1
|
Zhao B, Suh J, Zhang Y, Yin E, Kadota-Watanabe C, Chang IW, Yaung J, Lao-Ngo I, Young NM, Kim RH, Klein OD, Hong C. p75 neurotrophin receptor regulates craniofacial growth and morphology in postnatal development. Front Cell Dev Biol 2025; 13:1569533. [PMID: 40171227 PMCID: PMC11959563 DOI: 10.3389/fcell.2025.1569533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Accepted: 03/03/2025] [Indexed: 04/03/2025] Open
Abstract
Craniofacial abnormalities are among the most prevalent congenital defects, significantly affecting appearance, function, and quality of life. While the role of genetic mutations in craniofacial malformations is recognized, the underlying molecular mechanisms remain poorly understood. In this study, we investigate the role of p75 neurotrophin receptor (p75NTR) in craniofacial development by comparing wild-type (p75NTR+/+) mice against p75NTR-deficient (p75NTR-/-) knockout mice. We employed histology, micro-CT surface distance, volumetric analysis, and geometric morphometric analysis to assess craniofacial development and growth. On postnatal day 7 (P7), p75NTR-/- mice exhibited reduced skull length compared to wild-type controls. By P28, micro-CT analysis revealed significant reductions in calvarial bone volume and trabecular bone thickness in p75NTR-/- mice. Geometric morphometric analysis identified significant shape alterations in the nasal, parietal, and occipital regions, with p75NTR-/- mice showing a shortened cranium and tapered nasal bone morphology. These findings highlight the critical role of p75NTR in regulating postnatal craniofacial development. Disruption of p75NTR signaling impairs both the growth and morphological integrity of craniofacial structures, which may contribute to the pathogenesis of congenital craniofacial abnormalities. In the future, a better understanding of the molecular mechanisms through which p75NTR mediates craniofacial development may offer valuable insights for future targeted therapeutic strategies for craniofacial defects.
Collapse
Affiliation(s)
- Byron Zhao
- Division of Orthodontics, Department of Orofacial Sciences, University of California, San Francisco, San Francisco, CA, United States
| | - Jinsook Suh
- Division of Orthodontics, Department of Orofacial Sciences, University of California, San Francisco, San Francisco, CA, United States
| | - Yan Zhang
- Division of Orthodontics, Department of Orofacial Sciences, University of California, San Francisco, San Francisco, CA, United States
| | - Eric Yin
- Division of Orthodontics, Department of Orofacial Sciences, University of California, San Francisco, San Francisco, CA, United States
| | - Chiho Kadota-Watanabe
- Division of Orthodontics, Department of Orofacial Sciences, University of California, San Francisco, San Francisco, CA, United States
- Division of Maxillofacial and Neck Reconstruction, Department of Maxillofacial Orthognathics, Institute of Science Tokyo, Tokyo, Japan
| | - In Won Chang
- Shapiro Family Laboratory of Viral Oncology and Aging Research, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, United States
| | - Jun Yaung
- Shapiro Family Laboratory of Viral Oncology and Aging Research, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, United States
| | - Isabelle Lao-Ngo
- Division of Orthodontics, Department of Orofacial Sciences, University of California, San Francisco, San Francisco, CA, United States
| | - Nathan M. Young
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Reuben H. Kim
- Shapiro Family Laboratory of Viral Oncology and Aging Research, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, United States
| | - Ophir D. Klein
- Department of Orofacial Sciences, Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, United States
- Department of Pediatrics, Cedars-Sinai Guerin Children’s, Los Angeles, CA, United States
| | - Christine Hong
- Division of Orthodontics, Department of Orofacial Sciences, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
2
|
Cai S, Liu J, Chen Y, Yang X, Yan Y, Zheng D, Lu Y, Xu L. A unilateral increase in the occlusal vertical dimension of growing rats results in mandibular deviation. Orthod Craniofac Res 2025; 28:84-94. [PMID: 39193636 DOI: 10.1111/ocr.12851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 08/01/2024] [Accepted: 08/16/2024] [Indexed: 08/29/2024]
Abstract
OBJECTIVE The effects of unilateral increased occlusal vertical dimension (iOVD) on bilateral craniofacial, mandibular and alveolar development in growing rats were investigated via cone-beam computed tomography (CBCT). The role of Wnt/β-catenin signalling in this process was examined. MATERIALS AND METHODS Forty-eight female Sprague-Dawley rats were randomly allocated into unilateral iOVD and sham groups. At 2, 4 and 8 weeks, the rats were scanned via CBCT to analyse cranial, maxillary, mandibular and dental morphology. Changes in temporomandibular joint (TMJ) cartilage histology and Wnt/β-catenin signalling were assessed by histochemical and immunohistochemical staining and qRT-PCR. RESULTS Dorsal cephalograms revealed that the mandible in the iOVD group tilted approximately 4° to the right. Unilateral iOVD had little effect on cranial and maxillary growth but inhibited mandibular growth (mandibular length and ramal height), especially on the deviated side (DS). Moreover, unilateral iOVD increased the length of the lower incisors and decreased the height of the molars on the DS. Unilateral iOVD induced bilateral osteoarthritis-like changes in the bilateral TMJ condylar cartilage and activated Wnt/β-catenin signalling in the condylar cartilage, especially on the contralateral side (CLS). CONCLUSION Occlusion with unilateral iOVD induced mandibular deviation, significantly inhibited mandibular growth and produced compensatory changes in the alveolar bone. In the iOVD group, the mandibular body length and ramal height were greater on the CLS than on the DS. Moreover, the greater β-catenin protein expression in the TMJ condylar cartilage on the CLS than on the DS may account for the difference in asymmetrical mandibular development.
Collapse
Affiliation(s)
- Senxin Cai
- Department of Orthodontics, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Key Laboratory of Stomatology of Fujian Province, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Jing Liu
- Fujian Maternal and Child Health Hospital, Fuzhou, China
| | - Yao Chen
- Key Laboratory of Stomatology of Fujian Province, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Xinhan Yang
- Key Laboratory of Stomatology of Fujian Province, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Yuxiang Yan
- Key Laboratory of Stomatology of Fujian Province, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Dali Zheng
- Key Laboratory of Stomatology of Fujian Province, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Youguang Lu
- Department of Preventive Dentistry, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Linyu Xu
- Department of Orthodontics, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| |
Collapse
|
3
|
Gündemir O, Özaydin İ, Erkiliç EE, Öztürkler O, Büyükbaki B, Yilmaz A, Onar V, Aydin U, Aksoy Ö. Geometric morphometric analysis of red fox (Vulpes vulpes) skulls using radiometric techniques at three and six months of development. Ann Anat 2025; 258:152374. [PMID: 39756625 DOI: 10.1016/j.aanat.2025.152374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 12/20/2024] [Accepted: 12/30/2024] [Indexed: 01/07/2025]
Abstract
BACKGROUND Morphological growth naturally progresses with age; however, the rate of growth varies across different parts of an organism, with certain structures developing more rapidly than others. This study aimed to analyze and compare the skull development of red foxes (Vulpes vulpes) during two specific developmental stages: the 3rd and 6th months, which represent distinct growth phases in their early ontogeny. METHODS In this study, we aimed to analyze and compare skull development in red foxes (Vulpes vulpes) during two specific post-natal time points: the 3rd and 6th months. Shape analysis was performed using radiographic images of nine red foxes at both the third and sixth months. RESULTS Shape differences were observed in the skulls of red foxes (Vulpes vulpes) at these two ages. Our findings confirmed the hypothesis that skull shape changes over time, reflecting distinct morphological adaptations associated with age-related growth. In the measurements at the 3rd month, the neurocranial region exhibited a more distinct and developed structure compared to the facial bones. Toward the 6th month, the skull displayed a thinner and more elongated structure with the further development of the facial bones. CONCLUSIONS This difference indicates a period of rapid growth and development in the neurocranial area, suggesting that red foxes experience significant neurological and sensory development early in life. Future studies on skull shape variation across different developmental stages in red foxes can expand on these findings. Age-related morphological studies, such as this one, provide essential baseline data on the natural growth and development of wild species like red foxes. This knowledge is essential for identifying deviations from normal development, which could result from environmental stressors, habitat changes, or malnutrition.
Collapse
Affiliation(s)
- Ozan Gündemir
- Department of Anatomy, Faculty of Veterinary Medicine, Istanbul University-Cerrahpasa, Istanbul 34320, Türkiye.
| | - İsa Özaydin
- Department of Surgery, Faculty of Veterinary Medicine, Kafkas University, Kars 36100, Türkiye
| | - Ekin Emre Erkiliç
- Department of Internal Medicine, Faculty of Veterinary Medicine, Kafkas University, Kars 36100, Türkiye
| | - Olcay Öztürkler
- Department of Wildlife and Ecology, Faculty of Veterinary Medicine, Kafkas University, Kars 36100, Türkiye
| | - Burak Büyükbaki
- Department of Wildlife and Ecology, Faculty of Veterinary Medicine, Kafkas University, Kars 36100, Türkiye
| | - Alican Yilmaz
- Department of Surgery, Faculty of Veterinary Medicine, Kafkas University, Kars 36100, Türkiye
| | - Vedat Onar
- Department of Anatomy, Faculty of Veterinary Medicine, Istanbul University-Cerrahpasa, Istanbul 34320, Türkiye
| | - Uğur Aydin
- Department of Surgery, Faculty of Veterinary Medicine, Kafkas University, Kars 36100, Türkiye
| | - Özgür Aksoy
- Department of Surgery, Faculty of Veterinary Medicine, Kafkas University, Kars 36100, Türkiye
| |
Collapse
|
4
|
Raoul‐Duval J, Ganet A, Benichi S, Baixe P, Cornillon C, Eschapasse L, Geoffroy M, Paternoster G, James S, Laporte S, Blauwblomme T, Khonsari RH, Taverne M. Geometric growth of the normal human craniocervical junction from 0 to 18 years old. J Anat 2024; 245:842-863. [PMID: 38783688 PMCID: PMC11547232 DOI: 10.1111/joa.14067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 04/09/2024] [Accepted: 05/09/2024] [Indexed: 05/25/2024] Open
Abstract
The craniocervical junction (CCJ) forms the bridge between the skull and the spine, a highly mobile group of joints that allows the mobility of the head in every direction. The CCJ plays a major role in protecting the inferior brainstem (bulb) and spinal cord, therefore also requiring some stability. Children are subjected to multiple constitutive or acquired diseases involving the CCJ: primary bone diseases such as in FGFR-related craniosynostoses or acquired conditions such as congenital torticollis, cervical spine luxation, and neurological disorders. To design efficient treatment plans, it is crucial to understand the relationship between abnormalities of the craniofacial region and abnormalities of the CCJ. This can be approached by the study of control and abnormal growth patterns. Here we report a model of normal skull base growth by compiling a collection of geometric models in control children. Focused analyses highlighted specific developmental patterns for each CCJ bone, emphasizing rapid growth during infancy, followed by varying rates of growth and maturation during childhood and adolescence until reaching stability by 18 years of age. The focus was on the closure patterns of synchondroses and sutures in the occipital bone, revealing distinct closure trajectories for the anterior intra-occipital synchondroses and the occipitomastoid suture. The findings, although based on a limited dataset, showcased specific age-related changes in width and closure percentages, providing valuable insights into growth dynamics within the first 2 years of life. Integration analyses revealed intricate relationships between skull and neck structures, emphasizing coordinated growth at different stages. Specific bone covariation patterns, as found between the first and second cervical vertebrae (C1 and C2), indicated synchronized morphological changes. Our results provide initial data for designing inclusive CCJ geometric models to predict normal and abnormal growth dynamics.
Collapse
Affiliation(s)
- Juliette Raoul‐Duval
- Craniofacial Growth and FormHôpital Necker – Enfants Malades, Assistance Publique – Hôpitaux de ParisParisFrance
| | - Angèle Ganet
- Craniofacial Growth and FormHôpital Necker – Enfants Malades, Assistance Publique – Hôpitaux de ParisParisFrance
| | - Sandro Benichi
- Department of Paediatric NeurosurgeryHôpital Necker – Enfants Malades, Assistance Publique – Hôpitaux de ParisParisFrance
- CRMR C‐MAVEM, Filière NeuroSphinx, Hôpital Necker – Enfants Malades, Assistance Publique – Hôpitaux de ParisParisFrance
| | - Pauline Baixe
- Craniofacial Growth and FormHôpital Necker – Enfants Malades, Assistance Publique – Hôpitaux de ParisParisFrance
| | - Clara Cornillon
- Craniofacial Growth and FormHôpital Necker – Enfants Malades, Assistance Publique – Hôpitaux de ParisParisFrance
| | - Lou Eschapasse
- Craniofacial Growth and FormHôpital Necker – Enfants Malades, Assistance Publique – Hôpitaux de ParisParisFrance
| | - Maya Geoffroy
- Craniofacial Growth and FormHôpital Necker – Enfants Malades, Assistance Publique – Hôpitaux de ParisParisFrance
- Institut de Biomécanique Humaine Georges Charpak, Ecole Nationale Supérieure Des Arts et MétiersParisFrance
| | - Giovanna Paternoster
- Department of Paediatric NeurosurgeryHôpital Necker – Enfants Malades, Assistance Publique – Hôpitaux de ParisParisFrance
- CRMR C‐MAVEM, Filière NeuroSphinx, Hôpital Necker – Enfants Malades, Assistance Publique – Hôpitaux de ParisParisFrance
| | - Syril James
- Department of Paediatric NeurosurgeryHôpital Necker – Enfants Malades, Assistance Publique – Hôpitaux de ParisParisFrance
- CRMR C‐MAVEM, Filière NeuroSphinx, Hôpital Necker – Enfants Malades, Assistance Publique – Hôpitaux de ParisParisFrance
| | - Sébastien Laporte
- Institut de Biomécanique Humaine Georges Charpak, Ecole Nationale Supérieure Des Arts et MétiersParisFrance
| | - Thomas Blauwblomme
- Department of Paediatric NeurosurgeryHôpital Necker – Enfants Malades, Assistance Publique – Hôpitaux de ParisParisFrance
| | - Roman H. Khonsari
- Craniofacial Growth and FormHôpital Necker – Enfants Malades, Assistance Publique – Hôpitaux de ParisParisFrance
- Department of Paediatric Maxillofacial Surgery and Plastic SurgeryHôpital Necker – Enfants Malades, Assistance Publique – Hôpitaux de ParisParisFrance
- Faculté de MédecineUniversité Paris CitéParisFrance
- CRMR CRANIOST, Filière TeteCou, Hôpital Necker – Enfants Malades, Assistance Publique – Hôpitaux de ParisParisFrance
| | - Maxime Taverne
- Craniofacial Growth and FormHôpital Necker – Enfants Malades, Assistance Publique – Hôpitaux de ParisParisFrance
| |
Collapse
|
5
|
Rico-Llanos G, Spoutil F, Blahova E, Koudelka A, Prochazkova M, Czyrek A, Fafilek B, Prochazka J, Gonzalez Lopez M, Krivanek J, Sedlacek R, Krakow D, Nonaka Y, Nakamura Y, Krejci P. Achondroplasia: aligning mouse model with human clinical studies shows crucial importance of immediate postnatal start of the therapy. J Bone Miner Res 2024; 39:1783-1792. [PMID: 39423254 PMCID: PMC11638852 DOI: 10.1093/jbmr/zjae173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 10/02/2024] [Accepted: 10/17/2024] [Indexed: 10/21/2024]
Abstract
Achondroplasia is the most common form of human dwarfism caused by mutations in the FGFR3 receptor tyrosine kinase. Current therapy begins at 2 years of age and improves longitudinal growth but does not address the cranial malformations including midface hypoplasia and foramen magnum stenosis, which lead to significant otolaryngeal and neurologic compromise. A recent clinical trial found partial restoration of cranial defects with therapy starting at 3 months of age, but results are still inconclusive. The benefits of achondroplasia therapy are therefore controversial, increasing skepticism among the medical community and patients. We used a mouse model of achondroplasia to test treatment protocols aligned with human studies. Early postnatal treatment (from day 1) was compared with late postnatal treatment (from day 4, equivalent to ~5 months in humans). Animals were treated with the FGFR3 inhibitor infigratinib and the effect on skeleton was thoroughly examined. We show that premature fusion of the skull base synchondroses occurs immediately after birth and leads to defective cranial development and foramen magnum stenosis in the mouse model to achondroplasia. This phenotype appears significantly restored by early infigratinib administration when compared with late treatment, which provides weak to no rescue. In contrast, the long bone growth is similarly improved by both early and late protocols. We provide clear evidence that immediate postnatal therapy is critical for normalization of skeletal growth in both the cranial base and long bones and the prevention of sequelae associated with achondroplasia. We also describe the limitations of early postnatal therapy, providing a paradigm-shifting argument for the development of prenatal therapy for achondroplasia.
Collapse
Affiliation(s)
- Gustavo Rico-Llanos
- Department of Biology, Faculty of Medicine, Masaryk University, CZ-62500 Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital, CZ-65691 Brno, Czech Republic
| | - Frantisek Spoutil
- Czech Center for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, CZ-25250 Vestec, Czech Republic
| | - Eva Blahova
- Department of Biology, Faculty of Medicine, Masaryk University, CZ-62500 Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital, CZ-65691 Brno, Czech Republic
| | - Adolf Koudelka
- Department of Biology, Faculty of Medicine, Masaryk University, CZ-62500 Brno, Czech Republic
| | - Michaela Prochazkova
- Czech Center for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, CZ-25250 Vestec, Czech Republic
| | - Aleksandra Czyrek
- Department of Biology, Faculty of Medicine, Masaryk University, CZ-62500 Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital, CZ-65691 Brno, Czech Republic
| | - Bohumil Fafilek
- Department of Biology, Faculty of Medicine, Masaryk University, CZ-62500 Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital, CZ-65691 Brno, Czech Republic
- Institute of Animal Physiology and Genetics of the of the Czech Academy of Sciences, CZ-60200 Brno, Czech Republic
| | - Jan Prochazka
- Czech Center for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, CZ-25250 Vestec, Czech Republic
| | - Marcos Gonzalez Lopez
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, CZ-62500 Brno, Czech Republic
| | - Jan Krivanek
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, CZ-62500 Brno, Czech Republic
| | - Radislav Sedlacek
- Czech Center for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, CZ-25250 Vestec, Czech Republic
| | - Deborah Krakow
- Department of Orthopaedic Surgery, Human Genetics, and Obstetrics and Gynecology, University of California Los Angeles, Los Angeles, CA 90095, United States
| | | | - Yoshikazu Nakamura
- RIBOMIC Inc., Tokyo 108-0071, Japan
- Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | - Pavel Krejci
- Department of Biology, Faculty of Medicine, Masaryk University, CZ-62500 Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital, CZ-65691 Brno, Czech Republic
- Institute of Animal Physiology and Genetics of the of the Czech Academy of Sciences, CZ-60200 Brno, Czech Republic
| |
Collapse
|
6
|
Zhou H, Li F, Lin Z, Meng L, Chen D, Zhang Q, Niu L. Holographic Ultrasound Modulates Neural Activity in a 1-Methyl-4-Phenyl-1,2,3,6-Tetrahydropyridine-Induced Mouse Model of Parkinson's Disease. RESEARCH (WASHINGTON, D.C.) 2024; 7:0516. [PMID: 39507404 PMCID: PMC11538569 DOI: 10.34133/research.0516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 10/08/2024] [Accepted: 10/08/2024] [Indexed: 11/08/2024]
Abstract
Ultrasound (US) has emerged as a noninvasive neurostimulation method for motor control in Parkinson's disease (PD). Previous in vivo US neuromodulation studies for PD were single-target stimulation. However, the motor symptoms of PD are linked with neural circuit dysfunction, and multi-target stimulation is conducted in clinical treatment for PD. Thus, in the present study, we achieved multi-target US stimulation using holographic lens transducer based on the Rayleigh-Sommerfeld diffraction integral and time-reversal methods. We demonstrated that holographic US stimulation of the bilateral dorsal striatum (DS) could improve the motor function in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced mouse model of PD. The holographic US wave (fundamental frequency: 3 MHz, pulse repetition frequency: 500 Hz, duty cycle: 20%, tone-burst duration: 0.4 ms, sonication duration: 1 s, interstimulus interval: 4 s, spatial-peak temporal-average intensity: 180 mw/cm2) was delivered to the bilateral DS 20 min per day for consecutive 10 d after the last injection of MPTP. Immunohistochemical c-Fos staining demonstrated that holographic US significantly increased the c-Fos-positive neurons in the bilateral DS compared with the sham group (P = 0.003). Moreover, our results suggested that holographic US stimulation of the bilateral DS ameliorated motor dysfunction (P < 0.05) and protected the dopaminergic (DA) neurons (P < 0.001). The neuroprotective effect of holographic US was associated with the prevention of axon degeneration and the reinforcement of postsynaptic densities [growth associated protein-43 (P < 0.001), phosphorylated Akt (P = 0.001), β3-tubulin (P < 0.001), phosphorylated CRMP2 (P = 0.037), postsynaptic density (P = 0.023)]. These data suggested that holographic US-induced acoustic radiation force has the potential to achieve multi-target neuromodulation and could serve as a reliable tool for the treatment of PD.
Collapse
Affiliation(s)
- Hui Zhou
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology,
Chinese Academy of Sciences, Shenzhen, China.
- Tech X Academy,
Shenzhen Polytechnic University, Shenzhen, China
| | - Fei Li
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology,
Chinese Academy of Sciences, Shenzhen, China.
| | - Zhengrong Lin
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology,
Chinese Academy of Sciences, Shenzhen, China.
| | - Long Meng
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology,
Chinese Academy of Sciences, Shenzhen, China.
| | - Dan Chen
- Institute of Ultrasonic Technology, Institute of Intelligent Manufacturing Technology,
Shenzhen Polytechnic University, Shenzhen, China
| | - Qingping Zhang
- School of Electronic and Communication Engineering,
Shenzhen Polytechnic University, Shenzhen, China
| | - Lili Niu
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology,
Chinese Academy of Sciences, Shenzhen, China.
| |
Collapse
|
7
|
Tavitian A, Somech J, Chamlian B, Liberman A, Galindez C, Schipper HM. Craniofacial anomalies in schizophrenia-relevant GFAP.HMOX1 0-12m mice. Anat Rec (Hoboken) 2024; 307:3529-3547. [PMID: 38606671 DOI: 10.1002/ar.25449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 02/29/2024] [Accepted: 03/25/2024] [Indexed: 04/13/2024]
Abstract
Subtle craniofacial dysmorphology has been reported in schizophrenia patients. This dysmorphology includes midline facial elongation, frontonasal anomalies and a sexually dimorphic deviation from normal directional asymmetry of the face, with male patients showing reduced and female patients showing enhanced facial asymmetry relative to healthy control subjects. GFAP.HMOX10-12m transgenic mice (Mus musculus) that overexpress heme oxygenase-1 in astrocytes recapitulate many schizophrenia-relevant neurochemical, neuropathological and behavioral features. As morphogenesis of the brain, skull and face are highly interrelated, we hypothesized that GFAP.HMOX10-12m mice may exhibit craniofacial anomalies similar to those reported in persons with schizophrenia. We examined craniofacial anatomy in male GFAP.HMOX10-12m mice and wild-type control mice at the early adulthood age of 6-8 months. We used computer vision techniques for the extraction and analysis of mouse head shape parameters from systematically acquired 2D digital images, and confirmed our results with landmark-based geometric morphometrics. We performed skull bone morphometry using digital calipers to take linear distance measurements between known landmarks. Relative to controls, adult male GFAP.HMOX10-12m mice manifested craniofacial dysmorphology including elongation of the nasal bones, alteration of head shape anisotropy and reduction of directional asymmetry in facial shape features. These findings demonstrate that GFAP.HMOX10-12m mice exhibit craniofacial anomalies resembling those described in schizophrenia patients, implicating heme oxygenase-1 in their development. As a preclinical mouse model, GFAP.HMOX10-12m mice provide a novel opportunity for the study of the etiopathogenesis of craniofacial and other anomalies in schizophrenia and related disorders.
Collapse
Affiliation(s)
- Ayda Tavitian
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
| | - Joseph Somech
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
| | - Badrouyk Chamlian
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
| | - Adrienne Liberman
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
| | - Carmela Galindez
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
| | - Hyman M Schipper
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
| |
Collapse
|
8
|
Sousa SC, Aroso M, Bessa R, Veríssimo E, Ferreira da Silva T, Lopes CDF, Brites P, Vieira J, Vieira CP, Aguiar PC, Sousa MM. Stretch triggers microtubule stabilization and MARCKS-dependent membrane incorporation in the shaft of embryonic axons. Curr Biol 2024; 34:4577-4588.e8. [PMID: 39265571 DOI: 10.1016/j.cub.2024.08.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 05/28/2024] [Accepted: 08/13/2024] [Indexed: 09/14/2024]
Abstract
Neurons have a unique polarized nature that must adapt to environmental changes throughout their lifespan. During embryonic development, axon elongation is led by the growth cone,1 culminating in the formation of a presynaptic terminal. After synapses are formed, axons elongate in a growth cone-independent manner to accompany body growth while maintaining their ultrastructure and function.2,3,4,5,6 To further understand mechanical strains on the axon shaft, we developed a computer-controlled stretchable microfluidic platform compatible with multi-omics and live imaging. Our data show that sensory embryonic dorsal root ganglia (DRGs) neurons have high plasticity, with axon shaft microtubules decreasing polymerization rates, aligning with the direction of tension, and undergoing stabilization. Moreover, in embryonic DRGs, stretch triggers yes-associated protein (YAP) nuclear translocation, supporting its participation in the regulatory network that enables tension-driven axon growth. Other than cytoskeleton remodeling, stretch prompted MARCKS-dependent formation of plasmalemmal precursor vesicles (PPVs), resulting in new membrane incorporation throughout the axon shaft. In contrast, adolescent DRGs showed a less robust adaptation, with axonal microtubules being less responsive to stretch. Also, while adolescent DRGs were still amenable to strain-induced PPV formation at higher stretch rates, new membrane incorporation in the axon shaft failed to occur. In summary, we developed a new resource to study the biology of axon stretch growth. By unraveling cytoskeleton adaptation and membrane remodeling in the axon shaft of stretched neurons, we are moving forward in understanding axon growth.
Collapse
Affiliation(s)
- Sara C Sousa
- Nerve Regeneration Group, IBMC-Instituto de Biologia Molecular e Celular and i3S - Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, 4200-135 Porto, Portugal; Graduate Program in Molecular and Cell Biology, ICBAS - Instituto de Ciências Biomédicas Abel Salazar, University of Porto, 4050-313 Porto, Portugal
| | - Miguel Aroso
- Neuroengineering and Computational Neuroscience Group, i3S - Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal
| | - Rita Bessa
- Nerve Regeneration Group, IBMC-Instituto de Biologia Molecular e Celular and i3S - Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, 4200-135 Porto, Portugal
| | - Eduardo Veríssimo
- Nerve Regeneration Group, IBMC-Instituto de Biologia Molecular e Celular and i3S - Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, 4200-135 Porto, Portugal; Graduate Program in Molecular and Cell Biology, ICBAS - Instituto de Ciências Biomédicas Abel Salazar, University of Porto, 4050-313 Porto, Portugal
| | - Tiago Ferreira da Silva
- Neurolipid Biology Group, IBMC-Instituto de Biologia Celular e Molecular and i3S - Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal
| | - Cátia D F Lopes
- Neuroengineering and Computational Neuroscience Group, i3S - Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal
| | - Pedro Brites
- Neurolipid Biology Group, IBMC-Instituto de Biologia Celular e Molecular and i3S - Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal
| | - Jorge Vieira
- Phenotypic Evolution Group, IBMC-Instituto de Biologia Molecular e Celular and i3S - Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, 4200-135 Porto, Portugal
| | - Cristina P Vieira
- Phenotypic Evolution Group, IBMC-Instituto de Biologia Molecular e Celular and i3S - Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, 4200-135 Porto, Portugal
| | - Paulo C Aguiar
- Neuroengineering and Computational Neuroscience Group, i3S - Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal
| | - Monica M Sousa
- Nerve Regeneration Group, IBMC-Instituto de Biologia Molecular e Celular and i3S - Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, 4200-135 Porto, Portugal.
| |
Collapse
|
9
|
Adepoju TE, Fisher HB, Winston C, White BR. Intact-skull cranial windows for widefield optical imaging in juvenile mice: complications and consequences. JOURNAL OF THE OPTICAL SOCIETY OF AMERICA. A, OPTICS, IMAGE SCIENCE, AND VISION 2024; 41:1942-1947. [PMID: 39889018 PMCID: PMC11973661 DOI: 10.1364/josaa.531568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 09/04/2024] [Indexed: 02/02/2025]
Abstract
Functional neuroimaging with widefield optical imaging (WOI) is potentially useful for studying developmental disorders in juvenile mice. However, WOI requires an intact-skull cranial window, and the effects of such windows on young mice are unknown. We performed intact-skull cranial window placement on mice as young as P7 to study the effects of chronic placement. Cranial windows placed at young ages (P7 and P10) were not longitudinally stable, resulting in significant attrition. Windows placed at ages P14 or less resulted in significant impairment to skull growth, which in turn caused artifacts in resting-state functional connectivity analysis. Longitudinal cranial windows should likely be avoided under P30.
Collapse
Affiliation(s)
- Temilola E. Adepoju
- Department of Pediatrics, Division of Cardiology, The Children’s Hospital of Philadelphia, and the Perelman School of Medicine at the University of Pennsylvania 3615 Civic Center Blvd, Philadelphia, PA 19104, USA
| | - Hayden B. Fisher
- Department of Pediatrics, Division of Cardiology, The Children’s Hospital of Philadelphia, and the Perelman School of Medicine at the University of Pennsylvania 3615 Civic Center Blvd, Philadelphia, PA 19104, USA
| | - Chloe Winston
- Department of Pediatrics, Division of Cardiology, The Children’s Hospital of Philadelphia, and the Perelman School of Medicine at the University of Pennsylvania 3615 Civic Center Blvd, Philadelphia, PA 19104, USA
| | - Brian R. White
- Department of Pediatrics, Division of Cardiology, The Children’s Hospital of Philadelphia, and the Perelman School of Medicine at the University of Pennsylvania 3615 Civic Center Blvd, Philadelphia, PA 19104, USA
| |
Collapse
|
10
|
Fosséprez J, Roels T, Manicourt D, Behets C. Craniofacial dysmorphism of osteogenesis imperfecta mouse and effect of cathepsin K knockout: Preliminary craniometry observations. Morphologie 2024; 108:100785. [PMID: 38788496 DOI: 10.1016/j.morpho.2024.100785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 04/24/2024] [Accepted: 04/24/2024] [Indexed: 05/26/2024]
Abstract
OBJECTIVES In addition to bone fragility, patients with osteogenesis imperfecta (OI) type III have typical craniofacial abnormalities, such as a triangular face and maxillary micrognathism. However, in the osteogenesis imperfecta mouse (oim), a validated model of OI type III, few descriptions exist of craniofacial phenotype. Treatment of OI mostly consists of bisphosphonate administration. Cathepsin K inhibition has been tested as a promising therapeutic approach for osteoporosis and positive results were observed in long bones of cathepsin K knocked out oim (oim/CatK-/-). This craniometry study aimed to highlight the craniofacial characteristics of oim and Cathepsin K KO mouse. MATERIALS AND METHODS We analyzed the craniofacial skeleton of 51 mice distributed in 4 genotype groups: Wt (control), oim, CatK-/-, oim/CatK-/-. The mice were euthanized at 13 weeks and their heads were analyzed using densitometric (pQCT), X-ray cephalometric, and histomorphometric methods. RESULTS The craniofacial skeleton of the oim mouse is frailer than the Wt one, with a reduced thickness and mineral density of the cranial vault and mandibular ramus. Different cephalometric data attest a dysmorphism similar to the one observed in humans with OI type III. Those abnormalities were not improved in the oim/CatK-/- group. CONCLUSION These results suggest that oim mouse could serve as a complete model of the human OI type III, including the craniofacial skeleton. They also suggest that invalidation of cathepsin K has no impact on the craniofacial abnormalities of the oim model.
Collapse
Affiliation(s)
- J Fosséprez
- Pole of Morphology, institut de recherche expérimentale et clinique (IREC), université catholique de Louvain (UCLouvain), Brussels, Belgium.
| | - T Roels
- Pole of Morphology, institut de recherche expérimentale et clinique (IREC), université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - D Manicourt
- Pole of Rheumatic Diseases, IREC, UCLouvain, Brussels, Belgium
| | - C Behets
- Pole of Morphology, institut de recherche expérimentale et clinique (IREC), université catholique de Louvain (UCLouvain), Brussels, Belgium
| |
Collapse
|
11
|
Shang G, Lei L, Peng C. Bioinformatics Study on Mechanism of Postnatal Development of Craniofacial Bone. J Craniofac Surg 2024; 35:1368-1371. [PMID: 38847500 DOI: 10.1097/scs.0000000000010354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Accepted: 05/04/2024] [Indexed: 07/24/2024] Open
Abstract
OBJECTIVE The postnatal development of craniofacial bone plays a crucial role in shaping the overall structure and functionality of the skull and face. Understanding the underlying mechanisms of this intricate process is essential for both clinical and research purposes. In this study, the authors conducted a bioinformatics analysis using the Gene Expression Omnibus database to investigate the molecular pathways and regulatory networks involved in the postnatal development of craniofacial bone. METHODS In this study, the online Gene Expression Omnibus microarray expression profiling data set GSE27976 was used to identify differentially expressed genes (DEGs) in different age groups. Protein-Protein Interaction network analyses, functional enrichment, and hub genes analysis were performed. The differences in immune infiltration and microenvironment among different types of cells were also analyzed. RESULTS In total, 523 DEGs, including 287 upregulated and 236 downregulated genes, were identified. GO and KEGG analysis showed that the DEGs were significantly enriched in multiple signaling pathways, such as skeletal system morphogenesis, osteoblast differentiation, and stem cell differentiation. Immune infiltration and microenvironment characteristics analysis showed that there were significant differences in fibroblasts, mesenchymal stem cell, osteoblast, stroma score, and microenvironment score between the two groups. Five hub genes, including IGF1, IL1B, ICAM1, MMP2 , and brain-derived neurotrophic factor, were filled out. CONCLUSION The findings of this study showed a significant shift in gene expression towards osteogenesis during the first 12 months after birth. These findings emphasize the critical role of the postnatal period in craniofacial bone development and provide valuable insights into the molecular mechanisms underlying this process.
Collapse
Affiliation(s)
- Guangling Shang
- Department of Liver Transplantation and Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University
| | - Liu Lei
- Department of Burns and Plastic Surgery, The Second Hospital of Shandong University, Shandong University
| | - Changliang Peng
- Department of Spine Surgery, The Second Hospital of Shandong University, Shandong University, Jinan, Shandong Province, People's Republic of China
| |
Collapse
|
12
|
Pöpplau JA, Schwarze T, Dorofeikova M, Pochinok I, Günther A, Marquardt A, Hanganu-Opatz IL. Reorganization of adolescent prefrontal cortex circuitry is required for mouse cognitive maturation. Neuron 2024; 112:421-440.e7. [PMID: 37979584 PMCID: PMC10855252 DOI: 10.1016/j.neuron.2023.10.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 08/31/2023] [Accepted: 10/19/2023] [Indexed: 11/20/2023]
Abstract
Most cognitive functions involving the prefrontal cortex emerge during late development. Increasing evidence links this delayed maturation to the protracted timeline of prefrontal development, which likely does not reach full maturity before the end of adolescence. However, the underlying mechanisms that drive the emergence and fine-tuning of cognitive abilities during adolescence, caused by circuit wiring, are still unknown. Here, we continuously monitored prefrontal activity throughout the postnatal development of mice and showed that an initial activity increase was interrupted by an extensive microglia-mediated breakdown of activity, followed by the rewiring of circuit elements to achieve adult-like patterns and synchrony. Interfering with these processes during adolescence, but not adulthood, led to a long-lasting microglia-induced disruption of prefrontal activity and neuronal morphology and decreased cognitive abilities. These results identified a nonlinear reorganization of prefrontal circuits during adolescence and revealed its importance for adult network function and cognitive processing.
Collapse
Affiliation(s)
- Jastyn A Pöpplau
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology, Hamburg Center of Neuroscience (HCNS), University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - Timo Schwarze
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology, Hamburg Center of Neuroscience (HCNS), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Mariia Dorofeikova
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology, Hamburg Center of Neuroscience (HCNS), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Irina Pochinok
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology, Hamburg Center of Neuroscience (HCNS), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anne Günther
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology, Hamburg Center of Neuroscience (HCNS), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Annette Marquardt
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology, Hamburg Center of Neuroscience (HCNS), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ileana L Hanganu-Opatz
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology, Hamburg Center of Neuroscience (HCNS), University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
13
|
Sung HH, Spresser WJ, Hoffmann JP, Dai Z, Van der Kraan PM, Caird MS, Davidson EB, Kozloff KM. Collagen mutation and age contribute to differential craniofacial phenotypes in mouse models of osteogenesis imperfecta. JBMR Plus 2024; 8:ziad004. [PMID: 38690127 PMCID: PMC11059998 DOI: 10.1093/jbmrpl/ziad004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 10/23/2023] [Accepted: 11/01/2023] [Indexed: 05/02/2024] Open
Abstract
Craniofacial and dentoalveolar abnormalities are present in all types of osteogenesis imperfecta (OI). Mouse models of the disorder are critical to understand these abnormalities and underlying OI pathogenesis. Previous studies on severely affected OI mice report a broad spectrum of craniofacial phenotypes, exhibiting some similarities to the human disorder. The Brtl/+ and G610c/+ are moderately severe and mild-type IV OI, respectively. Little is known about the aging effects on the craniofacial bones of these models and their homology to human OI. This study aimed to analyze the Brtl/+ and G610c/+ craniofacial morphometries during aging to establish suitability for further OI craniofacial bone intervention studies. We performed morphological measurements on the micro-CT-scanned heads of 3-wk-old, 3-mo-old, and 6-mo-old female Brtl/+ and G610c/+ mice. We observed that Brtl/+ skulls are shorter in length than WT (P < .05), whereas G610c/+ skulls are similar in length to their WT counterparts. The Brtl/+ mice exhibit alveolar bone with a porotic-like appearance that is not observed in G610c/+. As they age, Brtl/+ mice show severe bone resorption in both the maxilla and mandible (P < .05). By contrast, G610c/+ mice experience mandibular resorption consistently across all ages, but maxillary resorption is only evident at 6 mo (P < .05). Western blot shows high osteoclastic activities in the Brtl/+ maxilla. Both models exhibit delayed pre-functional eruptions of the third molars (P < .05), which are similar to those observed in some bisphosphonate-treated OI subjects. Our study shows that the Brtl/+ and G610c/+ mice display clear features found in type IV OI patients; both show age-related changes in the craniofacial growth phenotype. Therefore, understanding the craniofacial features of these models and how they age will allow us to select the most accurate mouse model, mouse age, and bone structure for the specific craniofacial bone treatment of differing OI groups.
Collapse
Affiliation(s)
- Hsiao H Sung
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI 48109, United States
- Department of Oral and Maxillofacial Surgery, University of Michigan, Ann Arbor, MI 48109, United States
- Experimental Rheumatology, Department of Rheumatology, Radboud Medical Centre, Nijmegen, The Netherlands, 6525 GA
| | - Wyatt J Spresser
- Department of Oral and Maxillofacial Surgery, University of Michigan, Ann Arbor, MI 48109, United States
| | - Joseph P Hoffmann
- Department of Oral and Maxillofacial Surgery, University of Michigan, Ann Arbor, MI 48109, United States
| | - Zongrui Dai
- Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109, United States
| | - Peter M Van der Kraan
- Experimental Rheumatology, Department of Rheumatology, Radboud Medical Centre, Nijmegen, The Netherlands, 6525 GA
| | - Michelle S Caird
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI 48109, United States
| | - Esmeralda Blaney Davidson
- Experimental Rheumatology, Department of Rheumatology, Radboud Medical Centre, Nijmegen, The Netherlands, 6525 GA
| | - Kenneth M Kozloff
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI 48109, United States
| |
Collapse
|
14
|
Michel ZD, Aitken SF, Glover OD, Alejandro LO, Randazzo D, Dambkowski C, Martin D, Collins MT, Somerman MJ, Chu EY. Infigratinib, a selective FGFR1-3 tyrosine kinase inhibitor, alters dentoalveolar development at high doses. Dev Dyn 2023; 252:1428-1448. [PMID: 37435833 PMCID: PMC10784415 DOI: 10.1002/dvdy.642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 06/14/2023] [Accepted: 06/22/2023] [Indexed: 07/13/2023] Open
Abstract
BACKGROUND Fibroblast growth factor receptor-3 (FGFR3) gain-of-function mutations are linked to achondroplasia. Infigratinib, a FGFR1-3 tyrosine kinase inhibitor, improves skeletal growth in an achondroplasia mouse model. FGFs and their receptors have critical roles in developing teeth, yet effects of infigratinib on tooth development have not been assessed. Dentoalveolar and craniofacial phenotype of Wistar rats dosed with low (0.1 mg/kg) and high (1.0 mg/kg) dose infigratinib were evaluated using micro-computed tomography, histology, and immunohistochemistry. RESULTS Mandibular third molars were reduced in size and exhibited aberrant crown and root morphology in 100% of female rats and 80% of male rats at high doses. FGFR3 and FGF18 immunolocalization and extracellular matrix protein expression were unaffected, but cathepsin K (CTSK) was altered by infigratinib. Cranial vault bones exhibited alterations in dimension, volume, and density that were more pronounced in females. In both sexes, interfrontal sutures were significantly more patent with high dose vs vehicle. CONCLUSIONS High dose infigratinib administered to rats during early stages affects dental and craniofacial development. Changes in CTSK from infigratinib in female rats suggest FGFR roles in bone homeostasis. While dental and craniofacial disruptions are not expected at therapeutic doses, our findings confirm the importance of dental monitoring in clinical studies.
Collapse
Affiliation(s)
- Zachary D Michel
- Skeletal Disorders and Mineral Homeostasis Section, National Institute of Dental and Craniofacial Research (NIDCR), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Sarah F Aitken
- Laboratory of Oral Connective Tissue Biology, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health, Bethesda, Maryland, USA
| | - Omar D Glover
- Laboratory of Oral Connective Tissue Biology, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health, Bethesda, Maryland, USA
| | - Lucy O Alejandro
- Laboratory of Oral Connective Tissue Biology, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health, Bethesda, Maryland, USA
| | - Davide Randazzo
- Light Imaging Section, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | | | - David Martin
- QED Therapeutics, San Francisco, California, USA
| | - Michael T Collins
- Skeletal Disorders and Mineral Homeostasis Section, National Institute of Dental and Craniofacial Research (NIDCR), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Martha J Somerman
- Laboratory of Oral Connective Tissue Biology, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health, Bethesda, Maryland, USA
| | - Emily Y Chu
- Laboratory of Oral Connective Tissue Biology, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health, Bethesda, Maryland, USA
- Department of Comprehensive Dentistry, Division of Cariology and Operative Dentistry, University of Maryland School of Dentistry, Baltimore, Maryland, USA
| |
Collapse
|
15
|
Onwochei-Bolum NV, Kramer B, Hutchinson EF. Taking shape: A geometric morphometric analysis of the immature human palate in relation to dental eruption and growth. Morphologie 2023; 108:100722. [PMID: 39491423 DOI: 10.1016/j.morpho.2023.100722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 08/01/2023] [Accepted: 08/01/2023] [Indexed: 11/05/2024]
Abstract
BACKGROUND The immature human palate functions in suckling and swallowing but transitions to participate in mastication and sound production as the individual grows and matures. Functional alterations manifest as changes in the morphology of the palate and may result from dental eruption. The aim of this study was to analyse the effect of dental eruption on morphological changes of the palate in immature individuals using geometric morphometrics. METHODS Crania from 72 individuals were sourced from the Raymond A. Dart Collection of Human Skeletons, University of the Witwatersrand. Age groups corresponding to the eruption of the deciduous (birth - 5 years), mixed (6-12 years) and permanent dentition (13-20 years) respectively were used. Fourteen osteological landmarks were digitised across the oral surface of the palate and were converted into linear distances and 3D wireframes. Data analysis focused on alterations in shape and size relative to the state of the dentition. RESULTS A significant increase in the geometric mean, length and width was observed between the different states of the dentition. The palate shape was long and narrow in the permanent dentition group, resulting in a more acute angle anteriorly and a protruded orale. A protrusion of the staphylion and deeper palatal dome and change in the horizontal alignment of the alveolar region were also observed in the permanent dentition group. CONCLUSION The subtle changes identified in the growing palate during different stages of the dentition may assist forensic scientists during detailed case analysis.
Collapse
Affiliation(s)
- N V Onwochei-Bolum
- School of Anatomical Sciences, Faculty of Health Sciences, University of the Witwatersrand, 7, York Road, Parktown, 2193 Johannesburg, South Africa
| | - B Kramer
- School of Anatomical Sciences, Faculty of Health Sciences, University of the Witwatersrand, 7, York Road, Parktown, 2193 Johannesburg, South Africa
| | - E F Hutchinson
- School of Anatomical Sciences, Faculty of Health Sciences, University of the Witwatersrand, 7, York Road, Parktown, 2193 Johannesburg, South Africa.
| |
Collapse
|
16
|
Reis CLB, de Fátima Pereira Madureira M, Cunha CLR, Junior WCR, Araújo TH, Esteves A, Stuani MBS, Kirschneck C, Proff P, Matsumoto MAN, Küchler EC, Silva Barroso de Oliveira D. Testosterone suppression impacts craniofacial growth structures during puberty : An animal study. J Orofac Orthop 2023; 84:287-297. [PMID: 35147736 DOI: 10.1007/s00056-021-00373-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 10/10/2021] [Indexed: 10/19/2022]
Abstract
AIM Hormones play a crucial role in growth development; however, the impact of testosterone suppression (TS) on craniofacial growth during puberty remains inconclusive. This study aimed to evaluate the impact of TS during puberty on cephalometric measurements and histological characteristics of facial growth centers. MATERIALS AND METHODS Thirty-six heterogenic Wistar male rats were randomly allocated into experimental orchiectomy (ORX) and control (sham) groups. At an age of 23 days (prepubertal stage), orchiectomy and placebo surgery were performed. Cephalometric measurements were performed via lateral cephalograms during and after puberty. The animals were euthanized at an age of 45 days (pubertal stage) and 73 days (postpubertal stage). Histological slices of the growth centers (condyle, premaxilla, and median palatine suture) were stained with hematoxylin and eosin, and sirius red. Student's t or Mann-Whitney U tests were used to compare linear and angular cephalometric measurements across groups (α error = 5%). RESULTS Linear and angular measurements were statistically different in ORX animals (cranial bones, maxilla, and mandible) at 45 days and 73 days. Condylar histology showed a decrease in prechondroblast differentiation and a delay of mineralization in ORX animals. Vascularization of the medium palatine suture was lower in the ORX group at 45 days. Type I and III collagen fiber synthesis was lower in the ORX groups. In the premaxillary suture, collagen fibers were better organized in the sham groups. CONCLUSIONS Our results suggest that testosterone suppression affects craniofacial growth during puberty.
Collapse
Affiliation(s)
- Caio Luiz Bitencourt Reis
- Department of Pediatric Dentistry, School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
- Department of Clinic and Surgery, School of Dentistry, Federal University of Alfenas, Rua Gabriel Monteiro da Silva, 700, Centro, Alfenas, Minas Gerais, 37130-001, Alfenas, Brazil
| | - Magali de Fátima Pereira Madureira
- Department of Clinic and Surgery, School of Dentistry, Federal University of Alfenas, Rua Gabriel Monteiro da Silva, 700, Centro, Alfenas, Minas Gerais, 37130-001, Alfenas, Brazil
| | - Caio Luis Rocha Cunha
- Department of Clinic and Surgery, School of Dentistry, Federal University of Alfenas, Rua Gabriel Monteiro da Silva, 700, Centro, Alfenas, Minas Gerais, 37130-001, Alfenas, Brazil
| | - Wagner Costa Rossi Junior
- Institute of Biomedical Sciences, Department of Anatomy, Federal University of Alfenas, Alfenas, Brazil
| | - Tomaz Henrique Araújo
- Institute of Biomedical Sciences, Department Structural Biology, Federal University of Alfenas, Alfenas, Brazil
| | - Alessandra Esteves
- Institute of Biomedical Sciences, Department of Anatomy, Federal University of Alfenas, Alfenas, Brazil
| | - Maria Bernadete Sasso Stuani
- Department of Pediatric Dentistry, School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Christian Kirschneck
- Department of Orthodontics, University Medical Centre of Regensburg, Regensburg, Germany
| | - Peter Proff
- Department of Orthodontics, University Medical Centre of Regensburg, Regensburg, Germany
| | - Mírian Aiko Nakane Matsumoto
- Department of Pediatric Dentistry, School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Erika Calvano Küchler
- Department of Pediatric Dentistry, School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
- Department of Orthodontics, University Medical Centre of Regensburg, Regensburg, Germany
| | - Daniela Silva Barroso de Oliveira
- Department of Clinic and Surgery, School of Dentistry, Federal University of Alfenas, Rua Gabriel Monteiro da Silva, 700, Centro, Alfenas, Minas Gerais, 37130-001, Alfenas, Brazil.
| |
Collapse
|
17
|
Gerasco JE, Hathaway‐Schrader JD, Poulides NA, Carson MD, Okhura N, Westwater C, Hatch NE, Novince CM. Commensal Microbiota Effects on Craniofacial Skeletal Growth and Morphology. JBMR Plus 2023; 7:e10775. [PMID: 37614301 PMCID: PMC10443078 DOI: 10.1002/jbm4.10775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 05/07/2023] [Accepted: 05/11/2023] [Indexed: 08/25/2023] Open
Abstract
Microbes colonize anatomical sites in health to form commensal microbial communities (e.g., commensal gut microbiota, commensal skin microbiota, commensal oral microbiota). Commensal microbiota has indirect effects on host growth and maturation through interactions with the host immune system. The commensal microbiota was recently introduced as a novel regulator of skeletal growth and morphology at noncraniofacial sites. Further, we and others have shown that commensal gut microbes, such as segmented filamentous bacteria (SFB), contribute to noncraniofacial skeletal growth and maturation. However, commensal microbiota effects on craniofacial skeletal growth and morphology are unclear. To determine the commensal microbiota's role in craniofacial skeletal growth and morphology, we performed craniometric and bone mineral density analyses on skulls from 9-week-old female C57BL/6T germ-free (GF) mice (no microbes), excluded-flora (EF) specific-pathogen-free mice (commensal microbiota), and murine-pathogen-free (MPF) specific-pathogen-free mice (commensal microbiota with SFB). Investigations comparing EF and GF mice revealed that commensal microbiota impacted the size and shape of the craniofacial skeleton. EF versus GF mice exhibited an elongated gross skull length. Cranial bone length analyses normalized to skull length showed that EF versus GF mice had enhanced frontal bone length and reduced cranial base length. The shortened cranial base in EF mice was attributed to decreased presphenoid, basisphenoid, and basioccipital bone lengths. Investigations comparing MPF mice and EF mice demonstrated that commensal gut microbes played a role in craniofacial skeletal morphology. Cranial bone length analyses normalized to skull length showed that MPF versus EF mice had reduced frontal bone length and increased cranial base length. The elongated cranial base in MPF mice was due to enhanced presphenoid bone length. This work, which introduces the commensal microbiota as a contributor to craniofacial skeletal growth, underscores that noninvasive interventions in the gut microbiome could potentially be employed to modify craniofacial skeletal morphology. © 2023 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Joy E. Gerasco
- Department of Oral Health Sciences, College of Dental MedicineMedical University of South CarolinaCharlestonSCUSA
- Department of Stomatology‐Division of Periodontics, College of Dental MedicineMedical University of South CarolinaCharlestonSCUSA
- Department of Pediatrics‐Division of Endocrinology, College of MedicineMedical University of South CarolinaCharlestonSCUSA
- Department of Orthodontics, Adam's School of DentistryUniversity of North CarolinaChapel HillNCUSA
| | - Jessica D. Hathaway‐Schrader
- Department of Oral Health Sciences, College of Dental MedicineMedical University of South CarolinaCharlestonSCUSA
- Department of Stomatology‐Division of Periodontics, College of Dental MedicineMedical University of South CarolinaCharlestonSCUSA
- Department of Pediatrics‐Division of Endocrinology, College of MedicineMedical University of South CarolinaCharlestonSCUSA
| | - Nicole A. Poulides
- Department of Oral Health Sciences, College of Dental MedicineMedical University of South CarolinaCharlestonSCUSA
- Department of Stomatology‐Division of Periodontics, College of Dental MedicineMedical University of South CarolinaCharlestonSCUSA
- Department of Pediatrics‐Division of Endocrinology, College of MedicineMedical University of South CarolinaCharlestonSCUSA
| | - Matthew D. Carson
- Department of Oral Health Sciences, College of Dental MedicineMedical University of South CarolinaCharlestonSCUSA
- Department of Stomatology‐Division of Periodontics, College of Dental MedicineMedical University of South CarolinaCharlestonSCUSA
- Department of Pediatrics‐Division of Endocrinology, College of MedicineMedical University of South CarolinaCharlestonSCUSA
| | - Naoto Okhura
- Department of Orthodontics and Pediatric Dentistry, School of DentistryUniversity of MichiganAnn ArborMIUSA
| | - Caroline Westwater
- Department of Oral Health Sciences, College of Dental MedicineMedical University of South CarolinaCharlestonSCUSA
- Department of Microbiology and Immunology, College of MedicineMedical University of South CarolinaCharlestonSCUSA
| | - Nan E. Hatch
- Department of Orthodontics and Pediatric Dentistry, School of DentistryUniversity of MichiganAnn ArborMIUSA
| | - Chad M. Novince
- Department of Oral Health Sciences, College of Dental MedicineMedical University of South CarolinaCharlestonSCUSA
- Department of Stomatology‐Division of Periodontics, College of Dental MedicineMedical University of South CarolinaCharlestonSCUSA
- Department of Pediatrics‐Division of Endocrinology, College of MedicineMedical University of South CarolinaCharlestonSCUSA
| |
Collapse
|
18
|
Hoshino Y, Takechi M, Moazen M, Steacy M, Koyabu D, Furutera T, Ninomiya Y, Nuri T, Pauws E, Iseki S. Synchondrosis fusion contributes to the progression of postnatal craniofacial dysmorphology in syndromic craniosynostosis. J Anat 2023; 242:387-401. [PMID: 36394990 PMCID: PMC9919486 DOI: 10.1111/joa.13790] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 09/16/2022] [Accepted: 10/28/2022] [Indexed: 11/18/2022] Open
Abstract
Syndromic craniosynostosis (CS) patients exhibit early, bony fusion of calvarial sutures and cranial synchondroses, resulting in craniofacial dysmorphology. In this study, we chronologically evaluated skull morphology change after abnormal fusion of the sutures and synchondroses in mouse models of syndromic CS for further understanding of the disease. We found fusion of the inter-sphenoid synchondrosis (ISS) in Apert syndrome model mice (Fgfr2S252W/+ ) around 3 weeks old as seen in Crouzon syndrome model mice (Fgfr2cC342Y/+ ). We then examined ontogenic trajectories of CS mouse models after 3 weeks of age using geometric morphometrics analyses. Antero-ventral growth of the face was affected in Fgfr2S252W/+ and Fgfr2cC342Y/+ mice, while Saethre-Chotzen syndrome model mice (Twist1+/- ) did not show the ISS fusion and exhibited a similar growth pattern to that of control littermates. Further analysis revealed that the coronal suture synostosis in the CS mouse models induces only the brachycephalic phenotype as a shared morphological feature. Although previous studies suggest that the fusion of the facial sutures during neonatal period is associated with midface hypoplasia, the present study suggests that the progressive postnatal fusion of the cranial synchondrosis also contributes to craniofacial dysmorphology in mouse models of syndromic CS. These morphological trajectories increase our understanding of the progression of syndromic CS skull growth.
Collapse
Affiliation(s)
- Yukiko Hoshino
- Department of Molecular Craniofacial EmbryologyGraduate School of Medical and Dental SciencesTokyo Medical and Dental University (TMDU)TokyoJapan
- Office of New Drug V, Pharmaceuticals and Medical Devices Agency (PMDA)TokyoJapan
| | - Masaki Takechi
- Department of Molecular Craniofacial EmbryologyGraduate School of Medical and Dental SciencesTokyo Medical and Dental University (TMDU)TokyoJapan
- Department of Anatomy and Life StructureJuntendo University Graduate School of MedicineTokyoJapan
| | - Mehran Moazen
- Department of UCL Mechanical EngineeringUniversity College LondonLondonUK
| | - Miranda Steacy
- Institute of Child Health, Great Ormond StreetUniversity College LondonLondonUK
| | - Daisuke Koyabu
- Department of Molecular Craniofacial EmbryologyGraduate School of Medical and Dental SciencesTokyo Medical and Dental University (TMDU)TokyoJapan
- Research and Development Center for Precision MedicineTsukuba UniversityTsukubaJapan
| | - Toshiko Furutera
- Department of Molecular Craniofacial EmbryologyGraduate School of Medical and Dental SciencesTokyo Medical and Dental University (TMDU)TokyoJapan
- Department of Anatomy and Life StructureJuntendo University Graduate School of MedicineTokyoJapan
| | - Youichirou Ninomiya
- Research Organization of Information and SystemsNational Institute of InformaticsTokyoJapan
| | - Takashi Nuri
- Department of Plastic and Reconstructive SurgeryOsaka Medical and Pharmaceutical UniversityOsakaJapan
| | - Erwin Pauws
- Institute of Child Health, Great Ormond StreetUniversity College LondonLondonUK
| | - Sachiko Iseki
- Department of Molecular Craniofacial EmbryologyGraduate School of Medical and Dental SciencesTokyo Medical and Dental University (TMDU)TokyoJapan
| |
Collapse
|
19
|
Bui AT, Lukashova L, Verdelis K, Vasquez B, Bhogadi L, Gabe CM, Margolis HC, Beniash E. Identification of stages of amelogenesis in the continuously growing mandiblular incisor of C57BL/6J male mice throughout life using molar teeth as landmarks. Front Physiol 2023; 14:1144712. [PMID: 36846326 PMCID: PMC9950101 DOI: 10.3389/fphys.2023.1144712] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Accepted: 01/31/2023] [Indexed: 02/12/2023] Open
Abstract
Continuously growing mouse incisors are widely used to study amelogenesis, since all stages of this process (i.e., secretory, transition and maturation) are present in a spatially determined sequence at any given time. To study biological changes associated with enamel formation, it is important to develop reliable methods for collecting ameloblasts, the cells that regulate enamel formation, from different stages of amelogenesis. Micro-dissection, the key method for collecting distinct ameloblast populations from mouse incisors, relies on positions of molar teeth as landmarks for identifying critical stages of amelogenesis. However, the positions of mandibular incisors and their spatial relationships with molars change with age. Our goal was to identify with high precision these relationships throughout skeletal growth and in older, skeletally mature animals. Mandibles from 2, 4, 8, 12, 16, and 24-week-old, and 18-month-old C57BL/6J male mice, were collected and studied using micro-CT and histology to obtain incisal enamel mineralization profiles and to identify corresponding changes in ameloblast morphology during amelogenesis with respect to positions of molars. As reported here, we have found that throughout active skeletal growth (weeks 2-16) the apices of incisors and the onset of enamel mineralization move distally relative to molar teeth. The position of the transition stage also moves distally. To test the accuracy of the landmarks, we micro-dissected enamel epithelium from mandibular incisors of 12-week-old animals into five segments, including 1) secretory, 2) late secretory - transition - early maturation, 3) early maturation, 4) mid-maturation and 5) late maturation. Isolated segments were pooled and subjected to expression analyses of genes encoding key enamel matrix proteins (EMPs), Amelx, Enam, and Odam, using RT-qPCR. Amelx and Enam were strongly expressed during the secretory stage (segment 1), while their expression diminished during transition (segment 2) and ceased in maturation (segments 3, 4, and 5). In contrast, Odam's expression was very low during secretion and increased dramatically throughout transition and maturation stages. These expression profiles are consistent with the consensus understanding of enamel matrix proteins expression. Overall, our results demonstrate the high accuracy of our landmarking method and emphasize the importance of selecting age-appropriate landmarks for studies of amelogenesis in mouse incisors.
Collapse
Affiliation(s)
- Ai Thu Bui
- Department of Oral and Craniofacial Sciences, University of Pittsburgh School of Dental Medicine (UPSDM), Pittsburgh, PA, United States,Center for Craniofacial Regeneration, UPSDM, Pittsburgh, PA, United States
| | - Lyudmila Lukashova
- Center for Craniofacial Regeneration, UPSDM, Pittsburgh, PA, United States
| | - Kostas Verdelis
- Center for Craniofacial Regeneration, UPSDM, Pittsburgh, PA, United States,Department of Endodontics, UPSDM, Pittsburgh, PA, United States
| | - Brent Vasquez
- Department of Oral and Craniofacial Sciences, University of Pittsburgh School of Dental Medicine (UPSDM), Pittsburgh, PA, United States
| | - Lasya Bhogadi
- Department of Oral and Craniofacial Sciences, University of Pittsburgh School of Dental Medicine (UPSDM), Pittsburgh, PA, United States
| | - Claire M. Gabe
- Department of Oral and Craniofacial Sciences, University of Pittsburgh School of Dental Medicine (UPSDM), Pittsburgh, PA, United States,Center for Craniofacial Regeneration, UPSDM, Pittsburgh, PA, United States
| | - Henry C. Margolis
- Department of Oral and Craniofacial Sciences, University of Pittsburgh School of Dental Medicine (UPSDM), Pittsburgh, PA, United States,Center for Craniofacial Regeneration, UPSDM, Pittsburgh, PA, United States,Department of Periodontics and Preventive Dentistry, UPSDM, Pittsburgh, PA, United States
| | - Elia Beniash
- Department of Oral and Craniofacial Sciences, University of Pittsburgh School of Dental Medicine (UPSDM), Pittsburgh, PA, United States,Center for Craniofacial Regeneration, UPSDM, Pittsburgh, PA, United States,*Correspondence: Elia Beniash,
| |
Collapse
|
20
|
Hallett SA, Zhou A, Herzog C, Arbiv A, Ono W, Ono N. Cranial Base Synchondrosis Lacks PTHrP-Expressing Column-Forming Chondrocytes. Int J Mol Sci 2022; 23:7873. [PMID: 35887221 PMCID: PMC9315528 DOI: 10.3390/ijms23147873] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 07/12/2022] [Accepted: 07/16/2022] [Indexed: 02/04/2023] Open
Abstract
The cranial base contains a special type of growth plate termed the synchondrosis, which functions as the growth center of the skull. The synchondrosis is composed of bidirectional opposite-facing layers of resting, proliferating, and hypertrophic chondrocytes, and lacks the secondary ossification center. In long bones, the resting zone of the epiphyseal growth plate houses a population of parathyroid hormone-related protein (PTHrP)-expressing chondrocytes that contribute to the formation of columnar chondrocytes. Whether PTHrP+ chondrocytes in the synchondrosis possess similar functions remains undefined. Using Pthrp-mCherry knock-in mice, we found that PTHrP+ chondrocytes predominantly occupied the lateral wedge-shaped area of the synchondrosis, unlike those in the femoral growth plate that reside in the resting zone within the epiphysis. In vivo cell-lineage analyses using a tamoxifen-inducible Pthrp-creER line revealed that PTHrP+ chondrocytes failed to establish columnar chondrocytes in the synchondrosis. Therefore, PTHrP+ chondrocytes in the synchondrosis do not possess column-forming capabilities, unlike those in the resting zone of the long bone growth plate. These findings support the importance of the secondary ossification center within the long bone epiphysis in establishing the stem cell niche for PTHrP+ chondrocytes, the absence of which may explain the lack of column-forming capabilities of PTHrP+ chondrocytes in the cranial base synchondrosis.
Collapse
Affiliation(s)
- Shawn A. Hallett
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA; (S.A.H.); (A.Z.); (A.A.)
| | - Annabelle Zhou
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA; (S.A.H.); (A.Z.); (A.A.)
| | - Curtis Herzog
- Department of Oral and Maxillofacial Surgery, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA;
| | - Ariel Arbiv
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA; (S.A.H.); (A.Z.); (A.A.)
| | - Wanida Ono
- Department of Orthodontics, University of Texas Health Science Center at Houston School of Dentistry, Houston, TX 77054, USA;
| | - Noriaki Ono
- Department of Diagnostic and Biomedical Sciences, University of Texas Health Science Center at Houston School of Dentistry, Houston, TX 77054, USA
| |
Collapse
|
21
|
Maruoka H, Hasegawa T, Yoshino H, Abe M, Haraguchi-Kitakamae M, Yamamoto T, Hongo H, Nakanishi K, Nasoori A, Nakajima Y, Omaki M, Sato Y, Luiz de Fraitas PH, Li M. Immunolocalization of endomucin-reactive blood vessels and α-smooth muscle actin-positive cells in murine nasal conchae. J Oral Biosci 2022; 64:337-345. [PMID: 35589073 DOI: 10.1016/j.job.2022.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 05/08/2022] [Accepted: 05/09/2022] [Indexed: 11/30/2022]
Abstract
OBJECTIVES Recently, the biological functions of endomucin-positive blood vessels and closely associated αSMA-positive cells in long bones have been highlighted. The surrounding tissues of the flat bones, such as nasal bones covered with mucosa and lamina propria, are different from those of the long bones, indicating the different distributions of endomucin-positive blood vessels and αSMA-reactive cells in nasal bones. This study demonstrates the immunolocalization of endomucin-reactive blood vessels and αSMA-positive cells in the nasal conchae of 3- and 7-week-old mice. METHODS The nasal conchae of 3-week-old and 7-week-old male C57BL/6J mice were used for immunoreaction of endomucin, CD34, PDGFbb, TRAP, and c-kit. RESULTS While we identified abundant endomucin-reactive blood vessels in the lamina propria neighboring the bone, not all were positive for endomucin. More CD34-reactive cells and small blood vessels were observed in the nasal conchae of 3-week-old mice than in those of 7-week-old mice. Some αSMA-positive cells in the nasal conchae surrounded the blood vessels, indicating vascular smooth muscle cells, while other αSMA-immunopositive fibroblastic cells were detected throughout the lamina propria. αSMA-positive cells did not co-localize with C-kit-immunoreactivity, thereby indicating that the αSMA-positive cells may be myofibroblasts rather than undifferentiated mesenchymal cells. CONCLUSIONS Unlike long bones, nasal conchae contain endomucin-positive as well as endomucin-negative blood vessels and exhibit numerous αSMA-positive fibroblastic cells throughout the lamina propria neighboring the bone. Apparently, the distribution patterns of endomucin-positive blood vessels and αSMA-positive cells in nasal conchae are different from those in long bones.
Collapse
Affiliation(s)
| | | | | | - Miki Abe
- Developmental Biology of Hard Tissue
| | - Mai Haraguchi-Kitakamae
- Developmental Biology of Hard Tissue; Division of Craniofacial Development and Tissue Biology, Graduate School of Dentistry, Tohoku University, Sendai, Japan
| | - Tomomaya Yamamoto
- Developmental Biology of Hard Tissue; Northern Army Medical Unit, Camp Makomanai, Japan Ground Self-Defense Forces, Sapporo, Japan
| | | | | | | | | | | | - Yoshiaki Sato
- Orthodontics, Graduate School of Dental Medicine and Faculty of Dental Medicine, Hokkaido University, Sapporo, Japan
| | | | - Minqi Li
- Shandong Provincial Key Laboratory of Oral Biomedicine, The School of Stomatology, Shandong University, Jinan, China
| |
Collapse
|
22
|
Expression of Cre recombinase in chondrocytes causes abnormal craniofacial and skeletal development. Transgenic Res 2022; 31:399-411. [DOI: 10.1007/s11248-022-00308-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 04/07/2022] [Indexed: 10/18/2022]
|
23
|
Song X, Guo Y, Li H, Chen C, Lee JH, Zhang Y, Schmidt Z, Wang X. Mesoscopic landscape of cortical functions revealed by through-skull wide-field optical imaging in marmoset monkeys. Nat Commun 2022; 13:2238. [PMID: 35474064 PMCID: PMC9042927 DOI: 10.1038/s41467-022-29864-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 04/04/2022] [Indexed: 12/15/2022] Open
Abstract
The primate cerebral cortex is organized into specialized areas representing different modalities and functions along a continuous surface. The functional maps across the cortex, however, are often investigated a single modality at a time (e.g., audition or vision). To advance our understanding of the complex landscape of primate cortical functions, here we develop a polarization-gated wide-field optical imaging method for measuring cortical functions through the un-thinned intact skull in awake marmoset monkeys (Callithrix jacchus), a primate species featuring a smooth cortex. Using this method, adjacent auditory, visual, and somatosensory cortices are noninvasively parcellated in individual subjects with detailed tonotopy, retinotopy, and somatotopy. An additional pure-tone-responsive tonotopic gradient is discovered in auditory cortex and a face-patch sensitive to motion in the lower-center visual field is localized near an auditory region representing frequencies of conspecific vocalizations. This through-skull landscape-mapping approach provides new opportunities for understanding how the primate cortex is organized and coordinated to enable real-world behaviors.
Collapse
Affiliation(s)
- Xindong Song
- grid.21107.350000 0001 2171 9311Laboratory of Auditory Neurophysiology, Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
| | - Yueqi Guo
- grid.21107.350000 0001 2171 9311Laboratory of Auditory Neurophysiology, Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
| | - Hongbo Li
- grid.21107.350000 0001 2171 9311Laboratory of Auditory Neurophysiology, Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
| | - Chenggang Chen
- grid.21107.350000 0001 2171 9311Laboratory of Auditory Neurophysiology, Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
| | - Jong Hoon Lee
- grid.21107.350000 0001 2171 9311Laboratory of Auditory Neurophysiology, Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
| | - Yang Zhang
- grid.21107.350000 0001 2171 9311Laboratory of Auditory Neurophysiology, Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
| | - Zachary Schmidt
- grid.21107.350000 0001 2171 9311Laboratory of Auditory Neurophysiology, Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
| | - Xiaoqin Wang
- grid.21107.350000 0001 2171 9311Laboratory of Auditory Neurophysiology, Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
| |
Collapse
|
24
|
Lesciotto KM, Tomlinson L, Leonard S, Richtsmeier JT. Embryonic and Early Postnatal Cranial Bone Volume and Tissue Mineral Density Values for C57BL/6J Laboratory Mice. Dev Dyn 2022; 251:1196-1208. [PMID: 35092111 PMCID: PMC9250594 DOI: 10.1002/dvdy.458] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 01/18/2022] [Accepted: 01/20/2022] [Indexed: 11/27/2022] Open
Abstract
Background Laboratory mice are routinely used in craniofacial research based on the relatively close genetic relationship and conservation of developmental pathways between humans and mice. Since genetic perturbations and disease states may have localized effects, data from individual cranial bones are valuable for the interpretation of experimental assays. We employ high‐resolution microcomputed tomography to characterize cranial bones of C57BL/6J mice at embryonic day (E) 15.5 and E17.5, day of birth (P0), and postnatal day 7 (P7) and provide estimates of individual bone volume and tissue mineral density (TMD). Results Average volume and TMD values are reported for individual bones. Significant differences in volume and TMD during embryonic ages likely reflect early mineralization of cranial neural crest‐derived and intramembranously forming bones. Although bones of the face and vault had higher TMD values during embryonic ages, bones of the braincase floor had significantly higher TMD values by P7. Conclusions These ontogenetic data on cranial bone volume and TMD serve as a reference standard for future studies using mice bred on a C57BL/6J genetic background. Our findings also highlight the importance of differentiating “control” data from mice that are presented as “unaffected” littermates, particularly when carrying a single copy of a cre‐recombinase gene. Higher average volume and density of cranial neural crest‐derived and intramembranously‐forming bones during embryonic development. Higher average density in bones of the braincase floor during early postnatal development. Ontogenetic data on cranial bone volume and TMD serve as a reference standard for mice bred on a C57BL/6J genetic background.
Collapse
Affiliation(s)
- Kate M Lesciotto
- College of Osteopathic Medicine, Sam Houston State University, Conroe, TX, USA
| | | | - Steven Leonard
- College of Medicine, Drexel University, Philadelphia, PA, USA
| | - Joan T Richtsmeier
- Department of Anthropology, Pennsylvania State University, University Park, PA, USA
| |
Collapse
|
25
|
Hsieh YL, Wei X, Wang Y, Zhang H, Qi S, Xie D, Mishina Y, Mendonça D, Hatch N, Liu F. Chondrocyte Tsc1 controls cranial base bone development by restraining the premature differentiation of synchondroses. Bone 2021; 153:116142. [PMID: 34365025 PMCID: PMC8543925 DOI: 10.1016/j.bone.2021.116142] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 06/30/2021] [Accepted: 08/02/2021] [Indexed: 02/05/2023]
Abstract
Cranial base bones are formed through endochondral ossification. Synchondroses are growth plates located between cranial base bones that facilitate anterior-posterior growth of the skull. Coordinated proliferation and differentiation of chondrocytes in cranial base synchondroses is essential for cranial base bone growth. Herein, we report that constitutive activation of the mechanistic target of rapamycin complex 1 (mTORC1) signaling via Tsc1 (Tuberous sclerosis 1) deletion in chondrocytes causes abnormal skull development with decreased size and rounded shape. In contrast to decreased anterior-posterior growth of the cranial base, mutant mice also exhibited significant expansion of cranial base synchondroses including the intersphenoid synchondrosis (ISS) and the spheno-occipital synchondrosis (SOS). Cranial base synchondrosis expansion in TSC1-deficient mice was accounted for by an expansion of the resting zone due to increased cell number and size without alteration in cell proliferation. Furthermore, our data showed that mTORC1 activity is inhibited in the resting and proliferating zone chondrocytes of wild type mice, and Tsc1 deletion activated mTORC1 signaling of the chondrocytes in the resting zone area. Consequently, the chondrocytes in the resting zone of TSC1-deficient mice acquired characteristics generally attributed to pre-hypertrophic chondrocytes including high mTORC1 activity, increased cell size, and increased expression level of PTH1R (Parathyroid hormone 1 receptor) and IHH (Indian hedgehog). Lastly, treatment with rapamycin, an inhibitor of mTORC1, rescued the abnormality in synchondroses. Our results established an important role for TSC1-mTORC1 signaling in regulating cranial base bone development and showed that chondrocytes in the resting zone of synchondroses are maintained in an mTORC1-inhibitory environment.
Collapse
Affiliation(s)
- Yuan-Lynn Hsieh
- Department of Biologic & Materials Sciences and Prosthodontics, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA
| | - Xiaoxi Wei
- Department of Biologic & Materials Sciences and Prosthodontics, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA; Department of Orthodontics, Hospital of Stomatology Jilin University, Changchun, Jilin 130021, China
| | - Yating Wang
- Department of Biologic & Materials Sciences and Prosthodontics, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA; State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Other Research Platforms & Department of Orthodontics and Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Honghao Zhang
- Department of Biologic & Materials Sciences and Prosthodontics, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA
| | - Shuqun Qi
- Department of Biologic & Materials Sciences and Prosthodontics, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA
| | - Di Xie
- Department of Biologic & Materials Sciences and Prosthodontics, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA
| | - Yuji Mishina
- Department of Biologic & Materials Sciences and Prosthodontics, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA
| | - Daniela Mendonça
- Department of Biologic & Materials Sciences and Prosthodontics, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA
| | - Nan Hatch
- Department of Orthodontics and Pediatric Dentistry, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA
| | - Fei Liu
- Department of Biologic & Materials Sciences and Prosthodontics, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA.
| |
Collapse
|
26
|
Aguirre JI, Castillo EJ, Kimmel DB. Biologic and pathologic aspects of osteocytes in the setting of medication-related osteonecrosis of the jaw (MRONJ). Bone 2021; 153:116168. [PMID: 34487892 PMCID: PMC8478908 DOI: 10.1016/j.bone.2021.116168] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 08/02/2021] [Accepted: 08/31/2021] [Indexed: 02/08/2023]
Abstract
Medication-related osteonecrosis of the jaw (MRONJ) is a potentially severe, debilitating condition affecting patients with cancer and patients with osteoporosis who have been treated with powerful antiresorptives (pARs) or angiogenesis inhibitors (AgIs). Oral risk factors associated with the development of MRONJ include tooth extraction and inflammatory dental disease (e.g., periodontitis, periapical infection). In bone tissues, osteocytes play a bidirectional role in which they not only act as the "receiver" of systemic signals from blood vessels, such as hormones and drugs, or local signals from the mineralized matrix as it is deformed, but they also play a critical role as "transmitter" of signals to the cells that execute bone modeling and remodeling (osteoclasts, osteoblasts and lining cells). When the survival capacity of osteocytes is overwhelmed, they can die. Osteocyte death has been associated with several pathological conditions. Whereas the causes and mechanisms of osteocyte death have been studied in conditions like osteonecrosis of the femoral head (ONFH), few studies of the causes and mechanisms of osteocyte death have been done in MRONJ. The three forms of cell death that affect most of the different cells in the body (apoptosis, autophagy, and necrosis) have been recognized in osteocytes. Notably, necroptosis, a form of regulated cell death with "a necrotic cell death phenotype," has also been identified as a form of cell death in osteocytes under certain pathologic conditions. Improving the understanding of osteocyte death in MRONJ may be critical for preventing disease and developing treatment approaches. In this review, we intend to provide insight into the biology of osteocytes, cell death, in general, and osteocyte death, in particular, and discuss hypothetical mechanisms involved in osteocyte death associated with MRONJ.
Collapse
Affiliation(s)
- J I Aguirre
- Department of Physiological Sciences, University of Florida (UF), Gainesville, FL, United States of America.
| | - E J Castillo
- Department of Physiological Sciences, University of Florida (UF), Gainesville, FL, United States of America.
| | - D B Kimmel
- Department of Physiological Sciences, University of Florida (UF), Gainesville, FL, United States of America
| |
Collapse
|
27
|
Snider TN, Louie KW, Zuzo G, Ruellas ACDO, Solem RC, Cevidanes LHS, Zhang H, Mishina Y. Quantification of three-dimensional morphology of craniofacial mineralized tissue defects in Tgfbr2/Osx-Cre mice. ORAL SCIENCE INTERNATIONAL 2021; 18:193-202. [PMID: 34720652 PMCID: PMC8552916 DOI: 10.1002/osi2.1099] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Craniofacial morphology is affected by the growth, development, and three-dimensional (3D) relationship of mineralized structures including the skull, jaws, and teeth. Despite fulfilling different purposes within this region, cranial bones and tooth dentin are derived from mesenchymal cells that are affected by perturbations within the TGF-β signaling pathway. TGFBR2 encodes a transmembrane receptor that is part of the canonical, SMAD-dependent TGF-β signaling pathway and mutations within this gene are associated with Loeys-Dietz syndrome, a condition which often presents with craniofacial signs including craniosynostosis and cleft palate. To investigate the role of Tgfbr2 in immature, but committed, mineralized tissue forming cells, we analyzed postnatal craniofacial morphology in mice with conditional Tgfbr2 deletion in Osx-expressing cells. Novel application of a 3D shape-based comparative technique revealed that Tgfbr2 in Osx-expressing cells results in impaired postnatal molar root and anterior cranial growth. These findings support those from studies using similar Tgfbr2 conditional knockout models, highlight the anomalous facial and dental regions/structures using tomographic imaging-based techniques, and provide insight into the role of Tgfbr2 during postnatal craniofacial development.
Collapse
Affiliation(s)
- Taylor Nicholas Snider
- Department of Biologic and Materials Sciences & Prosthodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - Ke’ale W. Louie
- Department of Biologic and Materials Sciences & Prosthodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - Gabrielle Zuzo
- Department of Biologic and Materials Sciences & Prosthodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | | | - Richard Christian Solem
- Department of Pediatric and Orthodontic Dentistry, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - Lucia H. S. Cevidanes
- Department of Pediatric and Orthodontic Dentistry, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - Honghao Zhang
- Department of Biologic and Materials Sciences & Prosthodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - Yuji Mishina
- Department of Biologic and Materials Sciences & Prosthodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
28
|
Bone conducted responses in the neonatal rat auditory cortex. Sci Rep 2021; 11:16777. [PMID: 34408208 PMCID: PMC8373948 DOI: 10.1038/s41598-021-96188-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 07/22/2021] [Indexed: 12/14/2022] Open
Abstract
Rats are born deaf and start hearing at the end of the second postnatal week, when the ear canals open and low-intensity sounds start to evoke responses in the auditory cortex. Here, using μECoG electrode arrays and intracortical silicon probe recordings, we found that bone-conducted (BC) sounds evoked biphasic responses in the auditory cortex starting from postnatal day (P) 8. The initial phase of these responses, generated by thalamocortical input, was followed by intracortical propagation within supragranular layers. BC-evoked responses co-localized with the responses evoked by electrical stimulation of the cochlea and the deepest layers of the inferior colliculus prior to onset of low-threshold hearing (P13), as well as with the responses evoked by high-frequency (30 kHz) low-intensity (70 dB) air-conducted sounds after that. Thus, BC signals reach high-frequency processing regions of the auditory cortex well before the onset of low-threshold hearing, reflecting early integrity of the auditory system.
Collapse
|
29
|
Bryant JP, Chandrashekhar V, Cappadona AJ, Lookian PP, Chandrashekhar V, Donahue DR, Munasinghe JB, Kim HJ, Vortmeyer AO, Heiss JD, Zhuang Z, Rosenblum JS. Multimodal Atlas of the Murine Inner Ear: From Embryo to Adult. Front Neurol 2021; 12:699674. [PMID: 34335453 PMCID: PMC8319626 DOI: 10.3389/fneur.2021.699674] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 06/22/2021] [Indexed: 12/02/2022] Open
Abstract
The inner ear is a complex organ housed within the petrous bone of the skull. Its intimate relationship with the brain enables the transmission of auditory and vestibular signals via cranial nerves. Development of this structure from neural crest begins in utero and continues into early adulthood. However, the anatomy of the murine inner ear has only been well-characterized from early embryogenesis to post-natal day 6. Inner ear and skull base development continue into the post-natal period in mice and early adulthood in humans. Traditional methods used to evaluate the inner ear in animal models, such as histologic sectioning or paint-fill and corrosion, cannot visualize this complex anatomy in situ. Further, as the petrous bone ossifies in the postnatal period, these traditional techniques become increasingly difficult. Advances in modern imaging, including high resolution Micro-CT and MRI, now allow for 3D visualization of the in situ anatomy of organs such as the inner ear. Here, we present a longitudinal atlas of the murine inner ear using high resolution ex vivo Micro-CT and MRI.
Collapse
Affiliation(s)
- Jean-Paul Bryant
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Vikram Chandrashekhar
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States.,Center for Imaging Science, Johns Hopkins University, Baltimore, MD, United States
| | - Anthony J Cappadona
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Pashayar P Lookian
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States.,Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | | | - Danielle R Donahue
- Mouse Imaging Facility, National Institute of Neurological Disorders and Stroke, Bethesda, MD, United States
| | - Jeeva B Munasinghe
- Mouse Imaging Facility, National Institute of Neurological Disorders and Stroke, Bethesda, MD, United States
| | - H Jeffrey Kim
- Department of Otolaryngology, Georgetown University School of Medicine, Washington, DC, United States.,Office of Clinical Director, National Institute on Deafness and Other Communication Disorders, Bethesda, MD, United States
| | - Alexander O Vortmeyer
- Department of Pathology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - John D Heiss
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Zhengping Zhuang
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Jared S Rosenblum
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States.,Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
30
|
Küchler EC, de Lara RM, Omori MA, Marañón-Vásquez G, Baratto-Filho F, Nelson-Filho P, Stuani MBS, Blanck-Lubarsch M, Schroeder A, Proff P, Kirschneck C. Effects of estrogen deficiency during puberty on maxillary and mandibular growth and associated gene expression - an μCT study on rats. Head Face Med 2021; 17:14. [PMID: 33888144 PMCID: PMC8061017 DOI: 10.1186/s13005-021-00265-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 04/12/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Estrogen is a well-known and important hormone involved in skeletal homeostasis, which regulates genes involved in bone biology. Some studies support that estrogen is important for craniofacial growth and development. Therefore this in vivo animal study aimed to investigate, whether and in which way low estrogen levels in the prepubertal period affect craniofacial development in the postpubertal stage and to quantify the gene expression of RANK, RANKL and OPG in cranial growth sites in ovariectomized estrogen-deficient rats during puberty. METHODS Control (sham-operated, n = 18) and ovariectomy (OVX, n = 18) surgeries were performed on 21-days-old female Wistar rats. Animals euthanized at an age of 45 days (pubertal stage) were used for gene expression analyses (n = 6 per group) and immunohistochemistry of RANK, RANKL and OPG. Animals euthanized at 63 days of age (post-pubertal stage) were used for craniofacial two-dimensional and three-dimensional craniofacial measurements using μCT imaging (n = 12 per group). RESULTS In the μCT analysis of the mandible and maxilla many statistically significant differences between sham-operated and OVX groups were observed, such as increased maxillary and mandibular bone length in OVX animals (p < 0.05). Condylar volume was also significantly different between groups (p < 0.05). The sham-operated group showed a higher level of RANK expression in the midpalatal suture (p = 0.036) and the RANKL:OPG ratio levels were higher in the OVX group (p = 0.015). CONCLUSIONS Our results suggest that estrogen deficiency during the prepubertal period is associated with alterations in the maxillary and mandibular bone length and condylar growth.
Collapse
Affiliation(s)
- Erika Calvano Küchler
- Department of Orthodontics, University of Regensburg, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany. .,Department of Pediatric Dentistry, School of Dentistry of Ribeirão Preto, University of São Paulo, Avenida do Café, Ribeirão Preto, SP, 14040-904, Brazil.
| | | | - Marjorie Ayumi Omori
- Department of Pediatric Dentistry, School of Dentistry of Ribeirão Preto, University of São Paulo, Avenida do Café, Ribeirão Preto, SP, 14040-904, Brazil
| | - Guido Marañón-Vásquez
- Department of Pediatric Dentistry, School of Dentistry of Ribeirão Preto, University of São Paulo, Avenida do Café, Ribeirão Preto, SP, 14040-904, Brazil
| | - Flares Baratto-Filho
- School of Dentistry, Univille University, R. Paulo Malschitzki, Joinville, SC, 89219-710, Brazil
| | - Paulo Nelson-Filho
- Department of Pediatric Dentistry, School of Dentistry of Ribeirão Preto, University of São Paulo, Avenida do Café, Ribeirão Preto, SP, 14040-904, Brazil
| | - Maria Bernadete Sasso Stuani
- Department of Pediatric Dentistry, School of Dentistry of Ribeirão Preto, University of São Paulo, Avenida do Café, Ribeirão Preto, SP, 14040-904, Brazil
| | - Moritz Blanck-Lubarsch
- Department of Orthodontics, University of Muenster, Albert-Schweitzer-Campus 1, Building W30, 48149, Münster, Germany
| | - Agnes Schroeder
- Department of Orthodontics, University of Regensburg, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany
| | - Peter Proff
- Department of Orthodontics, University of Regensburg, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany
| | - Christian Kirschneck
- Department of Orthodontics, University of Regensburg, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany.
| |
Collapse
|
31
|
Kim BS, Shin HR, Kim HJ, Yoon H, Cho YD, Choi KY, Choi JY, Kim WJ, Ryoo HM. Septal chondrocyte hypertrophy contributes to midface deformity in a mouse model of Apert syndrome. Sci Rep 2021; 11:7979. [PMID: 33846505 PMCID: PMC8041873 DOI: 10.1038/s41598-021-87260-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 03/24/2021] [Indexed: 11/25/2022] Open
Abstract
Midface hypoplasia is a major manifestation of Apert syndrome. However, the tissue component responsible for midface hypoplasia has not been elucidated. We studied mice with a chondrocyte-specific Fgfr2S252W mutation (Col2a1-cre; Fgfr2S252W/+) to investigate the effect of cartilaginous components in midface hypoplasia of Apert syndrome. In Col2a1-cre; Fgfr2S252W/+ mice, skull shape was normal at birth, but hypoplastic phenotypes became evident with age. General dimensional changes of mutant mice were comparable with those of mice with mutations in EIIa-cre; Fgfr2S252W/+, a classic model of Apert syndrome in mice. Col2a1-cre; Fgfr2S252W/+ mice showed some unique facial phenotypes, such as elevated nasion, abnormal fusion of the suture between the premaxilla and the vomer, and decreased perpendicular plate of the ethmoid bone volume, which are related to the development of the nasal septal cartilage. Morphological and histological examination revealed that the presence of increased septal chondrocyte hypertrophy and abnormal thickening of nasal septum is causally related to midface deformities in nasal septum-associated structures. Our results suggest that careful examination and surgical correction of the nasal septal cartilage may improve the prognosis in the surgical treatment of midface hypoplasia and respiratory problems in patients with Apert syndrome.
Collapse
Affiliation(s)
- Bong-Soo Kim
- Department of Molecular Genetics and Dental Pharmacology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, South Korea
| | - Hye-Rim Shin
- Department of Molecular Genetics and Dental Pharmacology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, South Korea
| | - Hyun-Jung Kim
- Department of Molecular Genetics and Dental Pharmacology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, South Korea
| | - Heein Yoon
- Department of Molecular Genetics and Dental Pharmacology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, South Korea
| | - Young-Dan Cho
- Department of Periodontology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, South Korea
| | - Kang-Young Choi
- Department of Plastic and Reconstructive Surgery, School of Medicine, Kyungpook National University, Daegu, South Korea
| | - Je-Yong Choi
- Department of Biochemistry and Cell Biology, Cell and Matrix Research Institute, Skeletal Disease Analysis Center, Korea Mouse Phenotyping Center (KMPC), School of Medicine, Kyungpook National University, Daegu, South Korea
| | - Woo-Jin Kim
- Department of Molecular Genetics and Dental Pharmacology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, South Korea.
| | - Hyun-Mo Ryoo
- Department of Molecular Genetics and Dental Pharmacology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, South Korea.
| |
Collapse
|
32
|
The Skull's Girder: A Brief Review of the Cranial Base. J Dev Biol 2021; 9:jdb9010003. [PMID: 33498686 PMCID: PMC7838769 DOI: 10.3390/jdb9010003] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/14/2021] [Accepted: 01/19/2021] [Indexed: 12/11/2022] Open
Abstract
The cranial base is a multifunctional bony platform within the core of the cranium, spanning rostral to caudal ends. This structure provides support for the brain and skull vault above, serves as a link between the head and the vertebral column below, and seamlessly integrates with the facial skeleton at its rostral end. Unique from the majority of the cranial skeleton, the cranial base develops from a cartilage intermediate-the chondrocranium-through the process of endochondral ossification. Owing to the intimate association of the cranial base with nearly all aspects of the head, congenital birth defects impacting these structures often coincide with anomalies of the cranial base. Despite this critical importance, studies investigating the genetic control of cranial base development and associated disorders lags in comparison to other craniofacial structures. Here, we highlight and review developmental and genetic aspects of the cranial base, including its transition from cartilage to bone, dual embryological origins, and vignettes of transcription factors controlling its formation.
Collapse
|
33
|
Baddam P, Biancardi V, Roth DM, Eaton F, Thereza-Bussolaro C, Mandal R, Wishart DS, Barr A, MacLean J, Flores-Mir C, Pagliardini S, Graf D. Neural crest-specific deletion of Bmp7 leads to midfacial hypoplasia, nasal airway obstruction, and disordered breathing modelling Obstructive Sleep Apnea. Dis Model Mech 2021; 14:dmm.047738. [PMID: 33431521 PMCID: PMC7888714 DOI: 10.1242/dmm.047738] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 12/22/2020] [Indexed: 12/14/2022] Open
Abstract
Pediatric obstructive sleep apnea (OSA), a relatively common sleep-related breathing disorder (SRBD) affecting approximately 1-5% of children, is often caused by anatomical obstruction and/or collapse of the nasal and/or pharyngeal airways. The resulting sleep disruption and intermittent hypoxia lead to various systemic morbidities. Predicting the development of OSA from craniofacial features alone is currently not possible and a controversy remains if upper airway obstruction facilitates reduced midfacial growth or vice-versa. Currently, there is no rodent model that recapitulates both the development of craniofacial abnormalities and upper airway obstruction to address these questions. Here, we describe that mice with a neural crest-specific deletion of Bmp7 (Bmp7ncko) present with shorter, more acute angled cranial base, midfacial hypoplasia, nasal septum deviation, turbinate swelling and branching defects, and nasal airway obstruction. Interestingly, several of these craniofacial features develop after birth during periods of rapid midfacial growth and precede the development of an upper airway obstruction. We identified that in this rodent model, no single feature appeared to predict upper airway obstruction, but the sum of those features resulted in a reduced breathing frequency, apneas and overall reduced oxygen consumption. Metabolomics analysis of serum from peripheral blood identified increased levels of hydroxyproline, a metabolite upregulated under hypoxic conditions. As this model recapitulates many features observed in OSA, it offers unique opportunities for studying how upper airway obstruction affects breathing physiology and leads to systemic morbidities.
Collapse
Affiliation(s)
- Pranidhi Baddam
- School of Dentistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Vivian Biancardi
- Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
- Equal contributions
| | - Daniela M Roth
- School of Dentistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
- Equal contributions
| | - Farah Eaton
- School of Dentistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Claudine Thereza-Bussolaro
- School of Dentistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
- Department of Dentistry, Hospital dos Pinheiros, UNIFASIPE, Sinop, Mato Grosso, Brazil
| | - Rupasri Mandal
- The Metabolomics Innovation Centre, Department of Biological Sciences, Faculty of Science, University of Alberta, Edmonton, AB, Canada
| | - David S Wishart
- The Metabolomics Innovation Centre, Department of Biological Sciences, Faculty of Science, University of Alberta, Edmonton, AB, Canada
| | - Amy Barr
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Joanna MacLean
- Department of Pediatrics and the Women & Children's Health Research Institute, Faculty of Medicine and Dentistry, University of Alberta
- Stollery Children's Hospital; Edmonton, AB, Canada
| | - Carlos Flores-Mir
- School of Dentistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Silvia Pagliardini
- Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Daniel Graf
- School of Dentistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
34
|
Tehrani KF, Koukourakis N, Czarske J, Mortensen LJ. In situ measurement of the isoplanatic patch for imaging through intact bone. JOURNAL OF BIOPHOTONICS 2021; 14:e202000160. [PMID: 32844561 PMCID: PMC10599401 DOI: 10.1002/jbio.202000160] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 08/23/2020] [Accepted: 08/24/2020] [Indexed: 06/11/2023]
Abstract
Wavefront-shaping (WS) enables imaging through scattering tissues like bone, which is important for neuroscience and bone-regeneration research. WS corrects for the optical aberrations at a given depth and field-of-view (FOV) within the sample; the extent of the validity of which is limited to a region known as the isoplanatic patch (IP). Knowing this parameter helps to estimate the number of corrections needed for WS imaging over a given FOV. In this paper, we first present direct transmissive measurement of murine skull IP using digital optical phase conjugation based focusing. Second, we extend our previously reported phase accumulation ray tracing (PART) method to provide in-situ in-silico estimation of IP, called correlative PART (cPART). Our results show an IP range of 1 to 3 μm for mice within an age range of 8 to 14 days old and 1.00 ± 0.25 μm in a 12-week old adult skull. Consistency between the two measurement approaches indicates that cPART can be used to approximate the IP before a WS experiment, which can be used to calculate the number of corrections required within a given field of view.
Collapse
Affiliation(s)
- Kayvan Forouhesh Tehrani
- Regenerative Bioscience Center, Rhodes Center for ADS, University of Georgia, Athens, GA, 30602, USA
| | - Nektarios Koukourakis
- TU Dresden, Chair of Measurement and Sensor System Technique, Helmholtzstr. 18, 01062 Dresden, Germany
- Competence Center for Biomedical Computational Laser Systems (BIOLAS), TU Dresden, Germany
| | - Jürgen Czarske
- TU Dresden, Chair of Measurement and Sensor System Technique, Helmholtzstr. 18, 01062 Dresden, Germany
- Cluster of Excellence Physics of Life, TU Dresden, Germany
- Competence Center for Biomedical Computational Laser Systems (BIOLAS), TU Dresden, Germany
| | - Luke J Mortensen
- Regenerative Bioscience Center, Rhodes Center for ADS, University of Georgia, Athens, GA, 30602, USA
- School of Chemical, Materials and Biomedical Engineering, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
35
|
The role of postnatal estrogen deficiency on cranium dimensions. Clin Oral Investig 2020; 25:3249-3255. [PMID: 33099705 DOI: 10.1007/s00784-020-03655-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 10/20/2020] [Indexed: 10/23/2022]
Abstract
OBJECTIVES The aim of this study was investigate the cranium dimensions of adult female rats, who suffered estrogen deficiency during the prepubertal stage, to assess the impact of estrogen deficiency on craniofacial morphology. MATERIAL AND METHODS Twenty-two female Wistar rats were divided into ovariectomy (OVX) (n = 11) and sham-operated control (n = 11) groups. Bilateral ovariectomy were performed in both groups at 21 days old (prepubertal stage), and rats were euthanized at an age of 63 days (post-pubertal stage). Micro-CT scans were performed with rat skulls, and the cranium morphometric landmark measurements were taken in the dorsal, lateral, and ventral view positions. Differences in measurements between the OVX and sham control groups were assessed using t test with an established alpha error of 5%. RESULTS The measures of the rats' skull showed that the inter-zygomatic arch width and anterior cranial base length were significantly larger in OVX group (p = 0.020 and p = 0.050, respectively), whereas the length of parietal bone was significantly higher in the sham group (p = 0.026). For the remaining measurements no significant differences between groups were detected (p > 0.05). CONCLUSION This study provides evidence that ovariectomized rats had alterations in cranial bone dimensions, demonstrating that estrogens during puberty are important for skull morphology. CLINICAL RELEVANCE To understand the role of estrogen on the postnatal cranium development will impact the clinical diagnose and therapy during childhood and adolescence.
Collapse
|
36
|
Funato N. New Insights Into Cranial Synchondrosis Development: A Mini Review. Front Cell Dev Biol 2020; 8:706. [PMID: 32850826 PMCID: PMC7432265 DOI: 10.3389/fcell.2020.00706] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Accepted: 07/13/2020] [Indexed: 11/24/2022] Open
Abstract
The synchondroses formed via endochondral ossification in the cranial base are an important growth center for the neurocranium. Abnormalities in the synchondroses affect cranial base elongation and the development of adjacent regions, including the craniofacial bones. In the central region of the cranial base, there are two synchondroses present—the intersphenoid synchondrosis and the spheno-occipital synchondrosis. These synchondroses consist of mirror image bipolar growth plates. The cross-talk of several signaling pathways, including the parathyroid hormone-like hormone (PTHLH)/parathyroid hormone-related protein (PTHrP), Indian hedgehog (Ihh), Wnt/β-catenin, and fibroblast growth factor (FGF) pathways, as well as regulation by cilium assembly and the transcription factors encoded by the RUNX2, SIX1, SIX2, SIX4, and TBX1 genes, play critical roles in synchondrosis development. Deletions or activation of these gene products in mice causes the abnormal ossification of cranial synchondrosis and skeletal elements. Gene disruption leads to both similar and markedly different abnormalities in the development of intersphenoid synchondrosis and spheno-occipital synchondrosis, as well as in the phenotypes of synchondroses and skeletal bones. This paper reviews the development of cranial synchondroses, along with its regulation by the signaling pathways and transcription factors, highlighting the differences between intersphenoid synchondrosis and spheno-occipital synchondrosis.
Collapse
Affiliation(s)
- Noriko Funato
- Department of Signal Gene Regulation, Tokyo Medical and Dental University, Tokyo, Japan.,Research Core, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
37
|
Kim B, Shin H, Kim W, Kim H, Cho Y, Yoon H, Baek J, Woo K, Lee Y, Ryoo H. PIN1 Attenuation Improves Midface Hypoplasia in a Mouse Model of Apert Syndrome. J Dent Res 2019; 99:223-232. [PMID: 31869252 DOI: 10.1177/0022034519893656] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Premature fusion of the cranial suture and midface hypoplasia are common features of syndromic craniosynostosis caused by mutations in the FGFR2 gene. The only treatment for this condition involves a series of risky surgical procedures designed to correct defects in the craniofacial bones, which must be performed until brain growth has been completed. Several pharmacologic interventions directed at FGFR2 downstream signaling have been tested as potential treatments for premature coronal suture fusion in a mouse model of Apert syndrome. However, there are no published studies that have targeted for the pharmacologic treatment of midface hypoplasia. We used Fgfr2S252W/+ knock-in mice as a model of Apert syndrome and morphometric analyses to identify causal hypoplastic sites in the midface region. Three-dimensional geometric and linear analyses of Fgfr2S252W/+ mice at postnatal day 0 demonstrated distinct morphologic variance. The premature fusion of anterior facial bones, such as the maxilla, nasal, and frontal bones, rather than the cranium or cranial base, is the main contributing factor toward the anterior-posterior skull length shortening. The cranial base of the mouse model had a noticeable downward slant around the intersphenoid synchondrosis, which is related to distortion of the airway. Within a skull, the facial shape variance was highly correlated with the cranial base angle change along Fgfr2 S252W mutation-induced craniofacial anomalies. The inhibition of an FGFR2 downstream signaling enzyme, PIN1, via genetic knockdown or use of a PIN1 inhibitor, juglone, attenuated the aforementioned deformities in a mouse model of Apert syndrome. Overall, these results indicate that FGFR2 signaling is a key contributor toward abnormal anterior-posterior dimensional growth in the midface region. Our study suggests a novel therapeutic option for the prevention of craniofacial malformations induced by mutations in the FGFR2 gene.
Collapse
Affiliation(s)
- B Kim
- Department of Molecular Genetics and Dental Pharmacology, Seoul National University, Seoul, Republic of Korea
| | - H Shin
- Department of Molecular Genetics and Dental Pharmacology, Seoul National University, Seoul, Republic of Korea
| | - W Kim
- Department of Molecular Genetics and Dental Pharmacology, Seoul National University, Seoul, Republic of Korea
| | - H Kim
- Department of Molecular Genetics and Dental Pharmacology, Seoul National University, Seoul, Republic of Korea
| | - Y Cho
- Department of Molecular Genetics and Dental Pharmacology, Seoul National University, Seoul, Republic of Korea.,Department of Periodontology, School of Dentistry and Dental Research Institute, BK21 Program, Seoul National University, Seoul, Republic of Korea
| | - H Yoon
- Department of Molecular Genetics and Dental Pharmacology, Seoul National University, Seoul, Republic of Korea
| | - J Baek
- Department of Molecular Genetics and Dental Pharmacology, Seoul National University, Seoul, Republic of Korea
| | - K Woo
- Department of Molecular Genetics and Dental Pharmacology, Seoul National University, Seoul, Republic of Korea
| | - Y Lee
- Department of Molecular Genetics and Dental Pharmacology, Seoul National University, Seoul, Republic of Korea
| | - H Ryoo
- Department of Molecular Genetics and Dental Pharmacology, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
38
|
Thomas N, Choi HK, Wei X, Wang L, Mishina Y, Guan JL, Liu F. Autophagy Regulates Craniofacial Bone Acquisition. Calcif Tissue Int 2019; 105:518-530. [PMID: 31372669 PMCID: PMC6801085 DOI: 10.1007/s00223-019-00593-2] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 07/26/2019] [Indexed: 10/26/2022]
Abstract
Increasing evidence has demonstrated the important role of autophagy in skeletal homeostasis; however, the role of autophagy in craniofacial bone development and acquisition is largely unknown. In this study, we investigated the effect of autophagy suppression on craniofacial bone acquisition by deleting Fip200 or Atg5, two essential autophagy genes, using Osterix-Cre (Osx-Cre). We found that the Osx-Cre transgene mildly decreased the bone mass of parietal bone but not frontal bone, and did not affect cranial base bone mass in adult mice. In the cranial vault, Fip200 or Atg5 deletion similarly decreased 50% bone mass of neural crest-derived frontal bone; Atg5 deletion decreased 50% and Fip200 deletion decreased 30% bone mass of mesoderm-derived parietal bone. In the cranial base, Fip200 or Atg5 deletion similarly decreased 30% bone mass of neural crest-derived presphenoid bone; Atg5 deletion decreased 30% and Fip200 deletion decreased 16% bone mass of mesoderm-derive basioccipital bone. Lastly, we used doxycycline treatment to inhibit the Osx-Cre expression until 2 months of age and showed that postnatal Fip200 deletion led to cranial vault bone mass decrease in association with a small increase in both bone volume/tissue volume and tissue mineral density. Altogether, this study demonstrated the important role of autophagy in craniofacial bone acquisition during development and postnatal growth.
Collapse
Affiliation(s)
- Neil Thomas
- Department of Biologic and Materials Sciences & Prosthodontics, University of Michigan School of Dentistry, Ann Arbor, MI, 48109, USA
| | - Han Kyoung Choi
- Department of Biologic and Materials Sciences & Prosthodontics, University of Michigan School of Dentistry, Ann Arbor, MI, 48109, USA
| | - Xiaoxi Wei
- Department of Biologic and Materials Sciences & Prosthodontics, University of Michigan School of Dentistry, Ann Arbor, MI, 48109, USA
- Department of Orthodontics, Jilin University School and Hospital of Stomatology, Changchun, 130021, Jilin, China
| | - Li Wang
- Department of Biologic and Materials Sciences & Prosthodontics, University of Michigan School of Dentistry, Ann Arbor, MI, 48109, USA
| | - Yuji Mishina
- Department of Biologic and Materials Sciences & Prosthodontics, University of Michigan School of Dentistry, Ann Arbor, MI, 48109, USA
| | - Jun-Lin Guan
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - Fei Liu
- Department of Biologic and Materials Sciences & Prosthodontics, University of Michigan School of Dentistry, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
39
|
Kulkarni AK, Louie KW, Yatabe M, Ruellas ACDO, Mochida Y, Cevidanes LHS, Mishina Y, Zhang H. A Ciliary Protein EVC2/LIMBIN Plays a Critical Role in the Skull Base for Mid-Facial Development. Front Physiol 2018; 9:1484. [PMID: 30410447 PMCID: PMC6210651 DOI: 10.3389/fphys.2018.01484] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 10/01/2018] [Indexed: 11/26/2022] Open
Abstract
Ellis-van Creveld (EvC) syndrome is an autosomal recessive chondrodysplastic disorder. Affected patients present a wide spectrum of symptoms including short stature, postaxial polydactyly, and dental abnormalities. We previously disrupted Evc2, one of the causative genes for EvC syndrome, in mice using a neural crest-specific, Cre-mediated approach (i.e., P0-Cre, referred to as Evc2 P0 mutants). Despite the fact that P0-Cre predominantly targets the mid-facial region, we reported that many mid-facial defects identified in Evc2 global mutants are not present in Evc2 P0 mutants at postnatal day 8 (P8). In the current study, we used multiple Cre lines (P0-Cre and Wnt1-Cre, respectively), to specifically delete Evc2 in neural crest-derived tissues and compared the resulting mid-facial defects at multiple time points (P8 and P28, respectively). While both Cre lines indistinguishably targeted the mid-facial region, they differentially targeted the anterior portion of the skull base. By comprehensively analyzing the shapes of conditional mutant skulls, we detected differentially affected mid-facial defects in Evc2 P0 mutants and Evc2 Wnt1 mutants. Micro-CT analysis of the skull base further revealed that the Evc2 mutation leads to a differentially affected skull base, caused by premature closure of the intersphenoid synchondrosis (presphenoidal synchondrosis), which limited the elongation of the anterior skull base during the postnatal development of the skull. Given the importance of the skull base in mid-facial bone development, our results suggest that loss of function of Evc2 within the skull base secondarily leads to many aspects of the mid-facial defects developed by the EvC syndrome.
Collapse
Affiliation(s)
- Anshul K Kulkarni
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, Ann Arbor, MI, United States
| | - Ke'ale W Louie
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, Ann Arbor, MI, United States
| | - Marilia Yatabe
- Department of Orthodontics and Pediatric Dentistry, School of Dentistry, University of Michigan, Ann Arbor, MI, United States
| | | | - Yoshiyuki Mochida
- Department of Molecular and Cell Biology, Henry M. Goldman School of Dental Medicine, Boston University, Boston, MA, United States
| | - Lucia H S Cevidanes
- Department of Orthodontics and Pediatric Dentistry, School of Dentistry, University of Michigan, Ann Arbor, MI, United States
| | - Yuji Mishina
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, Ann Arbor, MI, United States
| | - Honghao Zhang
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
40
|
Mid-facial developmental defects caused by the widely used LacZ reporter gene when expressed in neural crest-derived cells. Transgenic Res 2018; 27:551-558. [PMID: 30136095 DOI: 10.1007/s11248-018-0091-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 08/17/2018] [Indexed: 10/28/2022]
Abstract
Reporter genes play important roles in transgenic research. LacZ is a widely used reporter gene that encodes Escherichia coli β-galactosidase, an enzyme that is well known for its ability to hydrolyze X-gal into a blue product. It is unknown whether transgenic LacZ has any adverse effects. R26R reporter mice, containing a LacZ reporter gene, were generated to monitor the in vivo recombination activity of various transgenic Cre recombinase via X-gal staining. P0-Cre is expressed in neural crest-derived cells, which give rise to the majority of the craniofacial bones. Herein, we report that 12% of the R26R reporter mice harboring P0-Cre had unexpected mid-facial developmental defects manifested by the asymmetrical growth of some facial bones, thus resulting in tilted mid-facial structure, shorter skull length, and malocclusion. Histological examination showed a disorganization of the frontomaxillary suture, which may at least partly explain the morphological defect in affected transgenic mice. Our data calls for the consideration of the potential in vivo adverse effects caused by transgenic β-galactosidase.
Collapse
|