1
|
Nagasaka A, Bando Y, Toda-Fujii M, Onozawa G, Suzuki K, Miyata T, Amano O. Differences in palatal shelf epithelial stiffness between the lingual/nasal and buccal/oral surfaces during palatal shelf elevation in developing mice. Dev Dyn 2025. [PMID: 40408552 DOI: 10.1002/dvdy.70044] [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: 12/25/2024] [Revised: 04/04/2025] [Accepted: 05/03/2025] [Indexed: 05/25/2025] Open
Abstract
BACKGROUND During secondary palate formation, bilateral palatal shelves grow vertically to a horizontal position. This morphological change of the palatal shelves, defined as the palatal shelf elevation, occurs from embryonic day (E)-13.5 to E14 in mice. Palatal shelves show regional differences in elevation patterns along the anterior-posterior (AP) axis; however, the underlying mechanisms remain unclear. Material properties of the lingual/nasal and buccal/oral surfaces, especially stiffness, possibly contribute to different elevation patterns. RESULTS Indentation test using atomic force microscopy was performed to measure the stiffness at the epithelial surface of the palatal shelf. Measurement of palatal shelf stiffness along the AP axis before and after elevation revealed that the lingual/nasal surface was softer than the buccal/oral surface in the posterior region before elevation and that the palatal shelf was stiffer after elevation than before elevation. Moreover, the thickness of epithelial cells on the lingual/nasal side was lower than that on the buccal/oral side before elevation. CONCLUSION Overall, our results suggest that epithelial cell thickness affects epithelial surface stiffness, causing regional differences in elevation patterns.
Collapse
Affiliation(s)
- Arata Nagasaka
- Division of Histology, Meikai University School of Dentistry, Sakado, Japan
| | - Yasuhiko Bando
- Division of Histology, Meikai University School of Dentistry, Sakado, Japan
| | - Miyuki Toda-Fujii
- Division of Histology, Meikai University School of Dentistry, Sakado, Japan
| | - Go Onozawa
- Division of Histology, Meikai University School of Dentistry, Sakado, Japan
- Division of Oral and Maxillofacial Surgery, Meikai University School of Dentistry, Sakado, Japan
| | - Kaito Suzuki
- Division of Histology, Meikai University School of Dentistry, Sakado, Japan
- Division of Oral and Maxillofacial Surgery, Meikai University School of Dentistry, Sakado, Japan
| | - Takaki Miyata
- Department of Anatomy and Cell Biology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Osamu Amano
- Division of Histology, Meikai University School of Dentistry, Sakado, Japan
| |
Collapse
|
2
|
Moore Zajic EL, Zhao R, McKinney MC, Yi K, Wood C, Trainor PA. Cell extrusion drives neural crest cell delamination. Proc Natl Acad Sci U S A 2025; 122:e2416566122. [PMID: 40063802 PMCID: PMC11929498 DOI: 10.1073/pnas.2416566122] [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: 08/26/2024] [Accepted: 01/17/2025] [Indexed: 03/15/2025] Open
Abstract
Neural crest cells (NCC) comprise a heterogeneous population of cells with variable potency that contribute to nearly every tissue and organ throughout the body. Considered unique to vertebrates, NCC are transiently generated within the dorsolateral region of the neural plate or neural tube during neurulation. Their delamination and migration are crucial for embryo development as NCC differentiation is influenced by their final resting locations. Previous work in avian and aquatic species revealed that NCC delaminate via an epithelial-mesenchymal transition (EMT), which transforms these progenitor cells from static polarized epithelial cells into migratory mesenchymal cells with fluid front and back polarity. However, the cellular and molecular mechanisms facilitating NCC delamination in mammals are poorly understood. Through time-lapse imaging of NCC delamination in mouse embryos, we identified a subset of cells that exit the neuroepithelium as isolated round cells, which then halt for a short period prior to acquiring the mesenchymal migratory morphology classically associated with delaminating NCC. High-magnification imaging and protein localization analyses of the cytoskeleton, together with measurements of pressure and tension of delaminating NCC and neighboring neuroepithelial cells, revealed that round NCC are extruded from the neuroepithelium prior to completion of EMT. Furthermore, cranial NCC are extruded through activation of the mechanosensitive ion channel, PIEZO1. Our results support a model in which cell density, pressure, and tension in the neuroepithelium result in activation of the live cell extrusion pathway and delamination of a subpopulation of NCC in parallel with EMT, which has implications for cell delamination in development and disease.
Collapse
Affiliation(s)
| | - Ruonan Zhao
- Stowers Institute for Medical Research, Kansas City, MO64110
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS66160
| | | | - Kexi Yi
- Stowers Institute for Medical Research, Kansas City, MO64110
| | | | - Paul A. Trainor
- Stowers Institute for Medical Research, Kansas City, MO64110
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS66160
| |
Collapse
|
3
|
Adelizzi E, Rhea L, Mitvalsky C, Pek S, Doolittle B, Dunnwald M. The ectodermal loss of ARHGAP29 alters epithelial morphology and organization and disrupts murine palatal development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.11.642653. [PMID: 40161602 PMCID: PMC11952475 DOI: 10.1101/2025.03.11.642653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Orofacial clefts, including cleft palate (CP), are among the most common types of birth defects. CP specifically, results from a failure of palatal shelf fusion during development. Previous studies have shown that mutations in RhoA GTPase Activating Protein 29 ( ARHGAP29) are linked to CP, yet the role and tissue-specific requirements for ARHGAP29 during palatogenesis remain unknown. Here, we use tissue-specific deletion of Arhgap29 in mice to provide the first direct evidence that ARHGAP29 is essential for proper palatal elevation and fusion. We demonstrate that ectodermal conditional loss of Arhgap29 induces a significant delay in the fusion of palatal shelves at embryonic (E) day 14.5 and an incomplete yet significantly penetrant cleft palate at E18.5 - neither of which are observed when Arhgap29 is lost later in development using K14-Cre. Phenotypic analyses of palatal shelves at E14.5 reveal a disorganized and thicker epithelium at the tip of the shelves. Loss of Arhgap29 increases palate epithelial cell area and upregulates alpha-smooth muscle actin and phospho-myosin regulatory light chain implicating cell morphology and contractility as drivers of CP. Summary statement This study in mice is the first direct evidence that ARHGAP29 is essential for proper palatal elevation and fusion. Loss of Arhgap29 alters oral epithelial morphology and upregulates contractility proteins.
Collapse
|
4
|
Im H, Song Y, Kim JK, Park DK, Kim DS, Kim H, Shin JO. Molecular Regulation of Palatogenesis and Clefting: An Integrative Analysis of Genetic, Epigenetic Networks, and Environmental Interactions. Int J Mol Sci 2025; 26:1382. [PMID: 39941150 PMCID: PMC11818578 DOI: 10.3390/ijms26031382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Revised: 02/03/2025] [Accepted: 02/04/2025] [Indexed: 02/16/2025] Open
Abstract
Palatogenesis is a complex developmental process requiring temporospatially coordinated cellular and molecular events. The following review focuses on genetic, epigenetic, and environmental aspects directing palatal formation and their implication in orofacial clefting genesis. Essential for palatal shelf development and elevation (TGF-β, BMP, FGF, and WNT), the subsequent processes of fusion (SHH) and proliferation, migration, differentiation, and apoptosis of neural crest-derived cells are controlled through signaling pathways. Interruptions to these processes may result in the birth defect cleft lip and/or palate (CL/P), which happens in approximately 1 in every 700 live births worldwide. Recent progress has emphasized epigenetic regulations via the class of non-coding RNAs with microRNAs based on critically important biological processes, such as proliferation, apoptosis, and epithelial-mesenchymal transition. These environmental risks (maternal smoking, alcohol, retinoic acid, and folate deficiency) interact with genetic and epigenetic factors during palatogenesis, while teratogens like dexamethasone and TCDD inhibit palatal fusion. In orofacial cleft, genetic, epigenetic, and environmental impact on the complex epidemiology. This is an extensive review, offering current perspectives on gene-environment interactions, as well as non-coding RNAs, in palatogenesis and emphasizing open questions regarding these interactions in palatal development.
Collapse
Affiliation(s)
- Hyuna Im
- Department of Anatomy, College of Medicine, Soonchunhyang University, Cheonan 33151, Republic of Korea (D.-K.P.); (D.-S.K.)
| | - Yujeong Song
- Department of Anatomy, College of Medicine, Soonchunhyang University, Cheonan 33151, Republic of Korea (D.-K.P.); (D.-S.K.)
| | - Jae Kyeom Kim
- Department of Food and Biotechnology, Korea University, Sejong 339770, Republic of Korea
- Department of Health Behavior and Nutrition Sciences, University of Delaware, Newark, DE 19711, USA
| | - Dae-Kyoon Park
- Department of Anatomy, College of Medicine, Soonchunhyang University, Cheonan 33151, Republic of Korea (D.-K.P.); (D.-S.K.)
| | - Duk-Soo Kim
- Department of Anatomy, College of Medicine, Soonchunhyang University, Cheonan 33151, Republic of Korea (D.-K.P.); (D.-S.K.)
| | - Hankyu Kim
- Department of Anatomy, College of Medicine, Soonchunhyang University, Cheonan 33151, Republic of Korea (D.-K.P.); (D.-S.K.)
| | - Jeong-Oh Shin
- Department of Anatomy, College of Medicine, Soonchunhyang University, Cheonan 33151, Republic of Korea (D.-K.P.); (D.-S.K.)
| |
Collapse
|
5
|
Tsujimoto T, Ou Y, Suzuki M, Murata Y, Inubushi T, Nagata M, Ishihara Y, Yonei A, Miyashita Y, Asano Y, Sakai N, Sakata Y, Ogino H, Yamashiro T, Kurosaka H. Compromised actin dynamics underlie the orofacial cleft in Baraitser-Winter Cerebrofrontofacial syndrome with a variant in ACTB. Hum Mol Genet 2024; 33:1975-1985. [PMID: 39271101 DOI: 10.1093/hmg/ddae133] [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: 04/15/2024] [Revised: 08/08/2024] [Accepted: 08/29/2024] [Indexed: 09/15/2024] Open
Abstract
Craniofacial anomalies encompassing the orofacial cleft are associated with > 30% of systemic congenital malformations. Baraitser-Winter Cerebrofrontofacial syndrome (BWCFF) is a rare genetic disorder attributed to variants in the actin beta (ACTB) or actin gamma genes that are correlated with a range of craniofacial abnormalities, including cleft lip and/or palate. The underlying pathological mechanism of BWCFF remains elusive, and it is necessary to investigate the etiology of orofacial clefts in patients with BWCFF. In this study, we identified a missense variant (c.1043C > T: p.S348L) in the ACTB gene of a patient with BWCFF and concomitant cleft lip and palate. Furthermore, we performed functional assessments of this variant using various disease models such as the MDCK cell line and Xenopus laevis. These models revealed a compromised capacity of mutated ACTB to localize to the epithelial junction, consequently affecting the behavior of epithelial cells. Additionally, we discovered that the mutated ACTB exhibited an impaired ability to bind PROFILIN1, a critical factor in actin polymerization. This defective ability may contribute to the molecular etiology of aberrant epithelial cell adhesion and migration, resulting in orofacial cleft formation in BWCFF.
Collapse
Affiliation(s)
- Takayuki Tsujimoto
- Department of Orthodontics and Dentofacial Orthopedics, Osaka University Graduate School of Dentistry, 1-8 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Yushi Ou
- Amphibian Research Center, Graduate School of Integrated Sciences for Life, Hiroshima University, 1-3-1 Kagamiyama, Higashi-hiroshima, Hiroshima 739-8526, Japan
| | - Makoto Suzuki
- Amphibian Research Center, Graduate School of Integrated Sciences for Life, Hiroshima University, 1-3-1 Kagamiyama, Higashi-hiroshima, Hiroshima 739-8526, Japan
| | - Yuka Murata
- Department of Orthodontics and Dentofacial Orthopedics, Osaka University Graduate School of Dentistry, 1-8 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Toshihiro Inubushi
- Department of Orthodontics and Dentofacial Orthopedics, Osaka University Graduate School of Dentistry, 1-8 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Miho Nagata
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Yasuki Ishihara
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Ayumi Yonei
- Department of Genetic Counseling, Osaka University Hospital, 2-15 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Yohei Miyashita
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Yoshihiro Asano
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Norio Sakai
- Department of Genetic Counseling, Osaka University Hospital, 2-15 Yamadaoka, Suita, Osaka 565-0871, Japan
- Department of Pediatrics, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
- Department of Health Science, Child Healthcare and Genetic Science, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Yasushi Sakata
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Hajime Ogino
- Amphibian Research Center, Graduate School of Integrated Sciences for Life, Hiroshima University, 1-3-1 Kagamiyama, Higashi-hiroshima, Hiroshima 739-8526, Japan
| | - Takashi Yamashiro
- Department of Orthodontics and Dentofacial Orthopedics, Osaka University Graduate School of Dentistry, 1-8 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Hiroshi Kurosaka
- Department of Orthodontics and Dentofacial Orthopedics, Osaka University Graduate School of Dentistry, 1-8 Yamadaoka, Suita, Osaka 565-0871, Japan
| |
Collapse
|
6
|
Siewert A, Hoeland S, Mangold E, Ludwig KU. Combining genetic and single-cell expression data reveals cell types and novel candidate genes for orofacial clefting. Sci Rep 2024; 14:26492. [PMID: 39489835 PMCID: PMC11532359 DOI: 10.1038/s41598-024-77724-9] [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: 08/22/2024] [Accepted: 10/24/2024] [Indexed: 11/05/2024] Open
Abstract
Non-syndromic cleft lip with/without cleft palate (nsCL/P) is one of the most common birth defects and has a multifactorial etiology. To date, over 45 loci harboring common risk variants have been identified. However, the effector genes at these loci, and the cell types that are affected by risk alleles, remain largely unknown. To address this, we combined genetic data from an nsCL/P genome-wide association study (GWAS) with single-cell RNA sequencing data obtained from the heads of unaffected human embryos. Using the recently developed single-cell disease relevance score (scDRS) approach, we identified two major cell types involved in nsCL/P development, namely the epithelium and the HAND2+ pharyngeal arches (PA). Combining scDRS with co-expression networks and differential gene expression analysis, we prioritized nsCL/P candidate genes, some of which were additionally supported by GWAS data (e.g., CTNND1, PRTG, RPL35A, RAB11FIP1, KRT19). Our results suggest that specific epithelial and PA sub-cell types are involved in nsCL/P development, and harbor a substantial fraction of the genetic risk for nsCL/P.
Collapse
Affiliation(s)
- Anna Siewert
- Institute of Human Genetics, School of Medicine & University Hospital Bonn, University of Bonn, Bonn, Germany.
| | - Simone Hoeland
- Institute of Human Genetics, School of Medicine & University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Elisabeth Mangold
- Institute of Human Genetics, School of Medicine & University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Kerstin U Ludwig
- Institute of Human Genetics, School of Medicine & University Hospital Bonn, University of Bonn, Bonn, Germany.
| |
Collapse
|
7
|
Bhaskar A, Astrof S. Identification of novel genes regulating the development of the palate. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.09.579685. [PMID: 38405938 PMCID: PMC10888939 DOI: 10.1101/2024.02.09.579685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
The International Mouse Phenotyping Consortium (IMPC) has generated thousands of knockout mouse lines, many of which exhibit embryonic or perinatal lethality. Using micro-computed tomography (micro-CT), the IMPC has created and publicly released 3D image datasets of embryos from these lethal and subviable lines. In this study, we leveraged this dataset to screen homozygous null mutants for anomalies in secondary palate development. We analyzed optical sections from 2,987 embryos at embryonic days E15.5 and E18.5, representing 484 homozygous mutant lines. Our analysis identified 45 novel genes implicated in palatogenesis. Gene set enrichment analysis highlighted biological processes and pathways relevant to palate development and uncovered 18 genes jointly regulating the development of the eye and the palate. These findings present a valuable resource for further research, offer novel insights into the molecular mechanisms underlying palatogenesis, and provide important context for understanding the etiology of rare human congenital disorders involving simultaneous malformations of the palate and other organs, including the eyes, ears, kidneys, and lungs.
Collapse
Affiliation(s)
- Ashwin Bhaskar
- Rutgers University, School of Arts and Sciences Honors Program, New Brunswick, NJ, 08901, USA
| | - Sophie Astrof
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers Biomedical and Health Sciences, 185 South Orange Ave, Newark, NJ, 07103, USA
| |
Collapse
|
8
|
Stanton E, Sheridan S, Urata M, Chai Y. From Bedside to Bench and Back: Advancing Our Understanding of the Pathophysiology of Cleft Palate and Implications for the Future. Cleft Palate Craniofac J 2024; 61:759-773. [PMID: 36457208 DOI: 10.1177/10556656221142098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2024] Open
Abstract
OBJECTIVE To provide a comprehensive understanding of the pathophysiology of cleft palate (CP) and future perspectives. DESIGN Literature review. SETTING Setting varied across studies by level of care and geographical locations. INTERVENTIONS No interventions were performed. MAIN OUTCOME MEASURE(S) Primary outcome measures were to summarize our current understanding of palatogenesis in humans and animal models, the pathophysiology of CP, and potential future treatment modalities. RESULTS Animal research has provided considerable insight into the pathophysiology, molecular and cellular mechanisms of CP that have allowed for the development of novel treatment strategies. However, much work has yet to be done to connect our mouse model investigations and discoveries to CP in humans. The success of innovative strategies for tissue regeneration in mice provides promise for an exciting new avenue for improved and more targeted management of cleft care with precision medicine in patients. However, significant barriers to clinical translation remain. Among the most notable challenges include the differences in some aspects of palatogenesis and tissue repair between mice and humans, suggesting that potential therapies that have worked in animal models may not provide similar benefits to humans. CONCLUSIONS Increased translation of pathophysiological and tissue regeneration studies to clinical trials will bridge a wide gap in knowledge between animal models and human disease. By enhancing interaction between basic scientists and clinicians, and employing our animal model findings of disease mechanisms in concert with what we glean in the clinic, we can generate a more targeted and improved treatment algorithm for patients with CP.
Collapse
Affiliation(s)
- Eloise Stanton
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA, USA
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Samuel Sheridan
- Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, USA
| | - Mark Urata
- Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, USA
- Division of Plastic and Maxillofacial Surgery, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Yang Chai
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA, USA
- Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
9
|
Zhang Z, Zhang Q, Liu Z, Wang C, Chen H, Luo X, Shen L, Long C, Wei G, Liu X. Rab25 is involved in hypospadias via the β1 integrin/EGFR pathway. Exp Cell Res 2024; 436:113980. [PMID: 38401686 DOI: 10.1016/j.yexcr.2024.113980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 02/18/2024] [Accepted: 02/19/2024] [Indexed: 02/26/2024]
Abstract
BACKGROUND Hypospadias is a common congenital abnormality of the penile. Abnormal regulation of critical genes involved in urethral development leads to hypospadias. We used the Rab25-/- mice and foreskin fibroblasts transfected with lentivirus in vitro and in vivo to investigate the role of Rab25 in hypospadias. METHODS The expression levels of various molecules in tissue samples and foreskin fibroblasts were confirmed using molecular biology methods (western blotting, PCR, immunohistochemistry, etc.). A scanning electron microscope (SEM) was used to visualize the external morphology of genital tubercles (GTs) of gestation day (GD) 18.5 male wild-type (WT) and Rab25-/- mice. RESULTS An expanded distal cleft and V-shaped urethral opening were observed in GD 18.5 Rab25-/- mice. We demonstrated that Rab25 mediated hypospadias through the β1 integrin/EGFR pathway. In addition, silencing Rab25 inhibited cell proliferation and migration and promoted apoptosis in the foreskin fibroblasts; Ki-67- and TUNEL-positive cells were mainly concentrated near the urethral seam. CONCLUSION These findings suggest that Rab25 plays an essential role in hypospadias by activation of β1 integrin/EGFR pathway, and Rab25 is a critical mediator of urethral seam formation in GD18.5 male fetal mice.
Collapse
Affiliation(s)
- Zhicheng Zhang
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China; Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China; Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China
| | - Qiang Zhang
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China; Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China; Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China
| | - Zhenmin Liu
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China; Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China; Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China
| | - Chong Wang
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China; Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China; Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China
| | - Hongsong Chen
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China; Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China; Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China
| | - Xingguo Luo
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China; Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China; Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China
| | - Lianju Shen
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China; Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China; Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China
| | - Chunlan Long
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China; Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China; Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China
| | - Guanghui Wei
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China; Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China; Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China
| | - Xing Liu
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China; Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China; Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China; Program for Youth Innovation in Future Medicine, Chongqing Medical University, Chongqing, 400014, PR China.
| |
Collapse
|
10
|
Moore E, Zhao R, McKinney MC, Yi K, Wood C, Trainor P. Cell extrusion - a novel mechanism driving neural crest cell delamination. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.09.584232. [PMID: 38559094 PMCID: PMC10979875 DOI: 10.1101/2024.03.09.584232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Neural crest cells (NCC) comprise a heterogeneous population of cells with variable potency, that contribute to nearly every tissue and organ system throughout the body. Considered unique to vertebrates, NCC are transiently generated within the dorsolateral region of the neural plate or neural tube, during neurulation. Their delamination and migration are crucial events in embryo development as the differentiation of NCC is heavily influenced by their final resting locations. Previous work in avian and aquatic species has shown that NCC delaminate via an epithelial-mesenchymal transition (EMT), which transforms these stem and progenitor cells from static polarized epithelial cells into migratory mesenchymal cells with fluid front and back polarity. However, the cellular and molecular drivers facilitating NCC delamination in mammals are poorly understood. We performed live timelapse imaging of NCC delamination in mouse embryos and discovered a group of cells that exit the neuroepithelium as isolated round cells, which then halt for a short period prior to acquiring the mesenchymal migratory morphology classically associated with most delaminating NCC. High magnification imaging and protein localization analyses of the cytoskeleton, together with measurements of pressure and tension of delaminating NCC and neighboring neuroepithelial cells, revealed these round NCC are extruded from the neuroepithelium prior to completion of EMT. Furthermore, we demonstrate that cranial NCC are extruded through activation of the mechanosensitive ion channel, PIEZO1, a key regulator of the live cell extrusion pathway, revealing a new role for PIEZO1 in neural crest cell development. Our results elucidating the cellular and molecular dynamics orchestrating NCC delamination support a model in which high pressure and tension in the neuroepithelium results in activation of the live cell extrusion pathway and delamination of a subpopulation of NCC in parallel with EMT. This model has broad implications for our understanding of cell delamination in development and disease.
Collapse
Affiliation(s)
- Emma Moore
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Ruonan Zhao
- Stowers Institute for Medical Research, Kansas City, MO, USA
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Mary C McKinney
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Kexi Yi
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | | | - Paul Trainor
- Stowers Institute for Medical Research, Kansas City, MO, USA
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
11
|
Sun B, Reynolds K, Saha SK, Zhang S, McMahon M, Zhou CJ. Ezh2-dependent methylation in oral epithelia promotes secondary palatogenesis. Birth Defects Res 2023; 115:1851-1865. [PMID: 37435868 PMCID: PMC10784412 DOI: 10.1002/bdr2.2216] [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: 03/31/2023] [Revised: 05/30/2023] [Accepted: 06/12/2023] [Indexed: 07/13/2023]
Abstract
BACKGROUND In addition to genomic risk variants and environmental influences, increasing evidence suggests epigenetic modifications are important for orofacial development and their alterations can contribute to orofacial clefts. Ezh2 encodes a core catalytic component of the Polycomb repressive complex responsible for addition of methyl marks to Histone H3 as a mechanism of repressing target genes. The role of Ezh2 in orofacial clefts remains unknown. AIMS To investigate the epithelial role of Ezh2-dependent methylation in secondary palatogenesis. METHODS We used conditional gene-targeting methods to ablate Ezh2 in the surface ectoderm-derived oral epithelium of mouse embryos. We then performed single-cell RNA sequencing combined with immunofluorescence and RT-qPCR to investigate gene expression in conditional mutant palate. We also employed double knockout analyses of Ezh1 and Ezh2 to address if they have synergistic roles in palatogenesis. RESULTS We found that conditional inactivation of Ezh2 in oral epithelia results in partially penetrant cleft palate. Double knockout analyses revealed that another family member Ezh1 is dispensable in orofacial development, and it does not have synergistic roles with Ezh2 in palatogenesis. Histochemistry and single-cell RNA-seq analyses revealed dysregulation of cell cycle regulators in the palatal epithelia of Ezh2 mutant mouse embryos disrupts palatogenesis. CONCLUSION Ezh2-dependent histone H3K27 methylation represses expression of cell cycle regulator Cdkn1a and promotes proliferation in the epithelium of the developing palatal shelves. Loss of this regulation may perturb movement of the palatal shelves, causing a delay in palate elevation which may result in failure of the secondary palate to close altogether.
Collapse
Affiliation(s)
| | | | - Subbroto Kuma Saha
- Institute for Pediatric Regenerative Medicine of Shriners Hospital for Children – Northern California & Department of Biochemistry and Molecular Medicine, School of Medicine, University of California at Davis, Sacramento, CA 95817, USA
| | - Shuwen Zhang
- Institute for Pediatric Regenerative Medicine of Shriners Hospital for Children – Northern California & Department of Biochemistry and Molecular Medicine, School of Medicine, University of California at Davis, Sacramento, CA 95817, USA
| | - Moira McMahon
- Institute for Pediatric Regenerative Medicine of Shriners Hospital for Children – Northern California & Department of Biochemistry and Molecular Medicine, School of Medicine, University of California at Davis, Sacramento, CA 95817, USA
| | - Chengji J Zhou
- Institute for Pediatric Regenerative Medicine of Shriners Hospital for Children – Northern California & Department of Biochemistry and Molecular Medicine, School of Medicine, University of California at Davis, Sacramento, CA 95817, USA
| |
Collapse
|
12
|
Wolf CJ, Fitzpatrick H, Becker C, Smith J, Wood C. An improved multicellular human organoid model for the study of chemical effects on palatal fusion. Birth Defects Res 2023; 115:1513-1533. [PMID: 37530699 PMCID: PMC11253831 DOI: 10.1002/bdr2.2229] [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: 04/07/2023] [Revised: 07/11/2023] [Accepted: 07/19/2023] [Indexed: 08/03/2023]
Abstract
BACKGROUND Tissue fusion is a mechanism involved in the development of the heart, iris, genital tubercle, neural tube, and palate during embryogenesis. Failed fusion of the palatal shelves could result in cleft palate (CP), a common birth defect. Organotypic models constructed of human cells offer an opportunity to investigate developmental processes in the human. Previously, our laboratory developed an organoid model of the human palate that contains human mesenchyme and epithelial progenitor cells to study the effects of chemicals on fusion. METHODS Here, we developed an organoid model more representative of the embryonic palate that includes three cell types: mesenchyme, endothelial, and epithelial cells. We measured fusion by a decrease in epithelial cells at the contact point between the organoids and compared the effects of CP teratogens on fusion and toxicity in the previous and current organoid models. We further tested additional suspect teratogens in our new model. RESULTS We found that the three-cell-type model is more sensitive to fusion inhibition by valproic acid and inhibitors of FGF, BMP, and TGFβRI/II. In this new model, we tested other suspect CP teratogens and found that nocodazole, topiramate, and Y27632 inhibit fusion at concentrations that do not induce toxicity. CONCLUSION This sensitive human three-cell-type organotypic model accurately evaluates chemicals for cleft palate teratogenicity.
Collapse
Affiliation(s)
- Cynthia J Wolf
- Center for Public Health and Environmental Assessment, Office of Research and Development, US Environmental Protection Agency, Research Triangle Park, North Carolina, USA
| | - Hunter Fitzpatrick
- Oak Ridge Institute for Science and Education, Oak Ridge, Tennessee, USA
| | - Carrie Becker
- Oak Ridge Institute for Science and Education, Oak Ridge, Tennessee, USA
| | - Jessica Smith
- Oak Ridge Institute for Science and Education, Oak Ridge, Tennessee, USA
| | - Carmen Wood
- Center for Public Health and Environmental Assessment, Office of Research and Development, US Environmental Protection Agency, Research Triangle Park, North Carolina, USA
| |
Collapse
|
13
|
Saroya GA, Siismets E, Hu M, Panaretos C, Rice A, Reynolds K, Zhou CJ, Kaartinen V. Canonical Wnt signaling is not required for Tgfb3 expression in the basal medial edge epithelium during palatogenesis. Front Physiol 2023; 14:704406. [PMID: 37250135 PMCID: PMC10213314 DOI: 10.3389/fphys.2023.704406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Accepted: 04/28/2023] [Indexed: 05/31/2023] Open
Abstract
The secondary palate forms from two lateral primordia called the palatal shelves which form a contact in the midline, become adherent at the fusing interface (medial edge epithelia, MEE) and subsequently fuse. The gene encoding transforming growth factor-ß3 (Tgfb3) is strongly and specifically expressed in MEE cells. Our previous study suggested that Tgfb3 expression is controlled via upstream cis-regulatory elements in and around the neighboring Ift43 gene. Another study suggested that the canonical Wnt signaling via ß-Catenin is responsible for the MEE-specific Tgfb3 gene expression, since deletion of the Ctnnb1 gene by a commonly used Keratin 14-Cre (K14Cre) mouse line almost completely abolished Tgfb3 expression in the MEE resulting in cleft palate. Here, we wanted to analyze whether Tcf/Lef consensus binding sites located in the previously identified regions of the Ift43 gene are responsible for the spatiotemporal control of Tgfb3 expression during palatogenesis. We show that contrary to the previous report, deletion of the Ctnnb1 gene in basal MEE cells by the K14Cre driver (the same K14Cre mouse line was used as in the previous study referenced above) does not affect the MEE-specific Tgfb3 expression or TGFß3-dependent palatal epithelial fusion. All mutant embryos showed a lack of palatal rugae accompanied by other craniofacial defects, e.g., a narrow snout and a small upper lip, while only a small subset (<5%) of Ctnnb1 mutants displayed a cleft palate. Moreover, the K14Cre:Ctnnb1 embryos showed reduced levels and altered patterns of Shh expression. Our present data imply that epithelial ß-catenin may not be required for MEE-specific Tgfb3 expression or palatal epithelial fusion.
Collapse
Affiliation(s)
- Ghazi-Abdullah Saroya
- Department of Biologic and Materials Sciences, University of Michigan School of Dentistry, Ann Arbor, MI, United States
| | - Erica Siismets
- Department of Biologic and Materials Sciences, University of Michigan School of Dentistry, Ann Arbor, MI, United States
- Oral Health Sciences PhD Program, University of Michigan School of Dentistry, Ann Arbor, MI, United States
| | - Max Hu
- Department of Biologic and Materials Sciences, University of Michigan School of Dentistry, Ann Arbor, MI, United States
- College of Literature, Sciences and the Arts, University of Michigan, Ann Arbor, MI, United States
| | - Christopher Panaretos
- Department of Biologic and Materials Sciences, University of Michigan School of Dentistry, Ann Arbor, MI, United States
| | - Adam Rice
- Department of Biologic and Materials Sciences, University of Michigan School of Dentistry, Ann Arbor, MI, United States
| | - Kurt Reynolds
- School of Medicine, Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children-Northern California, University of California at Davis, Sacramento, CA, United States
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California at Davis, Sacramento, CA, United States
| | - Chengji J. Zhou
- School of Medicine, Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children-Northern California, University of California at Davis, Sacramento, CA, United States
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California at Davis, Sacramento, CA, United States
| | - Vesa Kaartinen
- Department of Biologic and Materials Sciences, University of Michigan School of Dentistry, Ann Arbor, MI, United States
| |
Collapse
|
14
|
Li Q, Chen J, Faux P, Delgado ME, Bonfante B, Fuentes-Guajardo M, Mendoza-Revilla J, Chacón-Duque JC, Hurtado M, Villegas V, Granja V, Jaramillo C, Arias W, Barquera R, Everardo-Martínez P, Sánchez-Quinto M, Gómez-Valdés J, Villamil-Ramírez H, Silva de Cerqueira CC, Hünemeier T, Ramallo V, Wu S, Du S, Giardina A, Paria SS, Khokan MR, Gonzalez-José R, Schüler-Faccini L, Bortolini MC, Acuña-Alonzo V, Canizales-Quinteros S, Gallo C, Poletti G, Rojas W, Rothhammer F, Navarro N, Wang S, Adhikari K, Ruiz-Linares A. Automatic landmarking identifies new loci associated with face morphology and implicates Neanderthal introgression in human nasal shape. Commun Biol 2023; 6:481. [PMID: 37156940 PMCID: PMC10167347 DOI: 10.1038/s42003-023-04838-7] [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: 03/22/2022] [Accepted: 04/12/2023] [Indexed: 05/10/2023] Open
Abstract
We report a genome-wide association study of facial features in >6000 Latin Americans based on automatic landmarking of 2D portraits and testing for association with inter-landmark distances. We detected significant associations (P-value <5 × 10-8) at 42 genome regions, nine of which have been previously reported. In follow-up analyses, 26 of the 33 novel regions replicate in East Asians, Europeans, or Africans, and one mouse homologous region influences craniofacial morphology in mice. The novel region in 1q32.3 shows introgression from Neanderthals and we find that the introgressed tract increases nasal height (consistent with the differentiation between Neanderthals and modern humans). Novel regions include candidate genes and genome regulatory elements previously implicated in craniofacial development, and show preferential transcription in cranial neural crest cells. The automated approach used here should simplify the collection of large study samples from across the world, facilitating a cosmopolitan characterization of the genetics of facial features.
Collapse
Affiliation(s)
- Qing Li
- Ministry of Education Key Laboratory of Contemporary Anthropology and Collaborative Innovation Center of Genetics and Development, School of Life Sciences and Human Phenome Institute, Fudan University, Yangpu District, Shanghai, 200438, China
| | - Jieyi Chen
- Ministry of Education Key Laboratory of Contemporary Anthropology and Collaborative Innovation Center of Genetics and Development, School of Life Sciences and Human Phenome Institute, Fudan University, Yangpu District, Shanghai, 200438, China
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai, 200031, China
| | - Pierre Faux
- Aix-Marseille Université, CNRS, EFS, ADES, Marseille, 13005, France
| | - Miguel Eduardo Delgado
- Ministry of Education Key Laboratory of Contemporary Anthropology and Collaborative Innovation Center of Genetics and Development, School of Life Sciences and Human Phenome Institute, Fudan University, Yangpu District, Shanghai, 200438, China
- División Antropología, Facultad de Ciencias Naturales y Museo, Universidad Nacional de La Plata, La Plata, República Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET, Buenos Aires, República Argentina
| | - Betty Bonfante
- Aix-Marseille Université, CNRS, EFS, ADES, Marseille, 13005, France
| | - Macarena Fuentes-Guajardo
- Departamento de Tecnología Médica, Facultad de Ciencias de la Salud, Universidad de Tarapacá, Arica, 1000000, Chile
| | - Javier Mendoza-Revilla
- Laboratorios de Investigación y Desarrollo, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Lima, 31, Perú
- Unit of Human Evolutionary Genetics, Institut Pasteur, Paris, 75015, France
| | - J Camilo Chacón-Duque
- Division of Vertebrates and Anthropology, Department of Earth Sciences, Natural History Museum, London, SW7 5BD, UK
| | - Malena Hurtado
- Laboratorios de Investigación y Desarrollo, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Lima, 31, Perú
| | - Valeria Villegas
- Laboratorios de Investigación y Desarrollo, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Lima, 31, Perú
| | - Vanessa Granja
- Laboratorios de Investigación y Desarrollo, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Lima, 31, Perú
| | - Claudia Jaramillo
- GENMOL (Genética Molecular), Universidad de Antioquia, Medellín, 5001000, Colombia
| | - William Arias
- GENMOL (Genética Molecular), Universidad de Antioquia, Medellín, 5001000, Colombia
| | - Rodrigo Barquera
- Molecular Genetics Laboratory, National School of Anthropology and History, Mexico City, 14050, Mexico, 6600, Mexico
- Department of Archaeogenetics, Max Planck Institute for the Science of Human History (MPI-SHH), Jena, 07745, Germany
| | - Paola Everardo-Martínez
- Molecular Genetics Laboratory, National School of Anthropology and History, Mexico City, 14050, Mexico, 6600, Mexico
| | - Mirsha Sánchez-Quinto
- Forensic Science, Faculty of Medicine, UNAM (Universidad Nacional Autónoma de México), Mexico City, 06320, Mexico
| | - Jorge Gómez-Valdés
- Molecular Genetics Laboratory, National School of Anthropology and History, Mexico City, 14050, Mexico, 6600, Mexico
| | - Hugo Villamil-Ramírez
- Unidad de Genomica de Poblaciones Aplicada a la Salud, Facultad de Química, UNAM-Instituto Nacional de Medicina Genómica, Mexico City, 4510, Mexico
| | | | - Tábita Hünemeier
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, SP, 05508-090, Brazil
| | - Virginia Ramallo
- Departamento de Genética, Universidade Federal do Rio Grande do Sul, Porto Alegre, 90040-060, Brazil
- Instituto Patagónico de Ciencias Sociales y Humanas, Centro Nacional Patagónico, CONICET, Puerto Madryn, U9129ACD, Argentina
| | - Sijie Wu
- Ministry of Education Key Laboratory of Contemporary Anthropology and Collaborative Innovation Center of Genetics and Development, School of Life Sciences and Human Phenome Institute, Fudan University, Yangpu District, Shanghai, 200438, China
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai, 200031, China
| | - Siyuan Du
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai, 200031, China
| | - Andrea Giardina
- School of Mathematics and Statistics, Faculty of Science, Technology, Engineering and Mathematics, The Open University, Milton Keynes, MK7 6AA, United Kingdom
| | - Soumya Subhra Paria
- School of Mathematics and Statistics, Faculty of Science, Technology, Engineering and Mathematics, The Open University, Milton Keynes, MK7 6AA, United Kingdom
| | - Mahfuzur Rahman Khokan
- School of Mathematics and Statistics, Faculty of Science, Technology, Engineering and Mathematics, The Open University, Milton Keynes, MK7 6AA, United Kingdom
| | - Rolando Gonzalez-José
- Instituto Patagónico de Ciencias Sociales y Humanas, Centro Nacional Patagónico, CONICET, Puerto Madryn, U9129ACD, Argentina
| | - Lavinia Schüler-Faccini
- Departamento de Genética, Universidade Federal do Rio Grande do Sul, Porto Alegre, 90040-060, Brazil
| | - Maria-Cátira Bortolini
- Departamento de Genética, Universidade Federal do Rio Grande do Sul, Porto Alegre, 90040-060, Brazil
| | - Victor Acuña-Alonzo
- Molecular Genetics Laboratory, National School of Anthropology and History, Mexico City, 14050, Mexico, 6600, Mexico
| | - Samuel Canizales-Quinteros
- Unidad de Genomica de Poblaciones Aplicada a la Salud, Facultad de Química, UNAM-Instituto Nacional de Medicina Genómica, Mexico City, 4510, Mexico
| | - Carla Gallo
- Laboratorios de Investigación y Desarrollo, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Lima, 31, Perú
| | - Giovanni Poletti
- Laboratorios de Investigación y Desarrollo, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Lima, 31, Perú
| | - Winston Rojas
- GENMOL (Genética Molecular), Universidad de Antioquia, Medellín, 5001000, Colombia
| | - Francisco Rothhammer
- Instituto de Alta Investigación, Universidad de Tarapacá, Arica, Arica, 1000000, Chile
| | - Nicolas Navarro
- Biogéosciences, UMR 6282 CNRS, Université de Bourgogne, Dijon, 21000, France
- EPHE, PSL University, Paris, 75014, France
| | - Sijia Wang
- Ministry of Education Key Laboratory of Contemporary Anthropology and Collaborative Innovation Center of Genetics and Development, School of Life Sciences and Human Phenome Institute, Fudan University, Yangpu District, Shanghai, 200438, China
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai, 200031, China
| | - Kaustubh Adhikari
- School of Mathematics and Statistics, Faculty of Science, Technology, Engineering and Mathematics, The Open University, Milton Keynes, MK7 6AA, United Kingdom.
- Department of Genetics, Evolution and Environment, and UCL Genetics Institute, University College London, London, WC1E 6BT, UK.
| | - Andrés Ruiz-Linares
- Ministry of Education Key Laboratory of Contemporary Anthropology and Collaborative Innovation Center of Genetics and Development, School of Life Sciences and Human Phenome Institute, Fudan University, Yangpu District, Shanghai, 200438, China.
- Aix-Marseille Université, CNRS, EFS, ADES, Marseille, 13005, France.
- Department of Genetics, Evolution and Environment, and UCL Genetics Institute, University College London, London, WC1E 6BT, UK.
| |
Collapse
|
15
|
Saroya G, Hu J, Hu M, Panaretos C, Mann J, Kim S, Bush J, Kaartinen V. Periderm Fate during Palatogenesis: TGF-β and Periderm Dedifferentiation. J Dent Res 2023; 102:459-466. [PMID: 36751050 PMCID: PMC10041600 DOI: 10.1177/00220345221146454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023] Open
Abstract
Failure of palatogenesis results in cleft palate, one of the most common congenital disabilities in humans. During the final phases of palatogenesis, the protective function of the peridermal cell layer must be eliminated for the medial edge epithelia to adhere properly, which is a prerequisite for the successful fusion of the secondary palate. However, a deeper understanding of the role and fate of the periderm in palatal adherence and fusion has been hampered due to a lack of appropriate periderm-specific genetic tools to examine this cell type in vivo. Here we used the cytokeratin-6A (Krt-6a) locus to develop both constitutive (Krt6ai-Cre) and inducible (Krt6ai-CreERT2) periderm-specific Cre driver mouse lines. These novel lines allowed us to achieve both the spatial and temporal control needed to dissect the periderm fate on a cellular resolution during palatogenesis. Our studies suggest that, already before the opposing palatal shelves contact each other, at least some palatal periderm cells start to gradually lose their squamous periderm-like phenotype and dedifferentiate into cuboidal cells, reminiscent of the basal epithelial cells seen in the palatal midline seam. Moreover, we show that transforming growth factor-β (TGF-β) signaling plays a critical periderm-specific role in palatogenesis. Thirty-three percent of embryos lacking a gene encoding the TGF-β type I receptor (Tgfbr1) in the periderm display a complete cleft of the secondary palate. Our subsequent experiments demonstrated that Tgfbr1-deficient periderm fails to undergo appropriate dedifferentiation. These studies define the periderm cell fate during palatogenesis and reveal a novel, critical role for TGF-β signaling in periderm dedifferentiation, which is a prerequisite for appropriate palatal epithelial adhesion and fusion.
Collapse
Affiliation(s)
- G. Saroya
- Department of Biologic and Materials Sciences, University of Michigan School of Dentistry, Ann Arbor, MI, USA
| | - J. Hu
- Department of Biologic and Materials Sciences, University of Michigan School of Dentistry, Ann Arbor, MI, USA
- College of Literature, Sciences, and the Arts, University of Michigan, Ann Arbor, MI, USA
| | - M. Hu
- Department of Biologic and Materials Sciences, University of Michigan School of Dentistry, Ann Arbor, MI, USA
- College of Literature, Sciences, and the Arts, University of Michigan, Ann Arbor, MI, USA
| | - C. Panaretos
- Department of Biologic and Materials Sciences, University of Michigan School of Dentistry, Ann Arbor, MI, USA
| | - J. Mann
- Department of Biologic and Materials Sciences, University of Michigan School of Dentistry, Ann Arbor, MI, USA
| | - S. Kim
- Department of Cell and Tissue Biology and Program in Craniofacial Biology, University of California San Francisco, San Francisco, CA, USA
| | - J.O. Bush
- Department of Cell and Tissue Biology and Program in Craniofacial Biology, University of California San Francisco, San Francisco, CA, USA
| | - V. Kaartinen
- Department of Biologic and Materials Sciences, University of Michigan School of Dentistry, Ann Arbor, MI, USA
| |
Collapse
|
16
|
Ultra-Rare Variants Identify Biological Pathways and Candidate Genes in the Pathobiology of Non-Syndromic Cleft Palate Only. Biomolecules 2023; 13:biom13020236. [PMID: 36830605 PMCID: PMC9953608 DOI: 10.3390/biom13020236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 01/20/2023] [Accepted: 01/24/2023] [Indexed: 01/28/2023] Open
Abstract
In recent decades, many efforts have been made to elucidate the genetic causes of non-syndromic cleft palate (nsCPO), a complex congenital disease caused by the interaction of several genetic and environmental factors. Since genome-wide association studies have evidenced a minor contribution of common polymorphisms in nsCPO inheritance, we used whole exome sequencing data to explore the role of ultra-rare variants in this study. In a cohort of 35 nsCPO cases and 38 controls, we performed a gene set enrichment analysis (GSEA) and a hypergeometric test for assessing significant overlap between genes implicated in nsCPO pathobiology and genes enriched in ultra-rare variants in our cohort. GSEA highlighted an enrichment of ultra-rare variants in genes principally belonging to cytoskeletal protein binding pathway (Probability Density Function corrected p-value = 1.57 × 10-4); protein-containing complex binding pathway (p-value = 1.06 × 10-2); cell adhesion molecule binding pathway (p-value = 1.24 × 10-2); ECM-receptor interaction pathway (p-value = 1.69 × 10-2); and in the Integrin signaling pathway (p-value = 1.28 × 10-2). Two genes implicated in nsCPO pathobiology, namely COL2A1 and GLI3, ranked among the genes (n = 34) with nominal enrichment in the ultra-rare variant collapsing analysis (Fisher's exact test p-value < 0.05). These genes were also part of an independent list of genes highly relevant to nsCPO biology (n = 25). Significant overlap between the two sets of genes (hypergeometric test p-value = 5.86 × 10-3) indicated that enriched genes are likely to be implicated in physiological palate development and/or the pathological processes of oral clefting. In conclusion, ultra-rare variants collectively impinge on biological pathways crucial to nsCPO pathobiology and point to candidate genes that may contribute to the individual risk of disease. Sequencing can be an effective approach to identify candidate genes and pathways for nsCPO.
Collapse
|
17
|
Mukhopadhyay P, Smolenkova I, Seelan RS, Pisano MM, Greene RM. Spatiotemporal Expression and Functional Analysis of miRNA-22 in the Developing Secondary Palate. Cleft Palate Craniofac J 2023; 60:27-38. [PMID: 34730446 DOI: 10.1177/10556656211054004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVE Normal development of the embryonic orofacial region requires precise spatiotemporal coordination between numerous genes. MicroRNAs represent small, single-stranded, non-coding molecules that regulate gene expression. This study examines the role of microRNA-22 (miR-22) in murine orofacial ontogeny. METHODS Spatiotemporal and differential expression of miR-22 (mmu-miR-22-3p) within the developing secondary palate was determined by in situ hybridization and quantitative real-time PCR, respectively. Bioinformatic approaches were used to predict potential mRNA targets of miR-22 and analyze their association with cellular functions indispensable for normal orofacial ontogeny. An in vitro palate organ culture system was used to assess the role of miR-22 in secondary palate development. RESULTS There was a progressive increase in miR-22 expression from GD12.5 to GD14.5 in palatal processes. On GD12.5 and GD13.5, miR-22 was expressed in the future oral, nasal, and medial edge epithelia. On GD14.5, miR-22 expression was observed in the residual midline epithelial seam (MES), the nasal epithelium and the mesenchyme, but not in the oral epithelium. Inhibition of miR-22 activity in palate organ cultures resulted in failure of MES removal. Bioinformatic analyses revealed potential mRNA targets of miR-22 that may play significant roles in regulating apoptosis, migration, and/or convergence/extrusion, developmental processes that modulate MES removal during palatogenesis. CONCLUSIONS Results from the current study suggest a key role for miR-22 in the removal of the MES during palatogenesis and that miR-22 may represent a potential contributor to the etiology of cleft palate.
Collapse
Affiliation(s)
- Partha Mukhopadhyay
- Department of Oral Immunology and Infectious Diseases, Division of Craniofacial Development and Anomalies, School of Dentistry, 5170University of Louisville, Louisville, KY 40202
| | - Irina Smolenkova
- Department of Oral Immunology and Infectious Diseases, Division of Craniofacial Development and Anomalies, School of Dentistry, 5170University of Louisville, Louisville, KY 40202
| | - Ratnam S Seelan
- Department of Oral Immunology and Infectious Diseases, Division of Craniofacial Development and Anomalies, School of Dentistry, 5170University of Louisville, Louisville, KY 40202
| | - M Michele Pisano
- Department of Oral Immunology and Infectious Diseases, Division of Craniofacial Development and Anomalies, School of Dentistry, 5170University of Louisville, Louisville, KY 40202
| | - Robert M Greene
- Department of Oral Immunology and Infectious Diseases, Division of Craniofacial Development and Anomalies, School of Dentistry, 5170University of Louisville, Louisville, KY 40202
| |
Collapse
|
18
|
Ramachandran J, Zhou W, Bardenhagen AE, Nasr T, Yates ER, Zorn AM, Ji H, Vokes SA. Hedgehog regulation of epithelial cell state and morphogenesis in the larynx. eLife 2022; 11:e77055. [PMID: 36398878 PMCID: PMC9718526 DOI: 10.7554/elife.77055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 11/18/2022] [Indexed: 11/19/2022] Open
Abstract
The larynx enables speech while regulating swallowing and respiration. Larynx function hinges on the laryngeal epithelium which originates as part of the anterior foregut and undergoes extensive remodeling to separate from the esophagus and form vocal folds that interface with the adjacent trachea. Here we find that sonic hedgehog (SHH) is essential for epithelial integrity in the mouse larynx as well as the anterior foregut. During larynx-esophageal separation, low Shh expression marks specific domains of actively remodeling epithelium that undergo an epithelial-to-mesenchymal transition (EMT) characterized by the induction of N-Cadherin and movement of cells out of the epithelial layer. Consistent with a role for SHH signaling in regulating this process, Shh mutants undergo an abnormal EMT throughout the anterior foregut and larynx, marked by a cadherin switch, movement out of the epithelial layer and cell death. Unexpectedly, Shh mutant epithelial cells are replaced by a new population of FOXA2-negative cells that likely derive from adjacent pouch tissues and form a rudimentary epithelium. These findings have important implications for interpreting the etiology of HH-dependent birth defects within the foregut. We propose that SHH signaling has a default role in maintaining epithelial identity throughout the anterior foregut and that regionalized reductions in SHH trigger epithelial remodeling.
Collapse
Affiliation(s)
- Janani Ramachandran
- Department of Molecular Biosciences, The University of Texas at AustinAustinUnited States
| | - Weiqiang Zhou
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public HealthBaltimoreUnited States
| | - Anna E Bardenhagen
- Department of Molecular Biosciences, The University of Texas at AustinAustinUnited States
| | - Talia Nasr
- Center for Stem Cell and Organoid Medicine (CuSTOM), Division of Developmental Biology, and Perinatal Institute, Cincinnati Children’s Hospital Medical CenterCincinnatiUnited States
- Department of Pediatrics, University of Cincinnati College of MedicineCincinnatiUnited States
| | - Ellen R Yates
- Department of Molecular Biosciences, The University of Texas at AustinAustinUnited States
| | - Aaron M Zorn
- Center for Stem Cell and Organoid Medicine (CuSTOM), Division of Developmental Biology, and Perinatal Institute, Cincinnati Children’s Hospital Medical CenterCincinnatiUnited States
- Department of Pediatrics, University of Cincinnati College of MedicineCincinnatiUnited States
| | - Hongkai Ji
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public HealthBaltimoreUnited States
| | - Steven A Vokes
- Department of Molecular Biosciences, The University of Texas at AustinAustinUnited States
| |
Collapse
|
19
|
Hammond NL, Dixon MJ. Revisiting the embryogenesis of lip and palate development. Oral Dis 2022; 28:1306-1326. [PMID: 35226783 PMCID: PMC10234451 DOI: 10.1111/odi.14174] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 02/17/2022] [Accepted: 02/23/2022] [Indexed: 12/13/2022]
Abstract
Clefts of the lip and palate (CLP), the major causes of congenital facial malformation globally, result from failure of fusion of the facial processes during embryogenesis. With a prevalence of 1 in 500-2500 live births, CLP causes major morbidity throughout life as a result of problems with facial appearance, feeding, speaking, obstructive apnoea, hearing and social adjustment and requires complex, multi-disciplinary care at considerable cost to healthcare systems worldwide. Long-term outcomes for affected individuals include increased mortality compared with their unaffected siblings. The frequent occurrence and major healthcare burden imposed by CLP highlight the importance of dissecting the molecular mechanisms driving facial development. Identification of the genetic mutations underlying syndromic forms of CLP, where CLP occurs in association with non-cleft clinical features, allied to developmental studies using appropriate animal models is central to our understanding of the molecular events underlying development of the lip and palate and, ultimately, how these are disturbed in CLP.
Collapse
Affiliation(s)
- Nigel L. Hammond
- Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
| | - Michael J. Dixon
- Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
| |
Collapse
|
20
|
Teng T, Teng CS, Kaartinen V, Bush JO. A unique form of collective epithelial migration is crucial for tissue fusion in the secondary palate and can overcome loss of epithelial apoptosis. Development 2022; 149:275520. [PMID: 35593401 PMCID: PMC9188751 DOI: 10.1242/dev.200181] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 04/11/2022] [Indexed: 11/20/2022]
Abstract
Tissue fusion frequently requires the removal of an epithelium that intervenes distinct primordia to form one continuous structure. In the mammalian secondary palate, a midline epithelial seam (MES) forms between two palatal shelves and must be removed to allow mesenchymal confluence. Abundant apoptosis and cell extrusion support their importance in MES removal. However, genetically disrupting the intrinsic apoptotic regulators BAX and BAK within the MES results in complete loss of cell death and cell extrusion, but successful removal of the MES. Novel static- and live-imaging approaches reveal that the MES is removed through streaming migration of epithelial trails and islands to reach the oral and nasal epithelial surfaces. Epithelial trail cells that express the basal epithelial marker ΔNp63 begin to express periderm markers, suggesting that migration is concomitant with differentiation. Live imaging reveals anisotropic actomyosin contractility within epithelial trails, and genetic ablation of actomyosin contractility results in dispersion of epithelial collectives and failure of normal MES migration. These findings demonstrate redundancy between cellular mechanisms of morphogenesis, and reveal a crucial and unique form of collective epithelial migration during tissue fusion. Summary: Multiple cellular processes mediate secondary palate fusion, including a unique form of streaming collective epithelial migration driven by pulsatile actomyosin contractility.
Collapse
Affiliation(s)
- Teng Teng
- University of California San Francisco 1 Department of Cell and Tissue Biology , , San Francisco, CA 94143 , USA
- University of California San Francisco 2 Program in Craniofacial Biology , , San Francisco, CA 94143 , USA
- Institute for Human Genetics, University of California San Francisco 3 , San Francisco, CA 94143 , USA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco 4 , San Francisco, CA 94143 , USA
| | - Camilla S. Teng
- University of California San Francisco 1 Department of Cell and Tissue Biology , , San Francisco, CA 94143 , USA
- University of California San Francisco 2 Program in Craniofacial Biology , , San Francisco, CA 94143 , USA
- Institute for Human Genetics, University of California San Francisco 3 , San Francisco, CA 94143 , USA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco 4 , San Francisco, CA 94143 , USA
| | - Vesa Kaartinen
- University of Michigan School of Dentistry 5 Department of Biologic and Materials Sciences , , Ann Arbor, MI 48109 , USA
| | - Jeffrey O. Bush
- University of California San Francisco 1 Department of Cell and Tissue Biology , , San Francisco, CA 94143 , USA
- University of California San Francisco 2 Program in Craniofacial Biology , , San Francisco, CA 94143 , USA
- Institute for Human Genetics, University of California San Francisco 3 , San Francisco, CA 94143 , USA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco 4 , San Francisco, CA 94143 , USA
| |
Collapse
|
21
|
Lan Y, Jiang R. Mouse models in palate development and orofacial cleft research: Understanding the crucial role and regulation of epithelial integrity in facial and palate morphogenesis. Curr Top Dev Biol 2022; 148:13-50. [PMID: 35461563 PMCID: PMC9060390 DOI: 10.1016/bs.ctdb.2021.12.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cleft lip and cleft palate are common birth defects resulting from genetic and/or environmental perturbations of facial development in utero. Facial morphogenesis commences during early embryogenesis, with cranial neural crest cells interacting with the surface ectoderm to form initially partly separate facial primordia consisting of the medial and lateral nasal prominences, and paired maxillary and mandibular processes. As these facial primordia grow around the primitive oral cavity and merge toward the ventral midline, the surface ectoderm undergoes a critical differentiation step to form an outer layer of flattened and tightly connected periderm cells with a non-stick apical surface that prevents epithelial adhesion. Formation of the upper lip and palate requires spatiotemporally regulated inter-epithelial adhesions and subsequent dissolution of the intervening epithelial seam between the maxillary and medial/lateral nasal processes and between the palatal shelves. Proper regulation of epithelial integrity plays a paramount role during human facial development, as mutations in genes encoding epithelial adhesion molecules and their regulators have been associated with syndromic and non-syndromic orofacial clefts. In this chapter, we summarize mouse genetic studies that have been instrumental in unraveling the mechanisms regulating epithelial integrity and periderm differentiation during facial and palate development. Since proper epithelial integrity also plays crucial roles in wound healing and cancer, understanding the mechanisms regulating epithelial integrity during facial development have direct implications for improvement in clinical care of craniofacial patients.
Collapse
Affiliation(s)
- Yu Lan
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States; Division of Plastic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Rulang Jiang
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States; Division of Plastic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States.
| |
Collapse
|
22
|
To Stick or Not to Stick: Adhesions in Orofacial Clefts. BIOLOGY 2022; 11:biology11020153. [PMID: 35205020 PMCID: PMC8869391 DOI: 10.3390/biology11020153] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 01/11/2022] [Accepted: 01/12/2022] [Indexed: 11/17/2022]
Abstract
Morphogenesis requires a tight coordination between mechanical forces and biochemical signals to inform individual cellular behavior. For these developmental processes to happen correctly the organism requires precise spatial and temporal coordination of the adhesion, migration, growth, differentiation, and apoptosis of cells originating from the three key embryonic layers, namely the ectoderm, mesoderm, and endoderm. The cytoskeleton and its remodeling are essential to organize and amplify many of the signaling pathways required for proper morphogenesis. In particular, the interaction of the cell junctions with the cytoskeleton functions to amplify the behavior of individual cells into collective events that are critical for development. In this review we summarize the key morphogenic events that occur during the formation of the face and the palate, as well as the protein complexes required for cell-to-cell adhesions. We then integrate the current knowledge into a comprehensive review of how mutations in cell-to-cell adhesion genes lead to abnormal craniofacial development, with a particular focus on cleft lip with or without cleft palate.
Collapse
|
23
|
Seelan RS, Pisano MM, Greene RM. MicroRNAs as epigenetic regulators of orofacial development. Differentiation 2022; 124:1-16. [DOI: 10.1016/j.diff.2022.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 12/30/2021] [Accepted: 01/13/2022] [Indexed: 11/03/2022]
|
24
|
Zulueta-Coarasa T, Rosenblatt J. The role of tissue maturity and mechanical state in controlling cell extrusion. Curr Opin Genet Dev 2021; 72:1-7. [PMID: 34560388 PMCID: PMC8860846 DOI: 10.1016/j.gde.2021.09.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 09/01/2021] [Accepted: 09/03/2021] [Indexed: 11/03/2022]
Abstract
Epithelia remove dying or excess cells by extrusion, a process that seamlessly squeezes cells out of the layer without disrupting their barrier function. New studies shed light into the intricate relationship between extrusion, tissue mechanics, and development. They emphasize the importance of whole tissue-mechanics, rather than single cell-mechanics in controlling extrusion. Tissue compaction, stiffness, and cell-cell adhesion can impact the efficiency of cell extrusion and mechanisms that drive it, to adapt to different conditions during development or disease.
Collapse
Affiliation(s)
- Teresa Zulueta-Coarasa
- The Randall Centre for Cell & Molecular Biophysics, Faculty of Life Sciences & Medicine, Schools of Basic & Medical Biosciences and Cancer & Pharmaceutical Sciences, King's College London, United Kingdom
| | - Jody Rosenblatt
- The Randall Centre for Cell & Molecular Biophysics, Faculty of Life Sciences & Medicine, Schools of Basic & Medical Biosciences and Cancer & Pharmaceutical Sciences, King's College London, United Kingdom.
| |
Collapse
|
25
|
Phatak M, Kulkarni S, Miles LB, Anjum N, Dworkin S, Sonawane M. Grhl3 promotes retention of epidermal cells under endocytic stress to maintain epidermal architecture in zebrafish. PLoS Genet 2021; 17:e1009823. [PMID: 34570762 PMCID: PMC8496789 DOI: 10.1371/journal.pgen.1009823] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Revised: 10/07/2021] [Accepted: 09/11/2021] [Indexed: 11/19/2022] Open
Abstract
Epithelia such as epidermis cover large surfaces and are crucial for survival. Maintenance of tissue homeostasis by balancing cell proliferation, cell size, and cell extrusion ensures epidermal integrity. Although the mechanisms of cell extrusion are better understood, how epithelial cells that round up under developmental or perturbed genetic conditions are reintegrated in the epithelium to maintain homeostasis remains unclear. Here, we performed live imaging in zebrafish embryos to show that epidermal cells that round up due to membrane homeostasis defects in the absence of goosepimples/myosinVb (myoVb) function, are reintegrated into the epithelium. Transcriptome analysis and genetic interaction studies suggest that the transcription factor Grainyhead-like 3 (Grhl3) induces the retention of rounded cells by regulating E-cadherin levels. Moreover, Grhl3 facilitates the survival of MyoVb deficient embryos by regulating cell adhesion, cell retention, and epidermal architecture. Our analyses have unraveled a mechanism of retention of rounded cells and its importance in epithelial homeostasis.
Collapse
Affiliation(s)
- Mandar Phatak
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Shruti Kulkarni
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Lee B. Miles
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, Australia
| | - Nazma Anjum
- Center for Biotechnology, A.C. College of Technology, Anna University, Chennai, India
| | - Sebastian Dworkin
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, Australia
| | - Mahendra Sonawane
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| |
Collapse
|
26
|
Goering JP, Wenger LW, Stetsiv M, Moedritzer M, Hall EG, Isai DG, Jack BM, Umar Z, Rickabaugh MK, Czirok A, Saadi I. In-frame deletion of SPECC1L microtubule association domain results in gain-of-function phenotypes affecting embryonic tissue movement and fusion events. Hum Mol Genet 2021; 31:18-31. [PMID: 34302166 DOI: 10.1093/hmg/ddab211] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 07/16/2021] [Accepted: 07/19/2021] [Indexed: 11/12/2022] Open
Abstract
Patients with autosomal dominant SPECC1L variants show syndromic malformations, including hypertelorism, cleft palate and omphalocele. These SPECC1L variants largely cluster in the second coiled-coil domain (CCD2), which facilitates association with microtubules. To study SPECC1L function in mice, we first generated a null allele (Specc1lΔEx4) lacking the entire SPECC1L protein. Homozygous mutants for these truncations died perinatally without cleft palate or omphalocele. Given the clustering of human variants in CCD2, we hypothesized that targeted perturbation of CCD2 may be required. Indeed, homozygotes for in-frame deletions involving CCD2 (Specc1lΔCCD2) resulted in exencephaly, cleft palate and ventral body wall closure defects (omphalocele). Interestingly, exencephaly and cleft palate were never observed in the same embryo. Further examination revealed a narrower oral cavity in exencephalic embryos, which allowed palatal shelves to elevate and fuse despite their defect. In the cell, wildtype SPECC1L was evenly distributed throughout the cytoplasm and colocalized with both microtubules and filamentous actin. In contrast, mutant SPECC1L-ΔCCD2 protein showed abnormal perinuclear accumulation with diminished overlap with microtubules, indicating that SPECC1L used microtubule association for trafficking in the cell. The perinuclear accumulation in the mutant also resulted in abnormally increased actin and non-muscle myosin II bundles dislocated to the cell periphery. Disrupted actomyosin cytoskeletal organization in SPECC1L CCD2 mutants would affect cell alignment and coordinated movement during neural tube, palate and ventral body wall closure. Thus, we show that perturbation of CCD2 in the context of full SPECC1L protein affects tissue fusion dynamics, indicating that human SPECC1L CCD2 variants are gain-of-function.
Collapse
Affiliation(s)
- Jeremy P Goering
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Luke W Wenger
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Marta Stetsiv
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Michael Moedritzer
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Everett G Hall
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Dona Greta Isai
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Brittany M Jack
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Zaid Umar
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Madison K Rickabaugh
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Andras Czirok
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Department of Biological Physics, Eotvos University, Budapest 1053, Hungary
| | - Irfan Saadi
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
27
|
Chaturvedi V, Murray MJ. Netrins: Evolutionarily Conserved Regulators of Epithelial Fusion and Closure in Development and Wound Healing. Cells Tissues Organs 2021; 211:193-211. [PMID: 33691313 DOI: 10.1159/000513880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 12/18/2020] [Indexed: 11/19/2022] Open
Abstract
Epithelial remodelling plays a crucial role during development. The ability of epithelial sheets to temporarily lose their integrity as they fuse with other epithelial sheets underpins events such as the closure of the neural tube and palate. During fusion, epithelial cells undergo some degree of epithelial-mesenchymal transition (EMT), whereby cells from opposing sheets dissolve existing cell-cell junctions, degrade the basement membrane, extend motile processes to contact each other, and then re-establish cell-cell junctions as they fuse. Similar events occur when an epithelium is wounded. Cells at the edge of the wound undergo a partial EMT and migrate towards each other to close the gap. In this review, we highlight the emerging role of Netrins in these processes, and provide insights into the possible signalling pathways involved. Netrins are secreted, laminin-like proteins that are evolutionarily conserved throughout the animal kingdom. Although best known as axonal chemotropic guidance molecules, Netrins also regulate epithelial cells. For example, Netrins regulate branching morphogenesis of the lung and mammary gland, and promote EMT during Drosophila wing eversion. Netrins also control epithelial fusion during optic fissure closure and inner ear formation, and are strongly implicated in neural tube closure and secondary palate closure. Netrins are also upregulated in response to organ damage and epithelial wounding, and can protect against ischemia-reperfusion injury and speed wound healing in cornea and skin. Since Netrins also have immunomodulatory properties, and can promote angiogenesis and re-innervation, they hold great promise as potential factors in future wound healing therapies.
Collapse
Affiliation(s)
- Vishal Chaturvedi
- School of BioSciences, University of Melbourne, Melbourne, Victoria, Australia
| | - Michael J Murray
- School of BioSciences, University of Melbourne, Melbourne, Victoria, Australia,
| |
Collapse
|
28
|
Hof L, Moreth T, Koch M, Liebisch T, Kurtz M, Tarnick J, Lissek SM, Verstegen MMA, van der Laan LJW, Huch M, Matthäus F, Stelzer EHK, Pampaloni F. Long-term live imaging and multiscale analysis identify heterogeneity and core principles of epithelial organoid morphogenesis. BMC Biol 2021; 19:37. [PMID: 33627108 PMCID: PMC7903752 DOI: 10.1186/s12915-021-00958-w] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 01/12/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Organoids are morphologically heterogeneous three-dimensional cell culture systems and serve as an ideal model for understanding the principles of collective cell behaviour in mammalian organs during development, homeostasis, regeneration, and pathogenesis. To investigate the underlying cell organisation principles of organoids, we imaged hundreds of pancreas and cholangiocarcinoma organoids in parallel using light sheet and bright-field microscopy for up to 7 days. RESULTS We quantified organoid behaviour at single-cell (microscale), individual-organoid (mesoscale), and entire-culture (macroscale) levels. At single-cell resolution, we monitored formation, monolayer polarisation, and degeneration and identified diverse behaviours, including lumen expansion and decline (size oscillation), migration, rotation, and multi-organoid fusion. Detailed individual organoid quantifications lead to a mechanical 3D agent-based model. A derived scaling law and simulations support the hypotheses that size oscillations depend on organoid properties and cell division dynamics, which is confirmed by bright-field microscopy analysis of entire cultures. CONCLUSION Our multiscale analysis provides a systematic picture of the diversity of cell organisation in organoids by identifying and quantifying the core regulatory principles of organoid morphogenesis.
Collapse
Affiliation(s)
- Lotta Hof
- Physical Biology Group, Buchmann Institute for Molecular Life Sciences (BMLS), Goethe-Universität Frankfurt am Main, Frankfurt am Main, Germany
| | - Till Moreth
- Physical Biology Group, Buchmann Institute for Molecular Life Sciences (BMLS), Goethe-Universität Frankfurt am Main, Frankfurt am Main, Germany
| | - Michael Koch
- Physical Biology Group, Buchmann Institute for Molecular Life Sciences (BMLS), Goethe-Universität Frankfurt am Main, Frankfurt am Main, Germany
| | - Tim Liebisch
- Frankfurt Institute for Advanced Studies and Faculty of Biological Sciences, Goethe-Universität Frankfurt am Main, Frankfurt am Main, Germany
| | - Marina Kurtz
- Department of Physics, Goethe Universität Frankfurt am Main, Frankfurt am Main, Germany
| | - Julia Tarnick
- Deanery of Biomedical Science, University of Edinburgh, Edinburgh, UK
| | - Susanna M Lissek
- Experimental Medicine and Therapy Research, University of Regensburg, Regensburg, Germany
| | - Monique M A Verstegen
- Department of Surgery, Erasmus MC - University Medical Center, Rotterdam, The Netherlands
| | - Luc J W van der Laan
- Department of Surgery, Erasmus MC - University Medical Center, Rotterdam, The Netherlands
| | - Meritxell Huch
- The Wellcome Trust/CRUK Gurdon Institute, University of Cambridge, Cambridge, UK
- Present address: Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Franziska Matthäus
- Frankfurt Institute for Advanced Studies and Faculty of Biological Sciences, Goethe-Universität Frankfurt am Main, Frankfurt am Main, Germany
| | - Ernst H K Stelzer
- Physical Biology Group, Buchmann Institute for Molecular Life Sciences (BMLS), Goethe-Universität Frankfurt am Main, Frankfurt am Main, Germany
| | - Francesco Pampaloni
- Physical Biology Group, Buchmann Institute for Molecular Life Sciences (BMLS), Goethe-Universität Frankfurt am Main, Frankfurt am Main, Germany.
| |
Collapse
|
29
|
Du W, Bhojwani A, Hu JK. FACEts of mechanical regulation in the morphogenesis of craniofacial structures. Int J Oral Sci 2021; 13:4. [PMID: 33547271 PMCID: PMC7865003 DOI: 10.1038/s41368-020-00110-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 12/03/2020] [Accepted: 12/07/2020] [Indexed: 02/07/2023] Open
Abstract
During embryonic development, organs undergo distinct and programmed morphological changes as they develop into their functional forms. While genetics and biochemical signals are well recognized regulators of morphogenesis, mechanical forces and the physical properties of tissues are now emerging as integral parts of this process as well. These physical factors drive coordinated cell movements and reorganizations, shape and size changes, proliferation and differentiation, as well as gene expression changes, and ultimately sculpt any developing structure by guiding correct cellular architectures and compositions. In this review we focus on several craniofacial structures, including the tooth, the mandible, the palate, and the cranium. We discuss the spatiotemporal regulation of different mechanical cues at both the cellular and tissue scales during craniofacial development and examine how tissue mechanics control various aspects of cell biology and signaling to shape a developing craniofacial organ.
Collapse
Affiliation(s)
- Wei Du
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
- School of Dentistry, University of California Los Angeles, Los Angeles, CA, USA
| | - Arshia Bhojwani
- School of Dentistry, University of California Los Angeles, Los Angeles, CA, USA
| | - Jimmy K Hu
- School of Dentistry, University of California Los Angeles, Los Angeles, CA, USA.
- Molecular Biology Institute, University of California Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
30
|
Iwata J. Gene-Environment Interplay and MicroRNAs in Cleft Lip and Cleft Palate. ORAL SCIENCE INTERNATIONAL 2021; 18:3-13. [PMID: 36855534 PMCID: PMC9969970 DOI: 10.1002/osi2.1072] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Cleft lip (CL) with/without cleft palate (CP) (hereafter CL/P) is the second most common congenital birth defect, affecting 7.94 to 9.92 children per 10,000 live births worldwide, followed by Down syndrome. An increasing number of genes have been identified as affecting susceptibility and/or as causative genes for CL/P in mouse genetic and chemically-induced CL and CP studies, as well as in human genome-wide association studies and linkage analysis. While marked progress has been made in the identification of genetic and environmental risk factors for CL/P, the interplays between these factors are not yet fully understood. This review aims to summarize our current knowledge of CL and CP from genetically engineered mouse models and environmental factors that have been studied in mice. Understanding the regulatory mechanism(s) of craniofacial development may not only advance our understanding of craniofacial developmental biology, but could also provide approaches for the prevention of birth defects and for tissue engineering in craniofacial tissue regeneration.
Collapse
Affiliation(s)
- Junichi Iwata
- Department of Diagnostic & Biomedical Sciences, The University of Texas Health Science Center at Houston, School of Dentistry, Houston, Texas, 77054 USA.,Center for Craniofacial Research, The University of Texas Health Science Center at Houston, School of Dentistry, Houston, Texas, 77054 USA.,Pediatric Research Center, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, Texas, 77030 USA.,MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas, 77030 USA
| |
Collapse
|
31
|
Patel A, Anderson G, Galea GL, Balys M, Sowden JC. A molecular and cellular analysis of human embryonic optic fissure closure related to the eye malformation coloboma. Development 2020; 147:dev193649. [PMID: 33158926 DOI: 10.1242/dev.193649] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 10/30/2020] [Indexed: 12/28/2022]
Abstract
Ocular coloboma is a congenital eye malformation, resulting from a failure in optic fissure closure (OFC) and causing visual impairment. There has been little study of the epithelial fusion process underlying closure in the human embryo and coloboma aetiology remains poorly understood. We performed RNAseq of cell populations isolated using laser capture microdissection to identify novel human OFC signature genes and probe the expression profile of known coloboma genes, along with a comparative murine analysis. Gene set enrichment patterns showed conservation between species. Expression of genes involved in epithelial-to-mesenchymal transition was transiently enriched in the human fissure margins during OFC at days 41-44. Electron microscopy and histological analyses showed that cells transiently delaminate at the point of closure, and produce cytoplasmic protrusions, before rearranging to form two continuous epithelial layers. Apoptosis was not observed in the human fissure margins. These analyses support a model of human OFC in which epithelial cells at the fissure margins undergo a transient epithelial-to-mesenchymal-like transition, facilitating cell rearrangement to form a complete optic cup.
Collapse
Affiliation(s)
- Aara Patel
- UCL Great Ormond Street Institute of Child Health, and NIHR Great Ormond Street Hospital Biomedical Research Centre, University College London, London WC1N 1EH, UK
| | - Glenn Anderson
- Department of Histopathology, Great Ormond Street Hospital for Children NHS Foundation Trust, London WC1N 3JH, UK
| | - Gabriel L Galea
- UCL Great Ormond Street Institute of Child Health, and NIHR Great Ormond Street Hospital Biomedical Research Centre, University College London, London WC1N 1EH, UK
| | - Monika Balys
- Department of Histopathology, Great Ormond Street Hospital for Children NHS Foundation Trust, London WC1N 3JH, UK
| | - Jane C Sowden
- UCL Great Ormond Street Institute of Child Health, and NIHR Great Ormond Street Hospital Biomedical Research Centre, University College London, London WC1N 1EH, UK
| |
Collapse
|
32
|
Yamamoto S, Kurosaka H, Miura J, Aoyama G, Sarper SE, Oka A, Inubushi T, Nakatsugawa K, Usami Y, Toyosawa S, Yamashiro T. Observation of the Epithelial Cell Behavior in the Nasal Septum During Primary Palate Closure in Mice. Front Physiol 2020; 11:538835. [PMID: 33123019 PMCID: PMC7566916 DOI: 10.3389/fphys.2020.538835] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 09/10/2020] [Indexed: 11/23/2022] Open
Abstract
Epithelial fusion is critical in palatogenesis, and incomplete fusion results in various type of facial cleft, depending on the region that fails to fuse. In mammalian palatogenesis, the bilateral secondary palatal processes fuse in the middle of the face to form the secondary palate. Later, the dorsal side of the secondary palatal shelves fuses with the nasal septum to complete palatogenesis. Importantly, the anterior border of the secondary palatal shelf fuses with the primary palate, which is located at the anterior and ventral border of the nasal septum. While numerous studies have investigated the mechanism of fusion between secondary palatal shelves, very little is known about how the primary palate touches and fuses with the secondary palatal shelves. In this study, we investigate the possible epithelial cell behaviors on the surface of the primary palate using palatal explant cultures of K14-GFP mice. A time-lapse observation of the GFP-labeled epithelium and an SEM analysis revealed that the extrusion epithelium appeared at the region corresponding to the fusing area and expanded rostrally on the nasal septum surface in the absence of the secondary palatal processes. Unlike on the secondary palate surface, cellular migration and subsequent autonomous mesenchymal exposure were not evident on the nasal septum or the primary palate. TUNEL staining revealed that these extrusion epithelia were undergoing apoptosis. These findings indicated that extrusion with apoptosis was autonomously initiated at the presumptive region of the fusion without contact with the opposing secondary palate.
Collapse
Affiliation(s)
- Sayuri Yamamoto
- Department of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Osaka University, Osaka, Japan
| | - Hiroshi Kurosaka
- Department of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Osaka University, Osaka, Japan
| | - Jiro Miura
- Division for Interdisciplinary Dentistry, Dental Hospital, Osaka University, Osaka, Japan
| | - Gozo Aoyama
- Department of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Osaka University, Osaka, Japan
| | - Safiye Esra Sarper
- Department of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Osaka University, Osaka, Japan.,Department of Biological Sciences, Graduate School of Science, Osaka University, Osaka, Japan
| | - Ayaka Oka
- Department of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Osaka University, Osaka, Japan
| | - Toshihiro Inubushi
- Department of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Osaka University, Osaka, Japan
| | - Kohei Nakatsugawa
- Department of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Osaka University, Osaka, Japan
| | - Yu Usami
- Department of Oral Pathology, Graduate School of Dentistry, Osaka University, Osaka, Japan
| | - Satoru Toyosawa
- Department of Oral Pathology, Graduate School of Dentistry, Osaka University, Osaka, Japan
| | - Takashi Yamashiro
- Department of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Osaka University, Osaka, Japan
| |
Collapse
|
33
|
Ji Y, Garland MA, Sun B, Zhang S, Reynolds K, McMahon M, Rajakumar R, Islam MS, Liu Y, Chen Y, Zhou CJ. Cellular and developmental basis of orofacial clefts. Birth Defects Res 2020; 112:1558-1587. [PMID: 32725806 DOI: 10.1002/bdr2.1768] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 06/21/2020] [Accepted: 06/27/2020] [Indexed: 12/11/2022]
Abstract
During craniofacial development, defective growth and fusion of the upper lip and/or palate can cause orofacial clefts (OFCs), which are among the most common structural birth defects in humans. The developmental basis of OFCs includes morphogenesis of the upper lip, primary palate, secondary palate, and other orofacial structures, each consisting of diverse cell types originating from all three germ layers: the ectoderm, mesoderm, and endoderm. Cranial neural crest cells and orofacial epithelial cells are two major cell types that interact with various cell lineages and play key roles in orofacial development. The cellular basis of OFCs involves defective execution in any one or several of the following processes: neural crest induction, epithelial-mesenchymal transition, migration, proliferation, differentiation, apoptosis, primary cilia formation and its signaling transduction, epithelial seam formation and disappearance, periderm formation and peeling, convergence and extrusion of palatal epithelial seam cells, cell adhesion, cytoskeleton dynamics, and extracellular matrix function. The latest cellular and developmental findings may provide a basis for better understanding of the underlying genetic, epigenetic, environmental, and molecular mechanisms of OFCs.
Collapse
Affiliation(s)
- Yu Ji
- Department of Biochemistry and Molecular Medicine, University of California at Davis, School of Medicine, Sacramento, California, USA.,Institute for Pediatric Regenerative Medicine of Shriners Hospitals for Children, School of Medicine, University of California at Davis, Sacramento, California, USA.,Biochemistry, Molecular, Cellular, and Developmental Biology (BMCDB) graduate group, University of California, Davis, California, USA
| | - Michael A Garland
- Department of Biochemistry and Molecular Medicine, University of California at Davis, School of Medicine, Sacramento, California, USA.,Institute for Pediatric Regenerative Medicine of Shriners Hospitals for Children, School of Medicine, University of California at Davis, Sacramento, California, USA
| | - Bo Sun
- Department of Biochemistry and Molecular Medicine, University of California at Davis, School of Medicine, Sacramento, California, USA.,Institute for Pediatric Regenerative Medicine of Shriners Hospitals for Children, School of Medicine, University of California at Davis, Sacramento, California, USA
| | - Shuwen Zhang
- Department of Biochemistry and Molecular Medicine, University of California at Davis, School of Medicine, Sacramento, California, USA.,Institute for Pediatric Regenerative Medicine of Shriners Hospitals for Children, School of Medicine, University of California at Davis, Sacramento, California, USA
| | - Kurt Reynolds
- Department of Biochemistry and Molecular Medicine, University of California at Davis, School of Medicine, Sacramento, California, USA.,Institute for Pediatric Regenerative Medicine of Shriners Hospitals for Children, School of Medicine, University of California at Davis, Sacramento, California, USA.,Biochemistry, Molecular, Cellular, and Developmental Biology (BMCDB) graduate group, University of California, Davis, California, USA
| | - Moira McMahon
- Institute for Pediatric Regenerative Medicine of Shriners Hospitals for Children, School of Medicine, University of California at Davis, Sacramento, California, USA
| | - Ratheya Rajakumar
- Department of Biochemistry and Molecular Medicine, University of California at Davis, School of Medicine, Sacramento, California, USA.,Institute for Pediatric Regenerative Medicine of Shriners Hospitals for Children, School of Medicine, University of California at Davis, Sacramento, California, USA
| | - Mohammad S Islam
- Institute for Pediatric Regenerative Medicine of Shriners Hospitals for Children, School of Medicine, University of California at Davis, Sacramento, California, USA
| | - Yue Liu
- Institute for Pediatric Regenerative Medicine of Shriners Hospitals for Children, School of Medicine, University of California at Davis, Sacramento, California, USA
| | - YiPing Chen
- Department of Cell and Molecular Biology, Tulane University, New Orleans, Louisiana, USA
| | - Chengji J Zhou
- Department of Biochemistry and Molecular Medicine, University of California at Davis, School of Medicine, Sacramento, California, USA.,Institute for Pediatric Regenerative Medicine of Shriners Hospitals for Children, School of Medicine, University of California at Davis, Sacramento, California, USA.,Biochemistry, Molecular, Cellular, and Developmental Biology (BMCDB) graduate group, University of California, Davis, California, USA
| |
Collapse
|
34
|
Freiberger K, Hemker S, McAnally R, King R, Meyers-Wallen VN, Schutte BC, Fyfe JC. Secondary Palate Development in the Dog ( Canis lupus familiaris). Cleft Palate Craniofac J 2020; 58:230-236. [PMID: 32705901 DOI: 10.1177/1055665620943771] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
OBJECTIVE To investigate the gestational timing of morphologic events in normal canine secondary palate development as a baseline for studies in dog models of isolated cleft palate (CP). METHODS Beagle and beagle/cocker spaniel-hybrid fetal dogs were obtained by cesarean-section on various days of gestation, timed from the initial rise of serum progesterone concentration. Morphology of fetal heads was determined by examining serial coronal sections. RESULTS On gestational day 35 (d35), the palatal shelves pointed ventrally alongside the tongue. On d36, palatal shelves were elongated and elevated to a horizontal position above the tongue but were not touching. On d37, palatine shelves and vomer were touching, but the medial epithelial seam (MES) between the apposed shelves remained. Immunostaining with epithelial protein markers showed that the MES gradually dissolved and was replaced by mesenchyme during d37-d44, and palate fusion was complete by d44. Examination of remnant MES suggested that fusion of palatal shelves began in mid-palate and moved rostrally and caudally. CONCLUSION Palate development occurs in dogs in the steps described in humans and mice, but palate closure occurs at an intermediate time in gestation. These normative data will form the basis of future studies to determine pathophysiologic mechanisms in dog models of CP. Added clinical significance is the enhancement of dogs as a large animal model to test new approaches for palate repair, with the obvious advantage of achieving full maturity within 2 years rather than 2 decades.
Collapse
Affiliation(s)
- Katharina Freiberger
- Microbiology & Molecular Genetics, 3078Michigan State University, East Lansing, MI, USA
| | - Shelby Hemker
- Microbiology & Molecular Genetics, 3078Michigan State University, East Lansing, MI, USA
| | - Ryan McAnally
- Microbiology & Molecular Genetics, 3078Michigan State University, East Lansing, MI, USA
| | - Rachel King
- Microbiology & Molecular Genetics, 3078Michigan State University, East Lansing, MI, USA
| | | | - Brian C Schutte
- Microbiology & Molecular Genetics, 3078Michigan State University, East Lansing, MI, USA.,Pediatrics & Human Development, 3078Michigan State University, East Lansing, MI, USA
| | - John C Fyfe
- Microbiology & Molecular Genetics, 3078Michigan State University, East Lansing, MI, USA
| |
Collapse
|
35
|
Lough KJ, Spitzer DC, Bergman AJ, Wu JJ, Byrd KM, Williams SE. Disruption of the nectin-afadin complex recapitulates features of the human cleft lip/palate syndrome CLPED1. Development 2020; 147:dev.189241. [PMID: 32554531 DOI: 10.1242/dev.189241] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 06/02/2020] [Indexed: 01/19/2023]
Abstract
Cleft palate (CP), one of the most common congenital conditions, arises from failures in secondary palatogenesis during embryonic development. Several human genetic syndromes featuring CP and ectodermal dysplasia have been linked to mutations in genes regulating cell-cell adhesion, yet mouse models have largely failed to recapitulate these findings. Here, we use in utero lentiviral-mediated genetic approaches in mice to provide the first direct evidence that the nectin-afadin axis is essential for proper palate shelf elevation and fusion. Using this technique, we demonstrate that palatal epithelial conditional loss of afadin (Afdn) - an obligate nectin- and actin-binding protein - induces a high penetrance of CP, not observed when Afdn is targeted later using Krt14-Cre We implicate Nectin1 and Nectin4 as being crucially involved, as loss of either induces a low penetrance of mild palate closure defects, while loss of both causes severe CP with a frequency similar to Afdn loss. Finally, expression of the human disease mutant NECTIN1W185X causes CP with greater penetrance than Nectin1 loss, suggesting this alteration may drive CP via a dominant interfering mechanism.
Collapse
Affiliation(s)
- Kendall J Lough
- Departments of Pathology & Laboratory Medicine and Biology, Lineberger Comprehensive Cancer Center, The University of North Carolina, Chapel Hill, NC 27599, USA
| | - Danielle C Spitzer
- Departments of Pathology & Laboratory Medicine and Biology, Lineberger Comprehensive Cancer Center, The University of North Carolina, Chapel Hill, NC 27599, USA
| | - Abby J Bergman
- Departments of Pathology & Laboratory Medicine and Biology, Lineberger Comprehensive Cancer Center, The University of North Carolina, Chapel Hill, NC 27599, USA
| | - Jessica J Wu
- Departments of Pathology & Laboratory Medicine and Biology, Lineberger Comprehensive Cancer Center, The University of North Carolina, Chapel Hill, NC 27599, USA
| | - Kevin M Byrd
- Departments of Pathology & Laboratory Medicine and Biology, Lineberger Comprehensive Cancer Center, The University of North Carolina, Chapel Hill, NC 27599, USA.,Department of Oral & Craniofacial Health Sciences, The University of North Carolina School of Dentistry, Chapel Hill, NC 27599, USA
| | - Scott E Williams
- Departments of Pathology & Laboratory Medicine and Biology, Lineberger Comprehensive Cancer Center, The University of North Carolina, Chapel Hill, NC 27599, USA
| |
Collapse
|
36
|
Goodwin AF, Chen CP, Vo NT, Bush JO, Klein OD. YAP/TAZ Regulate Elevation and Bone Formation of the Mouse Secondary Palate. J Dent Res 2020; 99:1387-1396. [PMID: 32623954 DOI: 10.1177/0022034520935372] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Clefting of the secondary palate is one of the most common congenital anomalies, and the multiple corrective surgeries that individuals with isolated cleft palate undergo are associated with major costs and morbidities. Secondary palate development is a complex, multistep process that includes the elevation of the palatal shelves from a vertical to horizontal position, a process that is not well understood. The Hippo signaling cascade is a mechanosensory pathway that regulates morphogenesis, homeostasis, and regeneration by controlling cell proliferation, apoptosis, and differentiation, primarily via negative regulation of the downstream effectors, Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ). We deleted Yap/Taz throughout the palatal shelf mesenchyme as well as specifically in the posterior palatal shelf mesenchyme, using the Osr2Cre and Col2Cre drivers, respectively, which resulted in palatal shelf elevation delay and clefting of the secondary palate. In addition, the deletion resulted in undersized bones of the secondary palate. We next determined downstream targets of YAP/TAZ in the posterior palatal shelves, which included Ibsp and Phex, genes involved in mineralization, and Loxl4, which encodes a lysyl oxidase that catalyzes collagen crosslinking. Ibsp, Phex, and Loxl4 were expressed at decreased levels in the ossification region in the posterior palatal shelf mesenchyme upon deletion of Yap/Taz. Furthermore, collagen levels were decreased specifically in the same region prior to elevation. Thus, our data suggest that YAP/TAZ may regulate collagen crosslinking in the palatal shelf mesenchyme, thus controlling palatal shelf elevation, as well as mineralization of the bones of the secondary palate.
Collapse
Affiliation(s)
- A F Goodwin
- Department of Orofacial Sciences, University of California, San Francisco, CA, USA.,Program in Craniofacial Biology, University of California, San Francisco, CA, USA
| | - C P Chen
- Department of Orofacial Sciences, University of California, San Francisco, CA, USA.,Program in Craniofacial Biology, University of California, San Francisco, CA, USA
| | - N T Vo
- Department of Orofacial Sciences, University of California, San Francisco, CA, USA.,Program in Craniofacial Biology, University of California, San Francisco, CA, USA
| | - J O Bush
- Program in Craniofacial Biology, University of California, San Francisco, CA, USA.,Department of Cell and Tissue Biology, University of California, San Francisco, CA, USA.,Institute of Human Genetics, University of California, San Francisco, CA, USA
| | - O D Klein
- Department of Orofacial Sciences, University of California, San Francisco, CA, USA.,Program in Craniofacial Biology, University of California, San Francisco, CA, USA.,Institute of Human Genetics, University of California, San Francisco, CA, USA.,Department of Pediatrics, University of California, San Francisco, CA, USA
| |
Collapse
|
37
|
Ninomiya H, Intoh A, Ishimine H, Onuma Y, Ito Y, Michiue T, Tazaki A, Kato M. Application of a human mesoderm tissue elongation system in vitro derived from human induced pluripotent stem cells to risk assessment for teratogenic chemicals. CHEMOSPHERE 2020; 250:126124. [PMID: 32092576 DOI: 10.1016/j.chemosphere.2020.126124] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 01/27/2020] [Accepted: 02/03/2020] [Indexed: 06/10/2023]
Abstract
Toxic compounds from the mother's diet and medication in addition to genetic factors and infection during pregnancy remain risks for various congenital disorders and misbirth. To ensure the safety of food and drugs for pregnant women, establishment of an in vitro system that morphologically resembles human tissues has been long desired. In this study, we focused on dorsal mesoderm elongation, one of the critical early development events for trunk formation, and we established in vitro autonomous elongating tissues from human induced pluripotent stem cells (hiPSCs). This artificial tissue elongation is regulated by MYOSIN II and FGF signaling, and is diminished by methylmercury or retinoic acid (RA), similar to in vivo human developmental disabilities. Moreover, our method for differentiation of hiPSCs requires only a short culture period, and the elongation is cell number-independent. Therefore, our in vitro human tissue elongation system is a potential tool for risk assessment assays for identification of teratogenic chemicals via human tissue morphogenesis.
Collapse
Affiliation(s)
- Hiromasa Ninomiya
- Department of Occupational and Environmental Health, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan; Department of Cell Biology, Nagoya City University, Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, Aichi, 467-8601, Japan
| | - Atsushi Intoh
- Department of Occupational and Environmental Health, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan; Voluntary Body for International Health Care in Universities, Nagoya, Japan; Nara Institute of Science and Technology, Division of Biological Science, Stem Cell Technologies Lab, Takayama-cho, Ikoma 8916-5, Nara, 630-0192, Japan
| | - Hisako Ishimine
- Department of Cell Biology and Anatomy, Fujita Health University, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan
| | - Yasuko Onuma
- Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology (AIST), Central 6, 1-1-1 Higashi, Tsukuba, Ibaraki, 305-8566, Japan
| | - Yuzuru Ito
- Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology (AIST), Central 6, 1-1-1 Higashi, Tsukuba, Ibaraki, 305-8566, Japan
| | - Tatsuo Michiue
- Department of Life Sciences (Biology), Graduate School of Arts and Sciences, The University of Tokyo, Tokyo, Japan
| | - Akira Tazaki
- Department of Occupational and Environmental Health, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan; Voluntary Body for International Health Care in Universities, Nagoya, Japan
| | - Masashi Kato
- Department of Occupational and Environmental Health, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan; Voluntary Body for International Health Care in Universities, Nagoya, Japan.
| |
Collapse
|
38
|
Palmieri A, Scapoli L, Carrozzo M, Cura F, Morselli PG, Pannuto L, Nouri N, Carinci F, Lauritano D, Martinelli M. ROCK1 is associated with non-syndromic cleft palate. J Oral Pathol Med 2019; 49:164-168. [PMID: 31715657 DOI: 10.1111/jop.12973] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 10/01/2019] [Accepted: 11/08/2019] [Indexed: 01/01/2023]
Abstract
BACKGROUND Craniofacial morphogenesis is the result of an intricate multistep network of tightly controlled spatial and temporal signalling that involves several molecules and transcription factors organized into highly coordinated pathways. Any alteration in even one step of this delicate process can lead to congenital malformations such as cleft palate. One of the first steps in embryonal orofacial development is the migration of cells from the neural crests to the branchial arches. Next, the cells have to proliferate, differentiate, move and connect to each other in order to correctly form the palate. Cell contraction, promoted by the interaction of non-muscle myosin II and actin A, is a crucial step in morphogenesis and is regulated by ROCK1 protein. METHODS A family-based association study was carried out in order to verify whether or not genetic variants of ROCK1 were associated with non-syndromic cleft palate (nsCP). Two cohorts from Italy and Iran, a total of 189 nsCP cases and their parents were enrolled. RESULTS The rs35996865-G allele was under-transmitted in cases of nsCP [P = .006, odds ratio (OR) = 0.63 (95% CI 0.45-0.88)]. CONCLUSION This investigation reveals for the first time data supporting a role for ROCK1 in nsCP aetiology.
Collapse
Affiliation(s)
- Annalisa Palmieri
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Luca Scapoli
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Marco Carrozzo
- School of Dental Sciences, University of Newcastle upon Tyne, Newcastle upon Tyne, UK
| | - Francesca Cura
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Paolo Giovanni Morselli
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy.,Plastic Surgery Unit, University Hospital of Bologna Sant'Orsola Malpighi Polyclinic, Bologna, Italy
| | - Lucia Pannuto
- Plastic Surgery Unit, University Hospital of Bologna Sant'Orsola Malpighi Polyclinic, Bologna, Italy
| | - Nayereh Nouri
- Craniofacial and Cleft Research Center, Isfahan University of Medical Sciences, Isfahan, Iran.,Department of Genetics and Molecular Biology, Medical School, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Francesco Carinci
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Dorina Lauritano
- Department of Medicine and Surgery, Centre of Neuroscience of Milan, University of Milano-Bicocca, Milan, Italy
| | - Marcella Martinelli
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| |
Collapse
|
39
|
Logan SM, Ruest LB, Benson MD, Svoboda KKH. Extracellular Matrix in Secondary Palate Development. Anat Rec (Hoboken) 2019; 303:1543-1556. [PMID: 31513730 DOI: 10.1002/ar.24263] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 05/14/2019] [Accepted: 07/03/2019] [Indexed: 12/11/2022]
Abstract
The secondary palate arises from outgrowths of epithelia-covered embryonic mesenchyme that grow from the maxillary prominence, remodel to meet over the tongue, and fuse at the midline. These events require the coordination of cell proliferation, migration, and gene expression, all of which take place in the context of the extracellular matrix (ECM). Palatal cells generate their ECM, and then stiffen, degrade, or otherwise modify its properties to achieve the required cell movement and organization during palatogenesis. The ECM, in turn, acts on the cells through their matrix receptors to change their gene expression and thus their phenotype. The number of ECM-related gene mutations that cause cleft palate in mice and humans is a testament to the crucial role the matrix plays in palate development and a reminder that understanding that role is vital to our progress in treating palate deformities. This article will review the known ECM constituents at each stage of palatogenesis, the mechanisms of tissue reorganization and cell migration through the palatal ECM, the reciprocal relationship between the ECM and gene expression, and human syndromes with cleft palate that arise from mutations of ECM proteins and their regulators. Anat Rec, 2019. © 2019 American Association for Anatomy.
Collapse
Affiliation(s)
- Shaun M Logan
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, Texas
| | - L Bruno Ruest
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, Texas
| | - M Douglas Benson
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, Texas
| | - Kathy K H Svoboda
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, Texas
| |
Collapse
|
40
|
Mammadova A, Carels CEL, Zhou J, Gilissen C, Helmich MPAC, Bian Z, Zhou H, Von den Hoff JW. Deregulated Adhesion Program in Palatal Keratinocytes of Orofacial Cleft Patients. Genes (Basel) 2019; 10:genes10110836. [PMID: 31652793 PMCID: PMC6895790 DOI: 10.3390/genes10110836] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 10/17/2019] [Accepted: 10/19/2019] [Indexed: 01/10/2023] Open
Abstract
Orofacial clefts (OFCs) are the most frequent craniofacial birth defects. An orofacial cleft (OFC) occurs as a result of deviations in palatogenesis. Cell proliferation, differentiation, adhesion, migration and apoptosis are crucial in palatogenesis. We hypothesized that deregulation of these processes in oral keratinocytes contributes to OFC. We performed microarray expression analysis on palatal keratinocytes from OFC and non-OFC individuals. Principal component analysis showed a clear difference in gene expression with 24% and 17% for the first and second component, respectively. In OFC cells, 228 genes were differentially expressed (p < 0.001). Gene ontology analysis showed enrichment of genes involved in β1 integrin-mediated adhesion and migration, as well as in P-cadherin expression. A scratch assay demonstrated reduced migration of OFC keratinocytes (343.6 ± 29.62 μm) vs. non-OFC keratinocytes (503.4 ± 41.81 μm, p < 0.05). Our results indicate that adhesion and migration are deregulated in OFC keratinocytes, which might contribute to OFC pathogenesis.
Collapse
Affiliation(s)
- Aysel Mammadova
- Department of Dentistry, Section Orthodontics and Craniofacial Biology, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center, P.O. Box 9101, 6500 HB Nijmegen, The Netherlands.
| | - Carine E L Carels
- Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium.
- Department of Oral Health Sciences, KU Leuven, 3000 Leuven, Belgium.
| | - Jie Zhou
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, Wuhan University, Wuhan 430079, China.
| | - Christian Gilissen
- Department of Human Genetics, Radboud University Medical Center, P.O. Box 9101, 6500 HB Nijmegen, The Netherlands.
| | - Maria P A C Helmich
- Department of Dentistry, Section Orthodontics and Craniofacial Biology, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center, P.O. Box 9101, 6500 HB Nijmegen, The Netherlands.
| | - Zhuan Bian
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, Wuhan University, Wuhan 430079, China.
| | - Huiqing Zhou
- Department of Human Genetics, Radboud University Medical Center, P.O. Box 9101, 6500 HB Nijmegen, The Netherlands.
- Department of Molecular Developmental Biology, Radboud Institute for Molecular Life Sciences (RIMLS), P.O. Box 9101, 6500 HB Nijmegen, The Netherlands.
| | - Johannes W Von den Hoff
- Department of Dentistry, Section Orthodontics and Craniofacial Biology, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center, P.O. Box 9101, 6500 HB Nijmegen, The Netherlands.
| |
Collapse
|
41
|
Baker NC, Sipes NS, Franzosa J, Belair DG, Abbott BD, Judson RS, Knudsen TB. Characterizing cleft palate toxicants using ToxCast data, chemical structure, and the biomedical literature. Birth Defects Res 2019; 112:19-39. [PMID: 31471948 DOI: 10.1002/bdr2.1581] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 07/23/2019] [Accepted: 07/24/2019] [Indexed: 12/11/2022]
Abstract
Cleft palate has been linked to both genetic and environmental factors that perturb key events during palatal morphogenesis. As a developmental outcome, it presents a challenging, mechanistically complex endpoint for predictive modeling. A data set of 500 chemicals evaluated for their ability to induce cleft palate in animal prenatal developmental studies was compiled from Toxicity Reference Database and the biomedical literature, which included 63 cleft palate active and 437 inactive chemicals. To characterize the potential molecular targets for chemical-induced cleft palate, we mined the ToxCast high-throughput screening database for patterns and linkages in bioactivity profiles and chemical structural descriptors. ToxCast assay results were filtered for cytotoxicity and grouped by target gene activity to produce a "gene score." Following unsuccessful attempts to derive a global prediction model using structural and gene score descriptors, hierarchical clustering was applied to the set of 63 cleft palate positives to extract local structure-bioactivity clusters for follow-up study. Patterns of enrichment were confirmed on the complete data set, that is, including cleft palate inactives, and putative molecular initiating events identified. The clusters corresponded to ToxCast assays for cytochrome P450s, G-protein coupled receptors, retinoic acid receptors, the glucocorticoid receptor, and tyrosine kinases/phosphatases. These patterns and linkages were organized into preliminary decision trees and the resulting inferences were mapped to a putative adverse outcome pathway framework for cleft palate supported by literature evidence of current mechanistic understanding. This general data-driven approach offers a promising avenue for mining chemical-bioassay drivers of complex developmental endpoints where data are often limited.
Collapse
Affiliation(s)
| | - Nisha S Sipes
- NIEHS Division of the National Toxicology Program, Research Triangle Park, North Carolina
| | - Jill Franzosa
- IOAA CSS, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina
| | - David G Belair
- NHEERL, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina
| | - Barbara D Abbott
- NHEERL, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina
| | - Richard S Judson
- National Center for Computational Toxicology, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina
| | - Thomas B Knudsen
- National Center for Computational Toxicology, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina
| |
Collapse
|
42
|
Ke CY, Mei HH, Wong FH, Lo LJ. IRF6 and TAK1 coordinately promote the activation of HIPK2 to stimulate apoptosis during palate fusion. Sci Signal 2019; 12:12/593/eaav7666. [DOI: 10.1126/scisignal.aav7666] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cleft palate is a common craniofacial defect caused by a failure in palate fusion. The palatal shelves migrate toward one another and meet at the embryonic midline, creating a seam. Transforming growth factor–β3 (TGF-β3)–induced apoptosis of the medial edge epithelium (MEE), the cells located along the seam, is required for completion of palate fusion. The transcription factor interferon regulatory factor 6 (IRF6) promotes TGF-β3–induced MEE cell apoptosis by stimulating the degradation of the transcription factor ΔNp63 and promoting the expression of the gene encoding the cyclin-dependent kinase inhibitor p21. Because homeodomain-interacting protein kinase 2 (HIPK2) functions downstream of IRF6 in human cancer cells and is required for ΔNp63 protein degradation in keratinocytes, we investigated whether HIPK2 played a role in IRF6-induced ΔNp63 degradation in palate fusion. HIPK2 was present in the MEE cells of mouse palatal shelves during seam formation in vivo, and ectopic expression of IRF6 in palatal shelves cultured ex vivo stimulated the expression of Hipk2 and the accumulation of phosphorylated HIPK2. Knockdown and ectopic expression experiments in organ culture demonstrated that p21 was required for HIPK2- and IRF6-dependent activation of caspase 3, MEE apoptosis, and palate fusion. Contact between palatal shelves enhanced the phosphorylation of TGF-β–activated kinase 1 (TAK1), which promoted the phosphorylation of HIPK2 and palate fusion. Our findings demonstrate that HIPK2 promotes seam cell apoptosis and palate fusion downstream of IRF6 and that IRF6 and TAK1 appear to coordinately enhance the abundance and activation of HIPK2 during palate fusion.
Collapse
|
43
|
Logan SM, Benson MD. Medial epithelial seam cell migration during palatal fusion. J Cell Physiol 2019; 235:1417-1424. [PMID: 31264714 DOI: 10.1002/jcp.29061] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 06/13/2019] [Indexed: 12/13/2022]
Abstract
The mammalian secondary palate forms from two shelves of mesenchyme sheathed in a single-layered epithelium. These shelves meet during embryogenesis to form the midline epithelial seam (MES). Failure of MES degradation prevents mesenchymal confluence and results in a cleft palate. Previous studies indicated that MES cells undergo features of epithelial-to-mesenchymal transition (EMT) and may become migratory as part of the fusion mechanism. To detect MES cell movement over the course of fusion, we imaged the midline of fusing embryonic ephrin-B2/GFP mouse palates in real time using two-photon microscopy. These mice express an ephrin-B2-driven green fluorescent protein (GFP) that labels the palatal epithelium nuclei and persists in those cells through the time window necessary for fusion. We observed collective migration of MES cells toward the oral surface of the palatal shelf over 48 hr of imaging, and we confirmed histologically that the imaged palates had fused by the end of the imaged period. We previously reported that ephrin reverse signaling in the MES is required for palatal fusion. We therefore added recombinant EphA4/Fc protein to block this signaling in imaged palates. The blockage inhibited fusion, as expected, but did not change the observed migration of GFP-labeled cells. Thus, we uncoupled migration and fusion. Our data reveal that palatal MES cells undergo a collective, unidirectional movement during palatal fusion and that ephrin reverse signaling, though required for fusion, controls aspects of the fusion mechanism independent of migration.
Collapse
Affiliation(s)
- Shaun M Logan
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, Texas
| | - M Douglas Benson
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, Texas
| |
Collapse
|
44
|
Aoyama G, Kurosaka H, Oka A, Nakatsugawa K, Yamamoto S, Sarper SE, Usami Y, Toyosawa S, Inubushi T, Isogai Y, Yamashiro T. Observation of Dynamic Cellular Migration of the Medial Edge Epithelium of the Palatal Shelf in vitro. Front Physiol 2019; 10:698. [PMID: 31244674 PMCID: PMC6562562 DOI: 10.3389/fphys.2019.00698] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 05/20/2019] [Indexed: 12/22/2022] Open
Abstract
Palatal fusion is a critical step during palatogenesis. In this fusing interface, the epithelial sheets need to be removed in order to achieve mesenchymal continuity. Epithelial cellular migration is one of the possible mechanisms, and live imaging of the labeled epithelium could provide direct evidence for it. However, the removal of medial edge epithelium (MEE) between the bilateral processes takes place in the middle of the dorso-ventral axis of the palatal shelf, and thus it is challenging to capture the cellular behavior directly. Here, we evaluate cellular behavior of MEE cells using a live imaging technique with a mouse model which expresses GFP under the promoter of Keratin14 (K14-GFP) and unpaired palatal shelf culture. Using this approach, we successfully obtained live images of epithelial behavior and detected epithelial cell migration on the surface of the secondary palatal shelf without touching of the opposing shelf. Additionally, the pattern of epithelial elimination resulted in oval-shaped exposed mesenchyme, which recapitulated the situation during secondary palate fusion in vivo. Detailed image processing revealed that most of the MEE migrated in an outward direction at the boundary regions as the oval shape of the exposed mesenchyme expanded. The migration was preceded by the bulging of MEE, and disappearance of GFP signals was not evident in bulging or migrating MEE at the boundary regions. Furthermore, the MEE migration and the subsequent mesenchymal exposure were disturbed by application of ROCK inhibitor. Together, these findings indicated that epithelial cell migration contributed importantly to the MEE removal and the subsequent exposure of the underlying mesenchyme. Furthermore, they indicated that the migration of epithelial cells was regulated in a time- and space-specific manner, since unpaired palatal shelf culture exhibited these cellular behaviors even in the absence of the opposing shelf. Altogether, present data indicated that this new experimental system combining live imaging with GFP-labeled epithelium mice and unpaired palatal shelf culture enabled direct visualization of cellular migration of MEE in vitro and could be a powerful tool to investigate its cellular and molecular mechanisms.
Collapse
Affiliation(s)
- Gozo Aoyama
- Department of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Osaka University, Osaka, Japan
| | - Hiroshi Kurosaka
- Department of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Osaka University, Osaka, Japan
| | - Ayaka Oka
- Department of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Osaka University, Osaka, Japan
| | - Kohei Nakatsugawa
- Department of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Osaka University, Osaka, Japan
| | - Sayuri Yamamoto
- Department of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Osaka University, Osaka, Japan
| | - Safiye Esra Sarper
- Department of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Osaka University, Osaka, Japan.,Department of Biological Sciences, Graduate School of Science, Osaka University, Osaka, Japan
| | - Yu Usami
- Department of Oral Pathology, Graduate School of Dentistry, Osaka University, Osaka, Japan
| | - Satoru Toyosawa
- Department of Oral Pathology, Graduate School of Dentistry, Osaka University, Osaka, Japan
| | - Toshihiro Inubushi
- Department of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Osaka University, Osaka, Japan
| | - Yukako Isogai
- Department of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Osaka University, Osaka, Japan
| | - Takashi Yamashiro
- Department of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Osaka University, Osaka, Japan
| |
Collapse
|
45
|
Abstract
Deviations from the precisely coordinated programme of human head development can lead to craniofacial and orofacial malformations often including a variety of dental abnormalities too. Although the aetiology is still unknown in many cases, during the last decades different intracellular signalling pathways have been genetically linked to specific disorders. Among these pathways, the RAS/extracellular signal-regulated kinase (ERK) signalling cascade is the focus of this review since it encompasses a large group of genes that when mutated cause some of the most common and severe developmental anomalies in humans. We present the components of the RAS/ERK pathway implicated in craniofacial and orodental disorders through a series of human and animal studies. We attempt to unravel the specific molecular targets downstream of ERK that act on particular cell types and regulate key steps in the associated developmental processes. Finally we point to ambiguities in our current knowledge that need to be clarified before RAS/ERK-targeting therapeutic approaches can be implemented.
Collapse
|
46
|
Acebedo AR, Suzuki K, Hino S, Alcantara MC, Sato Y, Haga H, Matsumoto KI, Nakao M, Shimamura K, Takeo T, Nakagata N, Miyagawa S, Nishinakamura R, Adelstein RS, Yamada G. Mesenchymal actomyosin contractility is required for androgen-driven urethral masculinization in mice. Commun Biol 2019; 2:95. [PMID: 30886905 PMCID: PMC6408527 DOI: 10.1038/s42003-019-0336-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 02/01/2019] [Indexed: 01/23/2023] Open
Abstract
The morphogenesis of mammalian embryonic external genitalia (eExG) shows dynamic differences between males and females. In genotypic males, eExG are masculinized in response to androgen signaling. Disruption of this process can give rise to multiple male reproductive organ defects. Currently, mechanisms of androgen-driven sexually dimorphic organogenesis are still unclear. We show here that mesenchymal-derived actomyosin contractility, by MYH10, is essential for the masculinization of mouse eExG. MYH10 is expressed prominently in the bilateral mesenchyme of male eExG. Androgen induces MYH10 protein expression and actomyosin contractility in the bilateral mesenchyme. Inhibition of actomyosin contractility through blebbistatin treatment and mesenchymal genetic deletion induced defective urethral masculinization with reduced mesenchymal condensation. We also suggest that actomyosin contractility regulates androgen-dependent mesenchymal directional cell migration to form the condensation in the bilateral mesenchyme leading to changes in urethral plate shape to accomplish urethral masculinization. Thus, mesenchymal-derived actomyosin contractility is indispensable for androgen-driven urethral masculinization.
Collapse
Affiliation(s)
- Alvin R. Acebedo
- Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, 641-8509 Japan
| | - Kentaro Suzuki
- Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, 641-8509 Japan
| | - Shinjiro Hino
- Department of Medical Cell Biology, Institute of Molecular Embryology and Genetics (IMEG), Kumamoto University, Kumamoto, 860-0811 Japan
| | - Mellissa C. Alcantara
- Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, 641-8509 Japan
| | - Yuki Sato
- Department of Anatomy and Cell Biology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582 Japan
| | - Hisashi Haga
- Transdisciplinary Life Science Course, Faculty of Advanced Life Science, Hokkaido University, N10-W8, Kita-ku, Sapporo, 060-0810 Japan
| | - Ken-ichi Matsumoto
- Department of Biosignaling and Radioisotope Experiment, Interdisciplinary Center for Science Research, Organization for Research, Shimane University, Izumo, Shimane, 693-8501 Japan
| | - Mitsuyoshi Nakao
- Department of Medical Cell Biology, Institute of Molecular Embryology and Genetics (IMEG), Kumamoto University, Kumamoto, 860-0811 Japan
| | - Kenji Shimamura
- Department of Brain Morphogenesis, Institute of Molecular Embryology and Genetics (IMEG), Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto, 860-0811 Japan
| | - Toru Takeo
- Division of Reproductive Engineering, Center for Animal Resources and Development (CARD), Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto, 860-0811 Japan
| | - Naomi Nakagata
- Division of Reproductive Engineering, Center for Animal Resources and Development (CARD), Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto, 860-0811 Japan
| | - Shinichi Miyagawa
- Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, 641-8509 Japan
| | - Ryuichi Nishinakamura
- Department of Kidney Development, Institute of Molecular Embryology and Genetics (IMEG), Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto, 860-0811 Japan
| | - Robert S. Adelstein
- Laboratory of Molecular Cardiology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892-1762 USA
| | - Gen Yamada
- Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, 641-8509 Japan
| |
Collapse
|
47
|
Brock CK, Wallin ST, Ruiz OE, Samms KM, Mandal A, Sumner EA, Eisenhoffer GT. Stem cell proliferation is induced by apoptotic bodies from dying cells during epithelial tissue maintenance. Nat Commun 2019; 10:1044. [PMID: 30837472 PMCID: PMC6400930 DOI: 10.1038/s41467-019-09010-6] [Citation(s) in RCA: 130] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 02/13/2019] [Indexed: 12/15/2022] Open
Abstract
Epithelial tissues require the removal and replacement of damaged cells to sustain a functional barrier. Dying cells provide instructive cues that can influence surrounding cells to proliferate, but how these signals are transmitted to their healthy neighbors to control cellular behaviors during tissue homeostasis remains poorly understood. Here we show that dying stem cells facilitate communication with adjacent stem cells by caspase-dependent production of Wnt8a-containing apoptotic bodies to drive cellular turnover in living epithelia. Basal stem cells engulf apoptotic bodies, activate Wnt signaling, and are stimulated to divide to maintain tissue-wide cell numbers. Inhibition of either cell death or Wnt signaling eliminated the apoptosis-induced cell division, while overexpression of Wnt8a signaling combined with induced cell death led to an expansion of the stem cell population. We conclude that ingestion of apoptotic bodies represents a regulatory mechanism linking death and division to maintain overall stem cell numbers and epithelial tissue homeostasis. Damaged epithelial tissues are known to compensate for cell death through compensatory cell divisions to maintain epithelial integrity. Here, the authors show in living epithelia that dying cells stimulate adjacent stem cells to divide through caspase-dependent production of Wnt8a-containing apoptotic bodies.
Collapse
Affiliation(s)
- Courtney K Brock
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Stephen T Wallin
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Oscar E Ruiz
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Krystin M Samms
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Amrita Mandal
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Elizabeth A Sumner
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - George T Eisenhoffer
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA. .,Genetics and Epigenetics Graduate Program, The University of Texas Graduate School of Biomedical Sciences at Houston, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|
48
|
Yu K, Yonemitsu MA. In Vitro Analysis of Palatal Shelf Elevation During Secondary Palate Formation. Anat Rec (Hoboken) 2019; 302:1594-1604. [PMID: 30730607 DOI: 10.1002/ar.24076] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 09/19/2018] [Accepted: 10/03/2018] [Indexed: 01/05/2023]
Abstract
Palatal shelf elevation is an essential morphogenetic process during secondary palate formation. It has been proposed that shelf elevation results from an intrinsic elevating force and is regulated by extrinsic factors that are associated with development of other orofacial structures. Although dynamic palate culture is a common in vitro approach for studying shelf elevation, it requires the tongue or the tongue and mandible to be removed before culture, which prevents any determination of the role of the extrinsic factors in regulating shelf elevation. We showed that ex vivo removal of the tongue and mandible from unfixed embryonic heads led to spontaneous shelf movements that were more pronounced at late E13.5 and early E14.5 than those of E12.5 and early E13.5, suggesting that the strength of the elevating force increases over time during palate development. We further used a suspension culture technique to analyze palatal shelf movement in an intact oral cavity by culturing the orofacial portion of embryonic heads that include the maxilla, palatal shelves, mandible, and tongue (MPMT). MPMT explants were cultured in the serum-free medium with slow rotation for 24-48 hr. The palatal shelves successfully elevated during culture and displayed intermediate morphologies that closely resemble those of in vivo shelf elevation. We demonstrate that the tongue and mandible facilitate shelf medial movement/growth during shelf elevation and further suggest that the interaction of the palatal shelves and tongue could be one of the extrinsic factors that regulate the elevation process. Anat Rec, 302:1594-1604, 2019. © 2019 American Association for Anatomy.
Collapse
Affiliation(s)
- Kai Yu
- Division of Craniofacial Medicine, Department of Pediatrics, University of Washington and Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, Washington
| | - Marisa A Yonemitsu
- Division of Craniofacial Medicine, Department of Pediatrics, University of Washington and Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, Washington
| |
Collapse
|
49
|
Franco JJ, Atieh Y, Bryan CD, Kwan KM, Eisenhoffer GT. Cellular crowding influences extrusion and proliferation to facilitate epithelial tissue repair. Mol Biol Cell 2019; 30:1890-1899. [PMID: 30785842 PMCID: PMC6727764 DOI: 10.1091/mbc.e18-05-0295] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Epithelial wound healing requires a complex orchestration of cellular rearrangements and movements to restore tissue architecture and function after injury. While it is well known that mechanical forces can affect tissue morphogenesis and patterning, how the biophysical cues generated after injury influence cellular behaviors during tissue repair is not well understood. Using time-lapse confocal imaging of epithelial tissues in living zebrafish larvae, we provide evidence that localized increases in cellular crowding during wound closure promote the extrusion of nonapoptotic cells via mechanically regulated stretch-activated ion channels (SACs). Directed cell migration toward the injury site promoted rapid changes in cell number and generated shifts in tension at cellular interfaces over long spatial distances. Perturbation of SAC activity resulted in failed extrusion and increased proliferation in crowded areas of the tissue. Together, we conclude that localized cell number plays a key role in dictating cellular behaviors that facilitate wound closure and tissue repair.
Collapse
Affiliation(s)
- Jovany J Franco
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030.,Department of BioSciences, Rice University, Houston, TX 77251
| | - Youmna Atieh
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Chase D Bryan
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112
| | - Kristen M Kwan
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112
| | - George T Eisenhoffer
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030.,Genetics and Epigenetics Graduate Program, Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
| |
Collapse
|
50
|
TGF-β Signaling and the Epithelial-Mesenchymal Transition during Palatal Fusion. Int J Mol Sci 2018; 19:ijms19113638. [PMID: 30463190 PMCID: PMC6274911 DOI: 10.3390/ijms19113638] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 10/27/2018] [Accepted: 11/12/2018] [Indexed: 12/15/2022] Open
Abstract
Signaling by transforming growth factor (TGF)-β plays an important role in development, including in palatogenesis. The dynamic morphological process of palatal fusion occurs to achieve separation of the nasal and oral cavities. Critically and specifically important in palatal fusion are the medial edge epithelial (MEE) cells, which are initially present at the palatal midline seam and over the course of the palate fusion process are lost from the seam, due to cell migration, epithelial-mesenchymal transition (EMT), and/or programed cell death. In order to define the role of TGF-β signaling during this process, several approaches have been utilized, including a small interfering RNA (siRNA) strategy targeting TGF-β receptors in an organ culture context, the use of genetically engineered mice, such as Wnt1-cre/R26R double transgenic mice, and a cell fate tracing through utilization of cell lineage markers. These approaches have permitted investigators to distinguish some specific traits of well-defined cell populations throughout the palatogenic events. In this paper, we summarize the current understanding on the role of TGF-β signaling, and specifically its association with MEE cell fate during palatal fusion. TGF-β is highly regulated both temporally and spatially, with TGF-β3 and Smad2 being the preferentially expressed signaling molecules in the critical cells of the fusion processes. Interestingly, the accessory receptor, TGF-β type 3 receptor, is also critical for palatal fusion, with evidence for its significance provided by Cre-lox systems and siRNA approaches. This suggests the high demand of ligand for this fine-tuned signaling process. We discuss the new insights in the fate of MEE cells in the midline epithelial seam (MES) during the palate fusion process, with a particular focus on the role of TGF-β signaling.
Collapse
|