1
|
Parolini C. Pathophysiology of bone remodelling cycle: Role of immune system and lipids. Biochem Pharmacol 2025; 235:116844. [PMID: 40044049 DOI: 10.1016/j.bcp.2025.116844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 01/31/2025] [Accepted: 02/28/2025] [Indexed: 03/15/2025]
Abstract
Osteoporosis is the most common skeletal disease worldwide, characterized by low bone mineral density, resulting in weaker bones, and an increased risk of fragility fractures. The maintenance of bone mass relies on the precise balance between bone synthesis and resorption. The close relationship between the immune and skeletal systems, called "osteoimmunology", was coined to identify these overlapping "scientific worlds", and its function resides in the evaluation of the mutual effects of the skeletal and immune systems at the molecular and cellular levels, in both physiological and pathological states. Lipids play an essential role in skeletal metabolism and bone health. Indeed, bone marrow and its skeletal components demand a dramatic amount of daily energy to control hematopoietic turnover, acquire and maintain bone mass, and actively being involved in whole-body metabolism. Statins, the main therapeutic agents in lowering plasma cholesterol levels, are able to promote osteoblastogenesis and inhibit osteoclastogenesis. This review is meant to provide an updated overview of the pathophysiology of bone remodelling cycle, focusing on the interplay between bone, immune system and lipids. Novel therapeutic strategies for the management of osteoporosis are also discussed.
Collapse
Affiliation(s)
- Cinzia Parolini
- Department of Pharmacological and Biomolecular Sciences, 'Rodolfo Paoletti', via Balzaretti 9 - Università degli Studi di Milano 20133 Milano, Italy.
| |
Collapse
|
2
|
Gomes Velasque Gama F, Casciani C, Dutra EH. FGF18 induces chondrogenesis and anti-osteoarthritic effects in a mouse model for TMJ degeneration. PLoS One 2025; 20:e0317816. [PMID: 40273050 PMCID: PMC12021239 DOI: 10.1371/journal.pone.0317816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 01/06/2025] [Indexed: 04/26/2025] Open
Abstract
OBJECTIVE Temporomandibular Joint Osteoarthritis (TMJ-OA) is a degenerative disease characterized by progressive loss of cartilage and subchondral bone sclerosis. Currently there are no effective treatments for TMJ-OA. FGF18 is a member of the fibroblast growth factor family with essential roles for chondrogenesis, selectively binding to FGFR3 receptor. Studies have reported FGF18 attenuates cartilage degradation. Whereas the anti-osteoarthritic effects of FGF18 in the articular cartilage are known, the effects of FGF18 in a TMJ fibrocartilage degeneration mouse model remain to be determined. The goal of this project was to determine the effects of intra-articular injections of FGF18 in a mouse model for TMJ degeneration. METHOD Prosthesis tubes were bonded at the left lower incisor of 6-week-old triple collagen transgenic mice (Col1a1XCol2a1XCol10a1), creating unilateral crossbite and degeneration of the TMJ fibrocartilage. Six weeks after placement of prosthesis tubes, experimental and control mice received intra-articular injections of rmFGF18 (5µg/week) or saline, respectively, for 3 weeks. RESULTS Mice receiving saline intra-articular injections presented with a thinner cartilage layer with decreased proteoglycan distribution and Edu positive cells (chondrocyte proliferation marker), while mice injected with rmFGF18 presented with significant increased fibrocartilage thickness, remarkable proteoglycan distribution and chondrocyte proliferation, suggesting healing of the induced degeneration. Furthermore, reversal of the TMJ degeneration achieved by rmFGF18 injection was accompanied by a substantial reduction in Noggin (antagonist of BMP signaling), increase in TIMP1 (inhibitor of metalloproteinases such as MMP13) and decrease in MMP13 expression. CONCLUSION Our results postulate FGF18 as a powerful growth factor for the healing of TMJ fibrocartilage.
Collapse
Affiliation(s)
| | - Christina Casciani
- Department of Orthodontics, UConn Health, Farmington, Connecticut, United States of America
| | - Eliane Hermes Dutra
- Department of Orthodontics, UConn Health, Farmington, Connecticut, United States of America
| |
Collapse
|
3
|
Largo R, Mediero A, Villa-Gomez C, Bermejo-Alvarez I, Herrero-Beaumont G. Aberrant anabolism hinders constructive metabolism of chondrocytes by pharmacotherapy in osteoarthritis. Bone Joint Res 2025; 14:199-207. [PMID: 40042132 PMCID: PMC11881514 DOI: 10.1302/2046-3758.143.bjr-2024-0241.r1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/03/2025] Open
Abstract
Osteoarthritis (OA) is a highly prevalent and disabling disease with an unmet therapeutic need. The characteristic cartilage loss and alteration of other joint structures result from a complex interaction of multiple risk factors, with mechanical overload consistently playing a central role. This overload generates an inflammatory response in the cartilage due to the activation of the innate immune response in chondrocytes, which occurs through various cellular mechanisms. Moreover, risk factors associated with obesity, being overweight, and metabolic syndrome enhance the inflammatory response both locally and systemically. OA chondrocytes, the only cells present in articular cartilage, are therefore inflamed and initiate an anabolic process in an attempt to repair the damaged tissue, which ultimately results in an aberrant and dysfunctional process. Under these circumstances, where the cartilage continues to be subjected to chronic mechanical stress, proposing a treatment that stimulates the chondrocytes' anabolic response to restore tissue structure does not appear to be a therapeutic target with a high likelihood of success. In fact, anabolic drugs proposed for the treatment of OA have yet to demonstrate efficacy. By contrast, multiple therapeutic strategies focused on pharmacologically managing the inflammatory component, both at the joint and systemic levels, have shown promise. Therefore, prioritizing the control of chronic innate pro-inflammatory pathways presents the most viable and promising therapeutic strategy for the effective management of OA. As research continues, this approach may offer the best opportunity to alleviate the burden of this incapacitating disease.
Collapse
Affiliation(s)
- Raquel Largo
- Joint and Bone Research Unit, Service of Rheumatology, IIS Fundación Jiménez Díaz UAM, Madrid, Spain
| | - Aranzazu Mediero
- Joint and Bone Research Unit, Service of Rheumatology, IIS Fundación Jiménez Díaz UAM, Madrid, Spain
| | - Cristina Villa-Gomez
- Joint and Bone Research Unit, Service of Rheumatology, IIS Fundación Jiménez Díaz UAM, Madrid, Spain
| | - Ismael Bermejo-Alvarez
- Joint and Bone Research Unit, Service of Rheumatology, IIS Fundación Jiménez Díaz UAM, Madrid, Spain
| | - Gabriel Herrero-Beaumont
- Joint and Bone Research Unit, Service of Rheumatology, IIS Fundación Jiménez Díaz UAM, Madrid, Spain
| |
Collapse
|
4
|
Li K, Song Y, Fan Y, Zhang H, Chu M, Liu Y. Transcriptome integration analysis revealed that miR-103-3p regulates goat myoblast proliferation by targeting FGF18. BMC Genomics 2025; 26:16. [PMID: 39773020 PMCID: PMC11706129 DOI: 10.1186/s12864-024-11183-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 12/23/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Myoblasts serve as the fundamental building blocks of muscle fibers, and there is a positive correlation between the diameter of myofibers during the juvenile phase and the rate of muscle growth, which does not change in adulthood. However, the molecular mechanisms governing myofiber diameter across various developmental stages in goats remain largely unclear. RESULTS In this study, we examined miRNA expression in the longissimus dorsi muscle tissue of goats at two distinct ages: one month, a period characterized by robust muscle growth, and nine months, when muscle development plateaus in adulthood. A total of 408 known miRNAs and 86 novel miRNAs were identified, with 32 miRNAs exhibiting differential expression between the two groups. A functional enrichment analysis of these targeted genes revealed significant enrichment in pathways closely correlated with skeletal muscle growth, development, and senescence. Notably, chi-miR-103-3p was identified among the DE miRNAs and appeared to play an important role in skeletal myoblast proliferation. Bioinformatics analysis, complemented by dual luciferase activity assays revealed that chi-miR-103-3p specifically targets the 3'UTR of FGF18. Subsequent cell transfection experiments demonstrated that chi-miR-103-3p suppresses the expression of FGF18 in goat myoblasts, thereby inhibiting cell proliferation. Moreover, FGF18 was observed to enhance the proliferation of goat myoblasts. CONCLUSIONS Collectively, our data indicated that the elevated expression of chi-miR-103-3p in adult goat myoblasts significantly repressed FGF18 expression, thereby limiting rapid muscle growth. Proliferation and differentiation of myoblasts can affect myofiber number and cell volume expansion. These findings lay the foundation for further elucidation of the molecular mechanisms underlying muscle growth and development across different life stages of goats. Additionally, it could be a potential molecular marker for improving muscle production in goats.
Collapse
Affiliation(s)
- Kunyu Li
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Yize Song
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Yekai Fan
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Hui Zhang
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Mingxing Chu
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China.
| | - Yufang Liu
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China.
| |
Collapse
|
5
|
Hernández-García F, Fernández-Iglesias Á, Rodríguez Suárez J, Gil Peña H, López JM, Pérez RF. The Crosstalk Between Cartilage and Bone in Skeletal Growth. Biomedicines 2024; 12:2662. [PMID: 39767569 PMCID: PMC11727353 DOI: 10.3390/biomedicines12122662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Revised: 11/11/2024] [Accepted: 11/18/2024] [Indexed: 01/04/2025] Open
Abstract
While the flat bones of the face, most of the cranial bones, and the clavicles are formed directly from sheets of undifferentiated mesenchymal cells, most bones in the human body are first formed as cartilage templates. Cartilage is subsequently replaced by bone via a very tightly regulated process termed endochondral ossification, which is led by chondrocytes of the growth plate (GP). This process requires continuous communication between chondrocytes and invading cell populations, including osteoblasts, osteoclasts, and vascular cells. A deeper understanding of these signaling pathways is crucial not only for normal skeletal growth and maturation but also for their potential relevance to pathophysiological processes in bones and joints. Due to limited information on the communication between chondrocytes and other cell types in developing bones, this review examines the current knowledge of how interactions between chondrocytes and bone-forming cells modulate bone growth.
Collapse
Affiliation(s)
- Frank Hernández-García
- Departamento de Medicina, Oviedo University, 33003 Oviedo, Spain; (F.H.-G.); (J.R.S.)
- Grupo Investigación Pediatría, Instituto de Investigación Sanitaria del Principado de Asturias, 33011 Oviedo, Spain; (Á.F.-I.); (H.G.P.); (J.M.L.)
| | - Ángela Fernández-Iglesias
- Grupo Investigación Pediatría, Instituto de Investigación Sanitaria del Principado de Asturias, 33011 Oviedo, Spain; (Á.F.-I.); (H.G.P.); (J.M.L.)
| | - Julián Rodríguez Suárez
- Departamento de Medicina, Oviedo University, 33003 Oviedo, Spain; (F.H.-G.); (J.R.S.)
- Grupo Investigación Pediatría, Instituto de Investigación Sanitaria del Principado de Asturias, 33011 Oviedo, Spain; (Á.F.-I.); (H.G.P.); (J.M.L.)
- AGC de Infancia y Adolescencia, Hospital Universitario Central de Asturias, 33011 Oviedo, Spain
- RICORS-SAMID (RD21/0012), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Helena Gil Peña
- Grupo Investigación Pediatría, Instituto de Investigación Sanitaria del Principado de Asturias, 33011 Oviedo, Spain; (Á.F.-I.); (H.G.P.); (J.M.L.)
- AGC de Infancia y Adolescencia, Hospital Universitario Central de Asturias, 33011 Oviedo, Spain
- RICORS2040 (RD21/0005/0011), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - José M. López
- Grupo Investigación Pediatría, Instituto de Investigación Sanitaria del Principado de Asturias, 33011 Oviedo, Spain; (Á.F.-I.); (H.G.P.); (J.M.L.)
- Departamento de Morfología y Biología Celular, Oviedo University, 33003 Oviedo, Spain
| | - Rocío Fuente Pérez
- Universidad Europea de Madrid, Department of Nursing, Faculty of Medicine, Health and Sports, 28670 Madrid, Spain
| |
Collapse
|
6
|
Wang Y, Zhang H, Zhan Y, Li Z, Li S, Guo S. Comprehensive in silico analysis of prognostic and immune infiltrates for FGFs in human ovarian cancer. J Ovarian Res 2024; 17:197. [PMID: 39385288 PMCID: PMC11465590 DOI: 10.1186/s13048-024-01496-z] [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: 06/05/2024] [Accepted: 08/14/2024] [Indexed: 10/12/2024] Open
Abstract
BACKGROUND Fibroblast growth factors (FGFs) are cell signaling proteins that perform multiple biological processes in many biological processes (cell development, repair, and metabolism). The dynamics of tumor cells, such as angiogenesis, transformation, and proliferation, have a significant impact on neoplasia and are modulated by FGFs. FGFs' expression and prognostic significance in ovarian cancer (OC), however, remain unclear. METHODS Through a series of in silico analysis, we investigated the transcriptional, survival data, genetic variation, gene-gene interaction network, ferroptosis-related genes, and DNA methylation of FGFs in OC patients. RESULTS We discovered that while FGF18 expression levels were higher in OC tissues than in normal OC tissues, FGF2/7/10/17/22 expression levels were lower in the former, and that FGF1/19 expression was related to the tumor stage in OC patients. According to the survival analysis, the clinical prognosis of individuals with OC was associated with the aberrant expression of FGFs. The function of FGFs and their neighboring genes was mainly connected to the cellular response to FGF stimulus. There was a negative correlation between FGF expression and various immune cell infiltration. CONCLUSIONS This study clarifies the relationship between FGFs and OC, which might provide new insights into the choice of prognostic biomarkers of OC patients.
Collapse
Affiliation(s)
- Yu Wang
- Emergency Medicine Clinical Research Center, Beijing Chao-yang Hospital, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Capital Medical University, Beijing, 100020, P.R. China
| | - Haiyue Zhang
- Thrombosis research center, Beijing Jishuitan hospital, Capital Medical University, Beijing, China, Xicheng District, Beijing 100035, China
- Department of Clinical Laboratory, Beijing Jishuitan Hospital Affiliated to Capital Medical University, Xicheng District, Beijing, China
| | - Yuanyuan Zhan
- Department of Endocrinology, Beijing Chao-yang Hospital, Capital Medical University, Beijing, 100020, P.R. China
| | - Zhuoran Li
- Emergency Medicine Clinical Research Center, Beijing Chao-yang Hospital, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Capital Medical University, Beijing, 100020, P.R. China
| | - Sujing Li
- Department of Plastic Surgery, Zhengzhou First People's Hospital, Zhengzhou, China
| | - Shubin Guo
- Emergency Medicine Clinical Research Center, Beijing Chao-yang Hospital, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Capital Medical University, Beijing, 100020, P.R. China.
| |
Collapse
|
7
|
Hasan S, Naseer S, Zamzam M, Mohilldean H, Van Wagoner C, Hasan A, Saleh ES, Uhley V, Kamel-ElSayed S. Nutrient and Hormonal Effects on Long Bone Growth in Healthy and Obese Children: A Literature Review. CHILDREN (BASEL, SWITZERLAND) 2024; 11:817. [PMID: 39062266 PMCID: PMC11276385 DOI: 10.3390/children11070817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 06/28/2024] [Accepted: 07/01/2024] [Indexed: 07/28/2024]
Abstract
Longitudinal bone growth is mediated through several mechanisms including macro- and micronutrients, and endocrine and paracrine hormones. These mechanisms can be affected by childhood obesity as excess adiposity may affect signaling pathways, place undue stress on the body, and affect normal physiology. This review describes the physiology of the epiphyseal growth plate, its regulation under healthy weight and obesity parameters, and bone pathology following obesity. A literature review was performed utilizing PubMed, PMC, NIH, and the Cochrane Database of Systematic Reviews pertinent to hormonal and nutritional effects on bone development, child obesity, and pathologic bone development related to weight. The review indicates a complex network of nutrients, hormones, and multi-system interactions mediates long bone growth. As growth of long bones occurs during childhood and the pubertal growth spurt, pediatric bones require adequate levels of minerals, vitamins, amino acids, and a base caloric supply for energy. Recommendations should focus on a nutrient-dense dietary approach rather than restrictive caloric diets to maintain optimal health. In conclusion, childhood obesity has profound multifaceted effects on the developing musculoskeletal system, ultimately causing poor nutritional status during development. Weight loss, under medical supervision, with proper nutritional guidelines, can help counteract the ill effects of childhood obesity.
Collapse
Affiliation(s)
- Sazid Hasan
- School of Medicine, Oakland University William Beaumont, Rochester, MI 48309, USA
| | - Shahrukh Naseer
- School of Medicine, Oakland University William Beaumont, Rochester, MI 48309, USA
| | - Mazen Zamzam
- School of Medicine, Oakland University William Beaumont, Rochester, MI 48309, USA
| | - Hashem Mohilldean
- School of Medicine, Oakland University William Beaumont, Rochester, MI 48309, USA
| | - Colin Van Wagoner
- School of Medicine, Oakland University William Beaumont, Rochester, MI 48309, USA
| | - Ahmad Hasan
- Department of Orthopedic Surgery, Detroit Medical Center, Detroit, MI 48201, USA
| | - Ehab S. Saleh
- School of Medicine, Oakland University William Beaumont, Rochester, MI 48309, USA
- Department of Orthopedic Surgery, Beaumont Hospital, Royal Oak, MI 48073, USA
| | - Virginia Uhley
- School of Medicine, Oakland University William Beaumont, Rochester, MI 48309, USA
| | - Suzan Kamel-ElSayed
- School of Medicine, Oakland University William Beaumont, Rochester, MI 48309, USA
| |
Collapse
|
8
|
Li HZ, Zhang JL, Yuan DL, Xie WQ, Ladel CH, Mobasheri A, Li YS. Role of signaling pathways in age-related orthopedic diseases: focus on the fibroblast growth factor family. Mil Med Res 2024; 11:40. [PMID: 38902808 PMCID: PMC11191355 DOI: 10.1186/s40779-024-00544-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Accepted: 06/12/2024] [Indexed: 06/22/2024] Open
Abstract
Fibroblast growth factor (FGF) signaling encompasses a multitude of functions, including regulation of cell proliferation, differentiation, morphogenesis, and patterning. FGFs and their receptors (FGFR) are crucial for adult tissue repair processes. Aberrant FGF signal transduction is associated with various pathological conditions such as cartilage damage, bone loss, muscle reduction, and other core pathological changes observed in orthopedic degenerative diseases like osteoarthritis (OA), intervertebral disc degeneration (IVDD), osteoporosis (OP), and sarcopenia. In OA and IVDD pathologies specifically, FGF1, FGF2, FGF8, FGF9, FGF18, FGF21, and FGF23 regulate the synthesis, catabolism, and ossification of cartilage tissue. Additionally, the dysregulation of FGFR expression (FGFR1 and FGFR3) promotes the pathological process of cartilage degradation. In OP and sarcopenia, endocrine-derived FGFs (FGF19, FGF21, and FGF23) modulate bone mineral synthesis and decomposition as well as muscle tissues. FGF2 and other FGFs also exert regulatory roles. A growing body of research has focused on understanding the implications of FGF signaling in orthopedic degeneration. Moreover, an increasing number of potential targets within the FGF signaling have been identified, such as FGF9, FGF18, and FGF23. However, it should be noted that most of these discoveries are still in the experimental stage, and further studies are needed before clinical application can be considered. Presently, this review aims to document the association between the FGF signaling pathway and the development and progression of orthopedic diseases. Besides, current therapeutic strategies targeting the FGF signaling pathway to prevent and treat orthopedic degeneration will be evaluated.
Collapse
Affiliation(s)
- Heng-Zhen Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Jing-Lve Zhang
- Department of Plastic and Cosmetic Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
- Xiangya School of Medicine Central, South University, Changsha, 410083, China
| | - Dong-Liang Yuan
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008, China
- Xiangya School of Medicine Central, South University, Changsha, 410083, China
| | - Wen-Qing Xie
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | | | - Ali Mobasheri
- Faculty of Medicine, Research Unit of Health Sciences and Technology, University of Oulu, 90014, Oulu, Finland.
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, 08406, Vilnius, Lithuania.
- Department of Rheumatology and Clinical Immunology, Universitair Medisch Centrum Utrecht, Utrecht, 3508, GA, the Netherlands.
- Department of Joint Surgery, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China.
- World Health Organization Collaborating Centre for Public Health Aspects of Musculoskeletal Health and Aging, Université de Liège, B-4000, Liège, Belgium.
| | - Yu-Sheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China.
| |
Collapse
|
9
|
Wang H, Qi LL, Shema C, Jiang KY, Ren P, Wang H, Wang L. Advances in the role and mechanism of fibroblasts in fracture healing. Front Endocrinol (Lausanne) 2024; 15:1350958. [PMID: 38469138 PMCID: PMC10925620 DOI: 10.3389/fendo.2024.1350958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 02/01/2024] [Indexed: 03/13/2024] Open
Abstract
With the development of social population ageing, bone fracture has become a global public health problem due to its high morbidity, disability and mortality. Fracture healing is a complex phenomenon involving the coordinated participation of immigration, differentiation and proliferation of inflammatory cells, angioblasts, fibroblasts, chondroblasts and osteoblasts which synthesize and release bioactive substances of extracellular matrix components, Mortality caused by age-related bone fractures or osteoporosis is steadily increasing worldwide as the population ages. Fibroblasts play an important role in the process of fracture healing. However, it is not clear how the growth factors and extracellular matrix stiffness of the bone-regeneration microenvironment affects the function of osteoblasts and fibroblasts in healing process. Therefore, this article focuses on the role of fibroblasts in the process of fracture healing and mechanisms of research progress.
Collapse
Affiliation(s)
- Hui Wang
- Department of Orthopedics, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Li-li Qi
- Experimental Center for Teaching of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Clement Shema
- Department of Orthopedic Research Center, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- International Education College of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Kui-ying Jiang
- National Demonstration Center for Experimental Basic Medical Education, Capital Medical University, Beijing, China
| | - Ping Ren
- Experimental Center for Teaching of Hebei Medical University, Shijiazhuang, Hebei, China
| | - He Wang
- Department of Pathogenic Biology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Lei Wang
- Department of Human Anatomy, Institute of Medicine and Health, Hebei Medical University, Shijiazhuang, Hebei, China
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei, China
- Neuroscience Research Center, Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Key Laboratory of Neurodegenerative Disease Mechanism, Shijiazhuang, Hebei, China
| |
Collapse
|
10
|
Yang J, Dong X, Wen H, Li Y, Wang X, Yan S, Zuo C, Lyu L, Zhang K, Qi X. FGFs function in regulating myoblasts differentiation in spotted sea bass (Lateolabrax maculatus). Gen Comp Endocrinol 2024; 347:114426. [PMID: 38103843 DOI: 10.1016/j.ygcen.2023.114426] [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: 10/19/2023] [Revised: 12/11/2023] [Accepted: 12/11/2023] [Indexed: 12/19/2023]
Abstract
Fibroblast growth factors (FGFs) are a family of structurally related peptides that regulate processes such as cell proliferation, differentiation, and damage repair. In our previous study, fibroblast growth factor receptor 4 (fgfr4) was detected in the most significant quantitative trait loci (QTL), when identified of QTLs and genetic markers for growth-related traits in spotted sea bass. However, knowledge of the function of fgfr4 is lacking, even the legends to activate the receptor is unknown in fish. To remedy this problem, in the present study, a total of 33 fgfs were identified from the genomic and transcriptomic databases of spotted sea bass, of which 10 were expressed in the myoblasts. According to the expression pattern during myoblasts proliferation and differentiation, fgf6a, fgf6b and fgf18 were selected for further prokaryotic expression and purification. The recombinant proteins FGF6a, FGF6b and FGF18 were found to inhibit myoblast differentiation. Overall, our results provide a theoretical basis for the molecular mechanisms of growth regulation in economic fish such as spotted sea bass.
Collapse
Affiliation(s)
- Jing Yang
- Key Laboratory of Mariculture, Ministry of Education (KLMME), Ocean University of China, Qingdao 266003
| | - Ximeng Dong
- Key Laboratory of Mariculture, Ministry of Education (KLMME), Ocean University of China, Qingdao 266003
| | - Haishen Wen
- Key Laboratory of Mariculture, Ministry of Education (KLMME), Ocean University of China, Qingdao 266003
| | - Yun Li
- Key Laboratory of Mariculture, Ministry of Education (KLMME), Ocean University of China, Qingdao 266003
| | - Xiaojie Wang
- Key Laboratory of Mariculture, Ministry of Education (KLMME), Ocean University of China, Qingdao 266003
| | - Shaojing Yan
- Key Laboratory of Mariculture, Ministry of Education (KLMME), Ocean University of China, Qingdao 266003
| | - Chenpeng Zuo
- Key Laboratory of Mariculture, Ministry of Education (KLMME), Ocean University of China, Qingdao 266003
| | - Likang Lyu
- Key Laboratory of Mariculture, Ministry of Education (KLMME), Ocean University of China, Qingdao 266003
| | - Kaiqiang Zhang
- Key Laboratory of Mariculture, Ministry of Education (KLMME), Ocean University of China, Qingdao 266003
| | - Xin Qi
- Key Laboratory of Mariculture, Ministry of Education (KLMME), Ocean University of China, Qingdao 266003.
| |
Collapse
|
11
|
Ng JQ, Jafarov TH, Little CB, Wang T, Ali AM, Ma Y, Radford GA, Vrbanac L, Ichinose M, Whittle S, Hunter DJ, Lannagan TRM, Suzuki N, Goyne JM, Kobayashi H, Wang TC, Haynes DR, Menicanin D, Gronthos S, Worthley DL, Woods SL, Mukherjee S. Loss of Grem1-lineage chondrogenic progenitor cells causes osteoarthritis. Nat Commun 2023; 14:6909. [PMID: 37907525 PMCID: PMC10618187 DOI: 10.1038/s41467-023-42199-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 10/03/2023] [Indexed: 11/02/2023] Open
Abstract
Osteoarthritis (OA) is characterised by an irreversible degeneration of articular cartilage. Here we show that the BMP-antagonist Gremlin 1 (Grem1) marks a bipotent chondrogenic and osteogenic progenitor cell population within the articular surface. Notably, these progenitors are depleted by injury-induced OA and increasing age. OA is also caused by ablation of Grem1 cells in mice. Transcriptomic and functional analysis in mice found that articular surface Grem1-lineage cells are dependent on Foxo1 and ablation of Foxo1 in Grem1-lineage cells caused OA. FGFR3 signalling was confirmed as a promising therapeutic pathway by administration of pathway activator, FGF18, resulting in Grem1-lineage chondrocyte progenitor cell proliferation, increased cartilage thickness and reduced OA. These findings suggest that OA, in part, is caused by mechanical, developmental or age-related attrition of Grem1 expressing articular cartilage progenitor cells. These cells, and the FGFR3 signalling pathway that sustains them, may be effective future targets for biological management of OA.
Collapse
Affiliation(s)
- Jia Q Ng
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Toghrul H Jafarov
- Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Christopher B Little
- Raymond Purves Bone & Joint Research Laboratories, Kolling Institute, University of Sydney Faculty of Medicine and Health, Royal North Shore Hospital, St. Leonards, NSW, Australia
| | - Tongtong Wang
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
- Precision Cancer Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Abdullah M Ali
- Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Yan Ma
- Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Georgette A Radford
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Laura Vrbanac
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Mari Ichinose
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Samuel Whittle
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
- Rheumatology Unit, The Queen Elizabeth Hospital, Woodville South, SA, Australia
| | - David J Hunter
- Northern Clinical School, University of Sydney, St. Leonards, Sydney, NSW, Australia
| | - Tamsin R M Lannagan
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Nobumi Suzuki
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Jarrad M Goyne
- Precision Cancer Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Hiroki Kobayashi
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Timothy C Wang
- Department of Medicine and Irving Cancer Research Center, Columbia University, New York, NY, USA
| | - David R Haynes
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Danijela Menicanin
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Stan Gronthos
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
- School of Biomedicine, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Daniel L Worthley
- Precision Cancer Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia.
- Colonoscopy Clinic, Brisbane, QLD, Australia.
| | - Susan L Woods
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia.
- Precision Cancer Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia.
| | | |
Collapse
|
12
|
Jan Vilim, Ghazalova T, Petulova E, Horackova A, Stepankova V, Chaloupkova R, Bednar D, Damborsky J, Prokop Z. Computer-assisted stabilization of fibroblast growth factor FGF-18. Comput Struct Biotechnol J 2023; 21:5144-5152. [PMID: 37920818 PMCID: PMC10618113 DOI: 10.1016/j.csbj.2023.10.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 10/05/2023] [Accepted: 10/06/2023] [Indexed: 11/04/2023] Open
Abstract
The fibroblast growth factors (FGF) family holds significant potential for addressing chronic diseases. Specifically, recombinant FGF18 shows promise in treating osteoarthritis by stimulating cartilage formation. However, recent phase 2 clinical trial results of sprifermin (recombinant FGF18) indicate insufficient efficacy. Leveraging our expertise in rational protein engineering, we conducted a study to enhance the stability of FGF18. As a result, we obtained a stabilized variant called FGF18-E4, which exhibited improved stability with 16 °C higher melting temperature, resistance to trypsin and a 2.5-fold increase in production yields. Moreover, the FGF18-E4 maintained mitogenic activity after 1-week incubation at 37 °C and 1-day at 50 °C. Additionally, the inserted mutations did not affect its binding to the fibroblast growth factor receptors, making FGF18-E4 a promising candidate for advancing FGF-based osteoarthritis treatment.
Collapse
Affiliation(s)
- Jan Vilim
- Loschmidt Laboratories, Department of Experimental Biology and RECETOX, Faculty of Science, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic
- Enantis Ltd., INBIT, Kamenice 34, 625 00 Brno, Czech Republic
| | | | - Eliska Petulova
- Enantis Ltd., INBIT, Kamenice 34, 625 00 Brno, Czech Republic
| | - Aneta Horackova
- Enantis Ltd., INBIT, Kamenice 34, 625 00 Brno, Czech Republic
| | | | | | - David Bednar
- Loschmidt Laboratories, Department of Experimental Biology and RECETOX, Faculty of Science, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital Brno, Pekarska 53, 656 91 Brno, Czech Republic
| | - Jiri Damborsky
- Loschmidt Laboratories, Department of Experimental Biology and RECETOX, Faculty of Science, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital Brno, Pekarska 53, 656 91 Brno, Czech Republic
| | - Zbynek Prokop
- Loschmidt Laboratories, Department of Experimental Biology and RECETOX, Faculty of Science, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital Brno, Pekarska 53, 656 91 Brno, Czech Republic
| |
Collapse
|
13
|
Abstract
Angiogenesis in the bone is unique and involves distinctive signals. Whether they are created through intramembranous ossification or endochondral ossification, bones are highly vascularized tissues. Long bones undergo a sequence of processes known as endochondral osteogenesis. Angiogenesis occurs during the creation of endochondral bone and is mediated by a variety of cells and factors. An initially avascular cartilage template is invaded by blood vessels from the nearby subchondral bone thanks to the secreted angiogenic chemicals by hypertrophic chondrocytes. Vascular endothelial growth factor (VEGF), one of several angiogenic molecules, is a significant regulator of blood vessel invasion, cartilage remodeling, and ossification of freshly created bone matrix; chondrocyte proliferation and hypertrophy are facilitated by the production of VEGFA and VEGF receptor-2 (VEGFR-2), which is stimulated by fibroblast growth factors (FGFs). NOTCH signaling controls blood capillaries formation during bone maturation and regeneration, while hypoxia-inducible factor 1 alpha (HIF1-a) promotes chondrocyte development by switching to anaerobic metabolism. To control skeletal remodeling and repair, osteogenic cells release angiogenic factors, whereas endothelial cells secrete angiocrine factors. One of the better instances of functional blood vessels specialization for certain organs is the skeletal system. A subpopulation of capillary endothelial cells in the bone regulate the activity of osteoprogenitor cells, which in turn affects bone formation during development and adult homeostasis. Angiogenesis and osteogenesis are strictly connected, and their crosstalk is essential to guarantee bone formation and to maintain bone homeostasis. Additionally, pathological processes including inflammation, cancer, and aging include both bone endothelial cells and angiocrine factors. Therefore, the study and understanding of these mechanisms is fundamental, because molecules and factors involved may represent key targets for novel and advanced therapies.
Collapse
Affiliation(s)
- Domenico Ribatti
- Department of Translational Biomedicine and Neurosciences, University of Bari Medical School, Bari, Italy
| | | |
Collapse
|
14
|
Shigley C, Trivedi J, Meghani O, Owens BD, Jayasuriya CT. Suppressing Chondrocyte Hypertrophy to Build Better Cartilage. Bioengineering (Basel) 2023; 10:741. [PMID: 37370672 DOI: 10.3390/bioengineering10060741] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 06/13/2023] [Accepted: 06/15/2023] [Indexed: 06/29/2023] Open
Abstract
Current clinical strategies for restoring cartilage defects do not adequately consider taking the necessary steps to prevent the formation of hypertrophic tissue at injury sites. Chondrocyte hypertrophy inevitably causes both macroscopic and microscopic level changes in cartilage, resulting in adverse long-term outcomes following attempted restoration. Repairing/restoring articular cartilage while minimizing the risk of hypertrophic neo tissue formation represents an unmet clinical challenge. Previous investigations have extensively identified and characterized the biological mechanisms that regulate cartilage hypertrophy with preclinical studies now beginning to leverage this knowledge to help build better cartilage. In this comprehensive article, we will provide a summary of these biological mechanisms and systematically review the most cutting-edge strategies for circumventing this pathological hallmark of osteoarthritis.
Collapse
Affiliation(s)
- Christian Shigley
- The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
| | - Jay Trivedi
- Department of Orthopaedics, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI 02903, USA
| | - Ozair Meghani
- Department of Orthopaedics, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI 02903, USA
| | - Brett D Owens
- Department of Orthopaedics, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI 02903, USA
- Division of Sports Surgery, Department of Orthopaedic Surgery, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI 02903, USA
| | - Chathuraka T Jayasuriya
- Department of Orthopaedics, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI 02903, USA
| |
Collapse
|
15
|
Ng JQ, Jafarov TH, Little CB, Wang T, Ali A, Ma Y, Radford GA, Vrbanac L, Ichinose M, Whittle S, Hunter D, Lannagan TRM, Suzuki N, Goyne JM, Kobayashi H, Wang TC, Haynes D, Menicanin D, Gronthos S, Worthley DL, Woods SL, Mukherjee S. Loss of Grem1-articular cartilage progenitor cells causes osteoarthritis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.29.534651. [PMID: 37034712 PMCID: PMC10081168 DOI: 10.1101/2023.03.29.534651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Osteoarthritis (OA), which carries an enormous disease burden across the world, is characterised by irreversible degeneration of articular cartilage (AC), and subsequently bone. The cellular cause of OA is unknown. Here, using lineage tracing in mice, we show that the BMP-antagonist Gremlin 1 (Grem1) marks a novel chondrogenic progenitor (CP) cell population in the articular surface that generates joint cartilage and subchondral bone during development and adulthood. Notably, this CP population is depleted in injury-induced OA, and with age. OA is also induced by toxin-mediated ablation of Grem1 CP cells in young mice. Transcriptomic analysis and functional modelling in mice revealed articular surface Grem1-lineage cells are dependent on Foxo1; ablation of Foxo1 in Grem1-lineage cells led to early OA. This analysis identified FGFR3 signalling as a therapeutic target, and injection of its activator, FGF18, caused proliferation of Grem1-lineage CP cells, increased cartilage thickness, and reduced OA pathology. We propose that OA arises from the loss of CP cells at the articular surface secondary to an imbalance in progenitor cell homeostasis and present a new progenitor population as a locus for OA therapy.
Collapse
Affiliation(s)
- Jia Q. Ng
- Adelaide Medical School, Faculty of Health and Medical Sciences University of Adelaide, Adelaide, SA, Australia
- These authors contributed equally
| | - Toghrul H. Jafarov
- Department of Medicine, Columbia University Medical Center, New York, NY, USA
- These authors contributed equally
| | - Christopher B. Little
- Raymond Purves Bone & Joint Research Laboratories, Kolling Institute, University of Sydney Faculty of Medicine and Health, Royal North Shore Hospital, St. Leonards, NSW, Australia
| | - Tongtong Wang
- Adelaide Medical School, Faculty of Health and Medical Sciences University of Adelaide, Adelaide, SA, Australia
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Abdullah Ali
- Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Yan Ma
- Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Georgette A Radford
- Adelaide Medical School, Faculty of Health and Medical Sciences University of Adelaide, Adelaide, SA, Australia
| | - Laura Vrbanac
- Adelaide Medical School, Faculty of Health and Medical Sciences University of Adelaide, Adelaide, SA, Australia
| | - Mari Ichinose
- Adelaide Medical School, Faculty of Health and Medical Sciences University of Adelaide, Adelaide, SA, Australia
| | - Samuel Whittle
- Adelaide Medical School, Faculty of Health and Medical Sciences University of Adelaide, Adelaide, SA, Australia
- Rheumatology Unit, The Queen Elizabeth Hospital, Woodville South, SA, Australia
| | - David Hunter
- Northern Clinical School, University of Sydney, St. Leonards, Sydney, NSW, Australia
| | - Tamsin RM Lannagan
- Adelaide Medical School, Faculty of Health and Medical Sciences University of Adelaide, Adelaide, SA, Australia
| | - Nobumi Suzuki
- Adelaide Medical School, Faculty of Health and Medical Sciences University of Adelaide, Adelaide, SA, Australia
| | - Jarrad M. Goyne
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Hiroki Kobayashi
- Adelaide Medical School, Faculty of Health and Medical Sciences University of Adelaide, Adelaide, SA, Australia
| | - Timothy C. Wang
- Department of Medicine and Irving Cancer Research Center, Columbia University, New York, NY USA
| | - David Haynes
- Adelaide Medical School, Faculty of Health and Medical Sciences University of Adelaide, Adelaide, SA, Australia
| | - Danijela Menicanin
- Adelaide Medical School, Faculty of Health and Medical Sciences University of Adelaide, Adelaide, SA, Australia
| | - Stan Gronthos
- Adelaide Medical School, Faculty of Health and Medical Sciences University of Adelaide, Adelaide, SA, Australia
- School of Biomedicine, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Daniel L. Worthley
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
- Colonoscopy Clinic, Brisbane, Qld, Australia
- These authors contributed equally, corresponding authors
| | - Susan L. Woods
- Adelaide Medical School, Faculty of Health and Medical Sciences University of Adelaide, Adelaide, SA, Australia
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
- These authors contributed equally, corresponding authors
| | - Siddhartha Mukherjee
- Department of Medicine, Columbia University Medical Center, New York, NY, USA
- These authors contributed equally, corresponding authors
| |
Collapse
|
16
|
Fibroblast growth factor 18 alleviates stress-induced pathological cardiac hypertrophy in male mice. Nat Commun 2023; 14:1235. [PMID: 36871047 PMCID: PMC9985628 DOI: 10.1038/s41467-023-36895-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 02/22/2023] [Indexed: 03/06/2023] Open
Abstract
Fibroblast growth factor-18 (FGF18) has diverse organ development and damage repair roles. However, its role in cardiac homeostasis following hypertrophic stimulation remains unknown. Here we investigate the regulation and function of the FGF18 in pressure overload (PO)-induced pathological cardiac hypertrophy. FGF18 heterozygous (Fgf18+/-) and inducible cardiomyocyte-specific FGF18 knockout (Fgf18-CKO) male mice exposed to transverse aortic constriction (TAC) demonstrate exacerbated pathological cardiac hypertrophy with increased oxidative stress, cardiomyocyte death, fibrosis, and dysfunction. In contrast, cardiac-specific overexpression of FGF18 alleviates hypertrophy, decreased oxidative stress, attenuates cardiomyocyte apoptosis, and ameliorates fibrosis and cardiac function. Tyrosine-protein kinase FYN (FYN), the downstream factor of FGF18, was identified by bioinformatics analysis, LC-MS/MS and experiment validation. Mechanistic studies indicate that FGF18/FGFR3 promote FYN activity and expression and negatively regulate NADPH oxidase 4 (NOX4), thereby inhibiting reactive oxygen species (ROS) generation and alleviating pathological cardiac hypertrophy. This study uncovered the previously unknown cardioprotective effect of FGF18 mediated by the maintenance of redox homeostasis through the FYN/NOX4 signaling axis in male mice, suggesting a promising therapeutic target for the treatment of cardiac hypertrophy.
Collapse
|
17
|
DePhillipo NN, Hendesi H, Aman ZS, Lind DRG, Smith J, Dodge GR. Preclinical Use of FGF-18 Augmentation for Improving Cartilage Healing Following Surgical Repair: A Systematic Review. Cartilage 2023; 14:59-66. [PMID: 36541606 PMCID: PMC10076894 DOI: 10.1177/19476035221142010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVE To evaluate the efficacy of fibroblast growth factor-18 (FGF-18) augmentation for improving articular cartilage healing following surgical repair in preclinical (in vivo) animal models. DESIGN A systematic review was performed evaluating the efficacy of FGF-18 augmentation with cartilage surgery compared with cartilage surgery without FGF-18 augmentation in living animal models. Eligible intervention groups were FGF-18 treatment in conjunction with orthopedic procedures, including microfracture, osteochondral auto/allograft transplantation, and cellular-based repair. Outcome variables were: International Cartilage Repair Society (ICRS) score, modified O'Driscoll histology score, tissue infill score, qualitative histology, and adverse events. Descriptive statistics were recorded and summarized for each included study. RESULTS In total, 493 studies were identified and 4 studies were included in the final analysis. All studies were randomized controlled trials evaluating in vivo use of recombinant human FGF-18 (rhFGF-18). Animal models included ovine (n = 3) and equine (n = 1), with rhFGF-18 use following microfracture (n = 3) or osteochondral defect repair (n = 1). The rhFGF-18 was delivered via intra-articular injection (n = 2), collagen membrane scaffold (n = 1), or both (n = 1). All studies reported significant, positive improvements in cartilage defect repair with rhFGF-18 compared with controls based on ICRS score (n = 4), modified O'Driscoll score (n = 4), tissue infill (n = 3), and expression of collagen type II (n = 4) (P < 0.05). No adverse events were reported with the intra-articular administration of this growth factor, indicating short-term safety and efficacy of rhFGF-18 in vivo. CONCLUSION This systematic review provides evidence that rhFGF-18 significantly improves cartilage healing at 6 months postoperatively following microfracture or osteochondral defect repair in preclinical randomized controlled trials.
Collapse
Affiliation(s)
- Nicholas N DePhillipo
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA
- Mechano-Therapeutics LLC, Philadelphia, PA, USA
| | - Honey Hendesi
- Department of Orthopedics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Zachary S Aman
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Dane R G Lind
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA
| | - Joseph Smith
- Department of Health, Nutrition, and Exercise Sciences, North Dakota State University, Fargo, ND, USA
| | - George R Dodge
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA
- Mechano-Therapeutics LLC, Philadelphia, PA, USA
| |
Collapse
|
18
|
Meyrueix LP, Gharaibeh R, Xue J, Brouwer C, Jones C, Adair L, Norris SA, Ideraabdullah F. Gestational diabetes mellitus placentas exhibit epimutations at placental development genes. Epigenetics 2022; 17:2157-2177. [DOI: 10.1080/15592294.2022.2111751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Affiliation(s)
| | - Raad Gharaibeh
- Department of Bioinformatics and Genomics, University of North Carolina, Charlotte, NC, USA
- Bioinformatics Service Division, University of North Carolina, Charlotte, NC, USA
- Department of Medicine, Division of Gastroenterology, University of Florida, Gainesville, FL, USA
| | - Jing Xue
- Genetics Department, University of North Carolina, Chapel Hill, NC, USA
| | - Cory Brouwer
- Department of Bioinformatics and Genomics, University of North Carolina, Charlotte, NC, USA
- Bioinformatics Service Division, University of North Carolina, Charlotte, NC, USA
| | - Corbin Jones
- Department of Biology and Integrative Program for Biological and Genome Sciences, University of North Carolina, Chapel Hill, NC, USA
| | - Linda Adair
- Nutrition Department, University of North Carolina, Chapel Hill, NC, USA
| | - Shane A. Norris
- SAMRC Developmental Health Pathways for Health Research Unit, University of Witwatersrand, Johannesburg, South Africa
| | - Folami Ideraabdullah
- Nutrition Department, University of North Carolina, Chapel Hill, NC, USA
- Genetics Department, University of North Carolina, Chapel Hill, NC, USA
- SAMRC Developmental Health Pathways for Health Research Unit, University of Witwatersrand, Johannesburg, South Africa
- Nutrition Research Institute, University of North Carolina, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
19
|
Fibroblast Growth Factors and Cellular Communication Network Factors: Intimate Interplay by the Founding Members in Cartilage. Int J Mol Sci 2022; 23:ijms23158592. [PMID: 35955724 PMCID: PMC9369280 DOI: 10.3390/ijms23158592] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/28/2022] [Accepted: 07/28/2022] [Indexed: 02/04/2023] Open
Abstract
Fibroblast growth factors (FGFs) constitute a large family of signaling molecules that act in an autocrine/paracrine, endocrine, or intracrine manner, whereas the cellular communication network factors (CCN) family is composed of six members that manipulate extracellular signaling networks. FGFs and CCNs are structurally and functionally distinct, except for the common characteristics as matricellular proteins. Both play significant roles in the development of a variety of tissues and organs, including the skeletal system. In vertebrates, most of the skeletal parts are formed and grow through a process designated endochondral ossification, in which chondrocytes play the central role. The growth plate cartilage is the place where endochondral ossification occurs, and articular cartilage is left to support the locomotive function of joints. Several FGFs, including FGF-2, one of the founding members of this family, and all of the CCNs represented by CCN2, which is required for proper skeletal development, can be found therein. Research over a decade has revealed direct binding of CCN2 to FGFs and FGF receptors (FGFRs), which occasionally affect the biological outcome via FGF signaling. Moreover, a recent study uncovered an integrated regulation of FGF and CCN genes by FGF signaling. In this review, after a brief introduction of these two families, molecular and genetic interactions between CCN and FGF family members in cartilage, and their biological effects, are summarized. The molecular interplay represents the mutual involvement of the other in their molecular functions, leading to collaboration between CCN2 and FGFs during skeletal development.
Collapse
|
20
|
Expanding horizons of achondroplasia treatment: current options and future developments. Osteoarthritis Cartilage 2022; 30:535-544. [PMID: 34864168 DOI: 10.1016/j.joca.2021.11.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 11/23/2021] [Accepted: 11/28/2021] [Indexed: 02/02/2023]
Abstract
Activating mutations in the FGFR3 receptor tyrosine kinase lead to most prevalent form of genetic dwarfism in humans, the achondroplasia. Many features of the complex function of FGFR3 in growing skeleton were characterized, which facilitated identification of therapy targets, and drove progress toward treatment. In August 2021, the vosoritide was approved for treatment of achondroplasia, which is based on a stable variant of the C-natriuretic peptide. Other drugs may soon follow, as several conceptually different inhibitors of FGFR3 signaling progress through clinical trials. Here, we review the current achondroplasia therapeutics, describe their mechanisms, and illuminate motivations leading to their development. We also discuss perspectives of curing achondroplasia, and options for repurposing achondroplasia drugs for dwarfing conditions unrelated to FGFR3.
Collapse
|
21
|
Hendesi H, Stewart S, Gibison ML, Guehring H, Richardson DW, Dodge GR. Recombinant fibroblast growth factor-18 (sprifermin) enhances microfracture-induced cartilage healing. J Orthop Res 2022; 40:553-564. [PMID: 33934397 PMCID: PMC8560655 DOI: 10.1002/jor.25063] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 03/19/2021] [Accepted: 04/26/2021] [Indexed: 02/04/2023]
Abstract
Posttraumatic osteoarthritis is a disabling condition impacting the mostly young and active population. In the present study, we investigated the impact of intra-articular sprifermin, a recombinant truncated fibroblast growth factor 18, on the outcome of microfracture treatment, a widely used surgical technique to enhance cartilage healing at the site of injury. For this study, we created a cartilage defect and performed microfracture treatment in fetlock joints of 18 horses, treated joints with one of three doses of sprifermin (10, 30, or 100 μg) or with saline, hyaluronan, and evaluated animals functional and structural outcomes over 24 weeks. For primary outcome measures, we performed histological evaluations and gene expression analysis of aggrecan, collagen types I and II, and cartilage oligomeric matrix protein in three regions of interest. As secondary outcome measures, we examined animals' lameness, performed arthroscopic, radiographic, and computed tomography (CT) scan imaging and gross morphology assessment. We detected the highest treatment benefit following 100 μg sprifermin treatment. The overall histological assessment showed an improvement in the kissing region, and the expression of constitutive genes showed a concentration-dependent enhancement, especially in the peri-lesion area. We detected a significant improvement in lameness scores, arthroscopic evaluations, radiography, and CT scans following sprifermin treatment when results from three dose-treatment groups were combined. Our results demonstrated, for the first time, an enhancement on microfracture outcomes following sprifermin treatment suggesting a cartilage regenerative role and a potential benefit of sprifermin treatment in early cartilage injuries.
Collapse
Affiliation(s)
- Honey Hendesi
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA
| | - Suzanne Stewart
- Department of Clinical Studies, New Bolton Center, University of Pennsylvania School of Veterinary Medicine
| | - Michelle L Gibison
- Department of Clinical Studies, New Bolton Center, University of Pennsylvania School of Veterinary Medicine
| | | | - Dean W. Richardson
- Department of Clinical Studies, New Bolton Center, University of Pennsylvania School of Veterinary Medicine
| | - George R. Dodge
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA,Translational Musculoskeletal Research Center, Corporal Michael J Crescenz VA Medical Center, Philadelphia, PA,Corresponding author: George R. Dodge, Ph.D., McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, 379A Stemmler Hall, 36 Street and Hamilton Walk, Philadelphia, PA 19104, Phone: (215) 573-1514, Fax: (215) 573-2133,
| |
Collapse
|
22
|
Chen H, Cui Y, Zhang D, Xie J, Zhou X. The role of fibroblast growth factor 8 in cartilage development and disease. J Cell Mol Med 2022; 26:990-999. [PMID: 35001536 PMCID: PMC8831980 DOI: 10.1111/jcmm.17174] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 12/22/2021] [Accepted: 12/28/2021] [Indexed: 02/05/2023] Open
Abstract
Fibroblast growth factor 8 (FGF‐8), also known as androgen‐induced growth factor (AIGF), is presumed to be a potent mitogenic cytokine that plays important roles in early embryonic development, brain formation and limb development. In the bone environment, FGF‐8 produced or received by chondrocyte precursor cells binds to fibroblast growth factor receptor (FGFR), causing different levels of activation of downstream signalling pathways, such as phospholipase C gamma (PLCγ)/Ca2+, RAS/mitogen‐activated protein kinase‐extracellular regulated protein kinases (RAS/MAPK‐MEK‐ERK), and Wnt‐β‐catenin‐Axin2 signalling, and ultimately controlling chondrocyte proliferation, differentiation, cell survival and migration. However, the molecular mechanism of FGF‐8 in normal or pathological cartilage remains unclear, and thus, FGF‐8 represents a novel exploratory target for studies of chondrocyte development and cartilage disease progression. In this review, studies assessing the relationship between FGF‐8 and chondrocytes that have been published in the past 5 years are systematically summarized to determine the probable mechanism and physiological effect of FGF‐8 on chondrocytes. Based on the existing research results, a therapeutic regimen targeting FGF‐8 is proposed to explore the possibility of treating chondrocyte‐related diseases.
Collapse
Affiliation(s)
- Haoran Chen
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yujia Cui
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Demao Zhang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jing Xie
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
23
|
Impact of perlecan, a core component of basement membrane, on regeneration of cartilaginous tissues. Acta Biomater 2021; 135:13-26. [PMID: 34454085 DOI: 10.1016/j.actbio.2021.08.032] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 08/02/2021] [Accepted: 08/20/2021] [Indexed: 02/03/2023]
Abstract
As an indispensable component of the extracellular matrix, perlecan (Pln) plays an essential role in cartilaginous tissue function. Although there exist studies suggesting that Pln expressed by cartilaginous tissues is critical for chondrogenesis, few papers have discussed the potential impact Pln may have on cartilage regeneration. In this review, we delineate Pln structure, biomechanical properties, and interactive ligands-which together contribute to the effect Pln has on cartilaginous tissue development. We also review how the signaling pathways of Pln affect cartilage development and scrutinize the potential application of Pln to divisions of cartilage regeneration, spanning vascularization, stem cell differentiation, and biomaterial improvement. The aim of this review is to deepen our understanding of the spatial and temporal interactions that occur between Pln and cartilaginous tissue and ultimately apply Pln in scaffold design to improve cell-based cartilage engineering and regeneration. STATEMENT OF SIGNIFICANCE: As a key component of the basement membrane, Pln plays a critical role in tissue development and repair. Recent findings suggest that Pln existing in the pericellular matrix surrounding mature chondrocytes is actively involved in cartilage regeneration and functionality. We propose that Pln is essential to developing an in vitro matrix niche within biological scaffolds for cartilage tissue engineering.
Collapse
|
24
|
McNamara JT, Huntington KE, Borys S, Jayasuriya CT, Brossay L. SHP-2 deletion in CD4Cre expressing chondrocyte precursors leads to tumor development with wrist tropism. Sci Rep 2021; 11:20006. [PMID: 34625577 PMCID: PMC8501018 DOI: 10.1038/s41598-021-99339-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 09/20/2021] [Indexed: 11/16/2022] Open
Abstract
Due to redundancy with other tyrosine phosphatases, the ubiquitously expressed tyrosine phosphatase SHP-2 (encoded by Ptpn11) is not required for T cell development. However, Ptpn11 gene deletion driven by CD4 Cre recombinase leads to cartilage tumors in the wrist. Using a fate mapping system, we demonstrate that wrist tumor development correlates with increased frequency and numbers of non-hematopoietic lineage negative CD45 negative cells with a bone chondrocyte stromal cell precursor cell (BCSP) phenotype. Importantly, the BCSP subset has a history of CD4 expression and a marked wrist location tropism, explaining why the wrist is the main site of tumor development. Mechanistically, we found that in SHP-2 absence, SOX-9 is no longer regulated, leading to an uncontrolled proliferation of the BCSP subset. Altogether, these results identify a unique subset of chondrocyte precursors tightly regulated by SHP-2. These findings underscore the need for the development of methods to therapeutically target this subset of cells, which could potentially have an impact on treatment of SHP-2 dysfunction linked debilitating diseases.
Collapse
Affiliation(s)
- Jeffrey T McNamara
- Division of Biology and Medicine, Department of Molecular Microbiology and Immunology, Brown University Alpert Medical School, Providence, RI, 02912, USA
| | - Kelsey E Huntington
- Division of Biology and Medicine, Department of Molecular Microbiology and Immunology, Brown University Alpert Medical School, Providence, RI, 02912, USA
| | - Samantha Borys
- Division of Biology and Medicine, Department of Molecular Microbiology and Immunology, Brown University Alpert Medical School, Providence, RI, 02912, USA
| | - Chathuraka T Jayasuriya
- Department of Orthopaedics, Rhode Island Hospital and Brown University Alpert Medical School, Providence, RI, USA
| | - Laurent Brossay
- Division of Biology and Medicine, Department of Molecular Microbiology and Immunology, Brown University Alpert Medical School, Providence, RI, 02912, USA.
| |
Collapse
|
25
|
Novel FGFR1 Variants Are Associated with Congenital Scoliosis. Genes (Basel) 2021; 12:genes12081126. [PMID: 34440300 PMCID: PMC8393897 DOI: 10.3390/genes12081126] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 07/07/2021] [Accepted: 07/14/2021] [Indexed: 11/21/2022] Open
Abstract
FGFR1 encodes a transmembrane cytokine receptor, which is involved in the early development of the human embryo and plays an important role in gastrulation, organ specification and patterning of various tissues. Pathogenic FGFR1 variants have been associated with Kallmann syndrome and hypogonadotropic hypogonadism. In our congenital scoliosis (CS) patient series of 424 sporadic CS patients under the framework of the Deciphering disorders Involving Scoliosis and COmorbidities (DISCO) study, we identified four unrelated patients harboring FGFR1 variants, including one frameshift and three missense variants. These variants were predicted to be deleterious by in silico prediction and conservation analysis. Signaling activities and expression levels of the mutated protein were evaluated in vitro and compared to that of the wild type (WT) FGFR1. As a result, the overall protein expressions of c.2334dupC, c.2339T>C and c.1261A>G were reduced to 43.9%, 63.4% and 77.4%, respectively. By the reporter gene assay, we observed significantly reduced activity for c.2334dupC, c.2339T>C and c.1261A>G, indicating the diminished FGFR1 signaling pathway. In conclusion, FGFR1 variants identified in our patients led to only mild disruption to protein function, caused milder skeletal and cardiac phenotypes than those reported previously.
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: 28] [Impact Index Per Article: 7.0] [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
|
Coutinho de Almeida R, Mahfouz A, Mei H, Houtman E, den Hollander W, Soul J, Suchiman E, Lakenberg N, Meessen J, Huetink K, Nelissen RGHH, Ramos YFM, Reinders M, Meulenbelt I. Identification and characterization of two consistent osteoarthritis subtypes by transcriptome and clinical data integration. Rheumatology (Oxford) 2021; 60:1166-1175. [PMID: 32885253 PMCID: PMC7937023 DOI: 10.1093/rheumatology/keaa391] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 06/04/2020] [Indexed: 01/13/2023] Open
Abstract
OBJECTIVE To identify OA subtypes based on cartilage transcriptomic data in cartilage tissue and characterize their underlying pathophysiological processes and/or clinically relevant characteristics. METHODS This study includes n = 66 primary OA patients (41 knees and 25 hips), who underwent a joint replacement surgery, from which macroscopically unaffected (preserved, n = 56) and lesioned (n = 45) OA articular cartilage were collected [Research Arthritis and Articular Cartilage (RAAK) study]. Unsupervised hierarchical clustering analysis on preserved cartilage transcriptome followed by clinical data integration was performed. Protein-protein interaction (PPI) followed by pathway enrichment analysis were done for genes significant differentially expressed between subgroups with interactions in the PPI network. RESULTS Analysis of preserved samples (n = 56) resulted in two OA subtypes with n = 41 (cluster A) and n = 15 (cluster B) patients. The transcriptomic profile of cluster B cartilage, relative to cluster A (DE-AB genes) showed among others a pronounced upregulation of multiple genes involved in chemokine pathways. Nevertheless, upon investigating the OA pathophysiology in cluster B patients as reflected by differentially expressed genes between preserved and lesioned OA cartilage (DE-OA-B genes), the chemokine genes were significantly downregulated with OA pathophysiology. Upon integrating radiographic OA data, we showed that the OA phenotype among cluster B patients, relative to cluster A, may be characterized by higher joint space narrowing (JSN) scores and low osteophyte (OP) scores. CONCLUSION Based on whole-transcriptome profiling, we identified two robust OA subtypes characterized by unique OA, pathophysiological processes in cartilage as well as a clinical phenotype. We advocate that further characterization, confirmation and clinical data integration is a prerequisite to allow for development of treatments towards personalized care with concurrently more effective treatment response.
Collapse
Affiliation(s)
- Rodrigo Coutinho de Almeida
- Department of Biomedical Data Sciences, Section Molecular Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Ahmed Mahfouz
- Delft Bioinformatics Lab, Delft University of Technology, Delft, The Netherlands.,Leiden Computational Biology Center, Leiden, The Netherlands
| | - Hailiang Mei
- Sequence Analysis Support Core, Leiden University Medical Center, Leiden, The Netherlands
| | - Evelyn Houtman
- Department of Biomedical Data Sciences, Section Molecular Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Wouter den Hollander
- Department of Biomedical Data Sciences, Section Molecular Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jamie Soul
- Skeletal Research Group, Institute of Genetic Medicine, Newcastle University, Central Parkway, Newcastle upon Tyne, UK
| | - Eka Suchiman
- Department of Biomedical Data Sciences, Section Molecular Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Nico Lakenberg
- Department of Biomedical Data Sciences, Section Molecular Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jennifer Meessen
- Department of Biomedical Data Sciences, Section Molecular Epidemiology, Leiden University Medical Center, Leiden, The Netherlands.,Department Orthopaedics, Leiden University Medical Center, Leiden, The Netherlands
| | - Kasper Huetink
- Department Orthopaedics, Leiden University Medical Center, Leiden, The Netherlands
| | - Rob G H H Nelissen
- Department Orthopaedics, Leiden University Medical Center, Leiden, The Netherlands
| | - Yolande F M Ramos
- Department of Biomedical Data Sciences, Section Molecular Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Marcel Reinders
- Department of Biomedical Data Sciences, Section Molecular Epidemiology, Leiden University Medical Center, Leiden, The Netherlands.,Delft Bioinformatics Lab, Delft University of Technology, Delft, The Netherlands.,Leiden Computational Biology Center, Leiden, The Netherlands
| | - Ingrid Meulenbelt
- Department of Biomedical Data Sciences, Section Molecular Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
28
|
Guasto A, Cormier-Daire V. Signaling Pathways in Bone Development and Their Related Skeletal Dysplasia. Int J Mol Sci 2021; 22:4321. [PMID: 33919228 PMCID: PMC8122623 DOI: 10.3390/ijms22094321] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 04/12/2021] [Accepted: 04/19/2021] [Indexed: 12/19/2022] Open
Abstract
Bone development is a tightly regulated process. Several integrated signaling pathways including HH, PTHrP, WNT, NOTCH, TGF-β, BMP, FGF and the transcription factors SOX9, RUNX2 and OSX are essential for proper skeletal development. Misregulation of these signaling pathways can cause a large spectrum of congenital conditions categorized as skeletal dysplasia. Since the signaling pathways involved in skeletal dysplasia interact at multiple levels and have a different role depending on the time of action (early or late in chondrogenesis and osteoblastogenesis), it is still difficult to precisely explain the physiopathological mechanisms of skeletal disorders. However, in recent years, significant progress has been made in elucidating the mechanisms of these signaling pathways and genotype-phenotype correlations have helped to elucidate their role in skeletogenesis. Here, we review the principal signaling pathways involved in bone development and their associated skeletal dysplasia.
Collapse
Affiliation(s)
- Alessandra Guasto
- Imagine Institute, Université de Paris, Clinical Genetics, INSERM UMR 1163, Necker Enfants Malades Hospital, 75015 Paris, France;
| | - Valérie Cormier-Daire
- Imagine Institute, Université de Paris, Clinical Genetics, INSERM UMR 1163, Necker Enfants Malades Hospital, 75015 Paris, France;
- Centre de Référence Pour Les Maladies Osseuses Constitutionnelles, Service de Génétique Clinique, AP-HP, Hôpital Necker-Enfants Malades, 75015 Paris, France
| |
Collapse
|
29
|
Wang H, Yang J, Zhang K, Liu J, Li Y, Su W, Song N. Advances of Fibroblast Growth Factor/Receptor Signaling Pathway in Hepatocellular Carcinoma and its Pharmacotherapeutic Targets. Front Pharmacol 2021; 12:650388. [PMID: 33935756 PMCID: PMC8082422 DOI: 10.3389/fphar.2021.650388] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 03/11/2021] [Indexed: 12/12/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is a type of primary liver cancer with poor prognosis, and its incidence and mortality rate are increasing worldwide. It is refractory to conventional chemotherapy and radiotherapy owing to its high tumor heterogeneity. Accumulated genetic alterations and aberrant cell signaling pathway have been characterized in HCC. The fibroblast growth factor (FGF) family and their receptors (FGFRs) are involved in diverse biological activities, including embryonic development, proliferation, differentiation, survival, angiogenesis, and migration, etc. Data mining results of The Cancer Genome Atlas demonstrate high levels of FGF and/or FGFR expression in HCC tumors compared with normal tissues. Moreover, substantial evidence indicates that the FGF/FGFR signaling axis plays an important role in various mechanisms that contribute to HCC development. At present, several inhibitors targeting FGF/FGFR, such as multikinase inhibitors, specific FGFR4 inhibitors, and FGF ligand traps, exhibit antitumor activity in preclinical or early development phases in HCC. In this review, we summarize the research progress regarding the molecular implications of FGF/FGFR-mediated signaling and the development of FGFR-targeted therapeutics in hepatocarcinogenesis.
Collapse
Affiliation(s)
- Haijun Wang
- Key Laboratory of Clinical Molecular Pathology, Department of Pathology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China.,School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Jie Yang
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Ke Zhang
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Jia Liu
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Yushan Li
- School of Public Health, Xinxiang Medical University, Xinxiang, China
| | - Wei Su
- Key Laboratory of Clinical Molecular Pathology, Department of Pathology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Na Song
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China.,Institute of Precision Medicine, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
30
|
Faienza MF, Chiarito M, Brunetti G, D'Amato G. Growth plate gene involment and isolated short stature. Endocrine 2021; 71:28-34. [PMID: 32504378 DOI: 10.1007/s12020-020-02362-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 05/20/2020] [Indexed: 12/18/2022]
Abstract
PURPOSE Short stature is a common clinical presentation, thus it is widely accepted that it is a polygenic trait. However, genome wide association and next generation sequencing studies have recently challenged this view, suggesting that many of the children classified as idiopathic short stature could instead have monogenic defects. Linear growth is determined primarily by chondrogenesis at the growth plate. This process results from chondrocyte proliferation, hypertrophy, and extracellular matrix secretion, and it is perfectly coordinated by complex networks of local paracrine and endocrine factors. Alterations in genes which control growth plate development can explain a large number of cases of isolated short stature, allowing an etiological diagnosis. METHODS/RESULTS We reviewed recent data on the genetic alterations in fundamental cellular processes, paracrine signaling, and cartilage matrix formation associated with impaired growth plate chondrogenesis. In particular we focused on growth plate gene involvement in nonsyndromic short stature. CONCLUSIONS The identification of genetic basis of growth failure will have a significant impact on the care of children affected with short stature.
Collapse
Affiliation(s)
- Maria Felicia Faienza
- Paediatric Unit, Department of Biomedical Sciences and Human Oncology, University of Bari "Aldo Moro", Bari, Italy.
| | - Mariangela Chiarito
- Paediatric Unit, Department of Biomedical Sciences and Human Oncology, University of Bari "Aldo Moro", Bari, Italy
| | - Giacomina Brunetti
- Department of Basic Medical Sciences, Neuroscience and Sense Organs, Section of Human Anatomy and Histology, University of Bari "A. Moro", Bari, Italy
| | | |
Collapse
|
31
|
Zhou E, Lui J. Physiological regulation of bone length and skeletal proportion in mammals. Exp Physiol 2020; 106:389-395. [PMID: 33369789 DOI: 10.1113/ep089086] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 12/11/2020] [Indexed: 11/08/2022]
Abstract
NEW FINDINGS What is the topic of this review? Mechanisms regulating bone length and skeletal proportions What advances does it highlight? The study of differential bone length between leg and finger bones, metatarsals of the Egyptian jerboa and genomic analysis of giraffes. ABSTRACT Among mammalian species, skeletal structures vary greatly in size and shape, leading to a dramatic variety of body sizes and proportions. How different bones grow to different lengths, whether among different species, different individuals of the same species, or even in different anatomical parts of our the body, has always been a fascinating subject of research in biology and physiology. In the current review, we focus on some of the recent advances in the field and discuss how these provided important new insights into the mechanisms regulating bone length and skeletal proportions.
Collapse
Affiliation(s)
- Elaine Zhou
- Section on Growth and Development, National Institute of Child Health and Human Development, NIH, Bethesda, Maryland, USA
| | - Julian Lui
- Section on Growth and Development, National Institute of Child Health and Human Development, NIH, Bethesda, Maryland, USA
| |
Collapse
|
32
|
Chen J, Hendriks M, Chatzis A, Ramasamy SK, Kusumbe AP. Bone Vasculature and Bone Marrow Vascular Niches in Health and Disease. J Bone Miner Res 2020; 35:2103-2120. [PMID: 32845550 DOI: 10.1002/jbmr.4171] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 07/21/2020] [Accepted: 08/05/2020] [Indexed: 12/20/2022]
Abstract
Bone vasculature and bone marrow vascular niches supply oxygen, nutrients, and secrete angiocrine factors required for the survival, maintenance, and self-renewal of stem and progenitor cells. In the skeletal system, vasculature creates nurturing niches for bone and blood-forming stem cells. Blood vessels regulate hematopoiesis and drive bone formation during development, repair, and regeneration. Dysfunctional vascular niches induce skeletal aging, bone diseases, and hematological disorders. Recent cellular and molecular characterization of the bone marrow microenvironment has provided unprecedented insights into the complexity, heterogeneity, and functions of the bone vasculature and vascular niches. The bone vasculature is composed of distinct vessel subtypes that differentially regulate osteogenesis, hematopoiesis, and disease conditions in bones. Further, bone marrow vascular niches supporting stem cells are often complex microenvironments involving multiple different cell populations and vessel subtypes. This review provides an overview of the emerging vascular cell heterogeneity in bone and the new roles of the bone vasculature and associated vascular niches in health and disease. © 2020 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Junyu Chen
- Tissue and Tumor Microenvironments Group, The Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Michelle Hendriks
- Institute of Clinical Sciences, Imperial College London, London, UK
- MRC London Institute of Medical Sciences, Imperial College London, London, UK
| | - Alexandros Chatzis
- Tissue and Tumor Microenvironments Group, The Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Saravana K Ramasamy
- Institute of Clinical Sciences, Imperial College London, London, UK
- MRC London Institute of Medical Sciences, Imperial College London, London, UK
| | - Anjali P Kusumbe
- Tissue and Tumor Microenvironments Group, The Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| |
Collapse
|
33
|
Boylan M, Anderson MJ, Ornitz DM, Lewandoski M. The Fgf8 subfamily (Fgf8, Fgf17 and Fgf18) is required for closure of the embryonic ventral body wall. Development 2020; 147:dev189506. [PMID: 32907848 PMCID: PMC7595690 DOI: 10.1242/dev.189506] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 08/28/2020] [Indexed: 12/26/2022]
Abstract
The closure of the embryonic ventral body wall in amniotes is an important morphogenetic event and is essential for life. Defects in human ventral wall closure are a major class of birth defect and a significant health burden. Despite this, very little is understood about how the ventral body wall is formed. Here, we show that fibroblast growth factor (FGF) ligands FGF8, FGF17 and FGF18 are essential for this process. Conditional mouse mutants for these genes display subtle migratory defects in the abdominal muscles of the ventral body wall and an enlarged umbilical ring, through which the internal organs are extruded. By refining where and when these genes are required using different Cre lines, we show that Fgf8 and Fgf17 are required in the presomitic mesoderm, whereas Fgf18 is required in the somites. This study identifies complex and multifactorial origins of ventral wall defects and has important implications for understanding their origins during embryonic development.
Collapse
Affiliation(s)
- Michael Boylan
- Cancer and Developmental Biology Lab, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA
| | - Matthew J Anderson
- Cancer and Developmental Biology Lab, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA
| | - David M Ornitz
- Department of Developmental Biology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Mark Lewandoski
- Cancer and Developmental Biology Lab, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA
| |
Collapse
|
34
|
Wu Y, Hong Z, Xu W, Chen J, Wang Q, Chen J, Ni W, Mei Z, Xie Z, Ma Y, Wang J, Lu J, Chen C, Fan S, Shen S. Circular RNA circPDE4D Protects against Osteoarthritis by Binding to miR-103a-3p and Regulating FGF18. Mol Ther 2020; 29:308-323. [PMID: 33125858 PMCID: PMC7791010 DOI: 10.1016/j.ymthe.2020.09.002] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 06/05/2020] [Accepted: 09/01/2020] [Indexed: 01/22/2023] Open
Abstract
Osteoarthritis (OA) is a common, age-related, and painful disease characterized by cartilage destruction, osteophyte formation, and synovial hyperplasia. This study revealed that circPDE4D, a circular RNA derived from human linear PDE4D, plays a critical role in maintaining the extracellular cellular matrix (ECM) during OA progression. circPDE4D was significantly downregulated in OA cartilage tissues and during stimulation with inflammatory cytokines. The knockdown of circPDE4D predominantly contributed to Aggrecan loss and the upregulation of matrix catabolic enzymes, including MMP3, MMP13, ADAMTS4, and ADAMTS5, but not proliferation or apoptosis. In a murine model of destabilization of the medial meniscus (DMM), the intraarticular injection of circPDE4D alleviated DMM-induced cartilage impairments. Mechanistically, we found that circPDE4D exerted its effect by acting as a sponge for miR-103a-3p and thereby regulated FGF18 expression, which is a direct target of miR-103a-3p. In conclusion, our findings highlight a novel protective role of circPDE4D in OA pathogenesis and indicate that the targeting of the circPDE4D-miR-103a-3p-FGF18 axis might provide a potential and promising approach for OA therapy.
Collapse
Affiliation(s)
- Yizheng Wu
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Zhejiang, China; Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Zhejiang, China
| | - Zhenghua Hong
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Zhejiang, China; Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Zhejiang, China; Department of Orthopaedic Surgery, Taizhou Hospital of Zhejiang Province, Zhejiang University School of Medicine, Zhejiang, China
| | - Wenbin Xu
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Zhejiang, China; Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Zhejiang, China
| | - Junxin Chen
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Zhejiang, China; Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Zhejiang, China
| | - Qingxin Wang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Zhejiang, China; Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Zhejiang, China; Department of Orthopaedic Surgery, China Coast Guard Hospital of the People's Armed Police Force, Zhejiang, China
| | - Jiaxin Chen
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Zhejiang, China
| | - Weiyu Ni
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Zhejiang, China; Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Zhejiang, China
| | - Zixuan Mei
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Zhejiang, China; Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Zhejiang, China
| | - Ziang Xie
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Zhejiang, China; Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Zhejiang, China
| | - Yan Ma
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Zhejiang, China; Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Zhejiang, China
| | - Jiying Wang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Zhejiang, China; Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Zhejiang, China
| | - Jianhong Lu
- Department of Clinical Laboratory, China Coast Guard Hospital of the People's Armed Police Force, Zhejiang, China
| | - Chao Chen
- Department of Orthopaedic Surgery, School of Traditional Chinese Medicine, Southern Medical University, Guangdong, China
| | - Shunwu Fan
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Zhejiang, China; Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Zhejiang, China.
| | - Shuying Shen
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Zhejiang, China; Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Zhejiang, China.
| |
Collapse
|
35
|
Chiarelli F, Primavera M, Mastromauro C. Evaluation and management of a child with short stature. Minerva Pediatr 2020; 72:452-461. [PMID: 32686926 DOI: 10.23736/s0026-4946.20.05980-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Growth monitoring is a fundamental approach to evaluate a child's health and it is part of preventive programs to timely identify and treat a possible disease. Height and weight measurements, calculation of height velocity over time are main instruments to discover pathological deviations. Short stature is defined as a height that is greater than or equal 2 standard deviations (SDS) below the mean height for reference children comparable for sex and age. According to the International Classification of Pediatric Endocrine Diagnosis (ICPED) the possible causes of short stature could be divided into three groups: primary growth disorders (intrinsic diseases of the growth plate), secondary growth disorders (diseases that interfere on the growth plate setting) and the idiopathic short stature in which no possible cause is identified. The etiology of short stature is not always a disease, but it could be a variant of normal growth. Furthermore, to date there are new advances in the genetic causes of short stature. A detailed evaluation of a child with growth impairment should include an accurate history, a standardize physical examination, general and specific laboratory evaluations, radiologic investigations and genetic testing. Short stature could represent an important threat for physical and psychological health in a child, so a prompt identification of abnormal growth deviations offers the possibility to early treat the possible cause of shortness. This review aimed to discuss a practical approach to a child with short stature on the bases of the most recent scientific evidence.
Collapse
|
36
|
Abstract
PURPOSE OF REVIEW Bone elongation is a complex process driven by multiple intrinsic (hormones, growth factors) and extrinsic (nutrition, environment) variables. Bones grow in length by endochondral ossification in cartilaginous growth plates at ends of developing long bones. This review provides an updated overview of the important factors that influence this process. RECENT FINDINGS Insulin-like growth factor-1 (IGF-1) is the major hormone required for growth and a drug for treating pediatric skeletal disorders. Temperature is an underrecognized environmental variable that also impacts linear growth. This paper reviews the current state of knowledge regarding the interaction of IGF-1 and environmental factors on bone elongation. Understanding how internal and external variables regulate bone lengthening is essential for developing and improving treatments for an array of bone elongation disorders. Future studies may benefit from understanding how these unique relationships could offer realistic new approaches for increasing bone length in different growth-limiting conditions.
Collapse
Affiliation(s)
- Holly L Racine
- Department of Natural Sciences and Mathematics, West Liberty University, West Liberty, WV, 26074, USA
| | - Maria A Serrat
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, 1 John Marshall Drive, Huntington, WV, 25755, USA.
- Department of Clinical and Translational Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, 25755, USA.
- Department of Orthopaedics, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, 25755, USA.
| |
Collapse
|
37
|
Kagan BJ, Rosello‐Diez A. Integrating levels of bone growth control: From stem cells to body proportions. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2020; 10:e384. [DOI: 10.1002/wdev.384] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 02/09/2020] [Accepted: 04/16/2020] [Indexed: 12/23/2022]
Affiliation(s)
- Brett J. Kagan
- Australian Regenerative Medicine Institute Monash University Clayton Australia
| | | |
Collapse
|
38
|
Evaluation of Relationship Between Common Variants in FGF18 Gene and Knee Osteoarthritis Susceptibility. Arch Med Res 2020; 51:76-81. [PMID: 32109713 DOI: 10.1016/j.arcmed.2019.12.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 11/26/2019] [Accepted: 12/10/2019] [Indexed: 11/21/2022]
Abstract
BACKGROUND AND AIMS Osteoarthritis (OA) is the most common type of arthritis disease. Previous etiological studies indicated that both environmental and genetic factors play important roles in the occurrence and development of knee OA (KOA). In the present study, we aimed to investigate the association between the FGF18 gene and susceptibility to KOA in Han Chinese population. METHODS A total of 2,556 unrelated Han Chinese individuals, including 866 KOA patients and 1,688 healthy controls, were recruited. Nine tag SNPs located within the gene region of FGF18 were selected for genotyping. Logistic models were fitted for single marker-based association analyses. Age, gender and BMI were included in each model as covariates. To investigate the functional consequences of significant SNP, we extracted expression quantitative trait loci (eQTL) data from the GTEx database. RESULTS SNP rs3884606 was significantly associated with the risk of KOA (OR [95% CI] = 1.25 [1.11-1.41], p = 0.0002) after adjusting for age, gender and BMI. The G allele was significantly associated with an increased risk of KOA. No significant eQTL signals could be identified for SNP rs3884606 on FGF18. CONCLUSION In this study, we identified a genetic polymorphism in FGF18 that was significantly associated with the risk of KOA based on samples with Chinese Han ancestry. Our findings replicated the recent GWAS report and supported an association between KOA and FGF18.
Collapse
|
39
|
Ng J, Little CB, Woods S, Whittle S, Lee FY, Gronthos S, Mukherjee S, Hunter DJ, Worthley DL. Stem cell-directed therapies for osteoarthritis: The promise and the practice. Stem Cells 2020; 38:477-486. [PMID: 31837053 DOI: 10.1002/stem.3139] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 11/13/2019] [Accepted: 11/15/2019] [Indexed: 12/15/2022]
Abstract
Osteoarthritis (OA) is a disease of an entire synovial joint characterized by clinical symptoms and distortion of joint tissues, including cartilage, muscles, ligaments, and bone. Although OA is a disease of all joint tissues, it is a defined accessible compartment and is thus amenable to topical surgical and regenerative therapies, including stem cells. All tissues arise from stem progenitor cells, and the relative capacity of different cellular compartments, and different individuals, to renew tissues into adulthood may be important in the onset of many different degenerative diseases. OA is driven by both mechanical and inflammatory factors, but how these factors affect the proliferation and differentiation of cells into cartilage in vivo is largely unknown. Indeed, our very basic understanding of the physiological cellular kinetics and biology of the stem-progenitor cell unit of the articular cartilage, and how this is influenced by mechano-inflammatory injury, is largely unknown. OA seems, rather deceptively, to be the low-hanging fruit for stem cell therapy. Without the basic understanding of the stem cell and progenitor unit that generate and maintain articular cartilage in vivo, we will continue to waste opportunities to both prevent and manage this disease. In this review, we discuss the biology of chondrogenesis, the stem cell populations that support articular cartilage in health and disease, and future opportunities afforded through the translation of basic articular chondrocyte stem cell biology into new clinical therapies.
Collapse
Affiliation(s)
- Jia Ng
- Precision Medicine, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia.,Department of Medicine, University of Adelaide, Adelaide, South Australia, Australia
| | - Christopher B Little
- Northern Clinical School, University of Sydney, St. Leonards, Sydney, New South Wales, Australia.,Raymond Purves Bone & Joint Research Laboratories, Kolling Institute, St. Leonards, Sydney, New South Wales, Australia
| | - Susan Woods
- Precision Medicine, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia.,Department of Medicine, University of Adelaide, Adelaide, South Australia, Australia
| | - Samuel Whittle
- Department of Medicine, University of Adelaide, Adelaide, South Australia, Australia
| | - Francis Y Lee
- Rheumatology Department, Royal North Shore Hospital, Institute of Bone and Joint Research, Kolling Institute, University of Sydney, St. Leonards, New South Wales, Australia
| | - Stan Gronthos
- Precision Medicine, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia.,Mesenchymal Stem Cell Laboratory, University of Adelaide, Adelaide, South Australia, Australia
| | - Siddhartha Mukherjee
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York
| | - David J Hunter
- Northern Clinical School, University of Sydney, St. Leonards, Sydney, New South Wales, Australia
| | - Daniel L Worthley
- Precision Medicine, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| |
Collapse
|
40
|
Lopes FM, Roberts NA, Zeef LAH, Gardiner NJ, Woolf AS. Overactivity or blockade of transforming growth factor-β each generate a specific ureter malformation. J Pathol 2019; 249:472-484. [PMID: 31400222 PMCID: PMC6900140 DOI: 10.1002/path.5335] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 07/19/2019] [Accepted: 08/06/2019] [Indexed: 12/13/2022]
Abstract
Transforming growth factor-β (TGFβ) has been reported to be dysregulated in malformed ureters. There exists, however, little information on whether altered TGFβ levels actually perturb ureter development. We therefore hypothesised that TGFβ has functional effects on ureter morphogenesis. Tgfb1, Tgfb2 and Tgfb3 transcripts coding for TGFβ ligands, as well as Tgfbr1 and Tgfbr2 coding for TGFβ receptors, were detected by quantitative polymerase chain reaction in embryonic mouse ureters collected over a wide range of stages. As assessed by in situ hybridisation and immunohistochemistry, the two receptors were detected in embryonic urothelia. Next, TGFβ1 was added to serum-free cultures of embryonic day 15 mouse ureters. These organs contain immature smooth muscle and urothelial layers and their in vivo potential to grow and acquire peristaltic function can be replicated in serum-free organ culture. Such organs therefore constitute a suitable developmental stage with which to define roles of factors that affect ureter growth and functional differentiation. Exogenous TGFβ1 inhibited growth of the ureter tube and generated cocoon-like dysmorphogenesis. RNA sequencing suggested that altered levels of transcripts encoding certain fibroblast growth factors (FGFs) followed exposure to TGFβ. In serum-free organ culture exogenous FGF10 but not FGF18 abrogated certain dysmorphic effects mediated by exogenous TGFβ1. To assess whether an endogenous TGFβ axis functions in developing ureters, embryonic day 15 explants were exposed to TGFβ receptor chemical blockade; growth of the ureter was enhanced, and aberrant bud-like structures arose from the urothelial tube. The muscle layer was attenuated around these buds, and peristalsis was compromised. To determine whether TGFβ effects were limited to one stage, explants of mouse embryonic day 13 ureters, more primitive organs, were exposed to exogenous TGFβ1, again generating cocoon-like structures, and to TGFβ receptor blockade, again generating ectopic buds. As for the mouse studies, immunostaining of normal embryonic human ureters detected TGFβRI and TGFβRII in urothelia. Collectively, these observations reveal unsuspected regulatory roles for endogenous TGFβ in embryonic ureters, fine-tuning morphogenesis and functional differentiation. Our results also support the hypothesis that the TGFβ up-regulation reported in ureter malformations impacts on pathobiology. Further experiments are needed to unravel the intracellular signalling mechanisms involved in these dysmorphic responses. © 2019 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Filipa M Lopes
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and HealthUniversity of ManchesterManchesterUK
| | - Neil A Roberts
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and HealthUniversity of ManchesterManchesterUK
| | - Leo AH Zeef
- The Bioinformatics Core FacilityUniversity of ManchesterManchesterUK
| | - Natalie J Gardiner
- Division of Diabetes, Endocrinology and Gastroenterology, School of Medical Sciences, Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
| | - Adrian S Woolf
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and HealthUniversity of ManchesterManchesterUK
- Royal Manchester Children's HospitalManchester University NHS Foundation Trust, Manchester Academic Health Science CentreManchesterUK
| |
Collapse
|
41
|
McKenzie J, Smith C, Karuppaiah K, Langberg J, Silva MJ, Ornitz DM. Osteocyte Death and Bone Overgrowth in Mice Lacking Fibroblast Growth Factor Receptors 1 and 2 in Mature Osteoblasts and Osteocytes. J Bone Miner Res 2019; 34:1660-1675. [PMID: 31206783 PMCID: PMC6744314 DOI: 10.1002/jbmr.3742] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 03/27/2019] [Accepted: 04/05/2019] [Indexed: 01/11/2023]
Abstract
Fibroblast growth factor (FGF) signaling pathways have well-established roles in skeletal development, with essential functions in both chondrogenesis and osteogenesis. In mice, previous conditional knockout studies suggested distinct roles for FGF receptor 1 (FGFR1) signaling at different stages of osteogenesis and a role for FGFR2 in osteoblast maturation. However, the potential for redundancy among FGFRs and the mechanisms and consequences of stage-specific osteoblast lineage regulation were not addressed. Here, we conditionally inactivate Fgfr1 and Fgfr2 in mature osteoblasts with an Osteocalcin (OC)-Cre or Dentin matrix protein 1 (Dmp1)-CreER driver. We find that young mice lacking both receptors or only FGFR1 are phenotypically normal. However, between 6 and 12 weeks of age, OC-Cre Fgfr1/Fgfr2 double- and Fgfr1 single-conditional knockout mice develop a high bone mass phenotype with increased periosteal apposition, increased and disorganized endocortical bone with increased porosity, and biomechanical properties that reflect increased bone mass but impaired material properties. Histopathological and gene expression analyses show that this phenotype is preceded by a striking loss of osteocytes and accompanied by activation of the Wnt/β-catenin signaling pathway. These data identify a role for FGFR1 signaling in mature osteoblasts/osteocytes that is directly or indirectly required for osteocyte survival and regulation of bone mass during postnatal bone growth. © 2019 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Jennifer McKenzie
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, USA.,Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Craig Smith
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Kannan Karuppaiah
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Joshua Langberg
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Matthew J Silva
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, USA.,Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - David M Ornitz
- Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, MO, USA.,Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
42
|
Hagan AS, Boylan M, Smith C, Perez-Santamarina E, Kowalska K, Hung IH, Lewis RM, Hajihosseini MK, Lewandoski M, Ornitz DM. Generation and validation of novel conditional flox and inducible Cre alleles targeting fibroblast growth factor 18 (Fgf18). Dev Dyn 2019; 248:882-893. [PMID: 31290205 PMCID: PMC7029619 DOI: 10.1002/dvdy.85] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 06/24/2019] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Fibroblast growth factor 18 (FGF18) functions in the development of several tissues, including the lung, limb bud, palate, skeleton, central nervous system, and hair follicle. Mice containing a germline knockout of Fgf18 (Fgf18 -/- ) die shortly after birth. Postnatally, FGF18 is being evaluated for pathogenic roles in fibrosis and several types of cancer. The specific cell types that express FGF18 have been difficult to identify, and the function of FGF18 in postnatal development and tissue homeostasis has been hampered by the perinatal lethality of Fgf18 null mice. RESULTS We engineered a floxed allele of Fgf18 (Fgf18 flox ) that allows conditional gene inactivation and a CreERT2 knockin allele (Fgf18 CreERT2 ) that allows the precise identification of cells that express Fgf18 and their lineage. We validated the Fgf18 flox allele by targeting it in mesenchymal tissue and primary mesoderm during embryonic development, resulting in similar phenotypes to those observed in Fgf18 null mice. We also use the Fgf18 CreERT2 allele, in combination with a conditional fluorescent reporter to confirm known and identify new sites of Fgf18 expression. CONCLUSION These alleles will be useful to investigate FGF18 function during organogenesis and tissue homeostasis, and to target specific cell lineages at embryonic and postnatal time points.
Collapse
Affiliation(s)
- Andrew S. Hagan
- Department of Developmental Biology, Washington University School of Medicine, Saint Louis, Missouri
| | - Michael Boylan
- Cancer and Developmental Biology Lab, National Cancer Institute, National Institutes of Health, Frederick, Maryland
| | - Craig Smith
- Department of Developmental Biology, Washington University School of Medicine, Saint Louis, Missouri
| | | | - Karolina Kowalska
- School of Biological Sciences, University of East Anglia, Norwich, UK
| | - Irene H. Hung
- Department of Neurobiology & Anatomy, University of Utah School of Medicine, Salt Lake City, Utah
| | - Renate M. Lewis
- Department of Neurology, Washington University School of Medicine, Saint Louis, Missouri
| | | | - Mark Lewandoski
- Cancer and Developmental Biology Lab, National Cancer Institute, National Institutes of Health, Frederick, Maryland
| | - David M. Ornitz
- Department of Developmental Biology, Washington University School of Medicine, Saint Louis, Missouri
| |
Collapse
|
43
|
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
|
44
|
Shi R, Huang Y, Ma C, Wu C, Tian W. Current advances for bone regeneration based on tissue engineering strategies. Front Med 2018; 13:160-188. [PMID: 30047029 DOI: 10.1007/s11684-018-0629-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 12/14/2017] [Indexed: 01/07/2023]
Abstract
Bone tissue engineering (BTE) is a rapidly developing strategy for repairing critical-sized bone defects to address the unmet need for bone augmentation and skeletal repair. Effective therapies for bone regeneration primarily require the coordinated combination of innovative scaffolds, seed cells, and biological factors. However, current techniques in bone tissue engineering have not yet reached valid translation into clinical applications because of several limitations, such as weaker osteogenic differentiation, inadequate vascularization of scaffolds, and inefficient growth factor delivery. Therefore, further standardized protocols and innovative measures are required to overcome these shortcomings and facilitate the clinical application of these techniques to enhance bone regeneration. Given the deficiency of comprehensive studies in the development in BTE, our review systematically introduces the new types of biomimetic and bifunctional scaffolds. We describe the cell sources, biology of seed cells, growth factors, vascular development, and the interactions of relevant molecules. Furthermore, we discuss the challenges and perspectives that may propel the direction of future clinical delivery in bone regeneration.
Collapse
Affiliation(s)
- Rui Shi
- Institute of Traumatology and Orthopaedics, Beijing Laboratory of Biomedical Materials, Beijing Jishuitan Hospital, Beijing, 100035, China
| | - Yuelong Huang
- Department of Spine Surgery of Beijing Jishuitan Hospital, The Fourth Clinical Medical College of Peking University, Beijing, 100035, China
| | - Chi Ma
- Institute of Traumatology and Orthopaedics, Beijing Laboratory of Biomedical Materials, Beijing Jishuitan Hospital, Beijing, 100035, China
| | - Chengai Wu
- Institute of Traumatology and Orthopaedics, Beijing Laboratory of Biomedical Materials, Beijing Jishuitan Hospital, Beijing, 100035, China
| | - Wei Tian
- Institute of Traumatology and Orthopaedics, Beijing Laboratory of Biomedical Materials, Beijing Jishuitan Hospital, Beijing, 100035, China. .,Department of Spine Surgery of Beijing Jishuitan Hospital, The Fourth Clinical Medical College of Peking University, Beijing, 100035, China.
| |
Collapse
|
45
|
Schmidt L, Taiyab A, Melvin VS, Jones KL, Williams T. Increased FGF8 signaling promotes chondrogenic rather than osteogenic development in the embryonic skull. Dis Model Mech 2018; 11:dmm031526. [PMID: 29752281 PMCID: PMC6031357 DOI: 10.1242/dmm.031526] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Accepted: 05/01/2018] [Indexed: 12/13/2022] Open
Abstract
The bones of the cranial vault are formed directly from mesenchymal cells through intramembranous ossification rather than via a cartilage intermediate. Formation and growth of the skull bones involves the interaction of multiple cell-cell signaling pathways, with fibroblast growth factors (FGFs) and their receptors exerting a prominent influence. Mutations within the FGF signaling pathway are the most frequent cause of craniosynostosis, which is a common human craniofacial developmental abnormality characterized by the premature fusion of the cranial sutures. Here, we have developed new mouse models to investigate how different levels of increased FGF signaling can affect the formation of the calvarial bones and associated sutures. Whereas moderate Fgf8 overexpression resulted in delayed ossification followed by craniosynostosis of the coronal suture, higher Fgf8 levels promoted a loss of ossification and favored cartilage over bone formation across the skull. By contrast, endochondral bones were still able to form and ossify in the presence of increased levels of Fgf8, although the growth and mineralization of these bones were affected to varying extents. Expression analysis demonstrated that abnormal skull chondrogenesis was accompanied by changes in the genes required for Wnt signaling. Moreover, further analysis indicated that the pathology was associated with decreased Wnt signaling, as the reduction in ossification could be partially rescued by halving Axin2 gene dosage. Taken together, these findings indicate that mesenchymal cells of the skull are not fated to form bone, but can be forced into a chondrogenic fate through the manipulation of FGF8 signaling. These results have implications for evolution of the different methods of ossification as well as for therapeutic intervention in craniosynostosis.
Collapse
Affiliation(s)
- Linnea Schmidt
- Program of Reproductive Sciences and Integrated Physiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Craniofacial Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Aftab Taiyab
- Department of Craniofacial Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Vida Senkus Melvin
- Department of Craniofacial Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Kenneth L Jones
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Trevor Williams
- Department of Craniofacial Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Children's Hospital Colorado, Aurora, CO 80045, USA
| |
Collapse
|
46
|
Fischer J, Knoch N, Sims T, Rosshirt N, Richter W. Time-dependent contribution of BMP, FGF, IGF, and HH signaling to the proliferation of mesenchymal stroma cells during chondrogenesis. J Cell Physiol 2018; 233:8962-8970. [PMID: 29856487 DOI: 10.1002/jcp.26832] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 05/08/2018] [Indexed: 12/24/2022]
Abstract
Early loss of up to 50% of cells is common for in vitro chondrogenesis of mesenchymal stromal cells (MSC) in pellet culture, reducing the efficacy and the tissue yield for cartilage engineering. Enhanced proliferation could compensate for this unwanted effect, but relevant signaling pathways remain largely unknown. The aim of this study was to identify the contribution of bone morphogenetic protein (BMP), fibroblast growth factor (FGF), insulin-like growth factor (IGF), and hedgehog (HH) signaling toward cell proliferation during chondrogenesis and investigate whether a further mitogenic stimulation is possible and promising. Human MSC were subjected to chondrogenesis in the presence or absence of pathway inhibitors or activators up to Day 14 or from Days 14 to 28, before proliferation, DNA and proteoglycan content were quantified. [3H]-thymidine incorporation revealed arrest of proliferation on Day 3, after which cell division was reinitiated. Although BMP signaling was essential for proliferation throughout chondrogenesis, IGF signaling was relevant only up to Day 14. In contrast, FGF and HH signaling drove proliferation only from Day 14 onward. Early BMP4, IGF-1, or FGF18 treatment neither prevented early cell loss nor allowed further mitogenic stimulation. However, application of the HH-agonist purmorphamine from Day 14 increased proliferation 1.44-fold (p < 0.05) and late BMP4-application enhanced the DNA and proteoglycan content, with significant effects on tissue yield. Conclusively, a differential and phase-dependent contribution of the four pathways toward proliferation was uncovered and BMP4 treatment was promising to enhance tissue yield. Culture forms less prone to size limitations by nutrient/oxygen gradients and a focus on early apoptosis prevention may be considered as the next steps to further enhance chondrocyte formation from MSC.
Collapse
Affiliation(s)
- Jennifer Fischer
- Research Centre for Experimental Orthopedics, Orthopaedic University Hospital Heidelberg, Heidelberg, Germany
| | - Natalie Knoch
- Research Centre for Experimental Orthopedics, Orthopaedic University Hospital Heidelberg, Heidelberg, Germany
| | - Tanja Sims
- Research Centre for Experimental Orthopedics, Orthopaedic University Hospital Heidelberg, Heidelberg, Germany
| | - Nils Rosshirt
- Department of Orthopedics, Trauma Surgery and Spinal Cord Injury, Orthopaedic University Hospital Heidelberg, Heidelberg, Germany
| | - Wiltrud Richter
- Research Centre for Experimental Orthopedics, Orthopaedic University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
47
|
Moore ER, Jacobs CR. The primary cilium as a signaling nexus for growth plate function and subsequent skeletal development. J Orthop Res 2018; 36:533-545. [PMID: 28901584 PMCID: PMC5839937 DOI: 10.1002/jor.23732] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 09/07/2017] [Indexed: 02/04/2023]
Abstract
The primary cilium is a solitary, antenna-like sensory organelle with many important roles in cartilage and bone development, maintenance, and function. The primary cilium's potential role as a signaling nexus in the growth plate makes it an attractive therapeutic target for diseases and disorders associated with bone development and maintenance. Many signaling pathways that are mediated by the cilium-such as Hh, Wnt, Ihh/PTHrP, TGFβ, BMP, FGF, and Notch-are also known to influence endochondral ossification, primarily by directing growth plate formation and chondrocyte behavior. Although a few studies have demonstrated that these signaling pathways can be directly tied to the primary cilium, many pathways have yet to be evaluated in context of the cilium. This review serves to bridge this knowledge gap in the literature, as well as discuss the cilium's importance in the growth plate's ability to sense and respond to chemical and mechanical stimuli. Furthermore, we explore the importance of using the appropriate mechanism to study the cilium in vivo and suggest IFT88 deletion is the best available technique. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 36:533-545, 2018.
Collapse
Affiliation(s)
- Emily R. Moore
- Department of Biomedical Engineering; Columbia University; 351 Engineering Terrace, Mail Code 8904, 1210 Amsterdam Avenue New York 10027 New York
| | - Christopher R. Jacobs
- Department of Biomedical Engineering; Columbia University; 351 Engineering Terrace, Mail Code 8904, 1210 Amsterdam Avenue New York 10027 New York
| |
Collapse
|
48
|
Palade J, Djordjevic D, Hutchins ED, George RM, Cornelius JA, Rawls A, Ho JWK, Kusumi K, Wilson-Rawls J. Identification of satellite cells from anole lizard skeletal muscle and demonstration of expanded musculoskeletal potential. Dev Biol 2018; 433:344-356. [PMID: 29291980 PMCID: PMC6180209 DOI: 10.1016/j.ydbio.2017.08.037] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 08/22/2017] [Accepted: 08/29/2017] [Indexed: 10/18/2022]
Abstract
The lizards are evolutionarily the closest vertebrates to humans that demonstrate the ability to regenerate entire appendages containing cartilage, muscle, skin, and nervous tissue. We previously isolated PAX7-positive cells from muscle of the green anole lizard, Anolis carolinensis, that can differentiate into multinucleated myotubes and express the muscle structural protein, myosin heavy chain. Studying gene expression in these satellite/progenitor cell populations from A. carolinensis can provide insight into the mechanisms regulating tissue regeneration. We generated a transcriptome from proliferating lizard myoprogenitor cells and compared them to transcriptomes from the mouse and human tissues from the ENCODE project using XGSA, a statistical method for cross-species gene set analysis. These analyses determined that the lizard progenitor cell transcriptome was most similar to mammalian satellite cells. Further examination of specific GO categories of genes demonstrated that among genes with the highest level of expression in lizard satellite cells were an increased number of genetic regulators of chondrogenesis, as compared to mouse satellite cells. In micromass culture, lizard PAX7-positive cells formed Alcian blue and collagen 2a1 positive nodules, without the addition of exogenous morphogens, unlike their mouse counterparts. Subsequent quantitative RT-PCR confirmed up-regulation of expression of chondrogenic regulatory genes in lizard cells, including bmp2, sox9, runx2, and cartilage specific structural genes, aggrecan and collagen 2a1. Taken together, these data suggest that tail regeneration in lizards involves significant alterations in gene regulation with expanded musculoskeletal potency.
Collapse
Affiliation(s)
- Joanna Palade
- School of Life Sciences, Arizona State University, PO Box 874501, Tempe, AZ 85287-4501, USA.
| | - Djordje Djordjevic
- Bioinformatics and Systems Medicine Laboratory, Victor Chang Cardiac Research Institute and St. Vincent's Clinical School, The University of New South Wales, Darlinghurst, NSW 2010, Australia.
| | - Elizabeth D Hutchins
- School of Life Sciences, Arizona State University, PO Box 874501, Tempe, AZ 85287-4501, USA; Neurogenomics Division, Translational Genomics Research Institute, 455 N. Fifth Street Phoenix, 85004, AZ, USA.
| | - Rajani M George
- School of Life Sciences, Arizona State University, PO Box 874501, Tempe, AZ 85287-4501, USA.
| | - John A Cornelius
- School of Life Sciences, Arizona State University, PO Box 874501, Tempe, AZ 85287-4501, USA.
| | - Alan Rawls
- School of Life Sciences, Arizona State University, PO Box 874501, Tempe, AZ 85287-4501, USA.
| | - Joshua W K Ho
- Bioinformatics and Systems Medicine Laboratory, Victor Chang Cardiac Research Institute and St. Vincent's Clinical School, The University of New South Wales, Darlinghurst, NSW 2010, Australia.
| | - Kenro Kusumi
- School of Life Sciences, Arizona State University, PO Box 874501, Tempe, AZ 85287-4501, USA; Neurogenomics Division, Translational Genomics Research Institute, 455 N. Fifth Street Phoenix, 85004, AZ, USA.
| | - Jeanne Wilson-Rawls
- School of Life Sciences, Arizona State University, PO Box 874501, Tempe, AZ 85287-4501, USA.
| |
Collapse
|
49
|
Yano K, Washio K, Tsumanuma Y, Yamato M, Ohta K, Okano T, Izumi Y. The role of Tsukushi (TSK), a small leucine-rich repeat proteoglycan, in bone growth. Regen Ther 2017; 7:98-107. [PMID: 30271858 PMCID: PMC6147151 DOI: 10.1016/j.reth.2017.08.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 08/07/2017] [Accepted: 08/14/2017] [Indexed: 01/14/2023] Open
Abstract
INTRODUCTION Endochondral ossification is one of a key process for bone maturation. Tsukushi (TSK) is a novel member of the secreted small leucine-rich repeat proteoglycan (SLRP) family. SLRPs localize to skeletal regions and play significant roles during whole phases of bone development. Although prior evidence suggests that TSK may be involved in the regulation of bone formation, its role in skeletal development has not yet been elucidated. METHODS In the present study, we examined TSK's function during bone growth by comparing skeletal growth of TSK deficient (TSK-/-) mice and wild type (WT) mice. And an in vitro experiment using siRNA transfection of a chondrogenic cell line was performed. RESULTS TSK-/- mice exhibited decreased weight and short stature at 3 weeks of age due to decreased longitudinal bone growth coupled with low bone mass. Furthermore, an in vitro experiment using siRNA transfection into a chondrogenic cell line revealed that decreased TSK expression induced down-regulation of key chondrogenic marker gene expression and up-regulation of mid-to-late chondrogenic markers gene expression. CONCLUSIONS Our results reveal that TSK regulates bone elongation and bone mass by modulating growth plate chondrocyte function and consequently, overall body size.
Collapse
Key Words
- BMP, bone morphogenetic protein
- Chondrocyte
- ECM, extracellular matrix
- EDTA, ethylenediaminetetraacetic Acid
- Endochondral ossification
- FBS, fetal bovine serum
- FGF, fibroblast growth factor
- Growth plate
- ITS, insulin-transferrin-selenium supplements
- SLRP, small leucine-rich repeat proteoglycan
- SLRPs
- Skeletal development
- TGF, transforming growth factor
- TRAP, tartrate-resistant acid phosphatase
- TSK, Tsukushi
- Tsukushi
- WT, wild type
- β-gal, β-Galactosidase
Collapse
Affiliation(s)
- Kosei Yano
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
- Institute of Advanced Biomedical Engineering and Sciences, Tokyo Women's Medical University (TWIns), 8-1 Kawada-cho, Shinjuku, Tokyo 162-8666, Japan
| | - Kaoru Washio
- Institute of Advanced Biomedical Engineering and Sciences, Tokyo Women's Medical University (TWIns), 8-1 Kawada-cho, Shinjuku, Tokyo 162-8666, Japan
| | - Yuka Tsumanuma
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Masayuki Yamato
- Institute of Advanced Biomedical Engineering and Sciences, Tokyo Women's Medical University (TWIns), 8-1 Kawada-cho, Shinjuku, Tokyo 162-8666, Japan
| | - Kunimasa Ohta
- Department of Developmental Neurobiology, Graduate School of Life Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto 860-8556, Japan
| | - Teruo Okano
- Institute of Advanced Biomedical Engineering and Sciences, Tokyo Women's Medical University (TWIns), 8-1 Kawada-cho, Shinjuku, Tokyo 162-8666, Japan
| | - Yuichi Izumi
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| |
Collapse
|
50
|
Abstract
In addition to their conventional role as a conduit system for gases, nutrients, waste products or cells, blood vessels in the skeletal system play active roles in controlling multiple aspects of bone formation and provide niches for hematopoietic stem cells that reside within the bone marrow. In addition, recent studies have highlighted roles for blood vessels during bone healing. Here, we provide an overview of the architecture of the bone vasculature and discuss how blood vessels form within bone, how their formation is modulated, and how they function during development and fracture repair.
Collapse
Affiliation(s)
- Kishor K Sivaraj
- Max-Planck-Institute for Molecular Biomedicine, Department of Tissue Morphogenesis and University of Münster, Faculty of Medicine, Münster D-48149, Germany
| | - Ralf H Adams
- Max-Planck-Institute for Molecular Biomedicine, Department of Tissue Morphogenesis and University of Münster, Faculty of Medicine, Münster D-48149, Germany
| |
Collapse
|