51
|
Xie Y, Su N, Yang J, Tan Q, Huang S, Jin M, Ni Z, Zhang B, Zhang D, Luo F, Chen H, Sun X, Feng JQ, Qi H, Chen L. FGF/FGFR signaling in health and disease. Signal Transduct Target Ther 2020; 5:181. [PMID: 32879300 PMCID: PMC7468161 DOI: 10.1038/s41392-020-00222-7] [Citation(s) in RCA: 454] [Impact Index Per Article: 90.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 05/28/2020] [Accepted: 06/15/2020] [Indexed: 12/13/2022] Open
Abstract
Growing evidences suggest that the fibroblast growth factor/FGF receptor (FGF/FGFR) signaling has crucial roles in a multitude of processes during embryonic development and adult homeostasis by regulating cellular lineage commitment, differentiation, proliferation, and apoptosis of various types of cells. In this review, we provide a comprehensive overview of the current understanding of FGF signaling and its roles in organ development, injury repair, and the pathophysiology of spectrum of diseases, which is a consequence of FGF signaling dysregulation, including cancers and chronic kidney disease (CKD). In this context, the agonists and antagonists for FGF-FGFRs might have therapeutic benefits in multiple systems.
Collapse
Affiliation(s)
- Yangli Xie
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China.
| | - Nan Su
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Jing Yang
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Qiaoyan Tan
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Shuo Huang
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Min Jin
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Zhenhong Ni
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Bin Zhang
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Dali Zhang
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Fengtao Luo
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Hangang Chen
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Xianding Sun
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Jian Q Feng
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX, 75246, USA
| | - Huabing Qi
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China.
| | - Lin Chen
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China.
| |
Collapse
|
52
|
Dambroise E, Ktorza I, Brombin A, Abdessalem G, Edouard J, Luka M, Fiedler I, Binder O, Pelle O, Patton EE, Busse B, Menager M, Sohm F, Legeai-Mallet L. Fgfr3 Is a Positive Regulator of Osteoblast Expansion and Differentiation During Zebrafish Skull Vault Development. J Bone Miner Res 2020; 35:1782-1797. [PMID: 32379366 DOI: 10.1002/jbmr.4042] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 04/09/2020] [Accepted: 04/27/2020] [Indexed: 12/20/2022]
Abstract
Gain or loss-of-function mutations in fibroblast growth factor receptor 3 (FGFR3) result in cranial vault defects highlighting the protein's role in membranous ossification. Zebrafish express high levels of fgfr3 during skull development; in order to study FGFR3's role in cranial vault development, we generated the first fgfr3 loss-of-function zebrafish (fgfr3lof/lof ). The mutant fish exhibited major changes in the craniofacial skeleton, with a lack of sutures, abnormal frontal and parietal bones, and the presence of ectopic bones. Integrated analyses (in vivo imaging and single-cell RNA sequencing of the osteoblast lineage) of zebrafish fgfr3lof/lof revealed a delay in osteoblast expansion and differentiation, together with changes in the extracellular matrix. These findings demonstrate that fgfr3 is a positive regulator of osteogenesis. We conclude that changes in the extracellular matrix within growing bone might impair cell-cell communication, mineralization, and new osteoblast recruitment. © 2020 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Emilie Dambroise
- Laboratory of Molecular and Physiopathological Bases of Osteochondrodysplasia, INSERM UMR 1163, Université de Paris, Imagine Institute, Paris, France
| | - Ivan Ktorza
- Laboratory of Molecular and Physiopathological Bases of Osteochondrodysplasia, INSERM UMR 1163, Université de Paris, Imagine Institute, Paris, France
| | - Alessandro Brombin
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK.,Cancer Research UK Edinburgh Centre, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Ghaith Abdessalem
- Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, INSERM UMR 1163, Université de Paris, Imagine Institute, Paris, France
| | - Joanne Edouard
- UMS AMAGEN, CNRS, INRA, Université Paris-Saclay, Gif-sur-Yvette, France.,Institute for Integrative Biology of the Cell (I2BC)-CNRS, Gif-sur-Yvette, France
| | - Marine Luka
- Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, INSERM UMR 1163, Université de Paris, Imagine Institute, Paris, France
| | - Imke Fiedler
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Olivia Binder
- Laboratory of Molecular and Physiopathological Bases of Osteochondrodysplasia, INSERM UMR 1163, Université de Paris, Imagine Institute, Paris, France
| | - Olivier Pelle
- Flow Cytometry Core Facility, Structure Fédérative de Recherche Necker, INSERM US24/CNRS UMS3633, Paris, France
| | - E Elizabeth Patton
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK.,Cancer Research UK Edinburgh Centre, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Björn Busse
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Mickaël Menager
- Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, INSERM UMR 1163, Université de Paris, Imagine Institute, Paris, France
| | - Frederic Sohm
- UMS AMAGEN, CNRS, INRA, Université Paris-Saclay, Gif-sur-Yvette, France.,Institute for Integrative Biology of the Cell (I2BC)-CNRS, Gif-sur-Yvette, France.,Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Flow Cytometry Core Facility, Structure Fédérative de Recherche Necker, INSERM US24/CNRS UMS3633, Paris, France.,Functional Genomics Institute of Lyon, University of Lyon, CNRS, INRA, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Laurence Legeai-Mallet
- Laboratory of Molecular and Physiopathological Bases of Osteochondrodysplasia, INSERM UMR 1163, Université de Paris, Imagine Institute, Paris, France
| |
Collapse
|
53
|
Thorup AS, Dell'Accio F, Eldridge SE. Lessons from joint development for cartilage repair in the clinic. Dev Dyn 2020; 250:360-376. [PMID: 32738003 DOI: 10.1002/dvdy.228] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 07/23/2020] [Accepted: 07/24/2020] [Indexed: 12/19/2022] Open
Abstract
More than 250 years ago, William Hunter stated that when cartilage is destroyed it never recovers. In the last 20 years, the understanding of the mechanisms that lead to joint formation and the knowledge that some of these mechanisms are reactivated in the homeostatic responses of cartilage to injury has offered an unprecedented therapeutic opportunity to achieve cartilage regeneration. Very large investments in ambitious clinical trials are finally revealing that, although we do not have perfect medicines yet, disease modification is a feasible possibility for human osteoarthritis.
Collapse
Affiliation(s)
- Anne-Sophie Thorup
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Francesco Dell'Accio
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Suzanne E Eldridge
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
54
|
Sun X, Zhang R, Chen H, Du X, Chen S, Huang J, Liu M, Xu M, Luo F, Jin M, Su N, Qi H, Yang J, Tan Q, Zhang D, Ni Z, Liang S, Zhang B, Chen D, Zhang X, Luo L, Chen L, Xie Y. Fgfr3 mutation disrupts chondrogenesis and bone ossification in zebrafish model mimicking CATSHL syndrome partially via enhanced Wnt/β-catenin signaling. Theranostics 2020; 10:7111-7130. [PMID: 32641982 PMCID: PMC7330844 DOI: 10.7150/thno.45286] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 05/21/2020] [Indexed: 12/12/2022] Open
Abstract
CATSHL syndrome, characterized by camptodactyly, tall stature and hearing loss, is caused by loss-of-function mutations of fibroblast growth factor receptors 3 (FGFR3) gene. Most manifestations of patients with CATSHL syndrome start to develop in the embryonic stage, such as skeletal overgrowth, craniofacial abnormalities, however, the pathogenesis of these phenotypes especially the early maldevelopment remains incompletely understood. Furthermore, there are no effective therapeutic targets for this skeleton dysplasia. Methods: We generated fgfr3 knockout zebrafish by CRISPR/Cas9 technology to study the developmental mechanisms and therapeutic targets of CATSHL syndrome. Several zebrafish transgenic lines labeling osteoblasts and chondrocytes, and live Alizarin red staining were used to analyze the dynamical skeleton development in fgfr3 mutants. Western blotting, whole mount in situ hybridization, Edu labeling based cell proliferation assay and Wnt/β-catenin signaling antagonist were used to explore the potential mechanisms and therapeutic targets. Results: We found that fgfr3 mutant zebrafish, staring from early development stage, showed craniofacial bone malformation with microcephaly and delayed closure of cranial sutures, chondroma-like lesion and abnormal development of auditory sensory organs, partially resembling the clinical manifestations of patients with CATSHL syndrome. Further studies showed that fgfr3 regulates the patterning and shaping of pharyngeal arches and the timely ossification of craniofacial skeleton. The abnormal development of pharyngeal arch cartilage is related to the augmented hypertrophy and disordered arrangement of chondrocytes, while decreased proliferation, differentiation and mineralization of osteoblasts may be involved in the delayed maturation of skull bones. Furthermore, we revealed that deficiency of fgfr3 leads to enhanced IHH signaling and up-regulated canonical Wnt/β-catenin signaling, and pharmacological inhibition of Wnt/β-catenin could partially alleviate the phenotypes of fgfr3 mutants. Conclusions: Our study further reveals some novel phenotypes and underlying developmental mechanism of CATSHL syndrome, which deepens our understanding of the pathogenesis of CATSHL and the role of fgfr3 in skeleton development. Our findings provide evidence that modulation of Wnt/β-catenin activity could be a potential therapy for CATSHL syndrome and related skeleton diseases.
Collapse
Affiliation(s)
- Xianding Sun
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Ruobin Zhang
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Hangang Chen
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Xiaolan Du
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Shuai Chen
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Junlan Huang
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Mi Liu
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Meng Xu
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Fengtao Luo
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Min Jin
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Nan Su
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Huabing Qi
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Jing Yang
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Qiaoyan Tan
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Dali Zhang
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Zhenhong Ni
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Sen Liang
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Bin Zhang
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Di Chen
- Research Center for Human Tissues and Organs Degeneration, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Xin Zhang
- Departments of Ophthalmology, Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| | - Lingfei Luo
- Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, Laboratory of Molecular Developmental Biology, School of Life Sciences, Southwest University, Beibei, Chongqing 400715, China
| | - Lin Chen
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Yangli Xie
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Army Medical University, Chongqing 400042, China
| |
Collapse
|
55
|
Sobhani N, Fan C, O. Flores-Villanueva P, Generali D, Li Y. The Fibroblast Growth Factor Receptors in Breast Cancer: from Oncogenesis to Better Treatments. Int J Mol Sci 2020; 21:E2011. [PMID: 32188012 PMCID: PMC7139621 DOI: 10.3390/ijms21062011] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 03/11/2020] [Accepted: 03/14/2020] [Indexed: 01/09/2023] Open
Abstract
Breast cancer (BC) is the most frequent form of malignancy and second only to lung cancer as cause of deaths in women. Notwithstanding many progresses made in the field, metastatic BC has a very poor prognosis. As therapies are becoming more personalized to meet the needs of patients, a better knowledge of the molecular biology leading to the disease unfolds the possibility to project more precise compounds or antibodies targeting definite alteration at the molecular level and functioning on such cancer-causing molecules expressed in cancer cells of patients, or present as antigens on the surface of cancer cell membranes. Fibroblast growth factor receptor (FGFR) is one of such druggable targets, activated by its own ligands -namely the Fibroblast Growth Factors (FGFs). This pathway provides a vast range of interesting molecular targets pursued at different levels of clinical investigation. Herein we provide an update on the knowledge of genetic alterations of the receptors in breast cancer, their role in tumorigenesis and the most recent drugs against this particular receptor for the treatment of the disease.
Collapse
Affiliation(s)
- Navid Sobhani
- Section of Epidemiology and Population Science, Department of Medicine, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030, USA; (C.F.); (P.O.F.-V.)
- Department of Medical, Surgical and Health Sciences, University of Trieste, Cattinara Hospital, Strada Di Fiume 447, 34149 Trieste, Italy;
| | - Chunmei Fan
- Section of Epidemiology and Population Science, Department of Medicine, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030, USA; (C.F.); (P.O.F.-V.)
| | - Pedro O. Flores-Villanueva
- Section of Epidemiology and Population Science, Department of Medicine, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030, USA; (C.F.); (P.O.F.-V.)
| | - Daniele Generali
- Department of Medical, Surgical and Health Sciences, University of Trieste, Cattinara Hospital, Strada Di Fiume 447, 34149 Trieste, Italy;
| | - Yong Li
- Section of Epidemiology and Population Science, Department of Medicine, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030, USA; (C.F.); (P.O.F.-V.)
| |
Collapse
|
56
|
Hirotsune S, Kiyonari H, Jin M, Kumamoto K, Yoshida K, Shinohara M, Watanabe H, Wynshaw-Boris A, Matsuzaki F. Enhanced homologous recombination by the modulation of targeting vector ends. Sci Rep 2020; 10:2518. [PMID: 32054870 PMCID: PMC7018964 DOI: 10.1038/s41598-020-58893-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 12/11/2019] [Indexed: 11/17/2022] Open
Abstract
The field of genome editing was founded on the establishment of methods, such as the clustered regularly interspaced short palindromic repeat (CRISPR) and CRISPR-associated protein (CRISPR/Cas) system, used to target DNA double-strand breaks (DSBs). However, the efficiency of genome editing also largely depends on the endogenous cellular repair machinery. Here, we report that the specific modulation of targeting vectors to provide 3' overhangs at both ends increased the efficiency of homology-directed repair (HDR) in embryonic stem cells. We applied the modulated targeting vectors to produce homologous recombinant mice directly by pronuclear injection, but the frequency of HDR was low. Furthermore, we combined our method with the CRISPR/Cas9 system, resulting in a significant increase in HDR frequency. Thus, our HDR-based method, enhanced homologous recombination for genome targeting (eHOT), is a new and powerful method for genome engineering.
Collapse
Affiliation(s)
- Shinji Hirotsune
- Department of Genetic Disease Research, Osaka City University, Graduate School of Medicine, Asahi-machi 1-4-3, Abeno, Osaka, 545-8585, Japan.
| | - Hiroshi Kiyonari
- Animal Resource Development Unit, Genetic Engineering Team, Division of Bio-Function Dynamics Imaging, RIKEN Center for Life Science Technologies, 2-2-3 Minatojima-Minamimachi, Chuou-ku, Kobe, 650-0047, Japan
| | - Mingyue Jin
- Department of Genetic Disease Research, Osaka City University, Graduate School of Medicine, Asahi-machi 1-4-3, Abeno, Osaka, 545-8585, Japan
| | - Kanako Kumamoto
- Department of Genetic Disease Research, Osaka City University, Graduate School of Medicine, Asahi-machi 1-4-3, Abeno, Osaka, 545-8585, Japan
| | - Kayo Yoshida
- Laboratory Animal Science, Osaka City University, Graduate School of Medicine, Asahi-machi 1-4-3, Abeno, Osaka, 545-8585, Japan
| | - Miki Shinohara
- Institute for Protein Research, Osaka University 3-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
- Faculty of Agriculture, Department of Advanced Bioscience, Kindai University, 3327-204 Nakamachi, Nara-city, Nara, 631-8505, Japan
| | - Hitomi Watanabe
- Laboratory of Integrative Biological Science, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, 606-8507, Japan
| | - Anthony Wynshaw-Boris
- Department of Genetics and Genome Sciences, Case Western Reserve University, School of Medicine, University Hospitals Case Medical Center 10900 Euclid Avenue, BRB731, Cleveland, Ohio, 44106-4955, USA
| | - Fumio Matsuzaki
- RIKEN Center for Developmental Biology, 2-2-3 Minatojima-Minamimachi, Chuou-ku, Kobe, 650-0047, Japan
| |
Collapse
|
57
|
Westerfield JM, Barrera FN. Membrane receptor activation mechanisms and transmembrane peptide tools to elucidate them. J Biol Chem 2019; 295:1792-1814. [PMID: 31879273 DOI: 10.1074/jbc.rev119.009457] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Single-pass membrane receptors contain extracellular domains that respond to external stimuli and transmit information to intracellular domains through a single transmembrane (TM) α-helix. Because membrane receptors have various roles in homeostasis, signaling malfunctions of these receptors can cause disease. Despite their importance, there is still much to be understood mechanistically about how single-pass receptors are activated. In general, single-pass receptors respond to extracellular stimuli via alterations in their oligomeric state. The details of this process are still the focus of intense study, and several lines of evidence indicate that the TM domain (TMD) of the receptor plays a central role. We discuss three major mechanistic hypotheses for receptor activation: ligand-induced dimerization, ligand-induced rotation, and receptor clustering. Recent observations suggest that receptors can use a combination of these activation mechanisms and that technical limitations can bias interpretation. Short peptides derived from receptor TMDs, which can be identified by screening or rationally developed on the basis of the structure or sequence of their targets, have provided critical insights into receptor function. Here, we explore recent evidence that, depending on the target receptor, TMD peptides cannot only inhibit but also activate target receptors and can accommodate novel, bifunctional designs. Furthermore, we call for more sharing of negative results to inform the TMD peptide field, which is rapidly transforming into a suite of unique tools with the potential for future therapeutics.
Collapse
Affiliation(s)
- Justin M Westerfield
- Department of Biochemistry & Cellular and Molecular Biology, University of Tennessee, Knoxville, Tennessee 37996
| | - Francisco N Barrera
- Department of Biochemistry & Cellular and Molecular Biology, University of Tennessee, Knoxville, Tennessee 37996.
| |
Collapse
|
58
|
Matsuoka D, Kamiya M, Sato T, Sugita Y. Role of the N-Terminal Transmembrane Helix Contacts in the Activation of FGFR3. J Comput Chem 2019; 41:561-572. [PMID: 31804721 DOI: 10.1002/jcc.26122] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Revised: 10/31/2019] [Accepted: 11/08/2019] [Indexed: 12/16/2022]
Abstract
Fibroblast growth factor receptor 3 (FGFR3) is a member of receptor tyrosine kinases, which is involved in skeletal cell growth, differentiation, and migration. FGFR3 transduces biochemical signals from the extracellular ligand-binding domain to the intracellular kinase domain through the conformational changes of the transmembrane (TM) helix dimer. Here, we apply generalized replica exchange with solute tempering method to wild type (WT) and G380R mutant (G380R) of FGFR3. The dimer interface in G380R is different from WT and the simulation results are in good agreement with the solid-state nuclear magnetic resonance (NMR) spectroscopy. TM helices in G380R are extended more than WT, and thereby, G375 in G380R contacts near the N-termini of the TM helix dimer. Considering that both G380R and G375C show the constitutive activation, the formation of the N-terminal contacts of the TM helices can be generally important for the activation mechanism. © 2019 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Daisuke Matsuoka
- Theoretical Molecular Science Laboratory, RIKEN Cluster for Pioneering Research, Wako-shi, Saitama, 351-0198, Japan
| | - Motoshi Kamiya
- Computational Biophysics Research Team, RIKEN Center for Computational Science, Kobe, 650-0047, Japan
| | - Takeshi Sato
- Division of Liberal Arts and Science, Kyoto Pharmaceutical University, Kyoto, 607-8414, Japan
| | - Yuji Sugita
- Theoretical Molecular Science Laboratory, RIKEN Cluster for Pioneering Research, Wako-shi, Saitama, 351-0198, Japan.,Computational Biophysics Research Team, RIKEN Center for Computational Science, Kobe, 650-0047, Japan.,Laboratory for Biomolecular Function Simulation, RIKEN Center for Biosystem Dynamics Research, Kobe, 650-0047, Japan
| |
Collapse
|
59
|
Growth Plate Chondrocytes: Skeletal Development, Growth and Beyond. Int J Mol Sci 2019; 20:ijms20236009. [PMID: 31795305 PMCID: PMC6929081 DOI: 10.3390/ijms20236009] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 11/22/2019] [Accepted: 11/27/2019] [Indexed: 02/01/2023] Open
Abstract
Growth plate chondrocytes play central roles in the proper development and growth of endochondral bones. Particularly, a population of chondrocytes in the resting zone expressing parathyroid hormone-related protein (PTHrP) is now recognized as skeletal stem cells, defined by their ability to undergo self-renewal and clonally give rise to columnar chondrocytes in the postnatal growth plate. These chondrocytes also possess the ability to differentiate into a multitude of cell types including osteoblasts and bone marrow stromal cells during skeletal development. Using single-cell transcriptomic approaches and in vivo lineage tracing technology, it is now possible to further elucidate their molecular properties and cellular fate changes. By discovering the fundamental molecular characteristics of these cells, it may be possible to harness their functional characteristics for skeletal growth and regeneration. Here, we discuss our current understanding of the molecular signatures defining growth plate chondrocytes.
Collapse
|
60
|
Recent Insights into Long Bone Development: Central Role of Hedgehog Signaling Pathway in Regulating Growth Plate. Int J Mol Sci 2019; 20:ijms20235840. [PMID: 31757091 PMCID: PMC6928971 DOI: 10.3390/ijms20235840] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Revised: 11/17/2019] [Accepted: 11/18/2019] [Indexed: 12/30/2022] Open
Abstract
The longitudinal growth of long bone, regulated by an epiphyseal cartilaginous component known as the “growth plate”, is generated by epiphyseal chondrocytes. The growth plate provides a continuous supply of chondrocytes for endochondral ossification, a sequential bone replacement of cartilaginous tissue, and any failure in this process causes a wide range of skeletal disorders. Therefore, the cellular and molecular characteristics of the growth plate are of interest to many researchers. Hedgehog (Hh), well known as a mitogen and morphogen during development, is one of the best known regulatory signals in the developmental regulation of the growth plate. Numerous animal studies have revealed that signaling through the Hh pathway plays multiple roles in regulating the proliferation, differentiation, and maintenance of growth plate chondrocytes throughout the skeletal growth period. Furthermore, over the past few years, a growing body of evidence has emerged demonstrating that a limited number of growth plate chondrocytes transdifferentiate directly into the full osteogenic and multiple mesenchymal lineages during postnatal bone development and reside in the bone marrow until late adulthood. Current studies with the genetic fate mapping approach have shown that the commitment of growth plate chondrocytes into the skeletal lineage occurs under the influence of epiphyseal chondrocyte-derived Hh signals during endochondral bone formation. Here, we discuss the valuable observations on the role of the Hh signaling pathway in the growth plate based on mouse genetic studies, with some emphasis on recent advances.
Collapse
|
61
|
Delucchi Á, Toro L, Alzamora R, Barrientos V, González M, Andaur R, León P, Villanueva F, Galindo M, Las Heras F, Montecino M, Moena D, Lazcano A, Pinto V, Salas P, Reyes ML, Mericq V, Michea L. Glucocorticoids Decrease Longitudinal Bone Growth in Pediatric Kidney Transplant Recipients by Stimulating the FGF23/FGFR3 Signaling Pathway. J Bone Miner Res 2019; 34:1851-1861. [PMID: 31099911 DOI: 10.1002/jbmr.3761] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Revised: 05/05/2019] [Accepted: 05/11/2019] [Indexed: 12/11/2022]
Abstract
Renal transplantation (RTx) is an effective therapy to improve clinical outcomes in pediatric patients with terminal chronic kidney disease. However, chronic immunosuppression with glucocorticoids (GCs) reduces bone growth and BMD. The mechanisms causing GC-induced growth impairment have not been fully clarified. Fibroblast growth factor 23 (FGF23) is a peptide hormone that regulates phosphate homeostasis and bone growth. In pathological conditions, FGF23 excess or abnormal FGF receptors (FGFR) activity leads to bone growth impairment. Experimental data indicate that FGF23 expression is induced by chronic GC exposure. Therefore, we hypothesize that GCs impair bone growth by increasing FGF23 expression, which has direct effects on bone growth plate. In a post hoc analysis of a multicentric randomized clinical trial of prepubertal RTx children treated with early GC withdrawal or chronic GC treatment, we observed that GC withdrawal was associated with improvement in longitudinal growth and BMD, and lower plasma FGF23 levels as compared with a chronic GC group. In prepubertal rats, GC-induced bone growth retardation correlated with increased plasma FGF23 and bone FGF23 expression. Additionally, GC treatment decreased FGFR1 expression whereas it increased FGFR3 expression in mouse tibia explants. The GC-induced bone growth impairment in tibiae explants was prevented by blockade of FGF23 receptors using either a pan-FGFR antagonist (PD173074), a C-terminal FGF23 peptide (FGF23180-205) which blocks the binding of FGF23 to the FGFR-Klotho complex or a specific FGFR3 antagonist (P3). Finally, local administration of PD173074 into the tibia growth plate ameliorated cartilage growth impairment in GC-treated rats. These results show that GC treatment partially reduces longitudinal bone growth via upregulation of FGF23 and FGFR3 expression, thus suggesting that the FGF23/Klotho/FGFR3 axis at the growth plate could be a potential therapeutic target for the management of GC-induced growth impairment in children.
Collapse
Affiliation(s)
- Ángela Delucchi
- Division of Nephrology, Hospital Luis Calvo Mackenna, Santiago, Chile.,Division of Nephrology, Clínica Alemana de Santiago, Santiago, Chile
| | - Luis Toro
- Division of Nephrology, Department of Medicine, Hospital Clínico Universidad de Chile, Santiago, Chile.,Centro de Investigación Clínica Avanzada, Hospital Clínico Universidad de Chile, Santiago, Chile.,Clinica Las Condes, Santiago, Chile
| | - Rodrigo Alzamora
- Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile
| | - Victor Barrientos
- Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Magdalena González
- Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Rodrigo Andaur
- Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Pablo León
- Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Francisco Villanueva
- Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Mario Galindo
- Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy (MIII), Santiago, Chile
| | - Facundo Las Heras
- Clinica Las Condes, Santiago, Chile.,Department of Anatomic Pathology, Hospital Clínico Universidad de Chile, Santiago, Chile
| | - Martín Montecino
- Institute of Biomedical Sciences, Faculty of Medicine and Faculty of Life Sciences, Universidad Andrés Bello, Santiago, Chile.,FONDAP Center for Genome Regulation, Universidad Andres Bello, Santiago, Chile
| | - Daniel Moena
- Institute of Biomedical Sciences, Faculty of Medicine and Faculty of Life Sciences, Universidad Andrés Bello, Santiago, Chile.,FONDAP Center for Genome Regulation, Universidad Andres Bello, Santiago, Chile
| | - Andrea Lazcano
- Division of Nephrology, Clínica Alemana de Santiago, Santiago, Chile.,Division of Nephrology, Hospital de Niños Roberto del Río, Santiago, Chile
| | - Viola Pinto
- Clinica Las Condes, Santiago, Chile.,Pediatric Nephrology Unit, Hospital Doctor Exequiel González Cortés, Santiago, Chile
| | - Paulina Salas
- Pediatric Nephrology Unit, Hospital Doctor Exequiel González Cortés, Santiago, Chile
| | - María Loreto Reyes
- Pediatric Endocrinology Unit, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Verónica Mericq
- Clinica Las Condes, Santiago, Chile.,Institute of Maternal and Child Research, Universidad de Chile, Santiago, Chile
| | - Luis Michea
- Division of Nephrology, Department of Medicine, Hospital Clínico Universidad de Chile, Santiago, Chile.,Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy (MIII), Santiago, Chile
| |
Collapse
|
62
|
Kamatkar N, Levy M, Hébert JM. Development of a Monomeric Inhibitory RNA Aptamer Specific for FGFR3 that Acts as an Activator When Dimerized. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 17:530-539. [PMID: 31357131 PMCID: PMC6661505 DOI: 10.1016/j.omtn.2019.06.020] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 06/12/2019] [Accepted: 06/22/2019] [Indexed: 12/11/2022]
Abstract
There have been limited options for people who suffer from fibroblast growth factor receptor (FGFR) signaling disorders. In this study, we developed RNA aptamers specific for FGFR3 as potential therapeutic agents. Using a structured aptamer library, we performed ten rounds of SELEX (systematic evolution of ligands by exponential enrichment) against mouse FGFR3c protein. Using an engineered BaF3 cell line, one aptamer clone from round 6 of the selection inhibited FGF-dependent cell growth with a concentration at which 50% of growth is observed (IC50) of ∼260 nM and bound both mouse and human FGFR3 but not FGFR1 or FGFR2. This inhibitor of FGFR3 signaling (iR3), when dimerized using a template-driven approach, resulted in a functional activator of FGFR3 (aR3). We validated the activity and specificity of iR3 and aR3 on engineered BaF3 cell lines, mouse and human FGFR protein, and primary cultures of neuroepithelial precursor cells.
Collapse
Affiliation(s)
- Nachiket Kamatkar
- Departments of Neuroscience and Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | - Matthew Levy
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | - Jean M Hébert
- Departments of Neuroscience and Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| |
Collapse
|
63
|
FGF2-induced STAT3 activation regulates pathologic neovascularization. Exp Eye Res 2019; 187:107775. [PMID: 31449793 DOI: 10.1016/j.exer.2019.107775] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 07/29/2019] [Accepted: 08/22/2019] [Indexed: 12/23/2022]
Abstract
Cell-autonomous endothelial cell (EC) fibroblast growth factor receptor (FGFR) signaling through FGFR1/2 is essential for injury-induced wound vascularization and pathologic neovascularization as in blinding eye diseases such as age-related macular degeneration. Which FGF ligand(s) is critical in regulating angiogenesis is unknown. Utilizing ex vivo models of choroidal endothelial sprouting and in vivo models of choroidal neovascularization (CNV), we demonstrate here that only FGF2 is the essential ligand. Though FGF-FGFR signaling can activate multiple intracellular signaling pathways, we show that FGF2 regulates pathogenic angiogenesis via STAT3 activation. The identification of FGF2 as a critical mediator in aberrant neovascularization provides a new opportunity for developing multi-target therapies in blinding eye diseases especially given the limitations of anti-VEGF monotherapy.
Collapse
|
64
|
Leerberg DM, Hopton RE, Draper BW. Fibroblast Growth Factor Receptors Function Redundantly During Zebrafish Embryonic Development. Genetics 2019; 212:1301-1319. [PMID: 31175226 PMCID: PMC6707458 DOI: 10.1534/genetics.119.302345] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 05/29/2019] [Indexed: 01/08/2023] Open
Abstract
Fibroblast growth factor (Fgf) signaling regulates many processes during development. In most cases, one tissue layer secretes an Fgf ligand that binds and activates an Fgf receptor (Fgfr) expressed by a neighboring tissue. Although studies have identified the roles of specific Fgf ligands during development, less is known about the requirements for the receptors. We have generated null mutations in each of the five fgfr genes in zebrafish. Considering the diverse requirements for Fgf signaling throughout development, and that null mutations in the mouse Fgfr1 and Fgfr2 genes are embryonic lethal, it was surprising that all zebrafish homozygous mutants are viable and fertile, with no discernable embryonic defect. Instead, we find that multiple receptors are involved in coordinating most Fgf-dependent developmental processes. For example, mutations in the ligand fgf8a cause loss of the midbrain-hindbrain boundary, whereas, in the fgfr mutants, this phenotype is seen only in embryos that are triple mutant for fgfr1a;fgfr1b;fgfr2, but not in any single or double mutant combinations. We show that this apparent fgfr redundancy is also seen during the development of several other tissues, including posterior mesoderm, pectoral fins, viscerocranium, and neurocranium. These data are an essential step toward defining the specific Fgfrs that function with particular Fgf ligands to regulate important developmental processes in zebrafish.
Collapse
Affiliation(s)
- Dena M Leerberg
- Department of Molecular and Cellular Biology, University of California, Davis, California 95616
| | - Rachel E Hopton
- Department of Molecular and Cellular Biology, University of California, Davis, California 95616
| | - Bruce W Draper
- Department of Molecular and Cellular Biology, University of California, Davis, California 95616
| |
Collapse
|
65
|
Yang C, Zhang R, Lin H, Wang H. Insights into the molecular regulatory network of pathomechanisms in osteochondroma. J Cell Biochem 2019; 120:16362-16369. [PMID: 31211456 DOI: 10.1002/jcb.29155] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 05/17/2019] [Accepted: 05/22/2019] [Indexed: 12/21/2022]
Abstract
Osteochondroma is a benign autosomal dominant hereditary disease characterized by abnormal proliferation of cartilage in the long bone. It is divided into solitary osteochondroma and hereditary multiple exostoses (HMEs). The exostosin-1 (EXT-1) and exostosin-2 (EXT-2) gene mutations are well-defined molecular mechanisms in the pathogenesis of HME. EXT-1 and EXT-2 encode glycosyltransferases that are necessary for the synthesis of heparin sulfate. Accumulating evidence suggests that mutations in the EXT family induce changes in isolated hypogonadotropic hypogonadism-parathyroid hormone-related protein, bone morphogenetic protein, and fibroblast growth factor signaling pathways. Studies have also found that a large number of microRNAs (miRNAs) are abnormally expressed in osteochondroma tissues, and some of them also participate in several major signaling pathways. The regulation of miRNA expression could be another breakthrough in the treatment of osteochondroma. Although the pathogenesis of osteochondroma is very complicated, significant progress has been made in recent years. It is hoped that the pathogenesis of osteochondroma will be clearly understood and the most effective methods for the prevention and treatment of osteochondroma will be determined. This review provides an update on the recent progress in the interpretation of the underlying molecular mechanisms of osteochondroma.
Collapse
Affiliation(s)
- Congyi Yang
- School of Basic Medical Sciences, Nanchang University, Nanchang, Jiangxi, China.,Queen Mary School, Nanchang University, Nanchang, Jiangxi, China
| | - Ruiqian Zhang
- School of Basic Medical Sciences, Nanchang University, Nanchang, Jiangxi, China.,Queen Mary School, Nanchang University, Nanchang, Jiangxi, China
| | - Hui Lin
- School of Basic Medical Sciences, Nanchang University, Nanchang, Jiangxi, China
| | - Hongmei Wang
- School of Basic Medical Sciences, Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
66
|
Tuzon CT, Rigueur D, Merrill AE. Nuclear Fibroblast Growth Factor Receptor Signaling in Skeletal Development and Disease. Curr Osteoporos Rep 2019; 17:138-146. [PMID: 30982184 PMCID: PMC8221190 DOI: 10.1007/s11914-019-00512-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW Fibroblast growth factor receptor (FGFR) signaling regulates proliferation and differentiation during development and homeostasis. While membrane-bound FGFRs play a central role in these processes, the function of nuclear FGFRs is also critical. Here, we highlight mechanisms for nuclear FGFR translocation and the effects of nuclear FGFRs on skeletal development and disease. RECENT FINDINGS Full-length FGFRs, internalized by endocytosis, enter the nucleus through β-importin-dependent mechanisms that recognize the nuclear localization signal within FGFs. Alternatively, soluble FGFR intracellular fragments undergo nuclear translocation following their proteolytic release from the membrane. FGFRs enter the nucleus during the cellular transition between proliferation and differentiation. Once nuclear, FGFRs interact with chromatin remodelers to alter the epigenetic state and transcription of their target genes. Dysregulation of nuclear FGFR is linked to the etiology of congenital skeletal disorders and neoplastic transformation. Revealing the activities of nuclear FGFR will advance our understanding of 20 congenital skeletal disorders caused by FGFR mutations, as well as FGFR-related cancers.
Collapse
Affiliation(s)
- Creighton T Tuzon
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, 90033, USA
| | - Diana Rigueur
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, 90033, USA
| | - Amy E Merrill
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, 90033, USA.
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA.
| |
Collapse
|
67
|
Sims D, Onambélé-Pearson G, Burden A, Payton C, Morse C. Whole-body and segmental analysis of body composition in adult males with achondroplasia using dual X-ray absorptiometry. PLoS One 2019; 14:e0213806. [PMID: 30889196 PMCID: PMC6424418 DOI: 10.1371/journal.pone.0213806] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 02/15/2019] [Indexed: 12/05/2022] Open
Abstract
Achondroplasia is a condition characterized by a genetic mutation affecting long bone endplate development. Current data suggests that the bone mineral content (BMC) and bone mineral density (BMD) of achondroplasic populations are below age matched individuals of average stature (controls). Due to the disproportionate limb-to-torso length compared to controls however, the lower BMC and BMD may be nullified when appropriately presented. The aim of this study was to measure whole-body and segmental body composition in adult males with achondroplasia (N = 10, 22 ±3 yrs), present data relative to whole-body and whole-limb values and compare all values to age matched controls (N = 17, 22 ±2 yrs). Dual X-ray absorptiometry (DEXA) was used to measure the in vivo mass of the whole-body and 15 segments, from which BMD, BMC, fat free mass (FFM) and body fat mass were measured. BMC of lumbar vertebrae (L1-4) was also measured and presented as a volumetric BMD (BMDVOL). The achondroplasic group had less BMC, BMD and FFM, and more body fat mass than controls as a whole-body measure. The lower achondroplasic BMC and BMD was somewhat nullified when presented relative to whole-body and whole-limb values respectively. There was no difference in lumbar BMDVOL between groups. Whole-body BMD measures presented the achondroplasic group as 'osteopenic'. When relative to whole-limb measures however, achondroplasic BMD descriptions were normal. Further work is needed to create a body composition database for achondroplasic population's, or for clinicians to present achondroplasic body composition values relative to the whole-limb.
Collapse
Affiliation(s)
- David Sims
- Health, Exercise and Active Living Research, Manchester Metropolitan University, Manchester, England
| | - Gladys Onambélé-Pearson
- Health, Exercise and Active Living Research, Manchester Metropolitan University, Manchester, England
| | - Adrian Burden
- Health, Exercise and Active Living Research, Manchester Metropolitan University, Manchester, England
| | - Carl Payton
- Health, Exercise and Active Living Research, Manchester Metropolitan University, Manchester, England
| | - Christopher Morse
- Health, Exercise and Active Living Research, Manchester Metropolitan University, Manchester, England
| |
Collapse
|
68
|
Tu Y, Qu T, Chen F. Mutant hFGF23(A12D) stimulates osteoblast differentiation through FGFR3. J Cell Mol Med 2019; 23:2933-2942. [PMID: 30761743 PMCID: PMC6433671 DOI: 10.1111/jcmm.14201] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 01/13/2019] [Accepted: 01/14/2019] [Indexed: 01/01/2023] Open
Abstract
Fibroblast growth factor (FGF) 23 is a member of the FGF family involved in bone development by interacting with FGFRs. In a previous study, we discovered a mutant human FGF (hFGF) 23 (A12D) in the mandibular prognathism (MP) pedigree. However, the exact role of hFGF23(A12D) during bone formation remains unclear. The aim of this study was to identify the function of hFGF23(A12D) in bone formation. We infected isolated rat calvaria (RC) cells with the recombinant lentivirus containing mutant hFGF23(A12D) and WT hFGF23 respectively. Real‐time PCR, western blot and enzyme‐linked immunosorbent assay confirmed that hFGF23(A12D) failed to be secreted. We measured cell growth via the CCK‐8 assay based on Zsgreen expression, detected cell differentiation ability via alkaline phosphatase staining, performed RT‐PCR and found that hFGF23(A12D) inhibited proliferation of RC cells and stimulated the differentiation of RC cells to osteoblasts. Through RNA sequencing, RT‐PCR and western blot, we found increased expression of FGFR3. Through co‐immunoprecipitation assays and immunofluorescence staining, we revealed that hFGF23(A12D) activated the mitogen‐activated protein kinase signalling pathway through interactions with the intracellular domain of FGFR3. In summary, we determined the mechanisms of hFGF23(A12D) involved in osteoblast generation and formation which is specifically due to its interaction with FGFR3.
Collapse
Affiliation(s)
- Yilin Tu
- Laboratory of Oral Biomedical Science and Translational Medicine, School and Hospital of Stomatology, Tongji University, Shanghai, China
| | - Taoran Qu
- Laboratory of Oral Biomedical Science and Translational Medicine, School and Hospital of Stomatology, Tongji University, Shanghai, China
| | - Fengshan Chen
- Laboratory of Oral Biomedical Science and Translational Medicine, School and Hospital of Stomatology, Tongji University, Shanghai, China
| |
Collapse
|
69
|
Chen B, Tan Q, Chen H, Luo F, Xu M, Zhao J, Liu P, Sun X, Su N, Zhang D, Fan W, Liu M, Huang H, Wang Z, Huang J, Zhang R, Li C, Li F, Ni Z, Du X, Jin M, Yang J, Xie Y, Chen L. Imbalanced development of anterior and posterior thorax is a causative factor triggering scoliosis. J Orthop Translat 2019; 17:103-111. [PMID: 31194037 PMCID: PMC6551366 DOI: 10.1016/j.jot.2018.12.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Revised: 12/05/2018] [Accepted: 12/11/2018] [Indexed: 11/19/2022] Open
Abstract
Objective Scoliosis is a common disease characterized by spinal curvature with variable severities. There is no generally accepted theory about the physical origin of the spinal deformation of scoliosis. The aim of this study was to explore a new hypothesis suggesting that the curvatures in scoliosis may be associated with the imbalance growth between thoracic vertebral column and sternum. Methods We undertook a comparative computed tomography (CT) based morphology study of thoracic vertebrae and sternum of patients with adolescent idiopathic scoliosis (AIS) and age-gender matched normal subjects. We further measured the ratios between the lengths of the sternum and thoracic vertebra of mice with deficiency of fibroblast growth factor receptor 3 (FGFR3), which exhibit scoliosis. Three-week-old C57BL/6J mice were used to generate bipedal and sternal growth plate injury model. Radiographs and histological images were obtained to observe the presence of sternal and spinal deformity. Results There was a significant correlation between the severities of scoliosis and the ratios of the sternum to thoracic vertebral lengths. We also found that FGFR3 deficient mice showed smaller ratio of the sternum to thoracic vertebra lengths than that of the wild-type mice, which were similar with that of the AIS patients. Surgery-induced injuries of sternal growth plates can accelerate and aggravate the scoliosis in bipedal mice and imbalanced development of anterior and posterior thoracic occurred before the appearance of scoliosis. Conclusions Our findings suggest that the imbalanced growth between the thoracic vertebral column and the sternum is an important causative factor for the pathogenesis of scoliosis including AIS. The translational potential of this article Imbalanced growth between the thoracic vertebral column and the sternum is associated with scoliosis. Surgical or rehabilitation intervention for scoliosis should focus on all components involved in the pathogenesis of curvature to obtain better outcome.
Collapse
Affiliation(s)
- Bo Chen
- Laboratory for the Rehabilitation of Traumatic Injuries, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, 400042, China
- Department of Spine Surgery, Institute of Surgery Research, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Qiaoyan Tan
- Laboratory for the Rehabilitation of Traumatic Injuries, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Hangang Chen
- Laboratory for the Rehabilitation of Traumatic Injuries, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Fengtao Luo
- Laboratory for the Rehabilitation of Traumatic Injuries, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Meng Xu
- Laboratory for the Rehabilitation of Traumatic Injuries, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Jianhua Zhao
- Department of Spine Surgery, Institute of Surgery Research, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Peng Liu
- Department of Spine Surgery, Institute of Surgery Research, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Xianding Sun
- Laboratory for the Rehabilitation of Traumatic Injuries, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Nan Su
- Laboratory for the Rehabilitation of Traumatic Injuries, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Dali Zhang
- Laboratory for the Rehabilitation of Traumatic Injuries, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Weili Fan
- Department of Spine Surgery, Institute of Surgery Research, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Mingyong Liu
- Department of Spine Surgery, Institute of Surgery Research, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Haiyang Huang
- Department of Spine Surgery, Institute of Surgery Research, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Zuqiang Wang
- Laboratory for the Rehabilitation of Traumatic Injuries, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Junlan Huang
- Laboratory for the Rehabilitation of Traumatic Injuries, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Ruobin Zhang
- Laboratory for the Rehabilitation of Traumatic Injuries, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Can Li
- Laboratory for the Rehabilitation of Traumatic Injuries, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Fangfang Li
- Laboratory for the Rehabilitation of Traumatic Injuries, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Zhenhong Ni
- Laboratory for the Rehabilitation of Traumatic Injuries, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Xiaolan Du
- Laboratory for the Rehabilitation of Traumatic Injuries, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Min Jin
- Laboratory for the Rehabilitation of Traumatic Injuries, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Jing Yang
- Laboratory for the Rehabilitation of Traumatic Injuries, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Yangli Xie
- Laboratory for the Rehabilitation of Traumatic Injuries, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, 400042, China
- Corresponding author. Laboratory for the Rehabilitation of Traumatic Injuries, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, 400042, China.
| | - Lin Chen
- Laboratory for the Rehabilitation of Traumatic Injuries, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, 400042, China
- Corresponding author. Laboratory for the Rehabilitation of Traumatic Injuries, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, 400042, China.
| |
Collapse
|
70
|
Abstract
Fibroblast growth factors (FGFs) and their receptors (FGFRs) are expressed throughout all stages of skeletal development. In the limb bud and in cranial mesenchyme, FGF signaling is important for formation of mesenchymal condensations that give rise to bone. Once skeletal elements are initiated and patterned, FGFs regulate both endochondral and intramembranous ossification programs. In this chapter, we review functions of the FGF signaling pathway during these critical stages of skeletogenesis, and explore skeletal malformations in humans that are caused by mutations in FGF signaling molecules.
Collapse
Affiliation(s)
- David M Ornitz
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, United States.
| | - Pierre J Marie
- UMR-1132 Inserm (Institut national de la Santé et de la Recherche Médicale) and University Paris Diderot, Sorbonne Paris Cité, Hôpital Lariboisière, Paris, France
| |
Collapse
|
71
|
Marques P, Korbonits M. Pseudoacromegaly. Front Neuroendocrinol 2019; 52:113-143. [PMID: 30448536 DOI: 10.1016/j.yfrne.2018.11.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 10/30/2018] [Accepted: 11/14/2018] [Indexed: 01/19/2023]
Abstract
Individuals with acromegaloid physical appearance or tall stature may be referred to endocrinologists to exclude growth hormone (GH) excess. While some of these subjects could be healthy individuals with normal variants of growth or physical traits, others will have acromegaly or pituitary gigantism, which are, in general, straightforward diagnoses upon assessment of the GH/IGF-1 axis. However, some patients with physical features resembling acromegaly - usually affecting the face and extremities -, or gigantism - accelerated growth/tall stature - will have no abnormalities in the GH axis. This scenario is termed pseudoacromegaly, and its correct diagnosis can be challenging due to the rarity and variability of these conditions, as well as due to significant overlap in their characteristics. In this review we aim to provide a comprehensive overview of pseudoacromegaly conditions, highlighting their similarities and differences with acromegaly and pituitary gigantism, to aid physicians with the diagnosis of patients with pseudoacromegaly.
Collapse
Affiliation(s)
- Pedro Marques
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Márta Korbonits
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK.
| |
Collapse
|
72
|
Miao K, Zhang X, Su SM, Zeng J, Huang Z, Chan UI, Xu X, Deng CX. Optimizing CRISPR/Cas9 technology for precise correction of the Fgfr3-G374R mutation in achondroplasia in mice. J Biol Chem 2018; 294:1142-1151. [PMID: 30487289 DOI: 10.1074/jbc.ra118.006496] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 11/26/2018] [Indexed: 12/13/2022] Open
Abstract
CRISPR/Cas9 is a powerful technology widely used for genome editing, with the potential to be used for correcting a wide variety of deleterious disease-causing mutations. However, the technique tends to generate more indels (insertions and deletions) than precise modifications at the target sites, which might not resolve the mutation and could instead exacerbate the initial genetic disruption. We sought to develop an improved protocol for CRISPR/Cas9 that would correct mutations without unintended consequences. As a case study, we focused on achondroplasia, a common genetic form of dwarfism defined by missense mutation in the Fgfr3 gene that results in glycine to arginine substitution at position 374 in mice in fibroblast growth factor receptor 3 (Fgfr3-G374R), which corresponds to G380R in humans. First, we designed a GFP reporter system that can evaluate the cutting efficiency and specificity of single guide RNAs (sgRNAs). Using the sgRNA selected based on our GFP reporter system, we conducted targeted therapy of achondroplasia in mice. We found that we achieved higher frequency of precise correction of the Fgfr3-G374R mutation using Cas9 protein rather than Cas9 mRNA. We further demonstrated that targeting oligos of 100 and 200 nucleotides precisely corrected the mutation at equal efficiency. We showed that our strategy completely suppressed phenotypes of achondroplasia and whole genome sequencing detected no off-target effects. These data indicate that improved protocols can enable the precise CRISPR/Cas9-mediated correction of individual mutations with high fidelity.
Collapse
Affiliation(s)
- Kai Miao
- Cancer Center, Faculty of Health Sciences, Macau SAR; Centre for Precision Medicine Research and Training, Faculty of Health Sciences, University of Macau, Macau SAR
| | - Xin Zhang
- Cancer Center, Faculty of Health Sciences, Macau SAR; Transgenic and Knockout Core, Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Sek Man Su
- Cancer Center, Faculty of Health Sciences, Macau SAR; Centre for Precision Medicine Research and Training, Faculty of Health Sciences, University of Macau, Macau SAR
| | - Jianming Zeng
- Cancer Center, Faculty of Health Sciences, Macau SAR; Centre for Precision Medicine Research and Training, Faculty of Health Sciences, University of Macau, Macau SAR
| | - Zebin Huang
- Cancer Center, Faculty of Health Sciences, Macau SAR; Centre for Precision Medicine Research and Training, Faculty of Health Sciences, University of Macau, Macau SAR
| | - Un In Chan
- Cancer Center, Faculty of Health Sciences, Macau SAR; Transgenic and Knockout Core, Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Xiaoling Xu
- Cancer Center, Faculty of Health Sciences, Macau SAR; Centre for Precision Medicine Research and Training, Faculty of Health Sciences, University of Macau, Macau SAR; Transgenic and Knockout Core, Faculty of Health Sciences, University of Macau, Macau SAR, China.
| | - Chu-Xia Deng
- Cancer Center, Faculty of Health Sciences, Macau SAR; Centre for Precision Medicine Research and Training, Faculty of Health Sciences, University of Macau, Macau SAR.
| |
Collapse
|
73
|
Krishnan Y, Grodzinsky AJ. Cartilage diseases. Matrix Biol 2018; 71-72:51-69. [PMID: 29803938 PMCID: PMC6146013 DOI: 10.1016/j.matbio.2018.05.005] [Citation(s) in RCA: 267] [Impact Index Per Article: 38.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Revised: 05/14/2018] [Accepted: 05/15/2018] [Indexed: 01/13/2023]
Abstract
Hyaline cartilages, fibrocartilages and elastic cartilages play multiple roles in the human body including bearing loads in articular joints and intervertebral discs, providing joint lubrication, forming the external ears and nose, supporting the trachea, and forming the long bones during development and growth. The structure and organization of cartilage's extracellular matrix (ECM) are the primary determinants of normal function. Most diseases involving cartilage lead to dramatic changes in the ECM which can govern disease progression (e.g., in osteoarthritis), cause the main symptoms of the disease (e.g., dwarfism caused by genetically inherited mutations) or occur as collateral damage in pathological processes occurring in other nearby tissues (e.g., osteochondritis dissecans and inflammatory arthropathies). Challenges associated with cartilage diseases include poor understanding of the etiology and pathogenesis, delayed diagnoses due to the aneural nature of the tissue and drug delivery challenges due to the avascular nature of adult cartilages. This narrative review provides an overview of the clinical and pathological features as well as current treatment options available for various cartilage diseases. Late breaking advances are also described in the quest for development and delivery of effective disease modifying drugs for cartilage diseases including osteoarthritis, the most common form of arthritis that affects hundreds of millions of people worldwide.
Collapse
Affiliation(s)
- Yamini Krishnan
- Department of Chemical Engineering, MIT, Cambridge, MA 02139, USA
| | - Alan J Grodzinsky
- Department of Biological Engineering, MIT, Cambridge, MA 02139, USA; Department of Mechanical Engineering, MIT, Cambridge, MA 02139, USA; Department of Electrical Engineering and Computer Science, MIT, Cambridge, MA 02139, USA.
| |
Collapse
|
74
|
Chen PY, Simons M. Fibroblast growth factor-transforming growth factor beta dialogues, endothelial cell to mesenchymal transition, and atherosclerosis. Curr Opin Lipidol 2018; 29:397-403. [PMID: 30080704 PMCID: PMC6290915 DOI: 10.1097/mol.0000000000000542] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW Despite much effort, atherosclerosis remains an important public health problem, leading to substantial morbidity and mortality worldwide. The purpose of this review is to provide an understanding of the role of endothelial cell fate change in atherosclerosis process. RECENT FINDINGS Recent studies indicate that a process known as endothelial-to-mesenchymal transition (EndMT) may play an important role in atherosclerosis development. Transforming growth factor beta (TGFβ) has been shown to be an important driver of the endothelial cell phenotype transition. SUMMARY The current review deals with the current state of knowledge regarding EndMT's role in atherosclerosis and its regulation by fibroblast growth factor (FGF)-TGFβ cross-talk. A better understanding of FGF-TGFβ signaling in the regulation of endothelial cell phenotypes is key to the development of novel therapeutic agents.
Collapse
Affiliation(s)
- Pei-Yu Chen
- Yale Cardiovascular Research Center, Department of Internal Medicine
| | - Michael Simons
- Yale Cardiovascular Research Center, Department of Internal Medicine
- Department of Cell Biology, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
75
|
Abstract
Chromosomal rearrangements, including translocations, are early and essential events in the formation of many tumors. Previous studies that defined the genetic requirements for rearrangement formation have identified differences between murine and human cells, most notably in the role of classic and alternative nonhomologous end-joining (NHEJ) factors. We reported that poly(ADP)ribose polymerase 3 (PARP3) promotes chromosomal rearrangements induced by endonucleases in multiple human cell types. We show here that in contrast to classic (c-NHEJ) factors, Parp3 also promotes rearrangements in murine cells, including translocations in murine embryonic stem cells (mESCs), class-switch recombination in primary B cells, and inversions in tail fibroblasts that generate Eml4-Alk fusions. In mESCs, Parp3-deficient cells had shorter deletion lengths at translocation junctions. This was corroborated using next-generation sequencing of Eml4-Alk junctions in tail fibroblasts and is consistent with a role for Parp3 in promoting the processing of DNA double-strand breaks. We confirmed a previous report that Parp1 also promotes rearrangement formation. In contrast with Parp3, rearrangement junctions in the absence of Parp1 had longer deletion lengths, suggesting that Parp1 may suppress double-strand break processing. Together, these data indicate that Parp3 and Parp1 promote rearrangements with distinct phenotypes.
Collapse
|
76
|
Balek L, Buchtova M, Kunova Bosakova M, Varecha M, Foldynova-Trantirkova S, Gudernova I, Vesela I, Havlik J, Neburkova J, Turner S, Krzyscik MA, Zakrzewska M, Klimaschewski L, Claus P, Trantirek L, Cigler P, Krejci P. Nanodiamonds as “artificial proteins”: Regulation of a cell signalling system using low nanomolar solutions of inorganic nanocrystals. Biomaterials 2018; 176:106-121. [DOI: 10.1016/j.biomaterials.2018.05.030] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 03/31/2018] [Accepted: 05/19/2018] [Indexed: 12/14/2022]
|
77
|
Saucedo L, Sobarzo C, Brukman NG, Guidobaldi HA, Lustig L, Giojalas LC, Buffone MG, Vazquez-Levin MH, Marín-Briggiler C. Involvement of fibroblast growth factor 2 (FGF2) and its receptors in the regulation of mouse sperm physiology. Reproduction 2018; 156:163-172. [DOI: 10.1530/rep-18-0133] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 06/01/2018] [Indexed: 01/22/2023]
Abstract
Fibroblast growth factor 2 (FGF2) and its receptors (FGFRs) have been described in several tissues, where they regulate cellular proliferation, differentiation, motility and apoptosis. Although FGF2/FGFRs expression in the male reproductive tract has been reported, there is scarce evidence on their presence in the female reproductive tract and their involvement in the modulation of sperm function. Therefore, the objective of this study was to determine the expression of FGF2 in the female reproductive tract and to assess the role of the FGF2/FGFRs system in the regulation of sperm physiology using the murine model. FGF2 was detected in uterus and oviduct protein extracts, and it was immunolocalized in epithelial cells of the uterus,isthmusandampulla, as well as in thecumulus oophorus-oocyte complex. The receptors FGFR1, FGFR2, FGFR3 and FGFR4 were immunodetected in the flagellum and acrosomal region of sperm recovered from thecaudaepididymis. Analysis of testis sections showed the expression of FGFRs in germ cells at different stages of the spermatogenesis, suggesting the testicular origin of the sperm FGFRs. Sperm incubation with recombinant FGF2 (rFGF2) led to increased sperm motility and velocity and to enhanced intracellular Ca2+levels and acrosomal loss compared to the control. In conclusion, this study shows that FGF2 is expressed in tissues of the female reproductive tract. Also, the fact that functional FGFRs are present in mouse sperm and that rFGF2 affects sperm motility and acrosomal exocytosis, suggests the involvement of this system in thein vivoregulation of sperm function.
Collapse
|
78
|
Current Status of Fibroblast Growth Factor Receptor-Targeted Therapies in Breast Cancer. Cells 2018; 7:cells7070076. [PMID: 30011957 PMCID: PMC6071019 DOI: 10.3390/cells7070076] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 06/30/2018] [Accepted: 07/11/2018] [Indexed: 01/08/2023] Open
Abstract
Breast cancer (BC) is the most common malignancy and second only to lung cancer in terms of mortality in women. Despite the incredible progress made in this field, metastatic breast cancer has a poor prognosis. In an era of personalized medicine, there is an urgent need for better knowledge of the biology leading to the disease, which can lead to the design of increasingly accurate drugs against patients' specific molecular aberrations. Among one of the actionable targets is the fibroblast growth factor receptor (FGFR) pathway, triggered by specific ligands. The Fibroblast Growth Factor Receptors/Fibroblast Growth Factors (FGFRs/FGFs) axis offers interesting molecular targets to be pursued in clinical development. This mini-review will focus on the current knowledge of FGFR mutations, which lead to tumor formation and summarizes the state-of-the-art therapeutic strategies for targeted treatments against the FGFRs/FGFs axis in the context of BC.
Collapse
|
79
|
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
|
80
|
Liu H, Fang Q, Wang M, Wang W, Zhang M, Zhang D, He Y, Zhang Y, Wang H, Otero M, Ma T, Chen J. FGF8 and FGFR3 are up-regulated in hypertrophic chondrocytes: Association with chondrocyte death in deep zone of Kashin-Beck disease. Biochem Biophys Res Commun 2018; 500:184-190. [DOI: 10.1016/j.bbrc.2018.04.023] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 04/03/2018] [Indexed: 11/26/2022]
|
81
|
Tang Y, Tang LY, Xu X, Li C, Deng C, Zhang YE. Generation of Smurf2 Conditional Knockout Mice. Int J Biol Sci 2018; 14:542-548. [PMID: 29805305 PMCID: PMC5968846 DOI: 10.7150/ijbs.24303] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 02/27/2018] [Indexed: 01/17/2023] Open
Abstract
Smad ubiquitin regulatory factor 2 (Smurf2) is a HECT domain-containing E3 ubiquitin ligase. Together with its closely related homolog Smurf1, Smurf2 was initially recognized as a negative regulator of transforming growth factor-β (TGF-β) and bone morphogenetic protein (BMP) signaling, but subsequent studies have expanded its function to regulate many different signaling pathways and play important roles in genomic stability, cell polarity, tissue homeostasis and carcinogenesis. Mice with conventional knockout of Smurf1 or Smurf2 alleles are viable, but conventional Smurf1 and Smurf2 double knockout mice were early embryonic lethal. In order to study the physiological function of Smurfs during late stage of embryonic development or in adult animals, we generated Smurf2flox/flox mice carrying a targeted mutation for conditional Smurf2 gene inactivation. We demonstrated that Cre-mediated recombination using Alb-Cre, a Cre line expressed in hepatocyte, results in specific deletion of the gene in liver tissue. We also showed that Cre-mediated recombination in mouse embryonic fibroblasts (MEFs) with Smurf2flox/flox genotype resulted in generation of Smurf2 knockout MEFs, and Smurf2 deficiency affects multiple signaling pathways. Therefore, this animal model will be useful to study the distinct roles of Smurf2 in different tissues at different ages.
Collapse
Affiliation(s)
- Yi Tang
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Liu-Ya Tang
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Xuan Xu
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Cuiling Li
- Genetics of Development and Disease Branch, National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Chuxia Deng
- Genetics of Development and Disease Branch, National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA.,Present address: Faculty of Health Sciences, University of Macau, Avenida da Universidade, Taipa, Macau, China
| | - Ying E Zhang
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| |
Collapse
|
82
|
Coffin JD, Homer-Bouthiette C, Hurley MM. Fibroblast Growth Factor 2 and Its Receptors in Bone Biology and Disease. J Endocr Soc 2018; 2:657-671. [PMID: 29942929 PMCID: PMC6009610 DOI: 10.1210/js.2018-00105] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 05/23/2018] [Indexed: 01/24/2023] Open
Abstract
The fibroblast growth factor (FGF) regulatory axis is phylogenetically ancient, evolving into a large mammalian/human gene family of 22 ligands that bind to four receptor tyrosine kinases for a complex physiologic system controlling cell growth, differentiation, and metabolism. The tissue targets for the primary FGF function are mainly in cartilage and in bone for morphogenesis, mineralization, and metabolism. A multitude of complexities in the FGF ligand-receptor signaling pathways have made translation into therapies for FGF-related bone disorders such as osteomalacia, osteoarthritis, and osteoporosis difficult but not impossible.
Collapse
Affiliation(s)
| | | | - Marja Marie Hurley
- Department of Medicine, University of Connecticut School of Medicine, UCONN Health, Farmington, Connecticut
| |
Collapse
|
83
|
Yan J, Li J, Hu J, Zhang L, Wei C, Sultana N, Cai X, Zhang W, Cai CL. Smad4 deficiency impairs chondrocyte hypertrophy via the Runx2 transcription factor in mouse skeletal development. J Biol Chem 2018; 293:9162-9175. [PMID: 29735531 DOI: 10.1074/jbc.ra118.001825] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 04/20/2018] [Indexed: 12/25/2022] Open
Abstract
Chondrocyte hypertrophy is the terminal step in chondrocyte differentiation and is crucial for endochondral bone formation. How signaling pathways regulate chondrocyte hypertrophic differentiation remains incompletely understood. In this study, using a Tbx18:Cre (Tbx18Cre/+) gene-deletion approach, we selectively deleted the gene for the signaling protein SMAD family member 4 (Smad4f/f ) in the limbs of mice. We found that the Smad4-deficient mice develop a prominent shortened limb, with decreased expression of chondrocyte differentiation markers, including Col2a1 and Acan, in the humerus at mid-to-late gestation. The most striking defects in these mice were the absence of stylopod elements and failure of chondrocyte hypertrophy in the humerus. Moreover, expression levels of the chondrocyte hypertrophy-related markers Col10a1 and Panx3 were significantly decreased. Of note, we also observed that the expression of runt-related transcription factor 2 (Runx2), a critical mediator of chondrocyte hypertrophy, was also down-regulated in Smad4-deficient limbs. To determine how the skeletal defects arose in the mouse mutants, we performed RNA-Seq with ChIP-Seq analyses and found that Smad4 directly binds to regulatory elements in the Runx2 promoter. Our results suggest a new mechanism whereby Smad4 controls chondrocyte hypertrophy by up-regulating Runx2 expression during skeletal development. The regulatory mechanism involving Smad4-mediated Runx2 activation uncovered here provides critical insights into bone development and pathogenesis of chondrodysplasia.
Collapse
Affiliation(s)
- Jianyun Yan
- From the Department of Developmental and Regenerative Biology, The Mindich Child Health and Development Institute, and The Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029.,the Laboratory of Heart Center and Department of Cardiology, Heart Center, Zhujiang Hospital, Southern Medical University, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou 510280, China, and
| | - Jun Li
- From the Department of Developmental and Regenerative Biology, The Mindich Child Health and Development Institute, and The Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Jun Hu
- From the Department of Developmental and Regenerative Biology, The Mindich Child Health and Development Institute, and The Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Lu Zhang
- From the Department of Developmental and Regenerative Biology, The Mindich Child Health and Development Institute, and The Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Chengguo Wei
- the Renal Division of the Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Nishat Sultana
- From the Department of Developmental and Regenerative Biology, The Mindich Child Health and Development Institute, and The Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Xiaoqiang Cai
- From the Department of Developmental and Regenerative Biology, The Mindich Child Health and Development Institute, and The Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Weijia Zhang
- the Renal Division of the Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Chen-Leng Cai
- From the Department of Developmental and Regenerative Biology, The Mindich Child Health and Development Institute, and The Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029,
| |
Collapse
|
84
|
Ledwon JK, Turin SY, Gosain AK, Topczewska JM. The expression of fgfr3 in the zebrafish head. Gene Expr Patterns 2018; 29:32-38. [PMID: 29630949 DOI: 10.1016/j.gep.2018.04.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 03/08/2018] [Accepted: 04/03/2018] [Indexed: 12/29/2022]
Abstract
Fibroblast growth factor (FGF) signaling is essential for many developmental processes and plays a pivotal role in skeletal homeostasis, regeneration and wound healing. FGF signals through one of five tyrosine kinase receptors: Fgfr1a, -1b, -2, -3, -4. To characterize the expression of zebrafish fgfr3 from the larval stage to adulthood, we used RNAscope in situ hybridization on paraffin sections of the zebrafish head. Our study revealed spatial and temporal distribution of fgfr3 transcript in chondrocytes of the head cartilages, osteoblasts involved in bone formation, ventricular zone of the brain, undifferentiated mesenchymal cells of the skin, and lens epithelium of the eye. In general, the expression pattern of zebrafish fgfr3 is similar to the expression observed in higher vertebrates.
Collapse
Affiliation(s)
- Joanna K Ledwon
- Northwestern University Feinberg School of Medicine, Ann & Robert H. Lurie Children's Hospital of Chicago, Division of Plastic and Reconstructive Surgery, Stanley Manne Children's Research Institute, Chicago, IL, USA
| | - Sergey Y Turin
- Northwestern University Feinberg School of Medicine, Ann & Robert H. Lurie Children's Hospital of Chicago, Division of Plastic and Reconstructive Surgery, Stanley Manne Children's Research Institute, Chicago, IL, USA
| | - Arun K Gosain
- Northwestern University Feinberg School of Medicine, Ann & Robert H. Lurie Children's Hospital of Chicago, Division of Plastic and Reconstructive Surgery, Stanley Manne Children's Research Institute, Chicago, IL, USA
| | - Jolanta M Topczewska
- Northwestern University Feinberg School of Medicine, Ann & Robert H. Lurie Children's Hospital of Chicago, Division of Plastic and Reconstructive Surgery, Stanley Manne Children's Research Institute, Chicago, IL, USA.
| |
Collapse
|
85
|
Moore ER, Jacobs CR. The primary cilium as a signaling nexus for growth plate function and subsequent skeletal development. J Orthop Res 2018; 36:533-545. [PMID: 28901584 PMCID: PMC5839937 DOI: 10.1002/jor.23732] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 09/07/2017] [Indexed: 02/04/2023]
Abstract
The primary cilium is a solitary, antenna-like sensory organelle with many important roles in cartilage and bone development, maintenance, and function. The primary cilium's potential role as a signaling nexus in the growth plate makes it an attractive therapeutic target for diseases and disorders associated with bone development and maintenance. Many signaling pathways that are mediated by the cilium-such as Hh, Wnt, Ihh/PTHrP, TGFβ, BMP, FGF, and Notch-are also known to influence endochondral ossification, primarily by directing growth plate formation and chondrocyte behavior. Although a few studies have demonstrated that these signaling pathways can be directly tied to the primary cilium, many pathways have yet to be evaluated in context of the cilium. This review serves to bridge this knowledge gap in the literature, as well as discuss the cilium's importance in the growth plate's ability to sense and respond to chemical and mechanical stimuli. Furthermore, we explore the importance of using the appropriate mechanism to study the cilium in vivo and suggest IFT88 deletion is the best available technique. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 36:533-545, 2018.
Collapse
Affiliation(s)
- Emily R. Moore
- Department of Biomedical Engineering; Columbia University; 351 Engineering Terrace, Mail Code 8904, 1210 Amsterdam Avenue New York 10027 New York
| | - Christopher R. Jacobs
- Department of Biomedical Engineering; Columbia University; 351 Engineering Terrace, Mail Code 8904, 1210 Amsterdam Avenue New York 10027 New York
| |
Collapse
|
86
|
Shazeeb MS, Cox MK, Gupta A, Tang W, Singh K, Pryce CT, Fogle R, Mu Y, Weber WD, Bangari DS, Ying X, Sabbagh Y. Skeletal Characterization of the Fgfr3 Mouse Model of Achondroplasia Using Micro-CT and MRI Volumetric Imaging. Sci Rep 2018; 8:469. [PMID: 29323153 PMCID: PMC5765052 DOI: 10.1038/s41598-017-18801-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 12/18/2017] [Indexed: 01/16/2023] Open
Abstract
Achondroplasia, the most common form of dwarfism, affects more than a quarter million people worldwide and remains an unmet medical need. Achondroplasia is caused by mutations in the fibroblast growth factor receptor 3 (FGFR3) gene which results in over-activation of the receptor, interfering with normal skeletal development leading to disproportional short stature. Multiple mouse models have been generated to study achondroplasia. The characterization of these preclinical models has been primarily done with 2D measurements. In this study, we explored the transgenic model expressing mouse Fgfr3 containing the achondroplasia mutation G380R under the Col2 promoter (Ach). Survival and growth rate of the Ach mice were reduced compared to wild-type (WT) littermates. Axial skeletal defects and abnormalities of the sternebrae and vertebrae were observed in the Ach mice. Further evaluation of the Ach mouse model was performed by developing 3D parameters from micro-computed tomography (micro-CT) and magnetic resonance imaging (MRI). The 3-week-old mice showed greater differences between the Ach and WT groups compared to the 6-week-old mice for all parameters. Deeper understanding of skeletal abnormalities of this model will help guide future studies for evaluating novel and effective therapeutic approaches for the treatment of achondroplasia.
Collapse
Affiliation(s)
- Mohammed Salman Shazeeb
- Global Bioimaging Department, Translational In-vivo Models, Sanofi R&D Global Research Platform, 49 New York Avenue, Framingham, MA, 01701, United States
| | - Megan K Cox
- Rare Diseases, Sanofi, 49 New York Avenue, Framingham, MA, 01701, USA
| | - Anurag Gupta
- Global Bioimaging Department, Translational In-vivo Models, Sanofi R&D Global Research Platform, 49 New York Avenue, Framingham, MA, 01701, United States
| | - Wen Tang
- Rare Diseases, Sanofi, 49 New York Avenue, Framingham, MA, 01701, USA
| | - Kuldeep Singh
- Global Discovery Pathology, Translational In-vivo Models, Sanofi R&D Global Research Platform, 5 The Mountain Road, Framingham, MA, 01701, USA
| | - Cynthia T Pryce
- Translational Sciences, Sanofi R&D Global Research Platform, 49 New York avenue, Framingham, MA, 01701, United States
| | - Robert Fogle
- Global Bioimaging Department, Translational In-vivo Models, Sanofi R&D Global Research Platform, 49 New York Avenue, Framingham, MA, 01701, United States
| | - Ying Mu
- Global Bioimaging Department, Translational In-vivo Models, Sanofi R&D Global Research Platform, 49 New York Avenue, Framingham, MA, 01701, United States
| | - William D Weber
- Translational Sciences, Sanofi R&D Global Research Platform, 49 New York avenue, Framingham, MA, 01701, United States
| | - Dinesh S Bangari
- Global Discovery Pathology, Translational In-vivo Models, Sanofi R&D Global Research Platform, 5 The Mountain Road, Framingham, MA, 01701, USA
| | - Xiaoyou Ying
- Global Bioimaging Department, Translational In-vivo Models, Sanofi R&D Global Research Platform, 49 New York Avenue, Framingham, MA, 01701, United States.
| | - Yves Sabbagh
- Rare Diseases, Sanofi, 49 New York Avenue, Framingham, MA, 01701, USA.
| |
Collapse
|
87
|
Yu J, Liang F, Huang H, Pirttiniemi P, Yu D. Effects of loading on chondrocyte hypoxia, HIF-1α and VEGF in the mandibular condylar cartilage of young rats. Orthod Craniofac Res 2017; 21:41-47. [PMID: 29271061 DOI: 10.1111/ocr.12212] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/22/2017] [Indexed: 12/19/2022]
Abstract
OBJECTIVES To investigate hypoxia-inducible factor 1-alpha (HIF-1α) and vascular endothelial growth factor (VEGF) expression under altered loading, and to explore the relationship between loading and hypoxia in the mandibular condylar cartilage of young rats. SETTING AND SAMPLE POPULATION Eighty Sprague-Dawley rats. MATERIAL AND METHODS The reduced loading group was fed soft food, and their incisors were cut to avoid occlusal contact. The increased loading group was fed hard food and had forced jaw-opening. Ten rats from each group (n = 10) were sacrificed at 12, 24, 48, and 96 hours after initiation of the experiment. Pimonidazole hydrochloride (Hypoxyprobe-1, HP-1) was used as a hypoxia marker to confirm the hypoxic state. Hypoxic chondrocytes as indicated by HP-1, HIF-1α and VEGF protein expressions were recognized by immunohistochemical detection. HIF-1α and VEGF mRNA expressions were detected by semi-quantitative RT-PCR. RESULTS Hypoxyprobe-1 was confined in the upper layers of cartilage, and was most strongly expressed in the weight-bearing area of TMJ at 12 and 96 hours. Staining of HIF-1α and VEGF was most strongly expressed in the chondrocytes of the fibrous and proliferative layer at all time points. Furthermore, expressions were also displayed in the hypertrophic and calcified layers at 48 and 96 hours. The expressions of HIF-1α and VEGF mRNA were higher in the increased loading group than in the reduced loading group at 48 and 96 hours (P < . 05). CONCLUSION Mechanical loading seems to directly induce weight-bearing area hypoxia followed by new vessel formation, which indicates that these factors are related and important for the development of cartilage.
Collapse
Affiliation(s)
- J Yu
- Department of Oral Development and Orthodontics, Institute of Dentistry, University of Oulu, Oulu, Finland
| | - F Liang
- Department of Oral and Maxillofacial Surgery, College of Stomatology, Guangxi Medical University, Nanning, China
| | - H Huang
- Department of Oral and Maxillofacial Surgery, College of Stomatology, Guangxi Medical University, Nanning, China
| | - P Pirttiniemi
- Department of Oral Development and Orthodontics, Institute of Dentistry, University of Oulu, Oulu, Finland
| | - D Yu
- Department of Stomatology, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
88
|
Montone R, Romanelli MG, Baruzzi A, Ferrarini F, Liboi E, Lievens PMJ. Mutant FGFR3 associated with SADDAN disease causes cytoskeleton disorganization through PLCγ1/Src-mediated paxillin hyperphosphorylation. Int J Biochem Cell Biol 2017; 95:17-26. [PMID: 29242050 DOI: 10.1016/j.biocel.2017.12.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 11/15/2017] [Accepted: 12/08/2017] [Indexed: 10/18/2022]
Abstract
K650M/E substitutions in the Fibroblast growth factor receptor 3 (FGFR3) are associated with Severe Achondroplasia with Developmental Delay and Acanthosis Nigricans (SADDAN) and Thanatophoric Dysplasia type II (TDII), respectively. Both SADDAN and TDII present with affected endochondral ossification marked by impaired chondrocyte functions and growth plate disorganization. In vitro, K650M/E substitutions confer FGFR3 constitutive kinase activity leading to impaired biosynthesis and accumulation of immature receptors in endoplasmic reticulum (ER)/Golgi. From those compartments, both SADDAN-FGFR3 and TDII-FGFR3 receptors engender uncontrolled signalling, activating PLCγ1, signal transducer and activator of transcription 1, 3 and 5 (STAT1/3/5) and ERK1/2 effectors. Here, we investigated the impact of SADDAN-FGFR3 and TDII-FGFR3 signalling on cytoskeletal organization. We report that SADDAN-FGFR3, but not TDII-FGFR3, affects F-actin organization by inducing tyrosine hyperphosphorylation of paxillin, a key regulator of focal adhesions and actin dynamics. Paxillin phosphorylation was upregulated at tyrosine 118, a functional target of Src and FAK kinases. By using Src-deficient cells and a Src kinase inhibitor, we established a role played by Src activation in paxillin hyperphosphorylation. Moreover, we found that SADDAN-FGFR3 induced FAK phosphorylation at tyrosines 576/577, suggesting its involvement as a Src co-activator in paxillin phosphorylation. Interestingly, paxillin hyperphosphorylation by SADDAN-FGFR3 caused paxillin mislocalization and partial co-localization with the mutant receptor. Finally, the SADDAN-FGFR3 double mutant unable to bind PLCγ1 failed to promote paxillin hyperphosphorylation, pointing to PLCγ1 as an early player in mediating paxillin alterations. Overall, our findings contribute to elucidate the molecular mechanism leading to cell dysfunctions caused by SADDAN-FGFR3 signalling.
Collapse
Affiliation(s)
- R Montone
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biology and Genetics, University of Verona Medical School, Verona, Italy
| | - M G Romanelli
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biology and Genetics, University of Verona Medical School, Verona, Italy
| | - A Baruzzi
- Department of Pathology and Diagnostics, University of Verona Medical School, Verona, Italy
| | - F Ferrarini
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biology and Genetics, University of Verona Medical School, Verona, Italy
| | - E Liboi
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biology and Genetics, University of Verona Medical School, Verona, Italy
| | - P M-J Lievens
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biology and Genetics, University of Verona Medical School, Verona, Italy.
| |
Collapse
|
89
|
Shuhaibar LC, Robinson JW, Vigone G, Shuhaibar NP, Egbert JR, Baena V, Uliasz TF, Kaback D, Yee SP, Feil R, Fisher MC, Dealy CN, Potter LR, Jaffe LA. Dephosphorylation of the NPR2 guanylyl cyclase contributes to inhibition of bone growth by fibroblast growth factor. eLife 2017; 6:31343. [PMID: 29199951 PMCID: PMC5745078 DOI: 10.7554/elife.31343] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 12/02/2017] [Indexed: 01/17/2023] Open
Abstract
Activating mutations in fibroblast growth factor (FGF) receptor 3 and inactivating mutations in the NPR2 guanylyl cyclase both cause severe short stature, but how these two signaling systems interact to regulate bone growth is poorly understood. Here, we show that bone elongation is increased when NPR2 cannot be dephosphorylated and thus produces more cyclic GMP. By developing an in vivo imaging system to measure cyclic GMP production in intact tibia, we show that FGF-induced dephosphorylation of NPR2 decreases its guanylyl cyclase activity in growth plate chondrocytes in living bone. The dephosphorylation requires a PPP-family phosphatase. Thus FGF signaling lowers cyclic GMP production in the growth plate, which counteracts bone elongation. These results define a new component of the signaling network by which activating mutations in the FGF receptor inhibit bone growth. Between birth and puberty, the bones of mammals grow drastically in length. This process is controlled by many proteins, and mutations affecting these proteins can cause bones to either be too long or too short. For example, mutations of a protein called the fibroblast growth factor receptor, or FGF for short, and a protein called NPR2, can cause similar forms of dwarfism – a condition characterized by short stature. The FGF protein controls bone growth, and people with overactive receptors for FGF suffer from a form of dwarfism known as achondroplasia, while people that lack FGF receptors have longer bones. The NPR2 protein, on the other hand, produces a molecule called cGMP, which is necessary for the bones to grow. When NPR2 is blocked, less cGMP is produced, which results in shorter limbs. Previous studies of bone cells grown in the laboratory have shown that these two proteins are linked by a chain of chemical messages. When the FGF receptor is active, phosphate molecules are removed from the NPR2 protein, which reduces the amount of GMP produced. However, until now it was not known whether this mechanism also controls growth in actual bones. Here, Shuhaibar et al. used genetically modified mice in which the phosphate group could not be removed from their NPR2 enzyme. As a result, the bones of these mice were longer than usual. Shuhaibar et al. then developed an imaging technique to examine the region in the bone were growth happens. To see whether FGF reduces the amount of cGMP produced by NPR2 in these areas, cGMP was detected with a fluorescent sensor in order to be tracked. In normal mice, the FGF receptor reduced the rate at which cGMP was produced, but in mice with mutated NPR2, this did not happen. When the cells could not remove the phosphates from NPR2, cGMP levels stayed high and the bones grew longer. These findings reveal new insights into the molecular causes of dwarfism. The next step will be to identify the enzyme responsible for removing phosphate from NPR2. Blocking its activity could help to enhance bone growth. In the future, this could lead to new drug treatments for achondroplasia.
Collapse
Affiliation(s)
- Leia C Shuhaibar
- Department of Cell Biology, University of Connecticut Health Center, Farmington, United States
| | - Jerid W Robinson
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, United States
| | - Giulia Vigone
- Department of Cell Biology, University of Connecticut Health Center, Farmington, United States
| | - Ninna P Shuhaibar
- Department of Cell Biology, University of Connecticut Health Center, Farmington, United States
| | - Jeremy R Egbert
- Department of Cell Biology, University of Connecticut Health Center, Farmington, United States
| | - Valentina Baena
- Department of Cell Biology, University of Connecticut Health Center, Farmington, United States
| | - Tracy F Uliasz
- Department of Cell Biology, University of Connecticut Health Center, Farmington, United States
| | - Deborah Kaback
- Department of Cell Biology, University of Connecticut Health Center, Farmington, United States
| | - Siu-Pok Yee
- Department of Cell Biology, University of Connecticut Health Center, Farmington, United States
| | - Robert Feil
- Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany
| | - Melanie C Fisher
- Center for Regenerative Medicine and Skeletal Development, University of Connecticut Health Center, Farmington, United States
| | - Caroline N Dealy
- Center for Regenerative Medicine and Skeletal Development, University of Connecticut Health Center, Farmington, United States
| | - Lincoln R Potter
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, United States
| | - Laurinda A Jaffe
- Department of Cell Biology, University of Connecticut Health Center, Farmington, United States
| |
Collapse
|
90
|
Osawa Y, Matsushita M, Hasegawa S, Esaki R, Fujio M, Ohkawara B, Ishiguro N, Ohno K, Kitoh H. Activated FGFR3 promotes bone formation via accelerating endochondral ossification in mouse model of distraction osteogenesis. Bone 2017; 105:42-49. [PMID: 28802681 DOI: 10.1016/j.bone.2017.05.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 05/15/2017] [Accepted: 05/19/2017] [Indexed: 01/19/2023]
Abstract
Achondroplasia (ACH) is one of the most common short-limbed skeletal dysplasias caused by gain-of-function mutations in the fibroblast growth factor receptors 3 (FGFR3) gene. Distraction osteogenesis (DO) is a treatment option for short stature in ACH in some countries. Although the patients with ACH usually show faster healing in DO, details of the newly formed bone have not been examined. We have developed a mouse model of DO and analyzed new bone regenerates of the transgenic mice with ACH (Fgfr3ach mice) histologically and morphologically. We established two kinds of DO protocols, the short-DO consisted of 5days of latency period followed by 5days of distraction with a rate of 0.4mm per 24h, and the long-DO consisted of the same latency period followed by 7days of distraction with a rate of 0.3mm per 12h. The callus formation was evaluated radiologically by bone fill score and quantified by micro-CT scan in both protocols. The histomorphometric analysis was performed in the short-DO protocol by various stainings, including Villanueva Goldner, Safranin-O/Fast green, tartrate-resistant acid phosphatase, and type X collagen. Bone fill scores were significantly higher in Fgfr3ach mice than in wild-type mice in both protocols. The individual bone parameters, including bone volume and bone volume/tissue volume, were also significantly higher in Fgfr3ach mice than in wild-type mice in both protocols. The numbers of osteoblasts, as well as osteoclasts, around the trabecular bone were increased in Fgfr3ach mice. Cartilaginous tissues of the distraction region rapidly disappeared in Fgfr3ach mice compared to wild-type mice during the consolidation phase. Similarly, type X collagen-positive cells were markedly decreased in Fgfr3ach mice during the same period. Fgfr3ach mice exhibited accelerated bone regeneration after DO. Accelerated endochondral ossification could contribute to faster healing in Fgfr3ach mice.
Collapse
Affiliation(s)
- Yusuke Osawa
- Department of Orthopaedic Surgery, Nagoya University Graduate School of Medicine, Japan; Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Japan.
| | - Masaki Matsushita
- Department of Orthopaedic Surgery, Nagoya University Graduate School of Medicine, Japan; Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Japan
| | - Sachi Hasegawa
- Department of Orthopaedic Surgery, Aichi Prefectural Colony Central Hospital, Japan
| | - Ryusaku Esaki
- Department of Orthopaedic Surgery, Nagoya University Graduate School of Medicine, Japan; Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Japan
| | - Masahito Fujio
- Department of Oral and Maxillofacial Surgery, Nagoya University Graduate School of Medicine, Japan
| | - Bisei Ohkawara
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Japan
| | - Naoki Ishiguro
- Department of Orthopaedic Surgery, Nagoya University Graduate School of Medicine, Japan
| | - Kinji Ohno
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Japan
| | - Hiroshi Kitoh
- Department of Orthopaedic Surgery, Nagoya University Graduate School of Medicine, Japan
| |
Collapse
|
91
|
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
|
92
|
Regulation of human and mouse telomerase genes by genomic contexts and transcription factors during embryonic stem cell differentiation. Sci Rep 2017; 7:16444. [PMID: 29180668 PMCID: PMC5703907 DOI: 10.1038/s41598-017-16764-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 11/16/2017] [Indexed: 12/14/2022] Open
Abstract
Differential regulation of telomerase reverse transcriptase (TERT) genes contribute to distinct aging and tumorigenic processes in humans and mice. To study TERT regulation, we generated mouse embryonic stem cell (ESC) lines containing single-copy bacterial artificial chromosome (BAC) reporters, covering hTERT and mTERT genes and their neighboring loci, via recombinase-mediated BAC targeting. ESC lines with chimeric BACs, in which two TERT promoters were swapped, were also generated. Using these chromatinized BACs, we showed that hTERT silencing during differentiation to embryoid bodies (EBs) and to fibroblast-like cells was driven by the human-specific genomic context and accompanied by increases of repressive epigenetic marks, H3K9me3 and H3K27me3, near its promoter. Conversely, the mouse genomic context did not repress TERT transcription until late during differentiation. The hTERT promoter was more active than its mouse counterpart when compared in the same genomic contexts. Mutations of E-box and E2F consensus sites at the promoter had little effect on hTERT transcription in ESCs. However, the mutant promoters were rapidly silenced upon EB differentiation, indicating that transcription factors (TFs) bound to these sites were critical in maintaining hTERT transcription during differentiation. Together, our study revealed a dynamic hTERT regulation by chromatin environment and promoter-bound TFs during ESC differentiation.
Collapse
|
93
|
Li R, Herriges JC, Chen L, Mecham RP, Sun X. FGF receptors control alveolar elastogenesis. Development 2017; 144:4563-4572. [PMID: 29122839 DOI: 10.1242/dev.149443] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 09/28/2017] [Indexed: 12/21/2022]
Abstract
Alveologenesis, the final step of lung development, is characterized by the formation of millions of alveolar septa that constitute the vast gas-exchange surface area. The genetic network driving alveologenesis is poorly understood compared with earlier steps in lung development. FGF signaling through receptors Fgfr3 and Fgfr4 is crucial for alveologenesis, but the mechanisms through which they mediate this process remain unclear. Here we show that in Fgfr3;Fgfr4 (Fgfr3;4) global mutant mice, alveolar simplification is first observed at the onset of alveologenesis at postnatal day 3. This is preceded by disorganization of elastin, indicating defects in the extracellular matrix (ECM). Although Fgfr3 and Fgfr4 are expressed in the mesenchyme and epithelium, inactivation in the mesenchyme, but not the epithelium, recapitulated the defects. Expression analysis of components of the elastogenesis machinery revealed that Mfap5 (also known as Magp2), which encodes an elastin-microfibril bridging factor, is upregulated in Fgfr3;4 mutants. Mfap5 mutation in the Fgfr3;4 mutant background partially attenuated the alveologenesis defects. These data demonstrate that, during normal lung maturation, FGF signaling restricts expression of the elastogenic machinery in the lung mesenchyme to control orderly formation of the elastin ECM, thereby driving alveolar septa formation to increase the gas-exchange surface.
Collapse
Affiliation(s)
- Rongbo Li
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA.,Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - John C Herriges
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Lin Chen
- Department of Rehabilitation Medicine, Center of Bone Metabolism and Repair (CBMR), Trauma Center, State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Robert P Mecham
- Department of Cell Biology & Physiology, Washington University School of Medicine, Saint Louis, MO 631103, USA
| | - Xin Sun
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA .,Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI 53706, USA
| |
Collapse
|
94
|
Bombesin-like receptor 3 (Brs3) expression in glutamatergic, but not GABAergic, neurons is required for regulation of energy metabolism. Mol Metab 2017; 6:1540-1550. [PMID: 29107299 PMCID: PMC5681273 DOI: 10.1016/j.molmet.2017.08.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 08/24/2017] [Accepted: 08/28/2017] [Indexed: 02/03/2023] Open
Abstract
Objective Bombesin-like receptor 3 (BRS-3) is an orphan G protein-coupled receptor. Brs3 null mice have reduced resting metabolic rate and body temperature, increased food intake, and obesity. Here we study the role of Brs3 in different neuron types. Methods Mice able to undergo Cre recombinase-dependent inactivation or re-expression of Brs3 were generated, respectively Brs3fl/y and Brs3loxTB/y. We then studied four groups of mice with Brs3 selectively inactivated or re-expressed in cells expressing Vglut2-Cre or Vgat-Cre. Results Deletion of Brs3 in glutamatergic neurons expressing Vglut2 reproduced the global null phenotype for regulation of food intake, metabolic rate, body temperature, adiposity, and insulin resistance. These mice also no longer responded to a BRS-3 agonist, MK-5046. In contrast, deletion of Brs3 in GABAergic neurons produced no detectable phenotype. Conversely, the wild type phenotype was restored by selective re-expression of Brs3 in glutamatergic neurons, with no normalization achieved by re-expressing Brs3 in GABAergic neurons. Conclusions Brs3 expression in glutamatergic neurons is both necessary and sufficient for full Brs3 function in energy metabolism. In these experiments, no function was identified for Brs3 in GABAergic neurons. The data suggest that the anti-obesity pharmacologic actions of BRS-3 agonists occur via agonism of receptors on glutamatergic neurons. Brs3 in glutamatergic neurons regulates food intake, metabolic rate, and body weight. Brs3 in glutamatergic neurons is both necessary and sufficient for these functions. No phenotypes were identified by Brs3 loss or re-expression in GABAergic neurons. BRS-3 agonists likely act on glutamatergic neurons for their anti-obesity effects.
Collapse
|
95
|
Statins do not inhibit the FGFR signaling in chondrocytes. Osteoarthritis Cartilage 2017; 25:1522-1530. [PMID: 28583899 DOI: 10.1016/j.joca.2017.05.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 05/19/2017] [Accepted: 05/25/2017] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Statins are widely used drugs for cholesterol lowering, which were recently found to counteract the effects of aberrant fibroblast growth factor receptor (FGFR3) signaling in cell and animal models of FGFR3-related chondrodysplasia. This opened an intriguing therapeutic possibility for human dwarfing conditions caused by gain-of-function mutations in FGFR3, although the mechanism of statin action on FGFR3 remains unclear. Here, we determine the effect of statins on FGFR signaling in chondrocytes. DESIGN Cultured chondrocyte cell lines, mouse embryonic tibia cultures and limb bud micromasses were treated with FGF2 to activate FGFR signaling. The effects of atorvastatin, fluvastatin, lovastatin and pravastatin on FGFR3 protein stability and on FGFR-mediated chondrocyte growth-arrest, loss of extracellular matrix (ECM), induction of premature senescence and hypertrophic differentiation were evaluated. RESULTS Statins did not alter the level of FGFR3 protein expression nor produce any effect on FGFR-mediated inhibition of chondrocyte proliferation and hypertrophic differentiation in cultured chondrocyte cell lines, mouse tibia cultures or limb bud micromasses. CONCLUSION We conclude that statins do not inhibit the FGFR signaling in chondrocytes. Therefore the statin-mediated rescue of FGFR3-related chondrodysplasia, described before, is likely not intrinsic to the growth plate cartilage.
Collapse
|
96
|
Detection of G1138A Mutation of the FGFR3 Gene in Tooth Material from a 180-Year-Old Museological Achondroplastic Skeleton. Genes (Basel) 2017; 8:genes8090214. [PMID: 28850094 PMCID: PMC5615348 DOI: 10.3390/genes8090214] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 08/18/2017] [Accepted: 08/22/2017] [Indexed: 01/04/2023] Open
Abstract
Throughout the last four centuries, many anatomical museums across the world have collected teratological specimens that became precious objects. These can be regarded as spirits of the past which have captured the morphology of diseases through time. These valuable and irreplaceable specimens can be perfectly used in contemporary dysmorphological or genetic research. Unfortunately, due to the historical nature of these specimens and the regularly used aggressive preservation fluids, DNA degradation is often present. Furthermore, the use of material for DNA extraction is restricted to preserve the appearance of these valuable museological specimens. Thus, the most challenging part in this perspective is to harvest sufficient DNA of good quality for further testing without damaging the specimens. Besides fixated specimens, most teratological collections contain dried skeletal and teeth materials which are an excellent source to extract DNA. We here present a DNA-based method that enables genetic identification of the G1138A mutation of the FGFR3 gene in a 180-year-old achondroplastic skeleton, confirming the previously morphologically determined disease. Nuclear DNA was extracted from a premolar tooth and the mutation was found using Sanger sequencing of a small region of the FGFR3 gene.
Collapse
|
97
|
Xie Y, Luo F, Xu W, Wang Z, Sun X, Xu M, Huang J, Zhang D, Tan Q, Chen B, Jiang W, Du X, Chen L. FGFR3 deficient mice have accelerated fracture repair. Int J Biol Sci 2017; 13:1029-1037. [PMID: 28924384 PMCID: PMC5599908 DOI: 10.7150/ijbs.19309] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 03/29/2017] [Indexed: 12/22/2022] Open
Abstract
Bone fracture healing is processed through multiple biological stages that partly recapitulates the skeletal development process. FGFR3 is a negative regulator of chondrogenesis during embryonic stage and plays an important role in both chondrogenesis and osteogenesis. We have investigated the role of FGFR3 in fracture healing using unstabilized fracture model and found that gain-of-function mutation of FGFR3 inhibits the initiation of chondrogenesis during cartilage callus formation. Here, we created closed, stabilized proximal tibia fractures with an intramedullary pin in Fgfr3-/-mice and their littermate wild-type mice. Fracture healing was evaluated by radiography, micro-CT, histology, and real-time polymerase chain reaction (RT-PCR) analysis. The fractured Fgfr3-/- mice had increased formation of cartilaginous callus, more fracture callus, and more rapid endochondral ossification in fracture sites with up-regulated expressions of chondrogenesis related gene. The fractures of Fgfr3-/- mice healed faster with accelerated fracture callus mineralization and up-regulated expression of osteoblastogenic genes. The healing of fractures in Fgfr3-/- mice was accelerated in the stage of formation of cartilage and endochondral ossification. Downregulation of FGFR3 activity can be considered as a potential bio-therapeutic strategy for fracture treatment.
Collapse
Affiliation(s)
- Yangli Xie
- Department of Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Fengtao Luo
- Department of Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Wei Xu
- Department of Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Zuqiang Wang
- Department of Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Xianding Sun
- Department of Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Meng Xu
- Department of Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Junlan Huang
- Department of Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Dali Zhang
- Department of Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Qiaoyan Tan
- Department of Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Bo Chen
- Department of Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Wanling Jiang
- Department of Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Xiaolan Du
- Department of Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Lin Chen
- Department of Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| |
Collapse
|
98
|
Ajmal M, Mir A, Shoaib M, Malik SA, Nasir M. Identification and in silico characterization of p.G380R substitution in FGFR3, associated with achondroplasia in a non-consanguineous Pakistani family. Diagn Pathol 2017; 12:47. [PMID: 28679403 PMCID: PMC5499044 DOI: 10.1186/s13000-017-0642-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2017] [Accepted: 06/29/2017] [Indexed: 12/03/2022] Open
Abstract
Background The dimerization efficiency of FGFR3 transmembrane domain plays a critical role in the formation of a normal skeleton through the negative regulation of bone development. Recently, gain-of-function mutations in the transmembrane domain of FGFR3 has been described associated with an aberrant negative regulation, leading to the development of achondroplasia-group disorders, including achondroplasia (ACH), hypochondroplasia (HCH) and thanatophoric dysplasia (TD). Here, we describe a non-consanguineous Pakistani family with achondroplasia to explain hereditary basis of the disease. Methods PCR-based linkage analysis using microsatellite markers was employed to localize the disease gene. Gene specific intronic primers were used to amplify the genomic DNA from all affected as well as phenotypically healthy individuals. Amplified PCR products were then subjected to Sanger sequencing and RFLP analysis to identify a potentially pathogenic mutation. The impact of identified mutation on FGFR3 protein’s structure and stability was highlighted through different bioinformatics tools. Results Genetic screening of the family revealed a previously reported heterozygous c.1138 G > A (p.G380R) mutation in the coding exon 8 of FGFR3 gene. Identified genetic variation was confirmed in all affected individuals while healthy individuals and controls were found genotypically normal. The results were further validated by RFLP analysis as c.1138 G > A substitution generates a unique recognition site for SfcI endonuclease. Following SfcI digestion, the electrophoretic pattern of three bands/DNA fragments for each patient is indicative of heterozygous status of the disease allele. In silico studies of the mutant FGFR3 protein predicted to adversely affect the stability of FGFR3 protein. Conclusions Mutation in the transmembrane domain may adversely affect the dimerization efficiency and overall stability of the FGFR3, leading to a constitutively active protein. As a result, an uncontrolled intracellular signaling or negative bone growth regulation leads to achondroplasia. Our findings support the fact that p.G380R is a common mutation among diverse population of the world and like other countries, can be used as a molecular diagnosis marker for achondroplasia in Pakistan. Electronic supplementary material The online version of this article (doi:10.1186/s13000-017-0642-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Muhammad Ajmal
- Institute of Biomedical and Genetic Engineering, 24-Mauve area, G-9/1, Islamabad, 44000, Pakistan
| | - Asif Mir
- Department of Biotechnology, International Islamic University, Islamabad, Pakistan
| | - Muhammad Shoaib
- KRL General Hospital, Orthopedic Department, 24-Mauve area, G-9/1, Islamabad, 44000, Pakistan
| | - Salman Akbar Malik
- Department of Biochemistry, Quaid-i-Azam University, Islamabad, 44000, Pakistan
| | - Muhammad Nasir
- Institute of Biomedical and Genetic Engineering, 24-Mauve area, G-9/1, Islamabad, 44000, Pakistan.
| |
Collapse
|
99
|
Defects in chondrocyte maturation and secondary ossification in mouse knee joint epiphyses due to Snorc deficiency. Osteoarthritis Cartilage 2017; 25:1132-1142. [PMID: 28323137 DOI: 10.1016/j.joca.2017.03.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 03/05/2017] [Accepted: 03/09/2017] [Indexed: 02/02/2023]
Abstract
OBJECTIVE The role of Snorc, a novel cartilage specific transmembrane proteoglycan, was studied during skeletal development using two Snorc knockout mouse models. Hypothesizing that Snorc, like the other transmembrane proteoglycans, may be a coreceptor, we also studied its interaction with growth factors. METHODS Skeletal development was studied in wild type (WT) and Snorc knockout mice during postnatal development by whole mount staining, X-ray imaging, histomorphometry, immunohistochemistry and qRT-PCR. Snorc promoter activity was studied by applying the LacZ reporter expressed by the targeting construct. Slot blot binding and cell proliferation assays were used to study the interaction of Snorc with several growth factors. RESULTS Snorc expression was localized in the knee epiphyses especially to the prehypertrophic chondrocytes delineating the cartilage canals and secondary ossification center (SOC). Snorc was demonstrated to have a glycosaminoglycan independent affinity to FGF2 and it inhibited FGF2 dependent cell growth of C3H101/2 cells. In Snorc deficient mice, SOCs in knee epiphyses were smaller, and growth plate (GP) maturation was disturbed, but total bone length was not affected. Central proliferative and hypertrophic zones were enlarged with higher extracellular matrix (ECM) volume and rounded chondrocyte morphology at postnatal days P10 and P22. Increased levels of Ihh and Col10a1, and reduced Mmp13 mRNA expression were observed at P10. CONCLUSIONS These findings suggest a role of Snorc in regulation of chondrocyte maturation and postnatal endochondral ossification. The interaction identified between recombinant Snorc core protein and FGF2 suggest functions related to FGF signaling.
Collapse
|
100
|
Abstract
Most toxic physeal changes are characterized microscopically by altered chondrocyte development, proliferation, or maturation in the growth plate and eventually result in disordered appositional bone growth. Many therapeutic drugs directly or indirectly target proteins involved in chondrocytic differentiation and maturation pathways, so toxic physeal injury has become increasingly common in preclinical toxicologic pathology. While physeal dysplasia has been associated with several different drug classes including bisphosphonates, vascular endothelial growth factor receptor inhibitors, fibroblast growth factor receptor inhibitors, transforming growth factor beta receptor inhibitors, and vascular targeting agents, physeal changes often share similar morphologic features including thickening and disorganization of the hypertrophic layer, increased numbers of hypertrophic chondrocytes, altered mineralization of endochondral ossification, and/or increased thickness of subphyseal bone. Knowledge of genetic and nutritional diseases affecting bone growth has been important in helping to determine which specific target drugs may be affecting that could result in toxic physeal lesions. A pathophysiologic mechanism for most physeal toxicants has been determined in detail using a variety of investigative techniques. However, due to the signaling cross talk and the tight regulation required for chondrocyte maturation in the physis, several growth factor pathways are likely to be affected simultaneously with pharmacologic disruption of physeal homeostasis and inhibition of one factor necessary for chondrocyte function often affects others.
Collapse
|