201
|
Men Y, Wang Y, Yi Y, Jing D, Luo W, Shen B, Stenberg W, Chai Y, Ge WP, Feng JQ, Zhao H. Gli1+ Periodontium Stem Cells Are Regulated by Osteocytes and Occlusal Force. Dev Cell 2020; 54:639-654.e6. [PMID: 32652075 DOI: 10.1016/j.devcel.2020.06.006] [Citation(s) in RCA: 99] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 02/04/2020] [Accepted: 06/02/2020] [Indexed: 01/05/2023]
Abstract
Teeth are attached to alveolar bone by the periodontal ligament (PDL), which contains stem cells supporting tissue turnover. Here, we identified Gli1+ cells in adult mouse molar PDL as multi-potential stem cells (PDLSCs) giving rise to PDL, alveolar bone, and cementum. They support periodontium tissue turnover and injury repair. Gli1+ PDLSCs are surrounding the neurovascular bundle and more enriched in the apical region. Canonical Wnt signaling is essential for their activation. Alveolar bone osteocytes negatively regulate Gli1+ PDLSCs activity through sclerostin, a Wnt inhibitor. Blockage of sclerostin accelerates the PDLSCs lineage contribution rate in vivo. Sclerostin expression is modulated by physiological occlusal force. Removal of occlusal force upregulates sclerostin and inhibits PDLSCs activation. In summary, Gli1+ cells are the multipotential PDLSCs in vivo. Osteocytes provide negative feedback to PDLSCs and inhibit their activities through sclerostin. Physiological occlusal force indirectly regulates PDLSCs activities by fine-tuning this feedback loop.
Collapse
Affiliation(s)
- Yi Men
- Department of Comprehensive Dentistry, College of Dentistry, Texas A&M University, Dallas, TX 75246, USA; West China School of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yuhong Wang
- Department of Comprehensive Dentistry, College of Dentistry, Texas A&M University, Dallas, TX 75246, USA; West China School of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yating Yi
- Department of Comprehensive Dentistry, College of Dentistry, Texas A&M University, Dallas, TX 75246, USA
| | - Dian Jing
- Department of Comprehensive Dentistry, College of Dentistry, Texas A&M University, Dallas, TX 75246, USA
| | - Wenjing Luo
- Department of Biomedical Sciences, College of Dentistry, Texas A&M University, Dallas, TX 75246, USA
| | - Bo Shen
- Children's Research Institute, UT Southwestern Medical Center Dallas, TX 75235, USA
| | - William Stenberg
- Department of Comprehensive Dentistry, College of Dentistry, Texas A&M University, Dallas, TX 75246, USA
| | - Yang Chai
- Center for Craniofacial Molecular Biology, University of Southern California, Herman Ostrow School of Dentistry, Los Angeles, CA 90089, USA
| | - Woo-Ping Ge
- Chinese Institute for Brain Research, Beijing 102206, China
| | - Jian Q Feng
- Department of Biomedical Sciences, College of Dentistry, Texas A&M University, Dallas, TX 75246, USA
| | - Hu Zhao
- Department of Comprehensive Dentistry, College of Dentistry, Texas A&M University, Dallas, TX 75246, USA.
| |
Collapse
|
202
|
Zhang D, Zhang S, Wang J, Li Q, Xue H, Sheng R, Xiong Q, Qi X, Wen J, Fan Y, Zhou B, Yuan Q. LepR-Expressing Stem Cells Are Essential for Alveolar Bone Regeneration. J Dent Res 2020; 99:1279-1286. [PMID: 32585118 DOI: 10.1177/0022034520932834] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Stem cells play a critical role in bone regeneration. Multiple populations of skeletal stem cells have been identified in long bone, while their identity and functions in alveolar bone remain unclear. Here, we identified a quiescent leptin receptor–expressing (LepR+) cell population that contributed to intramembranous bone formation. Interestingly, these LepR+ cells became activated in response to tooth extraction and generated the majority of the newly formed bone in extraction sockets. In addition, genetic ablation of LepR+ cells attenuated extraction socket healing. The parabiosis experiments revealed that the LepR+ cells in the healing sockets were derived from resident tissue rather than peripheral blood circulation. Further studies on the mechanism suggested that these LepR+ cells were responsive to parathyroid hormone/parathyroid hormone 1 receptor (PTH/PTH1R) signaling. Collectively, we demonstrate that LepR+ cells, a postnatal skeletal stem cell population, are essential for alveolar bone regeneration of extraction sockets.
Collapse
Affiliation(s)
- D. Zhang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - S. Zhang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - J. Wang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Periodontology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Q. Li
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - H. Xue
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - R. Sheng
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Q. Xiong
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - X. Qi
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - J. Wen
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Y. Fan
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - B.O. Zhou
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai, China
| | - Q. Yuan
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
203
|
Bi R, Yin Q, Mei J, Chen K, Luo X, Fan Y, Zhu S. Identification of human temporomandibular joint fibrocartilage stem cells with distinct chondrogenic capacity. Osteoarthritis Cartilage 2020; 28:842-852. [PMID: 32147536 DOI: 10.1016/j.joca.2020.02.835] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 02/08/2020] [Accepted: 02/26/2020] [Indexed: 02/05/2023]
Abstract
OBJECTIVE This study was aimed to identify the residence of human fibrocartilage stem cells (hFCSCs), characterize their stem cell properties and investigate the functional mechanisms which regulate fibrocartilage stem cells (FCSCs) toward chondrogenic differentiation during cartilage homeostasis and repairing. METHODS Cytological characteristics of hFCSCs and human orofacial mesenchymal stem cells (hOFMSCs) were analyzed. Chondrogenic potential of hFCSCs was compared with hOFMSCs both in vitro and in vivo. Regulatory role of SOX9 during FCSCs chondrogenesis was studied by shRNA interference in vitro, and by GFP+ FCSCs treatment in rat condylar cartilage defect model. SOX9 expression was also examined in temporomandibular joint osteoarthritis (TMJOA) patients' cartilage surface. RESULTS hFCSCs exhibited typical mesenchymal stem cell characteristics, with significantly stronger chondrogenic capability compared to hOFMSCs. Moreover, hFCSCs showed remarkably increased expression of SOX9. During cartilage pellet culture, there was stronger SOX9 expression in hFCSCs than hOFMSCs. SOX9 shRNA interference downregulated chondrogenic capability of hFCSCs in vitro, as well as disrupting migration and chondrogenic differentiation of GFP+ FCSCs toward mature chondrocytes in rat condylar cartilage defect. Of note, SOX9 expression was also found suppressed in the condylar superficial zone of TMJOA patients. CONCLUSION We found the existence of FCSCs in human TMJ cartilage, and characterized their distinct stem cell features. SOX9 is essential for hFCSCs chondrogenic differentiation, and a comprehensive understanding of the regulatory role of SOX9 in hFCSCs would be important for exploring potential intervention strategy of condylar cartilage degradation during TMJ disorders.
Collapse
Affiliation(s)
- R Bi
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthognathic and TMJ Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - Q Yin
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthognathic and TMJ Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - J Mei
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthognathic and TMJ Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - K Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthognathic and TMJ Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - X Luo
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthognathic and TMJ Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - Y Fan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Operative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - S Zhu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthognathic and TMJ Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
204
|
Pezoa SA, Artinger KB, Niswander LA. GCN5 acetylation is required for craniofacial chondrocyte maturation. Dev Biol 2020; 464:24-34. [PMID: 32446700 DOI: 10.1016/j.ydbio.2020.05.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 04/21/2020] [Accepted: 05/13/2020] [Indexed: 02/06/2023]
Abstract
Development of the craniofacial structures requires the precise differentiation of cranial neural crest cells into osteoblasts or chondrocytes. Here, we explore the epigenetic and non-epigenetic mechanisms that are required for the development of craniofacial chondrocytes. We previously demonstrated that the acetyltransferase activity of the highly conserved acetyltransferase GCN5, or KAT2A, is required for murine craniofacial development. We show that Gcn5 is required cell autonomously in the cranial neural crest. Moreover, GCN5 is required for chondrocyte development following the arrival of the cranial neural crest within the pharyngeal arches. Using a combination of in vivo and in vitro inhibition of GCN5 acetyltransferase activity, we demonstrate that GCN5 is a potent activator of chondrocyte maturation, acting to control chondrocyte maturation and size increase during pre-hypertrophic maturation to hypertrophic chondrocytes. Rather than acting as an epigenetic regulator of histone H3K9 acetylation, our findings suggest GCN5 primarily acts as a non-histone acetyltransferase to regulate chondrocyte development. Here, we investigate the contribution of GCN5 acetylation to the activity of the mTORC1 pathway. Our findings indicate that GCN5 acetylation is required for activation of this pathway, either via direct activation of mTORC1 or through indirect mechanisms. We also investigate one possibility of how mTORC1 activity is regulated through RAPTOR acetylation, which is hypothesized to enhance mTORC1 downstream phosphorylation. This study contributes to our understanding of the specificity of acetyltransferases, and the cell type specific roles in which these enzymes function.
Collapse
Affiliation(s)
- Sofia A Pezoa
- Cell Biology, Stem Cells, and Developmental Biology Graduate Program. University of Colorado Anschutz School of Medicine, Aurora, CO, USA, 80045; Department of Molecular, Cellular, and Developmental Biology. University of Colorado Boulder, Boulder, CO, USA, 80309
| | - Kristin B Artinger
- Department of Craniofacial Biology, University of Colorado Anschutz School of Dentistry, Aurora, CO, USA, 80045
| | - Lee A Niswander
- Department of Molecular, Cellular, and Developmental Biology. University of Colorado Boulder, Boulder, CO, USA, 80309.
| |
Collapse
|
205
|
Melms H, Herrmann M, Förstner K, Bharti R, Schneider D, Mentrup B, Rudert M, Schlagenhauf U, Jakob F, Graser S. Novel molecular cues for dental defects in hypophosphatasia. Exp Cell Res 2020; 392:112026. [PMID: 32333908 DOI: 10.1016/j.yexcr.2020.112026] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 04/15/2020] [Accepted: 04/17/2020] [Indexed: 01/09/2023]
Abstract
Mineralization disorders with a broad range of etiological factors represent a huge challenge in dental diagnosis and therapy. Hypophosphatasia (HPP) belongs to the rare diseases affecting predominantly mineralized tissues, bones and teeth, and occurs due to mutations in the ALPL gene, which encodes tissue-nonspecific alkaline phosphatase (TNAP). Here we analyzed stem cells from bone marrow (BMSCs), dental pulp (DPSCs) and periodontal ligament (PDLSCs) in the absence and presence of efficient TNAP inhibitors. The differentiation capacity, expression of surface markers, and gene expression patterns of donor-matched dental cells were compared during this in vitro study. Differentiation assays showed efficient osteogenic but low adipogenic differentiation (aD) capacity of PDLSCs and DPSCs. TNAP inhibitor treatment completely abolished the mineralization process during osteogenic differentiation (oD). RNA-seq analysis in PDLSCs, comparing oD with and without TNAP inhibitor levamisole, showed clustered regulation of candidate molecular mechanisms that putatively impaired osteogenesis and mineralization, disequilibrated ECM production and turnover, and propagated inflammation. Combined alteration of cementum formation, mineralization, and elastic attachment of teeth to cementum via elastic fibers may explain dental key problems in HPP. Using this in vitro model of TNAP deficiency in DPSCs and PDLSCs, we provide novel putative target areas for research on molecular cues for specific dental problems in HPP.
Collapse
Affiliation(s)
- Hannah Melms
- Bernhard-Heine-Center for Locomotion Research, University of Würzburg, Würzburg, Germany; Department of Conservative Dentistry, School of Dental Medicine, University Hospital Tübingen, Tübingen, Germany
| | - Marietta Herrmann
- Bernhard-Heine-Center for Locomotion Research, University of Würzburg, Würzburg, Germany; IZKF Research Group Tissue Regeneration in Musculoskeletal Diseases, University Hospital Würzburg, Würzburg, Germany
| | - Konrad Förstner
- Core Unit SysMed, University of Würzburg, Würzburg, Germany; Institute for Information Sciences, University of Applied Sciences, Cologne, Germany; ZB MED - Information Centre for Life Science, Cologne, Germany
| | - Richa Bharti
- Core Unit SysMed, University of Würzburg, Würzburg, Germany; Department for Bioinformatics, Weihenstephan-Triesdorf University of Applied Sciences, TUM Campus Straubing for Biotechnology and Sustainability, Straubing, Germany
| | - Doris Schneider
- Bernhard-Heine-Center for Locomotion Research, University of Würzburg, Würzburg, Germany
| | - Birgit Mentrup
- Bernhard-Heine-Center for Locomotion Research, University of Würzburg, Würzburg, Germany; Department for Regenerative Musculoskeletal Medicine, University of Münster, Germany
| | - Maximilian Rudert
- Department of Orthopaedics, Orthopedic Department König-Ludwig-Haus, University of Würzburg, Würzburg, Germany
| | - Ulrich Schlagenhauf
- Department of Periodontology, University Clinics Würzburg, Würzburg, Germany
| | - Franz Jakob
- Bernhard-Heine-Center for Locomotion Research, University of Würzburg, Würzburg, Germany
| | - Stephanie Graser
- Bernhard-Heine-Center for Locomotion Research, University of Würzburg, Würzburg, Germany.
| |
Collapse
|
206
|
Pouraghaei S, Moztarzadeh F, Chen C, Ansari S, Moshaverinia A. Microenvironment Can Induce Development of Auditory Progenitor Cells from Human Gingival Mesenchymal Stem Cells. ACS Biomater Sci Eng 2020; 6:2263-2273. [PMID: 33455314 DOI: 10.1021/acsbiomaterials.9b01795] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Sensorineural hearing loss in mammals occurs due to irreversible damage to the sensory epithelia of the inner ear and has very limited treatment options. The ability to regenerate the auditory progenitor cells is a promising approach for the treatment of sensorineural hearing loss; therefore, finding an appropriate and easily accessible stem cell source for restoring the sense of hearing would be of great interest. Here, we proposed a novel easy-to-access source of cells with the ability to recover auditory progenitor cells. In this study, gingival mesenchymal stem cells (GMSCs) were utilized, as these cells have high self-renewal and multipotent differentiation capacity and can be obtained easily from the oral cavity or discarded tissue samples at dental clinics. To manipulate the biophysical properties of the cellular microenvironment for promoting GMSC differentiation toward the target cells, we also tried to propose a candidate biomaterial. GMSCs in combination with an appropriate scaffold material can, therefore, present advantageous therapeutic options for a number of conditions. Here, we report the potential of GMSCs to differentiate into auditory progenitor cells while supporting them with an optimized three-dimensional scaffold and certain growth factors. A hybrid hydrogel scaffold based on peptide modified alginate and Matrigel was used here in addition to the presence of fibroblast growth factor-basic (bFGF), insulin-like growth factor (IGF), and epidermal growth factor (EGF). Our in vitro and in vivo studies confirmed the auditory differentiation potential of GMSCs within the engineered microenvironment.
Collapse
Affiliation(s)
- Sevda Pouraghaei
- Department of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran
- Weintraub Center for Reconstructive Biotechnology, Division of Advanced Prosthodontics, School of Dentistry, University of California, Los Angeles, California, United States
| | - Fathollah Moztarzadeh
- Department of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran
| | - Chider Chen
- Department of Oral and Maxillofacial Surgery and Pharmacology, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Sahar Ansari
- Department of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran
| | - Alireza Moshaverinia
- Weintraub Center for Reconstructive Biotechnology, Division of Advanced Prosthodontics, School of Dentistry, University of California, Los Angeles, California, United States
- California NanoSystems Institute, University of California, Los Angeles, California, United States
| |
Collapse
|
207
|
Huang Q, Cohen MA, Alsina FC, Devlin G, Garrett A, McKey J, Havlik P, Rakhilin N, Wang E, Xiang K, Mathews P, Wang L, Bock C, Ruthig V, Wang Y, Negrete M, Wong CW, Murthy PKL, Zhang S, Daniel AR, Kirsch DG, Kang Y, Capel B, Asokan A, Silver DL, Jaenisch R, Shen X. Intravital imaging of mouse embryos. Science 2020; 368:181-186. [PMID: 32273467 PMCID: PMC7646360 DOI: 10.1126/science.aba0210] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 03/16/2020] [Indexed: 12/11/2022]
Abstract
Embryonic development is a complex process that is unamenable to direct observation. In this study, we implanted a window to the mouse uterus to visualize the developing embryo from embryonic day 9.5 to birth. This removable intravital window allowed manipulation and high-resolution imaging. In live mouse embryos, we observed transient neurotransmission and early vascularization of neural crest cell (NCC)-derived perivascular cells in the brain, autophagy in the retina, viral gene delivery, and chemical diffusion through the placenta. We combined the imaging window with in utero electroporation to label and track cell division and movement within embryos and observed that clusters of mouse NCC-derived cells expanded in interspecies chimeras, whereas adjacent human donor NCC-derived cells shrank. This technique can be combined with various tissue manipulation and microscopy methods to study the processes of development at unprecedented spatiotemporal resolution.
Collapse
Affiliation(s)
- Qiang Huang
- Department of Pediatric Surgery, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, USA
| | - Malkiel A Cohen
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - Fernando C Alsina
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC, USA
| | - Garth Devlin
- Department of Surgery, School of Medicine, Duke University, Durham, NC, USA
| | - Aliesha Garrett
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, USA
| | - Jennifer McKey
- Department of Cell Biology, School of Medicine, Duke University, Durham, NC, USA
| | - Patrick Havlik
- Department of Surgery, School of Medicine, Duke University, Durham, NC, USA
| | - Nikolai Rakhilin
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, USA
| | - Ergang Wang
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, USA
| | - Kun Xiang
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, USA
| | - Parker Mathews
- Division of Hematologic Malignancies and Cellular Therapy, Duke University Medical Center, Durham, NC, USA
| | - Lihua Wang
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, USA
| | - Cheryl Bock
- Duke Cancer Institute, School of Medicine, Duke University, Durham, NC, USA
| | - Victor Ruthig
- Department of Cell Biology, School of Medicine, Duke University, Durham, NC, USA
| | - Yi Wang
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, USA
| | - Marcos Negrete
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, USA
| | - Chi Wut Wong
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, USA
| | - Preetish K L Murthy
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, USA
| | - Shupei Zhang
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - Andrea R Daniel
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, USA
| | - David G Kirsch
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, USA
- Department of Pharmacology & Cancer Biology, Duke University Medical Center, Durham, NC, USA
| | - Yubin Kang
- Division of Hematologic Malignancies and Cellular Therapy, Duke University Medical Center, Durham, NC, USA
| | - Blanche Capel
- Department of Cell Biology, School of Medicine, Duke University, Durham, NC, USA
| | - Aravind Asokan
- Department of Surgery, School of Medicine, Duke University, Durham, NC, USA
| | - Debra L Silver
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC, USA
| | - Rudolf Jaenisch
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA.
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Xiling Shen
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, USA.
| |
Collapse
|
208
|
Chen X, Wang H, Yu M, Kim JK, Qi H, Ha P, Jiang W, Chen E, Luo X, Needle RB, Baik L, Yang C, Shi J, Kwak JH, Ting K, Zhang X, Soo C. Cumulative inactivation of Nell-1 in Wnt1 expressing cell lineages results in craniofacial skeletal hypoplasia and postnatal hydrocephalus. Cell Death Differ 2020; 27:1415-1430. [PMID: 31582804 PMCID: PMC7206096 DOI: 10.1038/s41418-019-0427-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Revised: 07/09/2019] [Accepted: 08/26/2019] [Indexed: 02/05/2023] Open
Abstract
Upregulation of Nell-1 has been associated with craniosynostosis (CS) in humans, and validated in a mouse transgenic Nell-1 overexpression model. Global Nell-1 inactivation in mice by N-ethyl-N-nitrosourea (ENU) mutagenesis results in neonatal lethality with skeletal abnormalities including cleidocranial dysplasia (CCD)-like calvarial bone defects. This study further defines the role of Nell-1 in craniofacial skeletogenesis by investigating specific inactivation of Nell-1 in Wnt1 expressing cell lineages due to the importance of cranial neural crest cells (CNCCs) in craniofacial tissue development. Nell-1flox/flox; Wnt1-Cre (Nell-1Wnt1 KO) mice were generated for comprehensive analysis, while the relevant reporter mice were created for CNCC lineage tracing. Nell-1Wnt1 KO mice were born alive, but revealed significant frontonasal and mandibular bone defects with complete penetrance. Immunostaining demonstrated that the affected craniofacial bones exhibited decreased osteogenic and Wnt/β-catenin markers (Osteocalcin and active-β-catenin). Nell-1-deficient CNCCs demonstrated a significant reduction in cell proliferation and osteogenic differentiation. Active-β-catenin levels were significantly low in Nell-1-deficient CNCCs, but were rescued along with osteogenic capacity to a level close to that of wild-type (WT) cells via exogenous Nell-1 protein. Surprisingly, 5.4% of young adult Nell-1Wnt1 KO mice developed hydrocephalus with premature ossification of the intrasphenoidal synchondrosis and widened frontal, sagittal, and coronal sutures. Furthermore, the epithelial cells of the choroid plexus and ependymal cells exhibited degenerative changes with misplaced expression of their respective markers, transthyretin and vimentin, as well as dysregulated Pit-2 expression in hydrocephalic Nell-1Wnt1 KO mice. Nell-1Wnt1 KO embryos at E9.5, 14.5, 17.5, and newborn mice did not exhibit hydrocephalic phenotypes grossly and/or histologically. Collectively, Nell-1 is a pivotal modulator of CNCCs that is essential for normal development and growth of the cranial vault and base, and mandibles partially via activating the Wnt/β-catenin pathway. Nell-1 may also be critically involved in regulating cerebrospinal fluid homeostasis and in the pathogenesis of postnatal hydrocephalus.
Collapse
Affiliation(s)
- Xiaoyan Chen
- Department of Orthodontics, Affiliated Hospital of Stomatology, Medical College, Zhejiang University, Hangzhou, Zhejiang, PR China
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, CA, USA
| | - Huiming Wang
- Department of Oral Implantology, Affiliated Hospital of Stomatology, Medical College, Zhejiang University, Hangzhou, Zhejiang, PR China
| | - Mengliu Yu
- Department of Oral Implantology, Affiliated Hospital of Stomatology, Medical College, Zhejiang University, Hangzhou, Zhejiang, PR China
- Center of Stomatology, China-Japan Friendship Hospital, 2nd Yinghuayuan East Street, Chaoyang District, Beijing, PR China
| | - Jong Kil Kim
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, CA, USA
| | - Huichuan Qi
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, CA, USA
- Department of Orthodontics, School and Hospital of Stomatology, Jilin University, Changchun, Jilin, PR China
| | - Pin Ha
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, CA, USA
| | - Wenlu Jiang
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, CA, USA
| | - Eric Chen
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, CA, USA
| | - Xiangyou Luo
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, CA, USA
- Department of Cleft Lip and Palate Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, PR China
| | - Ryan Brent Needle
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, CA, USA
| | - Lloyd Baik
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, CA, USA
| | - Cathryn Yang
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, CA, USA
| | - Jiejun Shi
- Department of Orthodontics, Affiliated Hospital of Stomatology, Medical College, Zhejiang University, Hangzhou, Zhejiang, PR China
| | - Jin Hee Kwak
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, CA, USA
| | - Kang Ting
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, CA, USA
| | - Xinli Zhang
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, CA, USA.
| | - Chia Soo
- Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, UCLA and Orthopaedic Hospital, University of California, Los Angeles, CA, USA
- UCLA Division of Plastic and Reconstructive Surgery and Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, University of California, Los Angeles, CA, USA
| |
Collapse
|
209
|
Pisciotta A, Bertoni L, Vallarola A, Bertani G, Mecugni D, Carnevale G. Neural crest derived stem cells from dental pulp and tooth-associated stem cells for peripheral nerve regeneration. Neural Regen Res 2020; 15:373-381. [PMID: 31571644 PMCID: PMC6921350 DOI: 10.4103/1673-5374.266043] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Accepted: 05/11/2019] [Indexed: 12/15/2022] Open
Abstract
The peripheral nerve injuries, representing some of the most common types of traumatic lesions affecting the nervous system, are highly invalidating for the patients besides being a huge social burden. Although peripheral nervous system owns a higher regenerative capacity than does central nervous system, mostly depending on Schwann cells intervention in injury repair, several factors determine the extent of functional outcome after healing. Based on the injury type, different therapeutic approaches have been investigated so far. Nerve grafting and Schwann cell transplantation have represented the gold standard treatment for peripheral nerve injuries, however these approaches own limitations, such as scarce donor nerve availability and donor site morbidity. Cell based therapies might provide a suitable tool for peripheral nerve regeneration, in fact, the ability of different stem cell types to differentiate towards Schwann cells in combination with the use of different scaffolds have been widely investigated in animal models of peripheral nerve injuries in the last decade. Dental pulp is a promising cell source for regenerative medicine, because of the ease of isolation procedures, stem cell proliferation and multipotency abilities, which are due to the embryological origin from neural crest. In this article we review the literature concerning the application of tooth derived stem cell populations combined with different conduits to peripheral nerve injuries animal models, highlighting their regenerative contribution exerted through either glial differentiation and neuroprotective/neurotrophic effects on the host tissue.
Collapse
Affiliation(s)
- Alessandra Pisciotta
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with Interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Laura Bertoni
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with Interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Antonio Vallarola
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Giulia Bertani
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with Interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Daniela Mecugni
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with Interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
- Azienda USL - Institute and Health Care (IRCCS) di Reggio Emilia, Reggio Emilia, Italy
| | - Gianluca Carnevale
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with Interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
210
|
Adachi N, Bilio M, Baldini A, Kelly RG. Cardiopharyngeal mesoderm origins of musculoskeletal and connective tissues in the mammalian pharynx. Development 2020; 147:147/3/dev185256. [PMID: 32014863 DOI: 10.1242/dev.185256] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 12/16/2019] [Indexed: 12/14/2022]
Abstract
Cardiopharyngeal mesoderm (CPM) gives rise to muscles of the head and heart. Using genetic lineage analysis in mice, we show that CPM develops into a broad range of pharyngeal structures and cell types encompassing musculoskeletal and connective tissues. We demonstrate that CPM contributes to medial pharyngeal skeletal and connective tissues associated with both branchiomeric and somite-derived neck muscles. CPM and neural crest cells (NCC) make complementary mediolateral contributions to pharyngeal structures, in a distribution established in the early embryo. We further show that biallelic expression of the CPM regulatory gene Tbx1, haploinsufficient in 22q11.2 deletion syndrome patients, is required for the correct patterning of muscles with CPM-derived connective tissue. Our results suggest that CPM plays a patterning role during muscle development, similar to that of NCC during craniofacial myogenesis. The broad lineage contributions of CPM to pharyngeal structures provide new insights into congenital disorders and evolution of the mammalian pharynx.
Collapse
Affiliation(s)
- Noritaka Adachi
- Aix-Marseille Université, CNRS UMR 7288, IBDM, 13009 Marseille, France
| | - Marchesa Bilio
- CNR Institute of Genetics and Biophysics Adriano Buzzati-Traverso, Via Pietro Castellino 111, 80131 Naples, Italy
| | - Antonio Baldini
- CNR Institute of Genetics and Biophysics Adriano Buzzati-Traverso, Via Pietro Castellino 111, 80131 Naples, Italy.,Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples 80131, Italy
| | - Robert G Kelly
- Aix-Marseille Université, CNRS UMR 7288, IBDM, 13009 Marseille, France
| |
Collapse
|
211
|
Characterization of designed directional polylactic acid 3D scaffolds for neural differentiation of human dental pulp stem cells. J Formos Med Assoc 2020; 119:268-275. [DOI: 10.1016/j.jfma.2019.05.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 05/07/2019] [Accepted: 05/10/2019] [Indexed: 11/22/2022] Open
|
212
|
Smith AJ, Sharpe PT. Biological tooth replacement and repair. PRINCIPLES OF TISSUE ENGINEERING 2020:1187-1199. [DOI: 10.1016/b978-0-12-818422-6.00066-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
213
|
Hu Z, Deng N, Liu K, Zeng W. DLK mediates the neuronal intrinsic immune response and regulates glial reaction and neuropathic pain. Exp Neurol 2019; 322:113056. [DOI: 10.1016/j.expneurol.2019.113056] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 08/13/2019] [Accepted: 09/03/2019] [Indexed: 12/18/2022]
|
214
|
Okuhara S, Birjandi AA, Adel Al-Lami H, Sagai T, Amano T, Shiroishi T, Xavier GM, Liu KJ, Cobourne MT, Iseki S. Temporospatial sonic hedgehog signalling is essential for neural crest-dependent patterning of the intrinsic tongue musculature. Development 2019; 146:146/21/dev180075. [PMID: 31719045 DOI: 10.1242/dev.180075] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 08/17/2019] [Indexed: 01/20/2023]
Abstract
The tongue is a highly specialised muscular organ with a complex anatomy required for normal function. We have utilised multiple genetic approaches to investigate local temporospatial requirements for sonic hedgehog (SHH) signalling during tongue development. Mice lacking a Shh cis-enhancer, MFCS4 (ShhMFCS4/-), with reduced SHH in dorsal tongue epithelium have perturbed lingual septum tendon formation and disrupted intrinsic muscle patterning, with these defects reproduced following global Shh deletion from E10.5 in pCag-CreERTM; Shhflox/flox embryos. SHH responsiveness was diminished in local cranial neural crest cell (CNCC) populations in both mutants, with SHH targeting these cells through the primary cilium. CNCC-specific deletion of orofaciodigital syndrome 1 (Ofd1), which encodes a ciliary protein, in Wnt1-Cre; Ofdfl/Y mice led to a complete loss of normal myotube arrangement and hypoglossia. In contrast, mesoderm-specific deletion of Ofd1 in Mesp1-Cre; Ofdfl/Y embryos resulted in normal intrinsic muscle arrangement. Collectively, these findings suggest key temporospatial requirements for local SHH signalling in tongue development (specifically, lingual tendon differentiation and intrinsic muscle patterning through signalling to CNCCs) and provide further mechanistic insight into the tongue anomalies seen in patients with disrupted hedgehog signalling.
Collapse
Affiliation(s)
- Shigeru Okuhara
- Section of Molecular Craniofacial Embryology, Graduate School of Dental and Medical Sciences, Tokyo Medical and Dental University (TMDU), Tokyo 113-8510, Japan
| | - Anahid A Birjandi
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London SE1 9RT, UK
| | - Hadeel Adel Al-Lami
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London SE1 9RT, UK
| | - Tomoko Sagai
- Mammalian Genetics Laboratory, National Institute of Genetics, Mishima 411-8540, Japan
| | - Takanori Amano
- Mammalian Genetics Laboratory, National Institute of Genetics, Mishima 411-8540, Japan
| | - Toshihiko Shiroishi
- Mammalian Genetics Laboratory, National Institute of Genetics, Mishima 411-8540, Japan
| | - Guilherme M Xavier
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London SE1 9RT, UK
| | - Karen J Liu
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London SE1 9RT, UK
| | - Martyn T Cobourne
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London SE1 9RT, UK
| | - Sachiko Iseki
- Section of Molecular Craniofacial Embryology, Graduate School of Dental and Medical Sciences, Tokyo Medical and Dental University (TMDU), Tokyo 113-8510, Japan
| |
Collapse
|
215
|
Zhao Y, Yuan X, Bellido T, Helms JA. A Correlation between Wnt/Beta-catenin Signaling and the Rate of Dentin Secretion. J Endod 2019; 45:1357-1364.e1. [PMID: 31522810 PMCID: PMC10900857 DOI: 10.1016/j.joen.2019.07.014] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 07/21/2019] [Accepted: 07/26/2019] [Indexed: 01/28/2023]
Abstract
INTRODUCTION Odontoblasts produce dentin throughout life and in response to trauma. The purpose of this study was to identify the roles of endogenous Wnt signaling in regulating the rate of dentin accumulation. METHODS Histology, immunohistochemistry, vital dye labeling, and histomorphometric assays were used to quantify the rate of dentin accumulation as a function of age. Two strains of Wnt reporter mice were used to identify and follow the distribution and number of Wnt-responsive odontoblasts as a function of age. To show a causal relationship between dentin secretion and Wnt signaling, dentin accumulation was monitored in a strain of mice in which Wnt signaling was aberrantly elevated. RESULTS Dentin deposition occurs throughout life, but the rate of accumulation slows with age. This decline in dentin secretion correlates with a decrease in endogenous Wnt signaling. In a genetically modified strain of mice, instead of tubular dentin, aberrantly elevated Wnt signaling resulted in accumulation of reparative dentin or osteodentin secreted from predontoblasts. CONCLUSIONS Wnt signaling regulates dentin secretion by odontoblasts, and the formation of reparative or osteodentin is the direct consequence of elevated Wnt signaling. These preclinical data have therapeutic implications for the development of a biologically based pulp capping medicant.
Collapse
Affiliation(s)
- Yuan Zhao
- Department of Cariology and Endodontology, School of Dentistry, Lanzhou University, Lanzhou, China; Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford School of Medicine, Stanford, California
| | - Xue Yuan
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford School of Medicine, Stanford, California
| | - Teresita Bellido
- Departments of Anatomy and Cell Biology and Medicine, Division of Endocrinology, Indiana University School of Medicine and Roudebush Veterans Administration Medical Center, Indianapolis, Indiana
| | - Jill A Helms
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford School of Medicine, Stanford, California.
| |
Collapse
|
216
|
Li X, Zheng Y, Hou L, Zhou Z, Huang Y, Zhang Y, Jia L, Li W. Exosomes derived from maxillary BMSCs enhanced the osteogenesis in iliac BMSCs. Oral Dis 2019; 26:131-144. [PMID: 31541596 DOI: 10.1111/odi.13202] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 07/16/2019] [Accepted: 08/19/2019] [Indexed: 12/14/2022]
Affiliation(s)
- Xiaobei Li
- Department of Orthodontics Peking University School and Hospital of Stomatology Beijing China
| | - Yunfei Zheng
- Department of Orthodontics Peking University School and Hospital of Stomatology Beijing China
| | - Liyu Hou
- Department of Stomatology Shenzhen People’s Hospital Shenzhen China
| | - Zhibo Zhou
- Department of Oral and Maxillofacial Surgery Peking University School and Hospital of Stomatology Beijing China
| | - Yiping Huang
- Department of Orthodontics Peking University School and Hospital of Stomatology Beijing China
| | - Yixin Zhang
- Department of Orthodontics Peking University School and Hospital of Stomatology Beijing China
| | - Lingfei Jia
- Department of Oral and Maxillofacial Surgery Peking University School and Hospital of Stomatology Beijing China
- Central Laboratory Peking University School and Hospital of Stomatology Beijing China
| | - Weiran Li
- Department of Orthodontics Peking University School and Hospital of Stomatology Beijing China
| |
Collapse
|
217
|
Marín-Llera JC, Garciadiego-Cázares D, Chimal-Monroy J. Understanding the Cellular and Molecular Mechanisms That Control Early Cell Fate Decisions During Appendicular Skeletogenesis. Front Genet 2019; 10:977. [PMID: 31681419 PMCID: PMC6797607 DOI: 10.3389/fgene.2019.00977] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 09/13/2019] [Indexed: 12/02/2022] Open
Abstract
The formation of the vertebrate skeleton is orchestrated in time and space by a number of gene regulatory networks that specify and position all skeletal tissues. During embryonic development, bones have two distinct origins: bone tissue differentiates directly from mesenchymal progenitors, whereas most long bones arise from cartilaginous templates through a process known as endochondral ossification. Before endochondral bone development takes place, chondrocytes form a cartilage analgen that will be sequentially segmented to form joints; thus, in the cartilage template, either the cartilage maturation programme or the joint formation programme is activated. Once the cartilage differentiation programme starts, the growth plate begins to form. In contrast, when the joint formation programme is activated, a capsule begins to form that contains special articular cartilage and synovium to generate a functional joint. In this review, we will discuss the mechanisms controlling the earliest molecular events that regulate cell fate during skeletogenesis in long bones. We will explore the initial processes that lead to the recruitment of mesenchymal stem/progenitor cells, the commitment of chondrocyte lineages, and the formation of skeletal elements during morphogenesis. Thereafter, we will review the process of joint specification and joint morphogenesis. We will discuss the links between transcription factor activity, cell–cell interactions, cell–extracellular matrix interactions, growth factor signalling, and other molecular interactions that control mesenchymal stem/progenitor cell fate during embryonic skeletogenesis.
Collapse
Affiliation(s)
- Jessica Cristina Marín-Llera
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico City, Mexico
| | | | - Jesús Chimal-Monroy
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico City, Mexico
| |
Collapse
|
218
|
Partially Penetrant Cardiac Neural Crest Defects in Hand1 Phosphomutant Mice: Dimer Choice That Is Not So Critical. Pediatr Cardiol 2019; 40:1339-1344. [PMID: 31338559 PMCID: PMC6786956 DOI: 10.1007/s00246-019-02162-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 07/16/2019] [Indexed: 10/26/2022]
Abstract
Hand1 is a basic Helix-loop-Helix transcription factor that exhibits post-translationally regulated dimer partner choice that allows for a diverse set of Hand1 transcriptional complexes. Indeed, when Hand1 phosphoregulation is altered, conditionally activated hypophorylation (Hand1PO4-) and phosphorylation mimic (Hand1PO4+) Hand1 alleles disrupt both craniofacial and limb morphogenesis with 100% penetrance. Interestingly, activation of conditional Hand1 Phosphomutant alleles within post-migratory neural crest cells produce heart defects that include ventricular septal defects, double-outlet right ventricle, persistent truncus arteriosus with partial penetrance. Single versus double-lobed thymus is a distinguishing feature between Wnt1-Cre;Hand1PO4-/+ and Wnt1-Cre;Hand1PO4+/+ mice. These data show that although Hand1 dimer regulation plays critical and consistent roles in disrupting craniofacial and limb morphogenesis, Hand1 dimer regulation during cardiac outflow track formation is less critical for normal morphogenesis. This review will present the OFT phenotypes observed in Hand1 Phosphomutant mice, and discuss possible mechanisms of how penetrance differences within the same tissues within the same embryos could be variable.
Collapse
|
219
|
Abstract
Los odontoblastos son células post-mitóticas de origen mesenquimal dispuestas en forma de palizada en la periferia de la pulpa dental y responsables de la formación de la dentina. Los odontoblastos derivan de la cresta neural y su diferenciación es la consecuencia de las interacciones epitelio-mesénquima entre las células de la papila dental y el epitelio dental interno. Este trabajo tiene como objetivo revisar los aspectos fisiológicos y patológicos de los odontoblastos, comprendiendo su origen, mecanismos de diferenciación y propiedades funcionales. Se realizó una búsqueda electrónica de literatura desde el año 2000 hasta Febrero de 2018, seleccionando 2889 artículos, de los cuales 52 artículos fueron analizados y discutidos. Los resultados exponen el origen, etapas y los factores relacionados con la diferenciación odontoblástica, junto con los aspectos principales de la organización estructural y funciones que desempeñan los odontoblastos. Esta revisión demuestra mediante la evidencia científica actual como los estudios concernientes a los odontoblastos se focalizan en comprender los mecanismos en la formación de la dentina reparativa, la respuesta inmunitaria y su rol en los procesos de inflamación y dolor. Trabajos futuros deberán esclarecer las diferentes señales involucradas en los procesos fisiopatológicos celulares y moleculares llevados a cabo por los odontoblastos.
Collapse
|
220
|
Human Amniotic Membrane as a Matrix for Endothelial Differentiation of VEGF-Treated Dental Stem Cells. Cell Mol Bioeng 2019. [DOI: 10.1007/s12195-019-00596-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
|
221
|
Sun Z, Yu S, Chen S, Liu H, Chen Z. SP1 regulates KLF4 via SP1 binding motif governed by DNA methylation during odontoblastic differentiation of human dental pulp cells. J Cell Biochem 2019; 120:14688-14699. [PMID: 31009133 PMCID: PMC8895433 DOI: 10.1002/jcb.28730] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Revised: 03/14/2019] [Accepted: 04/01/2019] [Indexed: 12/16/2022]
Abstract
OBJECTIVE DNA methylation is a critical epigenetic modulation in regulating gene expression in cell differentiation process, however, its detailed molecular mechanism during odontoblastic differentiation remains elusive. We aimed to study the global effect of DNA methylation on odontoblastic differentiation and how DNA methylation affects the transactivation of transcription factor (TF) on its target gene. METHODS DNA methyltransferase (DNMTs) inhibition assay and following odontoblastic differentiation assay were performed to evaluate the effect of DNA methylation inhibition on odontoblastic differentiation. Promoter DNA methylation microarray and motif enrichment assay were performed to predict the most DNA-methylation-affected TF motifs during odontoblastic differentiation. The enriched target sites and motifs were further analyzed by methylation-specific polymerase chain reaction (MS-PCR) and sequencing. The functional target sites were validated in vitro with Luciferase assay. The regulatory effect of DNA methylation on the enriched target sites in primary human dental pulp cells and motifs were confirmed by in vitro methylation assay. RESULTS Inhibition of DNMTs in preodontoblast cells increased the expression level of Klf4 as well as marker genes of odontoblastic differentiation including Dmp1 and Dspp, and enhanced the efficiency of odontoblastic differentiation. SP1/KLF4 binding motifs were found to be highly enriched in the promoter regions and showed demethylation during odontoblastic differentiation. Mutation of SP1 binding site at -75 within KLF4's promoter region significantly decreased the luciferase activity. The in vitro methylation of KLF4's promoter decreased the transactivation of SP1 on KLF4. CONCLUSION We confirmed that SP1 regulates KLF4 through binding site lying in a CpG island in KLF4's promoter region which demethylated during odontoblastic differentiation thus enhancing the efficiency of SP1's binding and transcriptional regulation on KLF4.
Collapse
Affiliation(s)
- Zheyi Sun
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory for Oral Biomedicine of Ministry of Education (KLOBM), School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Shuaitong Yu
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory for Oral Biomedicine of Ministry of Education (KLOBM), School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Shuo Chen
- Department of Developmental Dentistry, University of Texas Health Science Center, San Antonio, Texas
| | - Huan Liu
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory for Oral Biomedicine of Ministry of Education (KLOBM), School and Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Periodontology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Zhi Chen
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory for Oral Biomedicine of Ministry of Education (KLOBM), School and Hospital of Stomatology, Wuhan University, Wuhan, China
| |
Collapse
|
222
|
Tang W, Martik ML, Li Y, Bronner ME. Cardiac neural crest contributes to cardiomyocytes in amniotes and heart regeneration in zebrafish. eLife 2019; 8:47929. [PMID: 31393264 PMCID: PMC6721792 DOI: 10.7554/elife.47929] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 08/08/2019] [Indexed: 12/30/2022] Open
Abstract
Cardiac neural crest cells contribute to important portions of the cardiovascular system including the aorticopulmonary septum and cardiac ganglion. Using replication incompetent avian retroviruses for precise high-resolution lineage analysis, we uncover a previously undescribed neural crest contribution to cardiomyocytes of the ventricles in Gallus gallus, supported by Wnt1-Cre lineage analysis in Mus musculus. To test the intriguing possibility that neural crest cells contribute to heart repair, we examined Danio rerio adult heart regeneration in the neural crest transgenic line, Tg(−4.9sox10:eGFP). Whereas the adult heart has few sox10+ cells in the apex, sox10 and other neural crest regulatory network genes are upregulated in the regenerating myocardium after resection. The results suggest that neural crest cells contribute to many cardiovascular structures including cardiomyocytes across vertebrates and to the regenerating heart of teleost fish. Thus, understanding molecular mechanisms that control the normal development of the neural crest into cardiomyocytes and reactivation of the neural crest program upon regeneration may open potential therapeutic approaches to repair heart damage in amniotes. Before birth, unspecialized stem cells go through a process called differentiation to form the many types of cells found in the adult. Neural crest cells are a group of these stem cells found in all animals with backbones (i.e. vertebrates) including humans. These cells migrate extensively during development to form many different parts of the body. Due to their contributions to diverse organs and tissues, neural crest cells are very important for healthy development. The heart ventricle is one of the tissues to which neural crest cells contribute during embryonic development in fish and amphibians. However, it was unclear whether this is also the case for birds or mammals or whether neural crest cells have any roles in the regeneration of the adult heart after injury in fish and amphibians. To address these questions, Tang, Martik et al. used cell biology techniques to track neural crest cells in living animals. The experiments revealed that neural crest cells contribute to heart tissue in developing birds and mammals and help repair the heart in adult zebrafish. Further results showed that the contribution of neural crest cells to the heart is controlled by the same genes during both the growth of the embryonic heart and the repair of the adult heart. These results provide new insights into the repair and healing of damaged heart muscle in fish. They also show that similar processes could exist in mammals, including humans, suggesting that activating neural crest cells in the heart could treat damage caused by heart attacks and related conditions.
Collapse
Affiliation(s)
- Weiyi Tang
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, United States
| | - Megan L Martik
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, United States
| | - Yuwei Li
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, United States
| | - Marianne E Bronner
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, United States
| |
Collapse
|
223
|
Tao H, Lin H, Sun Z, Pei F, Zhang J, Chen S, Liu H, Chen Z. Klf4 Promotes Dentinogenesis and Odontoblastic Differentiation via Modulation of TGF-β Signaling Pathway and Interaction With Histone Acetylation. J Bone Miner Res 2019; 34:1502-1516. [PMID: 31112333 PMCID: PMC8895434 DOI: 10.1002/jbmr.3716] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Revised: 03/04/2019] [Accepted: 03/05/2019] [Indexed: 12/14/2022]
Abstract
Transcription factors bind to cell-specific cis-regulatory elements, such as enhancers and promoters, to initiate much of the gene expression program of different biological process. Odontoblast differentiation is a necessary step for tooth formation and is also governed by a complex gene regulatory network. Our previous in vitro experiments showed that Krüppel-like factor 4 (KLF4) can promote odontoblastic differentiation of both mouse dental papillary cells (mDPCs) and human dental pulp cells; however, its mechanism remains unclear. We first used Wnt1-Cre; KLF4fx/fx (Klf4 cKO) mice to examine the role of KLF4 during odontoblast differentiation in vivo and demonstrated significantly impaired dentin mineralization and enlarged pulp/root canals. Additionally, combinatory analysis using RNA-seq and ATAC-seq revealed genomewide direct regulatory targets of KLF4 in mouse odontoblasts. We found that KLF4 can directly activate the TGF-β signaling pathway at the beginning of odontoblast differentiation with Runx2 as a cofactor. Furthermore, we found that KLF4 can directly upregulate the expression levels of Dmp1 and Sp7, which are markers of odontoblastic differentiation, through binding to their promoters. Interestingly, as a transcription factor, KLF4 can also recruit histone acetylase as a regulatory companion to the downstream target genes to positively or negatively regulate transcription. To further investigate other regulatory companions of KLF4, we chose histone acetylase HDAC3 and P300. Immunoprecipitation demonstrated that KLF4 interacted with P300 and HDAC3. Next, ChIP analysis detected P300 and HDAC3 enrichment on the promoter region of KLF4 target genes Dmp1 and Sp7. HDAC3 mainly interacted with KLF4 on day 0 of odontoblastic induction, whereas P300 interacted on day 7 of induction. These temporal-specific interactions regulated Dmp1 and Sp7 transcription, thus regulating dentinogenesis. Taken together, these results demonstrated that KLF4 regulates Dmp1 and Sp7 transcription via the modulation of histone acetylation and is vital to dentinogenesis. © 2019 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Huangheng Tao
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory for Oral Biomedicine of Ministry of Education (KLOBM), School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Heng Lin
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory for Oral Biomedicine of Ministry of Education (KLOBM), School and Hospital of Stomatology, Wuhan University, Wuhan, China.,Department of Anesthesiology, University of Rochester, Rochester, NY, USA
| | - Zheyi Sun
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory for Oral Biomedicine of Ministry of Education (KLOBM), School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Fei Pei
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory for Oral Biomedicine of Ministry of Education (KLOBM), School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Jie Zhang
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory for Oral Biomedicine of Ministry of Education (KLOBM), School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Shuo Chen
- Department of Developmental Dentistry, University of Texas Health Science Center, San Antonio, TX, USA
| | - Huan Liu
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory for Oral Biomedicine of Ministry of Education (KLOBM), School and Hospital of Stomatology, Wuhan University, Wuhan, China.,Department of Periodontology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Zhi Chen
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory for Oral Biomedicine of Ministry of Education (KLOBM), School and Hospital of Stomatology, Wuhan University, Wuhan, China
| |
Collapse
|
224
|
Stage-dependent differential gene expression profiles of cranial neural crest-like cells derived from mouse-induced pluripotent stem cells. Med Mol Morphol 2019; 53:28-41. [PMID: 31297611 PMCID: PMC7033077 DOI: 10.1007/s00795-019-00229-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 06/26/2019] [Indexed: 12/13/2022]
Abstract
Cranial neural crest cells are multipotent cells that migrate into the pharyngeal arches of the vertebrate embryo and differentiate into various craniofacial organ derivatives. Therefore, migrating cranial neural crest cells are considered one of the most attractive candidate cell sources in regenerative medicine. We generated cranial neural crest like cell (cNCCs) using mouse-induced pluripotent stem cells cultured in neural crest-inducing medium for 14 days. Subsequently, we conducted RNA sequencing experiments to analyze gene expression profiles of cNCCs at different time points after induction. cNCCs expressed several neural crest specifier genes; however, some previously reported specifier genes such as paired box 3 and Forkhead box D3, which are essential for embryonic neural crest development, were not expressed. Moreover, ETS proto-oncogene 1, transcription factor and sex-determining region Y-box 10 were only expressed after 14 days of induction. Finally, cNCCs expressed multiple protocadherins and a disintegrin and metalloproteinase with thrombospondin motifs enzymes, which may be crucial for their migration.
Collapse
|
225
|
Chen Q, Zhao Y, Qian Y, Lu C, Shen G, Dai J. A genetic-phenotypic classification for syndromic micrognathia. J Hum Genet 2019; 64:875-883. [PMID: 31273320 DOI: 10.1038/s10038-019-0630-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 05/27/2019] [Accepted: 06/04/2019] [Indexed: 11/09/2022]
Abstract
Micrognathia is a common craniofacial deformity which represents hypoplastic development of the mandible, accompanied by retrognathia and consequent airway problems. Usually, micrognathia is accompanied by multiple systematic defects, known as syndromic micrognathia, and is in close association with genetic factors. Now, large quantities of pathogenic genes of syndromic micrognathia have been revealed. However, how these different pathogenic genes could lead to similar phenotypes, and whether there are some common characteristics among these pathogenic genes are still unknown. In this study, we proposed a genetic-phenotypic classification of syndromic micrognathia based on pathogenic genes information obtained from Phenolyzer, DAVID, OMIM, and PubMed database. Pathogenic genes of syndromic micrognathia could be divided into four groups based on gene function, including cellular processes and structures, cell metabolism, cartilage and bone development, and neuromuscular function. In addition, these four groups exhibited various clinical characteristics, and the affected systems, such as central nervous system, skeletal system, cardiovascular system, oral and dental system, respiratory system and muscle, were different in these four groups. This classification could provide meaningful insights into the pathogenesis of syndromic micrognathia, and offer some clues for understanding the molecular mechanism, as well as guiding precise clinical diagnosis and treatment for syndromic micrognathia.
Collapse
Affiliation(s)
- Qiming Chen
- Department of Oral & Cranio-maxillofacial Surgery, Shanghai Key laboratory of stomatology, Shanghai ninth People's Hospital, Shanghai JiaoTong University, School of Medicine, Shanghai, 200011, P.R. China
| | - Yan Zhao
- Department of Oral & Cranio-maxillofacial Surgery, Shanghai Key laboratory of stomatology, Shanghai ninth People's Hospital, Shanghai JiaoTong University, School of Medicine, Shanghai, 200011, P.R. China
| | - Yifeng Qian
- Department of Oral & Cranio-maxillofacial Surgery, Shanghai Key laboratory of stomatology, Shanghai ninth People's Hospital, Shanghai JiaoTong University, School of Medicine, Shanghai, 200011, P.R. China
| | - Chenpei Lu
- Department of Oral & Cranio-maxillofacial Surgery, Shanghai Key laboratory of stomatology, Shanghai ninth People's Hospital, Shanghai JiaoTong University, School of Medicine, Shanghai, 200011, P.R. China
| | - Guofang Shen
- Department of Oral & Cranio-maxillofacial Surgery, Shanghai Key laboratory of stomatology, Shanghai ninth People's Hospital, Shanghai JiaoTong University, School of Medicine, Shanghai, 200011, P.R. China.
| | - Jiewen Dai
- Department of Oral & Cranio-maxillofacial Surgery, Shanghai Key laboratory of stomatology, Shanghai ninth People's Hospital, Shanghai JiaoTong University, School of Medicine, Shanghai, 200011, P.R. China.
| |
Collapse
|
226
|
Cibi DM, Mia MM, Guna Shekeran S, Yun LS, Sandireddy R, Gupta P, Hota M, Sun L, Ghosh S, Singh MK. Neural crest-specific deletion of Rbfox2 in mice leads to craniofacial abnormalities including cleft palate. eLife 2019; 8:45418. [PMID: 31241461 PMCID: PMC6663295 DOI: 10.7554/elife.45418] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 06/25/2019] [Indexed: 12/17/2022] Open
Abstract
Alternative splicing (AS) creates proteomic diversity from a limited size genome by generating numerous transcripts from a single protein-coding gene. Tissue-specific regulators of AS are essential components of the gene regulatory network, required for normal cellular function, tissue patterning, and embryonic development. However, their cell-autonomous function in neural crest development has not been explored. Here, we demonstrate that splicing factor Rbfox2 is expressed in the neural crest cells (NCCs), and deletion of Rbfox2 in NCCs leads to cleft palate and defects in craniofacial bone development. RNA-Seq analysis revealed that Rbfox2 regulates splicing and expression of numerous genes essential for neural crest/craniofacial development. We demonstrate that Rbfox2-TGF-β-Tak1 signaling axis is deregulated by Rbfox2 deletion. Furthermore, restoration of TGF-β signaling by Tak1 overexpression can rescue the proliferation defect seen in Rbfox2 mutants. We also identified a positive feedback loop in which TGF-β signaling promotes expression of Rbfox2 in NCCs. Abnormalities affecting the head and face – such as a cleft lip or palate – are among the most common of all birth defects. These tissues normally develop from cells in the embryo known as the neural crest cells, and specifically a subset of these cells called the cranial neural crest cells. Most cases of cleft lip or palate are linked back to genes that affect the biology of this group of cells. The list of genes implicated in the impaired development of cranial neural crest cells code for proteins with a wide range of different activities. Some encode transcription factors – proteins that switch genes on or off. Others code for chromatin remodeling factors, which control how the DNA is packed inside cells. However, the role of another group of proteins – the splicing factors – remains unclear and warrants further investigation. When a gene is switched on its genetic code is first copied into a short-lived molecule called a transcript. These transcripts are then edited to form templates to build proteins. Splicing is one way that a transcript can be edited, which involves different pieces of the transcript being cut out and the remaining pieces being pasted together to form alternative versions of the final template. Splicing factors control this process. Cibi et al. now show that neural crest cells from mice make a splicing factor called Rbfox2 and that deleting this gene for this protein from only these cells leads to mice with a cleft palate and defects in the bones of their head and face. Further analysis helped to identify the transcripts that are spliced by Rbfox2, and the effects that these splicing events have on gene activity in mouse tissues that develop from cranial neural crest cells. Cibi et al. went on to find a signaling pathway that was impaired in the mutant cells that lacked Rbfox2. Forcing the mutant cells to over-produce one of the proteins involved in this signaling pathway (a protein named Tak1) was enough to compensate for the some of the defects caused by a lack of Rbfox2, suggesting it acts downstream of the splicing regulator. Lastly, Cibi et al. showed that another protein in this signaling pathway, called TGF-β, acted to increase how much Rbfox2 was made by neural crest cells. Together these findings may be relevant in human disease studies, given that altered TGF-β signaling is a common feature in many birth defects seen in humans.
Collapse
Affiliation(s)
- Dasan Mary Cibi
- Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore, Singapore
| | - Masum M Mia
- Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore, Singapore
| | - Shamini Guna Shekeran
- Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore, Singapore
| | - Lim Sze Yun
- National Heart Research Institute, National Heart Center, Singapore, Singapore
| | - Reddemma Sandireddy
- Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore, Singapore
| | - Priyanka Gupta
- Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore, Singapore
| | - Monalisa Hota
- Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore, Singapore
| | - Lei Sun
- Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore, Singapore
| | - Sujoy Ghosh
- Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore, Singapore
| | - Manvendra K Singh
- Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore, Singapore.,National Heart Research Institute, National Heart Center, Singapore, Singapore
| |
Collapse
|
227
|
Lukacs M, Roberts T, Chatuverdi P, Stottmann RW. Glycosylphosphatidylinositol biosynthesis and remodeling are required for neural tube closure, heart development, and cranial neural crest cell survival. eLife 2019; 8:45248. [PMID: 31232685 PMCID: PMC6611694 DOI: 10.7554/elife.45248] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 06/05/2019] [Indexed: 01/10/2023] Open
Abstract
Glycosylphosphatidylinositol (GPI) anchors attach nearly 150 proteins to the cell membrane. Patients with pathogenic variants in GPI biosynthesis genes develop diverse phenotypes including seizures, dysmorphic facial features and cleft palate through an unknown mechanism. We identified a novel mouse mutant (cleft lip/palate, edema and exencephaly; Clpex) with a hypo-morphic mutation in Post-Glycophosphatidylinositol Attachment to Proteins-2 (Pgap2), a component of the GPI biosynthesis pathway. The Clpex mutation decreases surface GPI expression. Surprisingly, Pgap2 showed tissue-specific expression with enrichment in the brain and face. We found the Clpex phenotype is due to apoptosis of neural crest cells (NCCs) and the cranial neuroepithelium. We showed folinic acid supplementation in utero can partially rescue the cleft lip phenotype. Finally, we generated a novel mouse model of NCC-specific total GPI deficiency. These mutants developed median cleft lip and palate demonstrating a previously undocumented cell autonomous role for GPI biosynthesis in NCC development.
Collapse
Affiliation(s)
- Marshall Lukacs
- Division of Human Genetics, Cincinnati Children's Medical Center, Cincinnati, United States.,Medical Scientist Training Program, Cincinnati Children's Medical Center, Cincinnati, United States
| | - Tia Roberts
- Division of Human Genetics, Cincinnati Children's Medical Center, Cincinnati, United States
| | - Praneet Chatuverdi
- Division of Developmental Biology, Cincinnati Children's Medical Center, Cincinnati, United States
| | - Rolf W Stottmann
- Division of Human Genetics, Cincinnati Children's Medical Center, Cincinnati, United States.,Medical Scientist Training Program, Cincinnati Children's Medical Center, Cincinnati, United States.,Division of Developmental Biology, Cincinnati Children's Medical Center, Cincinnati, United States.,Department of Pediatrics, University of Cincinnati, Cincinnati, United States
| |
Collapse
|
228
|
Fu M, Xu L, Chen X, Han W, Ruan C, Li J, Cai C, Ye M, Gao P. Neural Crest Cells Differentiate Into Brown Adipocytes and Contribute to Periaortic Arch Adipose Tissue Formation. Arterioscler Thromb Vasc Biol 2019; 39:1629-1644. [PMID: 31189430 DOI: 10.1161/atvbaha.119.312838] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
OBJECTIVE Periaortic arch adipose tissue (PAAT) plays critical roles in regulating vascular homeostasis; however, its anatomic features, developmental processes, and origins remain unclear. Approach and Results: Anatomic analysis and genetic lineage tracing of Wnt1 (wingless-type MMTV [mouse mammary tumor virus] integration site family member 1)-Cre+;Rosa26RFP/+ mice, Myf5 (myogenic factor 5)-Cre+;Rosa26RFP/+ mice, and SM22α-Cre+;Rosa26RFP/+ mice are performed, and the results show that PAAT has unique anatomic features, and the developmental processes of PAAT are independent of the others periaortic adipose tissues. PAAT adipocytes are mainly derived from neural crest cells (NCCs) rather than from Myf5+ progenitors. Most PAAT adipocyte progenitors expressed SM22α+ (smooth muscle protein 22-alpha) during development. Using Wnt1-Cre+;PPARγflox/flox mice, we found that knockout of PPAR (peroxisome proliferator-activated receptor)-γ in NCCs results in PAAT developmental delay and dysplasia, further confirming that NCCs contribute to PAAT formation. And we further indicated PAAT dysplasia aggravates Ang II (angiotensin II)-induced inflammation and remodeling of the common carotid artery close to aorta arch. We also found that NCCs can be differentiated into both brown and white adipocytes in vivo and in vitro. RNA sequencing results suggested NCC-derived adipose tissue displays a distinct transcriptional profile compared with the non-NCC-derived adipose tissue in PAAT. CONCLUSIONS PAAT has distinctive anatomic features and developmental processes. Most PAAT adipocytes are originated from NCCs which derive from ectoderm. NCCs are progenitors not only of white adipocytes but also of brown adipocytes. This study indicates that the PAAT is derived from multiple cell lineages, the adipocytes derived from different origins have distinct transcriptional profiles, and PAAT plays a critical role in Ang II-induced common carotid artery inflammation and remodeling.Visual OvervieW: An online visual overview is available for this article.
Collapse
Affiliation(s)
- Mengxia Fu
- From the State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Department of Hypertension, Ruijin Hospital and Shanghai Institute of Hypertension (M.F., L.X., X.C., W.H., C.R., M.Y., P.G.), Shanghai Jiao Tong University School of Medicine, China
| | - Lian Xu
- From the State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Department of Hypertension, Ruijin Hospital and Shanghai Institute of Hypertension (M.F., L.X., X.C., W.H., C.R., M.Y., P.G.), Shanghai Jiao Tong University School of Medicine, China
| | - Xiaohui Chen
- From the State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Department of Hypertension, Ruijin Hospital and Shanghai Institute of Hypertension (M.F., L.X., X.C., W.H., C.R., M.Y., P.G.), Shanghai Jiao Tong University School of Medicine, China
| | - Weiqing Han
- From the State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Department of Hypertension, Ruijin Hospital and Shanghai Institute of Hypertension (M.F., L.X., X.C., W.H., C.R., M.Y., P.G.), Shanghai Jiao Tong University School of Medicine, China
| | - Chengchao Ruan
- From the State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Department of Hypertension, Ruijin Hospital and Shanghai Institute of Hypertension (M.F., L.X., X.C., W.H., C.R., M.Y., P.G.), Shanghai Jiao Tong University School of Medicine, China
| | - Jun Li
- Department of Cardiology, Shanghai General Hospital (J.L.), Shanghai Jiao Tong University School of Medicine, China
| | - Chenleng Cai
- Riley Heart Research Center, and Herman Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis (C.C.)
| | - Maoqing Ye
- From the State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Department of Hypertension, Ruijin Hospital and Shanghai Institute of Hypertension (M.F., L.X., X.C., W.H., C.R., M.Y., P.G.), Shanghai Jiao Tong University School of Medicine, China
| | - Pingjin Gao
- From the State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Department of Hypertension, Ruijin Hospital and Shanghai Institute of Hypertension (M.F., L.X., X.C., W.H., C.R., M.Y., P.G.), Shanghai Jiao Tong University School of Medicine, China
| |
Collapse
|
229
|
Guo W, Fan Z, Wang S, Du J. ALK5 is essential for tooth germ differentiation during tooth development. Biotech Histochem 2019; 94:481-490. [PMID: 31144525 DOI: 10.1080/10520295.2018.1552018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The TGFβ superfamily of proteins participates in tooth development. TGFβ1 and TGFβ3 regulate odontoblast differentiation and dentin extracellular matrix synthesis. Although the expression of TGFβ family member ligands is well-characterized during mammalian tooth development, less is known about the TGFβ receptor, which is a heteromeric complex consisting of a type I and type II receptors. The molecular mechanism of ALK5 (TGFβR1) in the dental mesenchyme is not clear. We investigated the role of ALK5 in tooth germ mesenchymal cells (TGMCs) from the lower first molar tooth germs of day 15.5 embryonic mice. Human recombinant TGFβ3 protein or an ALK5 inhibitor (SD208) was added to the cells. Cell proliferation was inhibited by SD208 and promoted by TGFβ3. We found that SD208 inhibited TGMCs osteogenesis and dentinogenesis. Both canonical and noncanonical TGFβ signaling pathways participated in the process. TAK1, P-TAK1, p38 and P-p38 showed greater expression and SMAD4 showed less expression when ALK5 was inhibited. Our findings contribute to understanding the role of TGFβ signaling for the differentiation of mesenchymal stem cells derived from dental germ and suggest possible targets for optimizing the use of stem cells of dental origin for tissue regeneration.
Collapse
Affiliation(s)
- W Guo
- Laboratory of Molecular Signaling and Stem Cells Therapy, Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology , Beijing , China
| | - Z Fan
- Laboratory of Molecular Signaling and Stem Cells Therapy, Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology , Beijing , China
| | - S Wang
- Laboratory of Molecular Signaling and Stem Cells Therapy, Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology , Beijing , China.,Department of Biochemistry and Molecular Biology, Capital Medical University School of Basic Medical Sciences , Beijing , China
| | - J Du
- Laboratory of Molecular Signaling and Stem Cells Therapy, Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology , Beijing , China
| |
Collapse
|
230
|
Nakajima T, Ikeya M. Insights into the biology of fibrodysplasia ossificans progressiva using patient-derived induced pluripotent stem cells. Regen Ther 2019; 11:25-30. [PMID: 31193176 PMCID: PMC6517845 DOI: 10.1016/j.reth.2019.04.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 03/18/2019] [Accepted: 04/05/2019] [Indexed: 12/12/2022] Open
Abstract
The demand for development of new drugs remains on the upward trend because of the large number of patients suffering from intractable diseases for which effective treatment has not been established yet. Recently, several researchers have attempted to apply induced pluripotent stem cell (iPSC) technology as a powerful tool for studying the mechanisms underlying the onset of various diseases and for new drug screening. This technology has made an enormous breakthrough, since it permits us to recapitulate the disease phenotype in vitro, outside of the patient's body. Here, we discuss the latest findings that uncovered a mechanism underlying the pathology of a rare genetic musculoskeletal disease, fibrodysplasia ossificans progressiva (FOP), by modeling the phenotypes with FOP patient-derived iPSCs, and that discovered promising candidate drugs for FOP treatment. We also discussed future directions of FOP research.
Collapse
Affiliation(s)
- Taiki Nakajima
- Department of Life Science Frontiers, Center for iPS Cell Research and Application, Kyoto University, Kyoto, 606-8507, Japan
| | - Makoto Ikeya
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto, 606-8507, Japan
| |
Collapse
|
231
|
Sugiyama K, Nagashima K, Miwa T, Shimizu Y, Kawaguchi T, Iida K, Tamaoki N, Hatakeyama D, Aoki H, Abe C, Morita H, Kunisada T, Shibata T, Fukumitsu H, Tezuka KI. FGF2-responsive genes in human dental pulp cells assessed using a rat spinal cord injury model. J Bone Miner Metab 2019; 37:467-474. [PMID: 30187276 DOI: 10.1007/s00774-018-0954-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 08/22/2018] [Indexed: 02/06/2023]
Abstract
The central nervous system in adult mammals does not heal spontaneously after spinal cord injury (SCI). However, SCI treatment has been improved recently following the development of cell transplantation therapy. We recently reported that fibroblast growth factor (FGF) 2-pretreated human dental pulp cells (hDPCs) can improve recovery in a rat model of SCI. This study aimed to investigate mechanisms underlying the curative effect of SCI enhanced via FGF2 pretreatment; we selected three hDPC lines upon screening for the presence of mesenchymal stem cell markers and of their functionality in a rat model of SCI, as assessed using the Basso, Beattie, and Bresnahan score of locomotor functional scale, electrophysiological tests, and morphological analyses. We identified FGF2-responsive genes via gene expression analyses in these lines. FGF2 treatment upregulated GABRB1, MMP1, and DRD2, which suggested to contribute to SCI or central the nervous system. In an expanded screening of additional lines, GABRB1 displayed rather unique and interesting behavior; two lines with the lowest sensitivity of GABRB1 to FGF2 treatment displayed an extremely minor effect in the SCI model. These findings provide insights into the role of FGF2-responsive genes, especially GABRB1, in recovery from SCI, using hDPCs treated with FGF2.
Collapse
Affiliation(s)
- Ken Sugiyama
- Department of Oral and Maxillofacial Science, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, Gifu, 501-1194, Japan
| | - Kosuke Nagashima
- Laboratory of Molecular Biology, Gifu Pharmaceutical University, 1-25-4 Daigaku-Nishi, Gifu, Gifu, 501-1196, Japan
| | - Takahiro Miwa
- Laboratory of Molecular Biology, Gifu Pharmaceutical University, 1-25-4 Daigaku-Nishi, Gifu, Gifu, 501-1196, Japan
| | - Yuta Shimizu
- Department of Periodontology, Asahi University School of Dentistry, 1851-1 Hozumi, Mizuho, Gifu, 501-0296, Japan
| | - Tomoko Kawaguchi
- Department of Oral and Maxillofacial Science, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, Gifu, 501-1194, Japan
| | - Kazuki Iida
- Department of Oral and Maxillofacial Science, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, Gifu, 501-1194, Japan
| | - Naritaka Tamaoki
- Department of Oral and Maxillofacial Science, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, Gifu, 501-1194, Japan
| | - Daijiro Hatakeyama
- Department of Oral and Maxillofacial Science, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, Gifu, 501-1194, Japan
| | - Hitomi Aoki
- Department of Tissue and Organ Development, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, Gifu, 501-1194, Japan
| | - Chikara Abe
- Department of Physiology, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, Gifu, 501-1194, Japan
| | - Hironobu Morita
- Department of Physiology, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, Gifu, 501-1194, Japan
| | - Takahiro Kunisada
- Department of Tissue and Organ Development, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, Gifu, 501-1194, Japan
| | - Toshiyuki Shibata
- Department of Oral and Maxillofacial Science, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, Gifu, 501-1194, Japan
| | - Hidefumi Fukumitsu
- Laboratory of Molecular Biology, Gifu Pharmaceutical University, 1-25-4 Daigaku-Nishi, Gifu, Gifu, 501-1196, Japan
| | - Ken-Ichi Tezuka
- Department of Tissue and Organ Development, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, Gifu, 501-1194, Japan.
- Center for Highly Advanced Integration of Nano and Life Sciences, Gifu University (G-CHAIN), Gifu, Japan.
| |
Collapse
|
232
|
Gancheva MR, Kremer KL, Gronthos S, Koblar SA. Using Dental Pulp Stem Cells for Stroke Therapy. Front Neurol 2019; 10:422. [PMID: 31110489 PMCID: PMC6501465 DOI: 10.3389/fneur.2019.00422] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 04/08/2019] [Indexed: 12/26/2022] Open
Abstract
Stroke is a leading cause of permanent disability world-wide, but aside from rehabilitation, there is currently no clinically-proven pharmaceutical or biological agent to improve neurological disability. Cell-based therapies using stem cells, such as dental pulp stem cells, are a promising alternative for treatment of neurological diseases, including stroke. The ischaemic environment in stroke affects multiple cell populations, thus stem cells, which act through cellular and molecular mechanisms, are promising candidates. The most common stem cell population studied in the neurological setting has been mesenchymal stem cells due to their accessibility. However, it is believed that neural stem cells, the resident stem cell of the adult brain, would be most appropriate for brain repair. Using reprogramming strategies, alternative sources of neural stem and progenitor cells have been explored. We postulate that a cell of closer origin to the neural lineage would be a promising candidate for reprogramming and modification towards a neural stem or progenitor cell. One such candidate population is dental pulp stem cells, which reside in the root canal of teeth. This review will focus on the neural potential of dental pulp stem cells and their investigations in the stroke setting to date, and include an overview on the use of different sources of neural stem cells in preclinical studies and clinical trials of stroke.
Collapse
Affiliation(s)
- Maria R. Gancheva
- Stroke Research Programme Laboratory, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Karlea L. Kremer
- Stroke Research Programme Laboratory, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Stan Gronthos
- Mesenchymal Stem Cell Laboratory, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
- South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Simon A. Koblar
- Stroke Research Programme Laboratory, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
- South Australian Health and Medical Research Institute, Adelaide, SA, Australia
- Central Adelaide Local Health Network, Adelaide, SA, Australia
| |
Collapse
|
233
|
Diaz RE, Shylo NA, Roellig D, Bronner M, Trainor PA. Filling in the phylogenetic gaps: Induction, migration, and differentiation of neural crest cells in a squamate reptile, the veiled chameleon (Chamaeleo calyptratus). Dev Dyn 2019; 248:709-727. [DOI: 10.1002/dvdy.38] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 04/04/2019] [Accepted: 04/04/2019] [Indexed: 12/13/2022] Open
Affiliation(s)
- Raul E. Diaz
- Department of Biological Sciences, Southeastern Louisiana University Hammond Louisiana
- Natural History Museum of Los Angeles CountyDivision of Herpetology Los Angeles California
| | | | - Daniela Roellig
- Division of Biology and Biological Engineering, California Institute of Technology Pasadena California
| | - Marianne Bronner
- Division of Biology and Biological Engineering, California Institute of Technology Pasadena California
| | - Paul A. Trainor
- Stowers Institute for Medical Research Kansas City Missouri
- Department of Anatomy and Cell Biology, University of Kansas Medical Center Kansas City Kansas
| |
Collapse
|
234
|
Shakiba N, Fahmy A, Jayakumaran G, McGibbon S, David L, Trcka D, Elbaz J, Puri MC, Nagy A, van der Kooy D, Goyal S, Wrana JL, Zandstra PW. Cell competition during reprogramming gives rise to dominant clones. Science 2019; 364:science.aan0925. [DOI: 10.1126/science.aan0925] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 08/02/2018] [Accepted: 02/25/2019] [Indexed: 12/25/2022]
Abstract
The ability to generate induced pluripotent stem cells from differentiated cell types has enabled researchers to engineer cell states. Although studies have identified molecular networks that reprogram cells to pluripotency, the cellular dynamics of these processes remain poorly understood. Here, by combining cellular barcoding, mathematical modeling, and lineage tracing approaches, we demonstrate that reprogramming dynamics in heterogeneous populations are driven by dominant “elite” clones. Clones arise a priori from a population of poised mouse embryonic fibroblasts derived from Wnt1-expressing cells that may represent a neural crest–derived population. This work highlights the importance of cellular dynamics in fate programming outcomes and uncovers cell competition as a mechanism by which cells with eliteness emerge to occupy and dominate the reprogramming niche.
Collapse
|
235
|
Collignon AM, Castillo-Dali G, Gomez E, Guilbert T, Lesieur J, Nicoletti A, Acuna-Mendoza S, Letourneur D, Chaussain C, Rochefort GY, Poliard A. Mouse Wnt1-CRE
-Rosa
Tomato
Dental Pulp Stem Cells Directly Contribute to the Calvarial Bone Regeneration Process. Stem Cells 2019; 37:701-711. [DOI: 10.1002/stem.2973] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 12/14/2018] [Accepted: 12/19/2018] [Indexed: 01/08/2023]
Affiliation(s)
- Anne-Margaux Collignon
- EA 2496 Orofacial Pathologies, Imagery, and Biotherapies, Dental School Faculty; University Paris Descartes, and Life Imaging Platform (PIV); Montrouge France
- University Hospitals, AP-HP; Paris France
| | - Gabriel Castillo-Dali
- EA 2496 Orofacial Pathologies, Imagery, and Biotherapies, Dental School Faculty; University Paris Descartes, and Life Imaging Platform (PIV); Montrouge France
| | - Eduardo Gomez
- EA 2496 Orofacial Pathologies, Imagery, and Biotherapies, Dental School Faculty; University Paris Descartes, and Life Imaging Platform (PIV); Montrouge France
| | - Thomas Guilbert
- Plateforme IMAG'IC, Institut Cochin, Inserm U1016-CNRS UMR8104; University Paris Descartes; Paris France
| | - Julie Lesieur
- EA 2496 Orofacial Pathologies, Imagery, and Biotherapies, Dental School Faculty; University Paris Descartes, and Life Imaging Platform (PIV); Montrouge France
| | - Antonino Nicoletti
- INSERM U1148, Laboratory of Vascular Translational Science; University Paris Diderot, University Paris 13, Bichat Hospital, and Département Hospitalo-Universitaire (DHU) FIRE; Paris France
| | - Soledad Acuna-Mendoza
- EA 2496 Orofacial Pathologies, Imagery, and Biotherapies, Dental School Faculty; University Paris Descartes, and Life Imaging Platform (PIV); Montrouge France
| | - Didier Letourneur
- INSERM U1148, Laboratory of Vascular Translational Science; University Paris Diderot, University Paris 13, Bichat Hospital, and Département Hospitalo-Universitaire (DHU) FIRE; Paris France
| | - Catherine Chaussain
- EA 2496 Orofacial Pathologies, Imagery, and Biotherapies, Dental School Faculty; University Paris Descartes, and Life Imaging Platform (PIV); Montrouge France
- University Hospitals, AP-HP; Paris France
| | - Gael Y. Rochefort
- EA 2496 Orofacial Pathologies, Imagery, and Biotherapies, Dental School Faculty; University Paris Descartes, and Life Imaging Platform (PIV); Montrouge France
| | - Anne Poliard
- EA 2496 Orofacial Pathologies, Imagery, and Biotherapies, Dental School Faculty; University Paris Descartes, and Life Imaging Platform (PIV); Montrouge France
| |
Collapse
|
236
|
Etchevers HC, Dupin E, Le Douarin NM. The diverse neural crest: from embryology to human pathology. Development 2019; 146:146/5/dev169821. [PMID: 30858200 DOI: 10.1242/dev.169821] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 02/07/2019] [Indexed: 01/13/2023]
Abstract
We review here some of the historical highlights in exploratory studies of the vertebrate embryonic structure known as the neural crest. The study of the molecular properties of the cells that it produces, their migratory capacities and plasticity, and the still-growing list of tissues that depend on their presence for form and function, continue to enrich our understanding of congenital malformations, paediatric cancers and evolutionary biology. Developmental biology has been key to our understanding of the neural crest, starting with the early days of experimental embryology and through to today, when increasingly powerful technologies contribute to further insight into this fascinating vertebrate cell population.
Collapse
Affiliation(s)
- Heather C Etchevers
- Aix-Marseille Université, INSERM, MMG, U1251, 27 boulevard Jean Moulin 13005 Marseille, France
| | - Elisabeth Dupin
- Sorbonne Universités, UPMC Paris 06, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, 75012 Paris, France
| | - Nicole M Le Douarin
- Sorbonne Universités, UPMC Paris 06, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, 75012 Paris, France
| |
Collapse
|
237
|
Clinical Potential and Current Progress of Dental Pulp Stem Cells for Various Systemic Diseases in Regenerative Medicine: A Concise Review. Int J Mol Sci 2019; 20:ijms20051132. [PMID: 30845639 PMCID: PMC6429131 DOI: 10.3390/ijms20051132] [Citation(s) in RCA: 157] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Revised: 02/28/2019] [Accepted: 03/01/2019] [Indexed: 12/13/2022] Open
Abstract
Dental pulp stem cells (DPSCs) are mesenchymal stem cells (MSCs) that have multipotent differentiation and a self-renewal ability. They have been useful not only for dental diseases, but also for systemic diseases. Extensive studies have suggested that DPSCs are effective for various diseases, such as spinal cord injuries, Parkinson's disease, Alzheimer's disease, cerebral ischemia, myocardial infarction, muscular dystrophy, diabetes, liver diseases, eye diseases, immune diseases, and oral diseases. DPSCs have the potential for use in a cell-therapeutic paradigm shift to treat these diseases. It has also been reported that DPSCs have higher regenerative potential than the bone marrow-derived mesenchymal stem cells known as representative MSCs. Therefore, DPSCs have recently gathered much attention. In this review, the therapeutic potential of DPSCs, the latest progress in the pre-clinical study for treatment of these various systemic diseases, and the clinical applications of DPSCs in regenerative medicine, are all summarized. Although challenges, including mechanisms of the effects and establishment of cell processing and transplantation methods for clinical use, still remain, DPSCs could be promising stem cells sources for various clinical applications, because of their easy isolation by a noninvasive procedure without ethical concerns.
Collapse
|
238
|
Abstract
Jaw bones and teeth originate from the first pharyngeal arch and develop in closely related ways. Reciprocal epithelial-mesenchymal interactions are required for the early patterning and morphogenesis of both tissues. Here we review the cellular contribution during the development of the jaw bones and teeth. We also highlight signaling networks as well as transcription factors mediating tissue-tissue interactions that are essential for jaw bone and tooth development. Finally, we discuss the potential for stem cell mediated regenerative therapies to mitigate disorders and injuries that affect these organs.
Collapse
Affiliation(s)
- Yuan Yuan
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA, United States.
| | - Yang Chai
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA, United States.
| |
Collapse
|
239
|
Ankamreddy H, Min H, Kim JY, Yang X, Cho ES, Kim UK, Bok J. Region-specific endodermal signals direct neural crest cells to form the three middle ear ossicles. Development 2019; 146:dev.167965. [PMID: 30630826 DOI: 10.1242/dev.167965] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 12/24/2018] [Indexed: 11/20/2022]
Abstract
Defects in the middle ear ossicles - malleus, incus and stapes - can lead to conductive hearing loss. During development, neural crest cells (NCCs) migrate from the dorsal hindbrain to specific locations in pharyngeal arch (PA) 1 and 2, to form the malleus-incus and stapes, respectively. It is unclear how migratory NCCs reach their proper destination in the PA and initiate mesenchymal condensation to form specific ossicles. We show that secreted molecules sonic hedgehog (SHH) and bone morphogenetic protein 4 (BMP4) emanating from the pharyngeal endoderm are important in instructing region-specific NCC condensation to form malleus-incus and stapes, respectively, in mouse. Tissue-specific knockout of Shh in the pharyngeal endoderm or Smo (a transducer of SHH signaling) in NCCs causes the loss of malleus-incus condensation in PA1 but only affects the maintenance of stapes condensation in PA2. By contrast, knockout of Bmp4 in the pharyngeal endoderm or Smad4 (a transducer of TGFβ/BMP signaling) in the NCCs disrupts NCC migration into the stapes region in PA2, affecting stapes formation. These results indicate that region-specific endodermal signals direct formation of specific middle ear ossicles.
Collapse
Affiliation(s)
- Harinarayana Ankamreddy
- Department of Anatomy, Yonsei University College of Medicine, Seoul, South Korea.,BK21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Hyehyun Min
- Department of Anatomy, Yonsei University College of Medicine, Seoul, South Korea
| | - Jae Yoon Kim
- Department of Anatomy, Yonsei University College of Medicine, Seoul, South Korea.,BK21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Xiao Yang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Eui-Sic Cho
- Cluster for Craniofacial Development and Regeneration Research, Institute of Oral Biosciences, Chonbuk National University School of Dentistry, Jeonju, South Korea
| | - Un-Kyung Kim
- Department of Biology, College of Natural Sciences, Kyungpook National University, Daegu, South Korea.,School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu, South Korea
| | - Jinwoong Bok
- Department of Anatomy, Yonsei University College of Medicine, Seoul, South Korea .,BK21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea.,Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
240
|
Head to Knee: Cranial Neural Crest-Derived Cells as Promising Candidates for Human Cartilage Repair. Stem Cells Int 2019; 2019:9310318. [PMID: 30766608 PMCID: PMC6350557 DOI: 10.1155/2019/9310318] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 11/04/2018] [Accepted: 12/06/2018] [Indexed: 12/13/2022] Open
Abstract
A large array of therapeutic procedures is available to treat cartilage disorders caused by trauma or inflammatory disease. Most are invasive and may result in treatment failure or development of osteoarthritis due to extensive cartilage damage from repeated surgery. Despite encouraging results of early cell therapy trials that used chondrocytes collected during arthroscopic surgery, these approaches have serious disadvantages, including morbidity associated with cell harvesting and low predictive clinical outcomes. To overcome these limitations, adult stem cells derived from bone marrow and subsequently from other tissues are now considered as preferred sources of cells for cartilage regeneration. Moreover, with new evidence showing that the choice of cell source is one of the most important factors for successful cell therapy, there is growing interest in neural crest-derived cells in both the research and clinical communities. Neural crest-derived cells such as nasal chondrocytes and oral stem cells that exhibit chondrocyte-like properties seem particularly promising in cartilage repair. Here, we review the types of cells currently available for cartilage cell therapy, including articular chondrocytes and various mesenchymal stem cells, and then highlight recent developments in the use of neural crest-derived chondrocytes and oral stem cells for repair of cartilage lesions.
Collapse
|
241
|
Vps4b heterozygous mice do not develop tooth defects that replicate human dentin dysplasia I. BMC Genet 2019; 20:7. [PMID: 30634912 PMCID: PMC6330468 DOI: 10.1186/s12863-018-0699-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 11/22/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Vacuolar protein sorting-associated protein 4B (VPS4B) is a member of the ATP enzyme AAA protein family, and is mainly involved in protein degradation and cell membrane fusion. Recently, a dominant mutation in this gene was identified in human dentin dysplasia type I (DD-I). Herein, we report the generation of Vps4b knockout (Vps4b KO) mice; however, the homozygous Vps4b KO mutation was embryonic lethal at the early stages of embryo development, and we therefore report the results of heterozygous mutant mice. RESULTS Mice heterozygous for Vps4b did not develop tooth defects replicating human DD-I. Immunohistochemistry showed that gene KO was successful, as there was decreased expression of Vps4b in heterozygous mice; hematoxylin and eosin (H&E) staining also showed that the width of the pre-dentin zone was increased in heterozygous mice, although the arrangement of the odontoblasts was not significantly different from wild-type (WT) mice. However, H&E staining showed no obvious abnormalities in the bones of heterozygous mice. Moreover, stereomicroscopic and X-ray radiography results indicated no abnormal manifestations in teeth or bones. Furthermore, statistical analysis of the volume and density of dentin and enamel, as well as skeletal analysis, including the volume and separation of trabecular bone analyzed by micro-CT, all showed no differences between Vps4b heterozygotes and WT mice. In addition, there also were no significant differences in bone or cartilage mineralization as evaluated by Alcian blue-Alizarin red staining. CONCLUSIONS The heterozygous Vps4b KO mice do not develop tooth defects that replicate human DD-I and this is likely to be due to differences in tooth development between the two species. Consequently, further studies are needed to determine whether mice are an appropriate animal model for human tooth diseases.
Collapse
|
242
|
Michalski MN, Zweifler LE, Sinder BP, Koh AJ, Yamashita J, Roca H, McCauley LK. Clodronate-Loaded Liposome Treatment Has Site-Specific Skeletal Effects. J Dent Res 2019; 98:459-467. [PMID: 30626255 DOI: 10.1177/0022034518821685] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Ineffective oral wound healing is detrimental to patients' oral health-related quality of life. Delineating the cellular mechanisms involved in optimal healing will elicit better approaches to treating patients with compromised healing. Osteal macrophages have recently emerged as important positive regulators of bone turnover. The contributions of macrophages to long bone healing have been studied, but their role in oral osseous wound healing following tooth extraction is less clear. Clodronate-loaded liposomes were used as a tool to deplete macrophages in C57BL/6J mice and assess oral osseous bone fill after extraction. In addition to macrophage ablation, osteoclast ablation occurred. Interestingly, depletion of macrophages and osteoclasts via clodronate treatment had differential effects based on skeletal location. In the nonwounded tibiae, clodronate treatment significantly increased CD68+ cells and decreased F4/80+ cells in the marrow, which correlated with increased trabecular bone volume fraction after 7 and 14 d. Serum formation and resorptive markers P1NP and TRAcP 5b were decreased as were tibial TRAP+ osteoclasts. In healing extraction sockets, clodronate treatment increased extraction socket trabecular bone thickness at 14 d, which correlated with decreased TRAP+ osteoclasts and F4/80+ macrophages. Conversely, nonwounded maxillary interseptal bone was unaffected by clodronate treatment. Furthermore, the increase in extraction socket bone fill with clodronate was less than the large increase in trabecular bone observed in a nonwounded long bone. These data suggest a temporal and spatial specificity in the roles of macrophages and osteoclasts in normal turnover and healing.
Collapse
Affiliation(s)
- M N Michalski
- 1 Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - L E Zweifler
- 1 Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - B P Sinder
- 1 Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - A J Koh
- 1 Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - J Yamashita
- 2 Department of Oral and Maxillofacial Surgery, Fukuoka Dental College, Fukuoka, Japan
| | - H Roca
- 1 Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - L K McCauley
- 1 Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI, USA.,3 Department of Pathology, Medical School, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
243
|
|
244
|
An Integrative View of Lepidosaur Cranial Anatomy, Development, and Diversification. HEADS, JAWS, AND MUSCLES 2019. [DOI: 10.1007/978-3-319-93560-7_9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
245
|
Kindberg AA, Bush JO. Cellular organization and boundary formation in craniofacial development. Genesis 2019; 57:e23271. [PMID: 30548771 PMCID: PMC6503678 DOI: 10.1002/dvg.23271] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 12/07/2018] [Accepted: 12/10/2018] [Indexed: 12/24/2022]
Abstract
Craniofacial morphogenesis is a highly dynamic process that requires changes in the behaviors and physical properties of cells in order to achieve the proper organization of different craniofacial structures. Boundary formation is a critical process in cellular organization, patterning, and ultimately tissue separation. There are several recurring cellular mechanisms through which boundary formation and cellular organization occur including, transcriptional patterning, cell segregation, cell adhesion and migratory guidance. Disruption of normal boundary formation has dramatic morphological consequences, and can result in human craniofacial congenital anomalies. In this review we discuss boundary formation during craniofacial development, specifically focusing on the cellular behaviors and mechanisms underlying the self-organizing properties that are critical for craniofacial morphogenesis.
Collapse
Affiliation(s)
- Abigail A. Kindberg
- Department of Cell and Tissue Biology, Program in Craniofacial Biology, and Institute of Human Genetics, University of California at San Francisco, San Francisco, CA 94143, USA
| | - Jeffrey O. Bush
- Department of Cell and Tissue Biology, Program in Craniofacial Biology, and Institute of Human Genetics, University of California at San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
246
|
Kaneko T, Gu B, Sone PP, Zaw SYM, Murano H, Zaw ZCT, Okiji T. Dental Pulp Tissue Engineering Using Mesenchymal Stem Cells: a Review with a Protocol. Stem Cell Rev Rep 2018; 14:668-676. [PMID: 29804171 DOI: 10.1007/s12015-018-9826-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Mesenchymal stem cells (MSCs) are adult stem cells that can be isolated from human and animal sources such as rats. Recently, an in vivo protocol for pulp tissue engineering using implantation of bone marrow MSCs into rat pulpotomized molars was established by our research group. This coronal pulp regeneration model showed almost complete regeneration/healing with dentin bridge formation when the cavity was sealed with mineral trioxide aggregate (MTA) to create a biocompatible seal of the pulp. This method is a powerful tool for elucidating the processes of dental pulp tissue regeneration following implantation of MSCs. In the present review, we discuss the literature in the field of dental pulp tissue engineering using MSCs including dental pulp stem cells and stem cells from exfoliated deciduous teeth. In addition, we present a brief step-by-step protocol of the coronal pulp regeneration model focusing on the implantation of rat bone marrow MSCs, biodegradable scaffolds, and hydrogels in pulpotomized rat molars. The protocol may lay the foundation for studies aiming at defining further histological and molecular mechanism of the rat pulp tissue engineering.
Collapse
Affiliation(s)
- Tomoatsu Kaneko
- Department of Pulp Biology and Endodontics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Yushima 1-5-45, Bunkyo-Ku, Tokyo, 113-8549, Japan.
| | - Bin Gu
- Department of Pulp Biology and Endodontics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Yushima 1-5-45, Bunkyo-Ku, Tokyo, 113-8549, Japan
| | - Phyo Pyai Sone
- Department of Pulp Biology and Endodontics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Yushima 1-5-45, Bunkyo-Ku, Tokyo, 113-8549, Japan
| | - Su Yee Myo Zaw
- Department of Pulp Biology and Endodontics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Yushima 1-5-45, Bunkyo-Ku, Tokyo, 113-8549, Japan
| | - Hiroki Murano
- Department of Pulp Biology and Endodontics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Yushima 1-5-45, Bunkyo-Ku, Tokyo, 113-8549, Japan
| | - Zar Chi Thein Zaw
- Department of Pulp Biology and Endodontics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Yushima 1-5-45, Bunkyo-Ku, Tokyo, 113-8549, Japan
| | - Takashi Okiji
- Department of Pulp Biology and Endodontics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Yushima 1-5-45, Bunkyo-Ku, Tokyo, 113-8549, Japan
| |
Collapse
|
247
|
Calamari ZT, Kuang-Hsien Hu J, Klein OD. Tissue Mechanical Forces and Evolutionary Developmental Changes Act Through Space and Time to Shape Tooth Morphology and Function. Bioessays 2018; 40:e1800140. [PMID: 30387177 PMCID: PMC6516060 DOI: 10.1002/bies.201800140] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 10/06/2018] [Indexed: 12/24/2022]
Abstract
Efforts from diverse disciplines, including evolutionary studies and biomechanical experiments, have yielded new insights into the genetic, signaling, and mechanical control of tooth formation and functions. Evidence from fossils and non-model organisms has revealed that a common set of genes underlie tooth-forming potential of epithelia, and changes in signaling environments subsequently result in specialized dentitions, maintenance of dental stem cells, and other phenotypic adaptations. In addition to chemical signaling, tissue forces generated through epithelial contraction, differential growth, and skeletal constraints act in parallel to shape the tooth throughout development. Here recent advances in understanding dental development from these studies are reviewed and important gaps that can be filled through continued application of evolutionary and biomechanical approaches are discussed.
Collapse
Affiliation(s)
- Zachary T. Calamari
- Department of Natural Sciences, Baruch College, City University of New York, New York City, New York, 10010, USA
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, San Francisco, California, 94143, USA
| | - Jimmy Kuang-Hsien Hu
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, San Francisco, California, 94143, USA
| | - Ophir D. Klein
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, San Francisco, California, 94143, USA
- Department of Pediatrics and Institute for Human Genetics, University of California, San Francisco, San Francisco, California, 94143, USA
| |
Collapse
|
248
|
Betters E, Charney RM, Garcia-Castro MI. Early specification and development of rabbit neural crest cells. Dev Biol 2018; 444 Suppl 1:S181-S192. [PMID: 29932896 PMCID: PMC6685428 DOI: 10.1016/j.ydbio.2018.06.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 06/01/2018] [Accepted: 06/18/2018] [Indexed: 11/19/2022]
Abstract
The phenomenal migratory and differentiation capacity of neural crest cells has been well established across model organisms. While the earliest stages of neural crest development have been investigated in non-mammalian model systems such as Xenopus and Aves, the early specification of this cell population has not been evaluated in mammalian embryos, of which the murine model is the most prevalent. Towards a more comprehensive understanding of mammalian neural crest formation and human comparative studies, we have used the rabbit as a mammalian system for the study of early neural crest specification and development. We examine the expression profile of well-characterized neural crest markers in rabbit embryos across developmental time from early gastrula to later neurula stages, and provide a comparison to markers of migratory neural crest in the chick. Importantly, we apply explant specification assays to address the pivotal question of mammalian neural crest ontogeny, and provide the first evidence that a specified population of neural crest cells exists in the rabbit gastrula prior to the overt expression of neural crest markers. Finally, we demonstrate that FGF signaling is necessary for early rabbit neural crest formation, as SU5402 treatment strongly represses neural crest marker expression in explant assays. This study pioneers the rabbit as a model for neural crest development, and provides the first demonstration of mammalian neural crest specification and the requirement of FGF signaling in this process.
Collapse
Affiliation(s)
- Erin Betters
- School of Medicine Division of Biomedical Sciences, University of California, Riverside, CA 92521, USA
| | - Rebekah M Charney
- School of Medicine Division of Biomedical Sciences, University of California, Riverside, CA 92521, USA
| | - Martín I Garcia-Castro
- School of Medicine Division of Biomedical Sciences, University of California, Riverside, CA 92521, USA.
| |
Collapse
|
249
|
Le Douarin NM, Dupin E. The “beginnings” of the neural crest. Dev Biol 2018; 444 Suppl 1:S3-S13. [DOI: 10.1016/j.ydbio.2018.07.019] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 07/20/2018] [Accepted: 07/20/2018] [Indexed: 12/14/2022]
|
250
|
Collignon AM, Lesieur J, Anizan N, Azzouna RB, Poliard A, Gorin C, Letourneur D, Chaussain C, Rouzet F, Rochefort GY. Early angiogenesis detected by PET imaging with 64Cu-NODAGA-RGD is predictive of bone critical defect repair. Acta Biomater 2018; 82:111-121. [PMID: 30312778 DOI: 10.1016/j.actbio.2018.10.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Revised: 10/04/2018] [Accepted: 10/07/2018] [Indexed: 12/15/2022]
Abstract
Therapies using stem cells may be applicable to all fields of regenerative medicine, including craniomaxillofacial surgery. Dental pulp stem cells (DPSCs) have demonstrated in vitro and in vivo osteogenic and proangiogenic properties. The aim of the study was to evaluate whether early angiogenesis investigated by nuclear imaging can predict bone formation within a mouse critical bone defect. Two symmetrical calvarial critical-sized defects were created. Defects were left empty or filled with i) DPSC-containing dense collagen scaffold, ii) 5% hypoxia-primed DPSC-containing dense collagen scaffold, iii) acellular dense collagen scaffold, or iv) left empty. Early angiogenesis assessed by PET using 64Cu-NODAGA-RGD as a tracer was found to be correlated with bone formation determined by micro-CT within the defects from day 30, and to be correlated to the late calcium apposition observed at day 90 using 18F-Na PET. These results suggest that nuclear imaging of angiogenesis, a technique applicable in clinical practice, is a promising approach for early prediction of bone grafting outcome, thus potentially allowing to anticipate alternative regenerative strategies. STATEMENT OF SIGNIFICANCE: Bone defects are a major concern in medicine. As life expectancy increases, the number of bone lesions grows, and occurring complications lead to a delay or even lack of consolidation. Therefore, to be able to predict healing or the absence of scarring at early times would be very interesting. This would not "waste time" for the patient. We report here that early nuclear imaging of angiogenesis, using 64Cu-NODAGA-RGD as a tracer, associated with nuclear imaging of mineralization, using 18F-Na as a tracer, is correlated to late bone healing objectivized by classical histology and microtomography. This nuclear imaging represents a promising approach for early prediction of bone grafting outcome in clinical practice, thus potentially allowing to anticipate alternative regenerative strategies.
Collapse
Affiliation(s)
- Anne-Margaux Collignon
- EA 2496 Orofacial Pathologies, Imagery and Biotherapies, Dental School Faculty, University Paris Descartes and Life Imaging Platform (PIV), Montrouge, France; University Hospitals, AP-HP, Paris, France
| | - Julie Lesieur
- EA 2496 Orofacial Pathologies, Imagery and Biotherapies, Dental School Faculty, University Paris Descartes and Life Imaging Platform (PIV), Montrouge, France
| | - Nadège Anizan
- Fédération de Recherche en Imagerie Multimodale (FRIM), Inserm UMS-34, Université Paris Diderot, Paris, France
| | - Rana Ben Azzouna
- University Hospitals, AP-HP, Paris, France; Fédération de Recherche en Imagerie Multimodale (FRIM), Inserm UMS-34, Université Paris Diderot, Paris, France; INSERM U1148, Laboratory of Vascular Translational Science, University Paris Diderot, University Paris 13, X Bichat Hospital, and Département Hospitalo-Universitaire (DHU) FIRE, F-75018 Paris, France
| | - Anne Poliard
- EA 2496 Orofacial Pathologies, Imagery and Biotherapies, Dental School Faculty, University Paris Descartes and Life Imaging Platform (PIV), Montrouge, France
| | - Caroline Gorin
- EA 2496 Orofacial Pathologies, Imagery and Biotherapies, Dental School Faculty, University Paris Descartes and Life Imaging Platform (PIV), Montrouge, France; University Hospitals, AP-HP, Paris, France
| | - Didier Letourneur
- INSERM U1148, Laboratory of Vascular Translational Science, University Paris Diderot, University Paris 13, X Bichat Hospital, and Département Hospitalo-Universitaire (DHU) FIRE, F-75018 Paris, France
| | - Catherine Chaussain
- EA 2496 Orofacial Pathologies, Imagery and Biotherapies, Dental School Faculty, University Paris Descartes and Life Imaging Platform (PIV), Montrouge, France; University Hospitals, AP-HP, Paris, France
| | - Francois Rouzet
- University Hospitals, AP-HP, Paris, France; Fédération de Recherche en Imagerie Multimodale (FRIM), Inserm UMS-34, Université Paris Diderot, Paris, France; INSERM U1148, Laboratory of Vascular Translational Science, University Paris Diderot, University Paris 13, X Bichat Hospital, and Département Hospitalo-Universitaire (DHU) FIRE, F-75018 Paris, France.
| | - Gael Y Rochefort
- EA 2496 Orofacial Pathologies, Imagery and Biotherapies, Dental School Faculty, University Paris Descartes and Life Imaging Platform (PIV), Montrouge, France.
| |
Collapse
|