151
|
Abstract
Neural crest stem/progenitor cells arise early during vertebrate embryogenesis at the border of the forming central nervous system. They subsequently migrate throughout the body, eventually differentiating into diverse cell types ranging from neurons and glia of the peripheral nervous system to bones of the face, portions of the heart, and pigmentation of the skin. Along the body axis, the neural crest is heterogeneous, with different subpopulations arising in the head, neck, trunk, and tail regions, each characterized by distinct migratory patterns and developmental potential. Modern genomic approaches like single-cell RNA- and ATAC-sequencing (seq) have greatly enhanced our understanding of cell lineage trajectories and gene regulatory circuitry underlying the developmental progression of neural crest cells. Here, we discuss how genomic approaches have provided new insights into old questions in neural crest biology by elucidating transcriptional and posttranscriptional mechanisms that govern neural crest formation and the establishment of axial level identity. Expected final online publication date for the Annual Review of Genetics, Volume 55 is November 2021. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Shashank Gandhi
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91125, USA; ,
| | - Marianne E Bronner
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91125, USA; ,
| |
Collapse
|
152
|
Queiroz A, Pelissari C, Arana-Chavez VE, Trierveiler M. Temporo-spatial distribution of stem cell markers CD146 and p75NTR during odontogenesis in mice. J Appl Oral Sci 2021; 29:e20210138. [PMID: 34550167 PMCID: PMC8462488 DOI: 10.1590/1678-7757-2021-0138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 05/29/2021] [Accepted: 06/28/2021] [Indexed: 11/22/2022] Open
Abstract
Mesenchymal and epithelial stem cells were identified in dental tissues; however, knowledge about the odontogenic stem cells is limited, and there are some questions regarding their temporo-spatial dynamics in tooth development. OBJECTIVE Our study aimed to analyze the expression of the stem cell markers CD146 and p75NTR during the different stages of odontogenesis. METHODOLOGY The groups consisted of 13.5, 15.5, 17.5 days old embryos, and 14 days postnatal BALB/c mice. The expression of CD146 and p75NTR was evaluated by immunohistochemistry. RESULTS Our results showed that positive cells for both markers were present in all stages of tooth development, and the number of positive cells increased with the progression of this process. Cells of epithelial and ectomesenchymal origin were positive for CD146, and the expression of p75NTR was mainly detected in the dental papilla and dental follicle. In the postnatal group, dental pulp cells were positive for CD146, and the reduced enamel epithelium and the oral mucosa epithelium showed immunostaining for p75NTR. CONCLUSIONS These results suggest that the staining pattern of CD146 and p75NTR underwent temporal and spatial changes during odontogenesis and both markers were expressed by epithelial and mesenchymal cell types, which is relevant due to the significance of the epithelial-ectomesenchymal interactions in tooth development.
Collapse
Affiliation(s)
- Aline Queiroz
- Universidade de São Paulo, Faculdade de Odontologia, Departamento de Estomatologia, Disciplina de Patologia Oral e Maxilofacial, Laboratório de Biologia de Células-Tronco em Odontologia LABITRON, São Paulo, SP, Brasil
| | - Cibele Pelissari
- Universidade de São Paulo, Faculdade de Odontologia, Departamento de Estomatologia, Disciplina de Patologia Oral e Maxilofacial, Laboratório de Biologia de Células-Tronco em Odontologia LABITRON, São Paulo, SP, Brasil
| | - Victor Elias Arana-Chavez
- Universidade de São Paulo, Faculdade de Odontologia, Departamento de Biomateriais e Biologia Oral, São Paulo, SP, Brasil
| | - Marília Trierveiler
- Universidade de São Paulo, Faculdade de Odontologia, Departamento de Estomatologia, Disciplina de Patologia Oral e Maxilofacial, Laboratório de Biologia de Células-Tronco em Odontologia LABITRON, São Paulo, SP, Brasil
| |
Collapse
|
153
|
Thiha P, Higashihori N, Kano S, Moriyama K. Histone methyltransferase SET domain bifurcated 1 negatively regulates parathyroid hormone/parathyroid hormone-related peptide receptor to control chondrocyte proliferation in Meckel's cartilage. Arch Oral Biol 2021; 131:105251. [PMID: 34521010 DOI: 10.1016/j.archoralbio.2021.105251] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 07/15/2021] [Accepted: 08/27/2021] [Indexed: 10/20/2022]
Abstract
OBJECTIVE The aim of this study is to show that the proliferation of chondrocytes is regulated by SET domain bifurcated 1 (SETDB1) along with the downregulation of parathyroid hormone (PTH)/parathyroid hormone-related peptide (PTHrP) receptor in Meckel's cartilage. DESIGN Setdb1 was knocked down or overexpressed in a mouse chondrogenic ATDC5 cells, by transfecting the cells with short interfering RNA against Setdb1 or wild-type Setdb1 expression vector, respectively. Cell proliferation was detected by bromodeoxyuridine incorporation. Setdb1 was conditionally deleted in neural crest cells with Wnt1-Cre (Setdb1 conditional knockout mice). Immunofluorescence staining of paraffin sections of embryonic days 13.5 and 14.5 Setdb1 conditional knockout mice or transfected ATDC5 cells was performed to detect PTH/PTHrP receptor. Protein kinase B (AKT) phosphorylation inhibitor was added to both siRNA-transfected ATDC5 cultures to determine whether AKT activation induces PTH/PTHrP receptor expression after Setdb1 knockdown or vice versa. RESULTS Setdb1 knockdown in ATDC5 cells showed increased cell proliferation and parathyroid hormone receptor 1 expression. Contrasting results were observed in the Setdb1-overexpressed wild-type cells. Immunofluorescence staining showed the highly expressed PTH/PTHrP receptor in Setdb1-knocked down ATDC5 cells and in the chondrocytes of Setdb1 conditional knockout embryonic Meckel's cartilage, indicating the negative regulation of SETDB1 on PTH/PTHrP receptor. Strong staining of phosphorylated AKT was observed in Setdb1-knocked down ATDC5 cells. However, the inhibition of AKT phosphorylation significantly reduced both the PTH/PTHrP receptor staining and the Setdb1-knockdown-induced increase in ATDC5 cell proliferation. CONCLUSIONS Our findings contribute new insights on SETDB1 function in relation with AKT and PTH/PTHrP receptor during chondrocyte proliferation.
Collapse
Affiliation(s)
- Phyo Thiha
- Maxillofacial Orthognathics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Japan
| | - Norihisa Higashihori
- Maxillofacial Orthognathics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Japan.
| | - Sakurako Kano
- Maxillofacial Orthognathics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Japan
| | - Keiji Moriyama
- Maxillofacial Orthognathics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Japan
| |
Collapse
|
154
|
Kundrát M. Earliest migratory cephalic NC cells are potent to differentiate into dental ectomesenchyme of the two lungfish dentitions: tetrapodomorph ancestral condition of unconstrained capability of mesencephalic NC cells to form oral teeth. Naturwissenschaften 2021; 108:37. [PMID: 34448941 DOI: 10.1007/s00114-021-01750-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 05/29/2021] [Accepted: 08/16/2021] [Indexed: 11/29/2022]
Abstract
Reciprocal interactions between epithelial and neural crest-derived mesenchymal cells have been recognized in the evolutionary modulation of tetrapod odontodes, skeletal structures that include the teeth and tooth-integrated basal tissue. Using cell-tracking experiments, it has been demonstrated that mandibular neural crest cells, labelled during migration, extensively populate dental papillae of all tooth phenotypes of the lobe-finned fish, the Australian lungfish (Neoceratodus forsteri). Here, I report on an extension of this experimental study that earliest migrating NC cells are able to differentiate into odontogenic ectomesenchyme. Using vital dye cell-tracking to mark the mesencephalic neural crest prior to migration, I have found that the corresponding population of earliest migratory cells selectively relocated to dental papillae of both temporary and permanent dentitions of Neoceratodus. I noticed a gradient in distribution of the labelled cells which populated posterior teeth, pterygoid and prearticular (including associated trabecular and Meckelian cartilages; major relocation) much more densely than those in anterior marginal positions, temporary and vomeral permanent teeth (minor relocation). Contrary to mice and zebrafish, the odontogenic potency of mesencephalic neural crest cells is already programmed at the onset of the migration event in lungfish. This may imply that the morphogenic potential of mesencephalic neural crest cells to form teeth has been heterochronically shifted and constrained to later migratory populations of neural crest cells during the developmental evolution of derived tetrapods, or/and arrested in their expression in the oral development of some modern osteichthyans.
Collapse
Affiliation(s)
- Martin Kundrát
- Evolutionary Biodiversity Research Group, PaleoBioImaging Lab, Center for Interdisciplinary Biosciences, Technology and Innovation Park, Pavol Jozef Šafárik University, Jesenná 5, Košice, 04154, Slovak Republic.
| |
Collapse
|
155
|
Ide Y, Nakahara T, Fukada T, Nasu M. Local Irradiation of Mouse Tooth Germ Gives Insight into the Direct Effects of Irradiation on Root Development. Radiat Res 2021; 196:602-610. [PMID: 34388821 DOI: 10.1667/rade-20-00081.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 07/27/2021] [Indexed: 11/03/2022]
Abstract
To elucidate the mechanism underlying the failure of root formation after irradiation, we established a method of local irradiation of the molar tooth germ and demonstrated that radiation directly affected dental root development. In the current study, to locally irradiate the lower first molars of 5-day-old C57BL/6J mice, we used lead glass containing a hole as a collimator. We confirmed that our local irradiation method targeted only the tooth germ. The irradiated root was immature in terms of apical growth, and dentin formation was irregular along the outside of the root apices. Moreover, calcified tissue apically surrounded Hertwig's epithelial root sheath, which disappeared abnormally early. This method using a local irradiation experimental model will facilitate research into radiation-induced disorders of dental root formation.
Collapse
Affiliation(s)
- Yoshiaki Ide
- Department of Anatomy, The Nippon Dental University School of Life Dentistry at Tokyo.,Department of Developmental and Regenerative Dentistry, The Nippon Dental University School of Life Dentistry at Tokyo
| | - Taka Nakahara
- Department of Developmental and Regenerative Dentistry, The Nippon Dental University School of Life Dentistry at Tokyo
| | - Tetsuya Fukada
- Department of Pharmacology, The Nippon Dental University School of Life Dentistry at Tokyo
| | - Masanori Nasu
- Research Center for Odontology, The Nippon Dental University School of Life Dentistry at Tokyo
| |
Collapse
|
156
|
Dasgupta K, Cesario JM, Ha S, Asam K, Deacon LJ, Song AH, Kim J, Cobb J, Yoon JK, Jeong J. R-Spondin 3 Regulates Mammalian Dental and Craniofacial Development. J Dev Biol 2021; 9:jdb9030031. [PMID: 34449628 PMCID: PMC8395884 DOI: 10.3390/jdb9030031] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 08/06/2021] [Accepted: 08/07/2021] [Indexed: 12/14/2022] Open
Abstract
Development of the teeth requires complex signaling interactions between the mesenchyme and the epithelium mediated by multiple pathways. For example, canonical WNT signaling is essential to many aspects of odontogenesis, and inhibiting this pathway blocks tooth development at an early stage. R-spondins (RSPOs) are secreted proteins, and they mostly augment WNT signaling. Although RSPOs have been shown to play important roles in the development of many organs, their role in tooth development is unclear. A previous study reported that mutating Rspo2 in mice led to supernumerary lower molars, while teeth forming at the normal positions showed no significant anomalies. Because multiple Rspo genes are expressed in the orofacial region, it is possible that the relatively mild phenotype of Rspo2 mutants is due to functional compensation by other RSPO proteins. We found that inactivating Rspo3 in the craniofacial mesenchyme caused the loss of lower incisors, which did not progress beyond the bud stage. A simultaneous deletion of Rspo2 and Rspo3 caused severe disruption of craniofacial development from early stages, which was accompanied with impaired development of all teeth. Together, these results indicate that Rspo3 is an important regulator of mammalian dental and craniofacial development.
Collapse
Affiliation(s)
- Krishnakali Dasgupta
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA; (K.D.); (J.M.C.); (S.H.); (K.A.); (L.J.D.); (A.H.S.); (J.K.)
| | - Jeffry M. Cesario
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA; (K.D.); (J.M.C.); (S.H.); (K.A.); (L.J.D.); (A.H.S.); (J.K.)
| | - Sara Ha
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA; (K.D.); (J.M.C.); (S.H.); (K.A.); (L.J.D.); (A.H.S.); (J.K.)
| | - Kesava Asam
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA; (K.D.); (J.M.C.); (S.H.); (K.A.); (L.J.D.); (A.H.S.); (J.K.)
| | - Lindsay J. Deacon
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA; (K.D.); (J.M.C.); (S.H.); (K.A.); (L.J.D.); (A.H.S.); (J.K.)
| | - Ana H. Song
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA; (K.D.); (J.M.C.); (S.H.); (K.A.); (L.J.D.); (A.H.S.); (J.K.)
| | - Julie Kim
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA; (K.D.); (J.M.C.); (S.H.); (K.A.); (L.J.D.); (A.H.S.); (J.K.)
| | - John Cobb
- Department of Biological Sciences, University of Calgary, Calgary, AB T2N 1N4, Canada;
| | - Jeong Kyo Yoon
- Soonchunhyang Institute of Medi-Bio Science, Soonchunhyang University, Cheonan 31151, Korea;
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan 31151, Korea
| | - Juhee Jeong
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA; (K.D.); (J.M.C.); (S.H.); (K.A.); (L.J.D.); (A.H.S.); (J.K.)
- Correspondence:
| |
Collapse
|
157
|
Alotaibi RN, Howe BJ, Chernus JM, Mukhopadhyay N, Sanchez C, Deleyiannis FWB, Neiswanger K, Padilla C, Poletta FA, Orioli IM, Buxó CJ, Hecht JT, Wehby GL, Long RE, Vieira AR, Weinberg SM, Shaffer JR, Moreno Uribe LM, Marazita ML. Genome-Wide Association Study (GWAS) of dental caries in diverse populations. BMC Oral Health 2021; 21:377. [PMID: 34311721 PMCID: PMC8311973 DOI: 10.1186/s12903-021-01670-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 05/30/2021] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Dental caries is one of the most common chronic diseases and is influenced by a complex interplay of genetic and environmental factors. Most previous genetic studies of caries have focused on identifying genes that contribute to dental caries in specific ethnic groups, usually of European descent. METHODS The aim of this study is to conduct a genome-wide association study (GWAS) to identify associations affecting susceptibility to caries in a large multiethnic population from Argentina, the Philippines, Guatemala, Hungary, and the USA, originally recruited for studies of orofacial clefts (POFC, N = 3686). Ages of the participants ranged from 2 to 12 years for analysis of the primary dentition, and 18-60 years for analysis of the permanent dentition. For each participant, dental caries was assessed by counts of decayed and filled teeth (dft/DFT) and genetic variants (single nucleotide polymorphisms, SNPs) were genotyped or imputed across the entire genome. Caries was analyzed separately for the primary and permanent dentitions, with age, gender, and presence/absence of any type of OFC treated as covariates. Efficient Mixed-Model Association eXpedited (EMMAX) was used to test genetic association, while simultaneously accounting for relatedness and stratification. RESULTS We identified several suggestive loci (5 × 10-8 < P < 5 × 10-6) within or near genes with plausible biological roles for dental caries, including a cluster of taste receptor genes (TAS2R38, TAS2R3, TAS2R4, TASR25) on chromosome 7 for the permanent dentition analysis, and DLX3 and DLX4 on chromosome 17 for the primary dentition analysis. Genome-wide significant results were seen with SNPs in the primary dentition only; however, none of the identified genes near these variants have known roles in cariogenesis. CONCLUSION The results of this study warrant further investigation and may lead to a better understanding of cariogenesis in diverse populations, and help to improve dental caries prediction, prevention, and/or treatment in future.
Collapse
Affiliation(s)
- Rasha N Alotaibi
- Department of Oral and Craniofacial Sciences, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
- Center for Craniofacial and Dental Genetics, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
- Dental Health Department, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia.
| | - Brian J Howe
- Department of Family Dentistry, College of Dentistry, University of Iowa, Iowa City, IA, USA
- The Iowa Center for Oral Health Research, College of Dentistry, University of Iowa, Iowa City, IA, USA
| | - Jonathan M Chernus
- Department of Oral and Craniofacial Sciences, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Nandita Mukhopadhyay
- Department of Oral and Craniofacial Sciences, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Craniofacial and Dental Genetics, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Carla Sanchez
- Department of Oral and Craniofacial Sciences, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Craniofacial and Dental Genetics, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Katherine Neiswanger
- Department of Oral and Craniofacial Sciences, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Craniofacial and Dental Genetics, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Carmencita Padilla
- Department of Pediatrics, College of Medicine, University of the Philippines, Manila, Philippines
| | - Fernando A Poletta
- ECLAMC/INAGEMP At Center for Medical Education and Clinical Research (CEMIC-CONICET), Buenos Aires, Argentina
| | - Ieda M Orioli
- Department of Genetics, Institute of Biology, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Carmen J Buxó
- Dental and Craniofacial Genomics Core, School of Dental Medicine, University of Puerto Rico, San Juan, PR, USA
| | - Jacqueline T Hecht
- Department of Pediatrics, University of Texas Health Science Center At Houston, Houston, TX, USA
| | - George L Wehby
- Department of Health Management and Policy, College of Public Health, University of Iowa, Iowa City, IA, USA
| | - Ross E Long
- Lancaster Cleft Palate Clinic, Lancaster, PA, USA
| | - Alexandre R Vieira
- Department of Oral and Craniofacial Sciences, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Seth M Weinberg
- Department of Oral and Craniofacial Sciences, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Craniofacial and Dental Genetics, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - John R Shaffer
- Department of Oral and Craniofacial Sciences, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Craniofacial and Dental Genetics, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Lina M Moreno Uribe
- Department of Family Dentistry, College of Dentistry, University of Iowa, Iowa City, IA, USA
- Department of Orthodontics, School of Dentistry, University of Iowa, Iowa City, IA, USA
| | - Mary L Marazita
- Department of Oral and Craniofacial Sciences, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Craniofacial and Dental Genetics, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
158
|
Wang Q, Xu L, Miura J, Saha MK, Uemura Y, Sandell LL, Trainor PA, Yamashiro T, Kurosaka H. Branchiomeric Muscle Development Requires Proper Retinoic Acid Signaling. Front Cell Dev Biol 2021; 9:596838. [PMID: 34307338 PMCID: PMC8299418 DOI: 10.3389/fcell.2021.596838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 05/12/2021] [Indexed: 11/30/2022] Open
Abstract
The first and second branchiomeric (branchial arch) muscles are craniofacial muscles that derive from branchial arch mesoderm. In mammals, this set of muscles is indispensable for jaw movement and facial expression. Defects during embryonic development that result in congenital partial absence of these muscles can have significant impact on patients’ quality of life. However, the detailed molecular and cellular mechanisms that regulate branchiomeric muscle development remains poorly understood. Herein we investigated the role of retinoic acid (RA) signaling in developing branchiomeric muscles using mice as a model. We administered all-trans RA (25 mg/kg body weight) to Institute of Cancer Research (ICR) pregnant mice by gastric intubation from E8.5 to E10.5. In their embryos at E13.5, we found that muscles derived from the first branchial arch (temporalis, masseter) and second branchial arch (frontalis, orbicularis oculi) were severely affected or undetectable, while other craniofacial muscles were hypoplastic. We detected elevated cell death in the branchial arch mesoderm cells in RA-treated embryos, suggesting that excessive RA signaling reduces the survival of precursor cells of branchiomeric muscles, resulting in the development of hypoplastic craniofacial muscles. In order to uncover the signaling pathway(s) underlying this etiology, we focused on Pitx2, Tbx1, and MyoD1, which are critical for cranial muscle development. Noticeably reduced expression of all these genes was detected in the first and second branchial arch of RA-treated embryos. Moreover, elevated RA signaling resulted in a reduction in Dlx5 and Dlx6 expression in cranial neural crest cells (CNCCs), which disturbed their interactions with branchiomeric mesoderm cells. Altogether, we discovered that embryonic craniofacial muscle defects caused by excessive RA signaling were associated with the downregulation of Pitx2, Tbx1, MyoD1, and Dlx5/6, and reduced survival of cranial myogenic precursor cells.
Collapse
Affiliation(s)
- Qi Wang
- Department of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Osaka University, Suita, Japan.,The Affiliated Stomatology Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Oral Biomedical Research of Zhejiang Province, Zhejiang University School of Stomatology, Hangzhou, China
| | - Lin Xu
- Department of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Osaka University, Suita, Japan
| | - Jiro Miura
- Division for Interdisciplinary Dentistry, Osaka University Graduate School of Dentistry, Suita, Japan
| | - Mithun Kumar Saha
- Department of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Osaka University, Suita, Japan
| | - Yume Uemura
- Department of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Osaka University, Suita, Japan
| | - Lisa L Sandell
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, KY, United States
| | - Paul A Trainor
- Stowers Institute for Medical Research, Kansas City, MO, United States.,Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Takashi Yamashiro
- Department of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Osaka University, Suita, Japan
| | - Hiroshi Kurosaka
- Department of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Osaka University, Suita, Japan
| |
Collapse
|
159
|
Function of Dental Follicle Progenitor/Stem Cells and Their Potential in Regenerative Medicine: From Mechanisms to Applications. Biomolecules 2021; 11:biom11070997. [PMID: 34356621 PMCID: PMC8301812 DOI: 10.3390/biom11070997] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/04/2021] [Accepted: 07/04/2021] [Indexed: 02/06/2023] Open
Abstract
Dental follicle progenitor/stem cells (DFPCs) are a group of dental mesenchyme stem cells that lie in the dental follicle and play a critical role in tooth development and maintaining function. Originating from neural crest, DFPCs harbor a multipotential differentiation capacity. More importantly, they have superiorities, including the easy accessibility and abundant sources, active self-renewal ability and noncontroversial sources compared with other stem cells, making them an attractive candidate in the field of tissue engineering. Recent advances highlight the excellent properties of DFPCs in regeneration of orofacial tissues, including alveolar bone repair, periodontium regeneration and bio-root complex formation. Furthermore, they play a unique role in maintaining a favorable microenvironment for stem cells, immunomodulation and nervous related tissue regeneration. This review is intended to summarize the current knowledge of DFPCs, including their stem cell properties, physiological functions and clinical application potential. A deep understanding of DFPCs can thus inspire novel perspectives in regenerative medicine in the future.
Collapse
|
160
|
Borgmann D, Ciglieri E, Biglari N, Brandt C, Cremer AL, Backes H, Tittgemeyer M, Wunderlich FT, Brüning JC, Fenselau H. Gut-brain communication by distinct sensory neurons differently controls feeding and glucose metabolism. Cell Metab 2021; 33:1466-1482.e7. [PMID: 34043943 PMCID: PMC8280952 DOI: 10.1016/j.cmet.2021.05.002] [Citation(s) in RCA: 107] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 02/23/2021] [Accepted: 05/03/2021] [Indexed: 01/26/2023]
Abstract
Sensory neurons relay gut-derived signals to the brain, yet the molecular and functional organization of distinct populations remains unclear. Here, we employed intersectional genetic manipulations to probe the feeding and glucoregulatory function of distinct sensory neurons. We reconstruct the gut innervation patterns of numerous molecularly defined vagal and spinal afferents and identify their downstream brain targets. Bidirectional chemogenetic manipulations, coupled with behavioral and circuit mapping analysis, demonstrated that gut-innervating, glucagon-like peptide 1 receptor (GLP1R)-expressing vagal afferents relay anorexigenic signals to parabrachial nucleus neurons that control meal termination. Moreover, GLP1R vagal afferent activation improves glucose tolerance, and their inhibition elevates blood glucose levels independent of food intake. In contrast, gut-innervating, GPR65-expressing vagal afferent stimulation increases hepatic glucose production and activates parabrachial neurons that control normoglycemia, but they are dispensable for feeding regulation. Thus, distinct gut-innervating sensory neurons differentially control feeding and glucoregulatory neurocircuits and may provide specific targets for metabolic control.
Collapse
Affiliation(s)
- Diba Borgmann
- Synaptic Transmission in Energy Homeostasis Group, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany; Translational Neurocircuitry Group, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany; Center for Anatomy II, Neuroanatomy, University Hospital Cologne, Joseph-Stelzmann Str. 9, 50937 Cologne, Germany
| | - Elisa Ciglieri
- Synaptic Transmission in Energy Homeostasis Group, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany; Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Strasse 26, 50937 Cologne, Germany
| | - Nasim Biglari
- Max Planck Institute for Metabolism Research, Department of Neuronal Control of Metabolism, Gleueler Strasse 50, 50931 Cologne, Germany
| | - Claus Brandt
- Max Planck Institute for Metabolism Research, Department of Neuronal Control of Metabolism, Gleueler Strasse 50, 50931 Cologne, Germany
| | - Anna Lena Cremer
- Max Planck Institute for Metabolism Research, Department of Neuronal Control of Metabolism, Gleueler Strasse 50, 50931 Cologne, Germany
| | - Heiko Backes
- Max Planck Institute for Metabolism Research, Department of Neuronal Control of Metabolism, Gleueler Strasse 50, 50931 Cologne, Germany
| | - Marc Tittgemeyer
- Translational Neurocircuitry Group, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), University of Cologne, Joseph-Stelzmann-Straße 26, Cologne 50931, Germany
| | - F Thomas Wunderlich
- Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Strasse 26, 50937 Cologne, Germany; Max Planck Institute for Metabolism Research, Department of Neuronal Control of Metabolism, Gleueler Strasse 50, 50931 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), University of Cologne, Joseph-Stelzmann-Straße 26, Cologne 50931, Germany; Center of Molecular Medicine Cologne (CMMC), University of Cologne, Robert-Koch-Straße 21, 50931 Cologne, Germany
| | - Jens C Brüning
- Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Strasse 26, 50937 Cologne, Germany; Max Planck Institute for Metabolism Research, Department of Neuronal Control of Metabolism, Gleueler Strasse 50, 50931 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), University of Cologne, Joseph-Stelzmann-Straße 26, Cologne 50931, Germany; Center of Molecular Medicine Cologne (CMMC), University of Cologne, Robert-Koch-Straße 21, 50931 Cologne, Germany
| | - Henning Fenselau
- Synaptic Transmission in Energy Homeostasis Group, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany; Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Strasse 26, 50937 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), University of Cologne, Joseph-Stelzmann-Straße 26, Cologne 50931, Germany.
| |
Collapse
|
161
|
Zhang Z, Oh M, Sasaki JI, Nör JE. Inverse and reciprocal regulation of p53/p21 and Bmi-1 modulates vasculogenic differentiation of dental pulp stem cells. Cell Death Dis 2021; 12:644. [PMID: 34168122 PMCID: PMC8225874 DOI: 10.1038/s41419-021-03925-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 06/10/2021] [Accepted: 06/11/2021] [Indexed: 12/20/2022]
Abstract
Dental pulp stem cells (DPSC) are capable of differentiating into vascular endothelial cells. Although the capacity of vascular endothelial growth factor (VEGF) to induce endothelial differentiation of stem cells is well established, mechanisms that maintain stemness and prevent vasculogenic differentiation remain unclear. Here, we tested the hypothesis that p53 signaling through p21 and Bmi-1 maintains stemness and inhibits vasculogenic differentiation. To address this hypothesis, we used primary human DPSC from permanent teeth and Stem cells from Human Exfoliated Deciduous (SHED) teeth as models of postnatal mesenchymal stem cells. DPSC seeded in biodegradable scaffolds and transplanted into immunodeficient mice generated mature human blood vessels invested with smooth muscle actin-positive mural cells. Knockdown of p53 was sufficient to induce vasculogenic differentiation of DPSC (without vasculogenic differentiation medium containing VEGF), as shown by increased expression of endothelial markers (VEGFR2, Tie-2, CD31, VE-cadherin), increased capillary sprouting in vitro; and increased DPSC-derived blood vessel density in vivo. Conversely, induction of p53 expression with small molecule inhibitors of the p53-MDM2 binding (MI-773, APG-115) was sufficient to inhibit VEGF-induced vasculogenic differentiation. Considering that p21 is a major downstream effector of p53, we knocked down p21 in DPSC and observed an increase in capillary sprouting that mimicked results observed when p53 was knocked down. Stabilization of ubiquitin activity was sufficient to induce p53 and p21 expression and reduce capillary sprouting. Interestingly, we observed an inverse and reciprocal correlation between p53/p21 and the expression of Bmi-1, a major regulator of stem cell self-renewal. Further, direct inhibition of Bmi-1 with PTC-209 resulted in blockade of capillary-like sprout formation. Collectively, these data demonstrate that p53/p21 functions through Bmi-1 to prevent the vasculogenic differentiation of DPSC.
Collapse
Affiliation(s)
- Zhaocheng Zhang
- Angiogenesis Research Laboratory, Department of Cariology, Restorative Sciences and Endodontics, University of Michigan School of Dentistry, Ann Arbor, MI, 48109, USA
| | - Min Oh
- Angiogenesis Research Laboratory, Department of Cariology, Restorative Sciences and Endodontics, University of Michigan School of Dentistry, Ann Arbor, MI, 48109, USA
| | - Jun-Ichi Sasaki
- Angiogenesis Research Laboratory, Department of Cariology, Restorative Sciences and Endodontics, University of Michigan School of Dentistry, Ann Arbor, MI, 48109, USA
| | - Jacques E Nör
- Angiogenesis Research Laboratory, Department of Cariology, Restorative Sciences and Endodontics, University of Michigan School of Dentistry, Ann Arbor, MI, 48109, USA.
- Department of Biomedical Engineering, University of Michigan College of Engineering, Ann Arbor, MI, USA.
- Department of Otolaryngology, University of Michigan School of Medicine, Ann Arbor, MI, USA.
| |
Collapse
|
162
|
Influence of Dental Pulp Harvesting Method on the Viability and Differentiation Capacity of Adult Dental Pulp-Derived Mesenchymal Stem Cells. Stem Cells Int 2021; 2021:9952401. [PMID: 34239574 PMCID: PMC8238610 DOI: 10.1155/2021/9952401] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 06/04/2021] [Indexed: 01/09/2023] Open
Abstract
Objective To compare two pulp harvesting methods for stem cell expansion, namely, conservative pulpotomy and pulpectomy from exodontia. Method Ten freshly extracted sound third molars from five patients were selected. Five were used in the control group, where pulp harvesting was performed by exodontia and the remaining teeth were used in the test group, where the pulp was harvested by conservative pulpotomy (preserving the tooth). This was a split-mouth design study, where a third molar from one side was randomly allocated into the test group and the contralateral tooth in the control group. After pulp harvesting, the following evaluations were performed: cell morphology, sterility test, immunophenotyping, differentiation assays, first pass live cell counts, time to cryopreservation, and total number of expanded cells at the end of the fourth pass. Results Regarding morphology, the cells from both groups presented a fibroblastic phenotype. All samples were sterile. Immunophenotyping demonstrated a positive expression for CD105, CD90, and CD73 and negative expression for CD45 in both groups. Differentiation assays were positive for osteogenic and chondrogenic differentiation in both groups. Regarding live cell counts in the first passage, the control group had 95.8% live cells in the total count and the test group 91.2% (p < 0.05). The time required for cryopreservation was equivalent in both groups 51.6 days and 52.6 days, respectively (p > 0.05). The total number of cells at the end of the fourth passage was 5,286,782 and 5,736,862, respectively (p > 0.05). Conclusion These results suggest that adult stem cell harvesting from conservative pulpotomy is as effective as the traditional exodontia-based method.
Collapse
|
163
|
Kovacs CS, Chaussain C, Osdoby P, Brandi ML, Clarke B, Thakker RV. The role of biomineralization in disorders of skeletal development and tooth formation. Nat Rev Endocrinol 2021; 17:336-349. [PMID: 33948016 DOI: 10.1038/s41574-021-00488-z] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/19/2021] [Indexed: 02/03/2023]
Abstract
The major mineralized tissues are bone and teeth, which share several mechanisms governing their development and mineralization. This crossover includes the hormones that regulate circulating calcium and phosphate concentrations, and the genes that regulate the differentiation and transdifferentiation of cells. In developing endochondral bone and in developing teeth, parathyroid hormone-related protein (PTHrP) acts in chondrocytes to delay terminal differentiation, thereby increasing the pool of precursor cells. Chondrocytes and (in specific circumstances) pre-odontoblasts can also transdifferentiate into osteoblasts. Moreover, bone and teeth share outcomes when affected by systemic disorders of mineral homeostasis or of the extracellular matrix, and by adverse effects of treatments such as bisphosphonates and fluoride. Unlike bone, teeth have more permanent effects from systemic disorders because they are not remodelled after they are formed. This Review discusses the normal processes of bone and tooth development, followed by disorders that have effects on both bone and teeth, versus disorders that have effects in one without affecting the other. The takeaway message is that bone specialists should know when to screen for dental disorders, just as dental specialists should recognize when a tooth disorder should raise suspicions about a possible underlying bone disorder.
Collapse
Affiliation(s)
- Christopher S Kovacs
- Faculty of Medicine - Endocrinology, Memorial University of Newfoundland, St. John's, NL, Canada.
| | | | - Philip Osdoby
- Department of Biology, Washington University, St. Louis, MO, USA
| | - Maria Luisa Brandi
- Department of Biochemical, Experimental and Clinical Sciences, University of Florence, Florence, Italy
| | - Bart Clarke
- Mayo Clinic Division of Endocrinology, Diabetes, Metabolism, and Nutrition, Rochester, MN, USA
| | - Rajesh V Thakker
- Academic Endocrine Unit, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
164
|
Fitriasari S, Trainor PA. Diabetes, Oxidative Stress, and DNA Damage Modulate Cranial Neural Crest Cell Development and the Phenotype Variability of Craniofacial Disorders. Front Cell Dev Biol 2021; 9:644410. [PMID: 34095113 PMCID: PMC8174788 DOI: 10.3389/fcell.2021.644410] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 04/21/2021] [Indexed: 12/11/2022] Open
Abstract
Craniofacial malformations are among the most common birth defects in humans and they often have significant detrimental functional, aesthetic, and social consequences. To date, more than 700 distinct craniofacial disorders have been described. However, the genetic, environmental, and developmental origins of most of these conditions remain to be determined. This gap in our knowledge is hampered in part by the tremendous phenotypic diversity evident in craniofacial syndromes but is also due to our limited understanding of the signals and mechanisms governing normal craniofacial development and variation. The principles of Mendelian inheritance have uncovered the etiology of relatively few complex craniofacial traits and consequently, the variability of craniofacial syndromes and phenotypes both within families and between families is often attributed to variable gene expression and incomplete penetrance. However, it is becoming increasingly apparent that phenotypic variation is often the result of combinatorial genetic and non-genetic factors. Major non-genetic factors include environmental effectors such as pregestational maternal diabetes, which is well-known to increase the risk of craniofacial birth defects. The hyperglycemia characteristic of diabetes causes oxidative stress which in turn can result in genotoxic stress, DNA damage, metabolic alterations, and subsequently perturbed embryogenesis. In this review we explore the importance of gene-environment associations involving diabetes, oxidative stress, and DNA damage during cranial neural crest cell development, which may underpin the phenotypic variability observed in specific craniofacial syndromes.
Collapse
Affiliation(s)
| | - Paul A Trainor
- Stowers Institute for Medical Research, Kansas City, MO, United States.,Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, United States
| |
Collapse
|
165
|
Kim BJ, Zaveri HP, Kundert PN, Jordan VK, Scott TM, Carmichael J, Scott DA. RERE deficiency contributes to the development of orofacial clefts in humans and mice. Hum Mol Genet 2021; 30:595-602. [PMID: 33772547 DOI: 10.1093/hmg/ddab084] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/15/2021] [Accepted: 03/17/2021] [Indexed: 11/12/2022] Open
Abstract
Deletions of chromosome 1p36 are the most common telomeric deletions in humans and are associated with an increased risk of orofacial clefting. Deletion/phenotype mapping, combined with data from human and mouse studies, suggests the existence of multiple 1p36 genes associated with orofacial clefting including SKI, PRDM16, PAX7 and GRHL3. The arginine-glutamic acid dipeptide (RE) repeats gene (RERE) is located in the proximal critical region for 1p36 deletion syndrome and encodes a nuclear receptor co-regulator. Pathogenic RERE variants have been shown to cause neurodevelopmental disorder with or without anomalies of the brain, eye or heart (NEDBEH). Cleft lip has previously been described in one individual with NEDBEH. Here we report the first individual with NEDBEH to have a cleft palate. We confirm that RERE is broadly expressed in the palate during mouse embryonic development, and we demonstrate that the majority of RERE-deficient mouse embryos on C57BL/6 background have cleft palate. We go on to show that ablation of Rere in cranial neural crest (CNC) cells, mediated by a Wnt1-Cre, leads to delayed elevation of the palatal shelves and cleft palate and that proliferation of mesenchymal cells in the palatal shelves is significantly reduced in Rereflox/flox; Wnt1-Cre embryos. We conclude that loss of RERE function contributes to the development of orofacial clefts in individuals with proximal 1p36 deletions and NEDBEH and that RERE expression in CNC cells and their derivatives is required for normal palatal development.
Collapse
Affiliation(s)
- Bum Jun Kim
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Hitisha P Zaveri
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Peter N Kundert
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- Medical Scientist Training Program, Baylor College of Medicine, Houston, TX 77030, USA
| | - Valerie K Jordan
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Tiana M Scott
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT 84602, USA
| | - Jenny Carmichael
- LNR Genomic Medicine Service, Northampton General Hospital, Cliftonville, Northampton NN1 5BD, UK
| | - Daryl A Scott
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
166
|
Mizukoshi M, Kaku M, Thant L, Kitami K, Arai M, Saito I, Uoshima K. In vivo cell proliferation analysis and cell-tracing reveal the global cellular dynamics of periodontal ligament cells under mechanical-loading. Sci Rep 2021; 11:9813. [PMID: 33963224 PMCID: PMC8105403 DOI: 10.1038/s41598-021-89156-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 04/22/2021] [Indexed: 12/14/2022] Open
Abstract
Periodontal ligament (PDL) is a uniquely differentiated tissue that anchors the tooth to the alveolar bone socket and plays key roles in oral function. PDL cells can respond rapidly to mechanical stimuli, resulting in accelerated tissue remodeling. Cell proliferation is an initial event in tissue remodeling and participates in maintaining the cell supply; therefore, analyzing cell-proliferative activity might provide a comprehensive view of cellular dynamics at the tissue level. In this study, we investigated proliferating cells in mouse molar PDL during orthodontic tooth movement (OTM)-induced tissue remodeling. Our results demonstrated that the mechanical stimuli evoked a dynamic change in the proliferative-cell profile at the entire PDL. Additionally, cell-tracing analysis revealed that the proliferated cells underwent further division and subsequently contributed to tissue remodeling. Moreover, OTM-induced proliferating cells expressed various molecular markers that most likely arise from a wide range of cell types, indicating the lineage plasticity of PDL cells in vivo. Although further studies are required, these findings partially elucidated the global views of the cell trajectory in mouse molar PDL under mechanical-loading conditions, which is vital for understanding the cellular dynamics of the PDL and beneficial for dental treatment in humans.
Collapse
Affiliation(s)
- Masaru Mizukoshi
- Division of Orthodontics, Faculty of Dentistry and Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Masaru Kaku
- Division of Bio-Prosthodontics, Faculty of Dentistry and Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan.
| | - Lay Thant
- Division of Orthodontics, Faculty of Dentistry and Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Kohei Kitami
- Division of Orthodontics, Faculty of Dentistry and Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Moe Arai
- Division of Orthodontics, Faculty of Dentistry and Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Isao Saito
- Division of Orthodontics, Faculty of Dentistry and Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Katsumi Uoshima
- Division of Bio-Prosthodontics, Faculty of Dentistry and Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| |
Collapse
|
167
|
Huang CY, Vesvoranan O, Yin X, Montoya A, Londono V, Sawatari Y, Garcia-Godoy F. Anti-Inflammatory Effects of Conditioned Medium of Periodontal Ligament-Derived Stem Cells on Chondrocytes, Synoviocytes, and Meniscus Cells. Stem Cells Dev 2021; 30:537-547. [PMID: 33757298 DOI: 10.1089/scd.2021.0010] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Osteoarthritis (OA) is the most common type of arthritis, afflicting millions of people in the world. Elevation of inflammatory mediators and enzymatic matrix destruction is often associated with OA. Therefore, the objective of this study was to investigate the effects of conditioned medium from periodontal ligament-derived stem cells (PDLSCs) on inflammatory and catabolic gene expressions of chondrocytes, synoviocytes, and meniscus cells under in vitro inflammatory condition. Stem cells were isolated from human periodontal ligaments. Conditioned medium was collected and concentrated 20 × . Chondrocytes, synoviocytes, and meniscus cells were isolated from pig knees and divided into four experimental groups: serum-free media, serum-free media+interleukin-1β (IL-1β) (10 ng/mL), conditioned media (CM), and CM+IL-1β. Protein content and extracellular vesicle (EV) miRNAs of CM were analyzed by liquid chromatography-tandem mass spectrometry and RNA sequencing, respectively. It was found that the IL-1β treatment upregulated the expression of IL-1β, tumor necrosis factor-α (TNF-α), MMP-13, and ADAMTS-4 genes in the three cell types, whereas PDLSC-conditioned medium prevented the upregulation of gene expression by IL-1β in all three cell types. This study also found that there was consistency in anti-inflammatory effects of PDLSC CM across donors and cell subcultures, while PDLSCs released several anti-inflammatory factors and EV miRNAs at high levels. OA has been suggested as an inflammatory disease in which all intrasynovial tissues are involved. PDLSC-conditioned medium is a cocktail of trophic factors and EV miRNAs that could mediate different inflammatory processes in various tissues in the joint. Introducing PDLSC-conditioned medium to osteoarthritic joints could be a potential treatment to prevent OA progression by inhibiting inflammation.
Collapse
Affiliation(s)
- Chun-Yuh Huang
- Department of Biomedical Engineering, University of Miami, Coral Gables, Florida, USA
| | - Oraya Vesvoranan
- Department of Biomedical Engineering, University of Miami, Coral Gables, Florida, USA
| | - Xue Yin
- Department of Biomedical Engineering, University of Miami, Coral Gables, Florida, USA
| | - Amanda Montoya
- Department of Biomedical Engineering, University of Miami, Coral Gables, Florida, USA
| | - Valeria Londono
- Department of Biomedical Engineering, University of Miami, Coral Gables, Florida, USA
| | - Yoh Sawatari
- Division of Oral and Maxillofacial Surgery, University of Miami/Jackson Memorial Hospital, Miami, Florida, USA
| | - Franklin Garcia-Godoy
- Department of Bioscience Research, College of Dentistry, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| |
Collapse
|
168
|
Association of Polymorphic and Haplotype Variants of the MSX1 Gene and the Impacted Teeth Phenomenon. Genes (Basel) 2021; 12:genes12040577. [PMID: 33923458 PMCID: PMC8073557 DOI: 10.3390/genes12040577] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/10/2021] [Accepted: 04/14/2021] [Indexed: 01/14/2023] Open
Abstract
It is known that genetic factors determine odontogenesis; furthermore, studies have revealed that various genes in humans can regulate the development of different types and generations of teeth. In this study it has been assumed that tooth impaction—at least to some extent—also depends on the presence of specific genetic markers, especially allelic variants of the MSX1 gene. The primary objective of the study was to evaluate the suitability of selected molecular markers located within the MSX1 gene for the determination of the risk of tooth impaction in particular patients. The study participants were divided into two groups: (1) the study group—at least one secondary tooth was impacted in the jaws; (2) the control group—no impacted tooth in the jaws. Real-Time PCR and TaqMan probes were used to detect selected polymorphisms in the analyzed genes. Two single nucleotide polymorphisms of MSX1 were analyzed. After the two subgroups of patients were distinguished in the study group based on the number of impacted teeth, statistically significant differences in the frequency of genotypes described for rs12532 in the MSX1 gene were found.
Collapse
|
169
|
Xie X, Xu C, Zhao H, Wang J, Feng JQ. A Biphasic Feature of Gli1 +-Mesenchymal Progenitors during Cementogenesis That Is Positively Controlled by Wnt/β-Catenin Signaling. J Dent Res 2021; 100:1289-1298. [PMID: 33853427 DOI: 10.1177/00220345211007429] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Cementum, a specialized bony layer covering an entire molar root surface, anchors teeth into alveolar bone. Gli1, a key transcriptional activator in Hedgehog signaling, has been identified as a mesenchymal progenitor cell marker in various tissues, including the periodontal ligament (PDL). To address the mechanisms by which Gli1+ progenitor cells contribute to cementogenesis, we used the Gli1lacZ/+ knock-in line to mark Gli1+ progenitors and the Gli1CreERT2/+; R26RtdTomato/+ line (named Gli1Lin) to trace Gli1 progeny cells during cementogenesis. Our data unexpectedly displayed a biphasic feature of Gli1+ PDL progenitor cells and cementum growth: a negative relationship between Gli1+ progenitor cell number and cementogenesis but a positive correlation between Gli1-derived acellular and cellular cementoblast cell number and cementum growth. DTA-ablation of Gli1Lin cells led to a cementum hypoplasia, including a significant reduction of both acellular and cellular cementoblast cells. Gain-of-function studies (by constitutive stabilization of β-catenin in Gli1Lin cells) revealed a cementum hyperplasia. A loss of function (by conditional deletion of β-catenin in Gli1+ cells) resulted in a reduction of postnatal cementum growth. Together, our studies support a vital role of Gli1+ progenitor cells in contribution to both types of cementum, in which canonical Wnt/β-catenin signaling positively regulates the differentiation of Gli1+ progenitors to cementoblasts during cementogenesis.
Collapse
Affiliation(s)
- X Xie
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX, USA.,State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - C Xu
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX, USA.,State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - H Zhao
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX, USA
| | - J Wang
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX, USA.,State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - J Q Feng
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX, USA
| |
Collapse
|
170
|
Höving AL, Windmöller BA, Knabbe C, Kaltschmidt B, Kaltschmidt C, Greiner JFW. Between Fate Choice and Self-Renewal-Heterogeneity of Adult Neural Crest-Derived Stem Cells. Front Cell Dev Biol 2021; 9:662754. [PMID: 33898464 PMCID: PMC8060484 DOI: 10.3389/fcell.2021.662754] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 03/18/2021] [Indexed: 12/16/2022] Open
Abstract
Stem cells of the neural crest (NC) vitally participate to embryonic development, but also remain in distinct niches as quiescent neural crest-derived stem cell (NCSC) pools into adulthood. Although NCSC-populations share a high capacity for self-renewal and differentiation resulting in promising preclinical applications within the last two decades, inter- and intrapopulational differences exist in terms of their expression signatures and regenerative capability. Differentiation and self-renewal of stem cells in developmental and regenerative contexts are partially regulated by the niche or culture condition and further influenced by single cell decision processes, making cell-to-cell variation and heterogeneity critical for understanding adult stem cell populations. The present review summarizes current knowledge of the cellular heterogeneity within NCSC-populations located in distinct craniofacial and trunk niches including the nasal cavity, olfactory bulb, oral tissues or skin. We shed light on the impact of intrapopulational heterogeneity on fate specifications and plasticity of NCSCs in their niches in vivo as well as during in vitro culture. We further discuss underlying molecular regulators determining fate specifications of NCSCs, suggesting a regulatory network including NF-κB and NC-related transcription factors like SLUG and SOX9 accompanied by Wnt- and MAPK-signaling to orchestrate NCSC stemness and differentiation. In summary, adult NCSCs show a broad heterogeneity on the level of the donor and the donors' sex, the cell population and the single stem cell directly impacting their differentiation capability and fate choices in vivo and in vitro. The findings discussed here emphasize heterogeneity of NCSCs as a crucial parameter for understanding their role in tissue homeostasis and regeneration and for improving their applicability in regenerative medicine.
Collapse
Affiliation(s)
- Anna L. Höving
- Department of Cell Biology, University of Bielefeld, Bielefeld, Germany
- Institute for Laboratory- and Transfusion Medicine, Heart and Diabetes Centre North Rhine-Westphalia (NRW), Ruhr University Bochum, Bad Oeynhausen, Germany
| | - Beatrice A. Windmöller
- Department of Cell Biology, University of Bielefeld, Bielefeld, Germany
- Forschungsverbund BioMedizin Bielefeld FBMB e.V., Bielefeld, Germany
| | - Cornelius Knabbe
- Institute for Laboratory- and Transfusion Medicine, Heart and Diabetes Centre North Rhine-Westphalia (NRW), Ruhr University Bochum, Bad Oeynhausen, Germany
- Forschungsverbund BioMedizin Bielefeld FBMB e.V., Bielefeld, Germany
| | - Barbara Kaltschmidt
- Department of Cell Biology, University of Bielefeld, Bielefeld, Germany
- Forschungsverbund BioMedizin Bielefeld FBMB e.V., Bielefeld, Germany
- Molecular Neurobiology, University of Bielefeld, Bielefeld, Germany
| | - Christian Kaltschmidt
- Department of Cell Biology, University of Bielefeld, Bielefeld, Germany
- Forschungsverbund BioMedizin Bielefeld FBMB e.V., Bielefeld, Germany
| | - Johannes F. W. Greiner
- Department of Cell Biology, University of Bielefeld, Bielefeld, Germany
- Forschungsverbund BioMedizin Bielefeld FBMB e.V., Bielefeld, Germany
| |
Collapse
|
171
|
Du J, Jing J, Yuan Y, Feng J, Han X, Chen S, Li X, Peng W, Xu J, Ho TV, Jiang X, Chai Y. Arid1a-Plagl1-Hh signaling is indispensable for differentiation-associated cell cycle arrest of tooth root progenitors. Cell Rep 2021; 35:108964. [PMID: 33826897 PMCID: PMC8132592 DOI: 10.1016/j.celrep.2021.108964] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 02/10/2021] [Accepted: 03/17/2021] [Indexed: 12/04/2022] Open
Abstract
Chromatin remodelers often show broad expression patterns in multiple cell types yet can elicit cell-specific effects in development and diseases. Arid1a binds DNA and regulates gene expression during tissue development and homeostasis. However, it is unclear how Arid1a achieves its functional specificity in regulating progenitor cells. Using the tooth root as a model, we show that loss of Arid1a impairs the differentiation-associated cell cycle arrest of tooth root progenitors through Hedgehog (Hh) signaling regulation, leading to shortened roots. Our data suggest that Plagl1, as a co-factor, endows Arid1a with its cell-type/spatial functional specificity. Furthermore, we show that loss of Arid1a leads to increased expression of Arid1b, which is also indispensable for odontoblast differentiation but is not involved in regulation of Hh signaling. This study expands our knowledge of the intricate interactions among chromatin remodelers, transcription factors, and signaling molecules during progenitor cell fate determination and lineage commitment.
Collapse
Affiliation(s)
- Jiahui Du
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA 90033, USA; Department of Prosthodontics, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Junjun Jing
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA 90033, USA
| | - Yuan Yuan
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA 90033, USA
| | - Jifan Feng
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA 90033, USA
| | - Xia Han
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA 90033, USA
| | - Shuo Chen
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA 90033, USA
| | - Xiang Li
- Department of Physics, George Washington University, Washington, DC 20052, USA
| | - Weiqun Peng
- Department of Physics, George Washington University, Washington, DC 20052, USA
| | - Jian Xu
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA 90033, USA
| | - Thach-Vu Ho
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA 90033, USA
| | - Xinquan Jiang
- Department of Prosthodontics, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Yang Chai
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA 90033, USA.
| |
Collapse
|
172
|
E LL, Zhang R, Li CJ, Zhang S, Ma XC, Xiao R, Liu HC. Effects of rhBMP-2 on Bone Formation Capacity of Rat Dental Stem/Progenitor Cells from Dental Follicle and Alveolar Bone Marrow. Stem Cells Dev 2021; 30:441-457. [PMID: 33798004 DOI: 10.1089/scd.2020.0170] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Dental stem/progenitor cells are a promising cell sources for alveolar bone (AB) regeneration because of their same embryonic origin and superior osteogenic potential. However, their molecular processes during osteogenic differentiation remain unclear. The objective of this study was to identify the responsiveness of dental follicle cells (DFCs) and AB marrow-derived mesenchymal stem cells (ABM-MSCs) to recombinant human bone morphogenetic protein-2 (rhBMP-2). These cells expressed vimentin and MSC markers and did not express cytokeratin and hematopoietic stem cell markers and showed multilineage differentiation potential under specific culture conditions. DFCs exhibited higher proliferation and colony-forming unit-fibroblast efficiency than ABM-MSCs; rhBMP-2 induced DFCs to differentiate toward a cementoblast/osteoblast phenotype and ABM-MSCs to differentiate only toward a osteoblast phenotype; and rhBMP-2-induced DFCs exhibited higher osteogenic differentiation potential than ABM-MSCs. These cells adhered, grew, and produced extracellular matrix on nanohydroxyapatite/collagen/poly(l-lactide) (nHAC/PLA). During a 14-day culture on nHAC/PLA, the extracellular alkaline phosphatase (ALP) activity of DFCs decreased gradually and that of ABM-MSCs increased gradually; rhBMP-2 enhanced their extracellular ALP activity, intracellular osteocalcin (OCN), and osteopontin (OPN) protein expression; and DFCs exhibited higher extracellular ALP activity and intracellular OCN protein expression than ABM-MSCs. When implanted subcutaneously in severe combined immunodeficient mice for 3 months, DFCs+nHAC/PLA+rhBMP-2 obtained higher percentage of bone formation area, OCN, and cementum attachment protein expression and lower OPN expression than ABM-MSCs+nHAC/PLA+rhBMP-2. These results showed that DFCs possessed superior proliferation and osteogenic differentiation potential in vitro, and formed higher quantity and quality bones in vivo. It suggested that DFCs might exhibit a more sensitive responsiveness to rhBMP-2, so that DFCs enter a relatively mature stage of osteogenic differentiation earlier than ABM-MSCs after rhBMP-2 induction. The findings imply that these dental stem/progenitor cells are alternative sources for AB engineering in regenerative medicine, and developing dental tissue may provide better source for stem/progenitor cells.
Collapse
Affiliation(s)
- Ling-Ling E
- Institute of Stomatology, First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Rong Zhang
- Institute of Stomatology, First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Chuan-Jie Li
- Institute of Stomatology, First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Shuo Zhang
- Institute of Stomatology, First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Xiao-Cao Ma
- Institute of Stomatology, First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Rui Xiao
- Institute of Stomatology, First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Hong-Chen Liu
- Institute of Stomatology, First Medical Center of Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
173
|
Yoshida H, Suzawa T, Shibata Y, Takahashi M, Kawai R, Takami M, Maki K, Kamijo R. Neural crest-derived cells in nasal conchae of adult mice contribute to bone regeneration. Biochem Biophys Res Commun 2021; 554:173-178. [PMID: 33798944 DOI: 10.1016/j.bbrc.2021.03.079] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 03/15/2021] [Indexed: 01/02/2023]
Abstract
Neural crest-derived cells (NCDCs), a class of adult stem cells not restricted to embryonic tissues, are attractive tissue regenerative therapy candidates because of their ease of isolation, self-renewing properties, and multipotency. Although adult NCDCs can undergo osteogenic differentiation in vitro, whether they induce bone formation in vivo remains unclear. Previously, our group reported findings showing high amounts of NCDCs scattered throughout nasal concha tissues of adult mice. In the present study, NCDCs in nasal conchae labeled with enhanced green fluorescent protein (EGFP) were collected from adult P0-Cre/CAG-CAT-EGFP double transgenic mice, then cultured in serum-free medium to increase the number. Subsequently, NCDCs were harvested and suspended in type I atelocollagen gel, then an atelocollagen sponge was used as a scaffold for the cell suspension. Atelocollagen scaffolds with NCDCs were placed on bone defects created in a mouse calvarial bone defect model. Over the ensuing 12 weeks, micro-CT and histological analysis findings showed that mice with scaffolds containing NCDCs had slightly greater bone formation as compared to those with a scaffold alone. Furthermore, Raman spectroscopy revealed spectral properties of bone in mice that received scaffolds with NCDCs similar to those of native calvarial bone. Bone regeneration is important not only for gaining bone mass but also chemical properties. These results are the first to show the validity of biomolecule-free adult nasal concha-derived NCDCs for bone regeneration, including the chemical properties of regenerated bone tissue.
Collapse
Affiliation(s)
- Hiroshi Yoshida
- Department of Biochemistry, School of Dentistry, Showa University, Tokyo, Japan; Department of Orthodontics, School of Dentistry, Showa University, Tokyo, Japan
| | - Tetsuo Suzawa
- Department of Biochemistry, School of Dentistry, Showa University, Tokyo, Japan.
| | - Yo Shibata
- Department of Conservative Dentistry, Division of Biomaterials and Engineering, School of Dentistry, Showa University, Tokyo, Japan
| | - Masahiro Takahashi
- Department of Orthodontics, School of Dentistry, Showa University, Tokyo, Japan
| | - Ryota Kawai
- Department of Orthodontics, School of Dentistry, Showa University, Tokyo, Japan
| | - Masamichi Takami
- Department of Pharmacology, School of Dentistry, Showa University, Tokyo, Japan
| | - Koutaro Maki
- Department of Orthodontics, School of Dentistry, Showa University, Tokyo, Japan
| | - Ryutaro Kamijo
- Department of Biochemistry, School of Dentistry, Showa University, Tokyo, Japan
| |
Collapse
|
174
|
Yamaguchi H, Kitami K, Wu X, He L, Wang J, Wang B, Komatsu Y. Alteration of DNA Damage Response Causes Cleft Palate. Front Physiol 2021; 12:649492. [PMID: 33854442 PMCID: PMC8039291 DOI: 10.3389/fphys.2021.649492] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 03/08/2021] [Indexed: 12/13/2022] Open
Abstract
Cleft palate is one of the most common craniofacial birth defects, however, little is known about how changes in the DNA damage response (DDR) cause cleft palate. To determine the role of DDR during palatogenesis, the DDR process was altered using a pharmacological intervention approach. A compromised DDR caused by a poly (ADP-ribose) polymerase (PARP) enzyme inhibitor resulted in cleft palate in wild-type mouse embryos, with increased DNA damage and apoptosis. In addition, a mouse genetic approach was employed to disrupt breast cancer 1 (BRCA1) and breast cancer 2 (BRCA2), known as key players in DDR. An ectomesenchymal-specific deletion of Brca1 or Brca2 resulted in cleft palate due to attenuation of cell survival. This was supported by the phenotypes of the ectomesenchymal-specific Brca1/Brca2 double-knockout mice. The cleft palate phenotype was rescued by superimposing p53 null alleles, demonstrating that the BRCA1/2-p53 DDR pathway is critical for palatogenesis. Our study highlights the importance of DDR in palatogenesis.
Collapse
Affiliation(s)
- Hiroyuki Yamaguchi
- Department of Pediatrics, The University of Texas Medical School at Houston, Houston, TX, United States
| | - Kohei Kitami
- Department of Pediatrics, The University of Texas Medical School at Houston, Houston, TX, United States
| | - Xiao Wu
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Li He
- Department of Pediatrics, The University of Texas Medical School at Houston, Houston, TX, United States
| | - Jianbo Wang
- Department of Pediatrics, The University of Texas Medical School at Houston, Houston, TX, United States
| | - Bin Wang
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States.,Graduate Program in Genetics & Epigenetics, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, United States
| | - Yoshihiro Komatsu
- Department of Pediatrics, The University of Texas Medical School at Houston, Houston, TX, United States.,Graduate Program in Genetics & Epigenetics, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, United States
| |
Collapse
|
175
|
White HE, Goswami A, Tucker AS. The Intertwined Evolution and Development of Sutures and Cranial Morphology. Front Cell Dev Biol 2021; 9:653579. [PMID: 33842480 PMCID: PMC8033035 DOI: 10.3389/fcell.2021.653579] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 03/08/2021] [Indexed: 12/21/2022] Open
Abstract
Phenotypic variation across mammals is extensive and reflects their ecological diversification into a remarkable range of habitats on every continent and in every ocean. The skull performs many functions to enable each species to thrive within its unique ecological niche, from prey acquisition, feeding, sensory capture (supporting vision and hearing) to brain protection. Diversity of skull function is reflected by its complex and highly variable morphology. Cranial morphology can be quantified using geometric morphometric techniques to offer invaluable insights into evolutionary patterns, ecomorphology, development, taxonomy, and phylogenetics. Therefore, the skull is one of the best suited skeletal elements for developmental and evolutionary analyses. In contrast, less attention is dedicated to the fibrous sutural joints separating the cranial bones. Throughout postnatal craniofacial development, sutures function as sites of bone growth, accommodating expansion of a growing brain. As growth frontiers, cranial sutures are actively responsible for the size and shape of the cranial bones, with overall skull shape being altered by changes to both the level and time period of activity of a given cranial suture. In keeping with this, pathological premature closure of sutures postnatally causes profound misshaping of the skull (craniosynostosis). Beyond this crucial role, sutures also function postnatally to provide locomotive shock absorption, allow joint mobility during feeding, and, in later postnatal stages, suture fusion acts to protect the developed brain. All these sutural functions have a clear impact on overall cranial function, development and morphology, and highlight the importance that patterns of suture development have in shaping the diversity of cranial morphology across taxa. Here we focus on the mammalian cranial system and review the intrinsic relationship between suture development and morphology and cranial shape from an evolutionary developmental biology perspective, with a view to understanding the influence of sutures on evolutionary diversity. Future work integrating suture development into a comparative evolutionary framework will be instrumental to understanding how developmental mechanisms shaping sutures ultimately influence evolutionary diversity.
Collapse
Affiliation(s)
- Heather E White
- Department of Life Sciences, Natural History Museum, London, United Kingdom.,Centre for Craniofacial and Regenerative Biology, King's College London, London, United Kingdom.,Division of Biosciences, University College London, London, United Kingdom
| | - Anjali Goswami
- Department of Life Sciences, Natural History Museum, London, United Kingdom.,Division of Biosciences, University College London, London, United Kingdom
| | - Abigail S Tucker
- Centre for Craniofacial and Regenerative Biology, King's College London, London, United Kingdom
| |
Collapse
|
176
|
Lin W, Li Q, Zhang D, Zhang X, Qi X, Wang Q, Chen Y, Liu C, Li H, Zhang S, Wang Y, Shao B, Zhang L, Yuan Q. Mapping the immune microenvironment for mandibular alveolar bone homeostasis at single-cell resolution. Bone Res 2021; 9:17. [PMID: 33723232 PMCID: PMC7960742 DOI: 10.1038/s41413-021-00141-5] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/18/2021] [Accepted: 02/01/2021] [Indexed: 02/04/2023] Open
Abstract
Alveolar bone is the thickened ridge of jaw bone that supports teeth. It is subject to constant occlusal force and pathogens invasion, and is therefore under active bone remodeling and immunomodulation. Alveolar bone holds a distinct niche from long bone considering their different developmental origin and postnatal remodeling pattern. However, a systematic explanation of alveolar bone at single-cell level is still lacking. Here, we construct a single-cell atlas of mouse mandibular alveolar bone through single-cell RNA sequencing (scRNA-seq). A more active immune microenvironment is identified in alveolar bone, with a higher proportion of mature immune cells than in long bone. Among all immune cell populations, the monocyte/macrophage subpopulation most actively interacts with mesenchymal stem cells (MSCs) subpopulation. Alveolar bone monocytes/macrophages express a higher level of Oncostatin M (Osm) compared to long bone, which promotes osteogenic differentiation and inhibits adipogenic differentiation of MSCs. In summary, our study reveals a unique immune microenvironment of alveolar bone, which may provide a more precise immune-modulatory target for therapeutic treatment of oral diseases.
Collapse
Affiliation(s)
- Weimin Lin
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Qiwen Li
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Danting Zhang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xiaohan Zhang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xingying Qi
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Qian Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yaqian Chen
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Caojie Liu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Hanwen Li
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Shiwen Zhang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yuan Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Bin Shao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Li Zhang
- Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, Center of Precision Medicine, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
| | - Quan Yuan
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China. .,Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| |
Collapse
|
177
|
Takenoshita M, Takechi M, Vu Hoang T, Furutera T, Akagawa C, Namangkalakul W, Aoto K, Kume T, Miyashin M, Iwamoto T, Iseki S. Cell lineage- and expression-based inference of the roles of forkhead box transcription factor Foxc2 in craniofacial development. Dev Dyn 2021; 250:1125-1139. [PMID: 33667029 DOI: 10.1002/dvdy.324] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 02/08/2021] [Accepted: 02/24/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Foxc2 is a member of the winged helix/forkhead (Fox) box family of transcription factors. Loss of function of Foxc2 causes craniofacial abnormalities such as cleft palate and deformed cranial base, but its role during craniofacial development remains to be elucidated. RESULTS The contributions of Foxc2-positive and its descendant cells to the craniofacial structure at E18.5 were examined using a tamoxifen-inducible Cre driver mouse (Foxc2-CreERT2) crossed with the R26R-LacZ reporter mouse. Foxc2 expression at E8.5 is restricted to the cranial mesenchyme, contributing to specific components including the cranial base, sensory capsule, tongue, upper incisor, and middle ear. Expression at E10.5 was still positively regulated in most of those regions. In situ hybridization analysis of Foxc2 and its closely related gene, Foxc1, revealed that expression domains of these genes largely overlap in the cephalic mesenchyme. Meanwhile, the tongue expressed Foxc2 but not Foxc1, and its development was affected by the neural crest-specific deletion of Foxc2 in mice (Wnt1-Cre; Foxc2fl/fl ). CONCLUSIONS Foxc2 is expressed in cranial mesenchyme that contributes to specific craniofacial tissue components from an early stage, and it seems to be involved in their development in cooperation with Foxc1. Foxc2 also has its own role in tongue development.
Collapse
Affiliation(s)
- Manami Takenoshita
- Department of Molecular Craniofacial Embryology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan.,Department of Pediatric Dentistry and Special Needs Dentistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Masaki Takechi
- Department of Molecular Craniofacial Embryology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Tri Vu Hoang
- Department of Molecular Craniofacial Embryology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Toshiko Furutera
- Department of Molecular Craniofacial Embryology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Chisaki Akagawa
- Department of Molecular Craniofacial Embryology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Worachat Namangkalakul
- Department of Molecular Craniofacial Embryology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Kazushi Aoto
- Department of Biochemistry, Hamamatsu University School of Medicine, Tokyo, Japan
| | - Tsutomu Kume
- Feinberg Cardiovascular Research Institute, Development of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Michiyo Miyashin
- Department of Pediatric Dentistry and Special Needs Dentistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Tsutomu Iwamoto
- Department of Pediatric Dentistry and Special Needs Dentistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Sachiko Iseki
- Department of Molecular Craniofacial Embryology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| |
Collapse
|
178
|
Chen H, Kang J, Zhang F, Yan T, Fan W, He H, Huang F. SIRT4 regulates rat dental papilla cell differentiation by promoting mitochondrial functions. Int J Biochem Cell Biol 2021; 134:105962. [PMID: 33636397 DOI: 10.1016/j.biocel.2021.105962] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 01/29/2021] [Accepted: 02/18/2021] [Indexed: 12/12/2022]
Abstract
INTRODUCTION SIRT4 is a mitochondrial sirtuin. Owing to its dependance on the cofactor nicotinamide adenine dinucleotide (NAD+), SIRT4 can act as a mitochondrial metabolic sensor of cellular energy status. We have previously shown that enhancement of mitochondrial functions is vital for the odontogenic diff ;erentiation of dental papilla cells (DPCs) during dentinogenesis. However, whether SIRT4 serves as an effective regulator of DPC diff ;erentiation by affecting mitochondrial functions remains unexplored. METHODS Primary DPCs obtained from the first molar dental papilla of neonatal Sprague-Dawley rats were used in this study. The expression pattern of SIRT4 was observed by immunohistochemistry in the first molar of postnatal day 1 (P1) rats. The changes in SIRT4 expression during odontogenic DPC differentiation were evaluated using real-time quantitative polymerase chain reaction (PCR), western blotting, and immunofluorescence. DPCs with loss (small interfering RNA-mediated knockdown) and gain (plasmid transfection-induced overexpression) of SIRT4 function were used to explore the role of SIRT4 in odontogenic differentiation. Mitochondrial function assays were performed using ATP, reactive oxygen species (ROS), and NAD+/NADH kits to investigate the potential mechanisms involved in SIRT4-mediated dentinogenesis. RESULTS In the present study, we found that SIRT4 expression increased in a time-dependent manner during odontogenic differentiation bothin vivo and in vitro. Sirt4 knockdown resulted in reduced odontogenic differentiation and mineralization, whereas an opposite effect was observed with SIRT4 overexpression. Furthermore, our results verified that in addition to reducing DPC differentiation, Sirt4 knockdown could also significantly reduce ATP levels, elevate the NAD+/NADH ratio, and increase ROS levels. CONCLUSION SIRT4 regulates mitochondrial functions and the antioxidant capacity of DPCs, thereby influencing dentin formation and tooth development, a phenomenon that may provide a foundation for better understanding the specific molecular mechanisms underlying dentin regeneration.
Collapse
Affiliation(s)
- Haoling Chen
- Department of Pediatric Dentistry, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Jun Kang
- Department of Pediatric Dentistry, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Fuping Zhang
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Tong Yan
- Department of Pediatric Dentistry, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Wenguo Fan
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Hongwen He
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China.
| | - Fang Huang
- Department of Pediatric Dentistry, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China.
| |
Collapse
|
179
|
Masuda K, Han X, Kato H, Sato H, Zhang Y, Sun X, Hirofuji Y, Yamaza H, Yamada A, Fukumoto S. Dental Pulp-Derived Mesenchymal Stem Cells for Modeling Genetic Disorders. Int J Mol Sci 2021; 22:ijms22052269. [PMID: 33668763 PMCID: PMC7956585 DOI: 10.3390/ijms22052269] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 02/18/2021] [Accepted: 02/23/2021] [Indexed: 12/20/2022] Open
Abstract
A subpopulation of mesenchymal stem cells, developmentally derived from multipotent neural crest cells that form multiple facial tissues, resides within the dental pulp of human teeth. These stem cells show high proliferative capacity in vitro and are multipotent, including adipogenic, myogenic, osteogenic, chondrogenic, and neurogenic potential. Teeth containing viable cells are harvested via minimally invasive procedures, based on various clinical diagnoses, but then usually discarded as medical waste, indicating the relatively low ethical considerations to reuse these cells for medical applications. Previous studies have demonstrated that stem cells derived from healthy subjects are an excellent source for cell-based medicine, tissue regeneration, and bioengineering. Furthermore, stem cells donated by patients affected by genetic disorders can serve as in vitro models of disease-specific genetic variants, indicating additional applications of these stem cells with high plasticity. This review discusses the benefits, limitations, and perspectives of patient-derived dental pulp stem cells as alternatives that may complement other excellent, yet incomplete stem cell models, such as induced pluripotent stem cells, together with our recent data.
Collapse
Affiliation(s)
- Keiji Masuda
- Section of Oral Medicine for Children, Division of Oral Health, Growth and Development, Faculty of Dental Science, Kyushu University, Maidashi 3-1-1, Higashi-Ku, Fukuoka 812-8582, Japan; (X.H.); (H.S.); (Y.Z.); (X.S.); (Y.H.); (H.Y.)
- Correspondence: (K.M.); (S.F.); Tel.: +81-92-642-6402 (K.M. & S.F.)
| | - Xu Han
- Section of Oral Medicine for Children, Division of Oral Health, Growth and Development, Faculty of Dental Science, Kyushu University, Maidashi 3-1-1, Higashi-Ku, Fukuoka 812-8582, Japan; (X.H.); (H.S.); (Y.Z.); (X.S.); (Y.H.); (H.Y.)
| | - Hiroki Kato
- Department of Molecular Cell Biology and Oral Anatomy, Graduate School of Dental Science, Kyushu University, Maidashi 3-1-1, Higashi-Ku, Fukuoka 812-8582, Japan;
| | - Hiroshi Sato
- Section of Oral Medicine for Children, Division of Oral Health, Growth and Development, Faculty of Dental Science, Kyushu University, Maidashi 3-1-1, Higashi-Ku, Fukuoka 812-8582, Japan; (X.H.); (H.S.); (Y.Z.); (X.S.); (Y.H.); (H.Y.)
| | - Yu Zhang
- Section of Oral Medicine for Children, Division of Oral Health, Growth and Development, Faculty of Dental Science, Kyushu University, Maidashi 3-1-1, Higashi-Ku, Fukuoka 812-8582, Japan; (X.H.); (H.S.); (Y.Z.); (X.S.); (Y.H.); (H.Y.)
| | - Xiao Sun
- Section of Oral Medicine for Children, Division of Oral Health, Growth and Development, Faculty of Dental Science, Kyushu University, Maidashi 3-1-1, Higashi-Ku, Fukuoka 812-8582, Japan; (X.H.); (H.S.); (Y.Z.); (X.S.); (Y.H.); (H.Y.)
| | - Yuta Hirofuji
- Section of Oral Medicine for Children, Division of Oral Health, Growth and Development, Faculty of Dental Science, Kyushu University, Maidashi 3-1-1, Higashi-Ku, Fukuoka 812-8582, Japan; (X.H.); (H.S.); (Y.Z.); (X.S.); (Y.H.); (H.Y.)
| | - Haruyoshi Yamaza
- Section of Oral Medicine for Children, Division of Oral Health, Growth and Development, Faculty of Dental Science, Kyushu University, Maidashi 3-1-1, Higashi-Ku, Fukuoka 812-8582, Japan; (X.H.); (H.S.); (Y.Z.); (X.S.); (Y.H.); (H.Y.)
| | - Aya Yamada
- Division of Pediatric Dentistry, Department of Oral Health and Development Sciences, Graduate School of Dentistry, Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8577, Japan;
| | - Satoshi Fukumoto
- Section of Oral Medicine for Children, Division of Oral Health, Growth and Development, Faculty of Dental Science, Kyushu University, Maidashi 3-1-1, Higashi-Ku, Fukuoka 812-8582, Japan; (X.H.); (H.S.); (Y.Z.); (X.S.); (Y.H.); (H.Y.)
- Division of Pediatric Dentistry, Department of Oral Health and Development Sciences, Graduate School of Dentistry, Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8577, Japan;
- Correspondence: (K.M.); (S.F.); Tel.: +81-92-642-6402 (K.M. & S.F.)
| |
Collapse
|
180
|
Stundl J, Bertucci PY, Lauri A, Arendt D, Bronner ME. Evolution of new cell types at the lateral neural border. Curr Top Dev Biol 2021; 141:173-205. [PMID: 33602488 DOI: 10.1016/bs.ctdb.2020.11.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
During the course of evolution, animals have become increasingly complex by the addition of novel cell types and regulatory mechanisms. A prime example is represented by the lateral neural border, known as the neural plate border in vertebrates, a region of the developing ectoderm where presumptive neural and non-neural tissue meet. This region has been intensively studied as the source of two important embryonic cell types unique to vertebrates-the neural crest and the ectodermal placodes-which contribute to diverse differentiated cell types including the peripheral nervous system, pigment cells, bone, and cartilage. How did these multipotent progenitors originate in animal evolution? What triggered the elaboration of the border during the course of chordate evolution? How is the lateral neural border patterned in various bilaterians and what is its fate? Here, we review and compare the development and fate of the lateral neural border in vertebrates and invertebrates and we speculate about its evolutionary origin. Taken together, the data suggest that the lateral neural border existed in bilaterian ancestors prior to the origin of vertebrates and became a developmental source of exquisite evolutionary change that frequently enabled the acquisition of new cell types.
Collapse
Affiliation(s)
- Jan Stundl
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, United States
| | | | | | - Detlev Arendt
- European Molecular Biology Laboratory, Heidelberg, Germany.
| | - Marianne E Bronner
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, United States.
| |
Collapse
|
181
|
He J, Yan J, Wang J, Zhao L, Xin Q, Zeng Y, Sun Y, Zhang H, Bai Z, Li Z, Ni Y, Gong Y, Li Y, He H, Bian Z, Lan Y, Ma C, Bian L, Zhu H, Liu B, Yue R. Dissecting human embryonic skeletal stem cell ontogeny by single-cell transcriptomic and functional analyses. Cell Res 2021; 31:742-757. [PMID: 33473154 PMCID: PMC8249634 DOI: 10.1038/s41422-021-00467-z] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 12/22/2020] [Indexed: 01/15/2023] Open
Abstract
Human skeletal stem cells (SSCs) have been discovered in fetal and adult long bones. However, the spatiotemporal ontogeny of human embryonic SSCs during early skeletogenesis remains elusive. Here we map the transcriptional landscape of human limb buds and embryonic long bones at single-cell resolution to address this fundamental question. We found remarkable heterogeneity within human limb bud mesenchyme and epithelium, and aligned them along the proximal–distal and anterior–posterior axes using known marker genes. Osteo-chondrogenic progenitors first appeared in the core limb bud mesenchyme, which give rise to multiple populations of stem/progenitor cells in embryonic long bones undergoing endochondral ossification. Importantly, a perichondrial embryonic skeletal stem/progenitor cell (eSSPC) subset was identified, which could self-renew and generate the osteochondral lineage cells, but not adipocytes or hematopoietic stroma. eSSPCs are marked by the adhesion molecule CADM1 and highly enriched with FOXP1/2 transcriptional network. Interestingly, neural crest-derived cells with similar phenotypic markers and transcriptional networks were also found in the sagittal suture of human embryonic calvaria. Taken together, this study revealed the cellular heterogeneity and lineage hierarchy during human embryonic skeletogenesis, and identified distinct skeletal stem/progenitor cells that orchestrate endochondral and intramembranous ossification.
Collapse
Affiliation(s)
- Jian He
- State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, 100071, China
| | - Jing Yan
- State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, 100071, China
| | - Jianfang Wang
- Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Liangyu Zhao
- Department of Orthopedics, Changzheng Hospital, Naval Medical University, Shanghai, 200003, China
| | - Qian Xin
- State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, 100071, China
| | - Yang Zeng
- State Key Laboratory of Experimental Hematology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100071, China
| | - Yuxi Sun
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Han Zhang
- Department of Transfusion, Daping Hospital, Army Military Medical University, Chongqing, 400042, China
| | - Zhijie Bai
- State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, 100071, China
| | - Zongcheng Li
- State Key Laboratory of Experimental Hematology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100071, China
| | - Yanli Ni
- State Key Laboratory of Experimental Hematology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100071, China
| | - Yandong Gong
- State Key Laboratory of Experimental Hematology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100071, China
| | - Yunqiao Li
- State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, 100071, China
| | - Han He
- State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, 100071, China
| | - Zhilei Bian
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Yu Lan
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, Guangdong, 510632, China.,Guangzhou Regenerative Medicine and Health-Guangdong Laboratory (GRMH-GDL), Guangzhou, Guangdong, 510530, China
| | - Chunyu Ma
- Department of Gynecology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100071, China
| | - Lihong Bian
- Department of Gynecology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100071, China
| | - Heng Zhu
- Beijing Institute of Radiation Medicine, Beijing, 100850, China.
| | - Bing Liu
- State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, 100071, China. .,State Key Laboratory of Experimental Hematology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100071, China. .,Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, Guangdong, 510632, China.
| | - Rui Yue
- Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
| |
Collapse
|
182
|
Etchevers HC. Pericyte Ontogeny: The Use of Chimeras to Track a Cell Lineage of Diverse Germ Line Origins. Methods Mol Biol 2021; 2235:61-87. [PMID: 33576971 DOI: 10.1007/978-1-0716-1056-5_6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The goal of lineage tracing is to understand body formation over time by discovering which cells are the progeny of a specific, identified, ancestral progenitor. Subsidiary questions include unequivocal identification of what they have become, how many descendants develop, whether they live or die, and where they are located in the tissue or body at the end of the window examined. A classical approach in experimental embryology, lineage tracing continues to be used in developmental biology and stem cell and cancer research, wherever cellular potential and behavior need to be studied in multiple dimensions, of which one is time. Each technical approach has its advantages and drawbacks. This chapter, with some previously unpublished data, will concentrate nonexclusively on the use of interspecies chimeras to explore the origins of perivascular (or mural) cells, of which those adjacent to the vascular endothelium are termed pericytes for this purpose. These studies laid the groundwork for our understanding that pericytes derive from progenitor mesenchymal pools of multiple origins in the vertebrate embryo, some of which persist into adulthood. The results obtained through xenografting, like in the methodology described here, complement those obtained through genetic lineage-tracing techniques within a given species.
Collapse
|
183
|
Nizami MZI, Nishina Y. Recent Advances in Stem Cells for Dental Tissue Engineering. ENGINEERING MATERIALS FOR STEM CELL REGENERATION 2021:281-324. [DOI: 10.1007/978-981-16-4420-7_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
184
|
Jamal M, Bashir A, Al-Sayegh M, Huang GTJ. Oral tissues as sources for induced pluripotent stem cell derivation and their applications for neural, craniofacial, and dental tissue regeneration. CELL SOURCES FOR IPSCS 2021:71-106. [DOI: 10.1016/b978-0-12-822135-8.00007-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
185
|
Dental Mesenchymal Stem/Progenitor Cells: A New Prospect in Regenerative Medicine. Stem Cells 2021. [DOI: 10.1007/978-3-030-77052-5_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
186
|
Yan F, Jia P, Yoshioka H, Suzuki A, Iwata J, Zhao Z. A developmental stage-specific network approach for studying dynamic co-regulation of transcription factors and microRNAs during craniofacial development. Development 2020; 147:226075. [PMID: 33234712 DOI: 10.1242/dev.192948] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 11/10/2020] [Indexed: 12/21/2022]
Abstract
Craniofacial development is regulated through dynamic and complex mechanisms that involve various signaling cascades and gene regulations. Disruption of such regulations can result in craniofacial birth defects. Here, we propose the first developmental stage-specific network approach by integrating two crucial regulators, transcription factors (TFs) and microRNAs (miRNAs), to study their co-regulation during craniofacial development. Specifically, we used TFs, miRNAs and non-TF genes to form feed-forward loops (FFLs) using genomic data covering mouse embryonic days E10.5 to E14.5. We identified key novel regulators (TFs Foxm1, Hif1a, Zbtb16, Myog, Myod1 and Tcf7, and miRNAs miR-340-5p and miR-129-5p) and target genes (Col1a1, Sgms2 and Slc8a3) expression of which changed in a developmental stage-dependent manner. We found that the Wnt-FoxO-Hippo pathway (from E10.5 to E11.5), tissue remodeling (from E12.5 to E13.5) and miR-129-5p-mediated Col1a1 regulation (from E10.5 to E14.5) might play crucial roles in craniofacial development. Enrichment analyses further suggested their functions. Our experiments validated the regulatory roles of miR-340-5p and Foxm1 in the Wnt-FoxO-Hippo subnetwork, as well as the role of miR-129-5p in the miR-129-5p-Col1a1 subnetwork. Thus, our study helps understand the comprehensive regulatory mechanisms for craniofacial development.
Collapse
Affiliation(s)
- Fangfang Yan
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Peilin Jia
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Hiroki Yoshioka
- Department of Diagnostic and Biomedical Sciences, School of Dentistry, The University of Texas Health Science Center at Houston, Houston, TX 77054, USA.,Center for Craniofacial Research, The University of Texas Health Science Center at Houston, Houston, TX 77054, USA
| | - Akiko Suzuki
- Department of Diagnostic and Biomedical Sciences, School of Dentistry, The University of Texas Health Science Center at Houston, Houston, TX 77054, USA.,Center for Craniofacial Research, The University of Texas Health Science Center at Houston, Houston, TX 77054, USA
| | - Junichi Iwata
- Department of Diagnostic and Biomedical Sciences, School of Dentistry, The University of Texas Health Science Center at Houston, Houston, TX 77054, USA.,Center for Craniofacial Research, The University of Texas Health Science Center at Houston, Houston, TX 77054, USA.,MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Zhongming Zhao
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA.,MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA.,Human Genetics Center, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| |
Collapse
|
187
|
Hamline MY, Corcoran CM, Wamstad JA, Miletich I, Feng J, Lohr JL, Hemberger M, Sharpe PT, Gearhart MD, Bardwell VJ. OFCD syndrome and extraembryonic defects are revealed by conditional mutation of the Polycomb-group repressive complex 1.1 (PRC1.1) gene BCOR. Dev Biol 2020; 468:110-132. [PMID: 32692983 PMCID: PMC9583620 DOI: 10.1016/j.ydbio.2020.06.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 06/16/2020] [Accepted: 06/26/2020] [Indexed: 12/15/2022]
Abstract
BCOR is a critical regulator of human development. Heterozygous mutations of BCOR in females cause the X-linked developmental disorder Oculofaciocardiodental syndrome (OFCD), and hemizygous mutations of BCOR in males cause gestational lethality. BCOR associates with Polycomb group proteins to form one subfamily of the diverse Polycomb repressive complex 1 (PRC1) complexes, designated PRC1.1. Currently there is limited understanding of differing developmental roles of the various PRC1 complexes. We therefore generated a conditional exon 9-10 knockout Bcor allele and a transgenic conditional Bcor expression allele and used these to define multiple roles of Bcor, and by implication PRC1.1, in mouse development. Females heterozygous for Bcor exhibiting mosaic expression due to the X-linkage of the gene showed reduced postnatal viability and had OFCD-like defects. By contrast, Bcor hemizygosity in the entire male embryo resulted in embryonic lethality by E9.5. We further dissected the roles of Bcor, focusing on some of the tissues affected in OFCD through use of cell type specific Cre alleles. Mutation of Bcor in neural crest cells caused cleft palate, shortening of the mandible and tympanic bone, ectopic salivary glands and abnormal tongue musculature. We found that defects in the mandibular region, rather than in the palate itself, led to palatal clefting. Mutation of Bcor in hindlimb progenitor cells of the lateral mesoderm resulted in 2/3 syndactyly. Mutation of Bcor in Isl1-expressing lineages that contribute to the heart caused defects including persistent truncus arteriosus, ventricular septal defect and fetal lethality. Mutation of Bcor in extraembryonic lineages resulted in placental defects and midgestation lethality. Ubiquitous over expression of transgenic Bcor isoform A during development resulted in embryonic defects and midgestation lethality. The defects we have found in Bcor mutants provide insights into the etiology of the OFCD syndrome and how BCOR-containing PRC1 complexes function in development.
Collapse
Affiliation(s)
- Michelle Y Hamline
- The Molecular, Cellular, Developmental Biology and Genetics Graduate Program, University of Minnesota, Minneapolis, MN, 55455, USA; University of Minnesota Medical Scientist Training Program, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Connie M Corcoran
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Joseph A Wamstad
- The Molecular, Cellular, Developmental Biology and Genetics Graduate Program, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Isabelle Miletich
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, SE1 9RT, UK
| | - Jifan Feng
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, SE1 9RT, UK
| | - Jamie L Lohr
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Myriam Hemberger
- Department of Biochemistry and Molecular Biology, Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Paul T Sharpe
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, SE1 9RT, UK; Medical Research Council Centre for Transplantation, King's College London, London, SE1 9RT, UK
| | - Micah D Gearhart
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN, 55455, USA; Developmental Biology Center, University of Minnesota, Minneapolis, MN, 55455, USA.
| | - Vivian J Bardwell
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN, 55455, USA; Developmental Biology Center, University of Minnesota, Minneapolis, MN, 55455, USA; Masonic Cancer Center, University of Minnesota, Minneapolis, MN, 55455, USA.
| |
Collapse
|
188
|
Yuan Y, Loh YHE, Han X, Feng J, Ho TV, He J, Jing J, Groff K, Wu A, Chai Y. Spatiotemporal cellular movement and fate decisions during first pharyngeal arch morphogenesis. SCIENCE ADVANCES 2020; 6:eabb0119. [PMID: 33328221 PMCID: PMC7744069 DOI: 10.1126/sciadv.abb0119] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 10/27/2020] [Indexed: 06/02/2023]
Abstract
Cranial neural crest (CNC) cells contribute to different cell types during embryonic development. It is unknown whether postmigratory CNC cells undergo dynamic cellular movement and how the process of cell fate decision occurs within the first pharyngeal arch (FPA). Our investigations demonstrate notable heterogeneity within the CNC cells, refine the patterning domains, and identify progenitor cells within the FPA. These progenitor cells undergo fate bifurcation that separates them into common progenitors and mesenchymal cells, which are characterized by Cdk1 and Spry2/Notch2 expression, respectively. The common progenitors undergo further bifurcations to restrict them into osteogenic/odontogenic and chondrogenic/fibroblast lineages. Disruption of a patterning domain leads to specific mandible and tooth defects, validating the binary cell fate restriction process. Different from the compartment model of mandibular morphogenesis, our data redefine heterogeneous cellular domains within the FPA, reveal dynamic cellular movement in time, and describe a sequential series of binary cell fate decision-making process.
Collapse
Affiliation(s)
- Yuan Yuan
- Center for Craniofacial Molecular Biology, University of Southern California, 2250 Alcazar Street, CSA 103, Los Angeles, CA 90033, USA
| | - Yong-Hwee Eddie Loh
- Bioinformatics Service, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA
| | - Xia Han
- Center for Craniofacial Molecular Biology, University of Southern California, 2250 Alcazar Street, CSA 103, Los Angeles, CA 90033, USA
| | - Jifan Feng
- Center for Craniofacial Molecular Biology, University of Southern California, 2250 Alcazar Street, CSA 103, Los Angeles, CA 90033, USA
| | - Thach-Vu Ho
- Center for Craniofacial Molecular Biology, University of Southern California, 2250 Alcazar Street, CSA 103, Los Angeles, CA 90033, USA
| | - Jinzhi He
- Center for Craniofacial Molecular Biology, University of Southern California, 2250 Alcazar Street, CSA 103, Los Angeles, CA 90033, USA
| | - Junjun Jing
- Center for Craniofacial Molecular Biology, University of Southern California, 2250 Alcazar Street, CSA 103, Los Angeles, CA 90033, USA
| | - Kimberly Groff
- Center for Craniofacial Molecular Biology, University of Southern California, 2250 Alcazar Street, CSA 103, Los Angeles, CA 90033, USA
| | - Alan Wu
- Center for Craniofacial Molecular Biology, University of Southern California, 2250 Alcazar Street, CSA 103, Los Angeles, CA 90033, USA
| | - Yang Chai
- Center for Craniofacial Molecular Biology, University of Southern California, 2250 Alcazar Street, CSA 103, Los Angeles, CA 90033, USA.
| |
Collapse
|
189
|
Morice A, Cornette R, Giudice A, Collet C, Paternoster G, Arnaud É, Galliani E, Picard A, Legeai-Mallet L, Khonsari RH. Early mandibular morphological differences in patients with FGFR2 and FGFR3-related syndromic craniosynostoses: A 3D comparative study. Bone 2020; 141:115600. [PMID: 32822871 DOI: 10.1016/j.bone.2020.115600] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 08/10/2020] [Accepted: 08/14/2020] [Indexed: 01/04/2023]
Abstract
Syndromic craniosynostoses are defined by the premature fusion of one or more cranial and facial sutures, leading to skull vault deformation, and midfacial retrusion. More recently, mandibular shape modifications have been described in FGFR-related craniosynostoses, which represent almost 75% of the syndromic craniosynostoses. Here, further characterisation of the mandibular phenotype in FGFR-related craniosynostoses is provided in order to confirm mandibular shape modifications, as this could contribute to a better understanding of the involvement of the FGFR pathway in craniofacial development. The aim of our study was to analyse early mandibular morphology in a cohort of patients with FGFR2- (Crouzon and Apert) and FGFR3- (Muenke and Crouzonodermoskeletal) related syndromic craniosynostoses. We used a comparative geometric morphometric approach based on 3D imaging. Thirty-one anatomical landmarks and eleven curves with sliding semi-landmarks were defined to model the shape of the mandible. In total, 40 patients (12 with Crouzon, 12 with Apert, 12 with Muenke and 4 with Crouzonodermoskeletal syndromes) and 40 age and sex-matched controls were included (mean age: 13.7 months ±11.9). Mandibular shape differed significantly between controls and each patient group based on geometric morphometrics. Mandibular shape in FGFR2-craniosynostoses was characterized by open gonial angle, short ramus height, and high and prominent symphysis. Short ramus height appeared more pronounced in Apert than in Crouzon syndrome. Additionally, narrow inter-condylar and inter-gonial distances were observed in Crouzon syndrome. Mandibular shape in FGFR3-craniosynostoses was characterized by high and prominent symphysis and narrow inter-gonial distance. In addition, narrow condylar processes affected patients with Crouzonodermoskeletal syndrome. Statistical analysis of variance showed significant clustering of Apert and Crouzon, Crouzon and Muenke, and Apert and Muenke patients (p < 0.05). Our results confirm distinct mandibular shapes at early ages in FGFR2- (Crouzon and Apert syndromes) and FGFR3-related syndromic craniosynostoses (Muenke and Crouzonodermoskeletal syndromes) and reinforce the hypothesis of genotype-phenotype correspondence concerning mandibular morphology.
Collapse
Affiliation(s)
- A Morice
- Service de Chirurgie Maxillo-Faciale et Chirurgie Plastique, Hôpital Universitaire Necker - Enfants Malades, Assistance Publique - Hôpitaux de Paris, Centre de Référence Maladies Rares MAFACE Fentes et Malformations Faciales, Université de Paris, Paris, France; Laboratoire 'Bases Moléculaires et Physiopathologiques des Ostéochondrodysplasies', INSERM UMR 1163, Institut Imagine, Paris, France.
| | - R Cornette
- Institut de Systématique, Evolution, Biodiversité (ISYEB), Muséum National d'Histoire Naturelle, Sorbonne Université, Ecole Pratique des Hautes Etudes, Université des Antilles, CNRS, CP 50, 57 rue Cuvier, 75005 Paris, France
| | - A Giudice
- Università Degli Studi di Catanzaro 'Magna Graecia', Catanzaro, Italy
| | - C Collet
- BIOSCAR, INSERM U1132, Université de Paris, Hôpital Lariboisière, 75010 Paris, France; Service de Biochimie et Biologie Moléculaire, CHU-Paris-GH Saint Louis Lariboisière Widal, Paris, France
| | - G Paternoster
- Service de Neurochirurgie, Hôpital Universitaire Necker - Enfants Malades, Assistance Publique - Hôpitaux de Paris, Centre de Référence Maladies Rares CRANIOST Craniosténoses et Malformations Craniofaciales, Université de Paris, Paris, France
| | - É Arnaud
- Service de Neurochirurgie, Hôpital Universitaire Necker - Enfants Malades, Assistance Publique - Hôpitaux de Paris, Centre de Référence Maladies Rares CRANIOST Craniosténoses et Malformations Craniofaciales, Université de Paris, Paris, France
| | - E Galliani
- Service de Chirurgie Maxillo-Faciale et Chirurgie Plastique, Hôpital Universitaire Necker - Enfants Malades, Assistance Publique - Hôpitaux de Paris, Centre de Référence Maladies Rares MAFACE Fentes et Malformations Faciales, Université de Paris, Paris, France
| | - A Picard
- Service de Chirurgie Maxillo-Faciale et Chirurgie Plastique, Hôpital Universitaire Necker - Enfants Malades, Assistance Publique - Hôpitaux de Paris, Centre de Référence Maladies Rares MAFACE Fentes et Malformations Faciales, Université de Paris, Paris, France
| | - L Legeai-Mallet
- Laboratoire 'Bases Moléculaires et Physiopathologiques des Ostéochondrodysplasies', INSERM UMR 1163, Institut Imagine, Paris, France
| | - R H Khonsari
- Service de Chirurgie Maxillo-Faciale et Chirurgie Plastique, Hôpital Universitaire Necker - Enfants Malades, Assistance Publique - Hôpitaux de Paris, Centre de Référence Maladies Rares MAFACE Fentes et Malformations Faciales, Université de Paris, Paris, France; Laboratoire 'Bases Moléculaires et Physiopathologiques des Ostéochondrodysplasies', INSERM UMR 1163, Institut Imagine, Paris, France; Service de Neurochirurgie, Hôpital Universitaire Necker - Enfants Malades, Assistance Publique - Hôpitaux de Paris, Centre de Référence Maladies Rares CRANIOST Craniosténoses et Malformations Craniofaciales, Université de Paris, Paris, France
| |
Collapse
|
190
|
Rocha M, Beiriger A, Kushkowski EE, Miyashita T, Singh N, Venkataraman V, Prince VE. From head to tail: regionalization of the neural crest. Development 2020; 147:dev193888. [PMID: 33106325 PMCID: PMC7648597 DOI: 10.1242/dev.193888] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The neural crest is regionalized along the anteroposterior axis, as demonstrated by foundational lineage-tracing experiments that showed the restricted developmental potential of neural crest cells originating in the head. Here, we explore how recent studies of experimental embryology, genetic circuits and stem cell differentiation have shaped our understanding of the mechanisms that establish axial-specific populations of neural crest cells. Additionally, we evaluate how comparative, anatomical and genomic approaches have informed our current understanding of the evolution of the neural crest and its contribution to the vertebrate body.
Collapse
Affiliation(s)
- Manuel Rocha
- Committee on Development, Regeneration and Stem Cell Biology, The University of Chicago, Chicago, IL 60637, USA
| | - Anastasia Beiriger
- Department of Organismal Biology and Anatomy, The University of Chicago, Chicago, IL 60637, USA
| | - Elaine E Kushkowski
- Committee on Development, Regeneration and Stem Cell Biology, The University of Chicago, Chicago, IL 60637, USA
| | - Tetsuto Miyashita
- Department of Organismal Biology and Anatomy, The University of Chicago, Chicago, IL 60637, USA
- Canadian Museum of Nature, Ottawa, ON K1P 6P4, Canada
| | - Noor Singh
- Department of Organismal Biology and Anatomy, The University of Chicago, Chicago, IL 60637, USA
| | - Vishruth Venkataraman
- Department of Organismal Biology and Anatomy, The University of Chicago, Chicago, IL 60637, USA
| | - Victoria E Prince
- Committee on Development, Regeneration and Stem Cell Biology, The University of Chicago, Chicago, IL 60637, USA
- Department of Organismal Biology and Anatomy, The University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
191
|
Tang W, Bronner ME. Neural crest lineage analysis: from past to future trajectory. Development 2020; 147:147/20/dev193193. [PMID: 33097550 DOI: 10.1242/dev.193193] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Since its discovery 150 years ago, the neural crest has intrigued investigators owing to its remarkable developmental potential and extensive migratory ability. Cell lineage analysis has been an essential tool for exploring neural crest cell fate and migration routes. By marking progenitor cells, one can observe their subsequent locations and the cell types into which they differentiate. Here, we review major discoveries in neural crest lineage tracing from a historical perspective. We discuss how advancing technologies have refined lineage-tracing studies, and how clonal analysis can be applied to questions regarding multipotency. We also highlight how effective progenitor cell tracing, when combined with recently developed molecular and imaging tools, such as single-cell transcriptomics, single-molecule fluorescence in situ hybridization and high-resolution imaging, can extend the scope of neural crest lineage studies beyond development to regeneration and cancer initiation.
Collapse
Affiliation(s)
- Weiyi Tang
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Marianne E Bronner
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| |
Collapse
|
192
|
Potential Therapeutic Effects of Exosomes in Regenerative Endodontics. Arch Oral Biol 2020; 120:104946. [PMID: 33129129 DOI: 10.1016/j.archoralbio.2020.104946] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 09/07/2020] [Accepted: 10/04/2020] [Indexed: 02/05/2023]
Abstract
OBJECTIVE This review aims to describe the basic characteristics of exosomes, and summarize their possible source and potential biological effects in pulp regeneration, providing new insights into the therapeutic role of exosomes for regenerative endodontics in the future. DESIGN A comprehensive review of scientific literature related to exosomes potentially used for pulp regeneration was conducted. RESULTS Dental mesenchymal stem cells (MSCs) play an important role in dental pulp regeneration. MSC-derived exosomes, as important biotransmitters in intercellular communication, have been shown to replicate the therapeutic effects of their parental cells. These exosomes have better stability, lower immunogenicity, higher safety and clinical efficiency, making it possible to apply them in pulp regeneration. Existing research suggests that exosomes could trigger the regeneration of dentin/pulp-like tissue in vivo, which may attribute to their role in promoting pulp angiogenesis, regulating dental cell proliferation, migration and differentiation, and providing neuroprotection. CONCLUSIONS The applications of exosomes in the treatment of pulp regeneration have great potential, and exosomes may become ideal therapeutic biomaterial in regenerative endodontics.
Collapse
|
193
|
Pini J, Kueper J, Hu YD, Kawasaki K, Yeung P, Tsimbal C, Yoon B, Carmichael N, Maas RL, Cotney J, Grinblat Y, Liao EC. ALX1-related frontonasal dysplasia results from defective neural crest cell development and migration. EMBO Mol Med 2020; 12:e12013. [PMID: 32914578 PMCID: PMC7539331 DOI: 10.15252/emmm.202012013] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 08/12/2020] [Accepted: 08/13/2020] [Indexed: 01/02/2023] Open
Abstract
A pedigree of subjects presented with frontonasal dysplasia (FND). Genome sequencing and analysis identified a p.L165F missense variant in the homeodomain of the transcription factor ALX1 which was imputed to be pathogenic. Induced pluripotent stem cells (iPSC) were derived from the subjects and differentiated to neural crest cells (NCC). NCC derived from ALX1L165F/L165F iPSC were more sensitive to apoptosis, showed an elevated expression of several neural crest progenitor state markers, and exhibited impaired migration compared to wild-type controls. NCC migration was evaluated in vivo using lineage tracing in a zebrafish model, which revealed defective migration of the anterior NCC stream that contributes to the median portion of the anterior neurocranium, phenocopying the clinical presentation. Analysis of human NCC culture media revealed a change in the level of bone morphogenic proteins (BMP), with a low level of BMP2 and a high level of BMP9. Soluble BMP2 and BMP9 antagonist treatments were able to rescue the defective migration phenotype. Taken together, these results demonstrate a mechanistic requirement of ALX1 in NCC development and migration.
Collapse
Affiliation(s)
- Jonathan Pini
- Center for Regenerative MedicineDepartment of SurgeryMassachusetts General HospitalBostonMAUSA
- Shriners Hospital for ChildrenBostonMAUSA
| | - Janina Kueper
- Center for Regenerative MedicineDepartment of SurgeryMassachusetts General HospitalBostonMAUSA
- Shriners Hospital for ChildrenBostonMAUSA
- Life and Brain CenterUniversity of BonnBonnGermany
| | - Yiyuan David Hu
- Center for Regenerative MedicineDepartment of SurgeryMassachusetts General HospitalBostonMAUSA
- Shriners Hospital for ChildrenBostonMAUSA
| | - Kenta Kawasaki
- Center for Regenerative MedicineDepartment of SurgeryMassachusetts General HospitalBostonMAUSA
- Shriners Hospital for ChildrenBostonMAUSA
| | - Pan Yeung
- Center for Regenerative MedicineDepartment of SurgeryMassachusetts General HospitalBostonMAUSA
- Shriners Hospital for ChildrenBostonMAUSA
| | - Casey Tsimbal
- Center for Regenerative MedicineDepartment of SurgeryMassachusetts General HospitalBostonMAUSA
- Shriners Hospital for ChildrenBostonMAUSA
| | - Baul Yoon
- Departments of Integrative Biology, Neuroscience, and Genetics Ph.D. Training ProgramUniversity of Wisconsin‐MadisonMadisonWIUSA
| | - Nikkola Carmichael
- Department of GeneticsBrigham and Women's HospitalHarvard Medical SchoolBostonMAUSA
| | - Richard L Maas
- Department of GeneticsBrigham and Women's HospitalHarvard Medical SchoolBostonMAUSA
| | - Justin Cotney
- Genetics and Genome SciencesUConn HealthFarmingtonCTUSA
| | - Yevgenya Grinblat
- Departments of Integrative Biology, Neuroscience, and Genetics Ph.D. Training ProgramUniversity of Wisconsin‐MadisonMadisonWIUSA
| | - Eric C Liao
- Center for Regenerative MedicineDepartment of SurgeryMassachusetts General HospitalBostonMAUSA
- Shriners Hospital for ChildrenBostonMAUSA
| |
Collapse
|
194
|
Barqué A, Jan K, De La Fuente E, Nicholas CL, Hynes RO, Naba A. Knockout of the gene encoding the extracellular matrix protein SNED1 results in early neonatal lethality and craniofacial malformations. Dev Dyn 2020; 250:274-294. [PMID: 33012048 DOI: 10.1002/dvdy.258] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 09/10/2020] [Accepted: 09/28/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The extracellular matrix (ECM) is a fundamental component of multicellular organisms that orchestrates developmental processes and controls cell and tissue organization. We previously identified the novel ECM protein SNED1 as a promoter of breast cancer metastasis and showed that its level of expression negatively correlated with breast cancer patient survival. Here, we sought to identify the roles of SNED1 during murine development. RESULTS We generated two novel Sned1 knockout mouse strains and showed that Sned1 is essential since homozygous ablation of the gene led to early neonatal lethality. Phenotypic analysis of the surviving knockout mice revealed a role for SNED1 in the development of craniofacial and skeletal structures since Sned1 knockout resulted in growth defects, nasal cavity occlusion, and craniofacial malformations. Sned1 is widely expressed in embryos, notably by cell populations undergoing epithelial-to-mesenchymal transition, such as the neural crest cells. We further show that mice with a neural-crest-cell-specific deletion of Sned1 survive, but display facial anomalies partly phenocopying the global knockout mice. CONCLUSIONS Our results demonstrate requisite roles for SNED1 during development and neonatal survival. Importantly, the deletion of 2q37.3 in humans, a region that includes the SNED1 locus, has been associated with facial dysmorphism and short stature.
Collapse
Affiliation(s)
- Anna Barqué
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Kyleen Jan
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Emanuel De La Fuente
- Department of Orthodontics, College of Dentistry, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Christina L Nicholas
- Department of Orthodontics, College of Dentistry, University of Illinois at Chicago, Chicago, Illinois, USA
- Department of Anthropology, College of Liberal Arts and Sciences, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Richard O Hynes
- Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Alexandra Naba
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| |
Collapse
|
195
|
Samuels BD, Aho R, Brinkley JF, Bugacov A, Feingold E, Fisher S, Gonzalez-Reiche AS, Hacia JG, Hallgrimsson B, Hansen K, Harris MP, Ho TV, Holmes G, Hooper JE, Jabs EW, Jones KL, Kesselman C, Klein OD, Leslie EJ, Li H, Liao EC, Long H, Lu N, Maas RL, Marazita ML, Mohammed J, Prescott S, Schuler R, Selleri L, Spritz RA, Swigut T, van Bakel H, Visel A, Welsh I, Williams C, Williams TJ, Wysocka J, Yuan Y, Chai Y. FaceBase 3: analytical tools and FAIR resources for craniofacial and dental research. Development 2020; 147:dev191213. [PMID: 32958507 PMCID: PMC7522026 DOI: 10.1242/dev.191213] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 08/13/2020] [Indexed: 12/12/2022]
Abstract
The FaceBase Consortium was established by the National Institute of Dental and Craniofacial Research in 2009 as a 'big data' resource for the craniofacial research community. Over the past decade, researchers have deposited hundreds of annotated and curated datasets on both normal and disordered craniofacial development in FaceBase, all freely available to the research community on the FaceBase Hub website. The Hub has developed numerous visualization and analysis tools designed to promote integration of multidisciplinary data while remaining dedicated to the FAIR principles of data management (findability, accessibility, interoperability and reusability) and providing a faceted search infrastructure for locating desired data efficiently. Summaries of the datasets generated by the FaceBase projects from 2014 to 2019 are provided here. FaceBase 3 now welcomes contributions of data on craniofacial and dental development in humans, model organisms and cell lines. Collectively, the FaceBase Consortium, along with other NIH-supported data resources, provide a continuously growing, dynamic and current resource for the scientific community while improving data reproducibility and fulfilling data sharing requirements.
Collapse
Affiliation(s)
- Bridget D Samuels
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, CA 90033, USA
| | - Robert Aho
- Program in Craniofacial Biology, Departments of Orofacial Sciences and of Anatomy, Institute of Human Genetics, University of California San Francisco, San Francisco, CA 94143, USA
| | - James F Brinkley
- Structural Informatics Group, Department of Biological Structure, University of Washington, Seattle, WA 98195, USA
| | - Alejandro Bugacov
- Information Sciences Institute, Viterbi School of Engineering, University of Southern California, Marina del Rey, CA 90292, USA
| | - Eleanor Feingold
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Shannon Fisher
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA 02118, USA
| | - Ana S Gonzalez-Reiche
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Joseph G Hacia
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Benedikt Hallgrimsson
- Department of Cell Biology and Anatomy, Alberta Children's Hospital Research Institute, and McCaig Bone and Joint Institute, University of Calgary, Alberta, Canada
| | - Karissa Hansen
- Program in Craniofacial Biology, Departments of Orofacial Sciences and of Anatomy, Institute of Human Genetics, University of California San Francisco, San Francisco, CA 94143, USA
| | - Matthew P Harris
- Department of Orthopedic Research, Boston Children's Hospital and Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Thach-Vu Ho
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, CA 90033, USA
| | - Greg Holmes
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Joan E Hooper
- Department of Cell and Developmental Biology, School of Medicine, University of Colorado, Aurora, CO 80045, USA
| | - Ethylin Wang Jabs
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kenneth L Jones
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Colorado, Aurora, CO 80045, USA
| | - Carl Kesselman
- Information Sciences Institute, Viterbi School of Engineering, University of Southern California, Marina del Rey, CA 90292, USA
| | - Ophir D Klein
- Program in Craniofacial Biology, Departments of Orofacial Sciences and Pediatrics, Institute for Human Genetics, University of California San Francisco, San Francisco, CA 94143, USA
| | | | - Hong Li
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado, Aurora, CO 80045, USA
| | - Eric C Liao
- Massachusetts General Hospital, Plastic and Reconstructive Surgery, Boston, MA 02114, USA
| | - Hannah Long
- Departments of Chemical and Systems Biology and of Developmental Biology, Howard Hughes Medical Institute, School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Na Lu
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Richard L Maas
- Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Mary L Marazita
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Center for Craniofacial and Dental Genetics, Department of Oral Biology, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Clinical and Translational Science, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Jaaved Mohammed
- Departments of Chemical and Systems Biology and of Developmental Biology, Howard Hughes Medical Institute, School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Sara Prescott
- Departments of Chemical and Systems Biology and of Developmental Biology, Howard Hughes Medical Institute, School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Robert Schuler
- Information Sciences Institute, Viterbi School of Engineering, University of Southern California, Marina del Rey, CA 90292, USA
| | - Licia Selleri
- Program in Craniofacial Biology, Departments of Orofacial Sciences and of Anatomy, Institute of Human Genetics, University of California San Francisco, San Francisco, CA 94143, USA
| | - Richard A Spritz
- Human Medical Genetics and Genomics Program, School of Medicine, University of Colorado, Aurora, CO 80045, USA
| | - Tomek Swigut
- Departments of Chemical and Systems Biology and of Developmental Biology, Howard Hughes Medical Institute, School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Harm van Bakel
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Axel Visel
- Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
- U.S. Department of Energy Joint Genome Institute, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
- School of Natural Sciences, University of California Merced, Merced, CA 95343, USA
| | - Ian Welsh
- Program in Craniofacial Biology, Departments of Orofacial Sciences and of Anatomy, Institute of Human Genetics, University of California San Francisco, San Francisco, CA 94143, USA
| | - Cristina Williams
- Information Sciences Institute, Viterbi School of Engineering, University of Southern California, Marina del Rey, CA 90292, USA
| | - Trevor J Williams
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado, Aurora, CO 80045, USA
| | - Joanna Wysocka
- Departments of Chemical and Systems Biology and of Developmental Biology, Howard Hughes Medical Institute, School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Yuan Yuan
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, CA 90033, USA
| | - Yang Chai
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
196
|
Nagata M, Ono N, Ono W. Unveiling diversity of stem cells in dental pulp and apical papilla using mouse genetic models: a literature review. Cell Tissue Res 2020; 383:603-616. [PMID: 32803323 DOI: 10.1007/s00441-020-03271-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 07/29/2020] [Indexed: 12/16/2022]
Abstract
The dental pulp, a non-mineralized connective tissue uniquely encased within the cavity of the tooth, provides a niche for diverse arrays of dental mesenchymal stem cells. Stem cells in the dental pulp, including dental pulp stem cells (DPSCs), stem cells from human exfoliated deciduous teeth (SHEDs) and stem cells from apical papilla (SCAPs), have been isolated from human tissues with an emphasis on their potential application to regenerative therapies. Recent studies utilizing mouse genetic models shed light on the identities of these mesenchymal progenitor cells derived from neural crest cells (NCCs) in their native conditions, particularly regarding how they contribute to homeostasis and repair of the dental tissue. The current concept is that at least two distinct niches for stem cells exist in the dental pulp, e.g., the perivascular niche and the perineural niche. The precise identities of these stem cells and their niches are now beginning to be unraveled thanks to sophisticated mouse genetic models, which lead to better understanding of the fundamental properties of stem cells in the dental pulp and the apical papilla in humans. The new knowledge will be highly instrumental for developing more effective stem cell-based regenerative therapies to repair teeth in the future.
Collapse
Affiliation(s)
- Mizuki Nagata
- Department of Orthodontics and Pediatric Dentistry, University of Michigan School of Dentistry, Ann Arbor, MI, 48109, USA
| | - Noriaki Ono
- Department of Orthodontics and Pediatric Dentistry, University of Michigan School of Dentistry, Ann Arbor, MI, 48109, USA
| | - Wanida Ono
- Department of Orthodontics and Pediatric Dentistry, University of Michigan School of Dentistry, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
197
|
Human Dental Pulp Stem Cells and Gingival Mesenchymal Stem Cells Display Action Potential Capacity In Vitro after Neuronogenic Differentiation. Stem Cell Rev Rep 2020; 15:67-81. [PMID: 30324358 DOI: 10.1007/s12015-018-9854-5] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The potential of human mesenchymal stromal/stem cells (MSCs) including oral stem cells (OSCs) as a cell source to derive functional neurons has been inconclusive. Here we tested a number of human OSCs for their neurogenic potential compared to non-OSCs and employed various neurogenic induction methods. OSCs including dental pulp stem cells (DPSCs), gingiva-derived mesenchymal stem cells (GMSCs), stem cells from apical papilla and non-OSCs including bone marrow MSCs (BMMSCs), foreskin fibroblasts and dermal fibroblasts using non-neurosphere-mediated or neurosphere-mediated methods to guide them toward neuronal lineages. Cells were subjected to RT-qPCR, immunocytofluorescence to detect the expression of neurogenic genes or electrophysiological analysis at final stage of maturation. We found that induced DPSCs and GMSCs overall appeared to be more neurogenic compared to other cells either morphologically or levels of neurogenic gene expression. Nonetheless, of all the neural induction methods employed, only one neurosphere-mediated method yielded electrophysiological properties of functional neurons. Under this method, cells expressed increased neural stem cell markers, nestin and SOX1, in the first phase of differentiation. Neuronal-like cells expressed βIII-tubulin, CNPase, GFAP, MAP-2, NFM, pan-Nav, GAD67, Nav1.6, NF1, NSE, PSD95, and synapsin after the second phase of differentiation to maturity. Electrophysiological experiments revealed that 8.3% of DPSC-derived neuronal cells and 21.2% of GMSC-derived neuronal cells displayed action potential, although no spontaneous excitatory/inhibitory postsynaptic action potential was observed. We conclude that DPSCs and GMSCs have the potential to become neuronal cells in vitro, therefore, these cells may be used as a source for neural regeneration.
Collapse
|
198
|
Men Y, Wang Y, Yi Y, Jing D, Luo W, Shen B, Stenberg W, Chai Y, Ge WP, Feng JQ, Zhao H. Gli1+ Periodontium Stem Cells Are Regulated by Osteocytes and Occlusal Force. Dev Cell 2020; 54:639-654.e6. [PMID: 32652075 DOI: 10.1016/j.devcel.2020.06.006] [Citation(s) in RCA: 99] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 02/04/2020] [Accepted: 06/02/2020] [Indexed: 01/05/2023]
Abstract
Teeth are attached to alveolar bone by the periodontal ligament (PDL), which contains stem cells supporting tissue turnover. Here, we identified Gli1+ cells in adult mouse molar PDL as multi-potential stem cells (PDLSCs) giving rise to PDL, alveolar bone, and cementum. They support periodontium tissue turnover and injury repair. Gli1+ PDLSCs are surrounding the neurovascular bundle and more enriched in the apical region. Canonical Wnt signaling is essential for their activation. Alveolar bone osteocytes negatively regulate Gli1+ PDLSCs activity through sclerostin, a Wnt inhibitor. Blockage of sclerostin accelerates the PDLSCs lineage contribution rate in vivo. Sclerostin expression is modulated by physiological occlusal force. Removal of occlusal force upregulates sclerostin and inhibits PDLSCs activation. In summary, Gli1+ cells are the multipotential PDLSCs in vivo. Osteocytes provide negative feedback to PDLSCs and inhibit their activities through sclerostin. Physiological occlusal force indirectly regulates PDLSCs activities by fine-tuning this feedback loop.
Collapse
Affiliation(s)
- Yi Men
- Department of Comprehensive Dentistry, College of Dentistry, Texas A&M University, Dallas, TX 75246, USA; West China School of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yuhong Wang
- Department of Comprehensive Dentistry, College of Dentistry, Texas A&M University, Dallas, TX 75246, USA; West China School of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yating Yi
- Department of Comprehensive Dentistry, College of Dentistry, Texas A&M University, Dallas, TX 75246, USA
| | - Dian Jing
- Department of Comprehensive Dentistry, College of Dentistry, Texas A&M University, Dallas, TX 75246, USA
| | - Wenjing Luo
- Department of Biomedical Sciences, College of Dentistry, Texas A&M University, Dallas, TX 75246, USA
| | - Bo Shen
- Children's Research Institute, UT Southwestern Medical Center Dallas, TX 75235, USA
| | - William Stenberg
- Department of Comprehensive Dentistry, College of Dentistry, Texas A&M University, Dallas, TX 75246, USA
| | - Yang Chai
- Center for Craniofacial Molecular Biology, University of Southern California, Herman Ostrow School of Dentistry, Los Angeles, CA 90089, USA
| | - Woo-Ping Ge
- Chinese Institute for Brain Research, Beijing 102206, China
| | - Jian Q Feng
- Department of Biomedical Sciences, College of Dentistry, Texas A&M University, Dallas, TX 75246, USA
| | - Hu Zhao
- Department of Comprehensive Dentistry, College of Dentistry, Texas A&M University, Dallas, TX 75246, USA.
| |
Collapse
|
199
|
Zhang D, Zhang S, Wang J, Li Q, Xue H, Sheng R, Xiong Q, Qi X, Wen J, Fan Y, Zhou B, Yuan Q. LepR-Expressing Stem Cells Are Essential for Alveolar Bone Regeneration. J Dent Res 2020; 99:1279-1286. [PMID: 32585118 DOI: 10.1177/0022034520932834] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Stem cells play a critical role in bone regeneration. Multiple populations of skeletal stem cells have been identified in long bone, while their identity and functions in alveolar bone remain unclear. Here, we identified a quiescent leptin receptor–expressing (LepR+) cell population that contributed to intramembranous bone formation. Interestingly, these LepR+ cells became activated in response to tooth extraction and generated the majority of the newly formed bone in extraction sockets. In addition, genetic ablation of LepR+ cells attenuated extraction socket healing. The parabiosis experiments revealed that the LepR+ cells in the healing sockets were derived from resident tissue rather than peripheral blood circulation. Further studies on the mechanism suggested that these LepR+ cells were responsive to parathyroid hormone/parathyroid hormone 1 receptor (PTH/PTH1R) signaling. Collectively, we demonstrate that LepR+ cells, a postnatal skeletal stem cell population, are essential for alveolar bone regeneration of extraction sockets.
Collapse
Affiliation(s)
- D. Zhang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - S. Zhang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - J. Wang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Periodontology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Q. Li
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - H. Xue
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - R. Sheng
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Q. Xiong
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - X. Qi
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - J. Wen
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Y. Fan
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - B.O. Zhou
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai, China
| | - Q. Yuan
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
200
|
Bi R, Yin Q, Mei J, Chen K, Luo X, Fan Y, Zhu S. Identification of human temporomandibular joint fibrocartilage stem cells with distinct chondrogenic capacity. Osteoarthritis Cartilage 2020; 28:842-852. [PMID: 32147536 DOI: 10.1016/j.joca.2020.02.835] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 02/08/2020] [Accepted: 02/26/2020] [Indexed: 02/05/2023]
Abstract
OBJECTIVE This study was aimed to identify the residence of human fibrocartilage stem cells (hFCSCs), characterize their stem cell properties and investigate the functional mechanisms which regulate fibrocartilage stem cells (FCSCs) toward chondrogenic differentiation during cartilage homeostasis and repairing. METHODS Cytological characteristics of hFCSCs and human orofacial mesenchymal stem cells (hOFMSCs) were analyzed. Chondrogenic potential of hFCSCs was compared with hOFMSCs both in vitro and in vivo. Regulatory role of SOX9 during FCSCs chondrogenesis was studied by shRNA interference in vitro, and by GFP+ FCSCs treatment in rat condylar cartilage defect model. SOX9 expression was also examined in temporomandibular joint osteoarthritis (TMJOA) patients' cartilage surface. RESULTS hFCSCs exhibited typical mesenchymal stem cell characteristics, with significantly stronger chondrogenic capability compared to hOFMSCs. Moreover, hFCSCs showed remarkably increased expression of SOX9. During cartilage pellet culture, there was stronger SOX9 expression in hFCSCs than hOFMSCs. SOX9 shRNA interference downregulated chondrogenic capability of hFCSCs in vitro, as well as disrupting migration and chondrogenic differentiation of GFP+ FCSCs toward mature chondrocytes in rat condylar cartilage defect. Of note, SOX9 expression was also found suppressed in the condylar superficial zone of TMJOA patients. CONCLUSION We found the existence of FCSCs in human TMJ cartilage, and characterized their distinct stem cell features. SOX9 is essential for hFCSCs chondrogenic differentiation, and a comprehensive understanding of the regulatory role of SOX9 in hFCSCs would be important for exploring potential intervention strategy of condylar cartilage degradation during TMJ disorders.
Collapse
Affiliation(s)
- R Bi
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthognathic and TMJ Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - Q Yin
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthognathic and TMJ Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - J Mei
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthognathic and TMJ Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - K Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthognathic and TMJ Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - X Luo
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthognathic and TMJ Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - Y Fan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Operative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - S Zhu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthognathic and TMJ Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|