1
|
Chantre-justino M, Silvestre RT, De Castro TL, Luz E, Pinheiro RDCES, Caruso A, Lopes ACDS, Meohas W, Alves G, Ornellas MHF. Genetic profiling of osteosarcoma in an adolescent using a next‑generation sequencing panel and Sanger sequencing: A case report and review of the literature. Biomed Rep 2025; 22:42. [PMID: 39810900 PMCID: PMC11729137 DOI: 10.3892/br.2025.1920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 11/20/2024] [Indexed: 01/16/2025] Open
Abstract
Osteosarcoma (OS) is the most common malignant bone tumor affecting adolescents and young adults and it usually occurs in the long bones of the extremities. The detection of cancer-related genetic alterations has a growing effect in guiding diagnosis, prognosis and targeted therapies. However, little is known about the molecular aspects involved in the etiology and progression of OS, which limits options for targeted therapies. The present study described a case of an adolescent patient (16-years-old) who was diagnosed with conventional central OS in the right distal femur without the evidence of pulmonary metastases; the patient was treated with surgery and adjuvant chemotherapy. Genetic alterations in resected tumor tissue were investigated via next-generation sequencing (NGS) technology using a targeted NGS panel. Sanger sequencing was also performed to investigate somatic and germline TP53 mutations (exons 4-8). NGS analysis revealed an intratumor heterogeneity signature in OS tumor, including several single nucleotide variants identified in genes encoding tyrosine kinase proteins. No PCR products for TP53 exon 5 were detected in the tumor sample by PCR analysis prior to Sanger sequencing, suggesting a significant deletion in this exon. Sanger sequencing analysis revealed the missense variant TP53 c.712T>A (p.Cys238Ser) in tumor tissue sample, thus reinforcing the role of TP53 somatic mutations in OS development. Additionally, the TP53 c.215C>G (p.Pro72Arg) germline missense variant was identified in the peripheral blood sample. In conclusion, the findings provided new information on genetic aspects that may contribute to OS development, especially in pediatric patients.
Collapse
Affiliation(s)
- Mariana Chantre-justino
- Research Division, National Institute of Traumatology and Orthopedics, Rio de Janeiro 20940-070, Brazil
| | - Rafaele Tavares Silvestre
- Circulating Biomarkers Laboratory, Pathology Department, Faculty of Medical Sciences, Rio de Janeiro State University, Rio de Janeiro 20550-170, Brazil
| | - Thiago Luz De Castro
- Circulating Biomarkers Laboratory, Pathology Department, Faculty of Medical Sciences, Rio de Janeiro State University, Rio de Janeiro 20550-170, Brazil
| | - Eliane Luz
- Specialized Care Center for Orthopedic Oncology, National Institute of Traumatology and Orthopedics, Rio de Janeiro 20940-070, Brazil
| | - Rafael De Castro E Silva Pinheiro
- Specialized Care Center for Orthopedic Oncology, National Institute of Traumatology and Orthopedics, Rio de Janeiro 20940-070, Brazil
| | - Anabela Caruso
- Specialized Care Center for Orthopedic Oncology, National Institute of Traumatology and Orthopedics, Rio de Janeiro 20940-070, Brazil
| | - Ana Cristina De Sá Lopes
- Specialized Care Center for Orthopedic Oncology, National Institute of Traumatology and Orthopedics, Rio de Janeiro 20940-070, Brazil
| | - Walter Meohas
- Specialized Care Center for Orthopedic Oncology, National Institute of Traumatology and Orthopedics, Rio de Janeiro 20940-070, Brazil
| | - Gilda Alves
- Circulating Biomarkers Laboratory, Pathology Department, Faculty of Medical Sciences, Rio de Janeiro State University, Rio de Janeiro 20550-170, Brazil
| | - Maria Helena Faria Ornellas
- Circulating Biomarkers Laboratory, Pathology Department, Faculty of Medical Sciences, Rio de Janeiro State University, Rio de Janeiro 20550-170, Brazil
| |
Collapse
|
2
|
Taylor-Miller T, Savarirayan R. Progress in managing children with achondroplasia. Expert Rev Endocrinol Metab 2024; 19:479-486. [PMID: 39132812 DOI: 10.1080/17446651.2024.2390416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 08/06/2024] [Indexed: 08/13/2024]
Abstract
INTRODUCTION Achondroplasia is a heritable disorder of the skeleton that affects approximately 300,000 individuals worldwide. Until recently, treatment for this condition has been purely symptomatic. Efficacious treatment options for children are now approved or are in clinical trials. AREAS COVERED This review discusses key advances in the therapeutic management of children with achondroplasia, including vosoritide, the first approved drug, and other emerging precision therapies. These include navepegritide, a long-acting form of C-type natriuretic peptide, and infigratinib, a tyrosine kinase receptor inhibitor, summarizing trial outcomes to date. EXPERT OPINION The advent of the first approved precision therapy for achondroplasia in vosoritide has been a paradigm shifting advance for children affected by this condition. In addition to changing their natural growth history, it is hoped that it will decrease their medical complications and enhance functionality. These new treatment options highlight the importance of prompt prenatal identification and subsequent testing of a suspected fetus with achondroplasia and counseling of families. It is hoped that, in the near future, families will have the option to consider a range of effective targeted therapies that best suit their child with achondroplasia, starting from birth should they choose.
Collapse
Affiliation(s)
| | - Ravi Savarirayan
- Victorian Clinical Genetics Service, Melbourne, Parkville, Victoria, Australia
- Molecular Therapies, Murdoch Children's Research Institute, Parkville, Victoria, Australia
- Department of Paediatrics, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
3
|
Noeraparast M, Krajina K, Pichler R, Niedersüß‐Beke D, Shariat SF, Grünwald V, Ahyai S, Pichler M. FGFR3 alterations in bladder cancer: Sensitivity and resistance to targeted therapies. Cancer Commun (Lond) 2024; 44:1189-1208. [PMID: 39161208 PMCID: PMC11483561 DOI: 10.1002/cac2.12602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 07/24/2024] [Accepted: 08/06/2024] [Indexed: 08/21/2024] Open
Abstract
In this review, we revisit the pivotal role of fibroblast growth factor receptor 3 (FGFR3) in bladder cancer (BLCA), underscoring its prevalence in both non-muscle-invasive and muscle-invasive forms of the disease. FGFR3 mutations in up to half of BLCAs play a well-established role in tumorigenesis, shaping distinct tumor initiation patterns and impacting the tumor microenvironment (TME). Emphasizing the importance of considering epithelial-mesenchymal transition profile and TME status, we revisit their relevance in predicting responses to immune checkpoint inhibitors in FGFR3-mutated BLCAs. This writing highlights the initially promising yet transient efficacy of the FGFR inhibitor Erdafitinib on FGFR3-mutated BLCA, stressing the pressing need to unravel resistance mechanisms and identify co-targets for future combinatorial studies. A thorough analysis of recent preclinical and clinical evidence reveals resistance mechanisms, including secondary mutations, epigenetic alterations in pathway effectors, phenotypic heterogeneity, and population-specific variations within FGFR3 mutational status. Lastly, we discuss the potential of combinatorial treatments and concepts like synthetic lethality for discovering more effective targeted therapies against FGFR3-mutated BLCA.
Collapse
Affiliation(s)
- Maxim Noeraparast
- Translational OncologyII. Med Clinics Hematology and OncologyAugsburgGermany
| | - Katarina Krajina
- Translational OncologyII. Med Clinics Hematology and OncologyAugsburgGermany
| | - Renate Pichler
- Department of UrologyMedical University of InnsbruckInnsbruckAustria
| | | | | | - Viktor Grünwald
- Interdisciplinary Genitourinary OncologyClinic for Urology, Clinic for Medical OncologyUniversity Hospital Essen, Hufelandstraße 55EssenGermany
| | - Sascha Ahyai
- Department of UrologyMedical University of GrazGrazAustria
| | - Martin Pichler
- Translational OncologyII. Med Clinics Hematology and OncologyAugsburgGermany
| |
Collapse
|
4
|
Hudkins RL, Allen E, Balcer A, Hoffman ID, Iyer S, Neal M, Nelson KJ, Rideout M, Ye Q, Starrett JH, Patel P, Harris T, Swanson RV, Bensen DC. Discovery of TYRA-300: First Oral Selective FGFR3 Inhibitor for the Treatment of Urothelial Cancers and Achondroplasia. J Med Chem 2024; 67:16737-16756. [PMID: 39258897 DOI: 10.1021/acs.jmedchem.4c01531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
Activating FGFR3 alterations have been identified in up to 15-20% of muscle-invasive bladder cancer and metastatic urothelial carcinoma (mUC), and as high as 80% in nonmuscle invasive bladder cancers. FGFR3 germline mutations have also been associated with a variety of skeletal dysplasias. Achondroplasia, the most common form of dwarfism in humans, results from a G380R mutation in FGFR3. The pan-FGFR inhibitor erdafitinib was approved for the treatment of mUC with FGFR3 alterations but is limited due to FGFR isoform off-target toxicities and the development of on-target gatekeeper resistance mutations. TYRA-300 (22) was conceived using a structure-based approach as a potent FGFR3-selective inhibitor to avoid the toxicities associated with inhibition of FGFR1, FGFR2, and FGFR4, and to be agnostic for the FGFR3 gatekeeper mutations. TYRA-300 is being evaluated in a Phase 1 clinical trial in urothelial cancers and solid tumors, with intention to initiate Phase 2 studies in urothelial cancers and achondroplasia.
Collapse
Affiliation(s)
- Robert L Hudkins
- Tyra Biosciences, Inc., 2656 State Street, Carlsbad, California 92008, United States
| | - Eric Allen
- Tyra Biosciences, Inc., 2656 State Street, Carlsbad, California 92008, United States
| | - Alexandra Balcer
- Tyra Biosciences, Inc., 2656 State Street, Carlsbad, California 92008, United States
| | - Isaac D Hoffman
- Tyra Biosciences, Inc., 2656 State Street, Carlsbad, California 92008, United States
| | - Samhita Iyer
- Tyra Biosciences, Inc., 2656 State Street, Carlsbad, California 92008, United States
| | - Melissa Neal
- Tyra Biosciences, Inc., 2656 State Street, Carlsbad, California 92008, United States
| | - Kirk J Nelson
- Tyra Biosciences, Inc., 2656 State Street, Carlsbad, California 92008, United States
| | - Marc Rideout
- Tyra Biosciences, Inc., 2656 State Street, Carlsbad, California 92008, United States
| | - Qing Ye
- Tyra Biosciences, Inc., 2656 State Street, Carlsbad, California 92008, United States
| | - Jacqueline H Starrett
- Tyra Biosciences, Inc., 2656 State Street, Carlsbad, California 92008, United States
| | - Piyush Patel
- Tyra Biosciences, Inc., 2656 State Street, Carlsbad, California 92008, United States
| | - Todd Harris
- Tyra Biosciences, Inc., 2656 State Street, Carlsbad, California 92008, United States
| | - Ronald V Swanson
- Tyra Biosciences, Inc., 2656 State Street, Carlsbad, California 92008, United States
| | - Daniel C Bensen
- Tyra Biosciences, Inc., 2656 State Street, Carlsbad, California 92008, United States
| |
Collapse
|
5
|
Li X, Pan Y, Liu K, Yang Y, Ye Y, Xu Q, Fan M, Guo F. Identification and functional coordination analysis of gene co-expression networks in different tissues of XBP1 cartilage-specific deficient mice. Cell Signal 2024; 113:110929. [PMID: 37875231 DOI: 10.1016/j.cellsig.2023.110929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Revised: 09/21/2023] [Accepted: 10/14/2023] [Indexed: 10/26/2023]
Abstract
Abnormal differentiation and proliferation of chondrocytes leads to various diseases related to growth and development. The process of chondrocyte differentiation involves a series of complex cellular and molecular interactions. X-box binding protein 1 (XBP1), an essential molecule of the unfolded protein response (UPR) in Endoplasmic Reticulum (ER) stress, participated in cartilage development and causes other related diseases. We previously reported that XBP1 deficiency in cartilage impacts the function and associated diseases of many different tissues including cartilage. However, how differential expression of genes modulates the roles of cartilage and other tissues when XBP1 is lack of in chondrocytes remains unclear. We aimed to screen for differentially expressed (DE) genes in cartilage, brain, heart, and muscle by high-throughput sequencing in XBP1 cartilage-specific knockout (CKO) mice. Further, gene co-expression networks were constructed by weighted gene co-expression network analysis (WGCNA) algorithm and pivot genes were identified in the above four tissues. Protein detection, Hematoxylin-eosin (HE) staining and immunohistochemistry (IHC) experiments have proved that these differentially co-expressed genes participate in the downstream regulatory pathway of different tissues and affect tissue function.Significantly differentially expressed mRNAs [differentially expressed genes (DEGs)] were identified between XBP1 CKO mice and controls in cartilage, brain, heart, and muscle tissues, including 610, 126, 199 and 219 DEGs, respectively. 39 differentially co-expressed genes were identified in the above four tissues, and they were important pivot genes. Comprehensive analysis discovered that XBP1 deficiency in cartilage influences the difference of co-expressed genes between cartilage and other different tissues. These differentially co-expressed genes participate in downstream regulatory pathways of different tissues and affect tissue functions. Collectively, our conclusions may contribute potential biomarkers and molecular mechanisms for the mutual modulation between cartilage and different tissues and the diagnosis and treatment of diseases caused by abnormalities in different tissues. The analysis also provides meaningful insights for future genetic discoveries.
Collapse
Affiliation(s)
- Xiaoli Li
- Laboratory of Developmental Biology, Department of Cell Biology and Genetics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Yiming Pan
- Laboratory of Developmental Biology, Department of Cell Biology and Genetics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Kaiwen Liu
- Laboratory of Developmental Biology, Department of Cell Biology and Genetics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Yuyou Yang
- Laboratory of Developmental Biology, Department of Cell Biology and Genetics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Yuanlan Ye
- Laboratory of Developmental Biology, Department of Cell Biology and Genetics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Qingbo Xu
- School of Cardiovascular Medicine and Sciences, King's College London BHF Centre, London, United Kingdom
| | - Mengtian Fan
- Laboratory of Developmental Biology, Department of Cell Biology and Genetics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Fengjin Guo
- Laboratory of Developmental Biology, Department of Cell Biology and Genetics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
6
|
Loisay L, Komla-Ebri D, Morice A, Heuzé Y, Viaut C, de La Seiglière A, Kaci N, Chan D, Lamouroux A, Baujat G, Bassett JD, Williams GR, Legeai-Mallet L. Hypochondroplasia gain-of-function mutation in FGFR3 causes defective bone mineralization in mice. JCI Insight 2023; 8:e168796. [PMID: 37345656 PMCID: PMC10371252 DOI: 10.1172/jci.insight.168796] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 05/10/2023] [Indexed: 06/23/2023] Open
Abstract
Hypochondroplasia (HCH) is a mild dwarfism caused by missense mutations in fibroblast growth factor receptor 3 (FGFR3), with the majority of cases resulting from a heterozygous p.Asn540Lys gain-of-function mutation. Here, we report the generation and characterization of the first mouse model (Fgfr3Asn534Lys/+) of HCH to our knowledge. Fgfr3Asn534Lys/+ mice exhibited progressive dwarfism and impairment of the synchondroses of the cranial base, resulting in defective formation of the foramen magnum. The appendicular and axial skeletons were both severely affected and we demonstrated an important role of FGFR3 in regulation of cortical and trabecular bone structure. Trabecular bone mineral density (BMD) of long bones and vertebral bodies was decreased, but cortical BMD increased with age in both tibiae and femurs. These results demonstrate that bones in Fgfr3Asn534Lys/+ mice, due to FGFR3 activation, exhibit some characteristics of osteoporosis. The present findings emphasize the detrimental effect of gain-of-function mutations in the Fgfr3 gene on long bone modeling during both developmental and aging processes, with potential implications for the management of elderly patients with hypochondroplasia and osteoporosis.
Collapse
Affiliation(s)
- Léa Loisay
- Université de Paris Cité, Imagine Institute, Laboratory of Molecular and Physiopathological Bases of Osteochondrodysplasia, INSERM UMR1163, Paris, France
| | - Davide Komla-Ebri
- Molecular Endocrinology Laboratory, Department of Metabolism Digestion and Reproduction, Imperial College London, London, United Kingdom
- UCB Pharma, Slough, United Kingdom
| | - Anne Morice
- Université de Paris Cité, Imagine Institute, Laboratory of Molecular and Physiopathological Bases of Osteochondrodysplasia, INSERM UMR1163, Paris, France
| | - Yann Heuzé
- UMR5199 PACEA, CNRS, MC, Université de Bordeaux, Pessac, France
| | - Camille Viaut
- Université de Paris Cité, Imagine Institute, Laboratory of Molecular and Physiopathological Bases of Osteochondrodysplasia, INSERM UMR1163, Paris, France
| | - Amélie de La Seiglière
- Université de Paris Cité, Imagine Institute, Laboratory of Molecular and Physiopathological Bases of Osteochondrodysplasia, INSERM UMR1163, Paris, France
| | - Nabil Kaci
- Université de Paris Cité, Imagine Institute, Laboratory of Molecular and Physiopathological Bases of Osteochondrodysplasia, INSERM UMR1163, Paris, France
| | - Danny Chan
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Audrey Lamouroux
- Department of Medical Genetics, CHU Arnaud De Villeneuve, Montpellier, France
| | - Geneviève Baujat
- Université de Paris Cité, Imagine Institute, Laboratory of Molecular and Physiopathological Bases of Osteochondrodysplasia, INSERM UMR1163, Paris, France
- Department of Medical Genetics, French Reference Center for Skeletal Dysplasia, AP-HP, Necker Enfants Malades Hospital, Paris, France
| | - J.H. Duncan Bassett
- Molecular Endocrinology Laboratory, Department of Metabolism Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Graham R. Williams
- Molecular Endocrinology Laboratory, Department of Metabolism Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Laurence Legeai-Mallet
- Université de Paris Cité, Imagine Institute, Laboratory of Molecular and Physiopathological Bases of Osteochondrodysplasia, INSERM UMR1163, Paris, France
| |
Collapse
|
7
|
Luo F, Xie Y, Chen H, Huang J, Li C, Chen L, Yang J, Su N. Fgfr1 deficiency in osteocytes leads to increased bone mass by enhancing Wnt/β-catenin signaling. Bone 2023:116817. [PMID: 37268269 DOI: 10.1016/j.bone.2023.116817] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 05/23/2023] [Accepted: 05/26/2023] [Indexed: 06/04/2023]
Abstract
Osteoporosis (OP) is the most common skeletal disease in middle-aged and elderly people. A comprehensive understanding of the pathogenesis of osteoporosis is important. Fibroblast growth factor receptor 1 (FGFR1) is an important molecule for skeletal development and bone remodeling. Osteocytes are the most numerous cells in bone and play critical roles in bone homeostasis, however the effect of FGFR1 on osteocytes is still unclear. To clarify the direct effects of FGFR1 on osteocytes, we conditionally deleted Fgfr1 in osteocytes with Dentin matrix protein 1 (Dmp1)-Cre. We found that mice lacking Fgfr1 in osteocytes (Fgfr1f/f;Dmp-cre, MUT) showed increased trabecular bone mass at 2 and 6 months of age, which resulted from enhanced bone formation and decreased bone resorption. Furthermore, the cortical bone was thicker in WT mice than that in MUT mice at 2 and 6 months of age. Histological analysis showed that MUT mice had a decreased number of osteocytes but an increased number of osteocyte dendrites. We further found that mice lacking Fgfr1 in osteocytes showed enhanced activation of β-catenin signaling. The expression of sclerostin, an inhibitor of Wnt/β-catenin signaling, was obviously decreased in MUT mice. Furthermore, we found that FGFR1 can inhibit the expression of β-catenin and decrease the activity of β-catenin signaling. In brief, our study showed that FGFR1 in osteocytes can regulate bone mass by regulating Wnt/β-catenin signaling, providing genetic evidence that FGFR1 plays essential roles in osteocytes during bone remodeling and suggesting that FGFR1 is a potential therapeutic target for the prevention of bone loss.
Collapse
Affiliation(s)
- Fengtao Luo
- Center of Bone Metabolism and Repair, 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, PR China; Emergency Department of the 940th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou, Gansu, PR China
| | - Yangli Xie
- Center of Bone Metabolism and Repair, 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, PR China
| | - Hangang Chen
- Center of Bone Metabolism and Repair, 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, PR China
| | - Junlan Huang
- Center of Bone Metabolism and Repair, 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, PR China
| | - Can Li
- Center of Bone Metabolism and Repair, 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, PR China
| | - Lin Chen
- Center of Bone Metabolism and Repair, 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, PR China.
| | - Jing Yang
- Center of Bone Metabolism and Repair, 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, PR China.
| | - Nan Su
- Center of Bone Metabolism and Repair, 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, PR China.
| |
Collapse
|
8
|
Xu Q, Luo Y, Chao Z, Zhang J, Liu X, Tang Q, Wang K, Tan S, Fang M. Integrated Analysis of Transcriptome Expression Profiles Reveals miRNA-326-NKX3.2-Regulated Porcine Chondrocyte Differentiation. Int J Mol Sci 2023; 24:ijms24087257. [PMID: 37108419 PMCID: PMC10138716 DOI: 10.3390/ijms24087257] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 03/20/2023] [Accepted: 03/22/2023] [Indexed: 04/29/2023] Open
Abstract
The porcine body length trait is an essential factor affecting meat production and reproductive performance. It is evident that the development/lengthening of individual vertebrae is one of the main reasons for increases in body length; however, the underlying molecular mechanism remains unclear. In this study, RNA-seq analysis was used to profile the transcriptome (lncRNA, mRNA, and miRNA) of the thoracic intervertebral cartilage (TIC) at two time points (1 and 4 months) during vertebral column development in Yorkshire (Y) and Wuzhishan pigs (W). There were four groups: 1- (Y1) and 4-month-old (Y4) Yorkshire pigs and 1- (W1) and 4-month-old (W4) Wuzhishan pigs. In total, 161, 275, 86, and 126 differentially expressed (DE) lncRNAs, 1478, 2643, 404, and 750 DE genes (DEGs), and 74,51, 34, and 23 DE miRNAs (DE miRNAs) were identified in the Y4 vs. Y1, W4 vs. W1, Y4 vs. W4, and Y1 vs. W1 comparisons, respectively. Functional analysis of these DE transcripts (DETs) demonstrated that they had participated in various biological processes, such as cellular component organization or biogenesis, the developmental process, the metabolic process, bone development, and cartilage development. The crucial bone development-related candidate genes NK3 Homeobox 2 (NKX3.2), Wnt ligand secretion mediator (WLS), gremlin 1 (GREM1), fibroblast growth factor receptor 3 (FGFR3), hematopoietically expressed homeobox (HHEX), (collagen type XI alpha 1 chain (COL11A1), and Wnt Family Member 16 (WNT16)) were further identified by functional analysis. Moreover, lncRNA, miRNA, and gene interaction networks were constructed; a total of 55 lncRNAs, 6 miRNAs, and 7 genes formed lncRNA-gene, miRNA-gene, and lncRNA-miRNA-gene pairs, respectively. The aim was to demonstrate that coding and non-coding genes may co-regulate porcine spine development through interaction networks. NKX3.2 was identified as being specifically expressed in cartilage tissues, and it delayed chondrocyte differentiation. miRNA-326 regulated chondrocyte differentiation by targeting NKX3.2. The present study provides the first non-coding RNA and gene expression profiles in the porcine TIC, constructs the lncRNA-miRNA-gene interaction networks, and confirms the function of NKX3.2 in vertebral column development. These findings contribute to the understanding of the potential molecular mechanisms regulating pig vertebral column development. They expand our knowledge about the differences in body length between different pig species and provide a foundation for future studies.
Collapse
Affiliation(s)
- Qiao Xu
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, MOA Laboratory of Animal Genetics and Breeding, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Yabiao Luo
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, MOA Laboratory of Animal Genetics and Breeding, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Zhe Chao
- Institute of Animal Sciences and Veterinary, Hainan Academy of Agricultural Sciences, Haikou 571100, China
| | - Jibin Zhang
- Department of Molecular and Cellular Biology, Beckman Research Institute, City of Hope, Duarte, CA 91006, USA
| | - Ximing Liu
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, MOA Laboratory of Animal Genetics and Breeding, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Qiguo Tang
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, MOA Laboratory of Animal Genetics and Breeding, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Kejun Wang
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, MOA Laboratory of Animal Genetics and Breeding, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Shuyi Tan
- Institute of Animal Sciences and Veterinary, Hainan Academy of Agricultural Sciences, Haikou 571100, China
| | - Meiying Fang
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, MOA Laboratory of Animal Genetics and Breeding, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| |
Collapse
|
9
|
Kaur J, Singh A, Shah M, Chandrani P, Chougule A, Shetty O, Pai T, Menon S, Yadav S, Kapoor A, Mishra BK, Dutt A, Noronha V, Prabhash K. Erdafitinib for tumors with FGFR3 mutation: A promising targeted therapy. CANCER RESEARCH, STATISTICS, AND TREATMENT 2023; 6:288-295. [DOI: 10.4103/crst.crst_176_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 06/22/2023] [Indexed: 12/19/2024] Open
Affiliation(s)
- Jaspreet Kaur
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Dr. E Borges Road, Parel, Mumbai, Maharashtra, India
| | - Ajaykumar Singh
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Dr. E Borges Road, Parel, Mumbai, Maharashtra, India
| | - Minit Shah
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Dr. E Borges Road, Parel, Mumbai, Maharashtra, India
| | - Pratik Chandrani
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Dr. E Borges Road, Parel, Mumbai, Maharashtra, India
| | - Anuradha Chougule
- Department of Molecular Pathology, Tata Memorial Hospital, Homi Bhabha National Institute, Dr. E Borges Road, Parel, Mumbai, Maharashtra, India
| | - Omshree Shetty
- Department of Molecular Pathology, Tata Memorial Hospital, Homi Bhabha National Institute, Dr. E Borges Road, Parel, Mumbai, Maharashtra, India
| | - Trupti Pai
- Department of Molecular Pathology, Tata Memorial Hospital, Homi Bhabha National Institute, Dr. E Borges Road, Parel, Mumbai, Maharashtra, India
| | - Santosh Menon
- Department of Pathology, Tata Memorial Hospital, Homi Bhabha National Institute, Dr. E Borges Road, Parel, Mumbai, Maharashtra, India
| | - Subhash Yadav
- Department of Pathology, Tata Memorial Hospital, Homi Bhabha National Institute, Dr. E Borges Road, Parel, Mumbai, Maharashtra, India
| | - Akhil Kapoor
- Department of Medical Oncology, Mahamana Pandit Madan Mohan Malviya Cancer Centre and Homi Bhabha Cancer Hospital, Tata Memorial Centre, Varanasi, India
| | - B. K. Mishra
- Department of Medical Oncology, Mahamana Pandit Madan Mohan Malviya Cancer Centre and Homi Bhabha Cancer Hospital, Tata Memorial Centre, Varanasi, India
| | - Amit Dutt
- Department of Integrated Cancer Genomics, ACTREC, Tata Memorial Centre, Navi Mumbai, India
| | - Vanita Noronha
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Dr. E Borges Road, Parel, Mumbai, Maharashtra, India
| | - Kumar Prabhash
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Dr. E Borges Road, Parel, Mumbai, Maharashtra, India
| |
Collapse
|
10
|
Georgantzoglou N, Shen G, Jour G, Linos K. A case of FN1-fused calcified chondroid mesenchymal neoplasm of the hand with novel FGFR3 partner gene. Genes Chromosomes Cancer 2023; 62:237-241. [PMID: 36504176 DOI: 10.1002/gcc.23115] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 10/26/2022] [Accepted: 10/31/2022] [Indexed: 12/14/2022] Open
Abstract
Calcified chondroid neoplasms with FN1::FGFR1 or FGFR2 fusions constitute a recently described category of mesenchymal neoplasms mostly encountered in the extremities and temporomandibular joint. Herein, we report a case of FNI1-fused calcified chondroid neoplasm of the hand with a novel FGFR3 fusion partner. The tumor exhibited a multilobulated growth pattern composed of epithelioid cells embedded in abundant stroma with myxoid, chondroid, and fibrous areas and scattered osteoclast-like giant cells. RNA sequencing revealed an in-frame fusion between Exon 31 of FN1 and Exon 3 of FGFR3, which was subsequently confirmed by reverse transcription-polymerase chain reaction. Our findings expand on the spectrum of potential fusion partners in FN1-fused calcified chondroid neoplasms.
Collapse
Affiliation(s)
- Natalia Georgantzoglou
- Department of Pathology, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire, USA
| | - Guomiao Shen
- Department of Pathology, New York University, New York, New York, USA
| | - George Jour
- Department of Pathology, New York University, New York, New York, USA
| | - Konstantinos Linos
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| |
Collapse
|
11
|
Hua J, Huang J, Li G, Lin S, Cui L. Glucocorticoid induced bone disorders in children: Research progress in treatment mechanisms. Front Endocrinol (Lausanne) 2023; 14:1119427. [PMID: 37082116 PMCID: PMC10111257 DOI: 10.3389/fendo.2023.1119427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 03/20/2023] [Indexed: 04/22/2023] Open
Abstract
Long-term or supra-physiological dose of glucocorticoid (GC) application in clinic can lead to impaired bone growth and osteoporosis. The side effects of GC on the skeletal system are particularly serious in growing children, potentially causing growth retardation or even osteoporotic fractures. Children's bone growth is dependent on endochondral ossification of growth plate chondrocytes, and excessive GC can hinder the development of growth plate and longitudinal bone growth. Despite the availability of drugs for treating osteoporosis, they have failed to effectively prevent or treat longitudinal bone growth and development disorders caused by GCs. As of now, there is no specific drug to mitigate these severe side effects. Traditional Chinese Medicine shows potential as an alternative to the current treatments by eliminating the side effects of GC. In summary, this article comprehensively reviews the research frontiers concerning growth and development disorders resulting from supra-physiological levels of GC and discusses the future research and treatment directions for optimizing steroid therapy. This article may also provide theoretical and experimental insight into the research and development of novel drugs to prevent GC-related side effects.
Collapse
Affiliation(s)
- Junying Hua
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Zhanjiang, China
| | - Jianping Huang
- Department of Prosthodontics, Yonsei University College of Dentistry, Seoul, Republic of Korea
| | - Gang Li
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Sien Lin
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Orthopaedic Center, Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, China
- *Correspondence: Liao Cui, ; Sien Lin,
| | - Liao Cui
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Zhanjiang, China
- *Correspondence: Liao Cui, ; Sien Lin,
| |
Collapse
|
12
|
Natesan K, Srivalli T, Mohan H, Jayaprakash A, Ramalingam V. UPLC-ESI-Q-TOF-MS E-based metabolomics analysis of Acer mono sap and evaluation of osteogenic activity in mouse osteoblast cells. Food Funct 2022; 13:13002-13013. [PMID: 36449013 DOI: 10.1039/d2fo01948e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Investigation of phytochemicals and bioactive molecules is tremendously vital for the applications of new plant resources in chemistry, food, and medicine. In this study, the chemical profiling of sap of Acer mono (SAM), a Korean syrup known for its anti-osteoporosis effect, was performed using UPLC-ESI-Q-TOF-MSE analysis. A total of 23 compounds were identified based on the mass and fragmentation characteristics and most of the compounds have significant biomedical applications. The in vitro antioxidant assessment of SAM indicated excellent activity by scavenging DPPH and ABTS-free radicals and were found to be 23.35 mg mL-1 and 29.33 mg mL-1, respectively, as IC50 concentrations. As well, the in vitro proliferation effect of the SAM was assessed against mouse MC3T3-E1 cells, and the results showed that the SAM enhanced the proliferation of the cells, and 12.5 mg mL-1 and 25 mg mL-1 of SAM were selected for osteogenic differentiation. The morphological analysis clearly evidenced the SAM enhanced the osteogenic activity in MC3T3-E1 cells by the increased deposition of extracellular calcium and nodule formation. Moreover, the qRT-PCR analysis confirmed the increased expression of osteoblast marker gene expression including ALP, osteocalcin, osteopontin, collagen1α1, Runx2, and osterix in SAM-treated MC3T3-E1 cells. Together, these results suggest that SAM possesses osteogenic effects and can be used for bone regeneration and bone loss-associated diseases such as osteoporosis.
Collapse
Affiliation(s)
- Karthi Natesan
- School of Allied Health Sciences, REVA University, Bengaluru, India
| | - Thimmarayan Srivalli
- PG and Research Department of Biochemistry, Scared Heart College (Autonomous), Tirupattur - 635601, Tamil Nadu, India (Affiliated to Thiruvalluvar University, Serkkadu, Vellore - 632115, Tamil Nadu, India)
| | - Harshavardhan Mohan
- Department of Chemistry, Research Institute of Physics and Chemistry, Jeonbuk National University, Jeonju 54896, Republic of Korea
| | - Arul Jayaprakash
- PG and Research Department of Biochemistry, Scared Heart College (Autonomous), Tirupattur - 635601, Tamil Nadu, India (Affiliated to Thiruvalluvar University, Serkkadu, Vellore - 632115, Tamil Nadu, India)
| | - Vaikundamoorthy Ramalingam
- Centre for Natural Products & Traditional Knowledge, CSIR-Indian Institute of Chemical Technology, Hyderabad, India. .,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad - 201002, India
| |
Collapse
|
13
|
Ekinci S, Ülger Y, Acar MO, Ceran A, Aycan Z, Fitoz ÖS, Ilgın Ruhi H. Clinical and radiologic evaluation of a Turkish family with hypochondroplasia and a rare FGFR3 variant. J Pediatr Endocrinol Metab 2022; 35:1097-1101. [PMID: 35438268 DOI: 10.1515/jpem-2021-0773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 03/28/2022] [Indexed: 11/15/2022]
Abstract
OBJECTIVES Hypochondroplasia (HCH) is characterized by disproportionate short stature and regarded as a milder form of achondroplasia (ACH), which is another skeletal dysplasia, both caused by variants in fibroblast growth factor receptor 3 (FGFR3) gene. HCH diagnosis is based on the clinical features and skeletal survey findings. The most common FGFR3 variant in HCH affects the codon 540, leading to substitution of asparagine with lysine in about 70% of patients. CASE PRESENTATION Herein, we described the clinical and radiographical manifestations of HCH in affected members of a Turkish family with very rare Asn540Thr (c.1619A>C) variant within hot spot of the gene for this condition. CONCLUSIONS This is a very rarely reported variant in the literature and this report is the first case with this variant in Turkish population. The report also presents the phenotypic variability within a family with the same variant, which is inherent to HCH.
Collapse
Affiliation(s)
- Sadiye Ekinci
- Medical Genetics Department, Ankara University School of Medicine, Ankara, Turkey
| | - Yasemin Ülger
- Medical Genetics Department, Ankara University School of Medicine, Ankara, Turkey
| | - Mustafa Oğuz Acar
- Medical Genetics Department, Ankara University School of Medicine, Ankara, Turkey
| | - Ayşegül Ceran
- Pediatric Endocrinology Department, Ankara University School of Medicine, Ankara, Turkey
| | - Zehra Aycan
- Pediatric Endocrinology Department, Ankara University School of Medicine, Ankara, Turkey
| | - Ömer Suat Fitoz
- Radiology Department, Ankara University School of Medicine, Ankara, Turkey
| | - Hatice Ilgın Ruhi
- Medical Genetics Department, Ankara University School of Medicine, Ankara, Turkey
| |
Collapse
|
14
|
The Role of the Key Differentially Mutated Gene FGFR3 in the Immune Microenvironment of Bladder Cancer. J Immunol Res 2022; 2022:7952706. [PMID: 35991125 PMCID: PMC9391163 DOI: 10.1155/2022/7952706] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/17/2022] [Accepted: 07/29/2022] [Indexed: 11/17/2022] Open
Abstract
The tumor microenvironment (TME) has been a major focus of research in recent years as a crucial factor in the development and progression of bladder cancer. Unfortunately, the precise composition of TME, particularly the immunological and stromal components, remains unknown. In this work, we downloaded the RNA-seq expression profiles and somatic mutation data of 433 bladder cancer cases from The Cancer Genome Atlas (TCGA) and then employed a comprehensive bioinformatics approach to evaluate them. Firstly, the expression profiles were used to predict the scores and then the content of immune and stromal cells via the estimate package in R software. We then identified differentially expressed genes (DEGs) and differentially mutated genes (DMGs) according to the high-stromal score cohort and low-stromal score cohort. Finally, fibroblast growth factor receptor 3 (FGFR3) was the main differentially mutated gene in bladder carcinoma that we discovered after conducting a cross-study on DEGs and DMGs. Follow-up investigation revealed that FGFR3, whose expression correlated inversely with cancer progression stage, appeared to be a protective factor in bladder cancer. The method of Gene Set Enrichment Analysis (GSEA) was employed to, respectively, interpret the expression data of FGFR3 in high and low expression lists. We observed that the genes in the low FGFR3 expression list were strongly enriched in the biological processes associated with transplantation and cell adhesion, suggesting the possible role of FGFR3 in predicting TME metastasis status in bladder cancer. Therefore, this study is aimed at investigating whether FGFR3 is promising as a biomarker of TME remodeling to explain underlying mechanisms involved in tumorigenesis and metastasis, which may help to make decisions on treatments for bladder cancer.
Collapse
|
15
|
Ornitz DM, Itoh N. New developments in the biology of fibroblast growth factors. WIREs Mech Dis 2022; 14:e1549. [PMID: 35142107 PMCID: PMC10115509 DOI: 10.1002/wsbm.1549] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 11/08/2021] [Accepted: 11/09/2021] [Indexed: 01/28/2023]
Abstract
The fibroblast growth factor (FGF) family is composed of 18 secreted signaling proteins consisting of canonical FGFs and endocrine FGFs that activate four receptor tyrosine kinases (FGFRs 1-4) and four intracellular proteins (intracellular FGFs or iFGFs) that primarily function to regulate the activity of voltage-gated sodium channels and other molecules. The canonical FGFs, endocrine FGFs, and iFGFs have been reviewed extensively by us and others. In this review, we briefly summarize past reviews and then focus on new developments in the FGF field since our last review in 2015. Some of the highlights in the past 6 years include the use of optogenetic tools, viral vectors, and inducible transgenes to experimentally modulate FGF signaling, the clinical use of small molecule FGFR inhibitors, an expanded understanding of endocrine FGF signaling, functions for FGF signaling in stem cell pluripotency and differentiation, roles for FGF signaling in tissue homeostasis and regeneration, a continuing elaboration of mechanisms of FGF signaling in development, and an expanding appreciation of roles for FGF signaling in neuropsychiatric diseases. This article is categorized under: Cardiovascular Diseases > Molecular and Cellular Physiology Neurological Diseases > Molecular and Cellular Physiology Congenital Diseases > Stem Cells and Development Cancer > Stem Cells and Development.
Collapse
Affiliation(s)
- David M Ornitz
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Nobuyuki Itoh
- Kyoto University Graduate School of Pharmaceutical Sciences, Sakyo, Kyoto, Japan
| |
Collapse
|
16
|
Luo X, Li J, Xiao C, Sun L, Xiang W, Chen N, Lei C, Lei H, Long Y, Long T, Suolang Q, Yi K. Whole-Genome Resequencing of Xiangxi Cattle Identifies Genomic Diversity and Selection Signatures. Front Genet 2022; 13:816379. [PMID: 35711927 PMCID: PMC9196905 DOI: 10.3389/fgene.2022.816379] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 04/13/2022] [Indexed: 01/11/2023] Open
Abstract
Understanding the genetic diversity in Xiangxi cattle may facilitate our efforts toward further breeding programs. Here we compared 23 Xiangxi cattle with 78 published genomes of 6 worldwide representative breeds to characterize the genomic variations of Xiangxi cattle. Based on clustering models in population structure analysis, we displayed that Xiangxi cattle had a mutual genome ancestor with Chinese indicine, Indian indicine, and East Asian taurine. Population genetic diversity was analyzed by four methods (nucleotide diversity, inbreeding coefficient, linkage disequilibrium decay and runs of homozygosity), and we found that Xiangxi cattle had higher genomic diversity and weaker artificial selection than commercial breed cattle. Using four testing methods (θπ, CLR, FST, and XP-EHH), we explored positive selection regions harboring genes in Xiangxi cattle, which were related to reproduction, growth, meat quality, heat tolerance, and immune response. Our findings revealed the extent of sequence variation in Xiangxi cattle at the genome-wide level. All of our fruitful results can bring about a valuable genomic resource for genetic studies and breed protection in the future.
Collapse
Affiliation(s)
- Xiaoyu Luo
- Hunan Institute of Animal and Veterinary Science, Changsha, China.,Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Xianyang, China
| | - Jianbo Li
- Hunan Institute of Animal and Veterinary Science, Changsha, China.,Xiangxi Cattle Engineering Technology Center of Hunan Province, Huayuan, China
| | - Chentong Xiao
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Xianyang, China
| | - Luyang Sun
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Xianyang, China
| | - Weixuan Xiang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Xianyang, China.,School of Life Science, University of Bristol, Bristol, United Kingdom
| | - Ningbo Chen
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Xianyang, China
| | - Chuzhao Lei
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Xianyang, China
| | - Hong Lei
- Hunan Institute of Animal and Veterinary Science, Changsha, China.,Xiangxi Cattle Engineering Technology Center of Hunan Province, Huayuan, China
| | - Yun Long
- Xiangxi Cattle Engineering Technology Center of Hunan Province, Huayuan, China.,Hunan De Nong Animal Husbandry Group Co. Ltd., Huayuan, China
| | - Ting Long
- Xiangxi Cattle Engineering Technology Center of Hunan Province, Huayuan, China.,Hunan De Nong Animal Husbandry Group Co. Ltd., Huayuan, China
| | - Quji Suolang
- Institute of Animal Science, Tibet Academy of Agricultural and Animal Husbandry Science, Lhasa, China
| | - Kangle Yi
- Hunan Institute of Animal and Veterinary Science, Changsha, China.,Xiangxi Cattle Engineering Technology Center of Hunan Province, Huayuan, China
| |
Collapse
|
17
|
Periosteum and development of the tissue-engineered periosteum for guided bone regeneration. J Orthop Translat 2022; 33:41-54. [PMID: 35228996 PMCID: PMC8858911 DOI: 10.1016/j.jot.2022.01.002] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 01/02/2022] [Accepted: 01/17/2022] [Indexed: 12/11/2022] Open
Abstract
Background Periosteum plays a significant role in bone formation and regeneration by storing progenitor cells, and also acts as a source of local growth factors and a scaffold for recruiting cells and other growth factors. Recently, tissue-engineered periosteum has been studied extensively and shown to be important for osteogenesis and chondrogenesis. Using biomimetic methods for artificial periosteum synthesis, membranous tissues with similar function and structure to native periosteum are produced that significantly improve the efficacy of bone grafting and scaffold engineering, and can serve as direct replacements for native periosteum. Many problems involving bone defects can be solved by preparation of idealized periosteum from materials with different properties using various techniques. Methods This review summarizes the significance of periosteum for osteogenesis and chondrogenesis from the aspects of periosteum tissue structure, osteogenesis performance, clinical application, and development of periosteum tissue engineering. The advantages and disadvantages of different tissue engineering methods are also summarized. Results The fast-developing field of periosteum tissue engineering is aimed toward synthesis of bionic periosteum that can ensure or accelerate the repair of bone defects. Artificial periosteum materials can be similar to natural periosteum in both structure and function, and have good therapeutic potential. Induction of periosteum tissue regeneration and bone regeneration by biomimetic periosteum is the ideal process for bone repair. Conclusions Periosteum is essential for bone formation and regeneration, and it is indispensable in bone repair. Achieving personalized structure and composition in the construction of tissue engineering periosteum is in accordance with the design concept of both universality and emphasis on individual differences and ensures the combination of commonness and individuality, which are expected to meet the clinical needs of bone repair more effectively. The translational potential of this article To better understand the role of periosteum in bone repair, clarify the present research situation of periosteum and tissue engineering periosteum, and determine the development and optimization direction of tissue engineering periosteum in the future. It is hoped that periosteum tissue engineering will play a greater role in meeting the clinical needs of bone repair in the future, and makes it possible to achieve optimization of bone tissue therapy.
Collapse
|
18
|
Melatonin and the Programming of Stem Cells. Int J Mol Sci 2022; 23:ijms23041971. [PMID: 35216086 PMCID: PMC8879213 DOI: 10.3390/ijms23041971] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 02/03/2022] [Accepted: 02/08/2022] [Indexed: 02/06/2023] Open
Abstract
Melatonin interacts with various types of stem cells, in multiple ways that comprise stimulation of proliferation, maintenance of stemness and self-renewal, protection of survival, and programming toward functionally different cell lineages. These various properties are frequently intertwined but may not be always jointly present. Melatonin typically stimulates proliferation and transition to the mature cell type. For all sufficiently studied stem or progenitor cells, melatonin’s signaling pathways leading to expression of respective morphogenetic factors are discussed. The focus of this article will be laid on the aspect of programming, particularly in pluripotent cells. This is especially but not exclusively the case in neural stem cells (NSCs) and mesenchymal stem cells (MSCs). Concerning developmental bifurcations, decisions are not exclusively made by melatonin alone. In MSCs, melatonin promotes adipogenesis in a Wnt (Wingless-Integration-1)-independent mode, but chondrogenesis and osteogenesis Wnt-dependently. Melatonin upregulates Wnt, but not in the adipogenic lineage. This decision seems to depend on microenvironment and epigenetic memory. The decision for chondrogenesis instead of osteogenesis, both being Wnt-dependent, seems to involve fibroblast growth factor receptor 3. Stem cell-specific differences in melatonin and Wnt receptors, and contributions of transcription factors and noncoding RNAs are outlined, as well as possibilities and the medical importance of re-programming for transdifferentiation.
Collapse
|
19
|
The Evolution of Biomineralization through the Co-Option of Organic Scaffold Forming Networks. Cells 2022; 11:cells11040595. [PMID: 35203246 PMCID: PMC8870065 DOI: 10.3390/cells11040595] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 02/07/2022] [Accepted: 02/08/2022] [Indexed: 12/05/2022] Open
Abstract
Biomineralization is the process in which organisms use minerals to generate hard structures like teeth, skeletons and shells. Biomineralization is proposed to have evolved independently in different phyla through the co-option of pre-existing developmental programs. Comparing the gene regulatory networks (GRNs) that drive biomineralization in different species could illuminate the molecular evolution of biomineralization. Skeletogenesis in the sea urchin embryo was extensively studied and the underlying GRN shows high conservation within echinoderms, larval and adult skeletogenesis. The organic scaffold in which the calcite skeletal elements form in echinoderms is a tubular compartment generated by the syncytial skeletogenic cells. This is strictly different than the organic cartilaginous scaffold that vertebrates mineralize with hydroxyapatite to make their bones. Here I compare the GRNs that drive biomineralization and tubulogenesis in echinoderms and in vertebrates. The GRN that drives skeletogenesis in the sea urchin embryo shows little similarity to the GRN that drives bone formation and high resemblance to the GRN that drives vertebrates’ vascular tubulogenesis. On the other hand, vertebrates’ bone-GRNs show high similarity to the GRNs that operate in the cells that generate the cartilage-like tissues of basal chordate and invertebrates that do not produce mineralized tissue. These comparisons suggest that biomineralization in deuterostomes evolved through the phylum specific co-option of GRNs that control distinct organic scaffolds to mineralization.
Collapse
|
20
|
Takemoto G, Matsushita M, Okamoto T, Ito T, Matsuura Y, Takashima C, Chen-Yoshikawa TF, Ebi H, Imagama S, Kitoh H, Ohno K, Hosono Y. Meclozine Attenuates the MARK Pathway in Mammalian Chondrocytes and Ameliorates FGF2-Induced Bone Hyperossification in Larval Zebrafish. Front Cell Dev Biol 2022; 9:694018. [PMID: 35118060 PMCID: PMC8804316 DOI: 10.3389/fcell.2021.694018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 12/20/2021] [Indexed: 11/13/2022] Open
Abstract
Meclozine has been developed as an inhibitor of fibroblast growth factor receptor 3 (FGFR3) to treat achondroplasia (ACH). Extracellular signal regulated kinase (ERK) phosphorylation was attenuated by meclozine in FGF2-treated chondrocyte cell line, but the site of its action has not been elucidated. Although orally administered meclozine promoted longitudinal bone growth in a mouse model of ACH, its effect on craniofacial bone development during the early stage remains unknown. Herein, RNA-sequencing analysis was performed using murine chondrocytes from FGF2-treated cultured tibiae, which was significantly elongated by meclozine treatment. Gene set enrichment analysis demonstrated that FGF2 significantly increased the enrichment score of mitogen-activated protein kinase (MAPK) family signaling cascades in chondrocytes; however, meclozine reduced this enrichment. Next, we administered meclozine to FGF2-treated larval zebrafish from 8 h post-fertilization (hpf). We observed that FGF2 significantly increased the number of ossified vertebrae in larval zebrafish at 7 days post-fertilization (dpf), while meclozine delayed vertebral ossification in FGF2-induced zebrafish. Meclozine also reversed the FGF2-induced upregulation of ossified craniofacial bone area, including ceratohyal, hyomandibular, and quadrate. The current study provided additional evidence regarding the inhibitory effect of meclozine on the FGF2-induced upregulation of MAPK signaling in chondrocytes and FGF2-induced development of craniofacial and vertebral bones.
Collapse
Affiliation(s)
- Genta Takemoto
- Department of Orthopaedic Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masaki Matsushita
- Department of Orthopaedic Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
- *Correspondence: Masaki Matsushita, ; Yasuyuki Hosono,
| | - Takaaki Okamoto
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Toshinari Ito
- Division of Molecular Therapeutics, Aichi Cancer Center Research Institute, Nagoya, Japan
- Department of Thoracic Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yuki Matsuura
- Division of Molecular Therapeutics, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Chieko Takashima
- Division of Molecular Therapeutics, Aichi Cancer Center Research Institute, Nagoya, Japan
| | | | - Hiromichi Ebi
- Division of Molecular Therapeutics, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Shiro Imagama
- Department of Orthopaedic Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroshi Kitoh
- Department of Orthopaedic Surgery, Aichi Children’s Health and Medical Center, Obu, Japan
- Department of Comprehensive Pediatric Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kinji Ohno
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yasuyuki Hosono
- Division of Molecular Therapeutics, Aichi Cancer Center Research Institute, Nagoya, Japan
- Department of Pharmacology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
- *Correspondence: Masaki Matsushita, ; Yasuyuki Hosono,
| |
Collapse
|
21
|
Wells KM, Baumel M, McCusker CD. The Regulation of Growth in Developing, Homeostatic, and Regenerating Tetrapod Limbs: A Minireview. Front Cell Dev Biol 2022; 9:768505. [PMID: 35047496 PMCID: PMC8763381 DOI: 10.3389/fcell.2021.768505] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 11/19/2021] [Indexed: 01/29/2023] Open
Abstract
The size and shape of the tetrapod limb play central roles in their functionality and the overall physiology of the organism. In this minireview we will discuss observations on mutant animal models and humans, which show that the growth and final size of the limb is most impacted by factors that regulate either limb bud patterning or the elongation of the long bones. We will also apply the lessons that have been learned from embryos to how growth could be regulated in regenerating limb structures and outline the challenges that are unique to regenerating animals.
Collapse
|
22
|
Li X, Yang Y, Liang L, Fan M, Li X, Feng N, Pan Y, Tan Q, Xu Q, Xie Y, Guo F. Effect Of XBP1 Deficiency In Cartilage On The Regulatory Network Of LncRNA/circRNA-miRNA-mRNA. Int J Biol Sci 2022; 18:315-330. [PMID: 34975335 PMCID: PMC8692151 DOI: 10.7150/ijbs.64054] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Accepted: 10/29/2021] [Indexed: 12/13/2022] Open
Abstract
X-box binding protein 1(XBP1) is a critical component for unfolded protein response (UPR) in ER stress. According to previous studies performed with different XBP1-deficient mice, the XBP1 gene affects mouse cartilage development and causes other related diseases. However, how the complete transcriptome, including mRNA and ncRNAs, affects the function of cartilage and other tissues when XBP1 is deficient in chondrocytes is unclear. In this study, we aimed to screen the differentially expressed (DE) mRNAs, circRNAs, lncRNAs and miRNAs in XBP1 cartilage-specific knockout (CKO) mice using high throughput sequencing and construct the circRNA-miRNA-mRNA and lncRNA-miRNA-mRNA regulatory networks. DE LncRNAs (DE-LncRNAs), circRNAs (DE-circRNAs), miRNAs (DE-miRNAs), and mRNAs [differentially expressed genes (DEGs)] between the cartilage tissue of XBP1 CKO mice and controls were identified, including 441 DE-LncRNAs, 15 DE-circRNAs, 6 DE-miRNAs, and 477 DEGs. Further, 253,235 lncRNA-miRNA-mRNA networks and 1,822 circRNA-miRNA-mRNA networks were constructed based on the correlation between lncRNAs/circRNAs, miRNAs, mRNAs. The whole transcriptome analysis revealed that XBP1 deficiency in cartilage affects the function of cartilage and other different tissues, as well as associated diseases. Overall, our findings may provide potential biomarkers and mechanisms for the diagnosis and treatment of cartilage and other related diseases.
Collapse
Affiliation(s)
- Xiaoli Li
- Department of Cell Biology and Genetics, Core Facility of Development Biology, Chongqing Medical University, Chongqing 400016, China
| | - Yuyou Yang
- Department of Cell Biology and Genetics, Core Facility of Development Biology, Chongqing Medical University, Chongqing 400016, China
| | - Li Liang
- Department of Cell Biology and Genetics, Core Facility of Development Biology, Chongqing Medical University, Chongqing 400016, China
| | - Mengtian Fan
- Department of Cell Biology and Genetics, Core Facility of Development Biology, Chongqing Medical University, Chongqing 400016, China
| | - Xingyue Li
- Department of Cell Biology and Genetics, Core Facility of Development Biology, Chongqing Medical University, Chongqing 400016, China
| | - Naibo Feng
- Department of Cell Biology and Genetics, Core Facility of Development Biology, Chongqing Medical University, Chongqing 400016, China
| | - Yiming Pan
- Department of Cell Biology and Genetics, Core Facility of Development Biology, Chongqing Medical University, Chongqing 400016, 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 400042, China
| | - Qingbo Xu
- Cardiovascular Division, King's College London BHF Centre, London, United Kingdom
| | - 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 400042, China
| | - Fengjin Guo
- Department of Cell Biology and Genetics, Core Facility of Development Biology, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
23
|
Wells KM, Kelley K, Baumel M, Vieira WA, McCusker CD. Neural control of growth and size in the axolotl limb regenerate. eLife 2021; 10:68584. [PMID: 34779399 PMCID: PMC8716110 DOI: 10.7554/elife.68584] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 11/13/2021] [Indexed: 11/29/2022] Open
Abstract
The mechanisms that regulate growth and size of the regenerating limb in tetrapods such as the Mexican axolotl are unknown. Upon the completion of the developmental stages of regeneration, when the regenerative organ known as the blastema completes patterning and differentiation, the limb regenerate is proportionally small in size. It then undergoes a phase of regeneration that we have called the ‘tiny-limb’ stage, which is defined by rapid growth until the regenerate reaches the proportionally appropriate size. In the current study we have characterized this growth and have found that signaling from the limb nerves is required for its maintenance. Using the regenerative assay known as the accessory limb model (ALM), we have found that growth and size of the limb positively correlates with nerve abundance. We have additionally developed a new regenerative assay called the neural modified-ALM (NM-ALM), which decouples the source of the nerves from the regenerating host environment. Using the NM-ALM we discovered that non-neural extrinsic factors from differently sized host animals do not play a prominent role in determining the size of the regenerating limb. We have also discovered that the regulation of limb size is not autonomously regulated by the limb nerves. Together, these observations show that the limb nerves provide essential cues to regulate ontogenetic allometric growth and the final size of the regenerating limb. Humans’ ability to regrow lost or damaged body parts is relatively limited, but some animals, such as the axolotl (a Mexican salamander), can regenerate complex body parts, like legs, many times over their lives. Studying regeneration in these animals could help researchers enhance humans’ abilities to heal. One way to do this is using the Accessory Limb Model (ALM), where scientists wound an axolotl’s leg, and study the additional leg that grows from the wound. The first stage of limb regeneration creates a new leg that has the right structure and shape. The new leg is very small so the next phase involves growing the leg until its size matches the rest of the animal. This phase must be controlled so that the limb stops growing when it reaches the right size, but how this regulation works is unclear. Previous research suggests that the number of nerves in the new leg could be important. Wells et al. used a ALM to study how the size of regenerating limbs is controlled. They found that changing the number of nerves connected to the new leg altered its size, with more nerves leading to a larger leg. Next, Wells et al. created a system that used transplanted nerve bundles of different sizes to grow new legs in different sized axolotls. This showed that the size of the resulting leg is controlled by the number of nerves connecting it to the CNS. Wells et al. also showed that nerves can only control regeneration if they remain connected to the central nervous system. These results explain how size is controlled during limb regeneration in axolotls, highlighting the fact that regrowth is directly controlled by the number of nerves connected to a regenerating leg. Much more work is needed to reveal the details of this process and the signals nerves use to control growth. It will also be important to determine whether this control system is exclusive to axolotls, or whether other animals also use it.
Collapse
Affiliation(s)
- Kaylee M Wells
- Biology Department, University of Massachusetts Boston, Boston, United States
| | - Kristina Kelley
- Biology Department, University of Massachusetts Boston, Boston, United States
| | - Mary Baumel
- Biology Department, University of Massachusetts Boston, Boston, United States
| | - Warren A Vieira
- Biology Department, University of Massachusetts Boston, Boston, United States
| | | |
Collapse
|
24
|
Jiang A, Xu P, Sun S, Zhao Z, Tan Q, Li W, Song C, Leng H. Cellular alterations and crosstalk in the osteochondral joint in osteoarthritis and promising therapeutic strategies. Connect Tissue Res 2021; 62:709-719. [PMID: 33397157 DOI: 10.1080/03008207.2020.1870969] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 12/28/2020] [Indexed: 02/03/2023]
Abstract
Osteoarthritis (OA) is a joint disorder involving cartilage degeneration and subchondral bone sclerosis. The bone-cartilage interface is implicated in OA pathogenesis due to its susceptibility to mechanical and biological factors. The crosstalk between cartilage and the underlying subchondral bone is elevated in OA due to multiple factors, such as increased vascularization, porosity, microcracks and fissures. Changes in the osteochondral joint are traceable to alterations in chondrocytes and bone cells (osteoblasts, osteocytes and osteoclasts). The phenotypes of these cells can change with the progression of OA. Aberrant intercellular communications among bone cell-bone cell and bone cell-chondrocyte are of great importance and might be the factors promoting OA development. An appreciation of cellular phenotypic changes in OA and the mechanisms by which these cells communicate would be expected to lead to the development of targeted drugs with fewer side effects.
Collapse
Affiliation(s)
- Ai Jiang
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| | - Peng Xu
- State Key Laboratory of Systematic and Evolutionary Botany, Institute of Botany, Chinese Academy of Sciences, Beijing, China
| | - Shang Sun
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| | - Zhenda Zhao
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| | - Qizhao Tan
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| | - Weishi Li
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education Lisbon Portugal
| | - Chunli Song
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
- Beijing Key Lab of Spine Diseases, Beijing, China
| | - Huijie Leng
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| |
Collapse
|
25
|
Razmara E, Bitaraf A, Karimi B, Babashah S. Functions of the SNAI family in chondrocyte-to-osteocyte development. Ann N Y Acad Sci 2021; 1503:5-22. [PMID: 34403146 DOI: 10.1111/nyas.14668] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/22/2021] [Accepted: 07/02/2021] [Indexed: 12/12/2022]
Abstract
Different cellular mechanisms contribute to osteocyte development. And while critical roles for members of the zinc finger protein SNAI family (SNAIs) have been discussed in cancer-related models, there are few reviews summarizing their importance for chondrocyte-to-osteocyte development. To help fill this gap, we review the roles of SNAIs in the development of mature osteocytes from chondrocytes, including the regulation of chondro- and osteogenesis through different signaling pathways and in programmed cell death. We also discuss how epigenetic factors-including DNA methylation, histone methylation and acetylation, and noncoding RNAs-contribute differently to both chondrocyte and osteocyte development. To better grasp the important roles of SNAIs in bone development, we also review genotype-phenotype correlations in different animal models. We end with comments about the possible importance of the SNAI family in cartilage/bone development and the potential applications for therapeutic goals.
Collapse
Affiliation(s)
- Ehsan Razmara
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - Amirreza Bitaraf
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Behnaz Karimi
- Hematology/Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Science, Tehran, Iran
| | - Sadegh Babashah
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
26
|
Kimura T, Bosakova M, Nonaka Y, Hruba E, Yasuda K, Futakawa S, Kubota T, Fafilek B, Gregor T, Abraham SP, Gomolkova R, Belaskova S, Pesl M, Csukasi F, Duran I, Fujiwara M, Kavkova M, Zikmund T, Kaiser J, Buchtova M, Krakow D, Nakamura Y, Ozono K, Krejci P. An RNA aptamer restores defective bone growth in FGFR3-related skeletal dysplasia in mice. Sci Transl Med 2021; 13:13/592/eaba4226. [PMID: 33952673 DOI: 10.1126/scitranslmed.aba4226] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 12/30/2020] [Accepted: 04/16/2021] [Indexed: 01/04/2023]
Abstract
Achondroplasia is the most prevalent genetic form of dwarfism in humans and is caused by activating mutations in FGFR3 tyrosine kinase. The clinical need for a safe and effective inhibitor of FGFR3 is unmet, leaving achondroplasia currently incurable. Here, we evaluated RBM-007, an RNA aptamer previously developed to neutralize the FGFR3 ligand FGF2, for its activity against FGFR3. In cultured rat chondrocytes or mouse embryonal tibia organ culture, RBM-007 rescued the proliferation arrest, degradation of cartilaginous extracellular matrix, premature senescence, and impaired hypertrophic differentiation induced by FGFR3 signaling. In cartilage xenografts derived from induced pluripotent stem cells from individuals with achondroplasia, RBM-007 rescued impaired chondrocyte differentiation and maturation. When delivered by subcutaneous injection, RBM-007 restored defective skeletal growth in a mouse model of achondroplasia. We thus demonstrate a ligand-trap concept of targeting the cartilage FGFR3 and delineate a potential therapeutic approach for achondroplasia and other FGFR3-related skeletal dysplasias.
Collapse
Affiliation(s)
- Takeshi Kimura
- Department of Pediatrics, Osaka University Graduate School of Medicine, 565-0871 Osaka, Japan
| | - Michaela Bosakova
- 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.,Institute of Animal Physiology and Genetics, Czech Academy of Sciences, 60200 Brno, Czech Republic
| | | | - Eva Hruba
- Institute of Animal Physiology and Genetics, Czech Academy of Sciences, 60200 Brno, Czech Republic
| | - Kie Yasuda
- Department of Pediatrics, Osaka University Graduate School of Medicine, 565-0871 Osaka, Japan
| | | | - Takuo Kubota
- Department of Pediatrics, Osaka University Graduate School of Medicine, 565-0871 Osaka, Japan
| | - Bohumil Fafilek
- 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.,Institute of Animal Physiology and Genetics, Czech Academy of Sciences, 60200 Brno, Czech Republic
| | - Tomas Gregor
- 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
| | - Sara P Abraham
- Department of Biology, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic
| | - Regina Gomolkova
- Department of Biology, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic.,Institute of Animal Physiology and Genetics, Czech Academy of Sciences, 60200 Brno, Czech Republic
| | - Silvie Belaskova
- International Clinical Research Center, St. Anne's University Hospital, 65691 Brno, Czech Republic
| | - Martin Pesl
- 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.,First Department of Internal Medicine-Cardioangiology, St. Anne's University Hospital, Masaryk University, 65691 Brno, Czech Republic
| | - Fabiana Csukasi
- Department of Orthopaedic Surgery, University of California Los Angeles, Los Angeles, CA 90095, USA.,Networking Research Center on Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN)-LABRET, University of Málaga, IBIMA-BIONAND, 29071 Málaga, Spain
| | - Ivan Duran
- Department of Orthopaedic Surgery, University of California Los Angeles, Los Angeles, CA 90095, USA.,Networking Research Center on Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN)-LABRET, University of Málaga, IBIMA-BIONAND, 29071 Málaga, Spain
| | | | - Michaela Kavkova
- Central European Institute of Technology, Brno University of Technology, 61200 Brno, Czech Republic
| | - Tomas Zikmund
- Central European Institute of Technology, Brno University of Technology, 61200 Brno, Czech Republic
| | - Josef Kaiser
- Central European Institute of Technology, Brno University of Technology, 61200 Brno, Czech Republic
| | - Marcela Buchtova
- Institute of Animal Physiology and Genetics, Czech Academy of Sciences, 60200 Brno, Czech Republic.,Department of Experimental Biology, Faculty of Science, Masaryk University, 62500 Brno, Czech Republic
| | - Deborah Krakow
- Department of Orthopaedic Surgery, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Yoshikazu Nakamura
- RIBOMIC Inc., Tokyo 108-0071, Japan. .,Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | - Keiichi Ozono
- Department of Pediatrics, Osaka University Graduate School of Medicine, 565-0871 Osaka, Japan.
| | - 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.,Institute of Animal Physiology and Genetics, Czech Academy of Sciences, 60200 Brno, Czech Republic
| |
Collapse
|
27
|
Insights of fibroblast growth factor receptor 3 aberrations in pan-cancer and their roles in potential clinical treatment. Aging (Albany NY) 2021; 13:16541-16566. [PMID: 34160364 PMCID: PMC8266346 DOI: 10.18632/aging.203175] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 06/02/2021] [Indexed: 02/05/2023]
Abstract
Fibroblast growth factor receptor 3 (FGFR3) alters frequently across various cancer types and is a common therapeutic target in bladder urothelial carcinoma (BLCA) with FGFR3 variants. Although emerging evidence supports the role of FGFR3 in individual cancer types, no pan-cancer analysis is available. In this work, we used the open comprehensive datasets, covering a total of 10,953 patients with 10,967 samples across 32 TCGA cancer types, to identify the full alteration spectrum of FGFR3. FGFR3 abnormal expression, methylation patterns, alteration frequency, mutation location distribution, functional impact, and prognostic implications differed greatly from cancer to cancer. The overall alteration frequency of FGFR3 was relatively low in all cancers. Targetable mutations were mainly detected in BLCA, and S249C, Y373C, G370C, and R248C were hotspot mutations that could be targeted by an FDA approved erdafitinib. Genetic fusions were mainly observed in glioma, followed by BLCA. FGFR3-TACC3 was the most common fusion type which was proposed as novel therapeutic targets in glioma and was targetable with erdafitinib in BLCA. Lung adenocarcinoma (LUAD) and lung squamous cell carcinoma (LUSC) were two lung cancer subtypes, FGFR3 fusion and hotspot mutation like S249C were observed more commonly in LUSC but not in LUAD. DNA methylation was correlated with the expression of FGFR3 and its downstream genes in some tumors. FGFG3 abnormal expression and alterations exhibited clinical correlations with patient prognosis in several tumors. This work exhibited the full alteration spectrum of FGFR3 and indicated several new clues for their application as potential therapeutic targets and prognostic indicators.
Collapse
|
28
|
Wrobel W, Pach E, Ben-Skowronek I. Advantages and Disadvantages of Different Treatment Methods in Achondroplasia: A Review. Int J Mol Sci 2021; 22:ijms22115573. [PMID: 34070375 PMCID: PMC8197470 DOI: 10.3390/ijms22115573] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 05/20/2021] [Accepted: 05/22/2021] [Indexed: 02/06/2023] Open
Abstract
Achondroplasia (ACH) is a disease caused by a missense mutation in the FGFR3 (fibroblast growth factor receptor 3) gene, which is the most common cause of short stature in humans. The treatment of ACH is necessary and urgent because untreated achondroplasia has many complications, both orthopedic and neurological, which ultimately lead to disability. This review presents the current and potential pharmacological treatments for achondroplasia, highlighting the advantages and disadvantages of all the drugs that have been demonstrated in human and animal studies in different stages of clinical trials. The article includes the potential impacts of drugs on achondroplasia symptoms other than short stature, including their effects on spinal canal stenosis, the narrowing of the foramen magnum and the proportionality of body structure. Addressing these effects could significantly improve the quality of life of patients, possibly reducing the frequency and necessity of hospitalization and painful surgical procedures, which are currently the only therapeutic options used. The criteria for a good drug for achondroplasia are best met by recombinant human growth hormone at present and will potentially be met by vosoritide in the future, while the rest of the drugs are in the early stages of clinical trials.
Collapse
|
29
|
Biosse Duplan M, Dambroise E, Estibals V, Veziers J, Guicheux J, Legeai-Mallet L. An Fgfr3-activating mutation in immature murine osteoblasts affects the appendicular and craniofacial skeleton. Dis Model Mech 2021; 14:dmm048272. [PMID: 33737326 PMCID: PMC8084574 DOI: 10.1242/dmm.048272] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 03/03/2021] [Indexed: 12/17/2022] Open
Abstract
Achondroplasia (ACH), the most common form of dwarfism, is caused by a missense mutation in the gene coding for fibroblast growth factor receptor 3 (FGFR3). The resulting increase in FGFR3 signaling perturbs the proliferation and differentiation of chondrocytes (CCs), alters the process of endochondral ossification and thus reduces bone elongation. Increased FGFR3 signaling in osteoblasts (OBs) might also contribute to bone anomalies in ACH. In the present study of a mouse model of ACH, we sought to determine whether FGFR3 overactivation in OBs leads to bone modifications. The model carries an Fgfr3-activating mutation (Fgfr3Y367C/+) that accurately mimics ACH; we targeted the mutation to either immature OBs and hypertrophic CCs or to mature OBs by using the Osx-cre and collagen 1α1 (2.3 kb Col1a1)-cre mouse strains, respectively. We observed that Fgfr3 activation in immature OBs and hypertrophic CCs (Osx-Fgfr3) not only perturbed the hypertrophic cells of the growth plate (thus affecting long bone growth) but also led to osteopenia and low cortical thickness in long bones in adult (3-month-old) mice but not growing (3-week-old) mice. Importantly, craniofacial membranous bone defects were present in the adult mice. In contrast, activation of Fgfr3 in mature OBs (Col1-Fgfr3) had very limited effects on skeletal shape, size and micro-architecture. In vitro, we observed that Fgfr3 activation in immature OBs was associated with low mineralization activity. In conclusion, immature OBs appear to be affected by Fgfr3 overactivation, which might contribute to the bone modifications observed in ACH independently of CCs.
Collapse
Affiliation(s)
- Martin Biosse Duplan
- Laboratory of Molecular and Physiopathological Bases of Osteochondrodysplasia, INSERM UMR 1163, Imagine Institute, Paris 75015, France
- Université de Paris, Paris 75006, France
- Service de Médecine Bucco-Dentaire, Hôpital Bretonneau, AP-HP, Paris 75018, France
| | - Emilie Dambroise
- Laboratory of Molecular and Physiopathological Bases of Osteochondrodysplasia, INSERM UMR 1163, Imagine Institute, Paris 75015, France
- Université de Paris, Paris 75006, France
| | - Valentin Estibals
- Laboratory of Molecular and Physiopathological Bases of Osteochondrodysplasia, INSERM UMR 1163, Imagine Institute, Paris 75015, France
- Université de Paris, Paris 75006, France
| | - Joelle Veziers
- Inserm, UMR 1229, RMeS – Regenerative Medicine and Skeleton, Université de Nantes, ONIRIS, Nantes, F-44042, France
- SC3M, SFR Santé F. Bonamy, FED 4203, UMS Inserm 016, CNRS 3556, Nantes F-44042, France
- CHU Nantes, PHU4 OTONN, Nantes, F-44093, France
| | - Jérome Guicheux
- Inserm, UMR 1229, RMeS – Regenerative Medicine and Skeleton, Université de Nantes, ONIRIS, Nantes, F-44042, France
- SC3M, SFR Santé F. Bonamy, FED 4203, UMS Inserm 016, CNRS 3556, Nantes F-44042, France
- CHU Nantes, PHU4 OTONN, Nantes, F-44093, France
| | - Laurence Legeai-Mallet
- Laboratory of Molecular and Physiopathological Bases of Osteochondrodysplasia, INSERM UMR 1163, Imagine Institute, Paris 75015, France
- Université de Paris, Paris 75006, France
| |
Collapse
|
30
|
Mou TC, Feng JY. Research advances in cartilage stem cells markers and induced differentiation. HUA XI KOU QIANG YI XUE ZA ZHI = HUAXI KOUQIANG YIXUE ZAZHI = WEST CHINA JOURNAL OF STOMATOLOGY 2021; 39:108-114. [PMID: 33723946 DOI: 10.7518/hxkq.2021.01.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Cartilage stem cells (CSCs) are cells that self-proliferate, have surface antigen expression, and have multidirectional differentiation potential in the articular cartilage. CSCs, as an ideal source of stem cells, has a good application prospect in stem cell therapy. This article reviews the CSCs markers, cartilage differentiation signaling pathway, and clinical treatment of osteoarthritis.
Collapse
Affiliation(s)
- Ting-Chen Mou
- Dept. of Stomatological, Taizhou Central Hospital (Taizhou University Hospital), Taizhou 318000, China
| | - Jian-Ying Feng
- College of Stomatology, Zhejiang Chinese Medical University, Hangzhou 310053, China
| |
Collapse
|
31
|
Zhang Y, Wang X, Huang X, Shen L, Zhang L, Shou D, Fan X. Transcriptome sequencing profiling identifies miRNA-331-3p as an osteoblast-specific miRNA in infected bone nonunion. Bone 2021; 143:115619. [PMID: 32858253 DOI: 10.1016/j.bone.2020.115619] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 08/20/2020] [Accepted: 08/24/2020] [Indexed: 11/25/2022]
Abstract
Bone nonunion caused by bacterial infection accounts for bone fractures, bone trauma and bone transplantation surgeries. Severe consequences include delayed unions and amputation and result in functional limitations, work disability, and poor quality of life. However, the mechanism of bone nonunion remains unknown. In this study, we aimed to screen the miRNA biomarkers of bacterial bone infection and investigated whether miRNAs regulate the osteoblasts and thus contribute to bone nonunion. We established a miRNA-mRNA network based on high-throughput RNA sequencing to compare the model rabbits infected with Staphylococcus aureus with the control rabbits. After validation experiments, miRNA-331-3p and fibroblast growth factor 23 (FGF23) were found to be inversely correlated with the pathways of osteoblast mineralization and pathology of infected bone nonunion. In in vitro experiments, miRNA-331-3p was downregulated and FGF23 was upregulated in lipopolysaccharide (LPS)-induced mouse calvarial osteoblasts. Further studies of the mechanism showed that mutated of putative miRNA-331-3p can bind to FGF23 3'-untranslated region sites. MiRNA-331-3p acted as an osteoblast mineralization promoter by directly targeting FGF23. Downregulation of miRNA-331-3p led to inhibition of osteoblast mineralization by regulating the DKK1/β-catenin mediated signaling. Thus, we established an improved animal model and identified new bone-related biomarkers in the infected bone nonunion. The miRNA-331-3p biomarker was demonstrated to regulate osteoblast mineralization by targeting FGF23. The novel mechanism can be used as potential diagnostic biomarkers and therapeutic targets in the infected bone nonunion and other inflammatory bone disorders.
Collapse
Affiliation(s)
- Yang Zhang
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; Department of Medicine, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou 310007, China
| | - Xuping Wang
- Department of Medicine, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou 310007, China
| | - Xiaowen Huang
- Department of Medicine, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou 310007, China
| | - Lifeng Shen
- Department of Orthopaedic Surgery, Zhejiang Provincial Tongde Hospital, Hangzhou 310012, China
| | - Li Zhang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Dan Shou
- Department of Medicine, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou 310007, China.
| | - Xiaohui Fan
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
32
|
Kacew A, Sweis RF. FGFR3 Alterations in the Era of Immunotherapy for Urothelial Bladder Cancer. Front Immunol 2020; 11:575258. [PMID: 33224141 PMCID: PMC7674585 DOI: 10.3389/fimmu.2020.575258] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 10/14/2020] [Indexed: 12/15/2022] Open
Abstract
FGFR3 is a prognostic and predictive marker and is a validated therapeutic target in urothelial bladder cancer. Its utility as a marker and target in the context of immunotherapy is incompletely understood. We review the role of FGFR3 in bladder cancer and discuss preclinical and clinical clues of its effectiveness as a patient selection factor and therapeutic target in the era of immunotherapy.
Collapse
Affiliation(s)
- Alec Kacew
- Section of Hematology and Oncology, Department of Medicine, The University of Chicago, Chicago, IL, United States
| | - Randy F Sweis
- Section of Hematology and Oncology, Department of Medicine, The University of Chicago, Chicago, IL, United States.,Committee on Immunology, The University of Chicago, Chicago, IL, United States.,Comprehensive Cancer Center, The University of Chicago, Chicago, IL, United States
| |
Collapse
|
33
|
Actomyosin and the MRTF-SRF pathway downregulate FGFR1 in mesenchymal stromal cells. Commun Biol 2020; 3:576. [PMID: 33067523 PMCID: PMC7567845 DOI: 10.1038/s42003-020-01309-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 08/25/2020] [Indexed: 12/14/2022] Open
Abstract
Both biological and mechanical signals are known to influence cell proliferation. However, biological signals are mostly studied in two-dimensions (2D) and the interplay between these different pathways is largely unstudied. Here, we investigated the influence of the cell culture environment on the response to bFGF, a widely studied and important proliferation growth factor. We observed that human mesenchymal stromal cells (hMSCs), but not fibroblasts, lose the ability to respond to soluble or covalently bound bFGF when cultured on microfibrillar substrates. This behavior correlated with a downregulation of FGF receptor 1 (FGFR1) expression of hMSCs on microfibrillar substrates. Inhibition of actomyosin or the MRTF/SRF pathway decreased FGFR1 expression in hMSCs, fibroblasts and MG63 cells. To our knowledge, this is the first time FGFR1 expression is shown to be regulated through a mechanosensitive pathway in hMSCs. These results add to the sparse literature on FGFR1 regulation and potentially aid designing tissue engineering constructs that better control cell proliferation.
Collapse
|
34
|
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: 410] [Impact Index Per Article: 82.0] [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
|
35
|
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
|
36
|
Chijimatsu R, Saito T. Mechanisms of synovial joint and articular cartilage development. Cell Mol Life Sci 2019; 76:3939-3952. [PMID: 31201464 PMCID: PMC11105481 DOI: 10.1007/s00018-019-03191-5] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 05/30/2019] [Accepted: 06/11/2019] [Indexed: 12/29/2022]
Abstract
Articular cartilage is formed at the end of epiphyses in the synovial joint cavity and permanently contributes to the smooth movement of synovial joints. Most skeletal elements develop from transient cartilage by a biological process known as endochondral ossification. Accumulating evidence indicates that articular and growth plate cartilage are derived from different cell sources and that different molecules and signaling pathways regulate these two kinds of cartilage. As the first sign of joint development, the interzone emerges at the presumptive joint site within a pre-cartilage tissue. After that, joint cavitation occurs in the center of the interzone, and the cells in the interzone and its surroundings gradually form articular cartilage and the synovial joint. During joint development, the interzone cells continuously migrate out to the epiphyseal cartilage and the surrounding cells influx into the joint region. These complicated phenomena are regulated by various molecules and signaling pathways, including GDF5, Wnt, IHH, PTHrP, BMP, TGF-β, and FGF. Here, we summarize current literature and discuss the molecular mechanisms underlying joint formation and articular development.
Collapse
Affiliation(s)
- Ryota Chijimatsu
- Bone and Cartilage Regenerative Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Taku Saito
- Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.
| |
Collapse
|
37
|
Abstract
PURPOSE OF REVIEW The goal of the review is to provide a comprehensive overview of the current understanding of the mechanisms underlying variation in human stature. RECENT FINDINGS Human height is an anthropometric trait that varies considerably within human populations as well as across the globe. Historically, much research focus was placed on understanding the biology of growth plate chondrocytes and how modifications to core chondrocyte proliferation and differentiation pathways potentially shaped height attainment in normal as well as pathological contexts. Recently, much progress has been made to improve our understanding regarding the mechanisms underlying the normal and pathological range of height variation within as well as between human populations, and today, it is understood to reflect complex interactions among a myriad of genetic, environmental, and evolutionary factors. Indeed, recent improvements in genetics (e.g., GWAS) and breakthroughs in functional genomics (e.g., whole exome sequencing, DNA methylation analysis, ATAC-sequencing, and CRISPR) have shed light on previously unknown pathways/mechanisms governing pathological and common height variation. Additionally, the use of an evolutionary perspective has also revealed important mechanisms that have shaped height variation across the planet. This review provides an overview of the current knowledge of the biological mechanisms underlying height variation by highlighting new research findings on skeletal growth control with an emphasis on previously unknown pathways/mechanisms influencing pathological and common height variation. In this context, this review also discusses how evolutionary forces likely shaped the genomic architecture of height across the globe.
Collapse
Affiliation(s)
| | - Terence D Capellini
- Department of Human Evolutionary Biology, Harvard University, Cambridge, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
38
|
Wang J, Liu S, Li J, Yi Z. The role of the fibroblast growth factor family in bone-related diseases. Chem Biol Drug Des 2019; 94:1740-1749. [PMID: 31260189 DOI: 10.1111/cbdd.13588] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Revised: 04/25/2019] [Accepted: 06/17/2019] [Indexed: 12/16/2022]
Abstract
Fibroblast growth factor (FGF) family members are important regulators of cell growth, proliferation, differentiation, and regeneration. The abnormal expression of certain FGF family members can cause skeletal diseases, including achondroplasia, craniosynostosis syndrome, osteoarthritis, and Kashin-Beck disease. Accumulating evidence shows that FGFs play a crucial role in the growth and proliferation of bone and in the pathogenesis of certain bone-related diseases. Here, we review the involvement of FGFs in bone-related processes and diseases; FGF1 in the differentiation of human bone marrow mesenchymal stem cells and fracture repair; FGF2, FGF9, and FGF18 in osteoarthritis; FGF6 in bone and muscle injury; FGF8 in osteoarthritis and Kashin-Beck disease; and FGF21 and FGF23 on bone regulation. These findings indicate that FGFs are targets for novel therapeutic interventions for bone-related diseases.
Collapse
Affiliation(s)
- Jicheng Wang
- Department of Orthopaedics, Shaanxi Provincial People's Hospital, Xi'an, China.,Xi'an Medical University, Xi'an, China
| | - Shizhang Liu
- Department of Orthopaedics, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Jingyuan Li
- Department of Orthopaedics, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Zhi Yi
- Department of Orthopaedics, Shaanxi Provincial People's Hospital, Xi'an, China
| |
Collapse
|
39
|
Saw S, Aiken A, Fang H, McKee TD, Bregant S, Sanchez O, Chen Y, Weiss A, Dickson BC, Czarny B, Sinha A, Fosang A, Dive V, Waterhouse PD, Kislinger T, Khokha R. Metalloprotease inhibitor TIMP proteins control FGF-2 bioavailability and regulate skeletal growth. J Cell Biol 2019; 218:3134-3152. [PMID: 31371388 PMCID: PMC6719459 DOI: 10.1083/jcb.201906059] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 07/10/2019] [Accepted: 07/15/2019] [Indexed: 12/19/2022] Open
Abstract
Saw et al. show via the combinatorial deletion of Timp family members in mice that metalloprotease regulation of FGF-2 is a crucial event in the chondrocyte maturation program, underlying the growth plate development and bone elongation responsible for attaining proper body stature. Regulated growth plate activity is essential for postnatal bone development and body stature, yet the systems regulating epiphyseal fusion are poorly understood. Here, we show that the tissue inhibitors of metalloprotease (TIMP) gene family is essential for normal bone growth after birth. Whole-body quadruple-knockout mice lacking all four TIMPs have growth plate closure in long bones, precipitating limb shortening, epiphyseal distortion, and widespread chondrodysplasia. We identify TIMP/FGF-2/IHH as a novel nexus underlying bone lengthening where TIMPs negatively regulate the release of FGF-2 from chondrocytes to allow IHH expression. Using a knock-in approach that combines MMP-resistant or ADAMTS-resistant aggrecans with TIMP deficiency, we uncouple growth plate activity in axial and appendicular bones. Thus, natural metalloprotease inhibitors are crucial regulators of chondrocyte maturation program, growth plate integrity, and skeletal proportionality. Furthermore, individual and combinatorial TIMP-deficient mice demonstrate the redundancy of metalloprotease inhibitor function in embryonic and postnatal development.
Collapse
Affiliation(s)
- Sanjay Saw
- Princess Margaret Cancer Centre/Ontario Cancer Institute, University Health Network, Toronto, Canada
| | - Alison Aiken
- Princess Margaret Cancer Centre/Ontario Cancer Institute, University Health Network, Toronto, Canada
| | - Hui Fang
- Princess Margaret Cancer Centre/Ontario Cancer Institute, University Health Network, Toronto, Canada
| | - Trevor D McKee
- Princess Margaret Cancer Centre/Ontario Cancer Institute, University Health Network, Toronto, Canada
| | | | - Otto Sanchez
- University of Ontario Institute of Technology, Oshawa, Canada
| | - Yan Chen
- Princess Margaret Cancer Centre/Ontario Cancer Institute, University Health Network, Toronto, Canada
| | - Ashley Weiss
- Princess Margaret Cancer Centre/Ontario Cancer Institute, University Health Network, Toronto, Canada
| | | | | | - Ankit Sinha
- Princess Margaret Cancer Centre/Ontario Cancer Institute, University Health Network, Toronto, Canada
| | - Amanda Fosang
- University of Melbourne Department of Paediatrics and Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
| | - Vincent Dive
- Institute of Biology and Technology, Saclay, France
| | - Paul D Waterhouse
- Princess Margaret Cancer Centre/Ontario Cancer Institute, University Health Network, Toronto, Canada
| | - Thomas Kislinger
- Princess Margaret Cancer Centre/Ontario Cancer Institute, University Health Network, Toronto, Canada
| | - Rama Khokha
- Princess Margaret Cancer Centre/Ontario Cancer Institute, University Health Network, Toronto, Canada
| |
Collapse
|
40
|
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
|
41
|
Han X, Ross J, Kolumam G, Pi M, Sonoda J, King G, Quarles LD. Cardiovascular Effects of Renal Distal Tubule Deletion of the FGF Receptor 1 Gene. J Am Soc Nephrol 2018; 29:69-80. [PMID: 28993502 PMCID: PMC5748915 DOI: 10.1681/asn.2017040412] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 08/03/2017] [Indexed: 01/11/2023] Open
Abstract
The bone-derived hormone fibroblast growth factor-23 (FGF-23) activates complexes composed of FGF receptors (FGFRs), including FGFR1, and α-Klotho in the kidney distal tubule (DT), leading to increased sodium retention and hypertension. However, the role of FGFR1 in regulating renal processes linked to hypertension is unclear. Here, we investigated the effects of selective FGFR1 loss in the DT. Conditional knockout (cKO) of FGFR1 in the DT (FGFR1DT-cKO mice) resulted in left ventricular hypertrophy (LVH) and decreased kidney expression of α-Klotho in association with enhanced BP, decreased expression of angiotensin converting enzyme 2, and increased expression of the Na+-K+-2Cl- cotransporter. Notably, recombinant FGF-23 administration similarly decreased the kidney expression of α-Klotho and induced LVH in mice. Pharmacologic activation of FGFR1 with a monoclonal anti-FGFR1 antibody (R1MAb1) normalized BP and significantly attenuated LVH in the Hyp mouse model of excess FGF-23, but did not induce a response in FGFR1DT-cKO mice. The hearts of FGFR1DT-cKO mice showed increased expression of the transient receptor potential cation channel, subfamily C, member 6 (TRPC6), consistent with cardiac effects of soluble Klotho deficiency. Moreover, administration of recombinant soluble Klotho lowered BP in the Hyp mice. Thus, FGFR1 in the DT regulates systemic hemodynamic responses opposite to those predicted by the actions of FGF-23. These cardiovascular effects appear to be mediated by paracrine FGF control of kidney FGFR1 and subsequent regulation of soluble Klotho and TRPC6. FGFR1 in the kidney may provide a new molecular target for treating hypertension.
Collapse
MESH Headings
- Angiotensin-Converting Enzyme 2
- Animals
- Antibodies, Monoclonal/pharmacology
- Blood Pressure/drug effects
- Blood Pressure/genetics
- Female
- Fibroblast Growth Factor-23
- Fibroblast Growth Factors/pharmacology
- Glucuronidase/genetics
- Glucuronidase/metabolism
- Hypertension/genetics
- Hypertrophy, Left Ventricular/genetics
- Immunologic Factors/pharmacology
- Kidney Tubules, Distal
- Klotho Proteins
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Myocardium/metabolism
- Peptidyl-Dipeptidase A/genetics
- Peptidyl-Dipeptidase A/metabolism
- RNA, Messenger/metabolism
- Receptor, Fibroblast Growth Factor, Type 1/genetics
- Receptor, Fibroblast Growth Factor, Type 1/immunology
- Recombinant Proteins/pharmacology
- Sodium-Potassium-Chloride Symporters/genetics
- Sodium-Potassium-Chloride Symporters/metabolism
- TRPC Cation Channels/genetics
- TRPC Cation Channels/metabolism
- TRPC6 Cation Channel
Collapse
Affiliation(s)
- Xiaobin Han
- Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Jed Ross
- Department of Molecular Biology and Biomedical Imaging, Genentech, South San Francisco, California; and
| | - Ganesh Kolumam
- Department of Molecular Biology and Biomedical Imaging, Genentech, South San Francisco, California; and
| | - Min Pi
- Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Junichiro Sonoda
- Department of Molecular Biology and Biomedical Imaging, Genentech, South San Francisco, California; and
| | - Gwendalyn King
- Department of Neurobiology, University of Alabama in Birmingham, Birmingham, Alabama
| | - L Darryl Quarles
- Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee;
| |
Collapse
|
42
|
Xu W, Luo F, Wang Q, Tan Q, Huang J, Zhou S, Wang Z, Sun X, Kuang L, Jin M, Su N, Jiang W, Chen L, Qi H, Zhu Y, Chen B, Chen H, Chen S, Gao Y, Xu X, Deng C, Chen L, Xie Y, Du X. Inducible Activation of FGFR2 in Adult Mice Promotes Bone Formation After Bone Marrow Ablation. J Bone Miner Res 2017. [PMID: 28650109 DOI: 10.1002/jbmr.3204] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Apert syndrome is one of the most severe craniosynostoses, resulting from gain-of-function mutations in fibroblast growth factor receptor 2 (FGFR2). Previous studies have shown that gain-of-function mutations of FGFR2 (S252W or P253R) cause skull malformation of human Apert syndrome by affecting both chondrogenesis and osteogenesis, underscoring the key role of FGFR2 in bone development. However, the effects of FGFR2 on bone formation at the adult stage have not been fully investigated. To investigate the role of FGFR2 in bone formation, we generated mice with tamoxifen-inducible expression of mutant FGFR2 (P253R) at the adult stage. Mechanical bone marrow ablation (BMX) was performed in both wild-type and Fgfr2 mutant (MT) mice. Changes in newly formed trabecular bone were assessed by micro-computed tomography and bone histomorphometry. We found that MT mice exhibited increased trabecular bone formation and decreased bone resorption after BMX accompanied with a remarkable increase in bone marrow stromal cell recruitment and proliferation, osteoblast proliferation and differentiation, and enhanced Wnt/β-catenin activity. Furthermore, pharmacologically inhibiting Wnt/β-catenin signaling can partially reverse the increased trabecular bone formation and decreased bone resorption in MT mice after BMX. Our data demonstrate that gain-of-function mutation in FGFR2 exerts a Wnt/β-catenin-dependent anabolic effect on trabecular bone by promoting bone formation and inhibiting bone resorption at the adult stage. © 2017 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- 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, 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, China
| | - Quan 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, 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, 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, China
| | - Siru Zhou
- 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, 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, 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, China
| | - Liang Kuang
- 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, China
| | - Min Jin
- 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, China
| | - Nan Su
- 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, 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, China
| | - Liang 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, China
| | - Huabing Qi
- 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, China
| | - Ying Zhu
- 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, 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, China
| | - Hangang 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, China
| | - Shuai 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, China
| | - Yu Gao
- 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, China
| | - Xiaoling Xu
- Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Chuxia Deng
- Faculty of Health Sciences, University of Macau, Macau SAR, 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, China
| | - 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, 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, China
| |
Collapse
|
43
|
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
|
44
|
Sarabipour S. Parallels and Distinctions in FGFR, VEGFR, and EGFR Mechanisms of Transmembrane Signaling. Biochemistry 2017. [DOI: 10.1021/acs.biochem.7b00399] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Sarvenaz Sarabipour
- Institute for Computational
Medicine and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States
| |
Collapse
|
45
|
Ornitz DM, Legeai-Mallet L. Achondroplasia: Development, pathogenesis, and therapy. Dev Dyn 2017; 246:291-309. [PMID: 27987249 DOI: 10.1002/dvdy.24479] [Citation(s) in RCA: 142] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 12/04/2016] [Accepted: 12/05/2016] [Indexed: 12/11/2022] Open
Abstract
Autosomal dominant mutations in fibroblast growth factor receptor 3 (FGFR3) cause achondroplasia (Ach), the most common form of dwarfism in humans, and related chondrodysplasia syndromes that include hypochondroplasia (Hch), severe achondroplasia with developmental delay and acanthosis nigricans (SADDAN), and thanatophoric dysplasia (TD). FGFR3 is expressed in chondrocytes and mature osteoblasts where it functions to regulate bone growth. Analysis of the mutations in FGFR3 revealed increased signaling through a combination of mechanisms that include stabilization of the receptor, enhanced dimerization, and enhanced tyrosine kinase activity. Paradoxically, increased FGFR3 signaling profoundly suppresses proliferation and maturation of growth plate chondrocytes resulting in decreased growth plate size, reduced trabecular bone volume, and resulting decreased bone elongation. In this review, we discuss the molecular mechanisms that regulate growth plate chondrocytes, the pathogenesis of Ach, and therapeutic approaches that are being evaluated to improve endochondral bone growth in people with Ach and related conditions. Developmental Dynamics 246:291-309, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- David M Ornitz
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Laurence Legeai-Mallet
- Imagine Institute, Inserm U1163, Université Paris Descartes, Service de Génétique, Hôpital Necker-Enfants Malades, AP-HP, Paris, France
| |
Collapse
|