1
|
Hu S, Chen S, Zeng H, Ruan X, Lin X, Vlashi R, Zhou C, Wang H, Chen G. Ap-2β regulates cranial osteogenic potential via the activation of Wnt/β-catenin signaling pathway. Dev Biol 2023; 501:S0012-1606(23)00114-8. [PMID: 37355029 DOI: 10.1016/j.ydbio.2023.06.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 05/29/2023] [Accepted: 06/21/2023] [Indexed: 06/26/2023]
Abstract
The skull is a fundamental bone that protects the development of brain and consists of several bony elements, such as the frontal and parietal bones. Frontal bone exhibited superior in osteogenic potential and regeneration of cranial defects compared to parietal bone. However, how this regional difference is regulated remains largely unknown. In this study, we identified an Ap-2β transcriptional factor with a higher expression in frontal bone, but its molecular function in osteoblasts needs to be elucidated. We found that Ap-2β knockdown in preosteoblasts leads to reduced proliferation, increased cell death and impaired differentiation. Through RNA-seq analysis, we found that Ap-2β influences multiple signaling pathways including the Wnt pathway, and overexpression of Ap-2β showed increased nuclear β-catenin and its target genes expressions in osteoblasts. Pharmacological activation of Wnt/β-catenin signaling using LiCl treatment cannot rescue the reduced luciferase activities of the β-catenin/TCF/LEF reporter in Ap-2β knockdown preosteoblasts. Besides, transient expression of Ap-2β via the lentivirus system could sufficiently rescue the inferior osteogenic potential in parietal osteoblasts, while Ap-2β knockdown in frontal osteoblasts resulted in reduced osteoblast activity, reduced active β-catenin and target genes expressions. Taken together, our data demonstrated that Ap-2β modulates osteoblast proliferation and differentiation through the regulation of Wnt/β-catenin signaling pathway and plays an important role in regulating regional osteogenic potential in frontal and parietal bone.
Collapse
Affiliation(s)
- Sujie Hu
- Department of Biopharmacy, College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Sisi Chen
- Department of Biopharmacy, College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Haozu Zeng
- Department of Biopharmacy, College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Xinyi Ruan
- Department of Biopharmacy, College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Xinyi Lin
- Department of Biopharmacy, College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Rexhina Vlashi
- Department of Biopharmacy, College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Chenhe Zhou
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China
| | - Haidong Wang
- Department of Orthopedics, Jiaxing Hospital of Traditional Chinese Medicine, Jiaxing, 314001, China.
| | - Guiqian Chen
- Department of Biopharmacy, College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China.
| |
Collapse
|
2
|
Kemppainen AV, Finnilä MA, Heikkinen A, Härönen H, Izzi V, Kauppinen S, Saarakkala S, Pihlajaniemi T, Koivunen J. The CMS19 disease model specifies a pivotal role for collagen XIII in bone homeostasis. Sci Rep 2022; 12:5866. [PMID: 35393492 PMCID: PMC8990013 DOI: 10.1038/s41598-022-09653-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 03/21/2022] [Indexed: 11/13/2022] Open
Abstract
Mutations in the COL13A1 gene result in congenital myasthenic syndrome type 19 (CMS19), a disease of neuromuscular synapses and including various skeletal manifestations, particularly facial dysmorphisms. The phenotypic consequences in Col13a1 null mice (Col13a1−/−) recapitulate the muscle findings of the CMS19 patients. Collagen XIII (ColXIII) is exists as two forms, a transmembrane protein and a soluble molecule. While the Col13a1−/− mice have poorly formed neuromuscular junctions, the prevention of shedding of the ColXIII ectodomain in the Col13a1tm/tm mice results in acetylcholine receptor clusters of increased size and complexity. In view of the bone abnormalities in CMS19, we here studied the tubular and calvarial bone morphology of the Col13a1−/− mice. We discovered several craniofacial malformations, albeit less pronounced ones than in the human disease, and a reduction of cortical bone mass in aged mice. In the Col13a1tm/tm mice, where ColXIII is synthesized but the ectodomain shedding is prevented due to a mutation in a protease recognition sequence, the cortical bone mass decreased as well with age and the cephalometric analyses revealed significant craniofacial abnormalities but no clear phenotypical pattern. To conclude, our data indicates an intrinsic role for ColXIII, particularly the soluble form, in the upkeep of bone with aging and suggests the possibility of previously undiscovered bone pathologies in patients with CMS19.
Collapse
Affiliation(s)
- A V Kemppainen
- ECM-Hypoxia Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, P.O. Box 5400, 90014, Oulu, Finland
| | - M A Finnilä
- Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, P.O. Box 5000, 90014, Oulu, Finland
| | - A Heikkinen
- ECM-Hypoxia Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, P.O. Box 5400, 90014, Oulu, Finland
| | - H Härönen
- ECM-Hypoxia Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, P.O. Box 5400, 90014, Oulu, Finland
| | - V Izzi
- ECM-Hypoxia Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, P.O. Box 5400, 90014, Oulu, Finland.,Faculty of Medicine, University of Oulu, 90014, Oulu, Finland.,Foundation for the Finnish Cancer Institute, Tukholmankatu 8, 00130, Helsinki, Finland
| | - S Kauppinen
- Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, P.O. Box 5000, 90014, Oulu, Finland
| | - S Saarakkala
- Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, P.O. Box 5000, 90014, Oulu, Finland.,Department of Diagnostic Radiology, Oulu University Hospital, Oulu, Finland
| | - T Pihlajaniemi
- ECM-Hypoxia Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, P.O. Box 5400, 90014, Oulu, Finland
| | - J Koivunen
- ECM-Hypoxia Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, P.O. Box 5400, 90014, Oulu, Finland.
| |
Collapse
|
3
|
Dong L, Lu D, Chen R, Lin Y, Zhu H, Zhang Z, Cai S, Cui P, Song G, Rao D, Yi X, Wu Y, Song N, Liu F, Zou Y, Zhang S, Zhang X, Wang X, Qiu S, Zhou J, Wang S, Zhang X, Shi Y, Figeys D, Ding L, Wang P, Zhang B, Rodriguez H, Gao Q, Gao D, Zhou H, Fan J. Proteogenomic characterization identifies clinically relevant subgroups of intrahepatic cholangiocarcinoma. Cancer Cell 2022; 40:70-87.e15. [PMID: 34971568 DOI: 10.1016/j.ccell.2021.12.006] [Citation(s) in RCA: 149] [Impact Index Per Article: 49.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 07/19/2021] [Accepted: 12/08/2021] [Indexed: 02/08/2023]
Abstract
We performed proteogenomic characterization of intrahepatic cholangiocarcinoma (iCCA) using paired tumor and adjacent liver tissues from 262 patients. Integrated proteogenomic analyses prioritized genetic aberrations and revealed hallmarks of iCCA pathogenesis. Aflatoxin signature was associated with tumor initiation, proliferation, and immune suppression. Mutation-associated signaling profiles revealed that TP53 and KRAS co-mutations may contribute to iCCA metastasis via the integrin-FAK-SRC pathway. FGFR2 fusions activated the Rho GTPase pathway and could be a potential source of neoantigens. Proteomic profiling identified four patient subgroups (S1-S4) with subgroup-specific biomarkers. These proteomic subgroups had distinct features in prognosis, genetic alterations, microenvironment dysregulation, tumor microbiota composition, and potential therapeutics. SLC16A3 and HKDC1 were further identified as potential prognostic biomarkers associated with metabolic reprogramming of iCCA cells. This study provides a valuable resource for researchers and clinicians to further identify molecular pathogenesis and therapeutic opportunities in iCCA.
Collapse
Affiliation(s)
- Liangqing Dong
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai 200032, China
| | - Dayun Lu
- Department of Analytical Chemistry and CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China; University of Chinese Academy of Sciences, Number 19A Yuquan Road, Beijing 100049, China
| | - Ran Chen
- University of Chinese Academy of Sciences, Number 19A Yuquan Road, Beijing 100049, China; State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Youpei Lin
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai 200032, China
| | - Hongwen Zhu
- Department of Analytical Chemistry and CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China
| | - Zhou Zhang
- Burning Rock Biotech, Shanghai 201114, China
| | - Shangli Cai
- Burning Rock Biotech, Shanghai 201114, China
| | - Peng Cui
- Burning Rock Biotech, Shanghai 201114, China
| | - Guohe Song
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai 200032, China
| | - Dongning Rao
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai 200032, China
| | - Xinpei Yi
- Lester and Sue Smith Breast Center, Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Yingcheng Wu
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai 200032, China
| | - Nixue Song
- Department of Analytical Chemistry and CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China; University of Chinese Academy of Sciences, Number 19A Yuquan Road, Beijing 100049, China
| | - Fen Liu
- University of Chinese Academy of Sciences, Number 19A Yuquan Road, Beijing 100049, China; State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Yunhao Zou
- University of Chinese Academy of Sciences, Number 19A Yuquan Road, Beijing 100049, China; State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Shu Zhang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai 200032, China
| | - Xiaoming Zhang
- Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Xiaoying Wang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai 200032, China
| | - Shuangjian Qiu
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai 200032, China
| | - Jian Zhou
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai 200032, China; Key Laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Shisheng Wang
- Frontiers Science Center for Disease-related Molecular Network, Institutes for Systems Genetics, Key Lab of Transplant Engineering and Immunology, MOH, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xu Zhang
- Ottawa Institute of Systems Biology, Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada; Shanghai Institute of Materia Medica-University of Ottawa Joint Research Center in Systems and Personalized Pharmacology, 555 Zuchongzhi Road, Shanghai 201203, China
| | - Yongyong Shi
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), The Collaborative Innovation Center for Brain Science, Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, China
| | - Daniel Figeys
- Ottawa Institute of Systems Biology, Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada; Shanghai Institute of Materia Medica-University of Ottawa Joint Research Center in Systems and Personalized Pharmacology, 555 Zuchongzhi Road, Shanghai 201203, China
| | - Li Ding
- Department of Medicine, McDonnell Genome Institute, Siteman Cancer Center, Washington University, St. Louis, MI 63108, USA
| | - Pei Wang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, NewYork, NY 10029, USA
| | - Bing Zhang
- Lester and Sue Smith Breast Center, Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Henry Rodriguez
- Office of Cancer Clinical Proteomics Research, Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Qiang Gao
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai 200032, China; Key Laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China; State Key Laboratory of Genetic Engineering, Fudan University, Shanghai 200433, China.
| | - Daming Gao
- University of Chinese Academy of Sciences, Number 19A Yuquan Road, Beijing 100049, China; State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China; School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China.
| | - Hu Zhou
- Department of Analytical Chemistry and CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China; University of Chinese Academy of Sciences, Number 19A Yuquan Road, Beijing 100049, China; Shanghai Institute of Materia Medica-University of Ottawa Joint Research Center in Systems and Personalized Pharmacology, 555 Zuchongzhi Road, Shanghai 201203, China.
| | - Jia Fan
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai 200032, China; Key Laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China.
| |
Collapse
|
4
|
Oss-Ronen L, Cohen I. Epigenetic regulation and signalling pathways in Merkel cell development. Exp Dermatol 2021; 30:1051-1064. [PMID: 34152646 DOI: 10.1111/exd.14415] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 06/15/2021] [Accepted: 06/17/2021] [Indexed: 12/20/2022]
Abstract
Merkel cells are specialized epithelial cells connected to afferent nerve endings responsible for light-touch sensations, formed at specific locations in touch-sensitive regions of the mammalian skin. Although Merkel cells are descendants of the epidermal lineage, little is known about the mechanisms responsible for the development of these unique mechanosensory cells. Recent studies have highlighted that the Polycomb group (PcG) of proteins play a significant role in spatiotemporal regulation of Merkel cell formation. In addition, several of the major signalling pathways involved in skin development have been shown to regulate Merkel cell development as well. Here, we summarize the current understandings of the role of developmental regulators in Merkel cell formation, including the interplay between the epigenetic machinery and key signalling pathways, and the lineage-specific transcription factors involved in the regulation of Merkel cell development.
Collapse
Affiliation(s)
- Liat Oss-Ronen
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Science, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Idan Cohen
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Science, Ben-Gurion University of the Negev, Beer Sheva, Israel
| |
Collapse
|
5
|
p38-MAPK-mediated translation regulation during early blastocyst development is required for primitive endoderm differentiation in mice. Commun Biol 2021; 4:788. [PMID: 34172827 PMCID: PMC8233355 DOI: 10.1038/s42003-021-02290-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 06/02/2021] [Indexed: 02/07/2023] Open
Abstract
Successful specification of the two mouse blastocyst inner cell mass (ICM) lineages (the primitive endoderm (PrE) and epiblast) is a prerequisite for continued development and requires active fibroblast growth factor 4 (FGF4) signaling. Previously, we identified a role for p38 mitogen-activated protein kinases (p38-MAPKs) during PrE differentiation, but the underlying mechanisms have remained unresolved. Here, we report an early blastocyst window of p38-MAPK activity that is required to regulate ribosome-related gene expression, rRNA precursor processing, polysome formation and protein translation. We show that p38-MAPK inhibition-induced PrE phenotypes can be partially rescued by activating the translational regulator mTOR. However, similar PrE phenotypes associated with extracellular signal-regulated kinase (ERK) pathway inhibition targeting active FGF4 signaling are not affected by mTOR activation. These data indicate a specific role for p38-MAPKs in providing a permissive translational environment during mouse blastocyst PrE differentiation that is distinct from classically reported FGF4-based mechanisms.
Collapse
|
6
|
Epstein RJ, Tian LJ, Gu YF. 2b or Not 2b: How Opposing FGF Receptor Splice Variants Are Blocking Progress in Precision Oncology. JOURNAL OF ONCOLOGY 2021; 2021:9955456. [PMID: 34007277 PMCID: PMC8110382 DOI: 10.1155/2021/9955456] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 04/21/2021] [Indexed: 01/16/2023]
Abstract
More than ten thousand peer-reviewed studies have assessed the role of fibroblast growth factors (FGFs) and their receptors (FGFRs) in cancer, but few patients have yet benefited from drugs targeting this molecular family. Strategizing how best to use FGFR-targeted drugs is complicated by multiple variables, including RNA splicing events that alter the affinity of ligands for FGFRs and hence change the outcomes of stromal-epithelial interactions. The effects of splicing are most relevant to FGFR2; expression of the FGFR2b splice isoform can restore apoptotic sensitivity to cancer cells, whereas switching to FGFR2c may drive tumor progression by triggering epithelial-mesenchymal transition. The differentiating and regulatory actions of wild-type FGFR2b contrast with the proliferative actions of FGFR1 and FGFR3, and may be converted to mitogenicity either by splice switching or by silencing of tumor suppressor genes such as CDH1 or PTEN. Exclusive use of small-molecule pan-FGFR inhibitors may thus cause nonselective blockade of FGFR2 isoforms with opposing actions, undermining the rationale of FGFR2 drug targeting. This splice-dependent ability of FGFR2 to switch between tumor-suppressing and -driving functions highlights an unmet oncologic need for isoform-specific drug targeting, e.g., by antibody inhibition of ligand-FGFR2c binding, as well as for more nuanced molecular pathology prediction of FGFR2 actions in different stromal-tumor contexts.
Collapse
Affiliation(s)
- Richard J. Epstein
- New Hope Cancer Center, Beijing United Hospital, 9-11 Jiangtai West Rd, Chaoyang, Beijing 100015, China
- Garvan Institute of Medical Research and UNSW Clinical School, 84 Victoria St, Darlinghurst 2010 Sydney, Australia
| | - Li Jun Tian
- New Hope Cancer Center, Beijing United Hospital, 9-11 Jiangtai West Rd, Chaoyang, Beijing 100015, China
| | - Yan Fei Gu
- New Hope Cancer Center, Beijing United Hospital, 9-11 Jiangtai West Rd, Chaoyang, Beijing 100015, China
| |
Collapse
|
7
|
Cleary JM, Raghavan S, Wu Q, Li YY, Spurr LF, Gupta HV, Rubinson DA, Fetter IJ, Hornick JL, Nowak JA, Siravegna G, Goyal L, Shi L, Brais LK, Loftus M, Shinagare AB, Abrams TA, Clancy TE, Wang J, Patel AK, Brichory F, Vaslin Chessex A, Sullivan RJ, Keller RB, Denning S, Hill ER, Shapiro GI, Pokorska-Bocci A, Zanna C, Ng K, Schrag D, Janne PA, Hahn WC, Cherniack AD, Corcoran RB, Meyerson M, Daina A, Zoete V, Bardeesy N, Wolpin BM. FGFR2 Extracellular Domain In-Frame Deletions are Therapeutically Targetable Genomic Alterations that Function as Oncogenic Drivers in Cholangiocarcinoma. Cancer Discov 2021; 11:2488-2505. [PMID: 33926920 DOI: 10.1158/2159-8290.cd-20-1669] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 03/10/2021] [Accepted: 04/26/2021] [Indexed: 11/16/2022]
Abstract
We conducted next generation DNA sequencing on 335 biliary tract cancers and characterized the genomic landscape by anatomic site within the biliary tree. In addition to frequent FGFR2 fusions among patients with intrahepatic cholangiocarcinoma (IHCC), we identified FGFR2 extracellular domain in-frame deletions (EIDs) in 5 of 178 (2.8%) patients with IHCC, including two patients with FGFR2 p.H167_N173del. Expression of this FGFR2 EID in NIH3T3 cells resulted in constitutive FGFR2 activation, oncogenic transformation, and sensitivity to FGFR inhibitors. Three patients with FGFR2 EIDs were treated with Debio 1347, an oral FGFR-1/2/3 inhibitor, and all showed partial responses. One patient developed an acquired L618F FGFR2 kinase domain mutation at disease progression and experienced a further partial response for 17 months to an irreversible FGFR2 inhibitor, futibatinib. Together, these findings reveal FGFR2 EIDs as an alternative mechanism of FGFR2 activation in IHCC that predict sensitivity to FGFR inhibitors in the clinic.
Collapse
Affiliation(s)
- James M Cleary
- Department of Medical Oncology, Dana-Farber Cancer Institute
| | | | | | - Yvonne Y Li
- Department of Medical Oncology, Dana-Farber Cancer Institute
| | - Liam F Spurr
- Dana-Farber Cancer Institute, Harvard Medical School
| | - Hersh V Gupta
- Department of Medical Oncology, Dana-Farber Cancer Institute
| | | | | | - Jason L Hornick
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School
| | | | | | - Lipika Goyal
- Internal Medicine, Massachusetts General Hospital Cancer Center, Harvard Medical School
| | - Lei Shi
- Center for Cancer Research, Massachusetts General Hospital Cancer Center, Harvard Medical School
| | - Lauren K Brais
- Department of Medical Oncology, Dana-Farber Cancer Institute
| | | | - Atul B Shinagare
- Department of Radiology, Brigham and Women's Hospital/ Dana-Farber Cancer Institute
| | | | | | - Jiping Wang
- Department of Surgery, Brigham and Women's Hospital
| | - Anuj K Patel
- Department of Gastrointestinal Oncology, Dana-Farber Cancer Institute
| | | | | | - Ryan J Sullivan
- Center for Melanoma, Massachusetts General Hospital Cancer Center
| | | | | | - Emma R Hill
- Dana-Farber/Brigham and Women's Cancer Center
| | | | | | | | - Kimmie Ng
- Department of Medical Oncology, Dana-Farber Cancer Institute
| | | | - Pasi A Janne
- Lowe Center for Thoracic Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute
| | - William C Hahn
- Department of Medical Oncology, Dana-Farber Cancer Institute
| | - Andrew D Cherniack
- Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School
| | | | | | | | | | | | - Brian M Wolpin
- Department of Medical Oncology, Dana-Farber/Harvard Cancer Center
| |
Collapse
|
8
|
Lam AS, Liu CC, Deutsch GH, Rivera J, Perkins JA, Holmes G, Jabs EW, Cunningham ML, Dahl JP. Genotype-Phenotype Correlation of Tracheal Cartilaginous Sleeves and Fgfr2 Mutations in Mice. Laryngoscope 2020; 131:E1349-E1356. [PMID: 32886384 DOI: 10.1002/lary.29060] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 07/22/2020] [Accepted: 08/10/2020] [Indexed: 12/21/2022]
Abstract
OBJECTIVES To characterize tracheal cartilage morphology in mouse models of fibroblast growth factor receptor (Fgfr2)-related craniosynostosis syndromes. To establish relationships between specific Fgfr2 mutations and tracheal cartilaginous sleeve (TCS) phenotypes in these mouse models. METHODS Postnatal day 0 knock-in mouse lines with disease-specific genetic variations in the Fgfr2 gene (Fgfr2C342Y/C342Y , Fgfr2C342Y/+ , Fgfr2+/Y394C , Fgfr2+/S252W , and Fgfr2+/P253R ) as well as line-specific controls were utilized. Tracheal cartilage morphology as measured by gross analyses, microcomputed tomography (μCT), and histopathology were compared using Chi-squared and single-factor analysis of variance statistical tests. RESULTS A greater proportion of rings per trachea were abnormal in Fgfr2C342Y/+ tracheas (63%) than Fgfr2+/S252W (17%), Fgfr2+/P253R (17%), Fgfr2+/Y394C (12%), and controls (10%) (P < .001 for each vs. Fgfr2C342Y/+ ). TCS segments were found only in Fgfr2C342Y/C342Y (100%) and Fgfr2C342Y/+ (72%) tracheas. Cricoid and first-tracheal ring fusion was noted in all Fgfr2C342Y/C342Y and 94% of Fgfr2C342Y/+ samples. The Fgfr2C342Y/C342Y and Fgfr2C342Y/+ groups were found to have greater areas and volumes of cartilage than other lines on gross analysis and μCT. Histologic analyses confirmed TCS among the Fgfr2C342Y/C342Y and Fgfr2C342Y/+ groups, without appreciable differences in cartilage morphology, cell size, or density; no histologic differences were observed among other Fgfr2 lines compared to controls. CONCLUSION This study found TCS phenotypes only in the Fgfr2C342Y mouse lines. These lines also had increased tracheal cartilage compared to other mutant lines and controls. These data support further study of the Fgfr2 mouse lines and the investigation of other Fgfr2 variants to better understand their role in tracheal development and TCS formation. LEVEL OF EVIDENCE NA Laryngoscope, 131:E1349-E1356, 2021.
Collapse
Affiliation(s)
- Austin S Lam
- Department of Otolaryngology - Head & Neck Surgery, University of Washington School of Medicine, Seattle, Washington, U.S.A.,Division of Pediatric Otolaryngology - Head & Neck Surgery, Seattle Children's Hospital, Seattle, Washington, U.S.A.,Seattle Children's Research Institute, Center for Developmental Biology and Regenerative Medicine, Seattle, Washington, U.S.A
| | - Carrie C Liu
- Department of Otolaryngology - Head & Neck Surgery, University of Washington School of Medicine, Seattle, Washington, U.S.A.,Division of Pediatric Otolaryngology - Head & Neck Surgery, Seattle Children's Hospital, Seattle, Washington, U.S.A.,Current address: Divisions of Otolaryngology - Head and Neck Surgery, and Pediatric Surgery, Department of Surgery, University of Calgary, Calgary, Alberta, Canada
| | - Gail H Deutsch
- Department of Pathology, University of Washington School of Medicine, Seattle, Washington, U.S.A.,Department of Pathology, Seattle Children's Hospital, Seattle, Washington, U.S.A
| | - Joshua Rivera
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York City, New York, U.S.A.,Current address: Center for Personalized Cancer Therapy, University of Massachusetts, Boston, Massachusetts, U.S.A
| | - Jonathan A Perkins
- Department of Otolaryngology - Head & Neck Surgery, University of Washington School of Medicine, Seattle, Washington, U.S.A.,Division of Pediatric Otolaryngology - Head & Neck Surgery, Seattle Children's Hospital, Seattle, Washington, U.S.A.,Craniofacial Center, Seattle Children's Hospital, Seattle, Washington, U.S.A
| | - Greg Holmes
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York City, New York, U.S.A
| | - Ethylin W Jabs
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York City, New York, U.S.A
| | - Michael L Cunningham
- Seattle Children's Research Institute, Center for Developmental Biology and Regenerative Medicine, Seattle, Washington, U.S.A.,Craniofacial Center, Seattle Children's Hospital, Seattle, Washington, U.S.A.,Department of Pediatrics, University of Washington School of Medicine, Seattle, Washington, U.S.A
| | - John P Dahl
- Department of Otolaryngology - Head & Neck Surgery, University of Washington School of Medicine, Seattle, Washington, U.S.A.,Division of Pediatric Otolaryngology - Head & Neck Surgery, Seattle Children's Hospital, Seattle, Washington, U.S.A.,Craniofacial Center, Seattle Children's Hospital, Seattle, Washington, U.S.A
| |
Collapse
|
9
|
Dias MS, Samson T, Rizk EB, Governale LS, Richtsmeier JT. Identifying the Misshapen Head: Craniosynostosis and Related Disorders. Pediatrics 2020; 146:peds.2020-015511. [PMID: 32868470 DOI: 10.1542/peds.2020-015511] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Pediatric care providers, pediatricians, pediatric subspecialty physicians, and other health care providers should be able to recognize children with abnormal head shapes that occur as a result of both synostotic and deformational processes. The purpose of this clinical report is to review the characteristic head shape changes, as well as secondary craniofacial characteristics, that occur in the setting of the various primary craniosynostoses and deformations. As an introduction, the physiology and genetics of skull growth as well as the pathophysiology underlying craniosynostosis are reviewed. This is followed by a description of each type of primary craniosynostosis (metopic, unicoronal, bicoronal, sagittal, lambdoid, and frontosphenoidal) and their resultant head shape changes, with an emphasis on differentiating conditions that require surgical correction from those (bathrocephaly, deformational plagiocephaly/brachycephaly, and neonatal intensive care unit-associated skill deformation, known as NICUcephaly) that do not. The report ends with a brief discussion of microcephaly as it relates to craniosynostosis as well as fontanelle closure. The intent is to improve pediatric care providers' recognition and timely referral for craniosynostosis and their differentiation of synostotic from deformational and other nonoperative head shape changes.
Collapse
Affiliation(s)
- Mark S Dias
- Section of Pediatric Neurosurgery, Department of Neurosurgery and
| | - Thomas Samson
- Division of Plastic Surgery, Department of Surgery, College of Medicine and
| | - Elias B Rizk
- Section of Pediatric Neurosurgery, Department of Neurosurgery and
| | - Lance S Governale
- Lillian S. Wells Department of Neurosurgery, College of Medicine, University of Florida, Gainesville, Florida
| | - Joan T Richtsmeier
- Department of Anthropology, College of the Liberal Arts and Huck Institutes of the Life Sciences, Pennsylvania State University, State College, Pennsylvania; and
| | | |
Collapse
|
10
|
Kim WJ, Shin HL, Kim BS, Kim HJ, Ryoo HM. RUNX2-modifying enzymes: therapeutic targets for bone diseases. Exp Mol Med 2020; 52:1178-1184. [PMID: 32788656 PMCID: PMC8080656 DOI: 10.1038/s12276-020-0471-4] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 05/21/2020] [Accepted: 05/22/2020] [Indexed: 01/01/2023] Open
Abstract
RUNX2 is a master transcription factor of osteoblast differentiation. RUNX2 expression in the bone and osteogenic front of a suture is crucial for cranial suture closure and membranous bone morphogenesis. In this manner, the regulation of RUNX2 is precisely controlled by multiple posttranslational modifications (PTMs) mediated by the stepwise recruitment of multiple enzymes. Genetic defects in RUNX2 itself or in its PTM regulatory pathways result in craniofacial malformations. Haploinsufficiency in RUNX2 causes cleidocranial dysplasia (CCD), which is characterized by open fontanelle and hypoplastic clavicles. In contrast, gain-of-function mutations in FGFRs, which are known upstream stimulating signals of RUNX2 activity, cause craniosynostosis (CS) characterized by premature suture obliteration. The identification of these PTM cascades could suggest suitable drug targets for RUNX2 regulation. In this review, we will focus on the mechanism of RUNX2 regulation mediated by PTMs, such as phosphorylation, prolyl isomerization, acetylation, and ubiquitination, and we will summarize the therapeutics associated with each PTM enzyme for the treatment of congenital cranial suture anomalies.
Collapse
Affiliation(s)
- Woo-Jin Kim
- Basic Research Lab for "Epigenetic Regeneration of Aged Skeleto-Muscular System (ERASMUS)", Department of Molecular Genetics and Dental Pharmacology, School of Dentistry, Dental Research Institute, Seoul National University, Seoul, South Korea
| | - Hye-Lim Shin
- Basic Research Lab for "Epigenetic Regeneration of Aged Skeleto-Muscular System (ERASMUS)", Department of Molecular Genetics and Dental Pharmacology, School of Dentistry, Dental Research Institute, Seoul National University, Seoul, South Korea
| | - Bong-Soo Kim
- Basic Research Lab for "Epigenetic Regeneration of Aged Skeleto-Muscular System (ERASMUS)", Department of Molecular Genetics and Dental Pharmacology, School of Dentistry, Dental Research Institute, Seoul National University, Seoul, South Korea
| | - Hyun-Jung Kim
- Basic Research Lab for "Epigenetic Regeneration of Aged Skeleto-Muscular System (ERASMUS)", Department of Molecular Genetics and Dental Pharmacology, School of Dentistry, Dental Research Institute, Seoul National University, Seoul, South Korea
| | - Hyun-Mo Ryoo
- Basic Research Lab for "Epigenetic Regeneration of Aged Skeleto-Muscular System (ERASMUS)", Department of Molecular Genetics and Dental Pharmacology, School of Dentistry, Dental Research Institute, Seoul National University, Seoul, South Korea.
| |
Collapse
|
11
|
Dash S, Trainor PA. The development, patterning and evolution of neural crest cell differentiation into cartilage and bone. Bone 2020; 137:115409. [PMID: 32417535 DOI: 10.1016/j.bone.2020.115409] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 05/04/2020] [Indexed: 12/12/2022]
Abstract
Neural crest cells are a vertebrate-specific migratory, multipotent cell population that give rise to a diverse array of cells and tissues during development. Cranial neural crest cells, in particular, generate cartilage, bone, tendons and connective tissue in the head and face as well as neurons, glia and melanocytes. In this review, we focus on the chondrogenic and osteogenic potential of cranial neural crest cells and discuss the roles of Sox9, Runx2 and Msx1/2 transcription factors and WNT, FGF and TGFβ signaling pathways in regulating neural crest cell differentiation into cartilage and bone. We also describe cranioskeletal defects and disorders arising from gain or loss-of-function of genes that are required for patterning and differentiation of cranial neural crest cells. Finally, we discuss the evolution of skeletogenic potential in neural crest cells and their function as a conduit for intraspecies and interspecies variation, and the evolution of craniofacial novelties.
Collapse
Affiliation(s)
- Soma Dash
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Paul A Trainor
- Stowers Institute for Medical Research, Kansas City, MO, USA; Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, USA.
| |
Collapse
|
12
|
Hasan MR, Takatalo M, Ma H, Rice R, Mustonen T, Rice DP. RAB23 coordinates early osteogenesis by repressing FGF10-pERK1/2 and GLI1. eLife 2020; 9:55829. [PMID: 32662771 PMCID: PMC7423339 DOI: 10.7554/elife.55829] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 07/13/2020] [Indexed: 12/20/2022] Open
Abstract
Mutations in the gene encoding Ras-associated binding protein 23 (RAB23) cause Carpenter Syndrome, which is characterized by multiple developmental abnormalities including polysyndactyly and defects in skull morphogenesis. To understand how RAB23 regulates skull development, we generated Rab23-deficient mice that survive to an age where skeletal development can be studied. Along with polysyndactyly, these mice exhibit premature fusion of multiple sutures resultant from aberrant osteoprogenitor proliferation and elevated osteogenesis in the suture. FGF10-driven FGFR1 signaling is elevated in Rab23-/-sutures with a consequent imbalance in MAPK, Hedgehog signaling and RUNX2 expression. Inhibition of elevated pERK1/2 signaling results in the normalization of osteoprogenitor proliferation with a concomitant reduction of osteogenic gene expression, and prevention of craniosynostosis. Our results suggest a novel role for RAB23 as an upstream negative regulator of both FGFR and canonical Hh-GLI1 signaling, and additionally in the non-canonical regulation of GLI1 through pERK1/2.
Collapse
Affiliation(s)
- Md Rakibul Hasan
- Craniofacial Development and Malformations research group, Orthodontics, Oral and Maxillofacial Diseases, University of Helsinki, Helsinki, Finland
| | - Maarit Takatalo
- Craniofacial Development and Malformations research group, Orthodontics, Oral and Maxillofacial Diseases, University of Helsinki, Helsinki, Finland
| | - Hongqiang Ma
- Craniofacial Development and Malformations research group, Orthodontics, Oral and Maxillofacial Diseases, University of Helsinki, Helsinki, Finland
| | - Ritva Rice
- Craniofacial Development and Malformations research group, Orthodontics, Oral and Maxillofacial Diseases, University of Helsinki, Helsinki, Finland
| | - Tuija Mustonen
- Craniofacial Development and Malformations research group, Orthodontics, Oral and Maxillofacial Diseases, University of Helsinki, Helsinki, Finland
| | - David Pc Rice
- Craniofacial Development and Malformations research group, Orthodontics, Oral and Maxillofacial Diseases, University of Helsinki, Helsinki, Finland.,Oral and Maxillofacial Diseases, Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
13
|
Xie N, Tian C, Wu H, Yang X, Liu L, Li J, Xiao H, Gao J, Lu J, Hu X, Cao M, Shui Z, Tang Y, Wang X, Yang J, Hu ZY, Ouyang Q. FGFR aberrations increase the risk of brain metastases and predict poor prognosis in metastatic breast cancer patients. Ther Adv Med Oncol 2020; 12:1758835920915305. [PMID: 32499836 PMCID: PMC7243401 DOI: 10.1177/1758835920915305] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 02/05/2020] [Indexed: 02/06/2023] Open
Abstract
Background: The survival status of patients with breast cancer and brain metastasis (BCBM) receiving current treatments is poor. Method: We designed a real-world study to investigate using patients’ clinical and genetic aberrations to forecast the prognoses of BCBM patients. We recruited 146 BCBM patients and analyzed their clinical features to evaluate the overall survival (OS). For genetic testing, 30 BCBM and 165 non-brain-metastatic (BM) metastatic breast cancer (MBC) patients from Hunan Cancer Hospital, and 86 BCBM and 1416 non-BM MBC patients from the Geneplus database who received circulating tumor DNA testing, were compared and analyzed. Results: Ki67 >14% and >3 metastatic brain tumors were significant risk factors associated with poor OS, while chemotherapy and brain radiotherapy were beneficial factors for better OS. Compared with non-BM MBC patients, BCBM patients had more fibroblast growth factor receptor (FGFR) aberrations. The combination of FGFR, TP53 and FLT1 aberrations plus immunohistochemistry HER2-positive were associated with an increased risk of brain metastasis (AUC = 77.13%). FGFR aberration alone was not only a predictive factor (AUC = 67.90%), but also a significant risk factor for poor progression-free survival (Logrank p = 0.029). FGFR1 aberration was more frequent than other FGFR family genes in BCBM patients, and FGFR1 aberration was significantly higher in BCBM patients than non-BM MBC patients. Most FGFR1-amplified MBC patients progressed within 3 months of the late-line (>2 lines) treatment. Conclusion: A group of genetic events, including FGFR, TP53 and FLT1 genetic aberrations, and HER2-positivity, forecasted the occurrence of BM in breast cancers. FGFR genetic aberration alone predicted poor prognosis.
Collapse
Affiliation(s)
- Ning Xie
- Hunan Cancer Hospital, and the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha, China
| | - Can Tian
- Hunan Cancer Hospital, and the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha, China
| | - Hui Wu
- Hunan Cancer Hospital, and the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha, China
| | - Xiaohong Yang
- Hunan Cancer Hospital, and the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha, China
| | - Liping Liu
- Hunan Cancer Hospital, and the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha, China
| | - Jing Li
- Hunan Cancer Hospital, and the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha, China
| | - Huawu Xiao
- Hunan Cancer Hospital, and the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha, China
| | - Jianxiang Gao
- Hunan Cancer Hospital, and the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha, China
| | - Jun Lu
- Hunan Cancer Hospital, and the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha, China
| | - Xuming Hu
- Hunan Cancer Hospital, and the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha, China
| | - Min Cao
- Hunan Cancer Hospital, and the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha, China
| | - Zhengrong Shui
- Hunan Cancer Hospital, and the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha, China
| | - Yu Tang
- Hunan Cancer Hospital, and the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha, China
| | - Xiao Wang
- ICF, 3 Corporate Square NE., Atlanta, GA, USA
| | - Jianbo Yang
- Department of Otolaryngology, Medical School, University of Minnesota, Minneapolis, Minnesota, USA
| | - Zhe-Yu Hu
- Department of Breast Cancer Medical Oncology, Hunan Cancer Hospital, the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, No. 283, Tongzipo Road, Changsha, 410013, P.R. China
| | - Quchang Ouyang
- Department of Breast Cancer Medical Oncology, Hunan Cancer Hospital, the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, No. 283, Tongzipo Road, Changsha, 410013, P.R. China
| |
Collapse
|
14
|
Ferreira LC, Dantas Junior JH. Report of a Father With Congenital Bilateral Absence of the Vas Deferens Fathering a Child With Beare–Stevenson Syndrome. Front Genet 2020; 11:104. [PMID: 32158469 PMCID: PMC7052335 DOI: 10.3389/fgene.2020.00104] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 01/29/2020] [Indexed: 11/13/2022] Open
Affiliation(s)
- Leonardo C. Ferreira
- Department of Biochemistry, Federal University of Rio Grande do Norte, Natal, Brazil
- Institute of Tropical Medicine of Rio Grande do Norte, Federal University of Rio Grande do Norte, Natal, Brazil
- *Correspondence: Leonardo C. Ferreira,
| | - José H. Dantas Junior
- University Hospital Onofre Lopes, Urologic Unit, Federal University of Rio Grande do Norte, Natal, Brazil
| |
Collapse
|
15
|
Cornille M, Dambroise E, Komla-Ebri D, Kaci N, Biosse-Duplan M, Di Rocco F, Legeai-Mallet L. Animal models of craniosynostosis. Neurochirurgie 2019; 65:202-209. [PMID: 31563616 DOI: 10.1016/j.neuchi.2019.09.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 07/22/2019] [Accepted: 09/16/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND Various animal models mimicking craniosynostosis have been developed, using mutant zebrafish and mouse. The aim of this paper is to review the different animal models for syndromic craniosynostosis and analyze what insights they have provided in our understanding of the pathophysiology of these conditions. MATERIAL AND METHODS The relevant literature for animal models of craniosynostosis was reviewed. RESULTS Although few studies on craniosynostosis using zebrafish were published, this model appears useful in studying the suture formation mechanisms conserved across vertebrates. Conversely, several mouse models have been generated for the most common syndromic craniosynostoses, associated with mutations in FGFR1, FGFR2, FGFR3 and TWIST genes and also in MSX2, EFFNA, GLI3, FREM1, FGF3/4 genes. The mouse models have also been used to test pharmacological treatments to restore craniofacial growth. CONCLUSIONS Several zebrafish and mouse models have been developed in recent decades. These animal models have been helpful for our understanding of normal and pathological craniofacial growth. Mouse models mimicking craniosynostoses can be easily used for the screening of drugs as therapeutic candidates.
Collapse
Affiliation(s)
- M Cornille
- Inserm U1163, Paris university, institut Imagine, 75015 Paris, France
| | - E Dambroise
- Inserm U1163, Paris university, institut Imagine, 75015 Paris, France
| | - D Komla-Ebri
- Inserm U1163, Paris university, institut Imagine, 75015 Paris, France; Molecular Endocrinology Laboratory, Department of Medicine, Imperial College London, W12 ONNLondon, United Kingdom
| | - N Kaci
- Inserm U1163, Paris university, institut Imagine, 75015 Paris, France; Inovarion, 75013 Paris, France
| | - M Biosse-Duplan
- Inserm U1163, Paris university, institut Imagine, 75015 Paris, France
| | - F Di Rocco
- Centre de référence craniosténoses, université de Lyon, 69677 Bron France; Service de neurochirurgie pédiatrique, université Lyon, hôpital Femme-Mère-Enfant, 69677, Bron, France.
| | - L Legeai-Mallet
- Inserm U1163, Paris university, institut Imagine, 75015 Paris, France.
| |
Collapse
|
16
|
Rachwalski M, Khonsari RH, Paternoster G. Current Approaches in the Development of Molecular and Pharmacological Therapies in Craniosynostosis Utilizing Animal Models. Mol Syndromol 2019; 10:115-123. [PMID: 30976284 DOI: 10.1159/000493535] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The development of the craniofacial skeleton is a spatial and temporal process where cranial sutures play a role in the regulation of morphogenesis and growth. Disruption of these cellular and molecular interactions may lead to craniosynostosis, the premature obliteration of one or more cranial sutures, yielding skull growth restriction and malformation perpendicular to the affected suture. Facial deformity and various functional CNS anomalies are other frequent complications. Cranial vault expansion and reconstructive surgery remain the mainstay of treatment but pose an elevated risk of morbidity for the infant. While the etiology of nonsyndromic craniosynostosis remains to be deciphered, gain-of-function mutations in FGFR1-3 and TWIST1 were found to be responsible for more than 3/4 of the most commonly encountered craniofacial syndromes. Animal models have been invaluable to further dissect the role of genes within the cranial sutures and for the development of alternative nonsurgical treatment strategies. In this review, we will present various molecular and pharmacological approaches for the treatment of craniosynostosis that have been tested using in vitro and in vivo assays as well as discuss their potential application in humans focusing on the case of tyrosine kinase inhibitors.
Collapse
Affiliation(s)
- Martin Rachwalski
- Imagine Institute of Genetic Diseases, INSERM U1163, Université Paris Descartes, Sorbonne Paris Cité, Departments of Malades, Paris, France.,Pediatric Neurosurgery, Hôpital Universitaire Necker-Enfants Malades, Paris, France.,Maxillofacial and Plastic Surgery, Hôpital Universitaire Necker-Enfants Malades, Paris, France.,National Reference Center for Craniofacial Anomalies, Hôpital Universitaire Necker-Enfants Malades, Paris, France
| | - Roman H Khonsari
- Imagine Institute of Genetic Diseases, INSERM U1163, Université Paris Descartes, Sorbonne Paris Cité, Departments of Malades, Paris, France.,Maxillofacial and Plastic Surgery, Hôpital Universitaire Necker-Enfants Malades, Paris, France.,National Reference Center for Craniofacial Anomalies, Hôpital Universitaire Necker-Enfants Malades, Paris, France
| | - Giovanna Paternoster
- Pediatric Neurosurgery, Hôpital Universitaire Necker-Enfants Malades, Paris, France.,National Reference Center for Craniofacial Anomalies, Hôpital Universitaire Necker-Enfants Malades, Paris, France
| |
Collapse
|
17
|
Abstract
Deviations from the precisely coordinated programme of human head development can lead to craniofacial and orofacial malformations often including a variety of dental abnormalities too. Although the aetiology is still unknown in many cases, during the last decades different intracellular signalling pathways have been genetically linked to specific disorders. Among these pathways, the RAS/extracellular signal-regulated kinase (ERK) signalling cascade is the focus of this review since it encompasses a large group of genes that when mutated cause some of the most common and severe developmental anomalies in humans. We present the components of the RAS/ERK pathway implicated in craniofacial and orodental disorders through a series of human and animal studies. We attempt to unravel the specific molecular targets downstream of ERK that act on particular cell types and regulate key steps in the associated developmental processes. Finally we point to ambiguities in our current knowledge that need to be clarified before RAS/ERK-targeting therapeutic approaches can be implemented.
Collapse
|
18
|
Sewda A, White SR, Erazo M, Hao K, García-Fructuoso G, Fernández-Rodriguez I, Heuzé Y, Richtsmeier JT, Romitti PA, Reva B, Jabs EW, Peter I. Nonsyndromic craniosynostosis: novel coding variants. Pediatr Res 2019; 85:463-468. [PMID: 30651579 PMCID: PMC6398438 DOI: 10.1038/s41390-019-0274-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 11/09/2018] [Indexed: 11/10/2022]
Abstract
BACKGROUND Craniosynostosis (CS), the premature fusion of one or more neurocranial sutures, is associated with approximately 200 syndromes; however, about 65-85% of patients present with no additional major birth defects. METHODS We conducted targeted next-generation sequencing of 60 known syndromic and other candidate genes in patients with sagittal nonsyndromic CS (sNCS, n = 40) and coronal nonsyndromic CS (cNCS, n = 19). RESULTS We identified 18 previously published and 5 novel pathogenic variants, including three de novo variants. Novel variants included a paternally inherited c.2209C>G:p.(Leu737Val) variant in BBS9 of a patient with cNCS. Common variants in BBS9, a gene required for ciliogenesis during cranial suture development, have been associated with sNCS risk in a previous genome-wide association study. We also identified c.313G>T:p.(Glu105*) variant in EFNB1 and c.435G>C:p.(Lys145Asn) variant in TWIST1, both in patients with cNCS. Mutations in EFNB1 and TWIST1 have been linked to craniofrontonasal and Saethre-Chotzen syndrome, respectively; both present with coronal CS. CONCLUSIONS We provide additional evidence that variants in genes implicated in syndromic CS play a role in isolated CS, supporting their inclusion in genetic panels for screening patients with NCS. We also identified a novel BBS9 variant that further shows the potential involvement of BBS9 in the pathogenesis of CS.
Collapse
Affiliation(s)
- Anshuman Sewda
- Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Sierra R. White
- Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Monica Erazo
- Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Ke Hao
- Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | | | | | - Yann Heuzé
- University Bordeaux, CNRS, MCC, PACEA, UMR5199, Bordeaux Archaeological Sciences Cluster of Excellence, Pessac, France
| | - Joan T. Richtsmeier
- Department of Anthropology, Pennsylvania State University, University Park, Pennsylvania
| | - Paul A. Romitti
- Department of Epidemiology, College of Public Health, University of Iowa, Iowa City, Iowa
| | - Boris Reva
- Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Ethylin Wang Jabs
- Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Inga Peter
- Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
19
|
Holmes G, O'Rourke C, Motch Perrine SM, Lu N, van Bakel H, Richtsmeier JT, Jabs EW. Midface and upper airway dysgenesis in FGFR2-related craniosynostosis involves multiple tissue-specific and cell cycle effects. Development 2018; 145:dev.166488. [PMID: 30228104 DOI: 10.1242/dev.166488] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 09/03/2018] [Indexed: 12/23/2022]
Abstract
Midface dysgenesis is a feature of more than 200 genetic conditions in which upper airway anomalies frequently cause respiratory distress, but its etiology is poorly understood. Mouse models of Apert and Crouzon craniosynostosis syndromes exhibit midface dysgenesis similar to the human conditions. They carry activating mutations of Fgfr2, which is expressed in multiple craniofacial tissues during development. Magnetic resonance microscopy of three mouse models of Apert and Crouzon syndromes revealed decreased nasal passage volume in all models at birth. Histological analysis suggested overgrowth of the nasal cartilage in the two Apert syndrome mouse models. We used tissue-specific gene expression and transcriptome analysis to further dissect the structural, cellular and molecular alterations underlying midface and upper airway dysgenesis in Apert Fgfr2+/S252W mutants. Cartilage thickened progressively during embryogenesis because of increased chondrocyte proliferation in the presence of Fgf2 Oral epithelium expression of mutant Fgfr2, which resulted in a distinctive nasal septal fusion defect, and premature facial suture fusion contributed to the overall dysmorphology. Midface dysgenesis in Fgfr2-related craniosynostosis is a complex phenotype arising from the combined effects of aberrant signaling in multiple craniofacial tissues.
Collapse
Affiliation(s)
- Greg Holmes
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Courtney O'Rourke
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Susan M Motch Perrine
- Department of Anthropology, Pennsylvania State University, University Park, PA 16802, USA
| | - Na Lu
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Harm van Bakel
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Joan T Richtsmeier
- Department of Anthropology, Pennsylvania State University, University Park, PA 16802, USA
| | - Ethylin Wang Jabs
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
20
|
Abstract
Craniosynostosis is a common craniofacial birth defect. This review focusses on the advances that have been achieved through studying the pathogenesis of craniosynostosis using mouse models. Classic methods of gene targeting which generate individual gene knockout models have successfully identified numerous genes required for normal development of the skull bones and sutures. However, the study of syndromic craniosynostosis has largely benefited from the production of knockin models that precisely mimic human mutations. These have allowed the detailed investigation of downstream events at the cellular and molecular level following otherwise unpredictable gain-of-function effects. This has greatly enhanced our understanding of the pathogenesis of this disease and has the potential to translate into improvement of the clinical management of this condition in the future.
Collapse
Affiliation(s)
- Kevin K L Lee
- UCL Great Ormond Street Institute of Child Health, London, UK
| | - Philip Stanier
- UCL Great Ormond Street Institute of Child Health, London, UK
| | - Erwin Pauws
- UCL Great Ormond Street Institute of Child Health, London, UK
| |
Collapse
|
21
|
LeBlanc S, David D, Colley A, Buckley M, Roscioli T, Barnett C. Atypical Skin Manifestations in FGFR2-Related Craniosynostosis Syndromes Broaden the Phenotypic Spectrum. Mol Syndromol 2018; 9:149-153. [PMID: 29928180 DOI: 10.1159/000488439] [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] [Accepted: 11/09/2017] [Indexed: 01/29/2023] Open
Abstract
Crouzon syndrome (CS) and Beare-Stevenson syndrome (BSS) are craniosynostosis syndromes caused by mutations in the fibroblast growth factor 2 (FGFR2) gene. CS is more common (1 in 60,000 live births) than BSS, where fewer than 20 individuals have been reported. The cardinal features of BSS are craniosynostosis, cutis gyrata, acanthosis nigricans, skin furrows, skin tags, anogenital anomalies, and a prominent umbilical stump. Previously described individuals with BSS have typically had mutations in exon 11 of FGFR2. Here, we present 2 patients with CS who have significant skin manifestations and some phenotypic overlap with BSS. De novo mutations in exon 8 of FGFR2 were identified in both; one is a mutation (c.799T>C; p.Ser267Pro) previously identified in individuals with CS and the other a novel in-frame deletion (c.820_824delinsTT; p.Val274_Glu275delinsLeu). No mutations in exon 11 of FGFR2, where previously reported BSS mutations have been located, were identified. This case expands the phenotypic spectrum of CS and highlights the overlap between conditions caused by mutations in FGFR2.
Collapse
Affiliation(s)
- Shannon LeBlanc
- Paediatric and Reproductive Genetics Unit, South Australian Clinical Genetics Service, Women's and Children's Hospital/SA Pathology, SA, Australia
| | - David David
- Australian Craniofacial Unit, Women's and Children's Hospital, North Adelaide, SA, Australia
| | - Alison Colley
- Department of Clinical Genetics, Liverpool Hospital, Liverpool, NSW, Australia
| | - Michael Buckley
- Genetics Laboratory, NSW Health Pathology East, North Sydney, NSW, Australia
| | - Tony Roscioli
- Department of Medical Genetics, Sydney Children's Hospital Randwick, NSW, Australia.,Prince of Wales Clinical School, University of New South Wales, Randwick, NSW, Australia
| | - Christopher Barnett
- Paediatric and Reproductive Genetics Unit, South Australian Clinical Genetics Service, Women's and Children's Hospital/SA Pathology, SA, Australia
| |
Collapse
|
22
|
Richtsmeier JT. A century of development. AMERICAN JOURNAL OF PHYSICAL ANTHROPOLOGY 2018; 165:726-740. [PMID: 29574839 PMCID: PMC6007869 DOI: 10.1002/ajpa.23379] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 12/02/2017] [Accepted: 12/09/2017] [Indexed: 12/13/2022]
Affiliation(s)
- Joan T Richtsmeier
- Department of Anthropology, Pennsylvania State University, University Park, Pennsylvania 16802
| |
Collapse
|
23
|
Role of Notch Signaling in the Physiological Patterning of Posterofrontal and Sagittal Cranial Sutures. J Craniofac Surg 2018; 28:1620-1625. [PMID: 28692512 DOI: 10.1097/scs.0000000000003721] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND The mutations in a Notch signaling ligand, jagged 1, are associated with unilateral coronal craniosynostosis in humans. However, the underlying mechanisms of Notch signaling in cranial suture biology still remain unclear. METHODS The temporal and spatial patterns of Notch signaling expression were examined in the posterofrontal and sagittal sutures of Sprague-Dawley rats by real-time quantitative reverse-transcription polymerase chain reaction at postnatal ages of 2, 15, and 25 days. The role of Notch signaling in the proliferation and differentiation of osteoblasts isolated from calvarial was examined in vitro by EdU incorporation assays and real-time quantitative reverse-transcription polymerase chain reaction after activating and inhibiting Notch signaling. RESULTS The mRNA levels of Notch family members (including Jagged 1, Delta 1, 3, 4, Notch 1-4, Hes 1, and Hes 5) decreased during the posterofrontal cranial suture fusion in rat. However, in the patent sagittal sutures, the mRNA levels of Notch family members (Jagged 2, Delta 1, Notch 1, Notch 3, Hes 5, and Hey 1) increased during suture development. The EdU incorporation assays revealed that the induction of Notch signaling in calvaria osteobalsts using Jagged 1 promoted the proliferation rates in those cells in vitro. Further studies showed that activation of Notch signaling calvaria osteobalsts using Jagged 1 led to the suppression of late osteogenetic markers such as type I collagen and osteocalcin. CONCLUSIONS The regulation of Notch signaling is of crucial importance during the physiological patterning of posterofrontal and sagittal cranial sutures. Thus, targeting this pathway may prove significant for the development of future therapeutic applications in craniosynostosis.
Collapse
|
24
|
Balek L, Gudernova I, Vesela I, Hampl M, Oralova V, Kunova Bosakova M, Varecha M, Nemec P, Hall T, Abbadessa G, Hatch N, Buchtova M, Krejci P. ARQ 087 inhibits FGFR signaling and rescues aberrant cell proliferation and differentiation in experimental models of craniosynostoses and chondrodysplasias caused by activating mutations in FGFR1, FGFR2 and FGFR3. Bone 2017; 105:57-66. [PMID: 28826843 DOI: 10.1016/j.bone.2017.08.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2016] [Revised: 08/17/2017] [Accepted: 08/18/2017] [Indexed: 01/16/2023]
Abstract
Tyrosine kinase inhibitors are being developed for therapy of malignancies caused by oncogenic FGFR signaling but little is known about their effect in congenital chondrodysplasias or craniosynostoses that associate with activating FGFR mutations. Here, we investigated the effects of novel FGFR inhibitor, ARQ 087, in experimental models of aberrant FGFR3 signaling in cartilage. In cultured chondrocytes, ARQ 087 efficiently rescued all major effects of pathological FGFR3 activation, i.e. inhibition of chondrocyte proliferation, loss of extracellular matrix and induction of premature senescence. In ex vivo tibia organ cultures, ARQ 087 restored normal growth plate architecture and eliminated the suppressing FGFR3 effect on chondrocyte hypertrophic differentiation, suggesting that it targets the FGFR3 pathway specifically, i.e. without interference with other pro-growth pathways. Moreover, ARQ 087 inhibited activity of FGFR1 and FGFR2 mutants associated with Pfeiffer, Apert and Beare-Stevenson craniosynostoses, and rescued FGFR-driven excessive osteogenic differentiation in mouse mesenchymal micromass cultures or in ex vivo calvarial organ cultures. Our data warrant further development of ARQ 087 for clinical use in skeletal disorders caused by activating FGFR mutations.
Collapse
Affiliation(s)
- Lukas Balek
- Institute of Experimental Biology, Faculty of Sciences, Masaryk University, 62500 Brno, Czech Republic; Department of Biology, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic
| | - Iva Gudernova
- Department of Biology, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic
| | - Iva Vesela
- Institute of Animal Physiology and Genetics, Czech Academy of Sciences, 60200 Brno, Czech Republic
| | - Marek Hampl
- Institute of Experimental Biology, Faculty of Sciences, Masaryk University, 62500 Brno, Czech Republic; Institute of Animal Physiology and Genetics, Czech Academy of Sciences, 60200 Brno, Czech Republic
| | - Veronika Oralova
- Institute of Animal Physiology and Genetics, Czech Academy of Sciences, 60200 Brno, Czech Republic
| | | | - Miroslav Varecha
- Department of Biology, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic; International Clinical Research Center, St. Anne's University Hospital, 65691 Brno, Czech Republic
| | - Pavel Nemec
- Department of Biology, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic; International Clinical Research Center, St. Anne's University Hospital, 65691 Brno, Czech Republic
| | | | | | - Nan Hatch
- University of Michigan School of Dentistry, University of Michigan, Ann Arbor, MI 48109, USA
| | - Marcela Buchtova
- Institute of Experimental Biology, Faculty of Sciences, Masaryk University, 62500 Brno, Czech Republic; Institute of Animal Physiology and Genetics, Czech Academy of Sciences, 60200 Brno, Czech Republic.
| | - Pavel Krejci
- Department of Biology, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic; International Clinical Research Center, St. Anne's University Hospital, 65691 Brno, Czech Republic.
| |
Collapse
|
25
|
Azoury SC, Reddy S, Shukla V, Deng CX. Fibroblast Growth Factor Receptor 2 ( FGFR2) Mutation Related Syndromic Craniosynostosis. Int J Biol Sci 2017; 13:1479-1488. [PMID: 29230096 PMCID: PMC5723914 DOI: 10.7150/ijbs.22373] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 10/01/2017] [Indexed: 12/30/2022] Open
Abstract
Craniosynostosis results from the premature fusion of cranial sutures, with an incidence of 1 in 2,100-2,500 live births. The majority of cases are non-syndromic and involve single suture fusion, whereas syndromic cases often involve complex multiple suture fusion. The fibroblast growth factor receptor 2 (FGFR2) gene is perhaps the most extensively studied gene that is mutated in various craniosynostotic syndromes including Crouzon, Apert, Pfeiffer, Antley-Bixler, Beare-Stevenson cutis gyrata, Jackson-Weiss, Bent Bone Dysplasia, and Seathre-Chotzen-like syndromes. The majority of these mutations are missense mutations that result in constitutive activation of the receptor and downstream molecular pathways. Treatment involves a multidisciplinary approach with ultimate surgical fixation of the cranial deformity to prevent further sequelae. Understanding the molecular mechanisms has allowed for the investigation of different therapeutic agents that can potentially be used to prevent the disorders. Further research efforts are need to better understand screening and effective methods of early intervention and prevention. Herein, the authors provide a comprehensive update on FGFR2-related syndromic craniosynostosis.
Collapse
Affiliation(s)
- Saïd C. Azoury
- Department of Surgery, The Johns Hopkins Hospital, Baltimore, MD, USA
| | - Sashank Reddy
- Department of Plastic and Reconstructive Surgery, The Johns Hopkins Hospital, Baltimore, MD, USA
| | - Vivek Shukla
- TGIB, NCI, National Institutes of Health, Bethesda, MD, USA
| | - Chu-Xia Deng
- Faculty of Health Sciences, University of Macau, Macau SAR, China
| |
Collapse
|
26
|
Lattanzi W, Barba M, Di Pietro L, Boyadjiev SA. Genetic advances in craniosynostosis. Am J Med Genet A 2017; 173:1406-1429. [PMID: 28160402 DOI: 10.1002/ajmg.a.38159] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 12/30/2016] [Accepted: 01/06/2017] [Indexed: 12/22/2022]
Abstract
Craniosynostosis, the premature ossification of one or more skull sutures, is a clinically and genetically heterogeneous congenital anomaly affecting approximately one in 2,500 live births. In most cases, it occurs as an isolated congenital anomaly, that is, nonsyndromic craniosynostosis (NCS), the genetic, and environmental causes of which remain largely unknown. Recent data suggest that, at least some of the midline NCS cases may be explained by two loci inheritance. In approximately 25-30% of patients, craniosynostosis presents as a feature of a genetic syndrome due to chromosomal defects or mutations in genes within interconnected signaling pathways. The aim of this review is to provide a detailed and comprehensive update on the genetic and environmental factors associated with NCS, integrating the scientific findings achieved during the last decade. Focus on the neurodevelopmental, imaging, and treatment aspects of NCS is also provided.
Collapse
Affiliation(s)
- Wanda Lattanzi
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy.,Latium Musculoskeletal Tıssue Bank, Rome, Italy
| | - Marta Barba
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Lorena Di Pietro
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Simeon A Boyadjiev
- Division of Genomic Medicine, Department of Pediatrics, Davis Medical Center, University of California, Sacramento, California
| |
Collapse
|
27
|
Norrie JL, Li Q, Co S, Huang BL, Ding D, Uy JC, Ji Z, Mackem S, Bedford MT, Galli A, Ji H, Vokes SA. PRMT5 is essential for the maintenance of chondrogenic progenitor cells in the limb bud. Development 2016; 143:4608-4619. [PMID: 27827819 DOI: 10.1242/dev.140715] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 10/24/2016] [Indexed: 12/13/2022]
Abstract
During embryonic development, undifferentiated progenitor cells balance the generation of additional progenitor cells with differentiation. Within the developing limb, cartilage cells differentiate from mesodermal progenitors in an ordered process that results in the specification of the correct number of appropriately sized skeletal elements. The internal pathways by which these cells maintain an undifferentiated state while preserving their capacity to differentiate is unknown. Here, we report that the arginine methyltransferase PRMT5 has a crucial role in maintaining progenitor cells. Mouse embryonic buds lacking PRMT5 have severely truncated bones with wispy digits lacking joints. This novel phenotype is caused by widespread cell death that includes mesodermal progenitor cells that have begun to precociously differentiate into cartilage cells. We propose that PRMT5 maintains progenitor cells through its regulation of Bmp4 Intriguingly, adult and embryonic stem cells also require PRMT5 for maintaining pluripotency, suggesting that similar mechanisms might regulate lineage-restricted progenitor cells during organogenesis.
Collapse
Affiliation(s)
- Jacqueline L Norrie
- Department of Molecular Biosciences, University of Texas at Austin, 2500 Speedway Stop A4800, Austin, TX 78712, USA
| | - Qiang Li
- Department of Molecular Biosciences, University of Texas at Austin, 2500 Speedway Stop A4800, Austin, TX 78712, USA
| | - Swanie Co
- Department of Molecular Biosciences, University of Texas at Austin, 2500 Speedway Stop A4800, Austin, TX 78712, USA
| | - Bau-Lin Huang
- Cancer and Developmental Biology Laboratory, CCR, NCI, Frederick, MD 21702, USA
| | - Ding Ding
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, 615 North Wolfe Street, Room E3638, Baltimore, MD 21205, USA
| | - Jann C Uy
- Department of Molecular Biosciences, University of Texas at Austin, 2500 Speedway Stop A4800, Austin, TX 78712, USA
| | - Zhicheng Ji
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, 615 North Wolfe Street, Room E3638, Baltimore, MD 21205, USA
| | - Susan Mackem
- Cancer and Developmental Biology Laboratory, CCR, NCI, Frederick, MD 21702, USA
| | - Mark T Bedford
- Department of Epigenetics & Molecular Carcinogenesis, M.D. Anderson Cancer Center, 1808 Park Road 1C (P.O. Box 389), Smithville, TX 78957, USA
| | - Antonella Galli
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, UK
| | - Hongkai Ji
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, 615 North Wolfe Street, Room E3638, Baltimore, MD 21205, USA
| | - Steven A Vokes
- Department of Molecular Biosciences, University of Texas at Austin, 2500 Speedway Stop A4800, Austin, TX 78712, USA
| |
Collapse
|
28
|
Kawao N, Yano M, Tamura Y, Okumoto K, Okada K, Kaji H. Role of osteoclasts in heterotopic ossification enhanced by fibrodysplasia ossificans progressiva-related activin-like kinase 2 mutation in mice. J Bone Miner Metab 2016. [PMID: 26204847 DOI: 10.1007/s00774-015-0701-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Fibrodysplasia ossificans progressiva (FOP) is a disorder of skeletal malformations and progressive heterotopic ossification. The constitutively activating mutation (R206H) of the bone morphogenetic protein type 1 receptor, activin-like kinase 2 (ALK2), is responsible for the pathogenesis of FOP. Although transfection of the causal mutation of FOP into myoblasts enhances osteoclast formation by transforming growth factor-β (TGF-β), the role of osteoclasts in heterotopic ossification is unknown. We therefore examined the effects of alendronate, SB431542 and SB203580 on heterotopic ossification induced by the causal mutation of FOP. Total bone mineral content as well as numbers of tartrate-resistant acid phosphatase (TRAP)-positive multinucleated and alkaline phosphatase (ALP)-positive cells in heterotopic bone were significantly higher in muscle tissues implanted with ALK2 (R206H)-transfected mouse myoblastic C2C12 cells than in the tissues implanted with empty vector-transfected cells in nude mice. Alendronate, an aminobisphosphonate, did not affect total mineral content or numbers of TRAP-positive multinucleated and ALP-positive cells in heterotopic bone, which were enhanced by the implantation of ALK2 (R206H)-transfected C2C12 cells, although it significantly decreased serum levels of cross-linked C-telopeptide of type I collagen, a bone resorption index. Moreover, neither SB431542, an inhibitor of TGF-β receptor type I kinase, nor SB203580, an inhibitor of p38 mitogen-activated protein kinase, affected the increase in heterotopic ossification due to the implantation of ALK2 (R206H)-transfected C2C12 cells. In conclusion, the present study indicates that osteoclast inhibition does not affect heterotopic ossification enhanced by FOP-related mutation.
Collapse
Affiliation(s)
- Naoyuki Kawao
- Department of Physiology and Regenerative Medicine, Kinki University Faculty of Medicine, 377-2 Ohnohigashi, Osakasayama, 589-8511, Osaka, Japan
| | - Masato Yano
- Department of Physiology and Regenerative Medicine, Kinki University Faculty of Medicine, 377-2 Ohnohigashi, Osakasayama, 589-8511, Osaka, Japan
| | - Yukinori Tamura
- Department of Physiology and Regenerative Medicine, Kinki University Faculty of Medicine, 377-2 Ohnohigashi, Osakasayama, 589-8511, Osaka, Japan
| | - Katsumi Okumoto
- Life Science Research Institute, Kinki University, Osaka, Japan
| | - Kiyotaka Okada
- Department of Physiology and Regenerative Medicine, Kinki University Faculty of Medicine, 377-2 Ohnohigashi, Osakasayama, 589-8511, Osaka, Japan
| | - Hiroshi Kaji
- Department of Physiology and Regenerative Medicine, Kinki University Faculty of Medicine, 377-2 Ohnohigashi, Osakasayama, 589-8511, Osaka, Japan.
| |
Collapse
|
29
|
Tracheal cartilaginous sleeves in children with syndromic craniosynostosis. Genet Med 2016; 19:62-68. [PMID: 27228464 DOI: 10.1038/gim.2016.60] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 03/29/2016] [Indexed: 11/09/2022] Open
Abstract
PURPOSE Because a tracheal cartilaginous sleeve (TCS) confers a significant mortality risk that can be mitigated with appropriate intervention, we sought to describe the prevalence and associated genotypes in a large cohort of children with syndromic craniosynostosis. METHODS Chart review of patients with syndromic craniosynostosis across two institutions. RESULTS In a cohort of 86 patients with syndromic craniosynostosis, 31 required airway evaluation under anesthesia. TCS was found in 19, for an overall prevalence of 22%. FGFR2, TWIST1, and FGFR3 mutations were identified in children with TCS. All five children with a W290C mutation in FGFR2 had TCS, and most previously reported children with W290C had identification of TCS or early death. In contrast, TCS was not associated with other mutations at residue 290. CONCLUSION There is an association between TCS and syndromic craniosynostosis, and it appears to be particularly high in individuals with the W290C mutation in FGFR2. Referral to a pediatric otolaryngologist and consideration of operative airway evaluation (i.e., bronchoscopy or rigid endoscopy) in all patients with syndromic craniosynostosis should be considered to evaluate for TCS. Results from genetic testing may help providers weigh the risks and benefits of early airway evaluation and intervention in children with higher-risk genotypes.Genet Med 19 1, 62-68.
Collapse
|
30
|
Abstract
The fibroblast growth factor (Fgf) family of ligands and receptor tyrosine kinases is required throughout embryonic and postnatal development and also regulates multiple homeostatic functions in the adult. Aberrant Fgf signaling causes many congenital disorders and underlies multiple forms of cancer. Understanding the mechanisms that govern Fgf signaling is therefore important to appreciate many aspects of Fgf biology and disease. Here we review the mechanisms of Fgf signaling by focusing on genetic strategies that enable in vivo analysis. These studies support an important role for Erk1/2 as a mediator of Fgf signaling in many biological processes but have also provided strong evidence for additional signaling pathways in transmitting Fgf signaling in vivo.
Collapse
Affiliation(s)
- J Richard Brewer
- Department of Developmental and Regenerative Biology, Tisch Cancer Institute, Icahn School of Medicine at Mt. Sinai, New York, New York 10029, USA
| | - Pierre Mazot
- Department of Developmental and Regenerative Biology, Tisch Cancer Institute, Icahn School of Medicine at Mt. Sinai, New York, New York 10029, USA
| | - Philippe Soriano
- Department of Developmental and Regenerative Biology, Tisch Cancer Institute, Icahn School of Medicine at Mt. Sinai, New York, New York 10029, USA
| |
Collapse
|
31
|
Hu P, Huang BY, Xia X, Xuan Q, Hu B, Qin YH. Therapeutic effect of CNP on renal osteodystrophy by antagonizing the FGF-23/MAPK pathway. J Recept Signal Transduct Res 2015; 36:213-9. [DOI: 10.3109/10799893.2015.1075041] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
32
|
Twigg SRF, Wilkie AOM. A Genetic-Pathophysiological Framework for Craniosynostosis. Am J Hum Genet 2015; 97:359-77. [PMID: 26340332 PMCID: PMC4564941 DOI: 10.1016/j.ajhg.2015.07.006] [Citation(s) in RCA: 174] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Accepted: 07/14/2015] [Indexed: 12/24/2022] Open
Abstract
Craniosynostosis, the premature fusion of one or more cranial sutures of the skull, provides a paradigm for investigating the interplay of genetic and environmental factors leading to malformation. Over the past 20 years molecular genetic techniques have provided a new approach to dissect the underlying causes; success has mostly come from investigation of clinical samples, and recent advances in high-throughput DNA sequencing have dramatically enhanced the study of the human as the preferred "model organism." In parallel, however, we need a pathogenetic classification to describe the pathways and processes that lead to cranial suture fusion. Given the prenatal onset of most craniosynostosis, investigation of mechanisms requires more conventional model organisms; principally the mouse, because of similarities in cranial suture development. We present a framework for classifying genetic causes of craniosynostosis based on current understanding of cranial suture biology and molecular and developmental pathogenesis. Of note, few pathologies result from complete loss of gene function. Instead, biochemical mechanisms involving haploinsufficiency, dominant gain-of-function and recessive hypomorphic mutations, and an unusual X-linked cellular interference process have all been implicated. Although few of the genes involved could have been predicted based on expression patterns alone (because the genes play much wider roles in embryonic development or cellular homeostasis), we argue that they fit into a limited number of functional modules active at different stages of cranial suture development. This provides a useful approach both when defining the potential role of new candidate genes in craniosynostosis and, potentially, for devising pharmacological approaches to therapy.
Collapse
Affiliation(s)
- Stephen R F Twigg
- Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Headington, Oxford OX3 9DS, UK
| | - Andrew O M Wilkie
- Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Headington, Oxford OX3 9DS, UK; Craniofacial Unit, Department of Plastic and Reconstructive Surgery, Oxford University Hospitals NHS Trust, John Radcliffe Hospital, Headington, Oxford OX3 9DU, UK.
| |
Collapse
|
33
|
Xie Y, Zhou S, Chen H, Du X, Chen L. Recent research on the growth plate: Advances in fibroblast growth factor signaling in growth plate development and disorders. J Mol Endocrinol 2014; 53:T11-34. [PMID: 25114206 DOI: 10.1530/jme-14-0012] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Skeletons are formed through two distinct developmental actions, intramembranous ossification and endochondral ossification. During embryonic development, most bone is formed by endochondral ossification. The growth plate is the developmental center for endochondral ossification. Multiple signaling pathways participate in the regulation of endochondral ossification. Fibroblast growth factor (FGF)/FGF receptor (FGFR) signaling has been found to play a vital role in the development and maintenance of growth plates. Missense mutations in FGFs and FGFRs can cause multiple genetic skeletal diseases with disordered endochondral ossification. Clarifying the molecular mechanisms of FGFs/FGFRs signaling in skeletal development and genetic skeletal diseases will have implications for the development of therapies for FGF-signaling-related skeletal dysplasias and growth plate injuries. In this review, we summarize the recent advances in elucidating the role of FGFs/FGFRs signaling in growth plate development, genetic skeletal disorders, and the promising therapies for those genetic skeletal diseases resulting from FGFs/FGFRs dysfunction. Finally, we also examine the potential important research in this field in the future.
Collapse
Affiliation(s)
- Yangli Xie
- Department of Rehabilitation MedicineCenter of Bone Metabolism and Repair, Trauma Center, State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Siru Zhou
- Department of Rehabilitation MedicineCenter of Bone Metabolism and Repair, Trauma Center, State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Hangang Chen
- Department of Rehabilitation MedicineCenter of Bone Metabolism and Repair, Trauma Center, State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Xiaolan Du
- Department of Rehabilitation MedicineCenter of Bone Metabolism and Repair, Trauma Center, State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Lin Chen
- Department of Rehabilitation MedicineCenter of Bone Metabolism and Repair, Trauma Center, State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| |
Collapse
|
34
|
Closing the Gap: Genetic and Genomic Continuum from Syndromic to Nonsyndromic Craniosynostoses. CURRENT GENETIC MEDICINE REPORTS 2014; 2:135-145. [PMID: 26146596 DOI: 10.1007/s40142-014-0042-x] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Craniosynostosis, a condition that includes the premature fusion of one or multiple cranial sutures, is a relatively common birth defect in humans and the second most common craniofacial anomaly after orofacial clefts. There is a significant clinical variation among different sutural synostoses as well as significant variation within any given single-suture synostosis. Craniosynostosis can be isolated (i.e., nonsyndromic) or occurs as part of a genetic syndrome (e.g., Crouzon, Pfeiffer, Apert, Muenke, and Saethre-Chotzen syndromes). Approximately 85 % of all cases of craniosynostosis are nonsyndromic. Several recent genomic discoveries are elucidating the genetic basis for nonsyndromic cases and implicate the newly identified genes in signaling pathways previously found in syndromic craniosynostosis. Published epidemiologic and phenotypic studies clearly demonstrate that nonsyndromic craniosynostosis is a complex and heterogeneous condition supporting a strong genetic component accompanied by environmental factors that contribute to the pathogenetic network of this birth defect. Large population, rather than single-clinic or hospital-based studies is required with phenotypically homogeneous subsets of patients to further understand the complex genetic, maternal, environmental, and stochastic factors contributing to nonsyndromic craniosynostosis. Learning about these variables is a key in formulating the basis of multidisciplinary and lifelong care for patients with these conditions.
Collapse
|
35
|
Heuzé Y, Singh N, Basilico C, Jabs EW, Holmes G, Richtsmeier JT. Morphological comparison of the craniofacial phenotypes of mouse models expressing the Apert FGFR2 S252W mutation in neural crest- or mesoderm-derived tissues. Bone 2014; 63:101-9. [PMID: 24632501 PMCID: PMC4018479 DOI: 10.1016/j.bone.2014.03.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Revised: 03/04/2014] [Accepted: 03/06/2014] [Indexed: 11/20/2022]
Abstract
Bones of the craniofacial skeleton are derived from two distinct cell lineages, cranial neural crest and mesoderm, and articulate at sutures and synchondroses which represent major bone growth sites. Premature fusion of cranial suture(s) is associated with craniofacial dysmorphogenesis caused in part by alteration in the growth potential at sutures and can occur as an isolated birth defect or as part of a syndrome, such as Apert syndrome. Conditional expression of the Apert FGFR2 S252W mutation in cells derived from mesoderm was previously shown to be necessary and sufficient to cause coronal craniosynostosis. Here we used micro computed tomography images of mice expressing the Apert mutation constitutively in either mesoderm- or neural crest-derived cells to quantify craniofacial shape variation and suture fusion patterns, and to identify shape changes in craniofacial bones derived from the lineage not expressing the mutation, referred to here as secondary shape changes. Our results show that at postnatal day 0: (i) conditional expression of the FGFR2 S252W mutation in neural crest-derived tissues causes a more severe craniofacial phenotype than when expressed in mesoderm-derived tissues; and (ii) both mesoderm- and neural crest-specific mouse models display secondary shape changes. We also show that premature suture fusion is not necessarily dependent on the expression of the FGFR2 S252W mutation in the sutural mesenchyme. More specifically, it appears that suture fusion patterns in both mouse models are suture-specific resulting from a complex combination of the influence of primary abnormalities of biogenesis or signaling within the sutures, and timing.
Collapse
Affiliation(s)
- Yann Heuzé
- Department of Anthropology, Pennsylvania State University, University Park, PA, USA
| | - Nandini Singh
- Department of Anthropology, Pennsylvania State University, University Park, PA, USA
| | - Claudio Basilico
- Department of Microbiology, New York University School of Medicine, New York, NY, USA
| | - Ethylin Wang Jabs
- Department of Genetics and Genomic Sciences, Mount Sinai School of Medicine, New York, NY, USA
| | - Greg Holmes
- Department of Genetics and Genomic Sciences, Mount Sinai School of Medicine, New York, NY, USA
| | - Joan T Richtsmeier
- Department of Anthropology, Pennsylvania State University, University Park, PA, USA.
| |
Collapse
|
36
|
Su N, Jin M, Chen L. Role of FGF/FGFR signaling in skeletal development and homeostasis: learning from mouse models. Bone Res 2014; 2:14003. [PMID: 26273516 PMCID: PMC4472122 DOI: 10.1038/boneres.2014.3] [Citation(s) in RCA: 195] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2013] [Revised: 11/29/2013] [Accepted: 12/03/2013] [Indexed: 01/06/2023] Open
Abstract
Fibroblast growth factor (FGF)/fibroblast growth factor receptor (FGFR) signaling plays essential roles in bone development and diseases. Missense mutations in FGFs and FGFRs in humans can cause various congenital bone diseases, including chondrodysplasia syndromes, craniosynostosis syndromes and syndromes with dysregulated phosphate metabolism. FGF/FGFR signaling is also an important pathway involved in the maintenance of adult bone homeostasis. Multiple kinds of mouse models, mimicking human skeleton diseases caused by missense mutations in FGFs and FGFRs, have been established by knock-in/out and transgenic technologies. These genetically modified mice provide good models for studying the role of FGF/FGFR signaling in skeleton development and homeostasis. In this review, we summarize the mouse models of FGF signaling-related skeleton diseases and recent progresses regarding the molecular mechanisms, underlying the role of FGFs/FGFRs in the regulation of bone development and homeostasis. This review also provides a perspective view on future works to explore the roles of FGF signaling in skeletal development and homeostasis.
Collapse
Affiliation(s)
- Nan Su
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Institute of Surgery Research, Daping Hospital, Third Military Medical University , Chongqing, 400042, China
| | - Min Jin
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Institute of Surgery Research, Daping Hospital, Third Military Medical University , Chongqing, 400042, China
| | - Lin Chen
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Institute of Surgery Research, Daping Hospital, Third Military Medical University , Chongqing, 400042, China
| |
Collapse
|
37
|
Motch Perrine SM, Cole TM, Martínez-Abadías N, Aldridge K, Jabs EW, Richtsmeier JT. Craniofacial divergence by distinct prenatal growth patterns in Fgfr2 mutant mice. BMC DEVELOPMENTAL BIOLOGY 2014; 14:8. [PMID: 24580805 PMCID: PMC4101838 DOI: 10.1186/1471-213x-14-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Accepted: 02/05/2014] [Indexed: 11/12/2022]
Abstract
Background Differences in cranial morphology arise due to changes in fundamental cell processes like migration, proliferation, differentiation and cell death driven by genetic programs. Signaling between fibroblast growth factors (FGFs) and their receptors (FGFRs) affect these processes during head development and mutations in FGFRs result in congenital diseases including FGFR-related craniosynostosis syndromes. Current research in model organisms focuses primarily on how these mutations change cell function local to sutures under the hypothesis that prematurely closing cranial sutures contribute to skull dysmorphogenesis. Though these studies have provided fundamentally important information contributing to the understanding of craniosynostosis conditions, knowledge of changes in cell function local to the sutures leave change in overall three-dimensional cranial morphology largely unexplained. Here we investigate growth of the skull in two inbred mouse models each carrying one of two gain-of-function mutations in FGFR2 on neighboring amino acids (S252W and P253R) that in humans cause Apert syndrome, one of the most severe FGFR-related craniosynostosis syndromes. We examine late embryonic skull development and suture patency in Fgfr2 Apert syndrome mice between embryonic day 17.5 and birth and quantify the effects of these mutations on 3D skull morphology, suture patency and growth. Results We show in mice what studies in humans can only infer: specific cranial growth deviations occur prenatally and worsen with time in organisms carrying these FGFR2 mutations. We demonstrate that: 1) distinct skull morphologies of each mutation group are established by E17.5; 2) cranial suture patency patterns differ between mice carrying these mutations and their unaffected littermates; 3) the prenatal skull grows differently in each mutation group; and 4) unique Fgfr2-related cranial morphologies are exacerbated by late embryonic growth patterns. Conclusions Our analysis of mutation-driven changes in cranial growth provides a previously missing piece of knowledge necessary for explaining variation in emergent cranial morphologies and may ultimately be helpful in managing human cases carrying these same mutations. This information is critical to the understanding of craniofacial development, disease and evolution and may contribute to the evaluation of incipient therapeutic strategies.
Collapse
Affiliation(s)
| | | | | | | | | | - Joan T Richtsmeier
- Department of Anthropology, Pennsylvania State University, University Park, PA 16802, USA.
| |
Collapse
|
38
|
Rachwalski M, Wollnik B, Kress W. Klinik und Genetik syndromaler und nichtsyndromaler Kraniosynostosen. MED GENET-BERLIN 2013. [DOI: 10.1007/s11825-013-0412-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Zusammenfassung
Kraniosynostosen gehören mit einer Inzidenz von 1:2000 bis 1:3000 Geburten zu den häufigsten kraniofazialen Anomalien. Die durch die vorzeitige Verknöcherung einer oder mehrerer Schädelnähte verursachte Wachstumshemmung kann zu schweren Deformitäten des Schädel- und Gesichtsskeletts führen. Dies sorgt nicht nur für eine große ästhetische Beeinträchtigung, sondern hat auch funktionelle Auswirkungen für die Patienten. Hierzu können u. a. gehören: intrakranielle Drucksteigerung, Atrophie des N. opticus, Atem-, Hör- und Entwicklungsstörungen. Trotz großer Anstrengungen konnten bisher nur für einen Teil der autosomal-dominanten syndromalen Kraniosynostosen die ursächlichen Gene, z. B „fibroblast growth factor receptor 1-3“ (FGFR1-3), „twist basic helix-loop-helix transcription factor 1“ (TWIST1) etc., gefunden werden. Die Ätiologie der nichtsyndromalen Kraniosynostosen bleibt weiterhin ungeklärt. Aufgrund der verbreiteten Anwendung neuer Sequenziertechnologien zur Identifizierung neuer kausaler Gene bei Patienten mit Kraniosynostose kann in den nächsten Jahren mit der Entschlüsselung vieler weiterer krankheitsverursachender Gene gerechnet werden. Insbesondere die syndromalen Formen der Kraniosynostose bedürfen aufgrund ihrer klinischen Komplexität einer interdisziplinären Betreuung. Die einzige Therapieoption besteht derzeit in der kraniofazialen Chirurgie, welche aber die genetisch determinierten pathologischen Wachstumsmuster der komplexen syndromalen Kraniosynostosen langfristig oft nicht beheben kann.
Collapse
Affiliation(s)
- M. Rachwalski
- Aff1 grid.411097.a 000000008852305X Institut für Humangenetik Uniklinik Köln Kerpener Str. 34 50931 Köln Deutschland
| | - B. Wollnik
- Aff1 grid.411097.a 000000008852305X Institut für Humangenetik Uniklinik Köln Kerpener Str. 34 50931 Köln Deutschland
| | - W. Kress
- Aff2 grid.8379.5 0000000119588658 Institut für Humangenetik Universität Würzburg Würzburg Deutschland
| |
Collapse
|
39
|
Richtsmeier JT, Flaherty K. Hand in glove: brain and skull in development and dysmorphogenesis. Acta Neuropathol 2013; 125:469-89. [PMID: 23525521 PMCID: PMC3652528 DOI: 10.1007/s00401-013-1104-y] [Citation(s) in RCA: 159] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2012] [Revised: 02/28/2013] [Accepted: 03/02/2013] [Indexed: 01/02/2023]
Abstract
The brain originates relatively early in development from differentiated ectoderm that forms a hollow tube and takes on an exceedingly complex shape with development. The skull is made up of individual bony elements that form from neural crest- and mesoderm-derived mesenchyme that unite to provide support and protection for soft tissues and spaces of the head. The meninges provide a protective and permeable membrane between brain and skull. Across evolutionary and developmental time, dynamic changes in brain and skull shape track one another so that their integration is evidenced in two structures that fit soundly regardless of changes in biomechanical and physiologic functions. Evidence for this tight correspondence is also seen in diseases of the craniofacial complex that are often classified as diseases of the skull (e.g., craniosynostosis) or diseases of the brain (e.g., holoprosencephaly) even when both tissues are affected. Our review suggests a model that links brain and skull morphogenesis through coordinated integration of signaling pathways (e.g., FGF, TGFβ, Wnt) via processes that are not currently understood, perhaps involving the meninges. Differences in the earliest signaling of biological structure establish divergent designs that will be enhanced during morphogenesis. Signaling systems that pattern the developing brain are also active in patterning required for growth and assembly of the skull and some members of these signaling families have been indicated as causal for craniofacial diseases. Because cells of early brain and skull are sensitive to similar signaling families, variation in the strength or timing of signals or shifts in patterning boundaries that affect one system (neural or skull) could also affect the other system and appropriate co-adjustments in development would be made. Interactions of these signaling systems and of the tissues that they pattern are fundamental to the consistent but labile functional and structural association of brain and skull conserved over evolutionary time obvious in the study of development and disease.
Collapse
Affiliation(s)
- Joan T Richtsmeier
- Department of Anthropology, Pennsylvania State University, 409 Carpenter Building, University Park, PA 16802, USA.
| | | |
Collapse
|
40
|
The role of vertebrate models in understanding craniosynostosis. Childs Nerv Syst 2012; 28:1471-81. [PMID: 22872264 DOI: 10.1007/s00381-012-1844-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2012] [Accepted: 06/13/2012] [Indexed: 01/10/2023]
Abstract
BACKGROUND Craniosynostosis (CS), the premature fusion of cranial sutures, is a relatively common pediatric anomaly, occurring in isolation or as part of a syndrome. A growing number of genes with pathologic mutations have been identified for syndromic and nonsyndromic CS. The study of human sutural material obtained post-operatively is not sufficient to understand the etiology of CS, for which animal models are indispensable. DISCUSSION The similarity of the human and murine calvarial structure, our knowledge of mouse genetics and biology, and ability to manipulate the mouse genome make the mouse the most valuable model organism for CS research. A variety of mouse mutants are available that model specific human CS mutations or have CS phenotypes. These allow characterization of the biochemical and morphological events, often embryonic, which precede suture fusion. Other vertebrate organisms have less functional genetic utility than mice, but the rat, rabbit, chick, zebrafish, and frog provide alternative systems in which to validate or contrast molecular functions relevant to CS.
Collapse
|
41
|
Percival CJ, Wang Y, Zhou X, Jabs EW, Richtsmeier JT. The effect of a Beare-Stevenson syndrome Fgfr2 Y394C mutation on early craniofacial bone volume and relative bone mineral density in mice. J Anat 2012; 221:434-42. [PMID: 22881429 DOI: 10.1111/j.1469-7580.2012.01555.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/24/2012] [Indexed: 12/30/2022] Open
Abstract
Quantifying the craniofacial skeletal phenotype during development highlights potential effects of known mutations on bone maturation and is an informative first step for the analysis of animal models. We introduce a novel technique to easily and efficiently quantify individual cranial bone volume and relative bone mineral density across the murine skull from high resolution computed tomography images. The approach can be combined with existing quantitative morphometric methods to provide details of bone growth and bone quality, which can be used to make inferences about regulatory effects local to individual bones and identify locations and developmental times for which additional analyses are warranted. Analysis of the Fgfr2(+/Y394C) mouse model of Beare-Stevenson cutis gyrata syndrome, an FGFR-related craniosynostosis syndrome, is used to demonstrate the method. Mutants and unaffected littermates display similar bone volume and relative bone density at birth, followed by significant differences at postnatal day eight. The change in rates of bone volume growth occurs similarly for all bones of the skull, regardless of origin, location or association with craniosynostosis. These results suggest an association between low bone density, low bone volume, and Fgfr craniosynostosis mutations. Our novel technique provides an initial quantitative evaluation of local shifts in bone maturation across the skull of animal models.
Collapse
|