1
|
Khorasani E, Vahidi B. 3D-Printed Scaffolds for Cranial Bone Regeneration: A Systematic Review of Design, Materials, and Computational Optimization. Biotechnol Bioeng 2025. [PMID: 40289530 DOI: 10.1002/bit.28994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 03/26/2025] [Accepted: 04/04/2025] [Indexed: 04/30/2025]
Abstract
Cranial bone defects from trauma, congenital conditions, or surgery are challenging to treat due to the skull's limited regeneration. Traditional methods like autografts and allografts have drawbacks, including donor site issues and poor integration. 3D-printed scaffolds provide a patient-specific alternative, improving bone regeneration and integration. This review evaluates advancements in 3D-printed scaffolds for cranial bone regeneration, focusing on fabrication techniques, material innovations, and structural optimization while assessing their preclinical and clinical potential. A systematic literature search (2014-2024) was conducted using PubMed and other databases. Studies addressing scaffold properties such as porosity, pore interconnectivity, and mechanical stability were included, while non-cranial scaffold studies were excluded. Advances in 3D printing have enabled patient-specific scaffolds with optimized architecture to enhance bone regeneration, mechanical support, and nutrient transport. Bioceramics, polymers, and composites mimic native bone properties, while bioactive coatings further improve osteogenesis. However, limited clinical translation and insufficient customization remain challenges. Further preclinical and clinical trials are crucial to overcoming barriers in mechanical optimization and patient-specific scaffold fabrication, bridging the gap between research and clinical applications.
Collapse
Affiliation(s)
- Elnaz Khorasani
- Department of Medical Technology and Tissue Engineering, Faculty of Life Science Engineering, School of Interdisciplinary Science and Technology, University of Tehran, Tehran, Iran
| | - Bahman Vahidi
- Department of Medical Technology and Tissue Engineering, Faculty of Life Science Engineering, School of Interdisciplinary Science and Technology, University of Tehran, Tehran, Iran
| |
Collapse
|
2
|
Telizhenko V, Gol'din P. Evolution of accessory bones in cetacean skull coupled with decreasing rate of ossification of cranial sutures. Sci Rep 2025; 15:10268. [PMID: 40133709 PMCID: PMC11937560 DOI: 10.1038/s41598-025-95566-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 03/21/2025] [Indexed: 03/27/2025] Open
Abstract
The skulls of cetaceans (whales, dolphins and porpoises) are distinguished by incomplete ossification of cranial sutures during their lifetime. We suggested suture ossification may correlate with the evolution of rarely reported accessory bones and examine the evolutionary patterns of cranial suture ossification and the presence of accessory elements in the skulls of 47 cetacean species and, for comparison, 15 terrestrial artiodactyls (even-hoofed mammals). A strong phylogenetic signal was found for both suture ossification rate and accessory bone presence. Cetaceans were shown to have a lower ossification rate than most terrestrial artiodactyls, except deer (Cervidae) and mouse deer (Tragulidae), which showed somewhat similar patterns. There were also several types of accessory bony elements, some of them first reported in the skulls of cetaceans and in one case, a muntjac deer. These elements (bones and clefts) evolved in the crania with the least number of ossified sutures. They can be identified as novel elements or, some of them, as plesiomorphies rarely seen in mammals but existing in reptiles and other ancestral groups. This leads to breaking a general trend of mammalian evolution: a reversal of Williston's law (reduction in bone number) and the development of new accessory cranial bones. Slowing cranial development rates explain these trends, and feeding mechanics may be a driver of decreasing suture ossification in cetaceans.
Collapse
|
3
|
Werneburg I, Bronzati M. Trifold origin of the reptilian ear ossicle and its relation to the evolutionary modification of the temporal skull region. J Anat 2025; 246:402-414. [PMID: 39297283 PMCID: PMC11828744 DOI: 10.1111/joa.14105] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 06/14/2024] [Accepted: 06/16/2024] [Indexed: 02/16/2025] Open
Abstract
Whereas mammals are characterized by the presence of three middle ear ossicles, reptiles have only one, the columella (stapes). Nevertheless, there is a great diversity of columellar anatomy among sauropsids, especially in the unique and cartilaginous "extracolumella"-portion. Molecular studies revealed the "columella" of chicken and quails to be formed within the second pharyngeal arch, although conflicting evidence exists for the columellar footplate and distal parts of the columella in these birds. We studied columellar development in four turtles, one lizard, and one caiman species and argue, using early blastematous stages, that, distally, the so-called "extracolumella" in turtles is mainly of quadrate, that is, first pharyngeal arch origin. Differently, the dorsal aspect of the "extracolumella" of the lizard and a part of the "dorsal columella process" of the caiman are likely quadrate-derived. This indicates only a partial homology of the distal columellar compartments among reptiles. Moreover, we observed in most species that, at early stages, the footplate differentiates from the otic capsule, which confirms widespread experimental findings of mesodermal cells contributing to the proximal part of the columella. We provide a hypothetical framework for the changes in the columella and quadrate morphology in reptilian evolution. Originally, as evidenced by the fossil record, the columella served as a stabilizing brace between the quadrate and braincase. Associated with changes in the feeding mode of late Permian taxa, the quadrate was integrated along the stress flows from biting, and in early development part of the quadrate differentiated to differently contribute to the distal part of the "columella-complex," which now contacts the tympanic membrane. In addition, part of the original otic capsule contributes to the footplate of the mobile columella, providing a connection with the inner ear.
Collapse
Affiliation(s)
- Ingmar Werneburg
- Paläontologische SammlungSenckenberg Centre for Human Evolution and Palaeoenvironment (SHEP) an der Universität TübingenTübingenGermany
- Fachbereich GeowissenschaftenUniversität TübingenTübingenGermany
| | - Mario Bronzati
- Paläontologische SammlungSenckenberg Centre for Human Evolution and Palaeoenvironment (SHEP) an der Universität TübingenTübingenGermany
| |
Collapse
|
4
|
Kalev-Altman R, Fraggi-Rankis V, Monsonego-Ornan E, Sela-Donenfeld D. The bone Gla protein osteocalcin is expressed in cranial neural crest cells. BMC Res Notes 2024; 17:329. [PMID: 39501347 PMCID: PMC11539830 DOI: 10.1186/s13104-024-06990-7] [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: 06/20/2024] [Accepted: 10/28/2024] [Indexed: 11/08/2024] Open
Abstract
BACKGROUND Osteocalcin is a small protein abundant in the bone extracellular-matrix, that serves as a marker for mature osteoblasts. To become activated, osteocalcin undergoes a specific post-translational carboxylation. Osteocalcin is expressed at advanced stages of embryogenesis and after birth, when bone formation takes place. Neural crest cells (NCCs) are a unique cell population that evolves during early stages of development. While initially NCCs populate the dorsal neural-tube, later they undergo epithelial-to-mesenchymal-transition and migrate throughout the embryo in highly-regulated manner. NCCs give rise to multiple cell types including neurons and glia of the peripheral nervous system, chromaffin cells and skin melanocytes. Remarkably, in the head region, NCCs give rise to cartilage and bone. FINDING Here we report that osteocalcin is detected in cranial NCCs. Analysis of chick embryos at stages of cranial NCC migration revealed that osteocalcin mRNA and protein is expressed in pre-migratory and migratory NCCs in-vivo and ex-vivo. Addition of warfarin, an inhibitor of osteocalcin carboxylation, onto neural-tube explants, reduced the amount of NCC migration. These results provide the first evidence of osteocalcin presence in cranial NCCs, much before they give rise to craniofacial skeleton, and propose its possible involvement in the regulation of NCC migration.
Collapse
Affiliation(s)
- Rotem Kalev-Altman
- Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University, Rehovot, 76100, Israel
- The Institute of Biochemistry and Nutrition, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University, Rehovot, 76100, Israel
- The Section of Genetic Medicine, The Department of Medicine, The University of Chicago, Chicago, IL, 60637, USA
| | - Veatriki Fraggi-Rankis
- Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University, Rehovot, 76100, Israel
- Smart Assays Biotechnologies Ltd, Weizmann Science Park, Ness Ziona, 7414003, Israel
| | - Efrat Monsonego-Ornan
- The Institute of Biochemistry and Nutrition, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University, Rehovot, 76100, Israel
| | - Dalit Sela-Donenfeld
- Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University, Rehovot, 76100, Israel.
| |
Collapse
|
5
|
Koyabu D. Evolution, conservatism and overlooked homologies of the mammalian skull. Philos Trans R Soc Lond B Biol Sci 2023; 378:20220081. [PMID: 37183902 PMCID: PMC10184252 DOI: 10.1098/rstb.2022.0081] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 01/22/2023] [Indexed: 05/16/2023] Open
Abstract
In the last decade, studies integrating palaeontology, embryology and experimental developmental biology have markedly altered our homological understanding of the mammalian skull. Indeed, new evidence suggests that we should revisit and restructure the conventional anatomical terminology applied to the components of the mammalian skull. Notably, these are classical problems that have remained unresolved since the ninteenth century. In this review, I offer perspectives on the overlooked problems associated with the homology, development, and conservatism of the mammalian skull, aiming to encourage future studies in these areas. I emphasise that ossification patterns, bone fusion, cranial sutures and taxon-specific neomorphic bones in the skull are virtually unexplored, and further studies would improve our homological understanding of the mammalian skull. Lastly, I highlight that overlooked bones may exist in the skull that are not yet known to science and suggest that further search is needed. This article is part of the theme issue 'The mammalian skull: development, structure and function'.
Collapse
Affiliation(s)
- Daisuke Koyabu
- Research and Development Center for Precision Medicine, University of Tsukuba, Tsukuba, Japan
- Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Hong Kong, People's Republic of China
| |
Collapse
|
6
|
Maden M, Polvadore T, Polanco A, Barbazuk WB, Stanley E. Osteoderms in a mammal the spiny mouse Acomys and the independent evolution of dermal armor. iScience 2023; 26:106779. [PMID: 37378333 PMCID: PMC10291248 DOI: 10.1016/j.isci.2023.106779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 02/06/2023] [Accepted: 04/25/2023] [Indexed: 06/29/2023] Open
Abstract
Osteoderms are bony plates found in the skin of vertebrates, mostly commonly in reptiles where they have evolved independently multiple times, suggesting the presence of a gene regulatory network that is readily activated and inactivated. They are absent in birds and mammals except for the armadillo. However, we have discovered that in one subfamily of rodents, the Deomyinae, there are osteoderms in the skin of their tails. Osteoderm development begins in the proximal tail skin and is complete 6 weeks after birth. RNA sequencing has identified the gene networks involved in their differentiation. There is a widespread down-regulation of keratin genes and an up-regulation of osteoblast genes and a finely balanced expression of signaling pathways as the osteoderms differentiate. Future comparisons with reptilian osteoderms may allow us to understand how these structures have evolved and why they are so rare in mammals.
Collapse
Affiliation(s)
- Malcolm Maden
- Department of Biology & UF Genetics Institute, University of Florida, Gainesville, FL 32611, USA
| | - Trey Polvadore
- Department of Biology & UF Genetics Institute, University of Florida, Gainesville, FL 32611, USA
| | - Arod Polanco
- Department of Biology & UF Genetics Institute, University of Florida, Gainesville, FL 32611, USA
| | - W. Brad Barbazuk
- Department of Biology & UF Genetics Institute, University of Florida, Gainesville, FL 32611, USA
| | - Edward Stanley
- Florida Museum of Natural History, University of Florida, Museum Road, Gainesville, FL 32611, USA
| |
Collapse
|
7
|
Kuroda S, Adachi N, Kuratani S. A detailed redescription of the mesoderm/neural crest cell boundary in the murine orbitotemporal region integrates the mammalian cranium into a pan-amniote cranial configuration. Evol Dev 2023; 25:32-53. [PMID: 35909296 DOI: 10.1111/ede.12411] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 06/14/2022] [Accepted: 07/02/2022] [Indexed: 01/13/2023]
Abstract
The morphology of the mammalian chondrocranium appears to differ significantly from those of other amniotes, since the former possesses uniquely developed brain and cranial sensory organs. In particular, a question has long remained unanswered as to the developmental and evolutionary origins of a cartilaginous nodule called the ala hypochiasmatica. In this study, we investigated the embryonic origin of skeletal elements in the murine orbitotemporal region by combining genetic cell lineage analysis with detailed morphological observation. Our results showed that the mesodermal embryonic environment including the ala hypochiasmatica, which appeared as an isolated mesodermal distribution in the neural crest-derived prechordal region, is formed as a part of the mesoderm that continued from the chordal region during early chondrocranial development. The mesoderm/neural crest cell boundary in the head mesenchyme is modified through development, resulting in the secondary mesodermal expansion to invade into the prechordal region. We thus revealed that the ala hypochiasmatica develops as the frontier of the mesodermal sheet stretched along the cephalic flexure. These results suggest that the mammalian ala hypochiasmatica has evolved from a part of the mesodermal primary cranial wall in ancestral amniotes. In addition, the endoskeletal elements in the orbitotemporal region, such as the orbital cartilage, suprapterygoid articulation of the palatoquadrate, and trabecula, some of which were once believed to represent primitive traits of amniotes and to be lost in the mammalian lineage, have been confirmed to exist in the mammalian cranium. Consequently, the mammalian chondrocranium can now be explained in relation to the pan-amniote cranial configuration.
Collapse
Affiliation(s)
- Shunya Kuroda
- Laboratory for Evolutionary Morphology, RIKEN Center for Biosystems Dynamics Research (BDR), Kobe, Hyogo, Japan.,Department of Biology, Graduate School of Science, Kobe University, Kobe, Hyogo, Japan
| | - Noritaka Adachi
- Aix-Marseille Université, CNRS UMR 7288, IBDM, Marseille, France.,Laboratory for Evolutionary Morphology, RIKEN Cluster for Pioneering Research (CPR), Kobe, Hyogo, Japan
| | - Shigeru Kuratani
- Laboratory for Evolutionary Morphology, RIKEN Center for Biosystems Dynamics Research (BDR), Kobe, Hyogo, Japan
| |
Collapse
|
8
|
Rose CS. The cellular basis of cartilage growth and shape change in larval and metamorphosing Xenopus frogs. PLoS One 2023; 18:e0277110. [PMID: 36634116 PMCID: PMC9836273 DOI: 10.1371/journal.pone.0277110] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 10/19/2022] [Indexed: 01/13/2023] Open
Abstract
As the first and sometimes only skeletal tissue to appear, cartilage plays a fundamental role in the development and evolution of vertebrate body shapes. This is especially true for amphibians whose largely cartilaginous feeding skeleton exhibits unparalleled ontogenetic and phylogenetic diversification as a consequence of metamorphosis. Fully understanding the evolutionary history, evolvability and regenerative potential of cartilage requires in-depth analysis of how chondrocytes drive growth and shape change. This study is a cell-level description of the larval growth and postembryonic shape change of major cartilages of the feeding skeleton of a metamorphosing amphibian. Histology and immunohistochemistry are used to describe and quantify patterns and trends in chondrocyte size, shape, division, death, and arrangement, and in percent matrix from hatchling to froglet for the lower jaw, hyoid and branchial arch cartilages of Xenopus laevis. The results are interpreted and integrated into programs of cell behaviors that account for the larval growth and histology, and metamorphic remodeling of each element. These programs provide a baseline for investigating hormone-mediated remodeling, cartilage regeneration, and intrinsic shape regulating mechanisms. These programs also contain four features not previously described in vertebrates: hypertrophied chondrocytes being rejuvenated by rapid cell cycling to a prechondrogenic size and shape; chondrocytes dividing and rearranging to reshape a cartilage; cartilage that lacks a perichondrium and grows at single-cell dimensions; and an adult cartilage forming de novo in the center of a resorbing larval one. Also, the unexpected superimposition of cell behaviors for shape change onto ones for larval growth and the unprecedented exploitation of very large and small cell sizes provide new directions for investigating the development and evolution of skeletal shape and metamorphic ontogenies.
Collapse
Affiliation(s)
- Christopher S. Rose
- Department of Biology, James Madison University, Harrisonburg, Virginia, United States of America
- * E-mail:
| |
Collapse
|
9
|
Kierdorf U, Stock SR, Gomez S, Antipova O, Kierdorf H. Distribution, structure, and mineralization of calcified cartilage remnants in hard antlers. Bone Rep 2022; 16:101571. [PMID: 35519288 PMCID: PMC9065892 DOI: 10.1016/j.bonr.2022.101571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 04/18/2022] [Accepted: 04/21/2022] [Indexed: 11/05/2022] Open
Abstract
Antlers are paired deciduous bony cranial appendages of deer that undergo a regular cycle of growth, death and casting, and constitute the most rapidly growing bones in mammals. Antler growth occurs in an appositional mode and involves a modified form of endochondral ossification. In endochondral bones, calcified cartilage is typically a transient tissue that is eventually completely replaced by bone tissue. We studied the distribution and characteristics of calcified cartilage in hard antlers from three deer species (Capreolus capreolus, Cervus elaphus, Dama dama), i.e., in antlers from which the skin (velvet) had been shed. Remnants of calcified cartilage were regularly present as part of the trabecular framework in the late formed, distal antler portions in all three species, whereas this tissue was largely or completely missing in the more proximal antler portions. The presence of calcified cartilage remnants in the distal antler portions is attributed to the limited antler lifespan of only a few months, which is also the reason for the virtual lack of bone remodeling in antlers. The calcified cartilage matrix was more highly mineralized than the antler bone matrix. Mineralized deposits were observed in some chondrocyte lacunae and occasionally also in osteocyte lacunae, a phenomenon that has not previously been reported in antlers. Using synchrotron radiation-induced X-ray fluorescence (SR-XRF) mapping, we further demonstrated increased zinc concentrations in cement lines, along the inner borders of incompletely formed primary osteons, along the walls of partly or completely mineral-occluded chondrocyte and osteocyte lacunae, and in intralacunar mineralized deposits. The present study demonstrates that antlers are a promising model for studying the mineralization of cartilage and bone matrices and the formation of mineralized deposits in chondrocyte and osteocyte lacunae. Remnants of calcified cartilage are regularly present in hard antlers of deer. Preservation of calcified cartilage is caused by the short lifespan of antlers. Calcified cartilage of antlers is more highly mineralized than antler bone. Mineralized deposits were observed in chondrocyte and osteocyte lacunae of antlers. SR-XRF showed increased Zn-concentration in cement lines and intralacunar deposits.
Collapse
|
10
|
Lesciotto KM, Tomlinson L, Leonard S, Richtsmeier JT. Embryonic and Early Postnatal Cranial Bone Volume and Tissue Mineral Density Values for C57BL/6J Laboratory Mice. Dev Dyn 2022; 251:1196-1208. [PMID: 35092111 PMCID: PMC9250594 DOI: 10.1002/dvdy.458] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 01/18/2022] [Accepted: 01/20/2022] [Indexed: 11/27/2022] Open
Abstract
Background Laboratory mice are routinely used in craniofacial research based on the relatively close genetic relationship and conservation of developmental pathways between humans and mice. Since genetic perturbations and disease states may have localized effects, data from individual cranial bones are valuable for the interpretation of experimental assays. We employ high‐resolution microcomputed tomography to characterize cranial bones of C57BL/6J mice at embryonic day (E) 15.5 and E17.5, day of birth (P0), and postnatal day 7 (P7) and provide estimates of individual bone volume and tissue mineral density (TMD). Results Average volume and TMD values are reported for individual bones. Significant differences in volume and TMD during embryonic ages likely reflect early mineralization of cranial neural crest‐derived and intramembranously forming bones. Although bones of the face and vault had higher TMD values during embryonic ages, bones of the braincase floor had significantly higher TMD values by P7. Conclusions These ontogenetic data on cranial bone volume and TMD serve as a reference standard for future studies using mice bred on a C57BL/6J genetic background. Our findings also highlight the importance of differentiating “control” data from mice that are presented as “unaffected” littermates, particularly when carrying a single copy of a cre‐recombinase gene. Higher average volume and density of cranial neural crest‐derived and intramembranously‐forming bones during embryonic development. Higher average density in bones of the braincase floor during early postnatal development. Ontogenetic data on cranial bone volume and TMD serve as a reference standard for mice bred on a C57BL/6J genetic background.
Collapse
Affiliation(s)
- Kate M Lesciotto
- College of Osteopathic Medicine, Sam Houston State University, Conroe, TX, USA
| | | | - Steven Leonard
- College of Medicine, Drexel University, Philadelphia, PA, USA
| | - Joan T Richtsmeier
- Department of Anthropology, Pennsylvania State University, University Park, PA, USA
| |
Collapse
|
11
|
Sanger TJ, Harding L, Kyrkos J, Turnquist AJ, Epperlein L, Nunez SA, Lachance D, Dhindsa S, Stroud JT, Diaz RE, Czesny B. Environmental Thermal Stress Induces Neuronal Cell Death and Developmental Malformations in Reptiles. Integr Org Biol 2021; 3:obab033. [PMID: 34877473 PMCID: PMC8643577 DOI: 10.1093/iob/obab033] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 09/25/2021] [Accepted: 11/12/2021] [Indexed: 12/19/2022] Open
Abstract
Every stage of organismal life history is being challenged by global warming. Many species are already experiencing temperatures approaching their physiological limits; this is particularly true for ectothermic species, such as lizards. Embryos are markedly sensitive to thermal insult. Here, we demonstrate that temperatures currently experienced in natural nesting areas can modify gene expression levels and induce neural and craniofacial malformations in embryos of the lizard Anolis sagrei. Developmental abnormalities ranged from minor changes in facial structure to significant disruption of anterior face and forebrain. The first several days of postoviposition development are particularly sensitive to this thermal insult. These results raise new concern over the viability of ectothermic species under contemporary climate change. Herein, we propose and test a novel developmental hypothesis that describes the cellular and developmental origins of those malformations: cell death in the developing forebrain and abnormal facial induction due to disrupted Hedgehog signaling. Based on similarities in the embryonic response to thermal stress among distantly related species, we propose that this developmental hypothesis represents a common embryonic response to thermal insult among amniote embryos. Our results emphasize the importance of adopting a broad, multidisciplinary approach that includes both lab and field perspectives when trying to understand the future impacts of anthropogenic change on animal development.
Collapse
Affiliation(s)
- Thomas J Sanger
- Department of Biology, Loyola University Chicago, 1050 Sheridan Rd., Chicago, IL 60660, USA
| | - Laura Harding
- Department of Biology, Loyola University Chicago, 1050 Sheridan Rd., Chicago, IL 60660, USA
| | - Judith Kyrkos
- Department of Biology, Loyola University Chicago, 1050 Sheridan Rd., Chicago, IL 60660, USA
| | - Alexandrea J Turnquist
- Department of Biology, Loyola University Chicago, 1050 Sheridan Rd., Chicago, IL 60660, USA
| | - Lilian Epperlein
- Department of Biology, Loyola University Chicago, 1050 Sheridan Rd., Chicago, IL 60660, USA
| | - Sylvia A Nunez
- Department of Biology, Loyola University Chicago, 1050 Sheridan Rd., Chicago, IL 60660, USA
| | - Dryden Lachance
- Department of Biology, Loyola University Chicago, 1050 Sheridan Rd., Chicago, IL 60660, USA
| | - Seerat Dhindsa
- Department of Biology, Loyola University Chicago, 1050 Sheridan Rd., Chicago, IL 60660, USA
| | - James T Stroud
- Department of Biology, Washington University in St. Louis, Campus Box 1137. One Brookings Drive St. Louis, MO 63130-4899, USA
| | - Raul E Diaz
- Department of Biological Sciences, California State University, Los Angeles, 5151 State University Dr., Los Angeles, CA 90032, USA
| | - Beata Czesny
- Department of Biology, Loyola University Chicago, 1050 Sheridan Rd., Chicago, IL 60660, USA
| |
Collapse
|
12
|
Schnabl J, Litz MPH, Schneider C, PenkoffLidbeck N, Bashiruddin S, Schwartz MS, Alligood K, Devoto SH, Barresi MJF. Characterizing the diverse cells that associate with the developing commissures of the zebrafish forebrain. Dev Neurobiol 2021; 81:671-695. [PMID: 33314626 DOI: 10.1002/dneu.22801] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 10/20/2020] [Accepted: 12/08/2020] [Indexed: 01/02/2023]
Abstract
During embryonic development of bilaterally symmetrical organisms, neurons send axons across the midline at specific points to connect the two halves of the nervous system with a commissure. Little is known about the cells at the midline that facilitate this tightly regulated process. We exploit the conserved process of vertebrate embryonic development in the zebrafish model system to elucidate the identity of cells at the midline that may facilitate postoptic (POC) and anterior commissure (AC) development. We have discovered that three different gfap+ astroglial cell morphologies persist in contact with pathfinding axons throughout commissure formation. Similarly, olig2+ progenitor cells occupy delineated portions of the postoptic and anterior commissures where they act as multipotent, neural progenitors. Moreover, we conclude that both gfap+ and olig2+ progenitor cells give rise to neuronal populations in both the telencephalon and diencephalon; however, these varied cell populations showed significant developmental timing differences between the telencephalon and diencephalon. Lastly, we also showed that fli1a+ mesenchymal cells migrate along the presumptive commissure regions before and during midline axon crossing. Furthermore, following commissure maturation, specific blood vessels formed at the midline of the POC and immediately ventral and parallel to the AC. This comprehensive account of the cellular populations that correlate with the timing and position of commissural axon pathfinding has supported the conceptual modeling and identification of the early forebrain architecture that may be necessary for proper commissure development.
Collapse
Affiliation(s)
- Jake Schnabl
- Department of Molecular and Cellular Biology, University of Massachusetts, Amherst, MA, USA
| | - Mackenzie P H Litz
- Department of Biological Sciences, Smith College, Northampton, MA, USA.,Computational and Systems Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Caitlin Schneider
- Department of Biological Sciences, Smith College, Northampton, MA, USA.,McGill University, Montreal, QC, Canada
| | | | - Sarah Bashiruddin
- Department of Biological Sciences, Smith College, Northampton, MA, USA.,Family Medicine Assoc, Westfield, MA, USA
| | - Morgan S Schwartz
- Department of Biological Sciences, Smith College, Northampton, MA, USA.,Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Kristin Alligood
- Department of Biological Sciences, Smith College, Northampton, MA, USA.,Farmers Conservation Alliance, Hood River, OR, USA
| | | | - Michael J F Barresi
- Department of Molecular and Cellular Biology, University of Massachusetts, Amherst, MA, USA.,Department of Biological Sciences, Smith College, Northampton, MA, USA
| |
Collapse
|
13
|
Takenoshita M, Takechi M, Vu Hoang T, Furutera T, Akagawa C, Namangkalakul W, Aoto K, Kume T, Miyashin M, Iwamoto T, Iseki S. Cell lineage- and expression-based inference of the roles of forkhead box transcription factor Foxc2 in craniofacial development. Dev Dyn 2021; 250:1125-1139. [PMID: 33667029 DOI: 10.1002/dvdy.324] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 02/08/2021] [Accepted: 02/24/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Foxc2 is a member of the winged helix/forkhead (Fox) box family of transcription factors. Loss of function of Foxc2 causes craniofacial abnormalities such as cleft palate and deformed cranial base, but its role during craniofacial development remains to be elucidated. RESULTS The contributions of Foxc2-positive and its descendant cells to the craniofacial structure at E18.5 were examined using a tamoxifen-inducible Cre driver mouse (Foxc2-CreERT2) crossed with the R26R-LacZ reporter mouse. Foxc2 expression at E8.5 is restricted to the cranial mesenchyme, contributing to specific components including the cranial base, sensory capsule, tongue, upper incisor, and middle ear. Expression at E10.5 was still positively regulated in most of those regions. In situ hybridization analysis of Foxc2 and its closely related gene, Foxc1, revealed that expression domains of these genes largely overlap in the cephalic mesenchyme. Meanwhile, the tongue expressed Foxc2 but not Foxc1, and its development was affected by the neural crest-specific deletion of Foxc2 in mice (Wnt1-Cre; Foxc2fl/fl ). CONCLUSIONS Foxc2 is expressed in cranial mesenchyme that contributes to specific craniofacial tissue components from an early stage, and it seems to be involved in their development in cooperation with Foxc1. Foxc2 also has its own role in tongue development.
Collapse
Affiliation(s)
- Manami Takenoshita
- Department of Molecular Craniofacial Embryology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan.,Department of Pediatric Dentistry and Special Needs Dentistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Masaki Takechi
- Department of Molecular Craniofacial Embryology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Tri Vu Hoang
- Department of Molecular Craniofacial Embryology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Toshiko Furutera
- Department of Molecular Craniofacial Embryology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Chisaki Akagawa
- Department of Molecular Craniofacial Embryology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Worachat Namangkalakul
- Department of Molecular Craniofacial Embryology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Kazushi Aoto
- Department of Biochemistry, Hamamatsu University School of Medicine, Tokyo, Japan
| | - Tsutomu Kume
- Feinberg Cardiovascular Research Institute, Development of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Michiyo Miyashin
- Department of Pediatric Dentistry and Special Needs Dentistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Tsutomu Iwamoto
- Department of Pediatric Dentistry and Special Needs Dentistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Sachiko Iseki
- Department of Molecular Craniofacial Embryology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| |
Collapse
|
14
|
The Skull's Girder: A Brief Review of the Cranial Base. J Dev Biol 2021; 9:jdb9010003. [PMID: 33498686 PMCID: PMC7838769 DOI: 10.3390/jdb9010003] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/14/2021] [Accepted: 01/19/2021] [Indexed: 12/11/2022] Open
Abstract
The cranial base is a multifunctional bony platform within the core of the cranium, spanning rostral to caudal ends. This structure provides support for the brain and skull vault above, serves as a link between the head and the vertebral column below, and seamlessly integrates with the facial skeleton at its rostral end. Unique from the majority of the cranial skeleton, the cranial base develops from a cartilage intermediate-the chondrocranium-through the process of endochondral ossification. Owing to the intimate association of the cranial base with nearly all aspects of the head, congenital birth defects impacting these structures often coincide with anomalies of the cranial base. Despite this critical importance, studies investigating the genetic control of cranial base development and associated disorders lags in comparison to other craniofacial structures. Here, we highlight and review developmental and genetic aspects of the cranial base, including its transition from cartilage to bone, dual embryological origins, and vignettes of transcription factors controlling its formation.
Collapse
|
15
|
LaMantia AS. Why Does the Face Predict the Brain? Neural Crest Induction, Craniofacial Morphogenesis, and Neural Circuit Development. Front Physiol 2020; 11:610970. [PMID: 33362582 PMCID: PMC7759552 DOI: 10.3389/fphys.2020.610970] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 11/24/2020] [Indexed: 12/14/2022] Open
Abstract
Mesenchephalic and rhombencephalic neural crest cells generate the craniofacial skeleton, special sensory organs, and subsets of cranial sensory receptor neurons. They do so while preserving the anterior-posterior (A-P) identity of their neural tube origins. This organizational principle is paralleled by central nervous system circuits that receive and process information from facial structures whose A-P identity is in register with that in the brain. Prior to morphogenesis of the face and its circuits, however, neural crest cells act as "inductive ambassadors" from distinct regions of the neural tube to induce differentiation of target craniofacial domains and establish an initial interface between the brain and face. At every site of bilateral, non-axial secondary induction, neural crest constitutes all or some of the mesenchymal compartment for non-axial mesenchymal/epithelial (M/E) interactions. Thus, for epithelial domains in the craniofacial primordia, aortic arches, limbs, the spinal cord, and the forebrain (Fb), neural crest-derived mesenchymal cells establish local sources of inductive signaling molecules that drive morphogenesis and cellular differentiation. This common mechanism for building brains, faces, limbs, and hearts, A-P axis specified, neural crest-mediated M/E induction, coordinates differentiation of distal structures, peripheral neurons that provide their sensory or autonomic innervation in some cases, and central neural circuits that regulate their behavioral functions. The essential role of this neural crest-mediated mechanism identifies it as a prime target for pathogenesis in a broad range of neurodevelopmental disorders. Thus, the face and the brain "predict" one another, and this mutual developmental relationship provides a key target for disruption by developmental pathology.
Collapse
Affiliation(s)
- Anthony-Samuel LaMantia
- Laboratory of Developmental Disorders and Genetics and Center for Neurobiology Research, Fralin Biomedical Research Institute, Department of Pediatrics, Virginia Tech-Carilion School of Medicine, Virginia Tech, Roanoke, VA, United States.,Department of Biological Sciences, Virginia Tech, Blacksburg, VA, United States
| |
Collapse
|
16
|
Kalev-Altman R, Hanael E, Zelinger E, Blum M, Monsonego-Ornan E, Sela-Donenfeld D. Conserved role of matrix metalloproteases 2 and 9 in promoting the migration of neural crest cells in avian and mammalian embryos. FASEB J 2020; 34:5240-5261. [PMID: 32067275 DOI: 10.1096/fj.201901217rr] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 01/28/2020] [Accepted: 02/02/2020] [Indexed: 01/10/2023]
Abstract
Neural crest cells (NCCs) are a unique embryonic cell population that initially reside at the dorsal neural tube but later migrate in the embryo and differentiate into multiple types of derivatives. To acquire motility, NCCs undergo epithelial-to-mesenchymal transition and invade the surrounding extracellular matrix (ECM). Matrix metalloproteases (MMPs) are a large family of proteases which regulate migration of various embryonic and adult cells via ECM remodeling. The gelatinase's subgroup of MMPs is the most studied one due to its key role in metastasis. As it is composed of only two proteases, MMP2 and MMP9, it is important to understand whether each is indispensable or redundant in its biological function. Here we explored the role of the gelatinases in executing NCC migration, by determining whether MMP2 and/or MMP9 regulate migration across species in singular, combined, or redundant manners. Chick and mouse embryos were utilized to compare expression and activity of both MMPs using genetic and pharmacological approaches in multiple in vivo and ex vivo assays. Both MMPs were found to be expressed and active in mouse and chick NCCs. Inhibition of each MMP was sufficient to prevent NCC migration in both species. Yet, NCC migration was maintained in MMP2-/- or MMP9-/- mouse mutants due to compensation between the gelatinases, but reciprocal pharmacological inhibition in each mutant prevented NCC migration. This study reveals for the first time that both gelatinases are expressed in avian and mammalian NCCs, and demonstrates their fundamental and conserved role in promoting embryonic cell migration.
Collapse
Affiliation(s)
- Rotem Kalev-Altman
- Koret School of Veterinary Medicine, Faculty of Agriculture, Food and Environmental Sciences, The Hebrew University, Rehovot, Israel.,The Institute of Biochemistry and Nutrition, Faculty of Agriculture, Food and Environmental Sciences, The Hebrew University, Rehovot, Israel
| | - Erez Hanael
- Koret School of Veterinary Medicine, Faculty of Agriculture, Food and Environmental Sciences, The Hebrew University, Rehovot, Israel
| | - Einat Zelinger
- Core Facility Unit, Faculty of Agriculture, Food and Environmental Sciences, The Hebrew University, Rehovot, Israel
| | - Martin Blum
- Institute of Zoology, University of Hohenheim, Stuttgart, Germany
| | - Efrat Monsonego-Ornan
- The Institute of Biochemistry and Nutrition, Faculty of Agriculture, Food and Environmental Sciences, The Hebrew University, Rehovot, Israel
| | - Dalit Sela-Donenfeld
- Koret School of Veterinary Medicine, Faculty of Agriculture, Food and Environmental Sciences, The Hebrew University, Rehovot, Israel
| |
Collapse
|
17
|
Yamada T, Takechi M, Yokoyama N, Hiraoka Y, Ishikubo H, Usami T, Furutera T, Taga Y, Hirate Y, Kanai-Azuma M, Yoda T, Ogawa-Goto K, Iseki S. Heterozygous mutation of the splicing factor Sf3b4 affects development of the axial skeleton and forebrain in mouse. Dev Dyn 2020; 249:622-635. [PMID: 31900962 DOI: 10.1002/dvdy.148] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Revised: 12/22/2019] [Accepted: 12/26/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Splicing factor 3B subunit 4 (SF3B4) is a causative gene of an acrofacial dysostosis, Nager syndrome. Although in vitro analyses of SF3B4 have proposed multiple noncanonical functions unrelated to splicing, less information is available based on in vivo studies using model animals. RESULTS We performed expression and functional analyses of Sf3b4 in mice. The mouse Sf3b4 transcripts were found from two-cell stage, and were ubiquitously present during embryogenesis with high expression levels in several tissues such as forming craniofacial bones and brain. In contrast, expression of a pseudogene-like sequence of mouse Sf3b4 (Sf3b4_ps) found by in silico survey was not detected up to embryonic day 10. We generated a Sf3b4 knockout mouse using CRISPR-Cas9 system. The homozygous mutant mouse of Sf3b4 was embryonic lethal. The heterozygous mutant of Sf3b4 mouse (Sf3b4+/- ) exhibited smaller body size compared to the wild-type from postnatal to adult period, as well as homeotic posteriorization of the vertebral morphology and flattened calvaria. The flattened calvaria appears to be attributable to mild microcephaly due to a lower cell proliferation rate in the forebrain. CONCLUSIONS Our study suggests that Sf3b4 controls anterior-posterior patterning of the axial skeleton and guarantees cell proliferation for forebrain development in mice.
Collapse
Affiliation(s)
- Takahiko Yamada
- Section of Molecular Craniofacial Embryology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan.,Section of Maxillofacial Surgery, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Masaki Takechi
- Section of Molecular Craniofacial Embryology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Norisuke Yokoyama
- Section of Molecular Craniofacial Embryology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Yuichi Hiraoka
- Laboratory of Genome Editing for Biomedical Research, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan.,Laboratory of Molecular Neuroscience, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Harumi Ishikubo
- Laboratory of Genome Editing for Biomedical Research, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan.,Laboratory of Molecular Neuroscience, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Takako Usami
- Laboratory of Genome Editing for Biomedical Research, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan.,Laboratory of Molecular Neuroscience, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Toshiko Furutera
- Section of Molecular Craniofacial Embryology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Yuki Taga
- Nippi Research Institute of Biomatrix, Ibaraki, Japan
| | - Yoshikazu Hirate
- Department of Experimental Animal Model for Human Disease, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Masami Kanai-Azuma
- Department of Experimental Animal Model for Human Disease, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Tetsuya Yoda
- Section of Maxillofacial Surgery, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | | | - Sachiko Iseki
- Section of Molecular Craniofacial Embryology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| |
Collapse
|
18
|
Feigin CY, Newton AH, Pask AJ. Widespread cis-regulatory convergence between the extinct Tasmanian tiger and gray wolf. Genome Res 2019; 29:1648-1658. [PMID: 31533979 PMCID: PMC6771401 DOI: 10.1101/gr.244251.118] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 08/19/2019] [Indexed: 12/18/2022]
Abstract
The extinct marsupial Tasmanian tiger, or thylacine, and the eutherian gray wolf are among the most widely recognized examples of convergent evolution in mammals. Despite being distantly related, these large predators independently evolved extremely similar craniofacial morphologies, and evidence suggests that they filled similar ecological niches. Previous analyses revealed little evidence of adaptive convergence between their protein-coding genes. Thus, the genetic basis of their convergence is still unclear. Here, we identified candidate craniofacial cis-regulatory elements across vertebrates and compared their evolutionary rates in the thylacine and wolf, revealing abundant signatures of convergent positive selection. Craniofacial thylacine-wolf accelerated regions were enriched near genes involved in TGF beta (TGFB) and BMP signaling, both of which are key morphological signaling pathways with critical roles in establishing the identities and boundaries between craniofacial tissues. Similarly, enhancers of genes involved in craniofacial nerve development showed convergent selection and involvement in these pathways. Taken together, these results suggest that adaptation in cis-regulators of TGF beta and BMP signaling may provide a mechanism to explain the coevolution of developmentally and functionally integrated craniofacial structures in these species. We also found that despite major structural differences in marsupial and eutherian brains, accelerated regions in both species were common near genes with roles in brain development. Our findings support the hypothesis that, relative to protein-coding genes, positive selection on cis-regulatory elements is likely to be an essential driver of adaptive convergent evolution and may underpin thylacine-wolf phenotypic similarities.
Collapse
Affiliation(s)
- Charles Y Feigin
- School of BioSciences, The University of Melbourne, Parkville, Victoria 3010, Australia.,Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544, USA
| | - Axel H Newton
- School of BioSciences, The University of Melbourne, Parkville, Victoria 3010, Australia.,Museums Victoria, Melbourne, Victoria 3053, Australia
| | - Andrew J Pask
- School of BioSciences, The University of Melbourne, Parkville, Victoria 3010, Australia.,Museums Victoria, Melbourne, Victoria 3053, Australia
| |
Collapse
|
19
|
Woronowicz KC, Schneider RA. Molecular and cellular mechanisms underlying the evolution of form and function in the amniote jaw. EvoDevo 2019; 10:17. [PMID: 31417668 PMCID: PMC6691539 DOI: 10.1186/s13227-019-0131-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 07/22/2019] [Indexed: 01/16/2023] Open
Abstract
The amniote jaw complex is a remarkable amalgamation of derivatives from distinct embryonic cell lineages. During development, the cells in these lineages experience concerted movements, migrations, and signaling interactions that take them from their initial origins to their final destinations and imbue their derivatives with aspects of form including their axial orientation, anatomical identity, size, and shape. Perturbations along the way can produce defects and disease, but also generate the variation necessary for jaw evolution and adaptation. We focus on molecular and cellular mechanisms that regulate form in the amniote jaw complex, and that enable structural and functional integration. Special emphasis is placed on the role of cranial neural crest mesenchyme (NCM) during the species-specific patterning of bone, cartilage, tendon, muscle, and other jaw tissues. We also address the effects of biomechanical forces during jaw development and discuss ways in which certain molecular and cellular responses add adaptive and evolutionary plasticity to jaw morphology. Overall, we highlight how variation in molecular and cellular programs can promote the phenomenal diversity and functional morphology achieved during amniote jaw evolution or lead to the range of jaw defects and disease that affect the human condition.
Collapse
Affiliation(s)
- Katherine C Woronowicz
- 1Department of Orthopaedic Surgery, University of California at San Francisco, 513 Parnassus Avenue, S-1161, Box 0514, San Francisco, CA 94143-0514 USA.,2Present Address: Department of Genetics, Harvard Medical School, Orthopaedic Research Laboratories, Children's Hospital Boston, Boston, MA 02115 USA
| | - Richard A Schneider
- 1Department of Orthopaedic Surgery, University of California at San Francisco, 513 Parnassus Avenue, S-1161, Box 0514, San Francisco, CA 94143-0514 USA
| |
Collapse
|
20
|
Kaucka M, Adameyko I. Evolution and development of the cartilaginous skull: From a lancelet towards a human face. Semin Cell Dev Biol 2019; 91:2-12. [DOI: 10.1016/j.semcdb.2017.12.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2017] [Revised: 11/27/2017] [Accepted: 12/09/2017] [Indexed: 11/16/2022]
|
21
|
Diaz RE, Shylo NA, Roellig D, Bronner M, Trainor PA. Filling in the phylogenetic gaps: Induction, migration, and differentiation of neural crest cells in a squamate reptile, the veiled chameleon (Chamaeleo calyptratus). Dev Dyn 2019; 248:709-727. [DOI: 10.1002/dvdy.38] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 04/04/2019] [Accepted: 04/04/2019] [Indexed: 12/13/2022] Open
Affiliation(s)
- Raul E. Diaz
- Department of Biological Sciences, Southeastern Louisiana University Hammond Louisiana
- Natural History Museum of Los Angeles CountyDivision of Herpetology Los Angeles California
| | | | - Daniela Roellig
- Division of Biology and Biological Engineering, California Institute of Technology Pasadena California
| | - Marianne Bronner
- Division of Biology and Biological Engineering, California Institute of Technology Pasadena California
| | - Paul A. Trainor
- Stowers Institute for Medical Research Kansas City Missouri
- Department of Anatomy and Cell Biology, University of Kansas Medical Center Kansas City Kansas
| |
Collapse
|
22
|
Suga M, Furue MK. Neural Crest Cell Models of Development and Toxicity: Cytotoxicity Assay Using Human Pluripotent Stem Cell-Derived Cranial Neural Crest Cell Model. Methods Mol Biol 2019; 1965:35-48. [PMID: 31069667 DOI: 10.1007/978-1-4939-9182-2_4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Cranial neural crest cells (NCCs) migrate to the branchial arches and give rise to the majority of cranial mesenchyme that eventually differentiates into odontoblasts, cartilage, craniofacial bone, and connective tissue; a subset of these cells differentiate into cranial ganglia. Here we present a protocol that describes directed differentiation method of human pluripotent stem cells into cranial NCC-like cells and a cytotoxicity assay using hPSC-derived cranial NCC-like cells. This cell-based assay system allows for high-sensitive cytotoxicity detection of test chemicals. These methods can be applied to predict drug/chemical toxicity effect on early craniofacial development.
Collapse
Affiliation(s)
- Mika Suga
- Laboratory of Stem Cell Cultures, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Ibaraki, Japan
| | - Miho K Furue
- Laboratory of Stem Cell Cultures, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Ibaraki, Japan.
| |
Collapse
|
23
|
Wan Y, Lantz B, Cusack BJ, Szabo-Rogers HL. Prickle1 regulates differentiation of frontal bone osteoblasts. Sci Rep 2018; 8:18021. [PMID: 30575813 PMCID: PMC6303328 DOI: 10.1038/s41598-018-36742-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 11/27/2018] [Indexed: 11/08/2022] Open
Abstract
Enlarged fontanelles and smaller frontal bones result in a mechanically compromised skull. Both phenotypes could develop from defective migration and differentiation of osteoblasts in the skull bone primordia. The Wnt/Planar cell polarity (Wnt/PCP) signaling pathway regulates cell migration and movement in other tissues and led us to test the role of Prickle1, a core component of the Wnt/PCP pathway, in the skull. For these studies, we used the missense allele of Prickle1 named Prickle1Beetlejuice (Prickle1Bj). The Prickle1Bj/Bj mutants are microcephalic and develop enlarged fontanelles between insufficient frontal bones, while the parietal bones are normal. Prickle1Bj/Bj mutants have several other craniofacial defects including a midline cleft lip, incompletely penetrant cleft palate, and decreased proximal-distal growth of the head. We observed decreased Wnt/β-catenin and Hedgehog signaling in the frontal bone condensations of the Prickle1Bj/Bj mutants. Surprisingly, the smaller frontal bones do not result from defects in cell proliferation or death, but rather significantly delayed differentiation and decreased expression of migratory markers in the frontal bone osteoblast precursors. Our data suggests that Prickle1 protein function contributes to both the migration and differentiation of osteoblast precursors in the frontal bone.
Collapse
Affiliation(s)
- Yong Wan
- Center for Craniofacial Regeneration, Department of Oral Biology, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Brandi Lantz
- Center for Craniofacial Regeneration, Department of Oral Biology, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Brian J Cusack
- Center for Craniofacial Regeneration, Department of Oral Biology, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Heather L Szabo-Rogers
- Center for Craniofacial Regeneration, Department of Oral Biology, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Developmental Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
- McGowan Institute of Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
24
|
Kuratani S. The neural crest and origin of the neurocranium in vertebrates. Genesis 2018; 56:e23213. [PMID: 30134067 DOI: 10.1002/dvg.23213] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 04/16/2018] [Accepted: 04/22/2018] [Indexed: 01/20/2023]
Abstract
Cranium of jawed vertebrates is composed of dorsal moiety that encapsulates the brain, or the neurocranium, and the is called the neurocranium, and the ventral moiety, the viscerocranium, that supports the pharynx. In modern jawed vertebrates (crown gnathostomes), the viscerocranium is predominantly of neural crest origin, and for the neurocranium, the rostral part is derived from neural crest cells, whereas the posterior part from the mesoderm. In the cyclostome cranium, the mesoderm/neural crest boundary of the neurocranium used to be enigmatic, let alone the morphological comparison of neurocranial between two cyclostome groups, lampreys and hagfishes. By examining the hagfish development it has become clear that cyclostomes share a common craniofacial embryonic pattern that is not shared by modern gnathostomes, and cyclostome cranium can be compared among the group as developmental modular units with comparable mesoderm/neural crest boundary within the neuroranium. Also, the dual origin of the jawed vertebrate neurocranium has now turned out to represent a derived condition, and ancestrally, the neurocranium would likely have been predominantly of mesodermal origin. Enlargement of the forebrain and reorganization of the oral apparatus seem to have led to the involvement of the neural crest in the rostral neurocranium.
Collapse
Affiliation(s)
- Shigeru Kuratani
- Laboratory for Evolutionary Morphology, RIKEN Center for Biosystems Dynamics Research (BDR) and RIKEN Cluster for Pioneering Research (CPR), Kobe, Hyogo 650-0047, Japan
| |
Collapse
|
25
|
Abstract
PURPOSE To review the recent data about orbital development and sort out the controversies from the very early stages during embryonic life till final maturation of the orbit late in fetal life, and to appreciate the morphogenesis of all the definitive structures in the orbit in a methodical and timely fashion. METHODS The authors extensively review major studies detailing every aspect of human embryologic and fetal orbital morphogenesis including the development of extraocular muscles, orbital fat, vessels, nerves, and the supportive connective tissue framework as well as bone. These interdisciplinary studies span almost a century and a half, and include some significant controversial opposing points of view which the authors hopefully sort out. The authors also highlight a few of the most noteworthy molecular biologic studies regarding the multiple and interacting signaling pathways involved in regulating normal orbital morphogenesis. RESULTS Orbital morphogenesis involves a successive series of subtle yet tightly regulated morphogenetic events that could only be explained through the chronological narrative used by the authors. The processes that trigger and contribute to the formation of the orbits are complex and seem to be intricately regulated by multifaceted interactions and bidirectional cross-talk between a multitude of cellular building raw materials including the developing optic vesicles, neuroectoderm, cranial neural crest cells and mesoderm. CONCLUSIONS Development of the orbit is a collective enterprise necessitating interactions between, as well as contributions from different cell populations both within and beyond the realm of the orbit. A basic understanding of the processes underlying orbital ontogenesis is a crucial first step toward establishing a genetic basis or an embryologic link with orbital disease.
Collapse
|
26
|
Kumar A, Davies TG, Itasaki N. Developmental abnormalities of the otic capsule and inner ear following application of prolyl-hydroxylase inhibitors in chick embryos. Birth Defects Res 2018; 110:1194-1204. [PMID: 30079508 DOI: 10.1002/bdr2.1375] [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: 03/15/2018] [Revised: 06/30/2018] [Accepted: 07/08/2018] [Indexed: 11/10/2022]
Abstract
BACKGROUND Naturally hypoxic conditions in amniote embryos play important roles in normal development. We previously showed that a hypoxic condition is required to produce a sufficient amount of neural crest cells (NCCs) during embryogenesis and that promoting a hypoxic response by prolyl-hydroxylase (PHD) inhibitors increases NCCs. Given that PHD inhibitors are considered as a potential treatment for anemia and ischemic diseases, we investigated the phenotypic effect of PHD inhibitors on embryonic development. METHODS Chick embryos were administered with PHD inhibitors prior to the induction of NCCs on day 1.5. Three main events relating to hypoxia, NCCs induction, vasculogenesis and chondrogenesis, were examined. RESULTS PHD inhibitors caused an increase of Sox10-positive NCCs in vivo. Vasculogenesis was promoted temporarily, although rapid vasculogenesis diminished the effect by day 5 in cephalic and pharyngeal regions. Studies on chondrogenesis at day 7 showed advanced development of the otic capsule, a cartilaginous structure encapsulating the inner ear. Analysis by X-ray micro-computed-tomography (μCT) revealed smaller otic capsule, suggesting premature differentiation. This in turn, deformed the developing semicircular canals within it. Other skeletal structures such as the palate and jaw were unaffected. The localized effect on the otic capsule was considered a result of the multiple effects from the hypoxic responses, increased NCCs and promoted chondrogenesis. CONCLUSION Given the wide range of clinical applications being considered for PHD inhibitors, this study provides crucial information to caution and guide use of PHD inhibitors when treating women of childbearing age.
Collapse
Affiliation(s)
- Akshay Kumar
- Faculty of Health Sciences, University of Bristol, Bristol, United Kingdom
| | - Thomas G Davies
- School of Earth Sciences, University of Bristol, Bristol, United Kingdom
| | - Nobue Itasaki
- Faculty of Health Sciences, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
27
|
Womack MC, Fiero TS, Hoke KL. Trait independence primes convergent trait loss. Evolution 2018; 72:679-687. [PMID: 29383712 DOI: 10.1111/evo.13442] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 01/21/2018] [Indexed: 01/24/2023]
Abstract
The repeated, independent evolution of traits (convergent evolution) is often attributed to shared environmental selection pressures. However, developmental dependencies among traits can limit the phenotypic variation available to selection and bias evolutionary outcomes. Here, we determine how changes in developmentally correlated traits may impact convergent loss of the tympanic middle ear, a highly labile trait within toads that currently lack adaptive explanation. The middle ear's lability could reflect evolutionary trade-offs with other skull features under selection, or the middle ear may evolve independently of the rest of the skull, allowing it to be modified by active or passive processes without pleiotropic trade-offs with other skull features. We compare the skulls of 55 species (39 eared, 16 earless) within the family Bufonidae, spanning six hypothesized independent middle ear transitions. We test whether shared or lineage-specific changes in skull shape distinguish earless species from eared species and whether earless skulls lack other late-forming skull bones. We find no evidence for pleiotropic trade-offs between the middle ear and other skull structures. Instead, middle ear loss in anurans may provide a rare example of developmental independence contributing to evolutionary lability of a sensory system.
Collapse
Affiliation(s)
- Molly C Womack
- Department of Biology, Colorado State University, Fort Collins, Colorado 80523.,Department of Vertebrate Zoology, National Museum of Natural History, Smithsonian Institution, Washington, District of Columbia 20560
| | - Tyler S Fiero
- Department of Biology, Colorado State University, Fort Collins, Colorado 80523
| | - Kim L Hoke
- Department of Biology, Colorado State University, Fort Collins, Colorado 80523
| |
Collapse
|
28
|
Percival CJ, Green R, Roseman CC, Gatti DM, Morgan JL, Murray SA, Donahue LR, Mayeux JM, Pollard KM, Hua K, Pomp D, Marcucio R, Hallgrímsson B. Developmental constraint through negative pleiotropy in the zygomatic arch. EvoDevo 2018; 9:3. [PMID: 29423138 PMCID: PMC5787316 DOI: 10.1186/s13227-018-0092-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2017] [Accepted: 01/08/2018] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Previous analysis suggested that the relative contribution of individual bones to regional skull lengths differ between inbred mouse strains. If the negative correlation of adjacent bone lengths is associated with genetic variation in a heterogeneous population, it would be an example of negative pleiotropy, which occurs when a genetic factor leads to opposite effects in two phenotypes. Confirming negative pleiotropy and determining its basis may reveal important information about the maintenance of overall skull integration and developmental constraint on skull morphology. RESULTS We identified negative correlations between the lengths of the frontal and parietal bones in the midline cranial vault as well as the zygomatic bone and zygomatic process of the maxilla, which contribute to the zygomatic arch. Through gene association mapping of a large heterogeneous population of Diversity Outbred (DO) mice, we identified a quantitative trait locus on chromosome 17 driving the antagonistic contribution of these two zygomatic arch bones to total zygomatic arch length. Candidate genes in this region were identified and real-time PCR of the maxillary processes of DO founder strain embryos indicated differences in the RNA expression levels for two of the candidate genes, Camkmt and Six2. CONCLUSIONS A genomic region underlying negative pleiotropy of two zygomatic arch bones was identified, which provides a mechanism for antagonism in component bone lengths while constraining overall zygomatic arch length. This type of mechanism may have led to variation in the contribution of individual bones to the zygomatic arch noted across mammals. Given that similar genetic and developmental mechanisms may underlie negative correlations in other parts of the skull, these results provide an important step toward understanding the developmental basis of evolutionary variation and constraint in skull morphology.
Collapse
Affiliation(s)
| | - Rebecca Green
- Alberta Children’s Hospital Institute for Child and Maternal Health, University of Calgary, Calgary, AB Canada
- The McCaig Bone and Joint Institute, University of Calgary, Calgary, AB Canada
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB Canada
| | - Charles C. Roseman
- Program in Ecology Evolution and Conservation Biology, University of Illinois, Urbana, IL USA
| | | | | | | | | | - Jessica M. Mayeux
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA USA
| | - K. Michael Pollard
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA USA
| | - Kunjie Hua
- Department of Genetics, University of North Carolina Medical School, Chapel Hill, NC USA
| | - Daniel Pomp
- Department of Genetics, University of North Carolina Medical School, Chapel Hill, NC USA
| | - Ralph Marcucio
- The Orthopaedic Trauma Institute, Department of Orthopaedic Surgery, UCSF School of Medicine, San Francisco, CA USA
| | - Benedikt Hallgrímsson
- Alberta Children’s Hospital Institute for Child and Maternal Health, University of Calgary, Calgary, AB Canada
- The McCaig Bone and Joint Institute, University of Calgary, Calgary, AB Canada
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB Canada
| |
Collapse
|
29
|
Van Otterloo E, Li H, Jones KL, Williams T. AP-2α and AP-2β cooperatively orchestrate homeobox gene expression during branchial arch patterning. Development 2018; 145:dev157438. [PMID: 29229773 PMCID: PMC5825845 DOI: 10.1242/dev.157438] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 12/05/2017] [Indexed: 12/19/2022]
Abstract
The evolution of a hinged moveable jaw with variable morphology is considered a major factor behind the successful expansion of the vertebrates. DLX homeobox transcription factors are crucial for establishing the positional code that patterns the mandible, maxilla and intervening hinge domain, but how the genes encoding these proteins are regulated remains unclear. Herein, we demonstrate that the concerted action of the AP-2α and AP-2β transcription factors within the mouse neural crest is essential for jaw patterning. In the absence of these two proteins, the hinge domain is lost and there are alterations in the size and patterning of the jaws correlating with dysregulation of homeobox gene expression, with reduced levels of Emx, Msx and Dlx paralogs accompanied by an expansion of Six1 expression. Moreover, detailed analysis of morphological features and gene expression changes indicate significant overlap with various compound Dlx gene mutants. Together, these findings reveal that the AP-2 genes have a major function in mammalian neural crest development, influencing patterning of the craniofacial skeleton via the DLX code, an effect that has implications for vertebrate facial evolution, as well as for human craniofacial disorders.
Collapse
Affiliation(s)
- Eric Van Otterloo
- Department of Craniofacial Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Hong Li
- Department of Craniofacial Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Kenneth L Jones
- Department of Pediatrics, Section of Hematology, Oncology, and Bone Marrow Transplant, University of Colorado School of Medicine, Aurora, CO 80045 USA
| | - Trevor Williams
- Department of Craniofacial Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Children's Hospital Colorado, Aurora, CO 80045, USA
| |
Collapse
|
30
|
The skull roof tracks the brain during the evolution and development of reptiles including birds. Nat Ecol Evol 2017; 1:1543-1550. [PMID: 29185519 DOI: 10.1038/s41559-017-0288-2] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 07/24/2017] [Indexed: 01/05/2023]
Abstract
Major transformations in brain size and proportions, such as the enlargement of the brain during the evolution of birds, are accompanied by profound modifications to the skull roof. However, the hypothesis of concerted evolution of shape between brain and skull roof over major phylogenetic transitions, and in particular of an ontogenetic relationship between specific regions of the brain and the skull roof, has never been formally tested. We performed 3D morphometric analyses to examine the deep history of brain and skull-roof morphology in Reptilia, focusing on changes during the well-documented transition from early reptiles through archosauromorphs, including nonavian dinosaurs, to birds. Non-avialan taxa cluster tightly together in morphospace, whereas Archaeopteryx and crown birds occupy a separate region. There is a one-to-one correspondence between the forebrain and frontal bone and the midbrain and parietal bone. Furthermore, the position of the forebrain-midbrain boundary correlates significantly with the position of the frontoparietal suture across the phylogenetic breadth of Reptilia and during the ontogeny of individual taxa. Conservation of position and identity in the skull roof is apparent, and there is no support for previous hypotheses that the avian parietal is a transformed postparietal. The correlation and apparent developmental link between regions of the brain and bony skull elements are likely to be ancestral to Tetrapoda and may be fundamental to all of Osteichthyes, coeval with the origin of the dermatocranium.
Collapse
|
31
|
Li J, Cui Y, Xu J, Wang Q, Yang X, Li Y, Zhang X, Qiu M, Zhang Z, Zhang Z. Suppressor of Fused restraint of Hedgehog activity level is critical for osteogenic proliferation and differentiation during calvarial bone development. J Biol Chem 2017; 292:15814-15825. [PMID: 28794157 DOI: 10.1074/jbc.m117.777532] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Revised: 07/04/2017] [Indexed: 12/31/2022] Open
Abstract
Hedgehog signaling plays crucial roles in the development of calvarial bone, relying on the activation of Gli transcription factors. However, the molecular mechanism of the role of regulated Gli protein level in osteogenic specification of mesenchyme still remains elusive. Here, we show by conditionally inactivating Suppressor of Fused (Sufu), a critical repressor of Hedgehog signaling, in Wnt1-Cre-mediated cranial neural crest (CNC) or Dermo1-Cre-mediated mesodermal lineages that Sufu restraint of Hedgehog activity level is critical for differentiation of preosteogenic mesenchyme. Ablation of Sufu results in failure of calvarial bone formation, including CNC-derived bones and mesoderm-derived bones, depending on the Cre line being used. Although mesenchymal cells populate to frontonasal destinations, where they are then condensed, Sufu deletion significantly inhibits the proliferation of osteoprogenitor cells, and these cells no longer differentiate into osteoblasts. We show that there is suppression of Runx2 and Osterix, the osteogenic regulators, in calvarial mesenchyme in the Sufu mutant. We show that down-regulation of several genes upstream to Runx2 and Osterix is manifested within the calvarial primordia, including Bmp2 and its downstream genes Msx1/2 and Dlx5 By contrast, we find that Gli1, the Hedgehog activity readout gene, is excessively activated in mesenchyme. Deletion of Sufu in CNC leads to a discernible decrease in the repressive Gli3 form and an increase in the full-length Gli2. Finally, we demonstrate that simultaneous deletion of Gli2 and Sufu in CNC completely restores calvarial bone formation, suggesting that a sustained level of Hedgehog activity is critical in specification of the osteogenic mesenchymal cells.
Collapse
Affiliation(s)
- Jianying Li
- From the Zhejiang Key Laboratory for Organogenesis and Regenerative Technology, Institute of Life Sciences, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou 310036, China and
| | - Ying Cui
- From the Zhejiang Key Laboratory for Organogenesis and Regenerative Technology, Institute of Life Sciences, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou 310036, China and
| | - Jie Xu
- From the Zhejiang Key Laboratory for Organogenesis and Regenerative Technology, Institute of Life Sciences, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou 310036, China and
| | - Qihui Wang
- From the Zhejiang Key Laboratory for Organogenesis and Regenerative Technology, Institute of Life Sciences, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou 310036, China and
| | - Xueqin Yang
- From the Zhejiang Key Laboratory for Organogenesis and Regenerative Technology, Institute of Life Sciences, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou 310036, China and
| | - Yan Li
- From the Zhejiang Key Laboratory for Organogenesis and Regenerative Technology, Institute of Life Sciences, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou 310036, China and
| | - Xiaoyun Zhang
- From the Zhejiang Key Laboratory for Organogenesis and Regenerative Technology, Institute of Life Sciences, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou 310036, China and
| | - Mengsheng Qiu
- From the Zhejiang Key Laboratory for Organogenesis and Regenerative Technology, Institute of Life Sciences, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou 310036, China and
| | - Ze Zhang
- the Department of Ophthamology, Tulane Medical Center, Tulane University, New Orleans, Louisiana 70112
| | - Zunyi Zhang
- From the Zhejiang Key Laboratory for Organogenesis and Regenerative Technology, Institute of Life Sciences, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou 310036, China and
| |
Collapse
|
32
|
Heuzé Y, Kawasaki K, Schwarz T, Schoenebeck JJ, Richtsmeier JT. Developmental and Evolutionary Significance of the Zygomatic Bone. Anat Rec (Hoboken) 2017; 299:1616-1630. [PMID: 27870340 PMCID: PMC5111587 DOI: 10.1002/ar.23449] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2016] [Revised: 06/23/2016] [Accepted: 06/23/2016] [Indexed: 01/18/2023]
Abstract
The zygomatic bone is derived evolutionarily from the orbital series. In most modern mammals the zygomatic bone forms a large part of the face and usually serves as a bridge that connects the facial skeleton to the neurocranium. Our aim is to provide information on the contribution of the zygomatic bone to variation in midfacial protrusion using three samples; humans, domesticated dogs, and monkeys. In each case, variation in midface protrusion is a heritable trait produced by one of three classes of transmission: localized dysmorphology associated with single gene dysfunction, selective breeding, or long‐term evolution from a common ancestor. We hypothesize that the shape of the zygomatic bone reflects its role in stabilizing the connection between facial skeleton and neurocranium and consequently, changes in facial protrusion are more strongly reflected by the maxilla and premaxilla. Our geometric morphometric analyses support our hypothesis suggesting that the shape of the zygomatic bone has less to do with facial protrusion. By morphometrically dissecting the zygomatic bone we have determined a degree of modularity among parts of the midfacial skeleton suggesting that these components have the ability to vary independently and thus can evolve differentially. From these purely morphometric data, we propose that the neural crest cells that are fated to contribute to the zygomatic bone experience developmental cues that distinguish them from the maxilla and premaxilla. The spatiotemporal and molecular identity of the cues that impart zygoma progenitors with their identity remains an open question that will require alternative data sets. Anat Rec, 299:1616–1630, 2016. © 2016 The Authors The Anatomical Record Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Yann Heuzé
- UMR5199 PACEA, Bordeaux Archaeological Sciences Cluster of Excellence, Université De Bordeaux
| | - Kazuhiko Kawasaki
- Department of Anthropology, Pennsylvania State University, University Park, PA
| | - Tobias Schwarz
- Department of Veterinary Clinical Studies, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Veterinary Centre, Roslin, Midlothian, UK
| | - Jeffrey J Schoenebeck
- Division of Genetics and Genomics, The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian, UK
| | - Joan T Richtsmeier
- Department of Anthropology, Pennsylvania State University, University Park, PA
| |
Collapse
|
33
|
Warth P, Hilton EJ, Naumann B, Olsson L, Konstantinidis P. Development of the skull and pectoral girdle in Siberian sturgeon,Acipenser baerii, and Russian sturgeon,Acipenser gueldenstaedtii(Acipenseriformes: Acipenseridae). J Morphol 2017; 278:418-442. [DOI: 10.1002/jmor.20653] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 12/23/2016] [Accepted: 12/30/2016] [Indexed: 11/11/2022]
Affiliation(s)
- Peter Warth
- Institut für Spezielle Zoologie und Evolutionsbiologie mit Phyletischem Museum, Friedrich-Schiller-Universität Jena; Germany
| | - Eric J. Hilton
- Department of Fisheries Science; Virginia Institute of Marine Science, College of William & Mary; Gloucester Point Virginia
| | - Benjamin Naumann
- Institut für Spezielle Zoologie und Evolutionsbiologie mit Phyletischem Museum, Friedrich-Schiller-Universität Jena; Germany
| | - Lennart Olsson
- Institut für Spezielle Zoologie und Evolutionsbiologie mit Phyletischem Museum, Friedrich-Schiller-Universität Jena; Germany
| | | |
Collapse
|
34
|
Diogo R, Guinard G, Diaz RE. Dinosaurs, Chameleons, Humans, and Evo-Devo Path: Linking Étienne Geoffroy's Teratology, Waddington's Homeorhesis, Alberch's Logic of "Monsters," and Goldschmidt Hopeful "Monsters". JOURNAL OF EXPERIMENTAL ZOOLOGY PART B-MOLECULAR AND DEVELOPMENTAL EVOLUTION 2016; 328:207-229. [PMID: 28422426 DOI: 10.1002/jez.b.22709] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 09/29/2016] [Accepted: 10/03/2016] [Indexed: 12/12/2022]
Abstract
Since the rise of evo-devo (evolutionary developmental biology) in the 1980s, few authors have attempted to combine the increasing knowledge obtained from the study of model organisms and human medicine with data from comparative anatomy and evolutionary biology in order to investigate the links between development, pathology, and macroevolution. Fortunately, this situation is slowly changing, with a renewed interest in evolutionary developmental pathology (evo-devo-path) in the past decades, as evidenced by the idea to publish this special, and very timely, issue on "Developmental Evolution in Biomedical Research." As all of us have recently been involved, independently, in works related in some way or another with evolution and developmental anomalies, we decided to join our different perspectives and backgrounds in the present contribution for this special issue. Specifically, we provide a brief historical account on the study of the links between evolution, development, and pathologies, followed by a review of the recent work done by each of us, and then by a general discussion on the broader developmental and macroevolutionary implications of our studies and works recently done by other authors. Our primary aims are to highlight the strength of studying developmental anomalies within an evolutionary framework to understand morphological diversity and disease by connecting the recent work done by us and others with the research done and broader ideas proposed by authors such as Étienne Geoffroy Saint-Hilaire, Waddington, Goldschmidt, Gould, and Per Alberch, among many others to pave the way for further and much needed work regarding abnormal development and macroevolution.
Collapse
Affiliation(s)
- Rui Diogo
- Department of Anatomy, College of Medicine, Howard University, Washington, District of Columbia
| | - Geoffrey Guinard
- UMR CNRS 5561, Biogéosciences, Université de Bourgogne, Dijon, France
| | - Raul E Diaz
- Department of Biology, La Sierra University, Riverside, California.,Natural History Museum of Los Angeles County, Los Angeles, California
| |
Collapse
|
35
|
Genetics of Skeletal Evolution in Unusually Large Mice from Gough Island. Genetics 2016; 204:1559-1572. [PMID: 27694627 DOI: 10.1534/genetics.116.193805] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 09/26/2016] [Indexed: 11/18/2022] Open
Abstract
Organisms on islands often undergo rapid morphological evolution, providing a platform for understanding mechanisms of phenotypic change. Many examples of evolution on islands involve the vertebrate skeleton. Although the genetic basis of skeletal variation has been studied in laboratory strains, especially in the house mouse Mus musculus domesticus, the genetic determinants of skeletal evolution in natural populations remain poorly understood. We used house mice living on the remote Gough Island-the largest wild house mice on record-to understand the genetics of rapid skeletal evolution in nature. Compared to a mainland reference strain from the same subspecies (WSB/EiJ), the skeleton of Gough Island mice is considerably larger, with notable expansions of the pelvis and limbs. The Gough Island mouse skeleton also displays changes in shape, including elongations of the skull and the proximal vs. distal elements in the limbs. Quantitative trait locus (QTL) mapping in a large F2 intercross between Gough Island mice and WSB/EiJ reveals hundreds of QTL that control skeletal dimensions measured at 5, 10, and/or 16 weeks of age. QTL exhibit modest, mostly additive effects, and Gough Island alleles are associated with larger skeletal size at most QTL. The QTL with the largest effects are found on a few chromosomes and affect suites of skeletal traits. Many of these loci also colocalize with QTL for body weight. The high degree of QTL colocalization is consistent with an important contribution of pleiotropy to skeletal evolution. Our results provide a rare portrait of the genetic basis of skeletal evolution in an island population and position the Gough Island mouse as a model system for understanding mechanisms of rapid evolution in nature.
Collapse
|
36
|
Jourdeuil K, Franz-Odendaal TA. Gene expression analysis during the induction and patterning of the conjunctival papillae in the chick embryonic eye. Gene Expr Patterns 2016; 22:30-36. [DOI: 10.1016/j.gep.2016.09.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 07/25/2016] [Accepted: 09/20/2016] [Indexed: 11/28/2022]
|
37
|
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
|
38
|
Flaherty K, Singh N, Richtsmeier JT. Understanding craniosynostosis as a growth disorder. WILEY INTERDISCIPLINARY REVIEWS. DEVELOPMENTAL BIOLOGY 2016; 5:429-59. [PMID: 27002187 PMCID: PMC4911263 DOI: 10.1002/wdev.227] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Revised: 12/06/2015] [Accepted: 12/24/2015] [Indexed: 12/15/2022]
Abstract
Craniosynostosis is a condition of complex etiology that always involves the premature fusion of one or multiple cranial sutures and includes various anomalies of the soft and hard tissues of the head. Steady progress in the field has resulted in identifying gene mutations that recurrently cause craniosynostosis. There are now scores of mutations on many genes causally related to craniosynostosis syndromes, though the genetic basis for the majority of nonsyndromic cases is unknown. Identification of these genetic mutations has allowed significant progress in understanding the intrinsic properties of cranial sutures, including mechanisms responsible for normal suture patency and for pathogenesis of premature suture closure. An understanding of morphogenesis of cranial vault sutures is critical to understanding the pathophysiology of craniosynostosis conditions, but the field is now poised to recognize the repeated changes in additional skeletal and soft tissues of the head that typically accompany premature suture closure. We review the research that has brought an understanding of premature suture closure within our reach. We then enumerate the less well-studied, but equally challenging, nonsutural phenotypes of craniosynostosis conditions that are well characterized in available mouse models. We consider craniosynostosis as a complex growth disorder of multiple tissues of the developing head, whose growth is also targeted by identified mutations in ways that are poorly understood. Knowledge gained from studies of humans and mouse models for these conditions underscores the diverse, associated developmental anomalies of the head that contribute to the complex phenotypes of craniosynostosis conditions presenting novel challenges for future research. WIREs Dev Biol 2016, 5:429-459. doi: 10.1002/wdev.227 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Kevin Flaherty
- Department of Anthropology, Pennsylvania State University,
University Park, PA 16802
| | - Nandini Singh
- Department of Anthropology, Pennsylvania State University,
University Park, PA 16802
| | - Joan T. Richtsmeier
- Department of Anthropology, Pennsylvania State University,
University Park, PA 16802
| |
Collapse
|
39
|
Adams DS, Uzel SGM, Akagi J, Wlodkowic D, Andreeva V, Yelick PC, Devitt-Lee A, Pare JF, Levin M. Bioelectric signalling via potassium channels: a mechanism for craniofacial dysmorphogenesis in KCNJ2-associated Andersen-Tawil Syndrome. J Physiol 2016; 594:3245-70. [PMID: 26864374 PMCID: PMC4908029 DOI: 10.1113/jp271930] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 02/01/2016] [Indexed: 12/21/2022] Open
Abstract
KEY POINTS Xenopus laevis craniofacial development is a good system for the study of Andersen-Tawil Syndrome (ATS)-associated craniofacial anomalies (CFAs) because (1) Kcnj2 is expressed in the nascent face; (2) molecular-genetic and biophysical techniques are available for the study of ion-dependent signalling during craniofacial morphogenesis; (3) as in humans, expression of variant Kcnj2 forms in embryos causes a muscle phenotype; and (4) variant forms of Kcnj2 found in human patients, when injected into frog embryos, cause CFAs in the same cell lineages. Forced expression of WT or variant Kcnj2 changes the normal pattern of Vmem (resting potential) regionalization found in the ectoderm of neurulating embryos, and changes the normal pattern of expression of ten different genetic regulators of craniofacial development, including markers of cranial neural crest and of placodes. Expression of other potassium channels and two different light-activated channels, all of which have an effect on Vmem , causes CFAs like those induced by injection of Kcnj2 variants. In contrast, expression of Slc9A (NHE3), an electroneutral ion channel, and of GlyR, an inactive Cl(-) channel, do not cause CFAs, demonstrating that correct craniofacial development depends on a pattern of bioelectric states, not on ion- or channel-specific signalling. Using optogenetics to control both the location and the timing of ion flux in developing embryos, we show that affecting Vmem of the ectoderm and no other cell layers is sufficient to cause CFAs, but only during early neurula stages. Changes in Vmem induced late in neurulation do not affect craniofacial development. We interpret these data as strong evidence, consistent with our hypothesis, that ATS-associated CFAs are caused by the effect of variant Kcnj2 on the Vmem of ectodermal cells of the developing face. We predict that the critical time is early during neurulation, and the critical cells are the ectodermal cranial neural crest and placode lineages. This points to the potential utility of extant, ion flux-modifying drugs as treatments to prevent CFAs associated with channelopathies such as ATS. ABSTRACT Variants in potassium channel KCNJ2 cause Andersen-Tawil Syndrome (ATS); the induced craniofacial anomalies (CFAs) are entirely unexplained. We show that KCNJ2 is expressed in Xenopus and mouse during the earliest stages of craniofacial development. Misexpression in Xenopus of KCNJ2 carrying ATS-associated mutations causes CFAs in the same structures affected in humans, changes the normal pattern of membrane voltage potential regionalization in the developing face and disrupts expression of important craniofacial patterning genes, revealing the endogenous control of craniofacial patterning by bioelectric cell states. By altering cells' resting potentials using other ion translocators, we show that a change in ectodermal voltage, not tied to a specific protein or ion, is sufficient to cause CFAs. By adapting optogenetics for use in non-neural cells in embryos, we show that developmentally patterned K(+) flux is required for correct regionalization of the resting potentials and for establishment of endogenous early gene expression domains in the anterior ectoderm, and that variants in KCNJ2 disrupt this regionalization, leading to the CFAs seen in ATS patients.
Collapse
Affiliation(s)
- Dany Spencer Adams
- Department of Biology and Tufts Centre for Regenerative and Developmental Biology, Tufts University, 200 Boston Avenue, Medford, MA, 02155, USA
| | - Sebastien G M Uzel
- Department of Mechanical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Jin Akagi
- School of Applied Sciences, RMIT University, Melbourne, Australia
| | - Donald Wlodkowic
- School of Applied Sciences, RMIT University, Melbourne, Australia
| | - Viktoria Andreeva
- Department of Orthodontics, Division of Craniofacial and Molecular Genetics, Tufts University School of Dental Medicine, Boston, MA 02111, USA
| | - Pamela Crotty Yelick
- Department of Orthodontics, Division of Craniofacial and Molecular Genetics, Tufts University School of Dental Medicine, Boston, MA 02111, USA
| | - Adrian Devitt-Lee
- Department of Biology and Tufts Centre for Regenerative and Developmental Biology, Tufts University, 200 Boston Avenue, Medford, MA, 02155, USA
| | - Jean-Francois Pare
- Department of Biology and Tufts Centre for Regenerative and Developmental Biology, Tufts University, 200 Boston Avenue, Medford, MA, 02155, USA
| | - Michael Levin
- Department of Biology and Tufts Centre for Regenerative and Developmental Biology, Tufts University, 200 Boston Avenue, Medford, MA, 02155, USA
| |
Collapse
|
40
|
Lovely CB, Swartz ME, McCarthy N, Norrie JL, Eberhart JK. Bmp signaling mediates endoderm pouch morphogenesis by regulating Fgf signaling in zebrafish. Development 2016; 143:2000-11. [PMID: 27122171 DOI: 10.1242/dev.129379] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 04/12/2016] [Indexed: 02/03/2023]
Abstract
The endodermal pouches are a series of reiterated structures that segment the pharyngeal arches and help pattern the vertebrate face. Multiple pathways regulate the complex process of endodermal development, including the Bone morphogenetic protein (Bmp) pathway. However, the role of Bmp signaling in pouch morphogenesis is poorly understood. Using genetic and chemical inhibitor approaches, we show that pouch morphogenesis requires Bmp signaling from 10-18 h post-fertilization, immediately following gastrulation. Blocking Bmp signaling during this window results in morphological defects to the pouches and craniofacial skeleton. Using genetic chimeras we show that Bmp signals directly to the endoderm for proper morphogenesis. Time-lapse imaging and analysis of reporter transgenics show that Bmp signaling is necessary for pouch outpocketing via the Fibroblast growth factor (Fgf) pathway. Double loss-of-function analyses demonstrate that Bmp and Fgf signaling interact synergistically in craniofacial development. Collectively, our analyses shed light on the tissue and signaling interactions that regulate development of the vertebrate face.
Collapse
Affiliation(s)
- C Ben Lovely
- Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Mary E Swartz
- Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Neil McCarthy
- Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | | | - Johann K Eberhart
- Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
41
|
Adameyko I, Fried K. The Nervous System Orchestrates and Integrates Craniofacial Development: A Review. Front Physiol 2016; 7:49. [PMID: 26924989 PMCID: PMC4759458 DOI: 10.3389/fphys.2016.00049] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 02/02/2016] [Indexed: 01/14/2023] Open
Abstract
Development of a head is a dazzlingly complex process: a number of distinct cellular sources including cranial ecto- and endoderm, mesoderm and neural crest contribute to facial and other structures. In the head, an extremely fine-tuned developmental coordination of CNS, peripheral neural components, sensory organs and a musculo-skeletal apparatus occurs, which provides protection and functional integration. The face can to a large extent be considered as an assembly of sensory systems encased and functionally fused with appendages represented by jaws. Here we review how the developing brain, neurogenic placodes and peripheral nerves influence the morphogenesis of surrounding tissues as a part of various general integrative processes in the head. The mechanisms of this impact, as we understand it now, span from the targeted release of the morphogens necessary for shaping to providing a niche for cellular sources required in later development. In this review we also discuss the most recent findings and ideas related to how peripheral nerves and nerve-associated cells contribute to craniofacial development, including teeth, during the post- neural crest period and potentially in regeneration.
Collapse
Affiliation(s)
- Igor Adameyko
- Department of Physiology and Pharmacology, Karolinska InstitutetStockholm, Sweden; Department of Molecular Neurosciences, Center of Brain Research, Medical University of ViennaVienna, Austria
| | - Kaj Fried
- Department of Neuroscience, Karolinska Institutet Stockholm, Sweden
| |
Collapse
|
42
|
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
|
43
|
Van Otterloo E, Feng W, Jones KL, Hynes NE, Clouthier DE, Niswander L, Williams T. MEMO1 drives cranial endochondral ossification and palatogenesis. Dev Biol 2015; 415:278-295. [PMID: 26746790 DOI: 10.1016/j.ydbio.2015.12.024] [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: 07/02/2015] [Revised: 11/02/2015] [Accepted: 12/21/2015] [Indexed: 02/01/2023]
Abstract
The cranial base is a component of the neurocranium and has a central role in the structural integration of the face, brain and vertebral column. Consequently, alteration in the shape of the human cranial base has been intimately linked with primate evolution and defective development is associated with numerous human facial abnormalities. Here we describe a novel recessive mutant mouse strain that presented with a domed head and fully penetrant cleft secondary palate coupled with defects in the formation of the underlying cranial base. Mapping and non-complementation studies revealed a specific mutation in Memo1 - a gene originally associated with cell migration. Expression analysis of Memo1 identified robust expression in the perichondrium and periosteum of the developing cranial base, but only modest expression in the palatal shelves. Fittingly, although the palatal shelves failed to elevate in Memo1 mutants, expression changes were modest within the shelves themselves. In contrast, the cranial base, which forms via endochondral ossification had major reductions in the expression of genes responsible for bone formation, notably matrix metalloproteinases and markers of the osteoblast lineage, mirrored by an increase in markers of cartilage and extracellular matrix development. Concomitant with these changes, mutant cranial bases showed an increased zone of hypertrophic chondrocytes accompanied by a reduction in both vascular invasion and mineralization. Finally, neural crest cell-specific deletion of Memo1 caused a failure of anterior cranial base ossification indicating a cell autonomous role for MEMO1 in the development of these neural crest cell derived structures. However, palate formation was largely normal in these conditional mutants, suggesting a non-autonomous role for MEMO1 in palatal closure. Overall, these findings assign a new function to MEMO1 in driving endochondral ossification in the cranium, and also link abnormal development of the cranial base with more widespread effects on craniofacial shape relevant to human craniofacial dysmorphology.
Collapse
Affiliation(s)
- Eric Van Otterloo
- Department of Craniofacial Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.
| | - Weiguo Feng
- Department of Craniofacial Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Kenneth L Jones
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Nancy E Hynes
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, CH-4058 Basel, Switzerland; University of Basel, CH-4002 Basel, Switzerland
| | - David E Clouthier
- Department of Craniofacial Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Lee Niswander
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Department of Pediatrics, University of Colorado Anschutz Medical Campus, Children's Hospital Colorado, Aurora, CO 80045, USA
| | - Trevor Williams
- Department of Craniofacial Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Department of Pediatrics, University of Colorado Anschutz Medical Campus, Children's Hospital Colorado, Aurora, CO 80045, USA
| |
Collapse
|
44
|
Fish JL. Developmental mechanisms underlying variation in craniofacial disease and evolution. Dev Biol 2015; 415:188-197. [PMID: 26724698 DOI: 10.1016/j.ydbio.2015.12.019] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 12/17/2015] [Accepted: 12/21/2015] [Indexed: 01/14/2023]
Abstract
Craniofacial disease phenotypes exhibit significant variation in penetrance and severity. Although many genetic contributions to phenotypic variation have been identified, genotype-phenotype correlations remain imprecise. Recent work in evolutionary developmental biology has exposed intriguing developmental mechanisms that potentially explain incongruities in genotype-phenotype relationships. This review focuses on two observations from work in comparative and experimental animal model systems that highlight how development structures variation. First, multiple genetic inputs converge on relatively few developmental processes. Investigation of when and how variation in developmental processes occurs may therefore help predict potential genetic interactions and phenotypic outcomes. Second, genetic mutation is typically associated with an increase in phenotypic variance. Several models outlining developmental mechanisms underlying mutational increases in phenotypic variance are discussed using Satb2-mediated variation in jaw size as an example. These data highlight development as a critical mediator of genotype-phenotype correlations. Future research in evolutionary developmental biology focusing on tissue-level processes may help elucidate the "black box" between genotype and phenotype, potentially leading to novel treatment, earlier diagnoses, and better clinical consultations for individuals affected by craniofacial anomalies.
Collapse
Affiliation(s)
- Jennifer L Fish
- University of Massachusetts Lowell, Department of Biological Sciences, 198 Riverside Street, Olsen Hall, Room 619, Lowell, MA 01854, United States.
| |
Collapse
|
45
|
Sheng G. The developmental basis of mesenchymal stem/stromal cells (MSCs). BMC DEVELOPMENTAL BIOLOGY 2015; 15:44. [PMID: 26589542 PMCID: PMC4654913 DOI: 10.1186/s12861-015-0094-5] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 11/13/2015] [Indexed: 02/07/2023]
Abstract
BACKGROUND Mesenchymal Stem/Stromal Cells (MSCs) define a population of progenitor cells capable of giving rises to at least three mesodermal lineages in vitro, the chondrocytes, osteoblasts and adipocytes. The validity of MSCs in vivo has been questioned because their existence, either as a homogeneous progenitor cell population or as a stem cell lineage, has been difficult to prove. The wide use of primary MSCs in regenerative and therapeutic applications raises ethical and regulatory concerns in many countries. In contrast to hematopoietic stem cells, a parallel concept which carries an embryological emphasis from its outset, MSCs have attracted little interest among developmental biologists and the embryological basis for their existence, or lack thereof, has not been carefully evaluated. METHODS This article provides a brief, embryological overview of these three mesoderm cell lineages and offers a framework of ontological rationales for the potential existence of MSCs in vivo. RESULTS Emphasis is given to the common somatic lateral plate mesoderm origin of the majority of body's adipose and skeletal tissues and of the major sources used for MSC derivation clinically. Support for the MSC hypothesis also comes from a large body of molecular and lineage analysis data in vivo. CONCLUSIONS It is concluded that despite the lack of a definitive proof, the MSC concept has a firm embryological basis and that advances in MSC research can be facilitated by achieving a better integration with developmental biology.
Collapse
Affiliation(s)
- Guojun Sheng
- Sheng Laboratory, International Research Center for Medical Sciences, Kumamoto University, Kumamoto, 860-0811, Japan.
| |
Collapse
|
46
|
Anthwal N, Peters H, Tucker AS. Species-specific modifications of mandible shape reveal independent mechanisms for growth and initiation of the coronoid. EvoDevo 2015; 6:35. [PMID: 26568815 PMCID: PMC4644282 DOI: 10.1186/s13227-015-0030-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Accepted: 10/12/2015] [Indexed: 01/10/2023] Open
Abstract
Background The variation in mandibular morphology of mammals reflects specialisations for different diets. Omnivorous and carnivorous mammals posses large mandibular coronoid processes, while herbivorous mammals have proportionally smaller or absent coronoids. This is correlated with the relative size of the temporalis muscle that forms an attachment to the coronoid process. The role of this muscle attachment in the development of the variation of the coronoid is unclear. Results By comparative developmental biology and mouse knockout studies, we demonstrate here that the initiation and growth of the coronoid are two independent processes, with initiation being intrinsic to the ossifying bone and growth dependent upon the extrinsic effect of muscle attachment. A necessary component of the intrinsic patterning is identified as the paired domain transcription factor Pax9. We also demonstrate that Sox9 plays a role independent of chondrogenesis in the growth of the coronoid process in response to muscle interaction. Conclusions The mandibular coronoid process is initiated by intrinsic factors, but later growth is dependent on extrinsic signals from the muscle. These extrinsic influences are hypothesised to be the basis of the variation in coronoid length seen across the mammalian lineage.
Collapse
Affiliation(s)
- Neal Anthwal
- Department of Craniofacial Development and Stem Cell Biology, Dental Institute, King's College London, London, SE1 9RT UK
| | - Heiko Peters
- Institute of Genetic Medicine, International Centre for Life, Newcastle University, Newcastle upon Tyne, NE1 3BZ UK
| | - Abigail S Tucker
- Department of Craniofacial Development and Stem Cell Biology, Dental Institute, King's College London, London, SE1 9RT UK
| |
Collapse
|
47
|
Homayounfar N, Park SS, Afsharinejad Z, Bammler TK, MacDonald JW, Farin FM, Mecham BH, Cunningham ML. Transcriptional analysis of human cranial compartments with different embryonic origins. Arch Oral Biol 2015; 60:1450-60. [PMID: 26188427 PMCID: PMC4750879 DOI: 10.1016/j.archoralbio.2015.06.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Revised: 06/11/2015] [Accepted: 06/12/2015] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Previous investigations suggest that the embryonic origins of the calvarial tissues (neural crest or mesoderm) may account for the molecular mechanisms underlying sutural development. The aim of this study was to evaluate the differences in the gene expression of human cranial tissues and assess the presence of an expression signature reflecting their embryonic origins. METHODS Using microarray technology, we investigated global gene expression of cells from the frontal and parietal bones and the metopic and sagittal intrasutural mesenchyme (ISM) of four human foetal calvaria. qRT-PCR of a selected group of genes was done to validate the microarray analysis. Paired comparison and correlation analyses were performed on microarray results. RESULTS Of six paired comparisons, frontal and parietal compartments (distinct tissue types of calvaria, either bone or intrasutural mesenchyme) had the most different gene expression profiles despite being composed of the same tissue type (bone). Correlation analysis revealed two distinct gene expression profiles that separate frontal and metopic compartments from parietal and sagittal compartments. TFAP2A, TFAP2B, ICAM1, SULF1, TNC and FOXF2 were among differentially expressed genes. CONCLUSION Transcriptional profiles of two groups of tissues, frontal and metopic compartments vs. parietal and sagittal compartments, suggest differences in proliferation, differentiation and extracellular matrix production. Our data suggest that in the second trimester of human foetal development, a gene expression signature of neural crest origin still exists in frontal and metopic compartments while gene expression of parietal and sagittal compartments is more similar to mesoderm.
Collapse
Affiliation(s)
- Negar Homayounfar
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, 1900 - 9th Avenue, Seattle, WA 98101, United States; Department of Oral Health Sciences, Dental School, University of Washington, United States; Department of Endodontics, Prosthodontics and Operative Dentistry, School of Dentistry, University of Maryland, Baltimore, United States.
| | - Sarah S Park
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, 1900 - 9th Avenue, Seattle, WA 98101, United States
| | - Zahra Afsharinejad
- Department of Environmental and Occupational Health Sciences, University of Washington, 4225 Roosevelt Way NE, # 100, Seattle, WA 98105-6099, United States
| | - Theodor K Bammler
- Department of Environmental and Occupational Health Sciences, University of Washington, 4225 Roosevelt Way NE, # 100, Seattle, WA 98105-6099, United States
| | - James W MacDonald
- Department of Environmental and Occupational Health Sciences, University of Washington, 4225 Roosevelt Way NE, # 100, Seattle, WA 98105-6099, United States
| | - Federico M Farin
- Department of Environmental and Occupational Health Sciences, University of Washington, 4225 Roosevelt Way NE, # 100, Seattle, WA 98105-6099, United States
| | - Brigham H Mecham
- Trialomics, 1700 7th Avenue, # 116, Seattle, WA 98101, United States
| | - Michael L Cunningham
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, 1900 - 9th Avenue, Seattle, WA 98101, United States; Seattle Children's Craniofacial Center, 4800 Sand Point Way NE, Seattle, WA 98105, United States
| |
Collapse
|
48
|
Atkins JB, Franz-Odendaal TA. The evolutionary and morphological history of the parasphenoid bone in vertebrates. ACTA ZOOL-STOCKHOLM 2015. [DOI: 10.1111/azo.12131] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- Jade B. Atkins
- Saint Mary's University; 923 Robie Street Halifax NS B3H 3C3 Canada
| | | |
Collapse
|
49
|
Hirasawa T, Kuratani S. Evolution of the vertebrate skeleton: morphology, embryology, and development. ZOOLOGICAL LETTERS 2015; 1:2. [PMID: 26605047 PMCID: PMC4604106 DOI: 10.1186/s40851-014-0007-7] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Accepted: 02/19/2014] [Indexed: 05/08/2023]
Abstract
Two major skeletal systems-the endoskeleton and exoskeleton-are recognized in vertebrate evolution. Here, we propose that these two systems are distinguished primarily by their relative positions, not by differences in embryonic histogenesis or cell lineage of origin. Comparative embryologic analyses have shown that both types of skeleton have changed their mode of histogenesis during evolution. Although exoskeletons were thought to arise exclusively from the neural crest, recent experiments in teleosts have shown that exoskeletons in the trunk are mesodermal in origin. The enameloid and dentine-coated postcranial exoskeleton seen in many vertebrates does not appear to represent an ancestral condition, as previously hypothesized, but rather a derived condition, in which the enameloid and dentine tissues became accreted to bones. Recent data from placoderm fossils are compatible with this scenario. In contrast, the skull contains neural crest-derived bones in its rostral part. Recent developmental studies suggest that the boundary between neural crest- and mesoderm-derived bones may not be consistent throughout evolution. Rather, the relative positions of bony elements may be conserved, and homologies of bony elements have been retained, with opportunistic changes in the mechanisms and cell lineages of development.
Collapse
Affiliation(s)
- Tatsuya Hirasawa
- Evolutionary Morphology Laboratory, RIKEN, 2-2-3 Minatojima-minami, Chuo-ku, Kobe, Hyogo 650-0047 Japan
| | - Shigeru Kuratani
- Evolutionary Morphology Laboratory, RIKEN, 2-2-3 Minatojima-minami, Chuo-ku, Kobe, Hyogo 650-0047 Japan
| |
Collapse
|
50
|
Dupin E, Le Douarin NM. The neural crest, a multifaceted structure of the vertebrates. ACTA ACUST UNITED AC 2014; 102:187-209. [PMID: 25219958 DOI: 10.1002/bdrc.21080] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2014] [Accepted: 08/22/2014] [Indexed: 12/29/2022]
Abstract
In this review, several features of the cells originating from the lateral borders of the primitive neural anlagen, the neural crest (NC) are considered. Among them, their multipotentiality, which together with their migratory properties, leads them to colonize the developing body and to participate in the development of many tissues and organs. The in vitro analysis of the developmental capacities of single NC cells (NCC) showed that they present several analogies with the hematopoietic cells whose differentiation involves the activity of stem cells endowed with different arrays of developmental potentialities. The permanence of such NC stem cells in the adult organism raises the problem of their role at that stage of life. The NC has appeared during evolution in the vertebrate phylum and is absent in their Protocordates ancestors. The major role of the NCC in the development of the vertebrate head points to a critical role for this structure in the remarkable diversification and radiation of this group of animals.
Collapse
Affiliation(s)
- Elisabeth Dupin
- INSERM, U968, Paris, F-75012, France; Sorbonne Universités, UPMC Univ Paris 06, UMR_S 968, Institut de la Vision, Paris, F-75012, France; CNRS, UMR_7210, Paris, F-75012, France
| | | |
Collapse
|