helped establish some clear concepts in HPE etiology. A combination of mouse and human cohort studies should improve our understanding of this fascinating and medically important issue.
Collapse
Affiliation(s)
- Mingi Hong
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Robert S Krauss
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
25
|
Mllt10 knockout mouse model reveals critical role of Af10-dependent H3K79 methylation in midfacial development. Sci Rep 2017; 7:11922. [PMID: 28931923 PMCID: PMC5607342 DOI: 10.1038/s41598-017-11745-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 08/30/2017] [Indexed: 01/04/2023] Open
Abstract
Epigenetic regulation is required to ensure the precise spatial and temporal pattern of gene expression that is necessary for embryonic development. Although the roles of some epigenetic modifications in embryonic development have been investigated in depth, the role of methylation at lysine 79 (H3K79me) is poorly understood. Dot1L, a unique methyltransferase for H3K79, forms complexes with distinct sets of co-factors. To further understand the role of H3K79me in embryogenesis, we generated a mouse knockout of Mllt10, the gene encoding Af10, one Dot1L complex co-factor. We find homozygous Mllt10 knockout mutants (Mllt10-KO) exhibit midline facial cleft. The midfacial defects of Mllt10-KO embryos correspond to hyperterolism and are associated with reduced proliferation of mesenchyme in developing nasal processes and adjacent tissue. We demonstrate that H3K79me level is significantly decreased in nasal processes of Mllt10-KO embryos. Importantly, we find that expression of AP2α, a gene critical for midfacial development, is directly regulated by Af10-dependent H3K79me, and expression AP2α is reduced specifically in nasal processes of Mllt10-KO embryos. Suppression of H3K79me completely mimicked the Mllt10-KO phenotype. Together these data are the first to demonstrate that Af10-dependent H3K79me is essential for development of nasal processes and adjacent tissues, and consequent midfacial formation.
Collapse
|
26
|
Milstone ZJ, Lawson G, Trivedi CM. Histone deacetylase 1 and 2 are essential for murine neural crest proliferation, pharyngeal arch development, and craniofacial morphogenesis. Dev Dyn 2017; 246:1015-1026. [PMID: 28791750 DOI: 10.1002/dvdy.24563] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2017] [Revised: 08/02/2017] [Accepted: 08/07/2017] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Craniofacial anomalies involve defective pharyngeal arch development and neural crest function. Copy number variation at 1p35, containing histone deacetylase 1 (Hdac1), or 6q21-22, containing Hdac2, are implicated in patients with craniofacial defects, suggesting an important role in guiding neural crest development. However, the roles of Hdac1 and Hdac2 within neural crest cells remain unknown. RESULTS The neural crest and its derivatives express both Hdac1 and Hdac2 during early murine development. Ablation of Hdac1 and Hdac2 within murine neural crest progenitor cells cause severe hemorrhage, atrophic pharyngeal arches, defective head morphogenesis, and complete embryonic lethality. Embryos lacking Hdac1 and Hdac2 in the neural crest exhibit decreased proliferation and increased apoptosis in both the neural tube and the first pharyngeal arch. Mechanistically, loss of Hdac1 and Hdac2 upregulates cyclin-dependent kinase inhibitors Cdkn1a, Cdkn1b, Cdkn1c, Cdkn2b, Cdkn2c, and Tp53 within the first pharyngeal arch. CONCLUSIONS Our results show that Hdac1 and Hdac2 function redundantly within the neural crest to regulate proliferation and the development of the pharyngeal arches by means of repression of cyclin-dependent kinase inhibitors. Developmental Dynamics 246:1015-1026, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Zachary J Milstone
- Cardiovascular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts.,Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Grace Lawson
- Cardiovascular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Chinmay M Trivedi
- Cardiovascular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts.,Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts.,Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Medical School, Worcester, Massachusetts
| |
Collapse
|
27
|
Pelttari K, Mumme M, Barbero A, Martin I. Nasal chondrocytes as a neural crest-derived cell source for regenerative medicine. Curr Opin Biotechnol 2017; 47:1-6. [PMID: 28551498 DOI: 10.1016/j.copbio.2017.05.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 05/08/2017] [Indexed: 12/18/2022]
Abstract
Cells deriving from neural crest are generally acknowledged during embryonic development for their multipotency and plasticity, accounting for their capacity to generate various cell and tissue types even across germ layers. At least partial preservation of some of these properties in adulthood makes neural crest derived cells of large interest for regenerative purposes. Chondrocytes from fully mature nasal septum cartilage in adults are also derivatives of neural crest cells and were recently demonstrated to be able not only to maintain functionality across serial cloning, as surrogate self-renewal test, but also to respond and adapt to heterotopic transplantation sites. Based on these findings, cartilage grafts engineered by nasal chondrocytes were clinically used to reconstitute the nasal alar lobule and to repair articular cartilage defects. This article discusses further perspectives of potential clinical utility for nasal chondrocytes in musculoskeletal regeneration. It then highlights the need to derive deeper understanding of their biological properties in order to inform on possible therapeutic modes of action. This acquired knowledge will help to optimise manufacturing conditions to guarantee defined functional traits associated with safety and therapeutic potency of nasal chondrocytes in regenerative medicine.
Collapse
Affiliation(s)
- Karoliina Pelttari
- Department of Biomedicine, University of Basel, University Hospital of Basel, Switzerland
| | - Marcus Mumme
- Department of Biomedicine, University of Basel, University Hospital of Basel, Switzerland; Clinic for Orthopedics and Traumatology, University Hospital of Basel, Switzerland
| | - Andrea Barbero
- Department of Biomedicine, University of Basel, University Hospital of Basel, Switzerland
| | - Ivan Martin
- Department of Biomedicine, University of Basel, University Hospital of Basel, Switzerland.
| |
Collapse
|
28
|
Gallagher ER, Collett BR, Barron S, Romitti P, Ansley T, Wehby GL. Laterality of Oral Clefts and Academic Achievement. Pediatrics 2017; 139:peds.2016-2662. [PMID: 28053221 PMCID: PMC5260154 DOI: 10.1542/peds.2016-2662] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/28/2016] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Children with isolated oral clefts have lower academic performance when compared with unaffected peers, yet few studies have examined specific attributes of clefts that may modify this risk. Oral clefts have nonrandom laterality, with left-sided clefts being more common than right-sided clefts, a pattern that may be genetically or environmentally influenced. The objective of this study was to evaluate the association between cleft laterality and academic achievement in a population-based sample of children with and without isolated oral clefts. METHODS The study included 292 children with isolated unilateral cleft lip with or without cleft palate identified by using the Iowa Registry for Congenital and Inherited Disorders matched with 908 unaffected classmates. This group provided 1953 child-grade observations for cases and 6829 for classmates. Academic achievement was evaluated by using high-quality standardized test data on multiple academic domains as well as use of special education. RESULTS We found that children with right-sided clefts had similar achievement scores and usage of special education services compared with their unaffected classmates. Children with left-sided clefts had lower reading scores than children with right-sided clefts by nearly 7 percentiles (P < .05). They also had lower scores on all evaluated domains by 4 to 6 percentiles and greater use of special education services by 6 percentage points than their classmates. CONCLUSIONS Children with left-sided clefts had poorer academic performance than their classmates or children with right-sided clefts, who showed similar academic achievement compared with their unaffected classmates.
Collapse
Affiliation(s)
| | - Brent R. Collett
- Psychiatry and Behavioral Sciences, Child Psychiatry at Seattle Children’s Hospital, University of Washington, Seattle, Washington
| | | | | | - Timothy Ansley
- Iowa Testing Programs, and,Psychological and Quantitative Foundations, and
| | - George L. Wehby
- Health Management and Policy, Economics, and Preventive and Community Dentistry, and Public Policy Center, University of Iowa, Iowa City, Iowa; and,National Bureau of Economic Research, Cambridge, Massachusetts
| |
Collapse
|
29
|
Abstract
PURPOSE OF REVIEW Craniofacial disorders are among the most common human birth defects and present an enormous health care and social burden. The development of animal models has been instrumental to investigate fundamental questions in craniofacial biology and this knowledge is critical to understand the etiology and pathogenesis of these disorders. RECENT FINDINGS The vast majority of craniofacial disorders arise from abnormal development of the neural crest, a multipotent and migratory cell population. Therefore, defining the pathogenesis of these conditions starts with a deep understanding of the mechanisms that preside over neural crest formation and its role in craniofacial development. SUMMARY This review discusses several studies using Xenopus embryos to model human craniofacial conditions, and emphasizes the strength of this system to inform important biological processes as they relate to human craniofacial development and disease.
Collapse
|
30
|
Raja JV, Asha ML, Kumar GA, Sattigeri AV, Malhotra D. Craniofacial ciliopathies: An expanding oral disease spectrum - a review of literature and a case report. Indian J Dent 2016; 7:153-157. [PMID: 27795653 PMCID: PMC5015569 DOI: 10.4103/0975-962x.180315] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
For all intents and purposes, craniofacial development is initiated as soon as the anteroposterior axis of an embryo is established. Although the neural crest receives a significant amount of attention, craniofacial tissue has more patterning information than other tissues of the body. New studies have further clarifi ed the contribution of ciliary epithelia as a source of patterning information for the face. In this paper, we review the craniofacial anomalies in patients with ciliopathies, in which orofacial region is a pivotal recognition of the disorder. Also, a case report of a patient with suspected ciliopathy has been presented along with a logical approach for diagnosis of such disorders.
Collapse
Affiliation(s)
- Jigna V Raja
- Department of Oral Medicine and Radiology, Dr. Syamala Reddy Dental College Hospital and Research Center, Bengaluru, Karnataka, India
| | - M L Asha
- Department of Oral Medicine and Radiology, Dr. Syamala Reddy Dental College Hospital and Research Center, Bengaluru, Karnataka, India
| | - G Arun Kumar
- Department of Oral Medicine and Radiology, Dr. Syamala Reddy Dental College Hospital and Research Center, Bengaluru, Karnataka, India
| | - Anupama V Sattigeri
- Department of Oral Medicine and Radiology, Dr. Syamala Reddy Dental College Hospital and Research Center, Bengaluru, Karnataka, India
| | - Diksha Malhotra
- Department of Oral Medicine and Radiology, Dr. Syamala Reddy Dental College Hospital and Research Center, Bengaluru, Karnataka, India
| |
Collapse
|
31
|
Maddin HC, Piekarski N, Sefton EM, Hanken J. Homology of the cranial vault in birds: new insights based on embryonic fate-mapping and character analysis. ROYAL SOCIETY OPEN SCIENCE 2016; 3:160356. [PMID: 27853617 PMCID: PMC5108967 DOI: 10.1098/rsos.160356] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2016] [Accepted: 07/12/2016] [Indexed: 05/24/2023]
Abstract
Bones of the cranial vault appear to be highly conserved among tetrapod vertebrates. Moreover, bones identified with the same name are assumed to be evolutionarily homologous. However, recent developmental studies reveal a key difference in the embryonic origin of cranial vault bones between representatives of two amniote lineages, mammals and birds, thereby challenging this view. In the mouse, the frontal is derived from cranial neural crest (CNC) but the parietal is derived from mesoderm, placing the CNC-mesoderm boundary at the suture between these bones. In the chicken, this boundary is located within the frontal. This difference and related data have led several recent authors to suggest that bones of the avian cranial vault are misidentified and should be renamed. To elucidate this apparent conflict, we fate-mapped CNC and mesoderm in axolotl to reveal the contributions of these two embryonic cell populations to the cranial vault in a urodele amphibian. The CNC-mesoderm boundary in axolotl is located between the frontal and parietal bones, as in the mouse but unlike the chicken. If, however, the avian frontal is regarded instead as a fused frontal and parietal (i.e. frontoparietal) and the parietal as a postparietal, then the cranial vault of birds becomes developmentally and topologically congruent with those of urodeles and mammals. This alternative hypothesis of cranial vault homology is also phylogenetically consistent with data from the tetrapod fossil record, where frontal, parietal and postparietal bones are present in stem lineages of all extant taxa, including birds. It further implies that a postparietal may be present in most non-avian archosaurs, but fused to the parietal or supraoccipital as in many extant mammals.
Collapse
|
32
|
Frohwitter G, Buerger H, VAN Diest PJ, Korsching E, Kleinheinz J, Fillies T. Cytokeratin and protein expression patterns in squamous cell carcinoma of the oral cavity provide evidence for two distinct pathogenetic pathways. Oncol Lett 2016; 12:107-113. [PMID: 27347109 PMCID: PMC4906805 DOI: 10.3892/ol.2016.4588] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 03/24/2016] [Indexed: 01/21/2023] Open
Abstract
Squamous cell carcinoma (SCC) of the oral cavity is a morphological heterogeneous disease. Various cytokeratin (CK) expression patterns with different prognostic values have been described, but little is known concerning the underlying biological cell mechanisms. Therefore, the present study investigated 193 cases of oral SCCs using immunohistochemistry for α/β/γ-catenin, glucose transporter 1, caspase-3, X-linked inhibitor of apoptosis protein, hypoxia inducible factor-1α, carbonic anhydrase 9, heat shock protein (hsp) 70, mast/stem cell growth factor receptor, p21, p27, p16, p53, B-cell lymphoma 6, epidermal growth factor receptor, cyclin D1 and CK1, 5/6, 8/18, 10, 14 and 19. Expression patterns were analyzed with biomathematical permutation analysis. The present results revealed a significant association between the expression of low-molecular weight CK8/18 and 19 and a high-tumor grade, β and γ-catenin expression, deregulated cell cycle proteins and a predominant localization of the tumor on the floor of the mouth. By contrast, expression of high-molecular weight CK1, 5/6, 10 and 14 was significantly associated with the expression of p21 and hsp70. In conclusion, the current study presents evidence for the existence of two parallel pathogenetic pathways in oral SCCs, characterized by the expression of low- and high-molecular weight CKs. Additional studies are required to demonstrate the extent that these results may be used to improve therapeutic regimens.
Collapse
Affiliation(s)
| | - Horst Buerger
- Institute of Pathology, D-33098 Paderborn, Germany; Department of Pathology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Paul J VAN Diest
- Department of Pathology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Eberhard Korsching
- Institute of Bioinformatics, Faculty of Medicine, University of Münster, Germany
| | - Johannes Kleinheinz
- Department of Cranio and Maxillofacial Surgery, University Hospital Muenster, D-48149 Münster, Germany
| | - Thomas Fillies
- Department of Cranio and Maxillofacial Surgery, Marienhospital Stuttgart, D-70199 Stuttgart, Germany
| |
Collapse
|
33
|
Lei R, Zhang K, Wei Y, Chen M, Weinstein LS, Hong Y, Zhu M, Li H, Li H. G-Protein α-Subunit Gsα Is Required for Craniofacial Morphogenesis. PLoS One 2016; 11:e0147535. [PMID: 26859889 PMCID: PMC4747491 DOI: 10.1371/journal.pone.0147535] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 01/05/2016] [Indexed: 02/05/2023] Open
Abstract
The heterotrimeric G protein subunit Gsα couples receptors to activate adenylyl cyclase and is required for the intracellular cAMP response and protein kinase A (PKA) activation. Gsα is ubiquitously expressed in many cell types; however, the role of Gsα in neural crest cells (NCCs) remains unclear. Here we report that NCCs-specific Gsα knockout mice die within hours after birth and exhibit dramatic craniofacial malformations, including hypoplastic maxilla and mandible, cleft palate and craniofacial skeleton defects. Histological and anatomical analysis reveal that the cleft palate in Gsα knockout mice is a secondary defect resulting from craniofacial skeleton deficiencies. In Gsα knockout mice, the morphologies of NCCs-derived cranial nerves are normal, but the development of dorsal root and sympathetic ganglia are impaired. Furthermore, loss of Gsα in NCCs does not affect cranial NCCs migration or cell proliferation, but significantly accelerate osteochondrogenic differentiation. Taken together, our study suggests that Gsα is required for neural crest cells-derived craniofacial development.
Collapse
Affiliation(s)
- Run Lei
- West China Developmental & Stem Cell Institute, West China Second Hospital, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Shenzhen Key Laboratory for Molecular Biology of Neural Development, Laboratory of Developmental and Regenerative biology, Institute of Biomedicine & Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
- SARITEX Center for Stem Cell Engineering Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Chinese Academy of Sciences, Shanghai, China
| | - Ke Zhang
- West China Developmental & Stem Cell Institute, West China Second Hospital, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Shenzhen Key Laboratory for Molecular Biology of Neural Development, Laboratory of Developmental and Regenerative biology, Institute of Biomedicine & Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
- SARITEX Center for Stem Cell Engineering Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Chinese Academy of Sciences, Shanghai, China
| | - Yanxia Wei
- West China Developmental & Stem Cell Institute, West China Second Hospital, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Min Chen
- Metabolic Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Lee S. Weinstein
- Metabolic Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Yang Hong
- Department of Cell Biology & Physiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Minyan Zhu
- SARITEX Center for Stem Cell Engineering Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Chinese Academy of Sciences, Shanghai, China
| | - Hongchang Li
- Shenzhen Key Laboratory for Molecular Biology of Neural Development, Laboratory of Developmental and Regenerative biology, Institute of Biomedicine & Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
- * E-mail: Hongchang Li (HCL); Huashun Li (HSL)
| | - Huashun Li
- West China Developmental & Stem Cell Institute, West China Second Hospital, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- SARITEX Center for Stem Cell Engineering Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Chinese Academy of Sciences, Shanghai, China
- * E-mail: Hongchang Li (HCL); Huashun Li (HSL)
| |
Collapse
|
34
|
Namkoong B, Güven S, Ramesan S, Liaudanskaya V, Abzhanov A, Demirci U. Recapitulating cranial osteogenesis with neural crest cells in 3-D microenvironments. Acta Biomater 2016; 31:301-311. [PMID: 26675129 DOI: 10.1016/j.actbio.2015.12.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 11/11/2015] [Accepted: 12/02/2015] [Indexed: 01/20/2023]
Abstract
The experimental systems that recapitulate the complexity of native tissues and enable precise control over the microenvironment are becoming essential for the pre-clinical tests of therapeutics and tissue engineering. Here, we described a strategy to develop an in vitro platform to study the developmental biology of craniofacial osteogenesis. In this study, we directly osteo-differentiated cranial neural crest cells (CNCCs) in a 3-D in vitro bioengineered microenvironment. Cells were encapsulated in the gelatin-based photo-crosslinkable hydrogel and cultured up to three weeks. We demonstrated that this platform allows efficient differentiation of p75 positive CNCCs to cells expressing osteogenic markers corresponding to the sequential developmental phases of intramembranous ossification. During the course of culture, we observed a decrease in the expression of early osteogenic marker Runx2, while the other mature osteoblast and osteocyte markers such as Osterix, Osteocalcin, Osteopontin and Bone sialoprotein increased. We analyzed the ossification of the secreted matrix with alkaline phosphatase and quantified the newly secreted hydroxyapatite. The Field Emission Scanning Electron Microscope (FESEM) images of the bioengineered hydrogel constructs revealed the native-like osteocytes, mature osteoblasts, and cranial bone tissue morphologies with canaliculus-like intercellular connections. This platform provides a broadly applicable model system to potentially study diseases involving primarily embryonic craniofacial bone disorders, where direct diagnosis and adequate animal disease models are limited.
Collapse
Affiliation(s)
- Bumjin Namkoong
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA; Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Sinan Güven
- Demirci BAMM Labs, Canary Center at Stanford for Early Cancer Detection, Department of Radiology, Department of Electrical Engineering (By courtesy), Stanford School of Medicine, Palo Alto, CA 94304, USA; Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus, Balcova, 35350 Izmir, Turkey
| | - Shwathy Ramesan
- Demirci BAMM Labs, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Volha Liaudanskaya
- Demirci BAMM Labs, Canary Center at Stanford for Early Cancer Detection, Department of Radiology, Department of Electrical Engineering (By courtesy), Stanford School of Medicine, Palo Alto, CA 94304, USA
| | - Arhat Abzhanov
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA; Current address: Department of Life Sciences, Imperial College London, Silwood Park Campus Buckhurst Road, Ascot, Berkshire SL5 7PY, United Kingdom; Current address: Natural History Museum, Cromwell Road, London SW7 5BD, United Kingdom.
| | - Utkan Demirci
- Demirci BAMM Labs, Canary Center at Stanford for Early Cancer Detection, Department of Radiology, Department of Electrical Engineering (By courtesy), Stanford School of Medicine, Palo Alto, CA 94304, USA; Demirci BAMM Labs, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
35
|
Gross JB, Stahl BA, Powers AK, Carlson BM. Natural bone fragmentation in the blind cave-dwelling fish, Astyanax mexicanus: candidate gene identification through integrative comparative genomics. Evol Dev 2016; 18:7-18. [PMID: 26153732 PMCID: PMC5226847 DOI: 10.1111/ede.12131] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Animals that colonize dark and nutrient-poor subterranean environments evolve numerous extreme phenotypes. These include dramatic changes to the craniofacial complex, many of which are under genetic control. These phenotypes can demonstrate asymmetric genetic signals wherein a QTL is detected on one side of the face but not the other. The causative gene(s) underlying QTL are difficult to identify with limited genomic resources. We approached this task by searching for candidate genes mediating fragmentation of the third suborbital bone (SO3) directly inferior to the orbit of the eye. We integrated positional genomic information using emerging Astyanax resources, and linked these intervals to homologous (syntenic) regions of the Danio rerio genome. We identified a discrete, approximately 6 Mb, conserved region wherein the gene causing SO3 fragmentation likely resides. We interrogated this interval for genes demonstrating significant differential expression using mRNA-seq analysis of cave and surface morphs across life history. We then assessed genes with known roles in craniofacial evolution and development based on GO term annotation. Finally, we screened coding sequence alterations in this region, identifying two key genes: transforming growth factor β3 (tgfb3) and bone morphogenetic protein 4 (bmp4). Of these candidates, tgfb3 is most promising as it demonstrates significant differential expression across multiple stages of development, maps close (<1 Mb) to the fragmentation critical locus, and is implicated in a variety of other animal systems (including humans) in non-syndromic clefting and malformations of the cranial sutures. Both abnormalities are analogous to the failure-to-fuse phenotype that we observe in SO3 fragmentation. This integrative approach will enable discovery of the causative genetic lesions leading to complex craniofacial features analogous to human craniofacial disorders. This work underscores the value of cave-dwelling fish as a powerful evolutionary model of craniofacial disease, and demonstrates the power of integrative system-level studies for informing the genetic basis of craniofacial aberrations in nature.
Collapse
Affiliation(s)
- Joshua B. Gross
- Department of Biological Sciences, University of Cincinnati, 312 Clifton Court, Cincinnati, Ohio 45221, USA
| | - Bethany A. Stahl
- Department of Biological Sciences, University of Cincinnati, 312 Clifton Court, Cincinnati, Ohio 45221, USA
| | - Amanda K. Powers
- Department of Biological Sciences, University of Cincinnati, 312 Clifton Court, Cincinnati, Ohio 45221, USA
| | - Brian M. Carlson
- Department of Biological Sciences, University of Cincinnati, 312 Clifton Court, Cincinnati, Ohio 45221, USA
| |
Collapse
|
36
|
Suzuki A, Sangani DR, Ansari A, Iwata J. Molecular mechanisms of midfacial developmental defects. Dev Dyn 2015; 245:276-93. [PMID: 26562615 DOI: 10.1002/dvdy.24368] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Revised: 10/19/2015] [Accepted: 11/01/2015] [Indexed: 12/22/2022] Open
Abstract
The morphogenesis of midfacial processes requires the coordination of a variety of cellular functions of both mesenchymal and epithelial cells to develop complex structures. Any failure or delay in midfacial development as well as any abnormal fusion of the medial and lateral nasal and maxillary prominences will result in developmental defects in the midface with a varying degree of severity, including cleft, hypoplasia, and midline expansion. Despite the advances in human genome sequencing technology, the causes of nearly 70% of all birth defects, which include midfacial development defects, remain unknown. Recent studies in animal models have highlighted the importance of specific signaling cascades and genetic-environmental interactions in the development of the midfacial region. This review will summarize the current understanding of the morphogenetic processes and molecular mechanisms underlying midfacial birth defects based on mouse models with midfacial developmental abnormalities.
Collapse
Affiliation(s)
- Akiko Suzuki
- Department of Diagnostic & Biomedical Sciences, School of Dentistry, The University of Texas Health Science Center at Houston, Houston, Texas.,Center for Craniofacial Research, The University of Texas Health Science Center at Houston, Houston, Texas
| | - Dhruvee R Sangani
- Center for Craniofacial Research, The University of Texas Health Science Center at Houston, Houston, Texas
| | - Afreen Ansari
- Center for Craniofacial Research, The University of Texas Health Science Center at Houston, Houston, Texas
| | - Junichi Iwata
- Department of Diagnostic & Biomedical Sciences, School of Dentistry, The University of Texas Health Science Center at Houston, Houston, Texas.,Center for Craniofacial Research, The University of Texas Health Science Center at Houston, Houston, Texas.,The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas
| |
Collapse
|
37
|
Ahi EP, Steinhäuser SS, Pálsson A, Franzdóttir SR, Snorrason SS, Maier VH, Jónsson ZO. Differential expression of the aryl hydrocarbon receptor pathway associates with craniofacial polymorphism in sympatric Arctic charr. EvoDevo 2015; 6:27. [PMID: 26388986 PMCID: PMC4574265 DOI: 10.1186/s13227-015-0022-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 09/04/2015] [Indexed: 12/03/2022] Open
Abstract
Background The developmental basis of craniofacial morphology hinges on interactions of numerous signalling systems. Extensive craniofacial variation in the polymorphic Arctic charr, a member of the salmonid family, from Lake Thingvallavatn (Iceland), offers opportunities to find and study such signalling pathways and their key regulators, thereby shedding light on the developmental pathways, and the genetics of trophic divergence. Results To identify genes involved in the craniofacial differences between benthic and limnetic Arctic charr, we used transcriptome data from different morphs, spanning early development, together with data on craniofacial expression patterns and skeletogenesis in model vertebrate species. Out of 20 genes identified, 7 showed lower gene expression in benthic than in limnetic charr morphs. We had previously identified a conserved gene network involved in extracellular matrix (ECM) organization and skeletogenesis, showing higher expression in developing craniofacial elements of benthic than in limnetic Arctic charr morphs. The present study adds a second set of genes constituting an expanded gene network with strong, benthic–limnetic differential expression. To identify putative upstream regulators, we performed knowledge-based motif enrichment analyses on the regulatory sequences of the identified genes which yielded potential binding sites for a set of known transcription factors (TFs). Of the 8 TFs that we examined using qPCR, two (Ahr2b and Ap2) were found to be differentially expressed between benthic and limnetic charr. Expression analysis of several known AhR targets indicated higher activity of the AhR pathway during craniofacial development in benthic charr morphotypes. Conclusion These results suggest a key role of the aryl hydrocarbon receptor (AhR) pathway in the observed craniofacial differences between distinct charr morphotypes. Electronic supplementary material The online version of this article (doi:10.1186/s13227-015-0022-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ehsan Pashay Ahi
- Institute of Life and Environmental Sciences, University of Iceland, Sturlugata 7, 101 Reykjavik, Iceland
| | - Sophie S Steinhäuser
- Biomedical Center, University of Iceland, Vatnsmýrarvegur 16, 101 Reykjavik, Iceland
| | - Arnar Pálsson
- Institute of Life and Environmental Sciences, University of Iceland, Sturlugata 7, 101 Reykjavik, Iceland ; Biomedical Center, University of Iceland, Vatnsmýrarvegur 16, 101 Reykjavik, Iceland
| | - Sigrídur Rut Franzdóttir
- Institute of Life and Environmental Sciences, University of Iceland, Sturlugata 7, 101 Reykjavik, Iceland
| | - Sigurdur S Snorrason
- Institute of Life and Environmental Sciences, University of Iceland, Sturlugata 7, 101 Reykjavik, Iceland
| | - Valerie H Maier
- Institute of Life and Environmental Sciences, University of Iceland, Sturlugata 7, 101 Reykjavik, Iceland ; Biomedical Center, University of Iceland, Vatnsmýrarvegur 16, 101 Reykjavik, Iceland
| | - Zophonías O Jónsson
- Institute of Life and Environmental Sciences, University of Iceland, Sturlugata 7, 101 Reykjavik, Iceland ; Biomedical Center, University of Iceland, Vatnsmýrarvegur 16, 101 Reykjavik, Iceland
| |
Collapse
|
38
|
He F, Soriano P. Sox10ER(T2) CreER(T2) mice enable tracing of distinct neural crest cell populations. Dev Dyn 2015; 244:1394-403. [PMID: 26250625 DOI: 10.1002/dvdy.24320] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Revised: 07/28/2015] [Accepted: 07/28/2015] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Neural crest cells play an important role in craniofacial morphogenesis and many other developmental processes. The formation of neural crest cells (NCCs) in vivo is a highly dynamic process and remains to be fully understood. RESULTS To investigate the spatiotemporal patterning of NCCs in vivo, we have generated Sox10ER(T2) CreER(T2) (SECE) mice, a transgenic line driving inducible Cre expression in NCCs. Inducing Cre activity at different stages triggered reporter expression in distinct NCC populations in SECE; R26R mice. By optimizing the timing and dosage of tamoxifen administration, we controlled Cre expression specifically in cranial NCCs. Using this approach, we demonstrate an important role for PDGFRα in cranial NCCs mitosis within the mandibular processes. Further reducing Cre activity within the cranial NCCs of SECE; R26R embryos revealed that SECE labels preferentially progenitors of medial nasal process (MNP) rather than the lateral nasal process (LNP), before their formation from the frontonasal prominence (FNP). CONCLUSIONS Our results indicate that NCCs are formed sequentially from rostral to caudal regions along the neural tube. These findings also suggest that NCCs within the FNP become specified regionally and genetically before they divide into MNP and LNP.
Collapse
Affiliation(s)
- Fenglei He
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Philippe Soriano
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
39
|
Cortés CR, Metzis V, Wicking C. Unmasking the ciliopathies: craniofacial defects and the primary cilium. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2015; 4:637-53. [PMID: 26173831 DOI: 10.1002/wdev.199] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 05/19/2015] [Accepted: 05/30/2015] [Indexed: 12/29/2022]
Abstract
Over the past decade, the primary cilium has emerged as a pivotal sensory organelle that acts as a major signaling hub for a number of developmental signaling pathways. In that time, a vast number of proteins involved in trafficking and signaling have been linked to ciliary assembly and/or function, demonstrating the importance of this organelle during embryonic development. Given the central role of the primary cilium in regulating developmental signaling, it is not surprising that its dysfunction results in widespread defects in the embryo, leading to an expanding class of human congenital disorders known as ciliopathies. These disorders are individually rare and phenotypically variable, but together they affect virtually every vertebrate organ system. Features of ciliopathies that are often overlooked, but which are being reported with increasing frequency, are craniofacial abnormalities, ranging from subtle midline defects to full-blown orofacial clefting. The challenge moving forward is to understand the primary mechanism of disease given the link between the primary cilium and a number of developmental signaling pathways (such as hedgehog, platelet-derived growth factor, and WNT signaling) that are essential for craniofacial development. Here, we provide an overview of the diversity of craniofacial abnormalities present in the ciliopathy spectrum, and reveal those defects in common across multiple disorders. Further, we discuss the molecular defects and potential signaling perturbations underlying these anomalies. This provides insight into the mechanisms leading to ciliopathy phenotypes more generally and highlights the prevalence of widespread dysmorphologies resulting from cilia dysfunction.
Collapse
Affiliation(s)
- Claudio R Cortés
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Vicki Metzis
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Carol Wicking
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| |
Collapse
|
40
|
Pelttari K, Pippenger B, Mumme M, Feliciano S, Scotti C, Mainil-Varlet P, Procino A, von Rechenberg B, Schwamborn T, Jakob M, Cillo C, Barbero A, Martin I. Adult human neural crest-derived cells for articular cartilage repair. Sci Transl Med 2015; 6:251ra119. [PMID: 25163479 DOI: 10.1126/scitranslmed.3009688] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In embryonic models and stem cell systems, mesenchymal cells derived from the neuroectoderm can be distinguished from mesoderm-derived cells by their Hox-negative profile--a phenotype associated with enhanced capacity of tissue regeneration. We investigated whether developmental origin and Hox negativity correlated with self-renewal and environmental plasticity also in differentiated cells from adults. Using hyaline cartilage as a model, we showed that adult human neuroectoderm-derived nasal chondrocytes (NCs) can be constitutively distinguished from mesoderm-derived articular chondrocytes (ACs) by lack of expression of specific HOX genes, including HOXC4 and HOXD8. In contrast to ACs, serially cloned NCs could be continuously reverted from differentiated to dedifferentiated states, conserving the ability to form cartilage tissue in vitro and in vivo. NCs could also be reprogrammed to stably express Hox genes typical of ACs upon implantation into goat articular cartilage defects, directly contributing to cartilage repair. Our findings identify previously unrecognized regenerative properties of HOX-negative differentiated neuroectoderm cells in adults, implying a role for NCs in the unmet clinical challenge of articular cartilage repair. An ongoing phase 1 clinical trial preliminarily indicated the safety and feasibility of autologous NC-based engineered tissues for the treatment of traumatic articular cartilage lesions.
Collapse
Affiliation(s)
- Karoliina Pelttari
- Departments of Surgery and of Biomedicine, University Hospital Basel, University of Basel, Hebelstrasse 20, 4031 Basel, Switzerland
| | - Benjamin Pippenger
- Departments of Surgery and of Biomedicine, University Hospital Basel, University of Basel, Hebelstrasse 20, 4031 Basel, Switzerland
| | - Marcus Mumme
- Departments of Surgery and of Biomedicine, University Hospital Basel, University of Basel, Hebelstrasse 20, 4031 Basel, Switzerland
| | - Sandra Feliciano
- Departments of Surgery and of Biomedicine, University Hospital Basel, University of Basel, Hebelstrasse 20, 4031 Basel, Switzerland
| | - Celeste Scotti
- Istituto Di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Ortopedico Galeazzi, Via R. Galeazzi 4, 20161 Milano, Italy
| | - Pierre Mainil-Varlet
- AGINKO Research AG, Route de l'ancienne Papeterie, P. O. Box 30, 1723 Marly, Switzerland
| | - Alfredo Procino
- Department of Medicine and Surgery, Federico II Medical School, Via S. Pansini 5, 80131 Napoli, Italy
| | - Brigitte von Rechenberg
- Musculoskeletal Research Unit, Equine Hospital, University of Zurich, Winterthurerstrasse 260, 8057 Zurich, Switzerland
| | | | - Marcel Jakob
- Departments of Surgery and of Biomedicine, University Hospital Basel, University of Basel, Hebelstrasse 20, 4031 Basel, Switzerland
| | - Clemente Cillo
- Department of Medicine and Surgery, Federico II Medical School, Via S. Pansini 5, 80131 Napoli, Italy
| | - Andrea Barbero
- Departments of Surgery and of Biomedicine, University Hospital Basel, University of Basel, Hebelstrasse 20, 4031 Basel, Switzerland
| | - Ivan Martin
- Departments of Surgery and of Biomedicine, University Hospital Basel, University of Basel, Hebelstrasse 20, 4031 Basel, Switzerland.
| |
Collapse
|
41
|
Bhanu U, Kulkarni R, Boaz K, Srikant N. Pigmented odontogenic tumors: Adding color to diagnosis? J Oral Maxillofac Pathol 2015; 18:398-402. [PMID: 25948995 PMCID: PMC4409185 DOI: 10.4103/0973-029x.151325] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Accepted: 01/05/2015] [Indexed: 12/05/2022] Open
Abstract
Melanocytes are neural crest derivatives that exhibit a ubiquitous presence in the epidermis. They determine the complexion of an individual and most importantly, provide a barrier against ultraviolet radiations from the sun. Their presence in the oral cavity is a consistent finding, especially in the gingiva and buccal mucosa of the dark complexioned. Melanocytes occasionally form a part of the histology of a variety of odontogenic cysts and tumors. How these cells make their way into the lesional tissue and the diagnostic relevance of their presence remains elusive. This write up attempts to trace the path melanocytes take to find themselves within odontogenic tumors and also offer possible explanations for the same.
Collapse
Affiliation(s)
- Udhay Bhanu
- Department of Oral Pathology and Microbiology, Manipal College of Dental Sciences, Manipal University, Mangalore, India
| | - Rasika Kulkarni
- Department of Oral Pathology and Microbiology, Manipal College of Dental Sciences, Manipal University, Mangalore, India
| | - Karen Boaz
- Department of Oral Pathology and Microbiology, Manipal College of Dental Sciences, Manipal University, Mangalore, India
| | - N Srikant
- Department of Oral Pathology and Microbiology, Manipal College of Dental Sciences, Manipal University, Mangalore, India
| |
Collapse
|
42
|
Wu BT, Wen SH, Hwang SPL, Huang CJ, Kuan YS. Control of Wnt5b secretion by Wntless modulates chondrogenic cell proliferation through fine-tuning fgf3 expression. J Cell Sci 2015; 128:2328-39. [PMID: 25934698 DOI: 10.1242/jcs.167403] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 04/21/2015] [Indexed: 01/22/2023] Open
Abstract
Wnts and Fgfs regulate various tissues development in vertebrates. However, how regional Wnt or Fgf activities are established and how they interact in any given developmental event is elusive. Here, we investigated the Wnt-mediated craniofacial cartilage development in zebrafish and found that fgf3 expression in the pharyngeal pouches is differentially reduced along the anteroposterior axis in wnt5b mutants and wntless (wls) morphants, but its expression is normal in wnt9a and wnt11 morphants. Introducing fgf3 mRNAs rescued the cartilage defects in Wnt5b- and Wls-deficient larvae. In wls morphants, endogenous Wls expression is not detectable but maternally deposited Wls is present in eggs, which might account for the lack of axis defects in wls morphants. Secretion of endogenous Wnt5b but not Wnt11 was affected in the pharyngeal tissue of Wls morphants, indicating that Wls is not involved in every Wnt secretion event. Furthermore, cell proliferation but not apoptosis in the developing jaw was affected in Wnt5b- and Wls-deficient embryos. Therefore, Wnt5b requires Wls for its secretion and regulates the proliferation of chondrogenic cells through fine-tuning the expression of fgf3 during jaw cartilage development.
Collapse
Affiliation(s)
- Bo-Tsung Wu
- Institute of Biochemical Sciences, College of Life Science, National Taiwan University, Taipei 10617, Taiwan Institute of Biological Chemistry, Academia Sinica, Taipei 11529, Taiwan
| | - Shih-Hsien Wen
- Institute of Biochemical Sciences, College of Life Science, National Taiwan University, Taipei 10617, Taiwan Institute of Biological Chemistry, Academia Sinica, Taipei 11529, Taiwan
| | - Sheng-Ping L Hwang
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 11529, Taiwan
| | - Chang-Jen Huang
- Institute of Biochemical Sciences, College of Life Science, National Taiwan University, Taipei 10617, Taiwan Institute of Biological Chemistry, Academia Sinica, Taipei 11529, Taiwan
| | - Yung-Shu Kuan
- Institute of Biochemical Sciences, College of Life Science, National Taiwan University, Taipei 10617, Taiwan Institute of Biological Chemistry, Academia Sinica, Taipei 11529, Taiwan Center for System Biology, National Taiwan University, Taipei 10617, Taiwan
| |
Collapse
|
43
|
Differential Gene Expression in the Otic Capsule and the Middle Ear—An Annotation of Bone-Related Signaling Genes. Otol Neurotol 2015; 36:727-32. [DOI: 10.1097/mao.0000000000000664] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
44
|
Abstract
The developmental paths that lead to the formation of skeletal muscles in the head are distinct from those operating in the trunk. Craniofacial muscles are associated with head and neck structures. In the embryo, these structures derive from distinct mesoderm populations. Distinct genetic programs regulate different groups of muscles within the head to generate diverse muscle specifications. Developmental and lineage studies in vertebrates and invertebrates demonstrated an overlap in progenitor populations derived from the pharyngeal mesoderm that contribute to certain head muscles and the heart. These studies reveal that the genetic program controlling pharyngeal muscles overlaps with that of the heart. Indeed cardiac and craniofacial birth defects are often linked. Recent studies suggest that early chordates, the last common ancestor of tunicates and vertebrates, had an ancestral pharyngeal mesoderm lineage that later during evolution gave rise to both heart and craniofacial structures. This chapter summarizes studies related to the origins, signaling, genetics, and evolution of the head musculature, highlighting its heterogeneous characteristics in all these aspects.
Collapse
Affiliation(s)
- Eldad Tzahor
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, 76100, Israel,
| |
Collapse
|
45
|
Lindholm M. DNA Dispose, but Subjects Decide. Learning and the Extended Synthesis. BIOSEMIOTICS 2015; 8:443-461. [PMID: 26640605 PMCID: PMC4661179 DOI: 10.1007/s12304-015-9242-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 05/11/2015] [Indexed: 05/23/2023]
Abstract
Adaptation by means of natural selection depends on the ability of populations to maintain variation in heritable traits. According to the Modern Synthesis this variation is sustained by mutations and genetic drift. Epigenetics, evodevo, niche construction and cultural factors have more recently been shown to contribute to heritable variation, however, leading an increasing number of biologists to call for an extended view of speciation and evolution. An additional common feature across the animal kingdom is learning, defined as the ability to change behavior according to novel experiences or skills. Learning constitutes an additional source for phenotypic variation, and change in behavior may induce long lasting shifts in fitness, and hence favor evolutionary novelties. Based on published studies, I demonstrate how learning about food, mate choice and habitats has contributed substantially to speciation in the canonical story of Darwin's finches on the Galapagos Islands. Learning cannot be reduced to genetics, because it demands decisions, which requires a subject. Evolutionary novelties may hence emerge both from shifts in allelic frequencies and from shifts in learned, subject driven behavior. The existence of two principally different sources of variation also prevents the Modern Synthesis from self-referring explanations.
Collapse
Affiliation(s)
- Markus Lindholm
- />Norwegian Institute for Water Research (NIVA), Gaustadalléen 21, 0349 Oslo, Norway
- />Rudolf Steiner University College, Dahls gate 30, 0260 Oslo, Norway
| |
Collapse
|
46
|
Ahi EP, Kapralova KH, Pálsson A, Maier VH, Gudbrandsson J, Snorrason SS, Jónsson ZO, Franzdóttir SR. Transcriptional dynamics of a conserved gene expression network associated with craniofacial divergence in Arctic charr. EvoDevo 2014; 5:40. [PMID: 25419450 PMCID: PMC4240837 DOI: 10.1186/2041-9139-5-40] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 09/17/2014] [Indexed: 12/30/2022] Open
Abstract
Background Understanding the molecular basis of craniofacial variation can provide insights into key developmental mechanisms of adaptive changes and their role in trophic divergence and speciation. Arctic charr (Salvelinus alpinus) is a polymorphic fish species, and, in Lake Thingvallavatn in Iceland, four sympatric morphs have evolved distinct craniofacial structures. We conducted a gene expression study on candidates from a conserved gene coexpression network, focusing on the development of craniofacial elements in embryos of two contrasting Arctic charr morphotypes (benthic and limnetic). Results Four Arctic charr morphs were studied: one limnetic and two benthic morphs from Lake Thingvallavatn and a limnetic reference aquaculture morph. The presence of morphological differences at developmental stages before the onset of feeding was verified by morphometric analysis. Following up on our previous findings that Mmp2 and Sparc were differentially expressed between morphotypes, we identified a network of genes with conserved coexpression across diverse vertebrate species. A comparative expression study of candidates from this network in developing heads of the four Arctic charr morphs verified the coexpression relationship of these genes and revealed distinct transcriptional dynamics strongly correlated with contrasting craniofacial morphologies (benthic versus limnetic). A literature review and Gene Ontology analysis indicated that a significant proportion of the network genes play a role in extracellular matrix organization and skeletogenesis, and motif enrichment analysis of conserved noncoding regions of network candidates predicted a handful of transcription factors, including Ap1 and Ets2, as potential regulators of the gene network. The expression of Ets2 itself was also found to associate with network gene expression. Genes linked to glucocorticoid signalling were also studied, as both Mmp2 and Sparc are responsive to this pathway. Among those, several transcriptional targets and upstream regulators showed differential expression between the contrasting morphotypes. Interestingly, although selected network genes showed overlapping expression patterns in situ and no morph differences, Timp2 expression patterns differed between morphs. Conclusion Our comparative study of transcriptional dynamics in divergent craniofacial morphologies of Arctic charr revealed a conserved network of coexpressed genes sharing functional roles in structural morphogenesis. We also implicate transcriptional regulators of the network as targets for future functional studies. Electronic supplementary material The online version of this article (doi:10.1186/2041-9139-5-40) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ehsan Pashay Ahi
- Institute of Life and Environmental Sciences, University of Iceland, Sturlugata 7, 101 Reykjavik, Iceland
| | - Kalina Hristova Kapralova
- Institute of Life and Environmental Sciences, University of Iceland, Sturlugata 7, 101 Reykjavik, Iceland
| | - Arnar Pálsson
- Institute of Life and Environmental Sciences, University of Iceland, Sturlugata 7, 101 Reykjavik, Iceland ; Biomedical Center, University of Iceland, Vatnsmýrarvegur 16, 101 Reykjavik, Iceland
| | - Valerie Helene Maier
- Institute of Life and Environmental Sciences, University of Iceland, Sturlugata 7, 101 Reykjavik, Iceland
| | - Jóhannes Gudbrandsson
- Institute of Life and Environmental Sciences, University of Iceland, Sturlugata 7, 101 Reykjavik, Iceland
| | - Sigurdur S Snorrason
- Institute of Life and Environmental Sciences, University of Iceland, Sturlugata 7, 101 Reykjavik, Iceland
| | - Zophonías O Jónsson
- Institute of Life and Environmental Sciences, University of Iceland, Sturlugata 7, 101 Reykjavik, Iceland ; Biomedical Center, University of Iceland, Vatnsmýrarvegur 16, 101 Reykjavik, Iceland
| | - Sigrídur Rut Franzdóttir
- Institute of Life and Environmental Sciences, University of Iceland, Sturlugata 7, 101 Reykjavik, Iceland
| |
Collapse
|
47
|
Barbeito-Andrés J, Ventrice F, Anzelmo M, Pucciarelli HM, Sardi ML. Developmental covariation of human vault and base throughout postnatal ontogeny. Ann Anat 2014; 197:59-66. [PMID: 25458178 DOI: 10.1016/j.aanat.2014.10.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Revised: 09/30/2014] [Accepted: 10/02/2014] [Indexed: 12/23/2022]
Abstract
In the present study, we analyzed postnatal ontogenetic integration among morphological traits of the human neurocranium. Particularly, the covariation between the vault and the base during postnatal life was assessed. Since the association between these regions may depend on the generalized change produced by allometry, we tested its effect on their covariation. On a sample of adults and subadults ranging from 0 to 31 years, 3D coordinates of neurocranial landmarks and semilandmarks were digitized and geometric morphometric technics were applied. Main aspects of shape variation were examined using Principal Components analysis. Covariation between the vault and the base was examined by Partial Least Squares analysis. According to our results, the vault and the base covary strongly during postnatal ontogeny and their relation depends largely on allometry. Two size variables were studied: centroid size, which was obtained from the recorded morphometric points, and endocranial volume, taken as an estimation of brain size. Although growing brain was found to be a developmental process that contributes to covariation among neurocranial traits, there would be other factors that exert their influence during ontogeny. These results lead to reconsider cranial morphological evolution taking into account the developmental constraints given by ontogenetic patterns of integration and reinforcing the idea that in human evolution a suite of relevant characters may be fuelled by few developmental processes.
Collapse
Affiliation(s)
- Jimena Barbeito-Andrés
- División Antropología, Museo de La Plata, Facultad de Ciencias Naturales y Museo, Universidad Nacional de La Plata, Paseo del Bosque s/n, 1900, La Plata, Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina.
| | - Fernando Ventrice
- Laboratorio de Neuroimágenes, Departamento de Imágenes, Instituto de Investigaciones Neurológicas Raúl Carrea, FLENI, Montañeses 2325, 1428 Ciudad Autónoma de Buenos Aires (Buenos Aires), Argentina
| | - Marisol Anzelmo
- División Antropología, Museo de La Plata, Facultad de Ciencias Naturales y Museo, Universidad Nacional de La Plata, Paseo del Bosque s/n, 1900, La Plata, Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Héctor M Pucciarelli
- División Antropología, Museo de La Plata, Facultad de Ciencias Naturales y Museo, Universidad Nacional de La Plata, Paseo del Bosque s/n, 1900, La Plata, Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Marina L Sardi
- División Antropología, Museo de La Plata, Facultad de Ciencias Naturales y Museo, Universidad Nacional de La Plata, Paseo del Bosque s/n, 1900, La Plata, Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| |
Collapse
|
48
|
Shared developmental programme strongly constrains beak shape diversity in songbirds. Nat Commun 2014; 5:3700. [PMID: 24739280 DOI: 10.1038/ncomms4700] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Accepted: 03/20/2014] [Indexed: 01/30/2023] Open
Abstract
The striking diversity of bird beak shapes is an outcome of natural selection, yet the relative importance of the limitations imposed by the process of beak development on generating such variation is unclear. Untangling these factors requires mapping developmental mechanisms over a phylogeny far exceeding model systems studied thus far. We address this issue with a comparative morphometric analysis of beak shape in a diverse group of songbirds. Here we show that the dynamics of the proliferative growth zone must follow restrictive rules to explain the observed variation, with beak diversity constrained to a three parameter family of shapes, parameterized by length, depth and the degree of shear. We experimentally verify these predictions by analysing cell proliferation in the developing embryonic beaks of the zebra finch. Our findings indicate that beak shape variability in many songbirds is strongly constrained by shared properties of the developmental programme controlling the growth zone.
Collapse
|
49
|
Kennedy AE, Dickinson AJ. Quantitative analysis of orofacial development and median clefts in Xenopus laevis. Anat Rec (Hoboken) 2014; 297:834-55. [PMID: 24443252 DOI: 10.1002/ar.22864] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 11/06/2013] [Accepted: 11/25/2013] [Indexed: 01/01/2023]
Abstract
Xenopus has become a useful tool to study the molecular mechanisms underlying orofacial development. However, few quantitative analyses exist to describe the anatomy of this region. In this study we combine traditional facial measurements with geometric morphometrics to describe anatomical changes in the orofacial region during normal and abnormal development. Facial measurements and principal component (PC) analysis indicate that during early tadpole development the face expands primarily in the midface region accounting for the development of the upper jaw and primary palate. The mouth opening correspondingly becomes flatter and wider as it incorporates the jaw elements. A canonical variate analysis of orofacial and mouth opening shape emphasized that changes in the orofacial shape occur gradually. Orofacial anatomy was quantified after altered levels of retinoic acid using all-trans retinoic acid or an inhibitor of retinoic acid receptors or by injecting antisense oligos targeting RALDH2. Such perturbations resulted in major decreases in the width of the midface and the mouth opening illustrated in facial measurements and a PC analysis. The mouth opening shape also had a gap in the primary palate resulting in a median cleft in the mouth opening that was only illustrated quantitatively in the morphometric analysis. Finally, canonical and discriminant function analysis statistically distinguished the orofacial and mouth opening shape changes among the different modes used to alter retinoic acid signaling levels. By combining quantitative analyses with molecular studies of orofacial development we will be better equipped to understand the complex morphogenetic processes involved in palate development and clefting.
Collapse
Affiliation(s)
- Allyson E Kennedy
- Department of Biology, Virginia Commonwealth University, 1000 West Cary Street, Richmond, Virginia
| | | |
Collapse
|
50
|
Hünemeier T, Gómez-Valdés J, De Azevedo S, Quinto-Sánchez M, Passaglia L, Salzano FM, Sánchez-Mejorada G, Alonzo VA, Martínez-Abadías N, Bortolini MC, González-José R. FGFR1 signaling is associated with the magnitude of morphological integration in human head shape. Am J Hum Biol 2013; 26:164-75. [DOI: 10.1002/ajhb.22496] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Revised: 11/24/2013] [Accepted: 11/26/2013] [Indexed: 11/09/2022] Open
Affiliation(s)
- Tábita Hünemeier
- Departamento de Genética; Instituto de Biociências, Universidade Federal do Rio Grande do Sul. Caixa Postal 15053; 91501-970 Porto Alegre RS Brazil
| | - Jorge Gómez-Valdés
- Departamento de Anatomía; Facultad de Medicina; Universidad Nacional Autónoma de México, Circuito Interior, Ciudad Universitaria; 04510 Distrito Federal México
| | - Soledad De Azevedo
- Centro Nacional Patagónico-CONICET; Bvd. Brown 2915, U9120ACD Puerto Madryn Argentina
| | - Mirsha Quinto-Sánchez
- Centro Nacional Patagónico-CONICET; Bvd. Brown 2915, U9120ACD Puerto Madryn Argentina
| | - Luciane Passaglia
- Departamento de Genética; Instituto de Biociências, Universidade Federal do Rio Grande do Sul. Caixa Postal 15053; 91501-970 Porto Alegre RS Brazil
| | - Francisco M. Salzano
- Departamento de Genética; Instituto de Biociências, Universidade Federal do Rio Grande do Sul. Caixa Postal 15053; 91501-970 Porto Alegre RS Brazil
| | - Gabriela Sánchez-Mejorada
- Departamento de Anatomía; Facultad de Medicina; Universidad Nacional Autónoma de México, Circuito Interior, Ciudad Universitaria; 04510 Distrito Federal México
| | | | - Neus Martínez-Abadías
- EMBL-CRG Systems Biology Research Unit; Center for Genomic Regulation (CRG), Dr. Aiguader 88 08003 Barcelona Spain
| | - Maria-Cátira Bortolini
- Departamento de Genética; Instituto de Biociências, Universidade Federal do Rio Grande do Sul. Caixa Postal 15053; 91501-970 Porto Alegre RS Brazil
| | - Rolando González-José
- Centro Nacional Patagónico-CONICET; Bvd. Brown 2915, U9120ACD Puerto Madryn Argentina
| |
Collapse
|