1
|
Feng J, Janečková E, Guo T, Ziaei H, Zhang M, Geng JJ, Cha S, Araujo-Villalba A, Liu M, Ho TV, Chai Y. High-resolution spatial transcriptomics and cell lineage analysis reveal spatiotemporal cell fate determination during craniofacial development. Nat Commun 2025; 16:4396. [PMID: 40355462 DOI: 10.1038/s41467-025-59206-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 04/14/2025] [Indexed: 05/14/2025] Open
Abstract
The differentiation of post-migratory cranial neural crest cells (CNCCs) into distinct mesenchymal lineages is crucial for craniofacial development. Here we report a high-resolution spatiotemporal transcriptomic and cell-type atlas of CNCC-derived mesenchymal lineage diversification during mouse palatogenesis. We systematically defined each mesenchymal cell type by mapping their transcriptomic profiles to spatial identities. Integrative analysis of spatial transcriptomic data from E12.5 to E15.5 further revealed mesenchymal lineage establishment at or prior to initiation of palatogenesis. We also identified a heterogeneous Sox9+ mesenchymal progenitor population at the onset of palatal development, with subpopulations already activating early lineage-specific markers. In vivo lineage tracing using these early lineage-specific markers demonstrated that distinct mesenchymal populations are established as early as E10.5 to E11.5, preceding palatal development, and contribute to their respective lineages. Together, our findings reveal the comprehensive, dynamic molecular and cellular landscape of palate development and shed light on cell fate regulation during embryogenesis.
Collapse
Affiliation(s)
- Jifan Feng
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, California, 90033, USA
| | - Eva Janečková
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, California, 90033, USA
| | - Tingwei Guo
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, California, 90033, USA
| | - Heliya Ziaei
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, California, 90033, USA
| | - Mingyi Zhang
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, California, 90033, USA
| | - Jessica Junyan Geng
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, California, 90033, USA
| | - Sa Cha
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, California, 90033, USA
| | - Angelita Araujo-Villalba
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, California, 90033, USA
| | - Mengmeng Liu
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, California, 90033, USA
| | - Thach-Vu Ho
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, California, 90033, USA
| | - Yang Chai
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, California, 90033, USA.
| |
Collapse
|
2
|
Kanai SM, Garcia CR, Augustus MR, Sharafeldeen SA, Brooks EP, Sucharov J, Lencer ES, Nichols JT, Clouthier DE. The Gq/11 family of Gα subunits is necessary and sufficient for lower jaw development. Development 2025; 152:dev204396. [PMID: 40171762 PMCID: PMC12045641 DOI: 10.1242/dev.204396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 03/18/2025] [Indexed: 04/04/2025]
Abstract
Vertebrate jaw development is coordinated by highly conserved ligand-receptor systems such as the peptide ligand Endothelin 1 (Edn1) and Endothelin receptor type A (Ednra), which are required for patterning of lower jaw structures. The Edn1/Ednra signaling pathway establishes the identity of lower jaw progenitor cells by regulating expression of numerous patterning genes, but the intracellular signaling mechanisms linking receptor activation to gene regulation remain poorly understood. As a first step towards elucidating this mechanism, we examined the function of the Gq/11 family of Gα subunits in zebrafish using pharmacological inhibition and genetic ablation of Gq/11 activity, and transgenic induction of a constitutively active Gq protein in edn1-/- embryos. Genetic loss of Gq/11 activity fully recapitulated the edn1-/- phenotype, with genes encoding G11 being most essential. Furthermore, inducing Gq activity in edn1-/- embryos not only restored Edn1/Ednra-dependent jaw structures and gene expression signatures but also caused homeosis of the upper jaw structure into a lower jaw-like structure. These results indicate that Gq/11 is necessary and sufficient to mediate the lower jaw patterning mechanism for Ednra in zebrafish.
Collapse
Affiliation(s)
- Stanley M. Kanai
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80108, USA
| | - Chloe R. Garcia
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80108, USA
| | - MaCalia R. Augustus
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80108, USA
| | - Shujan A. Sharafeldeen
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80108, USA
| | - Elliott P. Brooks
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80108, USA
| | - Juliana Sucharov
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80108, USA
| | - Ezra S. Lencer
- Department of Biology, Lafayette College, Easton, PA 18042, USA
| | - James T. Nichols
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80108, USA
| | - David E. Clouthier
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80108, USA
| |
Collapse
|
3
|
Moore Zajic EL, Zhao R, McKinney MC, Yi K, Wood C, Trainor PA. Cell extrusion drives neural crest cell delamination. Proc Natl Acad Sci U S A 2025; 122:e2416566122. [PMID: 40063802 PMCID: PMC11929498 DOI: 10.1073/pnas.2416566122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 01/17/2025] [Indexed: 03/15/2025] Open
Abstract
Neural crest cells (NCC) comprise a heterogeneous population of cells with variable potency that contribute to nearly every tissue and organ throughout the body. Considered unique to vertebrates, NCC are transiently generated within the dorsolateral region of the neural plate or neural tube during neurulation. Their delamination and migration are crucial for embryo development as NCC differentiation is influenced by their final resting locations. Previous work in avian and aquatic species revealed that NCC delaminate via an epithelial-mesenchymal transition (EMT), which transforms these progenitor cells from static polarized epithelial cells into migratory mesenchymal cells with fluid front and back polarity. However, the cellular and molecular mechanisms facilitating NCC delamination in mammals are poorly understood. Through time-lapse imaging of NCC delamination in mouse embryos, we identified a subset of cells that exit the neuroepithelium as isolated round cells, which then halt for a short period prior to acquiring the mesenchymal migratory morphology classically associated with delaminating NCC. High-magnification imaging and protein localization analyses of the cytoskeleton, together with measurements of pressure and tension of delaminating NCC and neighboring neuroepithelial cells, revealed that round NCC are extruded from the neuroepithelium prior to completion of EMT. Furthermore, cranial NCC are extruded through activation of the mechanosensitive ion channel, PIEZO1. Our results support a model in which cell density, pressure, and tension in the neuroepithelium result in activation of the live cell extrusion pathway and delamination of a subpopulation of NCC in parallel with EMT, which has implications for cell delamination in development and disease.
Collapse
Affiliation(s)
| | - Ruonan Zhao
- Stowers Institute for Medical Research, Kansas City, MO64110
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS66160
| | | | - Kexi Yi
- Stowers Institute for Medical Research, Kansas City, MO64110
| | | | - Paul A. Trainor
- Stowers Institute for Medical Research, Kansas City, MO64110
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS66160
| |
Collapse
|
4
|
Aoki H, Tomita H, Hara A, Kunisada T. Conditional heterozygous loss of kit receptor tyrosine kinase in neural crest cell lineage is associated with midline cleft lip and bifid nose deformity. J Oral Biosci 2025; 67:100572. [PMID: 39426597 DOI: 10.1016/j.job.2024.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 10/15/2024] [Accepted: 10/16/2024] [Indexed: 10/21/2024]
Abstract
OBJECTIVES The receptor tyrosine kinase Kit is expressed in cells derived from the trunk neural crest (NC), such as melanocytes; however, its role in cranial NC cell development is not fully understood. METHODS We investigated the effects of the heterozygous loss of Kit in NC cells during embryonic development by mating Kit2lox/+ mice with Wnt1-Cre mice to produce Wnt1-Cre; Kit2lox/+ embryos. In addition, Wnt1-Cre mice were mated with Rosa26R-yellow fluorescent protein (YFP) mice to visualize the tissue regions expressing Cre recombinase. Histological studies of the craniofacial regions of these mice were performed using samples from embryonic day (E) 12.5 and postnatal day (P) 1. Cellular apoptosis and proliferation were both analyzed through the immunostaining of tissue sections collected on E13.5 and E14.5 using anti-cleaved caspase 3 (CC3) to detect apoptosis and anti-Ki67 to detect proliferation. Cells from YFP-positive tissue regions of the facial areas of Wnt1-Cre; Kit+/+; Rosa26R-YFP embryos and Wnt1-Cre; Kit2lox/+; Rosa26R-YFP embryos collected on E12.5 and E15.5 were cultured and evaluated for cell proliferation. RESULTS Compared with control littermates, Wnt1-Cre; Kit2lox/+ embryos exhibited midline cleft lip and bifid nose deformities. Substantial early (P1) postnatal lethality was observed in Wnt1-Cre; Kit2lox/+ mice, with none surviving to 3 weeks of age. YFP-positive cells from the maxillary regions of Wnt1-Cre; Kit2lox/+; Rosa26R-YFP embryos exhibited defective cell growth and self-renewal in vitro. CONCLUSION Conditional heterozygous loss of Kit in Wnt1-Cre; Kit2lox/+ embryos is associated with craniofacial dysplasia and exhibit defective NC development in vitro and in vivo.
Collapse
Affiliation(s)
- Hitomi Aoki
- Department of Stem Cell and Regenerative Medicine, Gifu University Graduate School of Medicine, 1-1, Yanagido, Gifu, 501-1194, Japan.
| | - Hiroyuki Tomita
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, 1-1, Yanagido, Gifu, 501-1194, Japan
| | - Akira Hara
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, 1-1, Yanagido, Gifu, 501-1194, Japan
| | - Takahiro Kunisada
- Department of Stem Cell and Regenerative Medicine, Gifu University Graduate School of Medicine, 1-1, Yanagido, Gifu, 501-1194, Japan
| |
Collapse
|
5
|
Morita K, Wang J, Okamoto K, Iwata T. The next generation of regenerative dentistry: From tooth development biology to periodontal tissue, dental pulp, and whole tooth reconstruction in the clinical setting. Regen Ther 2025; 28:333-344. [PMID: 39885872 PMCID: PMC11780712 DOI: 10.1016/j.reth.2025.01.002] [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: 09/29/2024] [Revised: 12/08/2024] [Accepted: 01/04/2025] [Indexed: 02/01/2025] Open
Abstract
In modern dentistry, prosthetic approaches such as implants and dentures have been developed as symptomatic solutions for tooth loss. However, the complete regeneration of teeth and periodontal tissue, an ultimate aspiration of humanity, remains unachieved. Recent advancements in fundamental scientific technologies, including single-cell RNA sequencing and spatial transcriptomics, have significantly advanced our molecular understanding of tooth development, paving the way toward achieving this goal. This review summarizes the fundamental processes of tooth development in humans and mice, recent findings from basic research, and current clinical applications in dental regenerative medicine, including periodontal, alveolar bone, and dental pulp regeneration using cellular approaches. Building on accumulated scientific knowledge, the complete regeneration of teeth and periodontal tissues may be achievable in the near future. We discuss the potential of emerging approaches, such as organoids derived from pluripotent stem cells and xenotransplantation using genetically modified animals, to transform dental medicine. These innovative concepts and integrated technologies hold the promise of enabling the regeneration of fully functional teeth and periodontal tissues.
Collapse
Affiliation(s)
- Kazuki Morita
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo (Science Tokyo), 1-5-45, Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Jiacheng Wang
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo (Science Tokyo), 1-5-45, Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Keisuke Okamoto
- Health Science Research and Development Center (HeRD), Institute of Science Tokyo (Science Tokyo), 1-5-45, Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Takanori Iwata
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo (Science Tokyo), 1-5-45, Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| |
Collapse
|
6
|
Samiei M, Harmsen MC, Abdolahinia ED, Barar J, Petridis X. Scaffold-Free Strategies in Dental Pulp/Dentine Tissue Engineering: Current Status and Implications for Regenerative Biological Processes. Bioengineering (Basel) 2025; 12:198. [PMID: 40001717 PMCID: PMC11851408 DOI: 10.3390/bioengineering12020198] [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: 01/31/2025] [Revised: 02/16/2025] [Accepted: 02/17/2025] [Indexed: 02/27/2025] Open
Abstract
Conventionally, root canal treatment is performed when the dental pulp is severely damaged or lost due to dental trauma or bacterial endodontic infections. This treatment involves removing the compromised or infected pulp tissue, disinfecting the root canal system, and sealing it with inert, non-degradable materials. However, contemporary endodontic treatment has shifted from merely obturating the root canal system with inert materials to guiding endodontic tissue regeneration through biological approaches. The ultimate goal of regenerative endodontics is to restore dental pulp tissue with structural organization and functional characteristics akin to the native pulp, leveraging advancements in tissue engineering and biomaterial sciences. Dental pulp tissue engineering commonly employs scaffold-based strategies, utilizing biomaterials as initial platforms for cell and growth factor delivery, which subsequently act as scaffolds for cell proliferation, differentiation and maturation. However, cells possess an intrinsic capacity for self-organization into spheroids and can generate their own extracellular matrix, eliminating the need for external scaffolds. This self-assembling property presents a promising alternative for scaffold-free dental pulp engineering, addressing limitations associated with biomaterial-based approaches. This review provides a comprehensive overview of cell-based, self-assembling and scaffold-free approaches in dental pulp tissue engineering, highlighting their potential advantages and challenges in advancing regenerative endodontics.
Collapse
Affiliation(s)
- Mohammad Samiei
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, P.O. Box 30.001, 9700 RB Groningen, The Netherlands; (M.S.); (M.C.H.)
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz 5165665811, Iran
| | - Martin Conrad Harmsen
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, P.O. Box 30.001, 9700 RB Groningen, The Netherlands; (M.S.); (M.C.H.)
| | - Elaheh Dalir Abdolahinia
- Department of Oral Science and Translation Research, College of Dental Medicine, Nova Southeastern University, Fort Lauderdale, FL 33314, USA;
| | - Jaleh Barar
- Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA;
| | - Xenos Petridis
- Department of Endodontics, Section of Dental Pathology & Therapeutics, School of Dentistry, National and Kapodistrian University of Athens, 115 27 Athens, Greece
- Department of Endodontology, Section of Fundamental Dentistry, Center for Dentistry and Oral Hygiene, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
| |
Collapse
|
7
|
Zhang J, Gao J, Zeng X, Wang Z, Chen C, Rong C, Li S, Cai L, Wang L, Zhang L, Tian Z. A novel Cdc42-YAP-fibronectin signaling axis regulates ameloblast differentiation during early enamel formation. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167570. [PMID: 39547518 DOI: 10.1016/j.bbadis.2024.167570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 09/22/2024] [Accepted: 11/07/2024] [Indexed: 11/17/2024]
Abstract
Enamel formation is a developmental event governed by intricate molecular signal pathways. Cdc42 is proven to regulate enamel development yet its underlying role and molecular mechanism in early amelogenesis remain elusive. The extracellular matrix of tooth germ basement membrane is critical for the regulation of ameloblast differentiation. Present study investigated whether Cdc42 influences amelogenesis by affecting ECM synthesis and how Cdc42 regulates ameloblasts differentiation. Epithelial-specific knockout of Cdc42 (Cdc42-cKO) mice model was employed to study the ECM expression including Fibronectin (Fn) and amelogenesis markers. Cdc42-cKO mice results in retarded ameloblast differentiation and enamel matrix decrease. Fn synthesis in the enamel organ and basal membrane was totally diminished along with Cdc42 knockdown. YAP acting as the Cdc42 downstream transcription factor, its distribution in ameloblasts was synchronously attenuated by Cdc42 knockdown and nuclear localization progressively decreased with tooth germ development. Cdc42 unidirectionally controls the Fn synthesis via YAP regulation. Overall, ameloblast differentiation inhibition by silencing of Cdc42 was successfully rescued by YAP activation. We demonstrated that Cdc42 as an initiator, mediated downstream pathway through transcriptional activator YAP, thereby affecting ameloblast differentiation by controlling Fn synthesis. The Cdc42-YAP-Fn signaling axis are elucidated to act critical role during the early amelogenesis.
Collapse
Affiliation(s)
- Jiayi Zhang
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China; School of Stomatology, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Jingyi Gao
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China; School of Stomatology, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Xiangliang Zeng
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China; School of Stomatology, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Zijie Wang
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China; School of Stomatology, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Chuying Chen
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China; School of Stomatology, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Chao Rong
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China; School of Stomatology, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Shaowei Li
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China; School of Stomatology, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Lingxuan Cai
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China; School of Stomatology, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Luchen Wang
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China; School of Stomatology, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Lin Zhang
- Department of Histology and Embryology, Guangdong Provincial Key Laboratory of Tissue Construction and Detection, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.
| | - Zhihui Tian
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China; School of Stomatology, Southern Medical University, Guangzhou, Guangdong Province, China.
| |
Collapse
|
8
|
Barlow LA. Development of ectodermal and endodermal taste buds. Dev Biol 2025; 518:20-27. [PMID: 39486632 PMCID: PMC11703678 DOI: 10.1016/j.ydbio.2024.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 09/20/2024] [Accepted: 10/27/2024] [Indexed: 11/04/2024]
Abstract
The sense of taste is mediated primarily by taste buds on the tongue. These multicellular sensory organs are induced, patterned and become innervated during embryogenesis such that a functional taste system is present at birth when animals begin to feed. While taste buds have been considered ectodermal appendages, this is only partly accurate as only fungiform taste buds in the anterior tongue arise from the ectoderm. Taste buds found in the posterior tongue actually derive from endoderm. Nonetheless, both anterior and posterior buds are functionally similar, despite their disparate embryonic origins. In this review, I compare the development of ectodermal vs endodermal taste buds, highlighting the many differences in the cellular and molecular genetic mechanisms governing their formation.
Collapse
Affiliation(s)
- Linda A Barlow
- Department of Cell and Developmental Biology, Rocky Mountain Taste and Smell Center, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA.
| |
Collapse
|
9
|
Shukla S, Jana S, Sanford N, Lee CY, Liu L, Cheng P, Quertermous T, Dichek DA. Single-Cell Transcriptomics Identifies Selective Lineage-Specific Regulation of Genes in Aortic Smooth Muscle Cells in Mice. Arterioscler Thromb Vasc Biol 2025; 45:e15-e29. [PMID: 39744838 PMCID: PMC11875902 DOI: 10.1161/atvbaha.124.321482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 12/18/2024] [Indexed: 02/11/2025]
Abstract
BACKGROUND Smooth muscle cells (SMCs) of the proximal thoracic aorta are derived from second heart field (SHF) and cardiac neural crest (CNC) lineages. Recent studies, both in vitro and in vivo, have implied relevance of lineage-specific SMC functions in the pathophysiology of thoracic aortic diseases; however, whether 2 lineage-derived SMCs have any predisposed transcriptional differences in the control aorta remains unexplored. METHODS Single-cell RNA sequencing and single-nucleus assay for transposase-accessible chromatin sequencing were performed on isolated cells from the aortic root and ascending aortas of 14-week-old SHF-traced (Mef2c-Cre+/0-Yfp+/0) and CNC-traced (Wnt1-Cre+/0-Yfp+/0) male mice. RNA in situ hybridization was performed for spatial expression of selected differentially expressed genes (DEGs) of both lineages. RESULTS Lineage stratification of SMCs in the proximal thoracic aorta was identified using antibody-based immunofluorescence staining. Single-cell RNA sequencing recognized 12 consistently upregulated DEGs (Des, Tnnt2, Hand2os1, Psd, Gpc3, Meis2, Dcn, Gm34030, Palld, Nrtn, Lum, and Cfh) in SHF-derived SMCs and 9 consistently upregulated DEGs (Ccn5, Ccdc42, Tes, Eln, Aebp1, Galnt6, Ccn2, Aopep, and Wtip) in CNC-derived SMCs. RNA in situ hybridization validated upregulated expressions of selective SHF-specific DEGs at the aortic root. We found SHF-derived SMCs contain a distinct, large subpopulation of SMCs that is enriched with Des and Tnnt2 expressions. Single-nucleus assay for transposase-accessible chromatin analysis further confirmed higher chromosomal accessibility for upregulated DEGs of SHF-derived SMCs. CONCLUSIONS The present study recognizes the presence of limited but distinct transcriptomic differences between CNC-derived and SHF-derived SMCs in the control proximal thoracic aorta.
Collapse
Affiliation(s)
- Shalabh Shukla
- Division of Cardiology, Department of Medicine, University of Washington, WA, USA
| | - Sayantan Jana
- Division of Cardiology, Department of Medicine, University of Washington, WA, USA
| | - Nicole Sanford
- Division of Cardiology, Department of Medicine, University of Washington, WA, USA
| | - Chloe Y Lee
- Division of Cardiology, Department of Medicine, University of Washington, WA, USA
| | - Li Liu
- Division of Cardiology, Department of Medicine, University of Washington, WA, USA
| | - Paul Cheng
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University, CA, USA
| | - Thomas Quertermous
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University, CA, USA
| | - David A Dichek
- Division of Cardiology, Department of Medicine, University of Washington, WA, USA
| |
Collapse
|
10
|
Ramos R, Pham KT, Prince RC, Leiser-Miller LB, Prasad MS, Wang X, Nordberg RC, Bielajew BJ, Hu JC, Yamaga K, Oh JW, Peng T, Datta R, Astrowskaja A, Almet AA, Burns JT, Liu Y, Guerrero-Juarez CF, Tran BQ, Chu YL, Nguyen AM, Hsi TC, Lim NTL, Schoeniger S, Liu R, Pai YL, Vadivel CK, Ingleby S, McKechnie AE, van Breukelen F, Hoehn KL, Rasweiler JJ, Kohara M, Loughry WJ, Weldy SH, Cosper R, Yang CC, Lin SJ, Cooper KL, Santana SE, Bradley JE, Kiebish MA, Digman M, James DE, Merrill AE, Nie Q, Schilling TF, Astrowski AA, Potma EO, García-Castro MI, Athanasiou KA, Behringer RR, Plikus MV. Superstable lipid vacuoles endow cartilage with its shape and biomechanics. Science 2025; 387:eads9960. [PMID: 39787221 DOI: 10.1126/science.ads9960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 11/13/2024] [Indexed: 01/12/2025]
Abstract
Conventionally, the size, shape, and biomechanics of cartilages are determined by their voluminous extracellular matrix. By contrast, we found that multiple murine cartilages consist of lipid-filled cells called lipochondrocytes. Despite resembling adipocytes, lipochondrocytes were molecularly distinct and produced lipids exclusively through de novo lipogenesis. Consequently, lipochondrocytes grew uniform lipid droplets that resisted systemic lipid surges and did not enlarge upon obesity. Lipochondrocytes also lacked lipid mobilization factors, which enabled exceptional vacuole stability and protected cartilage from shrinking upon starvation. Lipid droplets modulated lipocartilage biomechanics by decreasing the tissue's stiffness, strength, and resilience. Lipochondrocytes were found in multiple mammals, including humans, but not in nonmammalian tetrapods. Thus, analogous to bubble wrap, superstable lipid vacuoles confer skeletal tissue with cartilage-like properties without "packing foam-like" extracellular matrix.
Collapse
Affiliation(s)
- Raul Ramos
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA, USA
| | - Kim T Pham
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA, USA
| | - Richard C Prince
- Department of Chemistry, University of California, Irvine, Irvine, CA, USA
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, USA
| | | | - Maneeshi S Prasad
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, USA
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - Xiaojie Wang
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA, USA
| | - Rachel C Nordberg
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, USA
| | - Benjamin J Bielajew
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, USA
| | - Jerry C Hu
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, USA
| | - Kosuke Yamaga
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA, USA
| | - Ji Won Oh
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA, USA
- Department of Anatomy, College of Medicine, Yonsei University, Seoul, Republic of Korea
- Department of Anatomy, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- Biomedical Research Institute, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Tao Peng
- Department of Mathematics, University of California, Irvine, Irvine, CA, USA
- Center for Complex Biological Systems, University of California, Irvine, Irvine, CA, USA
| | - Rupsa Datta
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, USA
| | - Aksana Astrowskaja
- Scientific Research Laboratory of Molecular Medicine, Grodna State Medical University, Grodna, Belarus
| | - Axel A Almet
- Department of Mathematics, University of California, Irvine, Irvine, CA, USA
- NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA, USA
| | - John T Burns
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA, USA
| | - Yuchen Liu
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA, USA
| | - Christian Fernando Guerrero-Juarez
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA, USA
- Department of Mathematics, University of California, Irvine, Irvine, CA, USA
- Center for Complex Biological Systems, University of California, Irvine, Irvine, CA, USA
| | - Bryant Q Tran
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA, USA
| | - Yi-Lin Chu
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA, USA
| | - Anh M Nguyen
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA, USA
| | - Tsai-Ching Hsi
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA, USA
| | - Norman T-L Lim
- National Institute of Education, Singapore, Republic of Singapore
| | - Sandra Schoeniger
- Institute of Veterinary Pathology, Leipzig University, Leipzig, Germany
- Discovery Life Sciences Biomarker Services GmbH, Kassel, Germany
| | - Ruiqi Liu
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA, USA
| | - Yun-Ling Pai
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA, USA
- Institute of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, Taipei, Taiwan
| | - Chella K Vadivel
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA, USA
- LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | | | - Andrew E McKechnie
- Mammal Research Institute, Department of Zoology and Entomology, University of Pretoria, Hatfield, South Africa
- South African National Biodiversity Institute, Pretoria, South Africa
| | - Frank van Breukelen
- School of Life Sciences, University of Nevada, Las Vegas, Las Vegas, NV, USA
| | - Kyle L Hoehn
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, Australia
| | - John J Rasweiler
- Department of Obstetrics and Gynecology, SUNY Downstate Medical Center, New York, NY, USA
| | - Michinori Kohara
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | | | - Scott H Weldy
- Serrano Animal and Bird Hospital, Lake Forest, CA, USA
| | | | - Chao-Chun Yang
- Department of Dermatology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- International Center for Wound Repair and Regeneration, National Cheng Kung University, Tainan, Taiwan
| | - Sung-Jan Lin
- Institute of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, Taipei, Taiwan
| | - Kimberly L Cooper
- Department of Cell and Developmental Biology, University of California, San Diego, La Jolla, CA, USA
| | - Sharlene E Santana
- Department of Biology, University of Washington, Seattle, WA, USA
- Department of Mammalogy, Burke Museum, University of Washington, Seattle, WA, USA
| | - Jeffrey E Bradley
- Department of Mammalogy, Burke Museum, University of Washington, Seattle, WA, USA
| | | | - Michelle Digman
- Center for Complex Biological Systems, University of California, Irvine, Irvine, CA, USA
- Department of Chemical Engineering and Materials Science, University of California, Irvine, Irvine, CA, USA
| | - David E James
- Charles Perkins Centre, School of Life and Environmental Sciences and School of Medical Sciences, University of Sydney, Sydney, NSW, Australia
| | - Amy E Merrill
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, USA
| | - Qing Nie
- Department of Mathematics, University of California, Irvine, Irvine, CA, USA
- Center for Complex Biological Systems, University of California, Irvine, Irvine, CA, USA
- NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA, USA
| | - Thomas F Schilling
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
- Center for Complex Biological Systems, University of California, Irvine, Irvine, CA, USA
- NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA, USA
| | | | - Eric O Potma
- Department of Chemistry, University of California, Irvine, Irvine, CA, USA
| | - Martín I García-Castro
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, USA
| | - Kyriacos A Athanasiou
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, USA
| | - Richard R Behringer
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Maksim V Plikus
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA, USA
- Center for Complex Biological Systems, University of California, Irvine, Irvine, CA, USA
- NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA, USA
| |
Collapse
|
11
|
Takeuchi R, Takechi M, Namangkalakul W, Ninomiya Y, Furutera T, Aoto K, Koyabu D, Adachi N, Hayashi K, Okabe M, Iseki S. The role of sonic hedgehog signaling in the oropharyngeal epithelium during jaw development. Congenit Anom (Kyoto) 2025; 65:e70001. [PMID: 39727066 DOI: 10.1111/cga.70001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 11/29/2024] [Accepted: 12/07/2024] [Indexed: 12/28/2024]
Abstract
Sonic hedgehog (Shh) is expressed in the oropharyngeal epithelium, including the frontonasal ectodermal zone (FEZ), which is defined as the boundary between Shh and Fgf8 expression domains in the frontonasal epithelium. To investigate the role of SHH signaling from the oropharyngeal epithelium, we generated mice in which Shh expression is specifically deleted in the oropharyngeal epithelium (Isl1-Cre; Shhf/f). In the mutant mouse, Shh expression was excised in the oropharyngeal epithelium as well as FEZ and ventral forebrain, consistent with the expression pattern of Isl1. Isl1-Cre; Shhf/f mice exhibited a complete loss of lower jaw components and a malformed upper jaw with defects in the cranial base and secondary palate. Massive cell death was observed in the mandibular process at embryonic day (E) 9.5 and E10.5, while mild cell death was observed in the lambdoidal region (the fusion area in the maxillary, lateral nasal, and medial nasal processes) at E10.5. An RNA-seq analysis revealed that Satb2, a gene involved in cell survival during jaw formation, was downregulated in the lambdoidal region in Isl1-Cre; Shhf/f mice. These results suggest that Shh expression in the FEZ is required for cell survival and skeletogenesis in the lambdoidal region during the development of the upper jaw and that the developmental control governed by SHH signaling is different between upper and lower jaws.
Collapse
Affiliation(s)
- Rika Takeuchi
- Department of Molecular Craniofacial Embryology and Oral Histology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
- Department of Dentistry and Oral Surgery, The Jikei University School of Medicine, Tokyo, Japan
- Department of Anatomy, The Jikei University School of Medicine, Tokyo, Japan
| | - Masaki Takechi
- Department of Molecular Craniofacial Embryology and Oral Histology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
- Department of Anatomy and Life Structure, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Worachat Namangkalakul
- Department of Molecular Craniofacial Embryology and Oral Histology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
- Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Youichirou Ninomiya
- Research Center for Medical Bigdata, Research Organization of Information and Systems, National Institute of Informatics, Tokyo, Japan
| | - Toshiko Furutera
- Department of Molecular Craniofacial Embryology and Oral Histology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
- Department of Anatomy and Life Structure, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Kazushi Aoto
- Central Laboratory, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Daisuke Koyabu
- Department of Molecular Craniofacial Embryology and Oral Histology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
- Research and Development Center for Precision Medicine, University of Tsukuba, Ibaraki, Japan
| | - Noritaka Adachi
- Department of Molecular Craniofacial Embryology and Oral Histology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Katsuhiko Hayashi
- Department of Dentistry and Oral Surgery, The Jikei University School of Medicine, Tokyo, Japan
| | - Masataka Okabe
- Department of Anatomy, The Jikei University School of Medicine, Tokyo, Japan
| | - Sachiko Iseki
- Department of Molecular Craniofacial Embryology and Oral Histology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| |
Collapse
|
12
|
Uptegrove A, Chen C, Sahagun-Bisson M, Kulkarni AK, Louie KW, Ueharu H, Mishina Y, Omi-Sugihara M. Influence of bone morphogenetic protein (BMP) signaling and masticatory load on morphological alterations of the mouse mandible during postnatal development. Arch Oral Biol 2025; 169:106096. [PMID: 39341045 PMCID: PMC11609011 DOI: 10.1016/j.archoralbio.2024.106096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 07/15/2024] [Accepted: 09/23/2024] [Indexed: 09/30/2024]
Abstract
OBJECTIVE Bone homeostasis relies on several contributing factors, encompassing growth factors and mechanical stimuli. While bone morphogenetic protein (BMP) signaling is acknowledged for its essential role in skeletal development, its specific impact on mandibular morphogenesis remains unexplored. Here, we investigated the involvement of BMP signaling and mechanical loading through mastication in postnatal mandibular morphogenesis. DESIGN We employed conditional deletion of Bmpr1a in osteoblasts and chondrocytes via Osterix-Cre. Cre activity was induced at birth for the 3-week group and at three weeks for the 9-week and 12-week groups, respectively. The conditional knockout (cKO) and control mice were given either a regular diet (hard diet, HD) or a powdered diet (soft diet, SD) from 3 weeks until sample collection, followed by micro-CT and histological analysis. RESULTS The cKO mice exhibited shorter anterior lengths and a posteriorly inclined ramus across all age groups compared to the control mice. The cKO mice displayed an enlarged hypertrophic cartilage area along with fewer osteoclast numbers in the subchondral bone of the condyle compared to the control group at three weeks, followed by a reduction in the cartilage area in the posterior region at twelve weeks. Superimposed imaging and histomorphometrical analysis of the condyle revealed that BMP signaling primarily affects the posterior part of the condyle, while mastication affects the anterior part. CONCLUSIONS Using 3D landmark-based geometric morphometrics and histological assessments of the mandible, we demonstrated that BMP signaling and mechanical loading reciprocally contribute to the morphological alterations of the mandible and condyle during postnatal development.
Collapse
Affiliation(s)
- Amber Uptegrove
- Department of Biologic and Materials Sciences & Prosthodontics, University of Michigan School of Dentistry, Ann Arbor, USA
| | - Coral Chen
- Department of Biologic and Materials Sciences & Prosthodontics, University of Michigan School of Dentistry, Ann Arbor, USA
| | - Madison Sahagun-Bisson
- Department of Biologic and Materials Sciences & Prosthodontics, University of Michigan School of Dentistry, Ann Arbor, USA
| | - Anshul K Kulkarni
- Department of Biologic and Materials Sciences & Prosthodontics, University of Michigan School of Dentistry, Ann Arbor, USA
| | - Ke'ale W Louie
- Department of Biologic and Materials Sciences & Prosthodontics, University of Michigan School of Dentistry, Ann Arbor, USA
| | - Hiroki Ueharu
- Department of Biologic and Materials Sciences & Prosthodontics, University of Michigan School of Dentistry, Ann Arbor, USA
| | - Yuji Mishina
- Department of Biologic and Materials Sciences & Prosthodontics, University of Michigan School of Dentistry, Ann Arbor, USA.
| | - Maiko Omi-Sugihara
- Department of Biologic and Materials Sciences & Prosthodontics, University of Michigan School of Dentistry, Ann Arbor, USA; Department of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Osaka University, Osaka, Japan.
| |
Collapse
|
13
|
Sun M, Yu Y, Zhang W, Ding Y, Li A, Li Y. Extracellular vesicles derived from dental follicle stem cells regulate tooth eruption by inhibiting osteoclast differentiation. Front Cell Dev Biol 2024; 12:1503481. [PMID: 39834384 PMCID: PMC11744031 DOI: 10.3389/fcell.2024.1503481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 12/03/2024] [Indexed: 01/22/2025] Open
Abstract
Tooth eruption as a crucial part in tooth development and regeneration is accompanied by ongoing osteogenesis and osteoclast activity. The dental follicle (DF) surrounding the developing tooth harbors dental follicle stem cells (DFSCs) which play a crucial role in maintaining bone remodeling. However, the mechanisms through which they regulate the balance between osteogenesis and osteoclast activity during tooth eruption remain poorly understood. Notably, extracellular vesicles (EVs) in bone homeostasis are considered essential. Our study revealed that the DFSCs could modulate tooth eruption by inhibiting osteoclast differentiation via EVs. Further investigation showed that EVs from DFSCs could inhibit osteoclast differentiation through the ANXA1-PPARγ-CEBPα pathway. Animal experiments indicated that EVs from DFSCs and the cargo ANXA1 affected tooth eruption. In summary, this study suggests the critical role of the dental follicle in tooth eruption through EVs, which may provide therapeutic targets for abnormal tooth eruption and effective approaches for the eruption of regenerated teeth.
Collapse
Affiliation(s)
- Meng Sun
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi’an Jiaotong University, Xi’an, China
- Department of Periodontology, College of Stomatology, Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Yiru Yu
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi’an Jiaotong University, Xi’an, China
- Department of Periodontology, College of Stomatology, Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Weixing Zhang
- Department of Periodontology, College of Stomatology, Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Yi Ding
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi’an Jiaotong University, Xi’an, China
- Department of Periodontology, College of Stomatology, Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Ang Li
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi’an Jiaotong University, Xi’an, China
- Department of Periodontology, College of Stomatology, Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Ye Li
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
14
|
Fan Y, Lyu P, Wang J, Wei Y, Li Z, Zhang S, Ouchi T, Jing J, Yuan Q, Rosen CJ, Zhou C. Negative feedback between PTH1R and IGF1 through the Hedgehog pathway in mediating craniofacial bone remodeling. JCI Insight 2024; 10:e183684. [PMID: 39688917 PMCID: PMC11948590 DOI: 10.1172/jci.insight.183684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 12/13/2024] [Indexed: 12/18/2024] Open
Abstract
Regeneration of orofacial bone defects caused by inflammation-related diseases or trauma remains an unmet challenge. Parathyroid hormone 1 receptor (PTH1R) signaling is a key mediator of bone remodeling whereas the regulatory mechanisms of PTH1R signaling in oral bone under homeostatic or inflammatory conditions have not been demonstrated by direct genetic evidence. Here, we observed that deletion of PTH1R in Gli1+ progenitors led to increased osteogenesis and osteoclastogenesis. Single-cell and bulk RNA-Seq analysis revealed that PTH1R suppressed the osteogenic potential of Gli1+ progenitors during inflammation. Moreover, we identified upregulated IGF1 expression upon PTH1R deletion. Dual deletion of IGF1 and PTH1R ameliorated the bone-remodeling phenotypes in PTH1R-deficient mice. Furthermore, in vivo evidence revealed an inverse relationship between PTH1R and Hedgehog signaling, which was responsible for the upregulated IGF1 production. Our work underscored the negative feedback between PTH1R and IGF1 in craniofacial bone turnover and revealed mechanisms modulating orofacial bone remodeling.
Collapse
Affiliation(s)
- Yi Fan
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases
- Department of Cariology and Endodontics
| | - Ping Lyu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases
- Department of Cariology and Endodontics
| | - Jiahe Wang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases
- Department of Pediatric Dentistry, and
| | - Yali Wei
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases
- Department of Cariology and Endodontics
| | - Zucen Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases
- Department of Cariology and Endodontics
| | - Shiwen Zhang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Takehito Ouchi
- Department of Physiology, Tokyo Dental College, Tokyo, Japan
| | - Junjun Jing
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases
| | - Quan Yuan
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | | | - Chenchen Zhou
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases
- Department of Pediatric Dentistry, and
| |
Collapse
|
15
|
Nicklin EF, Cohen KE, Cooper RL, Mitchell G, Fraser GJ. Evolution, development, and regeneration of tooth-like epithelial appendages in sharks. Dev Biol 2024; 516:221-236. [PMID: 39154741 DOI: 10.1016/j.ydbio.2024.08.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 08/05/2024] [Accepted: 08/15/2024] [Indexed: 08/20/2024]
Abstract
Sharks and their relatives are typically covered in highly specialized epithelial appendages embedded in the skin called dermal denticles; ancient tooth-like units (odontodes) composed of dentine and enamel-like tissues. These 'skin teeth' are remarkably similar to oral teeth of vertebrates and share comparable morphological and genetic signatures. Here we review the histological and morphological data from embryonic sharks to uncover characters that unite all tooth-like elements (odontodes), including teeth and skin denticles in sharks. In addition, we review the differences between the skin and oral odontodes that reflect their varied capacity for renewal. Our observations have begun to decipher the developmental and genetic shifts that separate these seemingly similar dental units, including elements of the regenerative nature in both oral teeth and the emerging skin denticles from the small-spotted catshark (Scyliorhinus canicula) and other chondrichthyan models. Ultimately, we ask what defines a tooth at both the molecular and morphological level. These insights aim to help us understand how nature makes, replaces and evolves a vast array of odontodes.
Collapse
Affiliation(s)
- Ella F Nicklin
- Department of Biology, University of Florida, Gainesville, USA
| | - Karly E Cohen
- Department of Biology, University of Florida, Gainesville, USA; Department of Biology, California State University Fullerton, Fullerton, USA
| | - Rory L Cooper
- Department of Genetics and Evolution, University of Geneva, Switzerland
| | - Gianna Mitchell
- Department of Biology, University of Florida, Gainesville, USA
| | - Gareth J Fraser
- Department of Biology, University of Florida, Gainesville, USA.
| |
Collapse
|
16
|
Morriss‐Kay G. A journey in the world of craniofacial development: From 1968 to the future. J Anat 2024; 245:816-828. [PMID: 38760969 PMCID: PMC11547235 DOI: 10.1111/joa.14057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 04/23/2024] [Indexed: 05/20/2024] Open
Abstract
This article is based on my talk at the meeting "3rd Advances in Craniosynostosis: Basic Science to Clinical Practice", held at University College, London, on 25 August 2023. It describes my contribution, together with that of my research team and external collaborators, to the field of craniofacial development. This began with my PhD research on the effects of excess vitamin A in rat embryos, which led to a study of normal as well as abnormal formation of the cranial neural tube. Many techniques for analysing morphogenetic change became available to me over the years: whole embryo culture, scanning and transmission electron microscopy, cell division analysis, immunohistochemistry and biochemical analysis of the extracellular matrix. The molecular revolution of the 1980s, and key collaborations with international research teams, enabled functional interpretation of some of the earlier morphological observations and required a change of experimental species to the mouse. Interactions between the molecular and experimental analysis of craniofacial morphogenesis in my laboratory with specialists in molecular genetics and clinicians brought my research journey near to my original aim: to contribute to a better understanding of the causes of human congenital anomalies.
Collapse
Affiliation(s)
- Gillian Morriss‐Kay
- Emeritus Professor of Developmental Anatomy, Department of Physiology, Anatomy and GeneticsUniversity of OxfordOxfordUK
| |
Collapse
|
17
|
Wang Y, Turkstani H, Alfaifi A, Akintoye SO. Diagnostic and Therapeutic Approaches to Jaw Osteoradionecrosis. Diagnostics (Basel) 2024; 14:2676. [PMID: 39682583 DOI: 10.3390/diagnostics14232676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 11/22/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024] Open
Abstract
Jaw osteoradionecrosis (ORN) is a major complication of head and neck cancer radiotherapy. Treatment complications account for most of the poor outcomes for head and neck cancers and the associated racial health disparities in cancer survivorship. The global incidence of jaw ORN is improving due to pre-radiotherapy patient preparations and improved head and neck cancer radiotherapy protocols. The diagnosis and management of jaw ORN are based on the patient's history and clinical presentation combined with radiological and histopathological tests. Evidence-based jaw ORN therapies focus on preventive, palliative, and surgical principles. However, new and innovative therapeutic approaches based on the cellular and molecular pathophysiological processes of jaw ORN and the jawbone's susceptibility to radiation bone damage are limited. The rationale for this narrative review is to highlight the current diagnostic approaches to jaw ORN and the pathophysiological basis for new therapeutic options for ORN.
Collapse
Affiliation(s)
- Yufan Wang
- Department of Oral and Maxillofacial Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Heba Turkstani
- Department of Oral Medicine, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Afrah Alfaifi
- Department of Oral Medicine, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sunday O Akintoye
- Department of Oral Medicine, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
18
|
Pranter R, Feiner N. Spatiotemporal distribution of neural crest cells in the common wall lizard Podarcis muralis. Dev Dyn 2024. [PMID: 39560189 DOI: 10.1002/dvdy.758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 08/23/2024] [Accepted: 10/06/2024] [Indexed: 11/20/2024] Open
Abstract
BACKGROUND Neural crest cells (NCCs) are migratory embryonic stem cells that give rise to a diverse set of cell types. Here we describe the dynamic distribution of NCCs in developing embryos of the common wall lizard Podarcis muralis inferred from 10 markers. Our aim is to provide insights into the NCC development of lacertid lizards and to infer evolutionary modifications by comparisons to other tetrapods. RESULTS NCC migration is ongoing at oviposition, following three streams in the head and multiple in the trunk. From 21ss, we observe expression patterns indicating the beginning of differentiation toward mesenchymal and neuronal fates. By 35ss, migration is restricted to caudal levels, and fully differentiated chromaffin cells are observed. CONCLUSIONS We find that some markers show patterns that differ from other tetrapods. For example, the antibody HNK-1 labels three NCC streams from the hindbrain while some comparable reptile studies describe four. However, the information emerging from all markers combined shows that the overall spatiotemporal distribution of NCCs in the common wall lizard is largely conserved with that of other tetrapods. Our study highlights the dynamic nature of seemingly canonical marker genes and provides the first description of spatiotemporal NCC dynamics in a lacertid lizard.
Collapse
Affiliation(s)
- Robin Pranter
- Department of Biology, Lund University, Lund, Sweden
| | | |
Collapse
|
19
|
Rinotas V, Stamatakis A, Stergiopoulos A, Bornehag CG, Rüegg J, Armaka M, Kitraki E. Prenatal Exposure to a Human Relevant Mixture of Endocrine-Disrupting Chemicals Affects Mandibular Development in Mice. Int J Mol Sci 2024; 25:12312. [PMID: 39596379 PMCID: PMC11594603 DOI: 10.3390/ijms252212312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 11/05/2024] [Accepted: 11/13/2024] [Indexed: 11/28/2024] Open
Abstract
Mandible is a bony structure of neuroectodermal origin with unique characteristics that support dentition and jaw movements. In the present study, we investigated the effects of gestational exposure to a mixture of endocrine-disrupting chemicals (EDCs) on mandibular growth in mice. The mixture under study (Mixture N1) has been associated with neurodevelopmental effects in both a human cohort and animal studies. Pregnant mice were exposed throughout gestation to 0.5× (times of pregnant women's exposure levels), 10×, 100× and 500× of Mixture N1, or the vehicle, and the mandibles of the male offspring were studied in adulthood. Micro-CT analysis showed non-monotonic effects of Mixture N1 in the distances between specific mandibular landmarks and in the crown width of M1 molar, as well as changes in the mandibular bone characteristics. The alveolar bone volume was reduced, and the trabecular separation was increased in the 500× exposed mice. Bone volume in the condyle head was increased in all treated groups. Τhe Safranin-O-stained area of mature hypertrophic chondrocytes and the width of their zones were reduced in 0.5×, 10× and 100× exposed groups. This is the first indication that prenatal exposure to an epidemiologically defined EDC mixture, associated with neurodevelopmental impacts, can also affect mandibular growth in mammals.
Collapse
Affiliation(s)
- Vagelis Rinotas
- Institute for Fundamental Biomedical Research (IFBR), Biomedical Research Center “Al. Fleming”, 16672 Vari, Greece
| | - Antonios Stamatakis
- Biology-Biochemistry Laboratory, Faculty of Nursing, School of Health Sciences, National and Kapodistrian University of Athens (NKUA), 11527 Athens, Greece; (A.S.)
| | - Athanasios Stergiopoulos
- Biology-Biochemistry Laboratory, Faculty of Nursing, School of Health Sciences, National and Kapodistrian University of Athens (NKUA), 11527 Athens, Greece; (A.S.)
| | - Carl-Gustaf Bornehag
- Department of Health Sciences, Karlstad University, 65188 Karlstad, Sweden
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Joëlle Rüegg
- Department of Organismal Biology, Uppsala University, 75236 Uppsala, Sweden
| | - Marietta Armaka
- Institute for Fundamental Biomedical Research (IFBR), Biomedical Research Center “Al. Fleming”, 16672 Vari, Greece
| | - Efthymia Kitraki
- Laboratory of Basic Sciences, Faculty of Dentistry, School of Health Sciences, National and Kapodistrian University of Athens (NKUA), 11527 Athens, Greece
| |
Collapse
|
20
|
Calsa B, Menezes LDS, Neves JG, Gontijo JAR, Santamaria-Jr M, Boer PA. Mandible development under gestational protein restriction: cellular and molecular mechanisms. J Mol Histol 2024; 55:937-953. [PMID: 39105943 DOI: 10.1007/s10735-024-10242-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 07/31/2024] [Indexed: 08/07/2024]
Abstract
Insufficient evidence regarding how maternal undernutrition affects craniofacial bone development persists. With its unique focus on the impact of gestational protein restriction on calvaria and mandible osteogenesis, this study aims to fill, at least in part, this gap. Female mice were mated and randomized into NP (normal protein) or LP (low protein) groups. On the 18th gestational day (GD), male embryos were collected and submitted to microtomography (µCT), scanning electron microscopy (SEM), energy-dispersive X-ray spectroscopy (EDS), PCR, and autophagy dynamic analyses. The study shows that the LP offspring exhibited lower body mass than the NP group, with µCT analysis revealing no volumetric differences in fetus's head. EDS analysis showed lower calcium and higher phosphorus percentages in mandibles and calvaria. SEM assessment evidenced higher hydroxyapatite crystal-like (HC) deposition on the calvaria surface in LP fetus. Conversely, lower HC deposition was observed on the mandible surface, suggesting delayed matrix mineralization in LP fetuses with a higher percentage of collagen fibers in the mandible bone. The autophagy process was reduced in the mesenchyme of LP fetuses. PCR array analysis of 84 genes revealed 27 genes with differential expression in the LP progeny-moreover, increased mRNA levels of Akt1, Mtor, Nfkb, and Smad1 in the LP offspring. In conclusion, the results suggest that gestational protein restriction anticipated bone differentiation in utero, before 18GD, where this process is reduced compared to the control, leading to the reduction in bone area at 15 postnatal day previously observed. These findings provide insights into the molecular and cellular mechanisms of mandible development and suggest potential implications for the Developmental Origins of Health and Disease (DOHaD).
Collapse
Affiliation(s)
- Bruno Calsa
- Fetal Programming and Hydroelectrolyte Metabolism Laboratory, Department of Internal Medicine, FCM, Campinas State University (UNICAMP), Campinas, SP, Brazil
| | - Luan Dos Santos Menezes
- Department of Restorative Dentistry, Dental Materials Division, Piracicaba Dental School, UNICAMP, Piracicaba, SP, Brazil
| | - José Guilherme Neves
- Department of Restorative Dentistry, Dental Materials Division, Piracicaba Dental School, UNICAMP, Piracicaba, SP, Brazil
| | - José Antônio Rocha Gontijo
- Fetal Programming and Hydroelectrolyte Metabolism Laboratory, Department of Internal Medicine, FCM, Campinas State University (UNICAMP), Campinas, SP, Brazil
| | - Milton Santamaria-Jr
- Department of Social and Pediatric Dentistry, Institute of Science and Technology, College of Dentistry, São Paulo State University, São Jose dos Campos, Sao Paulo, Brazil
| | - Patrícia Aline Boer
- Fetal Programming and Hydroelectrolyte Metabolism Laboratory, Department of Internal Medicine, FCM, Campinas State University (UNICAMP), Campinas, SP, Brazil.
| |
Collapse
|
21
|
Kanai SM, Garcia CR, Augustus MR, Sharafeldeen SA, Brooks EP, Sucharov J, Lencer ES, Nichols JT, Clouthier DE. The Gq/11 family of Gα subunits is necessary and sufficient for lower jaw development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.17.611698. [PMID: 39345358 PMCID: PMC11430119 DOI: 10.1101/2024.09.17.611698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Vertebrate jaw development is coordinated by highly conserved ligand-receptor systems such as the peptide ligand Endothelin 1 (Edn1) and Endothelin receptor type A (Ednra), which are required for patterning of lower jaw structures. The Edn1/Ednra signaling pathway establishes the identity of lower jaw progenitor cells by regulating expression of numerous patterning genes, but the intracellular signaling mechanisms linking receptor activation to gene regulation remain poorly understood. As a first step towards elucidating this mechanism, we examined the function of the Gq/11 family of Gα subunits in zebrafish using pharmacological inhibition and genetic ablation of Gq/11 activity and transgenic induction of a constitutively active Gq protein in edn1 -/- embryos. Genetic loss of Gq/11 activity fully recapitulated the edn1 -/- phenotype, with genes encoding G11 being most essential. Furthermore, inducing Gq activity in edn1 -/- embryos not only restored Edn1/Ednra-dependent jaw structures and gene expression signatures but also caused homeosis of the upper jaw structure into a lower jaw-like structure. These results indicate that Gq/11 is necessary and sufficient to mediate the lower jaw patterning mechanism for Ednra in zebrafish.
Collapse
Affiliation(s)
- Stanley M. Kanai
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO USA
| | - Chloe R. Garcia
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO USA
| | - MaCalia R. Augustus
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO USA
| | - Shujan A. Sharafeldeen
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO USA
| | - Elliott P. Brooks
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO USA
| | - Juliana Sucharov
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO USA
| | - Ezra S. Lencer
- Department of Biology, Lafayette College, Easton, PA USA
| | - James T. Nichols
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO USA
| | - David E. Clouthier
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO USA
| |
Collapse
|
22
|
Tokavanich N, Chan B, Strauss K, Castro Andrade CD, Arai Y, Nagata M, Foretz M, Brooks DJ, Ono N, Ono W, Wein MN. Control of alveolar bone development, homeostasis, and socket healing by salt inducible kinases. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.04.611228. [PMID: 39282451 PMCID: PMC11398370 DOI: 10.1101/2024.09.04.611228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2025]
Abstract
Alveolar bone supports and anchors teeth. The parathyroid hormone-related protein (PTHrP) pathway plays a key role in alveolar bone biology. Salt inducible kinases (SIKs) are important downstream regulators of PTH/PTHrP signaling in the appendicular skeleton where SIK inhibition increases bone formation and trabecular bone mass. However, the function of these kinases in alveolar bone remains unknown. Here, we report a critical role for SIK2/SIK3 in alveolar bone development, homeostasis, and socket healing after tooth extraction. Inducible SIK2/SIK3 deletion led to dramatic alveolar bone defects without changes in tooth eruption. Ablating these kinases impairs alveolar bone formation due to disrupted osteoblast maturation, a finding associated with ectopic periostin expression by fibrous cells in regions of absent alveolar bone at steady state and following molar extraction. Distinct phenotypic consequences of SIK2/SIK3 deletion in appendicular versus craniofacial bones prompted us to identify a specific transcriptomic signature in alveolar versus long bone osteoblasts. Thus, SIK2/SIK3 deletion illuminates a key role for these kinases in alveolar bone biology and highlights the emerging concept that different osteoblast subsets utilize unique genetic programs. Summary statement SIK2/SIK3 deletion in alveolar bone reduces bone formation and mass by impairing osteoblast maturation, unlike in long bones, where it increases bone formation and mass.
Collapse
|
23
|
LaCombe JM, Sloan K, Thomas JR, Blackwell MP, Crawford I, Bishop F, Wallace JM, Roper RJ. Sex-specific trisomic Dyrk1a-related skeletal phenotypes during development in a Down syndrome model. Dis Model Mech 2024; 17:dmm050914. [PMID: 39136051 PMCID: PMC11449447 DOI: 10.1242/dmm.050914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 08/04/2024] [Indexed: 08/21/2024] Open
Abstract
Skeletal insufficiency affects all individuals with Down syndrome (DS) or trisomy 21 and may alter bone strength throughout development due to a reduced period of bone formation and early attainment of peak bone mass compared to those in typically developing individuals. Appendicular skeletal deficits also appear in males before females with DS. In femurs of male Ts65Dn DS model mice, cortical deficits were pronounced throughout development, but trabecular deficits and Dyrk1a overexpression were transitory until postnatal day (P) 30, when there were persistent trabecular and cortical deficits and Dyrk1a was trending toward overexpression. Correction of DS-related skeletal deficits by a purported DYRK1A inhibitor or through genetic means beginning at P21 was not effective at P30, but germline normalization of Dyrk1a improved male bone structure by P36. Trabecular and cortical deficits in female Ts65Dn mice were evident at P30 but subsided by P36, typifying periodic developmental skeletal normalizations that progressed to more prominent bone deficiencies. Sex-dependent differences in skeletal deficits with a delayed impact of trisomic Dyrk1a are important to find temporally specific treatment periods for bone and other phenotypes associated with trisomy 21.
Collapse
Affiliation(s)
- Jonathan M LaCombe
- Department of Biology, Indiana University Indianapolis, Indianapolis, IN 46202, USA
- Labcorp Early Development Laboratories, Inc., Greenfield, IN 46140, USA
| | - Kourtney Sloan
- Department of Biology, Indiana University Indianapolis, Indianapolis, IN 46202, USA
| | - Jared R Thomas
- Department of Biology, Indiana University Indianapolis, Indianapolis, IN 46202, USA
| | - Matthew P Blackwell
- Department of Biology, Indiana University Indianapolis, Indianapolis, IN 46202, USA
| | - Isabella Crawford
- Department of Biology, Indiana University Indianapolis, Indianapolis, IN 46202, USA
| | - Flannery Bishop
- Department of Biology, Indiana University Indianapolis, Indianapolis, IN 46202, USA
| | - Joseph M Wallace
- Department of Biomedical Engineering, Purdue University, Indianapolis, IN 46202, USA
| | - Randall J Roper
- Department of Biology, Indiana University Indianapolis, Indianapolis, IN 46202, USA
| |
Collapse
|
24
|
Yu W, Kastriti ME, Ishan M, Choudhary SK, Rashid MM, Kramer N, Do HGT, Wang Z, Xu T, Schwabe RF, Ye K, Adameyko I, Liu HX. The duct of von Ebner's glands is a source of Sox10 + taste bud progenitors and susceptible to pathogen infections. Front Cell Dev Biol 2024; 12:1460669. [PMID: 39247625 PMCID: PMC11377339 DOI: 10.3389/fcell.2024.1460669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Accepted: 08/01/2024] [Indexed: 09/10/2024] Open
Abstract
Introduction We have recently demonstrated that Sox10-expressing (Sox10 +) cells give rise to mainly type-III neuronal taste bud cells that are responsible for sour and salt taste. The two tissue compartments containing Sox10 + cells in the surrounding of taste buds include the connective tissue core of taste papillae and von Ebner's glands (vEGs) that are connected to the trench of circumvallate and foliate papillae. Methods In this study, we performed single cell RNA-sequencing of the epithelium of Sox10-Cre/tdT mouse circumvallate/vEG complex and used inducible Cre mouse models to map the cell lineages of vEGs and/or connective tissue (including stromal and Schwann cells). Results Transcriptomic analysis indicated that Sox10 expression was enriched in the cell clusters of vEG ducts that contained abundant proliferating cells, while Sox10-Cre/tdT expression was enriched in type-III taste bud cells and vEG ductal cells. In vivo lineage mapping showed that the traced cells were distributed in circumvallate taste buds concurrently with those in the vEGs, but not in the connective tissue. Moreover, multiple genes encoding pathogen receptors were enriched in the vEG ducts hosting Sox10 + cells. Discussion Our data supports that it is the vEGs, not connective tissue core, that serve as the niche of Sox10 + taste bud progenitors. If this is also true in humans, our data indicates that vEG duct is a source of Sox10 + taste bud progenitors and susceptible to pathogen infections.
Collapse
Affiliation(s)
- Wenxin Yu
- Department of Animal and Dairy Science, Regenerative Bioscience Center, College of Agricultural and Environmental Sciences, University of Georgia, Athens, GA, United States
| | | | - Mohamed Ishan
- Department of Animal and Dairy Science, Regenerative Bioscience Center, College of Agricultural and Environmental Sciences, University of Georgia, Athens, GA, United States
| | | | - Md Mamunur Rashid
- Department of Animal and Dairy Science, Regenerative Bioscience Center, College of Agricultural and Environmental Sciences, University of Georgia, Athens, GA, United States
| | - Naomi Kramer
- Department of Animal and Dairy Science, Regenerative Bioscience Center, College of Agricultural and Environmental Sciences, University of Georgia, Athens, GA, United States
| | - Hy Gia Truong Do
- Department of Genetics, University of Georgia, Athens, GA, United States
| | - Zhonghou Wang
- Department of Animal and Dairy Science, Regenerative Bioscience Center, College of Agricultural and Environmental Sciences, University of Georgia, Athens, GA, United States
| | - Ting Xu
- Department of Animal and Dairy Science, Regenerative Bioscience Center, College of Agricultural and Environmental Sciences, University of Georgia, Athens, GA, United States
| | - Robert F Schwabe
- Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY, United States
| | - Kaixiong Ye
- Institute of Bioinformatics, University of Georgia, Athens, GA, United States
- Department of Genetics, University of Georgia, Athens, GA, United States
| | - Igor Adameyko
- Department of Neuroimmunology, Medical University of Vienna, Vienna, Austria
- Department of Physiology and Pharmacology, Karolinska Institutet, Solna, Sweden
| | - Hong-Xiang Liu
- Department of Animal and Dairy Science, Regenerative Bioscience Center, College of Agricultural and Environmental Sciences, University of Georgia, Athens, GA, United States
| |
Collapse
|
25
|
Xu Y, Sun B, Wang H, Cai Y, Chu D, Cao R, Wang Z. Autophagy regulates age-related delayed jawbone regeneration and decreased osteoblast osteogenesis by degrading FABP3. FASEB J 2024; 38:e23824. [PMID: 39012304 DOI: 10.1096/fj.202400549rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 06/25/2024] [Accepted: 07/03/2024] [Indexed: 07/17/2024]
Abstract
The regenerative ability of limb bones after injury decreases during aging, but whether a similar phenomenon occurs in jawbones and whether autophagy plays a role in this process remain unclear. Through retrospective analysis of clinical data and studies on a mouse model of jawbone defects, we confirmed the presence of delayed or impaired bone regeneration in the jawbones of old individuals and mice. Subsequently, osteoblasts (OBs) derived from mouse jawbones were isolated, showing reduced osteogenesis in senescent osteoblasts (S-OBs). We observed a reduction in autophagy within both aged jawbones and S-OBs. Additionally, pharmacological inhibition of autophagy in normal OBs (N-OBs) led to cell aging and decreased osteogenesis, while autophagic activation reversed the aging phenotype of S-OBs. The activator rapamycin (RAPA) increased the autophagy level and bone regeneration in aged jawbones. Finally, we found that fatty acid-binding protein 3 (FABP3) was degraded by autolysosomes through its interaction with sequestosome 1 (P62/SQSTM1). Autophagy inhibition within senescent jawbones and S-OBs led to the excessive accumulation of FABP3, and FABP3 knockdown partially rescued the decreased osteogenesis in S-OBs and alleviated age-related compromised jawbone regeneration. In summary, we confirmed that autophagy inhibition plays an important role in delaying bone regeneration in aging jawbones. Autophagic activation or FABP3 knockdown can partially rescue the osteogenesis of S-OBs and the regeneration of aging jawbones, providing insight into jawbone aging.
Collapse
Affiliation(s)
- Yifan Xu
- Department of Implantology & Department of Oral and Maxillofacial Surgery, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| | - Bin Sun
- Department of Implantology & Department of Oral and Maxillofacial Surgery, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| | - Haicheng Wang
- Department of Implantology & Department of Oral and Maxillofacial Surgery, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| | - Yuyi Cai
- Department of Stomatology, Daping Hospital, Army Medical University (The Third Military Medical University), Chongqing, China
| | - Danna Chu
- Department of Implantology & Department of Oral and Maxillofacial Surgery, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| | - Rongkai Cao
- Department of Implantology & Department of Oral and Maxillofacial Surgery, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| | - Zuolin Wang
- Department of Implantology & Department of Oral and Maxillofacial Surgery, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| |
Collapse
|
26
|
Pi HJ, Huang B, Yuan Q, Jing JJ. Neural regulation of mesenchymal stem cells in craniofacial bone: development, homeostasis and repair. Front Physiol 2024; 15:1423539. [PMID: 39135707 PMCID: PMC11318092 DOI: 10.3389/fphys.2024.1423539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 07/15/2024] [Indexed: 08/15/2024] Open
Abstract
Mesenchymal stem cells endow various functions, including proliferation, multipotency, migration, etc. Craniofacial bones originate from the cranial neural crest and are developed mainly through intramembranous ossification, which are different from long bones. There are varied mesenchymal stem cells existing in the craniofacial bone, including Gli1 + cells, Axin2 + cells, Prx1 + cells, etc. Nerves distributed in craniofacial area are also derived from the neural crest, and the trigeminal nerve is the major sensory nerve in craniofacial area. The nerves and the skeleton are tightly linked spatially, and the skeleton is broadly innervated by sensory and sympathetic nerves, which also participate in bone development, homeostasis and healing process. In this review, we summarize mesenchymal stem cells located in craniofacial bone or, to be more specific, in jaws, temporomandibular joint and cranial sutures. Then we discuss the research advance concerning neural regulation of mesenchymal stem cells in craniofacial bone, mainly focused on development, homeostasis and repair. Discovery of neural regulation of mesenchymal stem cells may assist in treatment in the craniofacial bone diseases or injuries.
Collapse
Affiliation(s)
| | | | - Quan Yuan
- *Correspondence: Quan Yuan, ; Jun-Jun Jing,
| | | |
Collapse
|
27
|
Li J, Sun J, Xu M, Yang L, Yang N, Deng J, Ma Y, Qi Y, Liu Z, Ruan Q, Liu Y, Huang Y. Human cytomegalovirus infection impairs neural differentiation via repressing sterol regulatory element binding protein 2-mediated cholesterol biosynthesis. Cell Mol Life Sci 2024; 81:289. [PMID: 38970696 PMCID: PMC11335213 DOI: 10.1007/s00018-024-05278-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 05/11/2024] [Accepted: 05/13/2024] [Indexed: 07/08/2024]
Abstract
Congenital human cytomegalovirus (HCMV) infection is a major cause of abnormalities and disorders in the central nervous system (CNS) and/or the peripheral nervous system (PNS). However, the complete pathogenesis of neural differentiation disorders caused by HCMV infection remains to be fully elucidated. Stem cells from human exfoliated deciduous teeth (SHEDs) are mesenchymal stem cells (MSCs) with a high proliferation and neurogenic differentiation capacity. Since SHEDs originate from the neural crest of the early embryonic ectoderm, SHEDs were hypothesized to serve as a promising cell line for investigating the pathogenesis of neural differentiation disorders in the PNS caused by congenital HCMV infection. In this work, SHEDs were demonstrated to be fully permissive to HCMV infection and the virus was able to complete its life cycle in SHEDs. Under neurogenic inductive conditions, HCMV infection of SHEDs caused an abnormal neural morphology. The expression of stem/neural cell markers was also disturbed by HCMV infection. The impairment of neural differentiation was mainly due to a reduction of intracellular cholesterol levels caused by HCMV infection. Sterol regulatory element binding protein-2 (SREBP2) is a critical transcription regulator that guides cholesterol synthesis. HCMV infection was shown to hinder the migration of SREBP2 into nucleus and resulted in perinuclear aggregations of SREBP2 during neural differentiation. Our findings provide new insights into the prevention and treatment of nervous system diseases caused by congenital HCMV infection.
Collapse
Affiliation(s)
- Jianming Li
- Virology Laboratory, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Jingxuan Sun
- Virology Laboratory, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Mingyi Xu
- Virology Laboratory, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Lei Yang
- Department of Pediatric Dentistry, School and Hospital of Stomatology, China Medical University, Shenyang, Liaoning, China
- Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, Liaoning, China
| | - Ning Yang
- Department of Pediatric Dentistry, School and Hospital of Stomatology, China Medical University, Shenyang, Liaoning, China
- Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, Liaoning, China
| | - Jingui Deng
- Virology Laboratory, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
- Department of Microorganism Laboratory, Shenyang Center for Disease Control and Prevention, Shenyang, Liaoning, China
| | - Yanping Ma
- Virology Laboratory, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
- Departments of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Ying Qi
- Virology Laboratory, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
- Departments of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Zhongyang Liu
- Virology Laboratory, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
- Departments of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Qiang Ruan
- Virology Laboratory, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.
- Departments of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.
| | - Yao Liu
- Department of Pediatric Dentistry, School and Hospital of Stomatology, China Medical University, Shenyang, Liaoning, China.
- Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, Liaoning, China.
| | - Yujing Huang
- Virology Laboratory, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.
- Departments of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.
| |
Collapse
|
28
|
Rojas-Torres J, Quijón MEG, Henríquez-Vidal A, Devia-Rubio L, Martínez-Duran L. Permanent and decidua dentition as chronological biomarkers of heavy metal contamination: A review of the forensic literature. J Trace Elem Med Biol 2024; 84:127435. [PMID: 38547726 DOI: 10.1016/j.jtemb.2024.127435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 02/19/2024] [Accepted: 03/14/2024] [Indexed: 05/27/2024]
Abstract
STATEMENT OF PROBLEM Contamination with heavy metals (HM) has great environmental consequences in the environment due to lack of biodegradation, in addition, accumulation in living beings causes defects in tissues and organs, deteriorating their function and inducing a wide spectrum of diseases. Human biomonitoring consists of the periodic measurement of a certain chemical substance or metabolite in a particular population, using matrices that can be acute or chronic. Teeth are chronic matrices that have great characteristics of resistance and chronological storage of information. This review aims to identify the mechanisms, spatial location, and affinity of HM within teeth, along with understanding its applicability as a chronological record matrix in the face of HM contamination. MATERIAL AND METHODS A systematic search review was performed using the PubMed/Medline, Web of Science, and Scopus metasearch engines, and the terms "teeth" OR "dental" OR "tooth" AND "heavy metals" were intersected. Complete articles are included in Spanish, English and Portuguese without time restrictions, involving studies in humans or in vitro; Letters to the editor, editorials and those that did not refer to information on the incorporation and relationship of HM with the teeth were excluded. RESULTS 837 published articles were detected, 91 were adjusted to the search objective, and 6 were manually included. Teeth are structures with a great capacity for information retention in the face of HM contamination due to low physiological turnover and their long processes of marked formations by developmental biorhythm milestones such as the neonatal line (temporal reference indicator). The contamination mechanisms inside the tooth are linked to the affinity of hydroxyapatite for HM; this incorporation can be in the soft matrix during the apposition phase or as part of the chemical exchanges between hydroxyapatite and the elements of the environment. CONCLUSION The teeth present unique characteristics of great resistance and affinity for HM, as well as a chronological biomarker for human biomonitoring, so they can be used as means of expertise or evidence to confirm or rule out a fact of environmental characteristics in the legal field.
Collapse
Affiliation(s)
- Javier Rojas-Torres
- Forensic Dentistry Lab, Centro de Investigación en Odontología Legal y Forense -CIO-, Facultad de Odontología, Universidad de La Frontera, Temuco 4811230, Chile; Programa de Doctorado en Ciencias mención Biología Celular y Molecular Aplicada, Universidad de La Frontera, Temuco 4811230, Chile.
| | - María Eugenia González Quijón
- Chemical Engineering Department, Universidad de La Frontera, Francisco Salazar 01145, Temuco 4780000, Chile; Center of Waste Management and Bioenergy-BIOREN, University of La Frontera, Francisco Salazar 01145, Temuco 4780000, Chile
| | - Andrés Henríquez-Vidal
- Forensic Dentistry Lab, Centro de Investigación en Odontología Legal y Forense -CIO-, Facultad de Odontología, Universidad de La Frontera, Temuco 4811230, Chile
| | - Leslie Devia-Rubio
- Forensic Dentistry Lab, Centro de Investigación en Odontología Legal y Forense -CIO-, Facultad de Odontología, Universidad de La Frontera, Temuco 4811230, Chile
| | - Luis Martínez-Duran
- Programa de Doctorado en Ciencias mención Biología Celular y Molecular Aplicada, Universidad de La Frontera, Temuco 4811230, Chile; Laboratorio de Farmacología Molecular y Química medicinal, departamento de Ciencias Químicas y Recursos Naturales, Facultad de Ingeniería y Ciencias, Universidad de La Frontera, Temuco 4811230, Chile; Receptomics and Brain Disorders Lab, Department of Human Physiology, Sport and Exercise, Faculty of Medicine, University of Malaga, Edificio Lopez-Penalver, Jimenez Fraud 10, Málaga 29071, Spain
| |
Collapse
|
29
|
Knyazeva A, Dyachuk V. Neural crest and sons: role of neural crest cells and Schwann cell precursors in development and gland embryogenesis. Front Cell Dev Biol 2024; 12:1406199. [PMID: 38989061 PMCID: PMC11233730 DOI: 10.3389/fcell.2024.1406199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Accepted: 06/10/2024] [Indexed: 07/12/2024] Open
Abstract
In this review, we consider the multipotency of neural crest cells (NCCs), Schwann cell precursors (SCPs), and their role in embryogenesis base on genetic tracing and knock out model animals and single cell transcriptomic analysis. In particular, we summarize and analyze data on the contribution of NCCs and SCPs to the gland development and functions.
Collapse
|
30
|
Zhao R, Moore EL, Gogol MM, Unruh JR, Yu Z, Scott AR, Wang Y, Rajendran NK, Trainor PA. Identification and characterization of intermediate states in mammalian neural crest cell epithelial to mesenchymal transition and delamination. eLife 2024; 13:RP92844. [PMID: 38873887 PMCID: PMC11178358 DOI: 10.7554/elife.92844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2024] Open
Abstract
Epithelial to mesenchymal transition (EMT) is a cellular process that converts epithelial cells to mesenchymal cells with migratory potential in developmental and pathological processes. Although originally considered a binary event, EMT in cancer progression involves intermediate states between a fully epithelial and a fully mesenchymal phenotype, which are characterized by distinct combinations of epithelial and mesenchymal markers. This phenomenon has been termed epithelial to mesenchymal plasticity (EMP), however, the intermediate states remain poorly described and it's unclear whether they exist during developmental EMT. Neural crest cells (NCC) are an embryonic progenitor cell population that gives rise to numerous cell types and tissues in vertebrates, and their formation and delamination is a classic example of developmental EMT. However, whether intermediate states also exist during NCC EMT and delamination remains unknown. Through single-cell RNA sequencing of mouse embryos, we identified intermediate NCC states based on their transcriptional signature and then spatially defined their locations in situ in the dorsolateral neuroepithelium. Our results illustrate the importance of cell cycle regulation and functional role for the intermediate stage marker Dlc1 in facilitating mammalian cranial NCC delamination and may provide new insights into mechanisms regulating pathological EMP.
Collapse
Affiliation(s)
- Ruonan Zhao
- Stowers Institute for Medical ResearchKansas CityUnited States
- Department of Anatomy and Cell Biology, University of Kansas Medical CenterKansas CityUnited States
| | - Emma L Moore
- Stowers Institute for Medical ResearchKansas CityUnited States
| | | | - Jay R Unruh
- Stowers Institute for Medical ResearchKansas CityUnited States
| | - Zulin Yu
- Stowers Institute for Medical ResearchKansas CityUnited States
| | - Allison R Scott
- Stowers Institute for Medical ResearchKansas CityUnited States
| | - Yan Wang
- Stowers Institute for Medical ResearchKansas CityUnited States
| | | | - Paul A Trainor
- Stowers Institute for Medical ResearchKansas CityUnited States
- Department of Anatomy and Cell Biology, University of Kansas Medical CenterKansas CityUnited States
| |
Collapse
|
31
|
Parslow VR, Elmore SA, Cochran RZ, Bolon B, Mahler B, Sabio D, Lubeck BA. Histology Atlas of the Developing Mouse Respiratory System From Prenatal Day 9.0 Through Postnatal Day 30. Toxicol Pathol 2024; 52:153-227. [PMID: 39096105 DOI: 10.1177/01926233241252114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Respiratory diseases are one of the leading causes of death and disability around the world. Mice are commonly used as models of human respiratory disease. Phenotypic analysis of mice with spontaneous, congenital, inherited, or treatment-related respiratory tract abnormalities requires investigators to discriminate normal anatomic features of the respiratory system from those that have been altered by disease. Many publications describe individual aspects of normal respiratory tract development, primarily focusing on morphogenesis of the trachea and lung. However, a single reference providing detailed low- and high-magnification, high-resolution images of routine hematoxylin and eosin (H&E)-stained sections depicting all major structures of the entire developing murine respiratory system does not exist. The purpose of this atlas is to correct this deficiency by establishing one concise reference of high-resolution color photomicrographs from whole-slide scans of H&E-stained tissue sections. The atlas has detailed descriptions and well-annotated images of the developing mouse upper and lower respiratory tracts emphasizing embryonic days (E) 9.0 to 18.5 and major early postnatal events. The selected images illustrate the main structures and events at key developmental stages and thus should help investigators both confirm the chronological age of mouse embryos and distinguish normal morphology as well as structural (cellular and organ) abnormalities.
Collapse
Affiliation(s)
| | - Susan A Elmore
- Experimental Pathology Laboratories, Inc., Research Triangle Park, North Carolina, USA
| | - Robert Z Cochran
- National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | | | - Beth Mahler
- Experimental Pathology Laboratories, Inc., Research Triangle Park, North Carolina, USA
| | - David Sabio
- Experimental Pathology Laboratories, Inc., Research Triangle Park, North Carolina, USA
| | - Beth A Lubeck
- National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| |
Collapse
|
32
|
Zhao Y, Chen S, Liu X, Chen X, Yang D, Zhang J, Wu D, Zhang Y, Xie S, Li X, Wang Z, Feng B, Qin D, Pei D, Wang Y, Cai J. Single-cell RNA-seq of in vitro expanded cells from cranial neural crest reveals a rare odontogenic sub-population. Cell Prolif 2024; 57:e13598. [PMID: 38196265 DOI: 10.1111/cpr.13598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 11/21/2023] [Accepted: 12/27/2023] [Indexed: 01/11/2024] Open
Abstract
Ecto-mesenchymal cells of mammalian tooth germ develops from cranial neural crest cells. These cells are recognised as a promising source for tooth development and regeneration. Despite the high heterogeneity of the neural crest, the cellular landscape of in vitro cultured cranial neural crest cells (CNCCs) for odontogenesis remains unclear. In this study, we used large-scale single-cell RNA sequencing to analyse the cellular landscape of in vitro cultured mouse CNCCs for odontogenesis. We revealed distinct cell trajectories from primary cells to passage 5 and identified a rare Alx3+/Barx1+ sub-population in primary CNCCs that differentiated into two odontogenic clusters characterised by the up-regulation of Pax9/Bmp3 and Lhx6/Dmp1. We successfully induced whole tooth-like structures containing enamel, dentin, and pulp under the mouse renal capsule using in vitro cultured cells from both cranial and trunk neural crests with induction rates of 26.7% and 22.1%, respectively. Importantly, we confirmed only cells sorted from odontogenic path can induce tooth-like structures. Cell cycle and DNA replication genes were concomitantly upregulated in the cultured NCCs of the tooth induction groups. Our data provide valuable insights into the cell heterogeneity of in vitro cultured CNCCs and their potential as a source for tooth regeneration.
Collapse
Affiliation(s)
- Yifan Zhao
- Innovation Centre for Advanced Interdisciplinary Medicine, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong SAR, China
- Bioland Laboratory, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Shubin Chen
- Innovation Centre for Advanced Interdisciplinary Medicine, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Bioland Laboratory, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Xiaobo Liu
- Laboratory of Cancer Precision Medicine, The First Hospital of Jilin University, Changchun, China
| | - Xiaoming Chen
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Guangdong Provincial People's Hospital Ganzhou Hospital, Ganzhou Municipal Hospital, Ganzhou, China
| | - Dandan Yang
- Experimental Center of Pathogenobiology Immunology, Cytobiology and Genetics, Basic Medical College, Jilin University, Changchun, China
| | - Jiashu Zhang
- Innovation Centre for Translational Medicine, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Di Wu
- Bioland Laboratory, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Yanmei Zhang
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Si Xie
- Innovation Centre for Advanced Interdisciplinary Medicine, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong SAR, China
| | - Xiaomei Li
- Innovation Centre for Advanced Interdisciplinary Medicine, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhiyuan Wang
- Innovation Centre for Translational Medicine, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Bo Feng
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Dajiang Qin
- Innovation Centre for Advanced Interdisciplinary Medicine, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong SAR, China
- Bioland Laboratory, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| | - Duanqing Pei
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong SAR, China
- Laboratory of Cell Fate Control, School of Life Sciences, Westlake University, Hangzhou, China
| | - Yaofeng Wang
- Innovation Centre for Advanced Interdisciplinary Medicine, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong SAR, China
| | - Jinglei Cai
- Innovation Centre for Advanced Interdisciplinary Medicine, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong SAR, China
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Guangzhou Key Laboratory of Enhanced Recovery after Abdominal Surgery, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
33
|
Taguchi M, Yoshimoto S, Suyama K, Sumi S, Ohki S, Ogata K, Fujimoto R, Murata D, Nakayama K, Oka K. Creating 3D constructs with cranial neural crest-derived cell lines using a bio-3D printer. J Oral Biosci 2024; 66:339-348. [PMID: 38750954 DOI: 10.1016/j.job.2024.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 05/10/2024] [Accepted: 05/13/2024] [Indexed: 06/15/2024]
Abstract
OBJECTIVES The development of bio-three-dimensional (bio-3D) printers has led to significant advances in regenerative medicine. Three-dimensional constructs, including spheroids, are maintained by extracellular matrix proteins secreted by cells so that the cells can be cultured in conditions closer to the physiological environment. This study aimed to create a useful 3D construct as a model of the dentin-pulp complex. METHODS We examined the expression patterns of extracellular matrix proteins and cell proliferation areas in a 3D construct created using O9-1 cells derived from cranial neural crest cells of mice. The 3D construct was created by sticking the spheroid cultures onto a needle array using a bio-3D printer. RESULTS Cell proliferation areas along with characteristic expression of tenascin C and DMP1 were evaluated. The expression of tenascin C and DMP1 was significantly enhanced in the spheroids compared to that in two-dimensional cultures. Moreover, cell proliferation regions and tenascin C expression were confirmed in the outer layer of spheroids in the embryonic stem cell medium, with insignificant DMP1 expression being observed. Interestingly, in a 3D construct cultured in calcification-induction medium, DMP1 expression was promoted, and DMP1-positive cells existed in the outermost layer without overlapping with tenascin C expression. CONCLUSIONS The extracellular matrix proteins, tenascin C and DMP1, were expressed in a polarized manner in spheroids and 3D constructs, similar to the findings in the dental papilla. Therefore, these 3D constructs show potential as artificial models for studying odontogenesis.
Collapse
Affiliation(s)
- Masahide Taguchi
- Section of Pediatric Dentistry, Department of Oral Growth and Development, Fukuoka Dental College, Fukuoka, Japan.
| | - Shohei Yoshimoto
- Section of Pathology, Department of Morphological Biology, Fukuoka Dental College, Fukuoka, Japan; Oral Medicine Research Center, Fukuoka Dental College, Fukuoka, Japan.
| | - Kanako Suyama
- Section of Pediatric Dentistry, Department of Oral Growth and Development, Fukuoka Dental College, Fukuoka, Japan.
| | - Satoko Sumi
- Section of Pediatric Dentistry, Department of Oral Growth and Development, Fukuoka Dental College, Fukuoka, Japan.
| | - Shirabe Ohki
- Section of Pediatric Dentistry, Department of Oral Growth and Development, Fukuoka Dental College, Fukuoka, Japan.
| | - Kayoko Ogata
- Department of Legal Medicine, Graduate School of Medical Sciences, Kanazawa University, Japan.
| | - Ryota Fujimoto
- Center for Regenerative Medicine Research, Faculty of Medicine, Saga University, Saga, Japan.
| | - Daiki Murata
- Center for Regenerative Medicine Research, Faculty of Medicine, Saga University, Saga, Japan.
| | - Koichi Nakayama
- Center for Regenerative Medicine Research, Faculty of Medicine, Saga University, Saga, Japan.
| | - Kyoko Oka
- Section of Pediatric Dentistry, Department of Oral Growth and Development, Fukuoka Dental College, Fukuoka, Japan; Oral Medicine Research Center, Fukuoka Dental College, Fukuoka, Japan.
| |
Collapse
|
34
|
Wang F, Wang H, Zhang H, Sun B, Wang Z. A Novel Mechanism of MSCs Responding to Occlusal Force for Bone Homeostasis. J Dent Res 2024; 103:642-651. [PMID: 38665065 DOI: 10.1177/00220345241236120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/24/2024] Open
Abstract
Alveolar bone, as tooth-supporting bone for mastication, is sensitive to occlusal force. However, the mechanism of alveolar bone loss after losing occlusal force remains unclear. Here, we performed single-cell RNA sequencing of nonhematopoietic (CD45-) cells in mouse alveolar bone after removing the occlusal force. Mesenchymal stromal cells (MSCs) and endothelial cell (EC) subsets were significantly decreased in frequency, as confirmed by immunofluorescence and flow cytometry. The osteogenic and proangiogenic abilities of MSCs were impaired, and the expression of mechanotransducers yes associated protein 1 (Yap) and WW domain containing transcription regulator 1 (Taz) in MSCs decreased. Conditional deletion of Yap and Taz from LepR+ cells, which are enriched in MSCs that are important for adult bone homeostasis, significantly decreased alveolar bone mass and resisted any further changes in bone mass induced by occlusal force changes. Interestingly, LepR-Cre; Yapf/f; Tazf/f mice showed a decrease in CD31hi endomucin (Emcn)hi endothelium, and the expression of some EC-derived signals acting on osteoblastic cells was inhibited in alveolar bone. Mechanistically, conditional deletion of Yap and Taz in LepR+ cells inhibited the secretion of pleiotrophin (Ptn), which impaired the proangiogenic capacity of LepR+ cells. Knockdown in MSC-derived Ptn repressed human umbilical vein EC tube formation in vitro. More important, administration of recombinant PTN locally recovered the frequency of CD31hiEmcnhi endothelium and rescued the low bone mass phenotype of LepR-Cre; Yapf/f; Tazf/f mice. Taken together, these findings suggest that occlusal force governs MSC-regulated endothelium to maintain alveolar bone homeostasis through the Yap/Taz/Ptn axis, providing a reference for further understanding of the relationship between dysfunction and bone homeostasis.
Collapse
Affiliation(s)
- F Wang
- Department of Oral Implantology & Department of Oral and Maxillofacial Surgery, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| | - H Wang
- Department of Oral Implantology & Department of Oral and Maxillofacial Surgery, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| | - H Zhang
- Department of Oral Implantology & Department of Oral and Maxillofacial Surgery, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| | - B Sun
- Department of Oral Implantology & Department of Oral and Maxillofacial Surgery, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| | - Z Wang
- Department of Oral Implantology & Department of Oral and Maxillofacial Surgery, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| |
Collapse
|
35
|
Zhang M, Guo T, Pei F, Feng J, Jing J, Xu J, Yamada T, Ho TV, Du J, Sehgal P, Chai Y. ARID1B maintains mesenchymal stem cell quiescence via inhibition of BCL11B-mediated non-canonical Activin signaling. Nat Commun 2024; 15:4614. [PMID: 38816354 PMCID: PMC11139927 DOI: 10.1038/s41467-024-48285-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 04/24/2024] [Indexed: 06/01/2024] Open
Abstract
ARID1B haploinsufficiency in humans causes Coffin-Siris syndrome, associated with developmental delay, facial dysmorphism, and intellectual disability. The role of ARID1B has been widely studied in neuronal development, but whether it also regulates stem cells remains unknown. Here, we employ scRNA-seq and scATAC-seq to dissect the regulatory functions and mechanisms of ARID1B within mesenchymal stem cells (MSCs) using the mouse incisor model. We reveal that loss of Arid1b in the GLI1+ MSC lineage disturbs MSCs' quiescence and leads to their proliferation due to the ectopic activation of non-canonical Activin signaling via p-ERK. Furthermore, loss of Arid1b upregulates Bcl11b, which encodes a BAF complex subunit that modulates non-canonical Activin signaling by directly regulating the expression of activin A subunit, Inhba. Reduction of Bcl11b or non-canonical Activin signaling restores the MSC population in Arid1b mutant mice. Notably, we have identified that ARID1B suppresses Bcl11b expression via specific binding to its third intron, unveiling the direct inter-regulatory interactions among BAF subunits in MSCs. Our results demonstrate the vital role of ARID1B as an epigenetic modifier in maintaining MSC homeostasis and reveal its intricate mechanistic regulatory network in vivo, providing novel insights into the linkage between chromatin remodeling and stem cell fate determination.
Collapse
Affiliation(s)
- Mingyi Zhang
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA, 90033, USA
| | - Tingwei Guo
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA, 90033, USA
| | - Fei Pei
- 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
| | - Junjun Jing
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA, 90033, USA
| | - Jian Xu
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA, 90033, USA
| | - Takahiko Yamada
- 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
| | - Jiahui Du
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA, 90033, USA
| | - Prerna Sehgal
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA, 90033, USA
| | - Yang Chai
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA, 90033, USA.
| |
Collapse
|
36
|
LaCombe JM, Sloan K, Thomas JR, Blackwell MP, Crawford I, Wallace JM, Roper RJ. Sex specific emergence of trisomic Dyrk1a-related skeletal phenotypes in the development of a Down syndrome mouse model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.24.595804. [PMID: 38826419 PMCID: PMC11142220 DOI: 10.1101/2024.05.24.595804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Skeletal insufficiency affects all individuals with Down syndrome (DS) or Trisomy 21 (Ts21) and may alter bone strength throughout development due to a reduced period of bone formation and early attainment of peak bone mass compared to typically developing individuals. Appendicular skeletal deficits also appear in males before females with DS. In femurs of male Ts65Dn DS model mice, cortical deficits were pronounced throughout development, but trabecular deficits and Dyrk1a overexpression were transitory until postnatal day (P) 30 when there were persistent trabecular and cortical deficits and Dyrk1a was trending overexpression. Correction of DS-related skeletal deficits by a purported DYRK1A inhibitor or through genetic means beginning at P21 was not effective at P30, but germline normalization of Dyrk1a improved male bone structure by P36. Trabecular and cortical deficits in female Ts65Dn mice were evident at P30 but subsided by P36, typifying periodic developmental skeletal normalizations that progressed to more prominent bone deficiencies. Sex-dependent differences in skeletal deficits with a delayed impact of trisomic Dyrk1a are important to find temporally specific treatment periods for bone and other phenotypes associated with Ts21.
Collapse
Affiliation(s)
- Jonathan M. LaCombe
- Department of Biology, Indiana University-Indianapolis, IN, USA
- Labcorp Early Development Laboratories, Inc., Greenfield, IN, USA
| | - Kourtney Sloan
- Department of Biology, Indiana University-Indianapolis, IN, USA
| | - Jared R. Thomas
- Department of Biology, Indiana University-Indianapolis, IN, USA
| | | | | | - Joseph M. Wallace
- Department of Biomedical Engineering, Purdue University, Indianapolis, IN, USA
| | | |
Collapse
|
37
|
Yu W, Kastriti ME, Ishan M, Choudhary SK, Kramer N, Rashid MM, Truong Do HG, Wang Z, Xu T, Schwabe RF, Ye K, Adameyko I, Liu HX. The main duct of von Ebner's glands is a source of Sox10 + taste bud progenitors and susceptible to pathogen infections. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.14.594215. [PMID: 38798668 PMCID: PMC11118543 DOI: 10.1101/2024.05.14.594215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
We have recently demonstrated that Sox10 -expressing ( Sox10 + ) cells give rise to mainly type-III neuronal taste bud cells that are responsible for sour and salt taste. The two tissue compartments containing Sox10 + cells in the surrounding of taste buds include the connective tissue core of taste papillae and von Ebner's glands (vEGs) that are connected to the trench of circumvallate and foliate papillae. In this study, we used inducible Cre mouse models to map the cell lineages of connective tissue (including stromal and Schwann cells) and vEGs and performed single cell RNA-sequencing of the epithelium of Sox10-Cre/tdT mouse circumvallate/vEG complex. In vivo lineage mapping showed that the distribution of traced cells in circumvallate taste buds was closely linked with that in the vEGs, but not in the connective tissue. Sox10 , but not the known stem cells marker Lgr5 , expression was enriched in the cell clusters of main ducts of vEGs that contained abundant proliferating cells, while Sox10-Cre/tdT expression was enriched in type-III taste bud cells and excretory ductal cells. Moreover, multiple genes encoding pathogen receptors are enriched in the vEG main ducts. Our data indicate that the main duct of vEGs is a source of Sox10 + taste bud progenitors and susceptible to pathogen infections.
Collapse
|
38
|
Yoon JY, Vu HT, Lee JH, Shin JS, Kim HW, Lee HH, Kim JB, Lee JH. Evaluation of Human Platelet Lysate as an Alternative to Fetal Bovine Serum for Potential Clinical Applications of Stem Cells from Human Exfoliated Deciduous Teeth. Cells 2024; 13:847. [PMID: 38786069 PMCID: PMC11120611 DOI: 10.3390/cells13100847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 05/10/2024] [Accepted: 05/13/2024] [Indexed: 05/25/2024] Open
Abstract
In recent years, there has been a surge in demand for and research focus on cell therapy, driven by the tissue-regenerative and disease-treating potentials of stem cells. Among the candidates, dental pulp stem cells (DPSCs) or human exfoliated deciduous teeth (SHED) have garnered significant attention due to their easy accessibility (non-invasive), multi-lineage differentiation capability (especially neurogenesis), and low immunogenicity. Utilizing these stem cells for clinical purposes requires careful culture techniques such as excluding animal-derived supplements. Human platelet lysate (hPL) has emerged as a safer alternative to fetal bovine serum (FBS) for cell culture. In our study, we assessed the impact of hPL as a growth factor supplement for culture medium, also conducting a characterization of SHED cultured in hPL-supplemented medium (hPL-SHED). The results showed that hPL has effects in enhancing cell proliferation and migration and increasing cell survivability in oxidative stress conditions induced by H2O2. The morphology of hPL-SHED exhibited reduced size and elongation, with a differentiation capacity comparable to or even exceeding that of SHED cultured in a medium supplemented with fetal bovine serum (FBS-SHED). Moreover, no evidence of chromosome abnormalities or tumor formation was detected. In conclusion, hPL-SHED emerges as a promising candidate for cell therapy, exhibiting considerable potential for clinical investigation.
Collapse
Affiliation(s)
- Ji-Young Yoon
- Institute for Stem Cell & Matters, Cell & Matter Corporation, Cheonan 31116, Republic of Korea; (J.-Y.Y.); (J.H.L.); (H.-W.K.)
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, 119 Dandae-ro, Cheonan 31116, Republic of Korea; (H.T.V.); (H.-H.L.)
| | - Huong Thu Vu
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, 119 Dandae-ro, Cheonan 31116, Republic of Korea; (H.T.V.); (H.-H.L.)
- Department of Pediatric Dentistry, Faculty of Odonto-Stomatology, University of Medincine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City 17000, Vietnam
- Department of Pediatric Dentistry, College of Dentistry, Dankook University, 119 Dandae-ro, Cheonan 31116, Republic of Korea;
| | - Jun Hee Lee
- Institute for Stem Cell & Matters, Cell & Matter Corporation, Cheonan 31116, Republic of Korea; (J.-Y.Y.); (J.H.L.); (H.-W.K.)
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, 119 Dandae-ro, Cheonan 31116, Republic of Korea; (H.T.V.); (H.-H.L.)
- Department of Biomaterials Science, College of Dentistry, Dankook University, 119 Dandae-ro, Cheonan 31116, Republic of Korea
- Department of Nanobiomedical Science & BK21 FOUR NBM Global Research Center for Regenerative Medicine, Dankook University, 119 Dandae-ro, Cheonan 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, 119 Dandae-ro, Cheonan 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, 119 Dandae-ro, Cheonan 31116, Republic of Korea
- Cell & Matter Institute, Dankook University, 119 Dandae-ro, Cheonan 31116, Republic of Korea
| | - Ji-Sun Shin
- Department of Pediatric Dentistry, College of Dentistry, Dankook University, 119 Dandae-ro, Cheonan 31116, Republic of Korea;
| | - Hae-Won Kim
- Institute for Stem Cell & Matters, Cell & Matter Corporation, Cheonan 31116, Republic of Korea; (J.-Y.Y.); (J.H.L.); (H.-W.K.)
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, 119 Dandae-ro, Cheonan 31116, Republic of Korea; (H.T.V.); (H.-H.L.)
- Department of Biomaterials Science, College of Dentistry, Dankook University, 119 Dandae-ro, Cheonan 31116, Republic of Korea
- Department of Nanobiomedical Science & BK21 FOUR NBM Global Research Center for Regenerative Medicine, Dankook University, 119 Dandae-ro, Cheonan 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, 119 Dandae-ro, Cheonan 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, 119 Dandae-ro, Cheonan 31116, Republic of Korea
- Cell & Matter Institute, Dankook University, 119 Dandae-ro, Cheonan 31116, Republic of Korea
| | - Hae-Hyoung Lee
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, 119 Dandae-ro, Cheonan 31116, Republic of Korea; (H.T.V.); (H.-H.L.)
- Department of Biomaterials Science, College of Dentistry, Dankook University, 119 Dandae-ro, Cheonan 31116, Republic of Korea
- Department of Nanobiomedical Science & BK21 FOUR NBM Global Research Center for Regenerative Medicine, Dankook University, 119 Dandae-ro, Cheonan 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, 119 Dandae-ro, Cheonan 31116, Republic of Korea
| | - Jong-Bin Kim
- Department of Pediatric Dentistry, College of Dentistry, Dankook University, 119 Dandae-ro, Cheonan 31116, Republic of Korea;
| | - Jung-Hwan Lee
- Institute for Stem Cell & Matters, Cell & Matter Corporation, Cheonan 31116, Republic of Korea; (J.-Y.Y.); (J.H.L.); (H.-W.K.)
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, 119 Dandae-ro, Cheonan 31116, Republic of Korea; (H.T.V.); (H.-H.L.)
- Department of Biomaterials Science, College of Dentistry, Dankook University, 119 Dandae-ro, Cheonan 31116, Republic of Korea
- Department of Nanobiomedical Science & BK21 FOUR NBM Global Research Center for Regenerative Medicine, Dankook University, 119 Dandae-ro, Cheonan 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, 119 Dandae-ro, Cheonan 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, 119 Dandae-ro, Cheonan 31116, Republic of Korea
- Cell & Matter Institute, Dankook University, 119 Dandae-ro, Cheonan 31116, Republic of Korea
| |
Collapse
|
39
|
Stanton E, Sheridan S, Urata M, Chai Y. From Bedside to Bench and Back: Advancing Our Understanding of the Pathophysiology of Cleft Palate and Implications for the Future. Cleft Palate Craniofac J 2024; 61:759-773. [PMID: 36457208 DOI: 10.1177/10556656221142098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2024] Open
Abstract
OBJECTIVE To provide a comprehensive understanding of the pathophysiology of cleft palate (CP) and future perspectives. DESIGN Literature review. SETTING Setting varied across studies by level of care and geographical locations. INTERVENTIONS No interventions were performed. MAIN OUTCOME MEASURE(S) Primary outcome measures were to summarize our current understanding of palatogenesis in humans and animal models, the pathophysiology of CP, and potential future treatment modalities. RESULTS Animal research has provided considerable insight into the pathophysiology, molecular and cellular mechanisms of CP that have allowed for the development of novel treatment strategies. However, much work has yet to be done to connect our mouse model investigations and discoveries to CP in humans. The success of innovative strategies for tissue regeneration in mice provides promise for an exciting new avenue for improved and more targeted management of cleft care with precision medicine in patients. However, significant barriers to clinical translation remain. Among the most notable challenges include the differences in some aspects of palatogenesis and tissue repair between mice and humans, suggesting that potential therapies that have worked in animal models may not provide similar benefits to humans. CONCLUSIONS Increased translation of pathophysiological and tissue regeneration studies to clinical trials will bridge a wide gap in knowledge between animal models and human disease. By enhancing interaction between basic scientists and clinicians, and employing our animal model findings of disease mechanisms in concert with what we glean in the clinic, we can generate a more targeted and improved treatment algorithm for patients with CP.
Collapse
Affiliation(s)
- Eloise Stanton
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA, USA
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Samuel Sheridan
- Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, USA
| | - Mark Urata
- Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, USA
- Division of Plastic and Maxillofacial Surgery, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Yang Chai
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA, USA
- Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
40
|
Liu C, Zhang L, Zheng X, Zhu J, Jin L, Gao R. Pleiotrophin inhibited chondrogenic differentiation potential of dental pulp stem cells. Oral Dis 2024; 30:1439-1450. [PMID: 36840423 DOI: 10.1111/odi.14554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 02/13/2023] [Accepted: 02/22/2023] [Indexed: 02/26/2023]
Abstract
OBJECTIVE Studies have shown that the levels of pleiotrophin (PTN) are greatly elevated in the synovial fluid and cartilage in osteoarthritis. Therefore, the purpose of this study was to investigate the effect and mechanism of PTN on the chondrogenic differentiation of DPSCs in inflammatory and normal microenvironments. MATERIALS AND METHODS A lentiviral vector was used to deplete or overexpress PTN in DPSCs. The inflammatory microenvironment was simulated in vitro by the addition of IL-1β to the culture medium. The chondrogenic differentiation potential was assessed using Alcian Blue staining and the main chondrogenic markers. A dual-luciferase reporter assay was used to explore the relationship between miR-137 and PTN. RESULTS The results showed that 0.1 ng/mL IL-1β treatment during chondrogenic induction greatly impaired the chondrogenic differentiation of DPSCs. Supplementation with PTN and PTN overexpression inhibited chondrogenic differentiation of DPSCs, while PTN depletion promoted chondrogenic differentiation. MiR-137 negatively regulated the expression of PTN by binding to the 3'UTR of its mRNA. Moreover, miR-137 promoted chondrogenic differentiation of DPSCs in normal and inflammatory microenvironments. CONCLUSION Our results suggest that PTN may play an inhibitory role in the chondrogenic differentiation of DPSCs in normal and inflammatory microenvironments, which is regulated by miR-137.
Collapse
Affiliation(s)
- Chang Liu
- Department of Stomatology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Lili Zhang
- Department of General Dentistry and Integrated Emergency Dental Care, Beijing Stomatological Hospital, Capital Medical University, Beijing, China
| | - Xiaoyu Zheng
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China
| | - Jiaman Zhu
- Department of Stomatology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Luyuan Jin
- Department of General Dentistry and Integrated Emergency Dental Care, Beijing Stomatological Hospital, Capital Medical University, Beijing, China
| | - Runtao Gao
- Department of Stomatology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
41
|
Gancheva MR, Kremer K, Breen J, Arthur A, Hamilton-Bruce A, Thomas P, Gronthos S, Koblar S. Effect of Octamer-Binding Transcription Factor 4 Overexpression on the Neural Induction of Human Dental Pulp Stem Cells. Stem Cell Rev Rep 2024; 20:797-815. [PMID: 38316679 PMCID: PMC10984899 DOI: 10.1007/s12015-024-10678-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/08/2024] [Indexed: 02/07/2024]
Abstract
Stem cell-based therapy is a potential alternative strategy for brain repair, with neural stem cells (NSC) presenting as the most promising candidates. Obtaining sufficient quantities of NSC for clinical applications is challenging, therefore alternative cell types, such as neural crest-derived dental pulp stem cells (DPSC), may be considered. Human DPSC possess neurogenic potential, exerting positive effects in the damaged brain through paracrine effects. However, a method for conversion of DPSC into NSC has yet to be developed. Here, overexpression of octamer-binding transcription factor 4 (OCT4) in combination with neural inductive conditions was used to reprogram human DPSC along the neural lineage. The reprogrammed DPSC demonstrated a neuronal-like phenotype, with increased expression levels of neural markers, limited capacity for sphere formation, and enhanced neuronal but not glial differentiation. Transcriptomic analysis further highlighted the expression of genes associated with neural and neuronal functions. In vivo analysis using a developmental avian model showed that implanted DPSC survived in the developing central nervous system and respond to endogenous signals, displaying neuronal phenotypes. Therefore, OCT4 enhances the neural potential of DPSC, which exhibited characteristics aligning with neuronal progenitors. This method can be used to standardise DPSC neural induction and provide an alternative source of neural cell types.
Collapse
Affiliation(s)
- Maria R Gancheva
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, 5005, Australia.
- School of Biological Sciences, Faculty of Science, Engineering and Technology, The University of Adelaide, Adelaide, 5005, Australia.
| | - Karlea Kremer
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, 5005, Australia
| | - James Breen
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, 5005, Australia
- School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, 5005, Australia
| | - Agnes Arthur
- School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, 5005, Australia
| | - Anne Hamilton-Bruce
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, 5005, Australia
- Stroke Research Programme, Basil Hetzel Institute, The Queen Elizabeth Hospital, Central Adelaide Local Health Network, Woodville South, 5011, Australia
| | - Paul Thomas
- School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, 5005, Australia
- South Australian Health and Medical Research Institute, Adelaide, 5000, Australia
| | - Stan Gronthos
- School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, 5005, Australia
- South Australian Health and Medical Research Institute, Adelaide, 5000, Australia
| | - Simon Koblar
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, 5005, Australia
| |
Collapse
|
42
|
Zhao Q, Ren H, Wang N, Yuan X, Zhao Y, Wen Q. NOTUM plays a bidirectionally modulatory role in the odontoblastic differentiation of human stem cells from the apical papilla through the WNT/β-catenin signaling pathway. Arch Oral Biol 2024; 160:105896. [PMID: 38278124 DOI: 10.1016/j.archoralbio.2024.105896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 12/29/2023] [Accepted: 01/17/2024] [Indexed: 01/28/2024]
Abstract
OBJECTIVE Notum is a secreted deacylase, which is crucial for tooth dentin development in mice. This study aimed to investigate the effect of NOTUM on the odontoblastic differentiation of human stem cells from the apical papilla (hSCAPs), to reveal the potential value of NOTUM in pulp-dentin complex regeneration. DESIGN The expression pattern of NOTUM in human tooth germs and during in vitro odontoblastic differentiation of hSCAPs was evaluated by immunohistochemical staining, and quantitative polymerase chain reaction, respectively. To manipulate the extracellular NOTUM level, ABC99 or small interfering RNA was used to down-regulate it, while recombinant NOTUM protein was added to up-regulate it. The effects of changing NOTUM level on the odontoblastic differentiation of hSCAPs and its interaction with the WNT/β-catenin signaling pathway were studied using alkaline phosphatase staining, alizarin red staining, quantitative polymerase chain reaction, and western blot. RESULTS NOTUM was observed in the apical papilla of human tooth germs. During in vitro odontoblastic differentiation of hSCAPs, NOTUM expression initially increased, while the WNT/β-catenin pathway was activated. Downregulation of NOTUM hindered odontoblastic differentiation. Recombinant NOTUM protein had varying effects on odontoblastic differentiation depending on exposure duration. Continuous addition of the protein inhibited both odontoblastic differentiation and the WNT/β-catenin pathway. However, applying the protein solely in the first 3 days enhanced odontoblastic differentiation and up-regulated the WNT/β-catenin pathway. CONCLUSION NOTUM demonstrated a bidirectional impact on in vitro odontoblastic differentiation of hSCAPs, potentially mediated by the WNT/β-catenin pathway. These findings suggest its promising potential for pulp-dentin complex regeneration.
Collapse
Affiliation(s)
- Qingxuan Zhao
- Department of Pediatric Dentistry, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, No.22, Zhongguancun South Avenue, Haidian District, Beijing 100081, PR China
| | - Huihui Ren
- Department of Pediatric Dentistry, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, No.22, Zhongguancun South Avenue, Haidian District, Beijing 100081, PR China
| | - Nan Wang
- Department of Pediatric Dentistry, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, No.22, Zhongguancun South Avenue, Haidian District, Beijing 100081, PR China
| | - Xiaojing Yuan
- Department of Pediatric Dentistry, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, No.22, Zhongguancun South Avenue, Haidian District, Beijing 100081, PR China
| | - Yuming Zhao
- Department of Pediatric Dentistry, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, No.22, Zhongguancun South Avenue, Haidian District, Beijing 100081, PR China.
| | - Quan Wen
- First Clinical Division, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, 37A Xishiku Street, Xicheng District, Beijing 100034, PR China.
| |
Collapse
|
43
|
Yang D, Jeong Y, Ortinau L, Solidum J, Park D. Mx1 -labeled pulp progenitor cells are main contributors to postnatal odontoblasts and pulp cells in murine molars. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.21.586156. [PMID: 38585950 PMCID: PMC10996506 DOI: 10.1101/2024.03.21.586156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Regeneration of dentin and odontoblasts from dental pulp stem cells (DPSCs) is essential for permanent tooth maintenance. However, the in vivo identity and role of endogenous DPSCs in reparative dentinogenesis are elusive. Here, using pulp single-cell analysis before and after molar eruption, we revealed that endogenous DPSCs are enriched in Cxcl12- GFP + coronal papilla-like cells with Mx1- Cre labeling. These Mx1 + Cxcl12- GFP + cells are long-term repopulating cells that contribute to the majority of pulp cells and new odontoblasts after eruption. Upon molar injury, Mx1 + DPSCs localize into the injury site and differentiate into new odontoblasts, forming scleraxis -GFP + and osteocalcin -GFP + dentinal tubules and reparative dentin. Single-cell and FACS analysis showed that Mx1 + Cxcl12- GFP + DPSCs are the most primitive cells with stem cell marker expression and odontoblast differentiation. Taken together, our findings demonstrate that Mx1 labels postnatal DSPCs, which are the main source of pulp cells and new odontoblasts with reparative dentinogenesis in vivo .
Collapse
|
44
|
Wang W, Yang N, Wang L, Zhu Y, Chu X, Xu W, Li Y, Xu Y, Gao L, Zhang B, Zhang G, Sun Q, Wang W, Wang Q, Zhang W, Chen D. The TET-Sall4-BMP regulatory axis controls craniofacial cartilage development. Cell Rep 2024; 43:113873. [PMID: 38427557 DOI: 10.1016/j.celrep.2024.113873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 07/25/2023] [Accepted: 02/12/2024] [Indexed: 03/03/2024] Open
Abstract
Craniofacial microsomia (CFM) is a congenital defect that usually results from aberrant development of embryonic pharyngeal arches. However, the molecular basis of CFM pathogenesis is largely unknown. Here, we employ the zebrafish model to investigate mechanisms of CFM pathogenesis. In early embryos, tet2 and tet3 are essential for pharyngeal cartilage development. Single-cell RNA sequencing reveals that loss of Tet2/3 impairs chondrocyte differentiation due to insufficient BMP signaling. Moreover, biochemical and genetic evidence reveals that the sequence-specific 5mC/5hmC-binding protein, Sall4, binds the promoter of bmp4 to activate bmp4 expression and control pharyngeal cartilage development. Mechanistically, Sall4 directs co-phase separation of Tet2/3 with Sall4 to form condensates that mediate 5mC oxidation on the bmp4 promoter, thereby promoting bmp4 expression and enabling sufficient BMP signaling. These findings suggest the TET-BMP-Sall4 regulatory axis is critical for pharyngeal cartilage development. Collectively, our study provides insights into understanding craniofacial development and CFM pathogenesis.
Collapse
Affiliation(s)
- Weigang Wang
- Institute of Biomedical Research, Yunnan University, Kunming, China
| | - Na Yang
- Institute of Biomedical Research, Yunnan University, Kunming, China; Department of Ultrasound, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Liangliang Wang
- Institute of Biomedical Research, Yunnan University, Kunming, China
| | - Yuanxiang Zhu
- Institute of Biomedical Research, Yunnan University, Kunming, China
| | - Xiao Chu
- Institute of Biomedical Research, Yunnan University, Kunming, China
| | - Weijie Xu
- Institute of Biomedical Research, Yunnan University, Kunming, China
| | - Yawei Li
- Institute of Biomedical Research, Yunnan University, Kunming, China
| | - Yihai Xu
- Institute of Biomedical Research, Yunnan University, Kunming, China
| | - Lina Gao
- Institute of Biomedical Research, Yunnan University, Kunming, China
| | - Beibei Zhang
- Institute of Biomedical Research, Yunnan University, Kunming, China
| | - Guoqiang Zhang
- Institute of Biomedical Research, Yunnan University, Kunming, China
| | - Qinmiao Sun
- Institute of Stem Cells and Regeneration, Chinese Academy of Sciences, Beijing, China
| | - Weihong Wang
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatology Hospital of Kunming Medical University, Kunming, China.
| | - Qiang Wang
- Division of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou, China.
| | - Wenxin Zhang
- Institute of Biomedical Research, Yunnan University, Kunming, China.
| | - Dahua Chen
- Institute of Biomedical Research, Yunnan University, Kunming, China; Southwest United Graduate School, Kunming, China.
| |
Collapse
|
45
|
Moore E, Zhao R, McKinney MC, Yi K, Wood C, Trainor P. Cell extrusion - a novel mechanism driving neural crest cell delamination. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.09.584232. [PMID: 38559094 PMCID: PMC10979875 DOI: 10.1101/2024.03.09.584232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Neural crest cells (NCC) comprise a heterogeneous population of cells with variable potency, that contribute to nearly every tissue and organ system throughout the body. Considered unique to vertebrates, NCC are transiently generated within the dorsolateral region of the neural plate or neural tube, during neurulation. Their delamination and migration are crucial events in embryo development as the differentiation of NCC is heavily influenced by their final resting locations. Previous work in avian and aquatic species has shown that NCC delaminate via an epithelial-mesenchymal transition (EMT), which transforms these stem and progenitor cells from static polarized epithelial cells into migratory mesenchymal cells with fluid front and back polarity. However, the cellular and molecular drivers facilitating NCC delamination in mammals are poorly understood. We performed live timelapse imaging of NCC delamination in mouse embryos and discovered a group of cells that exit the neuroepithelium as isolated round cells, which then halt for a short period prior to acquiring the mesenchymal migratory morphology classically associated with most delaminating NCC. High magnification imaging and protein localization analyses of the cytoskeleton, together with measurements of pressure and tension of delaminating NCC and neighboring neuroepithelial cells, revealed these round NCC are extruded from the neuroepithelium prior to completion of EMT. Furthermore, we demonstrate that cranial NCC are extruded through activation of the mechanosensitive ion channel, PIEZO1, a key regulator of the live cell extrusion pathway, revealing a new role for PIEZO1 in neural crest cell development. Our results elucidating the cellular and molecular dynamics orchestrating NCC delamination support a model in which high pressure and tension in the neuroepithelium results in activation of the live cell extrusion pathway and delamination of a subpopulation of NCC in parallel with EMT. This model has broad implications for our understanding of cell delamination in development and disease.
Collapse
Affiliation(s)
- Emma Moore
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Ruonan Zhao
- Stowers Institute for Medical Research, Kansas City, MO, USA
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Mary C McKinney
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Kexi Yi
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | | | - Paul Trainor
- Stowers Institute for Medical Research, Kansas City, MO, USA
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
46
|
Kong Q, Wang Y, Jiang N, Wang Y, Wang R, Hu X, Mao J, Shi X. Exosomes as Promising Therapeutic Tools for Regenerative Endodontic Therapy. Biomolecules 2024; 14:330. [PMID: 38540750 PMCID: PMC10967740 DOI: 10.3390/biom14030330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 03/03/2024] [Accepted: 03/08/2024] [Indexed: 11/06/2024] Open
Abstract
Pulpitis is a common and frequent disease in dental clinics. Although vital pulp therapy and root canal treatment can stop the progression of inflammation, they do not allow for genuine structural regeneration and functional reconstruction of the pulp-dentin complex. In recent years, with the development of tissue engineering and regenerative medicine, research on stem cell-based regenerative endodontic therapy (RET) has achieved satisfactory preliminary results, significantly enhancing its clinical translational prospects. As one of the crucial paracrine effectors, the roles and functions of exosomes in pulp-dentin complex regeneration have gained considerable attention. Due to their advantages of cost-effectiveness, extensive sources, favorable biocompatibility, and high safety, exosomes are considered promising therapeutic tools to promote dental pulp regeneration. Accordingly, in this article, we first focus on the biological properties of exosomes, including their biogenesis, uptake, isolation, and characterization. Then, from the perspectives of cell proliferation, migration, odontogenesis, angiogenesis, and neurogenesis, we aim to reveal the roles and mechanisms of exosomes involved in regenerative endodontics. Lastly, immense efforts are made to illustrate the clinical strategies and influencing factors of exosomes applied in dental pulp regeneration, such as types of parental cells, culture conditions of parent cells, exosome concentrations, and scaffold materials, in an attempt to lay a solid foundation for exploring and facilitating the therapeutic strategy of exosome-based regenerative endodontic procedures.
Collapse
Affiliation(s)
- Qingyue Kong
- Center of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Q.K.); (Y.W.); (Y.W.); (R.W.)
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Yujie Wang
- Center of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Q.K.); (Y.W.); (Y.W.); (R.W.)
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Nan Jiang
- Central Laboratory, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing 100081, China;
| | - Yifan Wang
- Center of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Q.K.); (Y.W.); (Y.W.); (R.W.)
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Rui Wang
- Center of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Q.K.); (Y.W.); (Y.W.); (R.W.)
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Xiaohan Hu
- Outpatient Department Office, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China;
| | - Jing Mao
- Center of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Q.K.); (Y.W.); (Y.W.); (R.W.)
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Xin Shi
- Center of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Q.K.); (Y.W.); (Y.W.); (R.W.)
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| |
Collapse
|
47
|
Feng Y, Wang H, Xu S, Huang J, Pei Q, Wang Z. The detection of Gper1 as an important gene promoting jawbone regeneration in the context of estrogen deficiency. Bone 2024; 180:116990. [PMID: 38141748 DOI: 10.1016/j.bone.2023.116990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 12/13/2023] [Accepted: 12/18/2023] [Indexed: 12/25/2023]
Abstract
Numerous studies have demonstrated that estrogen deficiency inhibit the proliferation and differentiation of pre-osteoblasts in skeleton by affecting osteogenic signaling, lead to decreased bone mass and impaired regeneration. To explore the mechanisms maintaining bone regeneration under estrogen deficiency, we randomly selected 1102 clinical cases, in which female patients aged between 18 and 75 have underwent tooth extraction in Stomatological Hospital of Tongji University, there is little difference in the healing effect of extraction defects, suggesting that to some extent, the regeneration of jawbone is insensitive to the decreased estrogen level. To illuminate the mechanisms promoting jawbone regeneration under estrogen deficiency, a tooth extraction defect model was established in the maxilla of female rats who underwent ovariectomy (OVX) or sham surgery, and jawbone marrow stromal cells (BMSCs) were isolated for single-cell sequencing. Further quantitative PCR, RNA interference, alizarin red staining, immunohistochemistry and western blotting experiments demonstrated that in the context of ovariectomy, maxillary defects promoted G protein-coupled estrogen receptor 1 (Gper1) expression, stimulate downstream cAMP/PKA/pCREB signaling, and facilitate cell proliferation, and thus provided sufficient progenitors for osteogenesis and enhanced the regeneration capacity of the jawbone. Correspondingly, the heterozygous deletion of the Gper1 gene attenuated the phosphorylation of CREB, led to decreased cell proliferation, and impaired the restoration of maxillary defects. This study demonstrates the importance of Gper1 in maintaining jawbone regeneration, especially in the context of estrogen deficiency.
Collapse
Affiliation(s)
- Yuan Feng
- Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, No.399 Middle Yanchang Road, Shanghai 200072, PR China
| | - Haicheng Wang
- Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, No.399 Middle Yanchang Road, Shanghai 200072, PR China
| | - Shuyu Xu
- Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, No.399 Middle Yanchang Road, Shanghai 200072, PR China
| | - Jie Huang
- Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, No.399 Middle Yanchang Road, Shanghai 200072, PR China
| | - Qingguo Pei
- Department of Stomatology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No.100 Haining Road, Shanghai 200080, PR China
| | - Zuolin Wang
- Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, No.399 Middle Yanchang Road, Shanghai 200072, PR China.
| |
Collapse
|
48
|
Dinwoodie OM, Tucker AS, Fons JM. Tracking cell layer contribution during repair of the tympanic membrane. Dis Model Mech 2024; 17:dmm050466. [PMID: 38390727 PMCID: PMC10985735 DOI: 10.1242/dmm.050466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 02/17/2024] [Indexed: 02/24/2024] Open
Abstract
The tympanic membrane (i.e. eardrum) sits at the interface between the middle and external ear. The tympanic membrane is composed of three layers: an outer ectoderm-derived layer, a middle neural crest-derived fibroblast layer with contribution from the mesoderm-derived vasculature, and an inner endoderm-derived mucosal layer. These layers form a thin sandwich that is often perforated following trauma, pressure changes or middle ear inflammation. During healing, cells need to bridge the perforation in the absence of an initial scaffold. Here, we assessed the contribution, timing and interaction of the different layers during membrane repair by using markers and reporter mice. We showed that the ectodermal layer is retracted after perforation, before proliferating away from the wound edge, with keratin 5 basal cells migrating over the hole to bridge the gap. The mesenchymal and mucosal layers then used this scaffold to complete the repair, followed by advancement of the vasculature. Finally, differentiation of the epithelium led to formation of a scab. Our results reveal the dynamics and interconnections between the embryonic germ layers during repair and highlight how defects might occur.
Collapse
Affiliation(s)
- Olivia M. Dinwoodie
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London SE1 9RT, UK
| | - Abigail S. Tucker
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London SE1 9RT, UK
| | - Juan M. Fons
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London SE1 9RT, UK
| |
Collapse
|
49
|
Zhao R, Moore EL, Gogol MM, Unruh JR, Yu Z, Scott A, Wang Y, Rajendran NK, Trainor PA. Identification and characterization of intermediate states in mammalian neural crest cell epithelial to mesenchymal transition and delamination. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.10.26.564204. [PMID: 37961316 PMCID: PMC10634855 DOI: 10.1101/2023.10.26.564204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Epithelial to mesenchymal transition (EMT) is a cellular process that converts epithelial cells to mesenchymal cells with migratory potential in both developmental and pathological processes. Although originally considered a binary event, EMT in cancer progression involves intermediate states between a fully epithelial and a fully mesenchymal phenotype, which are characterized by distinct combinations of epithelial and mesenchymal markers. This phenomenon has been termed epithelial to mesenchymal plasticity (EMP), however, the intermediate states remain poorly described and it's unclear whether they exist during developmental EMT. Neural crest cells (NCC) are an embryonic progenitor cell population that gives rise to numerous cell types and tissues in vertebrates, and their formation is a classic example of developmental EMT. An important feature of NCC development is their delamination from the neuroepithelium via EMT, following which NCC migrate throughout the embryo and undergo differentiation. NCC delamination shares similar changes in cellular state and structure with cancer cell invasion. However, whether intermediate states also exist during NCC EMT and delamination remains unknown. Through single cell RNA sequencing, we identified intermediate NCC states based on their transcriptional signature and then spatially defined their locations in situ in the dorsolateral neuroepithelium. Our results illustrate the progressive transcriptional and spatial transitions from premigratory to migratory cranial NCC during EMT and delamination. Of note gene expression and trajectory analysis indicate that distinct intermediate populations of NCC delaminate in either S phase or G2/M phase of the cell cycle, and the importance of cell cycle regulation in facilitating mammalian cranial NCC delamination was confirmed through cell cycle inhibition studies. Additionally, transcriptional knockdown revealed a functional role for the intermediate stage marker Dlc1 in regulating NCC delamination and migration. Overall, our work identifying and characterizing the intermediate cellular states, processes, and molecular signals that regulate mammalian NCC EMT and delamination furthers our understanding of developmental EMP and may provide new insights into mechanisms regulating pathological EMP.
Collapse
Affiliation(s)
- Ruonan Zhao
- Stowers Institute for Medical Research, Kansas City, MO, USA
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Emma L. Moore
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | | | - Jay R. Unruh
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Zulin Yu
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Allison Scott
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Yan Wang
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | | | - Paul A. Trainor
- Stowers Institute for Medical Research, Kansas City, MO, USA
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
50
|
Edens BM, Bronner ME. Making developmental sense of the senses, their origin and function. Curr Top Dev Biol 2024; 159:132-167. [PMID: 38729675 DOI: 10.1016/bs.ctdb.2024.01.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2024]
Abstract
The primary senses-touch, taste, sight, smell, and hearing-connect animals with their environments and with one another. Aside from the eyes, the primary sense organs of vertebrates and the peripheral sensory pathways that relay their inputs arise from two transient stem cell populations: the neural crest and the cranial placodes. In this chapter we consider the senses from historical and cultural perspectives, and discuss the senses as biological faculties. We begin with the embryonic origin of the neural crest and cranial placodes from within the neural plate border of the ectodermal germ layer. Then, we describe the major chemical (i.e. olfactory and gustatory) and mechanical (i.e. vestibulo-auditory and somatosensory) senses, with an emphasis on the developmental interactions between neural crest and cranial placodes that shape their structures and functions.
Collapse
Affiliation(s)
- Brittany M Edens
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, United States
| | - Marianne E Bronner
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, United States.
| |
Collapse
|