1
|
Dalecka L, Hruba E, Andrasova M, Steklikova K, Pavlikova Z, Kucerova K, Szotkowska T, Bartos M, Buchtova M, Tucker AS, Hovorakova M. Sprouty2/4 deficiency disrupts early signaling centers impacting chondrogenesis in the mouse forelimb. JBMR Plus 2025; 9:ziaf002. [PMID: 39906257 PMCID: PMC11792080 DOI: 10.1093/jbmrpl/ziaf002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 12/18/2024] [Accepted: 01/08/2025] [Indexed: 02/06/2025] Open
Abstract
The FGF signaling pathway plays an important role in the regulation of limb development, controlling cell migration, proliferation, differentiation, and apoptosis. Sprouty proteins act as antagonists of the FGF pathway and control the extent of FGF signaling as part of a negative feedback loop. Sprouty2/4 deficient mice evince defects in endochondral bone formation and digit patterning in their forelimbs, with pathogenesis recently related to ciliopathies. To understand the mechanisms behind these pathologies, the limb defects in Sprouty2+/-;Sprouty4-/- male and female mice were characterized and correlated to the dynamic expression patterns of Sprouty2 and Sprouty4, and the impact on the main signaling centers of the limb bud was assessed. Sprouty2 and Sprouty4 exhibited dynamic expressions during limb development. Interestingly, despite similar expression patterns in all limbs, the hindlimbs did not evince any obvious alterations in development, while the forelimbs showed consistent phenotypes of variable severity. Prenatally as well as postnatally, the left forelimb was significantly more severely affected than the right one. A broad variety of pathologies was present in the autopodium of the forelimb, including changes in digit number, size, shape, and number of bones, hand clefts, and digit fusions. Ectopic ossification of bones and abnormal bone fusions detected in micro-CT scans were frequently observed in the digital as well as in the carpal and metacarpal areas. Sprouty2+/-;Sprouty4-/- limb buds showed patchy loss of Fgf8 expression in the apical ectodermal ridge, and a loss of tissue underlying these regions. The zone of polarizing activity was also impacted, with lineage analysis highlighting a change in the contribution of Sonic hedgehog expressing cells. These findings support the link between Sproutys and Hedgehog signaling during limb development and highlight the importance of Sprouty2 and Sprouty4 in controlling early signaling centers in the limb.
Collapse
Affiliation(s)
- Linda Dalecka
- First Faculty of Medicine, Institute of Histology and Embryology, Charles University, 128 00 Prague, Czech Republic
- Department of Cell Biology, Faculty of Science, Charles University, 128 00 Prague, Czech Republic
| | - Eva Hruba
- Institute of Animal Physiology and Genetics, Czech Academy of Sciences, 602 00 Brno, Czech Republic
- Department of Experimental Biology, Faculty of Science, Masaryk University, 625 00 Brno, Czech Republic
| | - Marketa Andrasova
- First Faculty of Medicine, Institute of Histology and Embryology, Charles University, 128 00 Prague, Czech Republic
- Department of Anthropology and Human Genetics, Faculty of Science, Charles University, 128 00 Prague, Czech Republic
| | - Klara Steklikova
- First Faculty of Medicine, Institute of Histology and Embryology, Charles University, 128 00 Prague, Czech Republic
- Department of Cell Biology, Faculty of Science, Charles University, 128 00 Prague, Czech Republic
| | - Zuzana Pavlikova
- First Faculty of Medicine, Institute of Histology and Embryology, Charles University, 128 00 Prague, Czech Republic
- Department of Anthropology and Human Genetics, Faculty of Science, Charles University, 128 00 Prague, Czech Republic
| | - Klara Kucerova
- Institute of Animal Physiology and Genetics, Czech Academy of Sciences, 602 00 Brno, Czech Republic
- Department of Experimental Biology, Faculty of Science, Masaryk University, 625 00 Brno, Czech Republic
| | - Tereza Szotkowska
- Institute of Animal Physiology and Genetics, Czech Academy of Sciences, 602 00 Brno, Czech Republic
| | - Martin Bartos
- First Faculty of Medicine, General University Hospital, Institute of Dental Medicine, 121 08 Prague, Czech Republic
- First Faculty of Medicine, Institute of Anatomy, Charles University, 128 00 Prague, Czech Republic
| | - Marcela Buchtova
- Institute of Animal Physiology and Genetics, Czech Academy of Sciences, 602 00 Brno, Czech Republic
- Department of Experimental Biology, Faculty of Science, Masaryk University, 625 00 Brno, Czech Republic
| | - Abigail Saffron Tucker
- First Faculty of Medicine, Institute of Histology and Embryology, Charles University, 128 00 Prague, Czech Republic
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral and Craniofacial Sciences, King’s College London, Guys Hospital, London, TN8 7LR, United Kingdom
| | - Maria Hovorakova
- First Faculty of Medicine, Institute of Histology and Embryology, Charles University, 128 00 Prague, Czech Republic
| |
Collapse
|
2
|
Maldonado E, Martínez-Sanz E, Catón J, Arráez-Aybar LA, Barrio MC, Naredo E, Murillo-González JA, Mérida-Velasco JR. Development of the Interosseous Muscles of the Human Hand: Morphological and Functional Aspects of the Terminal Insertion. Cells Tissues Organs 2024; 214:104-113. [PMID: 39106842 DOI: 10.1159/000540760] [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: 09/07/2023] [Accepted: 07/23/2024] [Indexed: 08/09/2024] Open
Abstract
INTRODUCTION To date, there have been no studies conducted on the development of interosseous muscles (IO) in the human hand. This study aimed to investigate the development of these muscles in order to clarify their terminal insertions and their relationship with the metacarpophalangeal joints. METHODS Serial sections of 25 human specimens (9 embryos and 16 fetuses) between the 7th and 14th weeks of development, sourced from the Collection of the Department of Anatomy and Embryology at UCM Faculty of Medicine, were analyzed bilaterally using a conventional optical microscope. RESULTS Our findings revealed that, during the 7th week of development, the metacarpophalangeal interzone mesenchyme extended into the extensor apparatus of the fingers. Furthermore, we observed that the joint capsule and the tendon of the IO derive from the articular interzone mesenchyme. By the end of the 7th week, corresponding to Carnegie stage 21, the myotendinous junction appeared, initiating cavitation of the metacarpophalangeal joint. During the fetal period, the terminal insertions of the IO were identified: both the dorsal interosseous (DI) and palmar interosseous (PI) muscles insert into the metacarpophalangeal joint capsule and establish a connection with the volar plate located at the base of the proximal phalanx and the extensor apparatus. Some muscle fibers also attach to the joint capsule at the level of the proximal synovial cul-de-sac. The functional implications of these findings are discussed within this work. CONCLUSION This study provides the first detailed description of the development of the interosseous muscles in the human hand.
Collapse
Affiliation(s)
- Estela Maldonado
- Department of Anatomy and Embryology, Faculty of Medicine, Universidad Complutense de Madrid (UCM), Madrid, Spain,
- UCM Research Group No. 920202, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Madrid, Spain,
| | - Elena Martínez-Sanz
- Department of Anatomy and Embryology, Faculty of Medicine, Universidad Complutense de Madrid (UCM), Madrid, Spain
- UCM Research Group No. 920202, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Madrid, Spain
| | - Javier Catón
- Department of Anatomy and Embryology, Faculty of Medicine, Universidad Complutense de Madrid (UCM), Madrid, Spain
- UCM Research Group No. 920202, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Madrid, Spain
| | - Luis A Arráez-Aybar
- Department of Anatomy and Embryology, Faculty of Medicine, Universidad Complutense de Madrid (UCM), Madrid, Spain
- UCM Research Group No. 920202, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Madrid, Spain
| | - María Carmen Barrio
- UCM Research Group No. 920202, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Madrid, Spain
- Department of Anatomy and Embryology, Faculty of Optics and Optometry, Universidad Complutense de Madrid (UCM), Madrid, Spain
| | - Esperanza Naredo
- Department of Rheumatology, Hospital Universitario Fundación Jiménez Díaz, Madrid, Spain
- Bone and Joint Research Unit, IIS-Fundación Jiménez Díaz, Universidad Autónoma of Madrid (UAM), Madrid, Spain
| | - Jorge A Murillo-González
- Department of Anatomy and Embryology, Faculty of Medicine, Universidad Complutense de Madrid (UCM), Madrid, Spain
- UCM Research Group No. 920202, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Madrid, Spain
| | - José Ramón Mérida-Velasco
- Department of Anatomy and Embryology, Faculty of Medicine, Universidad Complutense de Madrid (UCM), Madrid, Spain
- UCM Research Group No. 920202, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Madrid, Spain
| |
Collapse
|
3
|
Hirsinger E, Blavet C, Bonnin MA, Bellenger L, Gharsalli T, Duprez D. Limb connective tissue is organized in a continuum of promiscuous fibroblast identities during development. iScience 2024; 27:110305. [PMID: 39050702 PMCID: PMC11267076 DOI: 10.1016/j.isci.2024.110305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 05/21/2024] [Accepted: 06/17/2024] [Indexed: 07/27/2024] Open
Abstract
Connective tissue (CT), which includes tendon and muscle CT, plays critical roles in development, in particular as positional cue provider. Nonetheless, our understanding of fibroblast developmental programs is hampered because fibroblasts are highly heterogeneous and poorly characterized. Combining single-cell RNA-sequencing-based strategies including trajectory inference and in situ hybridization analyses, we address the diversity of fibroblasts and their developmental trajectories during chicken limb fetal development. We show that fibroblasts switch from a positional information to a lineage diversification program at the fetal period onset. Muscle CT and tendon are composed of several fibroblast populations that emerge asynchronously. Once the final muscle pattern is set, transcriptionally close populations are found in neighboring locations in limbs, prefiguring the adult fibroblast layers. We propose that the limb CT is organized in a continuum of promiscuous fibroblast identities, allowing for the robust and efficient connection of muscle to bone and skin.
Collapse
Affiliation(s)
- Estelle Hirsinger
- Sorbonne Université, Institut Biologie Paris Seine, CNRS UMR7622, Developmental Biology Laboratory, Inserm U1156, 75005 Paris, France
| | - Cédrine Blavet
- Sorbonne Université, Institut Biologie Paris Seine, CNRS UMR7622, Developmental Biology Laboratory, Inserm U1156, 75005 Paris, France
| | - Marie-Ange Bonnin
- Sorbonne Université, Institut Biologie Paris Seine, CNRS UMR7622, Developmental Biology Laboratory, Inserm U1156, 75005 Paris, France
| | - Léa Bellenger
- Sorbonne Université, CNRS FR3631, Inserm U1156, Institut de Biologie Paris Seine (IBPS), ARTbio Bioinformatics Analysis Facility, Paris, Institut Français de Bioinformatique (IFB), 75005 Paris, France
| | - Tarek Gharsalli
- Sorbonne Université, Institut Biologie Paris Seine, CNRS UMR7622, Developmental Biology Laboratory, Inserm U1156, 75005 Paris, France
- Inovarion, 75005 Paris, France
| | - Delphine Duprez
- Sorbonne Université, Institut Biologie Paris Seine, CNRS UMR7622, Developmental Biology Laboratory, Inserm U1156, 75005 Paris, France
| |
Collapse
|
4
|
Usami Y, Iijima H, Kokubun T. Exploring the role of mechanical forces on tendon development using in vivo model: A scoping review. Dev Dyn 2024; 253:550-565. [PMID: 37947268 DOI: 10.1002/dvdy.673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 08/25/2023] [Accepted: 09/27/2023] [Indexed: 11/12/2023] Open
Abstract
Tendons transmit the muscle contraction forces to bones and drive joint movement throughout life. While extensive research have indicated the essentiality of mechanical forces on tendon development, a comprehensive understanding of the fundamental role of mechanical forces still needs to be impaerted. This scoping review aimed to summarize the current knowledge about the role of mechanical forces during the tendon developmental phase. The electronic database search using PubMed, performed in May 2023, yielded 651 articles, of which 16 met the prespecified inclusion criteria. We summarized and divided the methods to reduce the mechanical force into three groups: loss of muscle, muscle dysfunction, and weight-bearing regulation. In contrast, there were few studies to analyze the increased mechanical force model. Most studies suggested that mechanical force has some roles in tendon development in the embryo to postnatal phase. However, we identified species variability and methodological heterogeneity to modulate mechanical force. To establish a comprehensive understanding, methodological commonality to modulate the mechanical force is needed in this field. Additionally, summarizing chronological changes in developmental processes across animal species helps to understand the essence of developmental tendon mechanobiology. We expect that the findings summarized in the current review serve as a groundwork for future study in the fields of tendon developmantal biology and mechanobiology.
Collapse
Affiliation(s)
- Yuna Usami
- Graduate School of Health, Medicine, and Welfare, Saitama Prefectural University, Koshigaya, Japan
| | - Hirotaka Iijima
- Discovery Center for Musculoskeletal Recovery, Schoen Adams Research Institute at Spaulding, Charlestown, Massachusetts, USA
- Department of Physical Medicine & Rehabilitation, Harvard Medical School, Boston, Massachusetts, USA
| | - Takanori Kokubun
- Graduate School of Health, Medicine, and Welfare, Saitama Prefectural University, Koshigaya, Japan
- Department of Physical Therapy, School of Health and Social Services, Saitama Prefectural University, Koshigaya, Japan
| |
Collapse
|
5
|
Lv Y, Wang Q, Lin C, Zheng X, Zhang Y, Hu X. Overexpression of Fgf18 in cranial neural crest cells recapitulates Pierre Robin sequence in mice. Front Cell Dev Biol 2024; 12:1376814. [PMID: 38694818 PMCID: PMC11061347 DOI: 10.3389/fcell.2024.1376814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 04/05/2024] [Indexed: 05/04/2024] Open
Abstract
The pivotal role of FGF18 in the regulation of craniofacial and skeletal development has been well established. Previous studies have demonstrated that mice with deficiency in Fgf18 exhibit severe craniofacial dysplasia. Recent clinical reports have revealed that the duplication of chromosome 5q32-35.3, which encompasses the Fgf18 gene, can lead to cranial bone dysplasia and congenital craniosynostosis, implicating the consequence of possible overdosed FGF18 signaling. This study aimed to test the effects of augmented FGF18 signaling by specifically overexpressing the Fgf18 gene in cranial neural crest cells using the Wnt1-Cre;pMes-Fgf18 mouse model. The results showed that overexpression of Fgf18 leads to craniofacial abnormalities in mice similar to the Pierre Robin sequence in humans, including abnormal tongue morphology, micrognathia, and cleft palate. Further examination revealed that elevated levels of Fgf18 activated the Akt and Erk signaling pathways, leading to an increase in the proliferation level of tongue tendon cells and alterations in the contraction pattern of the genioglossus muscle. Additionally, we observed that excessive FGF18 signaling contributed to the reduction in the length of Meckel's cartilage and disrupted the development of condylar cartilage, ultimately resulting in mandibular defects. These anomalies involve changes in several downstream signals, including Runx2, p21, Akt, Erk, p38, Wnt, and Ihh. This study highlights the crucial role of maintaining the balance of endogenous FGF18 signaling for proper craniofacial development and offers insights into potential formation mechanisms of the Pierre Robin sequence.
Collapse
Affiliation(s)
| | | | | | | | | | - Xuefeng Hu
- Fujian Key Laboratory of Developmental and Neural Biology, College of Life Sciences, Fujian Normal University, Fuzhou, China
| |
Collapse
|
6
|
Liu C, Zhou N, Li N, Xu T, Chen X, Zhou H, Xie A, Liu H, Zhu L, Wang S, Xiao J. Disrupted tenogenesis in masseter as a potential cause of micrognathia. Int J Oral Sci 2022; 14:50. [PMID: 36257937 PMCID: PMC9579150 DOI: 10.1038/s41368-022-00196-y] [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: 06/21/2022] [Revised: 08/01/2022] [Accepted: 08/04/2022] [Indexed: 11/09/2022] Open
Abstract
Micrognathia is a severe craniofacial deformity affecting appearance and survival. Previous studies revealed that multiple factors involved in the osteogenesis of mandibular bone have contributed to micrognathia, but concerned little on factors other than osteogenesis. In the current study, we found that ectopic activation of Fgf8 by Osr2-cre in the presumptive mesenchyme for masseter tendon in mice led to micrognathia, masseter regression, and the disrupted patterning and differentiation of masseter tendon. Since Myf5-cre;Rosa26R-Fgf8 mice exhibited the normal masseter and mandibular bone, the possibility that the micrognathia and masseter regression resulted directly from the over-expressed Fgf8 was excluded. Further investigation disclosed that a series of chondrogenic markers were ectopically activated in the developing Osr2-cre;Rosa26R-Fgf8 masseter tendon, while the mechanical sensing in the masseter and mandibular bone was obviously reduced. Thus, it suggested that the micrognathia in Osr2-cre;Rosa26R-Fgf8 mice resulted secondarily from the reduced mechanical force transmitted to mandibular bone. Consistently, when tenogenic or myogenic components were deleted from the developing mandibles, both the micrognathia and masseter degeneration took place with the decreased mechanical sensing in mandibular bone, which verified that the loss of mechanical force transmitted by masseter tendon could result in micrognathia. Furthermore, it appeared that the micrognathia resulting from the disrupted tenogenesis was attributed to the impaired osteogenic specification, instead of the differentiation in the periosteal progenitors. Our findings disclose a novel mechanism for mandibular morphogenesis, and shed light on the prevention and treatment for micrognathia.
Collapse
Affiliation(s)
- Chao Liu
- Department of Oral Pathology, Dalian Medical University School of Stomatology, Dalian, China.,Academician Laboratory of Immunology and Oral Development & Regeneration, Dalian Medical University, Dalian, China
| | - Nan Zhou
- Department of Oral Pathology, Dalian Medical University School of Stomatology, Dalian, China
| | - Nan Li
- Department of Oral Pathology, Dalian Medical University School of Stomatology, Dalian, China.,Academician Laboratory of Immunology and Oral Development & Regeneration, Dalian Medical University, Dalian, China
| | - Tian Xu
- Department of Oral Pathology, Dalian Medical University School of Stomatology, Dalian, China
| | - Xiaoyan Chen
- Department of Oral Pathology, Dalian Medical University School of Stomatology, Dalian, China
| | - Hailing Zhou
- Department of Oral Pathology, Dalian Medical University School of Stomatology, Dalian, China
| | - Ailun Xie
- Department of Oral Pathology, Dalian Medical University School of Stomatology, Dalian, China
| | - Han Liu
- Department of Oral Pathology, Dalian Medical University School of Stomatology, Dalian, China.,Academician Laboratory of Immunology and Oral Development & Regeneration, Dalian Medical University, Dalian, China
| | - Lei Zhu
- Department of Oral Pathology, Dalian Medical University School of Stomatology, Dalian, China.,Academician Laboratory of Immunology and Oral Development & Regeneration, Dalian Medical University, Dalian, China
| | - Songlin Wang
- Academician Laboratory of Immunology and Oral Development & Regeneration, Dalian Medical University, Dalian, China. .,Beijing Laboratory of Oral Health, Capital Medical University, Beijing, China.
| | - Jing Xiao
- Department of Oral Pathology, Dalian Medical University School of Stomatology, Dalian, China. .,Academician Laboratory of Immunology and Oral Development & Regeneration, Dalian Medical University, Dalian, China.
| |
Collapse
|
7
|
Korntner SH, Jana A, Kinnard E, Leo E, Beane T, Li X, Sengupta R, Becker L, Kuo CK. Craniofacial tendon development-Characterization of extracellular matrix morphology and spatiotemporal protein distribution. Front Cell Dev Biol 2022; 10:944126. [PMID: 36158210 PMCID: PMC9490420 DOI: 10.3389/fcell.2022.944126] [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] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 08/10/2022] [Indexed: 11/13/2022] Open
Abstract
Craniofacial (CF) tendons are often affected by traumatic injuries and painful disorders that can severely compromise critical jaw functions, such as mastication and talking. Unfortunately, tendons lack the ability to regenerate, and there are no solutions to restore their native properties or function. An understanding of jaw tendon development could inform tendon regeneration strategies to restore jaw function, however CF tendon development has been relatively unexplored. Using the chick embryo, we identified the jaw-closing Tendon of the musculus Adductor Mandibulae Externus (TmAM) and the jaw-opening Tendon of the musculus Depressor Mandibulae (TmDM) that have similar functions to the masticatory tendons in humans. Using histological and immunohistochemical (IHC) analyses, we characterized the TmAM and TmDM on the basis of cell and extracellular matrix (ECM) morphology and spatiotemporal protein distribution from early to late embryonic development. The TmAM and TmDM were detectable as early as embryonic day (d) 9 based on histological staining and tenascin-C (TNC) protein distribution. Collagen content increased and became more organized, cell density decreased, and cell nuclei elongated over time during development in both the TmAM and TmDM. The TmAM and TmDM exhibited similar spatiotemporal patterns for collagen type III (COL3), but differential spatiotemporal patterns for TNC, lysyl oxidase (LOX), and matrix metalloproteinases (MMPs). Our results demonstrate markers that play a role in limb tendon formation are also present in jaw tendons during embryonic development, implicate COL3, TNC, LOX, MMP2, and MMP9 in jaw tendon development, and suggest TmAM and TmDM possess different developmental programs. Taken together, our study suggests the chick embryo may be used as a model with which to study CF tendon extracellular matrix development, the results of which could ultimately inform therapeutic approaches for CF tendon injuries and disorders.
Collapse
Affiliation(s)
- Stefanie H Korntner
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, United States
| | - Aniket Jana
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, United States
| | - Elizabeth Kinnard
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, United States
| | - Emily Leo
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, United States
| | - Timothy Beane
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
| | - Xianmu Li
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
| | - Rohit Sengupta
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, United States
| | - Lauren Becker
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, United States
| | - Catherine K Kuo
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, United States
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States
- Department of Orthopaedics, University of Maryland Medical Center, Baltimore, MD, United States
| |
Collapse
|
8
|
Abstract
In organisms from flies to mammals, the initial formation of a functional tendon is completely dependent on chemical signals from muscle (myokines). However, how myokines affect the maturation, maintenance, and regeneration of tendons as a function of age is completely unstudied. Here we discuss the role of four myokines - fibroblast growth factors (FGF), myostatin, the secreted protein acidic and rich in cysteine (SPARC), and miR-29 - in tendon development and hypothesize a role for these factors in the progressive changes in tendon structure and function as a result of muscle wasting (disuse, aging and disease). Because of the close relationship between mechanical loading and muscle and tendon regulation, disentangling muscle-tendon crosstalk from simple mechanical loading is experimentally quite difficult. Therefore, we propose an experimental framework that hopefully will be useful in demonstrating muscle-tendon crosstalk in vivo. Though understudied, the promise of a better understanding of muscle-tendon crosstalk is the development of new interventions that will improve tendon development, regeneration, and function throughout the lifespan.
Collapse
Affiliation(s)
- Alec M Avey
- Functional Molecular Biology Laboratory, University of California, Davis, CA, United States.,Molecular, Cellular and Integrative Physiology, University of California Davis, Davis, CA, United States
| | - Keith Baar
- Functional Molecular Biology Laboratory, University of California, Davis, CA, United States.,Neurobiology, Physiology and Behavior, University of California Davis, Davis, CA, United States.,Physiology and Membrane Biology, University of California Davis Health, Sacramento, CA, United States.,VA Northern California Health Care System, Mather, CA, United States
| |
Collapse
|
9
|
Nakamichi R, Asahara H. Regulation of tendon and ligament differentiation. Bone 2021; 143:115609. [PMID: 32829041 PMCID: PMC7770025 DOI: 10.1016/j.bone.2020.115609] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 08/06/2020] [Accepted: 08/17/2020] [Indexed: 02/08/2023]
Abstract
Tendons transmit power from muscles to bones, and ligaments maintain the stability of joints, thus producing smooth and flexible movements of articular joints. However, tendons have poor self-healing ability upon damage due to injuries, diseases, or aging. To maintain homeostasis or promote regeneration of the tendon/ligament, it is critical to understand the mechanism responsible for the coordination of tendon/ligament-specific gene expression and subsequent cell differentiation. In this review, we have discussed the core molecular mechanisms involved in the development and homeostasis of tendons and ligaments, with particular focus on transcription factors, signaling, and mechanical stress.
Collapse
Affiliation(s)
- Ryo Nakamichi
- Department of Molecular Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, MBB-102, , La Jolla, CA 92037, USA; Department of Orthopaedic Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Hiroshi Asahara
- Department of Molecular Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, MBB-102, , La Jolla, CA 92037, USA; Department of Systems Biomedicine, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan.
| |
Collapse
|
10
|
Anthwal N, Tucker AS. The TMJ Disc Is a Common Ancestral Feature in All Mammals, as Evidenced by the Presence of a Rudimentary Disc During Monotreme Development. Front Cell Dev Biol 2020; 8:356. [PMID: 32509783 PMCID: PMC7248220 DOI: 10.3389/fcell.2020.00356] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 04/21/2020] [Indexed: 12/14/2022] Open
Abstract
The novel mammalian jaw joint, known in humans as the temporomandibular joint or TMJ, is cushioned by a fibrocartilage disc. This disc is secondarily absent in therian mammals that have lost their dentition, such as giant anteaters and some baleen whales. The disc is also absent in all monotremes. However, it is not known if the absence in monotremes is secondary to the loss of dentition, or if it is an ancestral absence. We use museum held platypus and echidna histological sections to demonstrate that the developing monotreme jaw joint forms a disc primordium that fails to mature and become separated from the mandibular condyle. We then show that monotreme developmental anatomy is similar to that observed in transgenic mouse mutants with reduced cranial musculature. We therefore suggest that the absence of the disc on monotremes is a consequence of the changes in jaw musculature associated with the loss of adult teeth. Taken together, these data indicate that the ancestors of extant monotremes likely had a jaw joint disc, and that the disc evolved in the last common ancestor of all mammals.
Collapse
Affiliation(s)
- Neal Anthwal
- Centre for Craniofacial and Regenerative Biology, King’s College London, London, United Kingdom
| | | |
Collapse
|
11
|
Flexor Tendon: Development, Healing, Adhesion Formation, and Contributing Growth Factors. Plast Reconstr Surg 2020; 144:639e-647e. [PMID: 31568303 DOI: 10.1097/prs.0000000000006048] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Management of flexor tendon injuries of the hand remains a major clinical problem. Even with intricate repair, adhesion formation remains a common complication. Significant progress has been made to better understand the mechanisms of healing and adhesion formation. However, there has been slow progress in the clinical prevention and reversal of flexor tendon adhesions. The goal of this article is to discuss recent literature relating to tendon development, tendon healing, and adhesion formation to identify areas in need of further research. Additional research is needed to understand and compare the molecular, cellular, and genetic mechanisms involved in flexor tendon morphogenesis, postoperative healing, and mechanical loading. Such knowledge is critical to determine how to improve repair outcomes and identify new therapeutic strategies to promote tissue regeneration and prevent adhesion formation.
Collapse
|
12
|
Wang J, Khodabukus A, Rao L, Vandusen K, Abutaleb N, Bursac N. Engineered skeletal muscles for disease modeling and drug discovery. Biomaterials 2019; 221:119416. [PMID: 31419653 DOI: 10.1016/j.biomaterials.2019.119416] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Revised: 08/01/2019] [Accepted: 08/05/2019] [Indexed: 01/04/2023]
Abstract
Skeletal muscle is the largest organ of human body with several important roles in everyday movement and metabolic homeostasis. The limited ability of small animal models of muscle disease to accurately predict drug efficacy and toxicity in humans has prompted the development in vitro models of human skeletal muscle that fatefully recapitulate cell and tissue level functions and drug responses. We first review methods for development of three-dimensional engineered muscle tissues and organ-on-a-chip microphysiological systems and discuss their potential utility in drug discovery research and development of new regenerative therapies. Furthermore, we describe strategies to increase the functional maturation of engineered muscle, and motivate the importance of incorporating multiple tissue types on the same chip to model organ cross-talk and generate more predictive drug development platforms. Finally, we review the ability of available in vitro systems to model diseases such as type II diabetes, Duchenne muscular dystrophy, Pompe disease, and dysferlinopathy.
Collapse
Affiliation(s)
- Jason Wang
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | | | - Lingjun Rao
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Keith Vandusen
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Nadia Abutaleb
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Nenad Bursac
- Department of Biomedical Engineering, Duke University, Durham, NC, USA.
| |
Collapse
|
13
|
Salva JE, Roberts RR, Stucky TS, Merrill AE. Nuclear FGFR2 regulates musculoskeletal integration within the developing limb. Dev Dyn 2019; 248:233-246. [PMID: 30620790 DOI: 10.1002/dvdy.9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 11/29/2018] [Accepted: 12/18/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Bent bone dysplasia syndrome (BBDS), a congenital skeletal disorder caused by dominant mutations in fibroblast growth factor receptor 2 (FGFR2), is characterized by bowed long bones within the limbs. We previously showed that the FGFR2 mutations in BBDS enhance nuclear and nucleolar localization of the receptor; however, exactly how shifts in subcellular distribution of FGFR2 affect limb development remained unknown. RESULTS Targeted expression of the BBDS mutations in the lateral plate mesoderm of the developing chick induced angulated hindlimbs, a hallmark feature of the disease. Whole-mount analysis of the underlying skeleton revealed bent long bones with shortened bone collars and, in severe cases, dysmorphic epiphyses. Epiphyseal changes were also correlated with joint dislocations and contractures. Histological analysis revealed that bent long bones and joint defects were closely associated with irregularities in skeletal muscle patterning and tendon-to-bone attachment. The spectrum of limb phenotypes induced by the BBDS mutations were recapitulated by targeted expression of wild-type FGFR2 appended with nuclear and nucleolar localization signals. CONCLUSIONS Our results indicate that the bent long bones in BBDS arise from disruptions in musculoskeletal integration and that increased nuclear and nucleolar localization of FGFR2 plays a mechanistic role in the disease phenotype. 248:233-246, 2019. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Joanna E Salva
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, California
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Ryan R Roberts
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, California
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Taylor S Stucky
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, California
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Amy E Merrill
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, California
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California
| |
Collapse
|
14
|
Márquez-Flórez K, Shefelbine S, Ramírez-Martínez A, Garzón-Alvarado D. Computational model for the patella onset. PLoS One 2018; 13:e0207770. [PMID: 30533045 PMCID: PMC6289436 DOI: 10.1371/journal.pone.0207770] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 11/06/2018] [Indexed: 11/23/2022] Open
Abstract
The patella is a sesamoid bone embedded within the quadriceps tendon and the patellar tendon that articulates with the femur. However, how is it formed is still unknown. Therefore, here we have evaluated, computationally, how three theories explain, independently, the patella onset. The first theory was proposed recently, in 2015. This theory suggested that the patella is initially formed as a bone eminence, attached to the anterodistal surface of the femur, while the quadriceps tendon is forming. Thereafter, a joint develops between the eminence and the femur, regulated by mechanical load. We evaluated this theory by simulating the biochemical environment that surrounds the tendon development. As a result, we obtained a patella-like structure embedded within the tendon, especially for larger flexion angles. The second and third theories are the most accepted until now. They state that the patella develops within tendons in response to the mechanical environment provided by the attaching muscles. The second theory analyzed the mechanical conditions (high hydrostatic stress) that (according to previous Carter theories) lead to the differentiation from tendon to fibrocartilage, and then, to bone. The last theory was evaluated using the self-optimizing capability of biological tissue. It was considered that the development of the patella, due to tissue topological optimization of the developing quadriceps tendon, is a feasible explanation of the patella appearance. For both theories, a patella onset was obtained as a structure embedded within the tendon. This model provided information about the relationship between the flexion angle and the patella size and shape. In conclusion, the computational models used to evaluate and analyze the selected theories allow determining that the patella onset may be the result of a combination of biochemical and mechanical factors that surround the patellar tendon development.
Collapse
Affiliation(s)
- Kalenia Márquez-Flórez
- Biomimetics Laboratory, Instituto de Biotecnología, Universidad Nacional de Colombia, Bogotá, Ciudad Universitaria, Colombia
- Numerical Methods and Modeling Research Group (GNUM), Universidad Nacional de Colombia, Bogotá, Ciudad Universitaria, Colombia
| | - Sandra Shefelbine
- Department of Mechanical Engineering, Northeastern University, Boston, MA, United States of America
| | | | - Diego Garzón-Alvarado
- Biomimetics Laboratory, Instituto de Biotecnología, Universidad Nacional de Colombia, Bogotá, Ciudad Universitaria, Colombia
- Numerical Methods and Modeling Research Group (GNUM), Universidad Nacional de Colombia, Bogotá, Ciudad Universitaria, Colombia
| |
Collapse
|
15
|
Woronowicz KC, Gline SE, Herfat ST, Fields AJ, Schneider RA. FGF and TGFβ signaling link form and function during jaw development and evolution. Dev Biol 2018; 444 Suppl 1:S219-S236. [PMID: 29753626 PMCID: PMC6239991 DOI: 10.1016/j.ydbio.2018.05.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 04/20/2018] [Accepted: 05/06/2018] [Indexed: 12/14/2022]
Abstract
How does form arise during development and change during evolution? How does form relate to function, and what enables embryonic structures to presage their later use in adults? To address these questions, we leverage the distinct functional morphology of the jaw in duck, chick, and quail. In connection with their specialized mode of feeding, duck develop a secondary cartilage at the tendon insertion of their jaw adductor muscle on the mandible. An equivalent cartilage is absent in chick and quail. We hypothesize that species-specific jaw architecture and mechanical forces promote secondary cartilage in duck through the differential regulation of FGF and TGFβ signaling. First, we perform transplants between chick and duck embryos and demonstrate that the ability of neural crest mesenchyme (NCM) to direct the species-specific insertion of muscle and the formation of secondary cartilage depends upon the amount and spatial distribution of NCM-derived connective tissues. Second, we quantify motility and build finite element models of the jaw complex in duck and quail, which reveals a link between species-specific jaw architecture and the predicted mechanical force environment. Third, we investigate the extent to which mechanical load mediates FGF and TGFβ signaling in the duck jaw adductor insertion, and discover that both pathways are mechano-responsive and required for secondary cartilage formation. Additionally, we find that FGF and TGFβ signaling can also induce secondary cartilage in the absence of mechanical force or in the adductor insertion of quail embryos. Thus, our results provide novel insights on molecular, cellular, and biomechanical mechanisms that couple musculoskeletal form and function during development and evolution.
Collapse
Affiliation(s)
- Katherine C Woronowicz
- Department of Orthopaedic Surgery, University of California, San Francisco, 513 Parnassus Avenue, S-1161, San Francisco, CA 94143-0514, USA
| | - Stephanie E Gline
- Department of Orthopaedic Surgery, University of California, San Francisco, 513 Parnassus Avenue, S-1161, San Francisco, CA 94143-0514, USA
| | - Safa T Herfat
- Department of Orthopaedic Surgery, University of California, San Francisco, 513 Parnassus Avenue, S-1161, San Francisco, CA 94143-0514, USA
| | - Aaron J Fields
- Department of Orthopaedic Surgery, University of California, San Francisco, 513 Parnassus Avenue, S-1161, San Francisco, CA 94143-0514, USA
| | - Richard A Schneider
- Department of Orthopaedic Surgery, University of California, San Francisco, 513 Parnassus Avenue, S-1161, San Francisco, CA 94143-0514, USA.
| |
Collapse
|
16
|
Hirasawa T, Kuratani S. Evolution of the muscular system in tetrapod limbs. ZOOLOGICAL LETTERS 2018; 4:27. [PMID: 30258652 PMCID: PMC6148784 DOI: 10.1186/s40851-018-0110-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 09/04/2018] [Indexed: 05/16/2023]
Abstract
While skeletal evolution has been extensively studied, the evolution of limb muscles and brachial plexus has received less attention. In this review, we focus on the tempo and mode of evolution of forelimb muscles in the vertebrate history, and on the developmental mechanisms that have affected the evolution of their morphology. Tetrapod limb muscles develop from diffuse migrating cells derived from dermomyotomes, and the limb-innervating nerves lose their segmental patterns to form the brachial plexus distally. Despite such seemingly disorganized developmental processes, limb muscle homology has been highly conserved in tetrapod evolution, with the apparent exception of the mammalian diaphragm. The limb mesenchyme of lateral plate mesoderm likely plays a pivotal role in the subdivision of the myogenic cell population into individual muscles through the formation of interstitial muscle connective tissues. Interactions with tendons and motoneuron axons are involved in the early and late phases of limb muscle morphogenesis, respectively. The mechanism underlying the recurrent generation of limb muscle homology likely resides in these developmental processes, which should be studied from an evolutionary perspective in the future.
Collapse
Affiliation(s)
- Tatsuya Hirasawa
- Laboratory for Evolutionary Morphology, RIKEN Center for Biosystems Dynamics Research (BDR), 2-2-3 Minatojima-minami, Chuo-ku, Kobe, Hyogo 650-0047 Japan
| | - Shigeru Kuratani
- Laboratory for Evolutionary Morphology, RIKEN Center for Biosystems Dynamics Research (BDR), 2-2-3 Minatojima-minami, Chuo-ku, Kobe, Hyogo 650-0047 Japan
- Evolutionary Morphology Laboratory, RIKEN Cluster for Pioneering Research (CPR), 2-2-3 Minatojima-minami, Chuo-ku, Kobe, Hyogo 650-0047 Japan
| |
Collapse
|
17
|
Hou Y, Ni M, Lin S, Sun Y, Lin W, Liu Y, Wang H, He W, Li G, Xu L. Tenomodulin highly expressing MSCs as a better cell source for tendon injury healing. Oncotarget 2017; 8:77424-77435. [PMID: 29100398 PMCID: PMC5652790 DOI: 10.18632/oncotarget.20495] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 07/13/2017] [Indexed: 02/06/2023] Open
Abstract
Tendon injuries are common orthopedic problems which may cause severe morbidity. MSCs (mesenchymal stem cells) have shown promising effect on tissue engineering and have been used for the treatment of tendon injury. But the low tenogenic differentiation capacity of MSCs have hindered their application. In the present study, we have constructed the Tenomodulin (Tnmd) promoter-driven GFP expression lentiviral plasmid. After transduced into BMSCs, the expression of GFP was used to select BMSCs highly expressing Tnmd by flow cytometry. We found that MSCs with higher level of Tnmd expression had stronger tenogenic differentiation ability. Furthermore, RNA sequencing was performed to identify the molecular difference between BMSCs expressing higher and lower levels of Tnmd. And finally we demonstrated that GDF7 was upregulated in BMSCs highly expressing Tnmd and played an vital role in promoting tenogenic differentiation of BMSCs. GDF7 was mainly accounted for the elevated tenogenic differentiation ability of BMSCs with higher Tnmd expression as silencing the endogenous GDF7 significantly inhibited tenogenesis in BMSCs. In addition, the effect of BMSCs with higher Tnmd level on tendon healing was evaluated by a rat patellar tendon injury model. Taken together, our study showed that Tnmd could be used as an ideal cell surface marker to select cells with higher tenogenic differentiation ability from BMSCs, and GDF7 was indispensable for tenogenesis of MSCs.
Collapse
Affiliation(s)
- Yonghui Hou
- Key Laboratory of Orthopaedics & Traumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, P.R. China
- Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, P.R. China
| | - Ming Ni
- Department of Orthopaedics, PLA General Hospital, Beijing, P.R. China
| | - Sien Lin
- Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, P.R. China
| | - Yuxin Sun
- Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, P.R. China
| | - Weiping Lin
- Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, P.R. China
| | - Yamei Liu
- Department of Diagnostics of Traditional Chinese Medicine, Guangzhou University of Traditional Chinese Medicine, Guangzhou, P.R. China
| | - Haibin Wang
- Key Laboratory of Orthopaedics & Traumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, P.R. China
| | - Wei He
- Key Laboratory of Orthopaedics & Traumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, P.R. China
| | - Gang Li
- Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, P.R. China
- Stem Cells and Regenerative Medicine Laboratory, Lui Che Woo Institute of Innovative Medicine, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, P.R. China
| | - Liangliang Xu
- Key Laboratory of Orthopaedics & Traumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, P.R. China
- Laboratory of Orthopaedics & Traumatology, Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, P.R. China
| |
Collapse
|
18
|
Asahara H, Inui M, Lotz MK. Tendons and Ligaments: Connecting Developmental Biology to Musculoskeletal Disease Pathogenesis. J Bone Miner Res 2017; 32:1773-1782. [PMID: 28621492 PMCID: PMC5585011 DOI: 10.1002/jbmr.3199] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 06/08/2017] [Accepted: 06/14/2017] [Indexed: 01/09/2023]
Abstract
Tendons and ligaments provide connections between muscle and bone or bone and bone to enable locomotion. Damage to tendons and ligaments caused by acute or chronic injury or associated with aging and arthritis is a prevalent cause of disability. Improvements in approaches for the treatment of these conditions depend on a better understanding of tendon and ligament development, cell biology, and pathophysiology. This review focuses on recent advances in the discovery of transcription factors that control ligament and tendon cell differentiation, how cell and extracellular matrix homeostasis are altered in disease, and how this new insight can lead to novel therapeutic approaches. © 2017 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Hiroshi Asahara
- Department of Molecular Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, USA
- Department of Systems BioMedicine, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Masafumi Inui
- Laboratory of Animal Regeneration Systemology, Department of Life Science, School of Agriculture, Meiji University, Kanagawa, 214-8571
| | - Martin K. Lotz
- Department of Molecular Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, USA
| |
Collapse
|
19
|
Nassari S, Duprez D, Fournier-Thibault C. Non-myogenic Contribution to Muscle Development and Homeostasis: The Role of Connective Tissues. Front Cell Dev Biol 2017; 5:22. [PMID: 28386539 PMCID: PMC5362625 DOI: 10.3389/fcell.2017.00022] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 03/07/2017] [Indexed: 12/22/2022] Open
Abstract
Skeletal muscles belong to the musculoskeletal system, which is composed of bone, tendon, ligament and irregular connective tissue, and closely associated with motor nerves and blood vessels. The intrinsic molecular signals regulating myogenesis have been extensively investigated. However, muscle development, homeostasis and regeneration require interactions with surrounding tissues and the cellular and molecular aspects of this dialogue have not been completely elucidated. During development and adult life, myogenic cells are closely associated with the different types of connective tissue. Connective tissues are defined as specialized (bone and cartilage), dense regular (tendon and ligament) and dense irregular connective tissue. The role of connective tissue in muscle morphogenesis has been investigated, thanks to the identification of transcription factors that characterize the different types of connective tissues. Here, we review the development of the various connective tissues in the context of the musculoskeletal system and highlight their important role in delivering information necessary for correct muscle morphogenesis, from the early step of myoblast differentiation to the late stage of muscle maturation. Interactions between muscle and connective tissue are also critical in the adult during muscle regeneration, as impairment of the regenerative potential after injury or in neuromuscular diseases results in the progressive replacement of the muscle mass by fibrotic tissue. We conclude that bi-directional communication between muscle and connective tissue is critical for a correct assembly of the musculoskeletal system during development as well as to maintain its homeostasis in the adult.
Collapse
Affiliation(s)
- Sonya Nassari
- Developmental Biology Laboratory, IBPS, Centre National de la Recherche Scientifique UMR7622, Institut National de la Santé Et de la Recherche Médicale U1156, Université Pierre et Marie Curie, Sorbonne Universités Paris, France
| | - Delphine Duprez
- Developmental Biology Laboratory, IBPS, Centre National de la Recherche Scientifique UMR7622, Institut National de la Santé Et de la Recherche Médicale U1156, Université Pierre et Marie Curie, Sorbonne Universités Paris, France
| | - Claire Fournier-Thibault
- Developmental Biology Laboratory, IBPS, Centre National de la Recherche Scientifique UMR7622, Institut National de la Santé Et de la Recherche Médicale U1156, Université Pierre et Marie Curie, Sorbonne Universités Paris, France
| |
Collapse
|
20
|
Havis E, Bonnin MA, Esteves de Lima J, Charvet B, Milet C, Duprez D. TGFβ and FGF promote tendon progenitor fate and act downstream of muscle contraction to regulate tendon differentiation during chick limb development. Development 2016; 143:3839-3851. [PMID: 27624906 DOI: 10.1242/dev.136242] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 08/25/2016] [Indexed: 01/02/2023]
Abstract
The molecular programme underlying tendon development has not been fully identified. Interactions with components of the musculoskeletal system are important for limb tendon formation. Limb tendons initiate their development independently of muscles; however, muscles are required for further tendon differentiation. We show that both FGF/ERK MAPK and TGFβ/SMAD2/3 signalling pathways are required and sufficient for SCX expression in chick undifferentiated limb cells, whereas the FGF/ERK MAPK pathway inhibits Scx expression in mouse undifferentiated limb mesodermal cells. During differentiation, muscle contraction is required to maintain SCX, TNMD and THBS2 expression in chick limbs. The activities of FGF/ERK MAPK and TGFβ/SMAD2/3 signalling pathways are decreased in tendons under immobilisation conditions. Application of FGF4 or TGFβ2 ligands prevents SCX downregulation in immobilised limbs. TGFβ2 but not FGF4 prevent TNMD and THBS2 downregulation under immobilisation conditions. We did not identify any intracellular crosstalk between both signalling pathways in their positive effect on SCX expression. Independently of each other, both FGF and TGFβ promote tendon commitment of limb mesodermal cells and act downstream of mechanical forces to regulate tendon differentiation during chick limb development.
Collapse
Affiliation(s)
- Emmanuelle Havis
- Sorbonne Universités, UPMC Univ Paris 06, CNRS UMR 7622, Inserm U1156, IBPS-Developmental Biology Laboratory, Paris F-75005, France
| | - Marie-Ange Bonnin
- Sorbonne Universités, UPMC Univ Paris 06, CNRS UMR 7622, Inserm U1156, IBPS-Developmental Biology Laboratory, Paris F-75005, France
| | - Joana Esteves de Lima
- Sorbonne Universités, UPMC Univ Paris 06, CNRS UMR 7622, Inserm U1156, IBPS-Developmental Biology Laboratory, Paris F-75005, France
| | - Benjamin Charvet
- Sorbonne Universités, UPMC Univ Paris 06, CNRS UMR 7622, Inserm U1156, IBPS-Developmental Biology Laboratory, Paris F-75005, France
| | - Cécile Milet
- Sorbonne Universités, UPMC Univ Paris 06, CNRS UMR 7622, Inserm U1156, IBPS-Developmental Biology Laboratory, Paris F-75005, France
| | - Delphine Duprez
- Sorbonne Universités, UPMC Univ Paris 06, CNRS UMR 7622, Inserm U1156, IBPS-Developmental Biology Laboratory, Paris F-75005, France
| |
Collapse
|
21
|
Satoh A, Makanae A, Nishimoto Y, Mitogawa K. FGF and BMP derived from dorsal root ganglia regulate blastema induction in limb regeneration in Ambystoma mexicanum. Dev Biol 2016; 417:114-25. [PMID: 27432514 DOI: 10.1016/j.ydbio.2016.07.005] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 06/14/2016] [Accepted: 07/11/2016] [Indexed: 10/21/2022]
Abstract
Urodele amphibians have a remarkable organ regeneration ability that is regulated by neural inputs. The identification of these neural inputs has been a challenge. Recently, Fibroblast growth factor (Fgf) and Bone morphogenic protein (Bmp) were shown to substitute for nerve functions in limb and tail regeneration in urodele amphibians. However, direct evidence of Fgf and Bmp being secreted from nerve endings and regulating regeneration has not yet been shown. Thus, it remained uncertain whether they were the nerve factors responsible for successful limb regeneration. To gather experimental evidence, the technical difficulties involved in the usage of axolotls had to be overcome. We achieved this by modifying the electroporation method. When Fgf8-AcGFP or Bmp7-AcGFP was electroporated into the axolotl dorsal root ganglia (DRG), GFP signals were detectable in the regenerating limb region. This suggested that Fgf8 and Bmp7 synthesized in neural cells in the DRG were delivered to the limbs through the long axons. Further knockdown experiments with double-stranded RNA interference resulted in impaired limb regeneration ability. These results strongly suggest that Fgf and Bmp are the major neural inputs that control the organ regeneration ability.
Collapse
Affiliation(s)
- Akira Satoh
- Okayama University, Research Core for Interdisciplinary Sciences (RCIS), 3-1-1 Tsushimanaka, Kita-ku, Okayama 700-8530, Japan.
| | - Aki Makanae
- Okayama University, Research Core for Interdisciplinary Sciences (RCIS), 3-1-1 Tsushimanaka, Kita-ku, Okayama 700-8530, Japan
| | - Yurie Nishimoto
- Okayama University, Research Core for Interdisciplinary Sciences (RCIS), 3-1-1 Tsushimanaka, Kita-ku, Okayama 700-8530, Japan
| | - Kazumasa Mitogawa
- Okayama University, Research Core for Interdisciplinary Sciences (RCIS), 3-1-1 Tsushimanaka, Kita-ku, Okayama 700-8530, Japan
| |
Collapse
|
22
|
Shukunami C, Yoshimoto Y, Takimoto A, Yamashita H, Hiraki Y. Molecular characterization and function of tenomodulin, a marker of tendons and ligaments that integrate musculoskeletal components. JAPANESE DENTAL SCIENCE REVIEW 2016; 52:84-92. [PMID: 28408960 PMCID: PMC5390337 DOI: 10.1016/j.jdsr.2016.04.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 03/16/2016] [Accepted: 04/01/2016] [Indexed: 01/14/2023] Open
Abstract
Tendons and ligaments are dense fibrous bands of connective tissue that integrate musculoskeletal components in vertebrates. Tendons connect skeletal muscles to the bone and function as mechanical force transmitters, whereas ligaments bind adjacent bones together to stabilize joints and restrict unwanted joint movement. Fibroblasts residing in tendons and ligaments are called tenocytes and ligamentocytes, respectively. Tenomodulin (Tnmd) is a type II transmembrane glycoprotein that is expressed at high levels in tenocytes and ligamentocytes, and is also present in periodontal ligament cells and tendon stem/progenitor cells. Tnmd is related to chondromodulin-1 (Chm1), a cartilage-derived angiogenesis inhibitor, and both Tnmd and Chm1 are expressed in the CD31− avascular mesenchyme. The conserved C-terminal hydrophobic domain of these proteins, which is characterized by the eight Cys residues to form four disulfide bonds, may have an anti-angiogenic function. This review highlights the molecular characterization and function of Tnmd, a specific marker of tendons and ligaments.
Collapse
Affiliation(s)
- Chisa Shukunami
- Department of Molecular Biology and Biochemistry, Division of Basic Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan
| | - Yuki Yoshimoto
- Department of Molecular Biology and Biochemistry, Division of Basic Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan
| | - Aki Takimoto
- Department of Cellular Differentiation, Institute for Frontier Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - Hiroshi Yamashita
- Department of Molecular Biology and Biochemistry, Division of Basic Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan
| | - Yuji Hiraki
- Department of Cellular Differentiation, Institute for Frontier Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| |
Collapse
|
23
|
Dex S, Lin D, Shukunami C, Docheva D. Tenogenic modulating insider factor: Systematic assessment on the functions of tenomodulin gene. Gene 2016; 587:1-17. [PMID: 27129941 DOI: 10.1016/j.gene.2016.04.051] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 04/20/2016] [Accepted: 04/25/2016] [Indexed: 02/08/2023]
Abstract
Tenomodulin (TNMD, Tnmd) is a gene highly expressed in tendon known to be important for tendon maturation with key implications for the residing tendon stem/progenitor cells as well as for the regulation of endothelial cell migration in chordae tendineae cordis in the heart and in experimental tumour models. This review aims at providing an encompassing overview of this gene and its protein. In addition, its known expression pattern as well as putative signalling pathways will be described. A chronological overview of the discovered functions of this gene in tendon and other tissues and cells is provided as well as its use as a tendon and ligament lineage marker is assessed in detail and discussed. Last, information about the possible connections between TNMD genomic mutations and mRNA expression to various diseases is delivered. Taken together this review offers a solid synopsis on the up-to-date information available about TNMD and aids at directing and focusing the future research to fully uncover the roles and implications of this interesting gene.
Collapse
Affiliation(s)
- Sarah Dex
- Experimental Surgery and Regenerative Medicine, Department of Surgery, Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Dasheng Lin
- Experimental Surgery and Regenerative Medicine, Department of Surgery, Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Chisa Shukunami
- Department of Molecular Biology and Biochemistry, Division of Basic Life Sciences, Institute of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Denitsa Docheva
- Experimental Surgery and Regenerative Medicine, Department of Surgery, Ludwig-Maximilians-University (LMU), Munich, Germany; Department of Medical Biology, Medical University-Plovdiv, Plovdiv, Bulgaria.
| |
Collapse
|
24
|
Booker BM, Friedrich T, Mason MK, VanderMeer JE, Zhao J, Eckalbar WL, Logan M, Illing N, Pollard KS, Ahituv N. Bat Accelerated Regions Identify a Bat Forelimb Specific Enhancer in the HoxD Locus. PLoS Genet 2016; 12:e1005738. [PMID: 27019019 PMCID: PMC4809552 DOI: 10.1371/journal.pgen.1005738] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 11/23/2015] [Indexed: 02/06/2023] Open
Abstract
The molecular events leading to the development of the bat wing remain largely unknown, and are thought to be caused, in part, by changes in gene expression during limb development. These expression changes could be instigated by variations in gene regulatory enhancers. Here, we used a comparative genomics approach to identify regions that evolved rapidly in the bat ancestor, but are highly conserved in other vertebrates. We discovered 166 bat accelerated regions (BARs) that overlap H3K27ac and p300 ChIP-seq peaks in developing mouse limbs. Using a mouse enhancer assay, we show that five Myotis lucifugus BARs drive gene expression in the developing mouse limb, with the majority showing differential enhancer activity compared to the mouse orthologous BAR sequences. These include BAR116, which is located telomeric to the HoxD cluster and had robust forelimb expression for the M. lucifugus sequence and no activity for the mouse sequence at embryonic day 12.5. Developing limb expression analysis of Hoxd10-Hoxd13 in Miniopterus natalensis bats showed a high-forelimb weak-hindlimb expression for Hoxd10-Hoxd11, similar to the expression trend observed for M. lucifugus BAR116 in mice, suggesting that it could be involved in the regulation of the bat HoxD complex. Combined, our results highlight novel regulatory regions that could be instrumental for the morphological differences leading to the development of the bat wing.
Collapse
Affiliation(s)
- Betty M. Booker
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, United States of America
- Institute for Human Genetics, University of California San Francisco, San Francisco, California, United States of America
| | - Tara Friedrich
- Institute for Human Genetics, University of California San Francisco, San Francisco, California, United States of America
- Gladstone Institutes, San Francisco, California, United States of America
| | - Mandy K. Mason
- Department of Molecular and Cell Biology, University of Cape Town, Cape Town, South Africa
| | - Julia E. VanderMeer
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, United States of America
- Institute for Human Genetics, University of California San Francisco, San Francisco, California, United States of America
| | - Jingjing Zhao
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, United States of America
- Institute for Human Genetics, University of California San Francisco, San Francisco, California, United States of America
- Key Laboratory of Advanced Control and Optimization for Chemical Processes of the Ministry of Education, East China University of Science and Technology, Shanghai, China
| | - Walter L. Eckalbar
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, United States of America
- Institute for Human Genetics, University of California San Francisco, San Francisco, California, United States of America
| | - Malcolm Logan
- Division of Developmental Biology, MRC-National Institute for Medical Research, Mill Hill, London, United Kingdom
- Randall Division of Cell and Molecular Biophysics, King’s College London, Guys Campus, London, United Kingdom
| | - Nicola Illing
- Department of Molecular and Cell Biology, University of Cape Town, Cape Town, South Africa
| | - Katherine S. Pollard
- Institute for Human Genetics, University of California San Francisco, San Francisco, California, United States of America
- Gladstone Institutes, San Francisco, California, United States of America
- Division of Biostatistics, University of California San Francisco, San Francisco, California, United States of America
- * E-mail: (KSP); (NA)
| | - Nadav Ahituv
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, United States of America
- Institute for Human Genetics, University of California San Francisco, San Francisco, California, United States of America
- * E-mail: (KSP); (NA)
| |
Collapse
|
25
|
Eckalbar WL, Schlebusch SA, Mason MK, Gill Z, Parker AV, Booker BM, Nishizaki S, Muswamba-Nday C, Terhune E, Nevonen KA, Makki N, Friedrich T, VanderMeer JE, Pollard KS, Carbone L, Wall JD, Illing N, Ahituv N. Transcriptomic and epigenomic characterization of the developing bat wing. Nat Genet 2016; 48:528-36. [PMID: 27019111 PMCID: PMC4848140 DOI: 10.1038/ng.3537] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 03/04/2016] [Indexed: 12/21/2022]
Abstract
Bats are the only mammals capable of powered flight, but little is known about the genetic determinants that shape their wings. Here we generated a genome for Miniopterus natalensis and performed RNA-seq and ChIP-seq (H3K27ac and H3K27me3) analyses on its developing forelimb and hindlimb autopods at sequential embryonic stages to decipher the molecular events that underlie bat wing development. Over 7,000 genes and several long noncoding RNAs, including Tbx5-as1 and Hottip, were differentially expressed between forelimb and hindlimb, and across different stages. ChIP-seq analysis identified thousands of regions that are differentially modified in forelimb and hindlimb. Comparative genomics found 2,796 bat-accelerated regions within H3K27ac peaks, several of which cluster near limb-associated genes. Pathway analyses highlighted multiple ribosomal proteins and known limb patterning signaling pathways as differentially regulated and implicated increased forelimb mesenchymal condensation in differential growth. In combination, our work outlines multiple genetic components that likely contribute to bat wing formation, providing insights into this morphological innovation.
Collapse
Affiliation(s)
- Walter L Eckalbar
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, California, USA.,Institute for Human Genetics, University of California, San Francisco, San Francisco, California, USA
| | - Stephen A Schlebusch
- Department of Molecular and Cell Biology, University of Cape Town, Cape Town, South Africa
| | - Mandy K Mason
- Department of Molecular and Cell Biology, University of Cape Town, Cape Town, South Africa
| | - Zoe Gill
- Department of Molecular and Cell Biology, University of Cape Town, Cape Town, South Africa
| | - Ash V Parker
- Department of Molecular and Cell Biology, University of Cape Town, Cape Town, South Africa
| | - Betty M Booker
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, California, USA.,Institute for Human Genetics, University of California, San Francisco, San Francisco, California, USA
| | - Sierra Nishizaki
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, California, USA.,Institute for Human Genetics, University of California, San Francisco, San Francisco, California, USA
| | | | - Elizabeth Terhune
- Oregon National Primate Research Center, Division of Neuroscience, Primate Genetics Section, Beaverton, Oregon, USA.,Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, Colorado, USA
| | - Kimberly A Nevonen
- Oregon National Primate Research Center, Division of Neuroscience, Primate Genetics Section, Beaverton, Oregon, USA
| | - Nadja Makki
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, California, USA.,Institute for Human Genetics, University of California, San Francisco, San Francisco, California, USA
| | - Tara Friedrich
- Institute for Human Genetics, University of California, San Francisco, San Francisco, California, USA.,Gladstone Institutes, San Francisco, California, USA
| | - Julia E VanderMeer
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, California, USA.,Institute for Human Genetics, University of California, San Francisco, San Francisco, California, USA
| | - Katherine S Pollard
- Institute for Human Genetics, University of California, San Francisco, San Francisco, California, USA.,Gladstone Institutes, San Francisco, California, USA.,Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, California, USA
| | - Lucia Carbone
- Oregon National Primate Research Center, Division of Neuroscience, Primate Genetics Section, Beaverton, Oregon, USA.,Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, Oregon, USA
| | - Jeff D Wall
- Institute for Human Genetics, University of California, San Francisco, San Francisco, California, USA.,Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, California, USA
| | - Nicola Illing
- Department of Molecular and Cell Biology, University of Cape Town, Cape Town, South Africa
| | - Nadav Ahituv
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, California, USA.,Institute for Human Genetics, University of California, San Francisco, San Francisco, California, USA
| |
Collapse
|
26
|
Tsutsumi R, Yamada S, Agata K. Functional joint regeneration is achieved using reintegration mechanism in Xenopus laevis. REGENERATION (OXFORD, ENGLAND) 2016; 3:26-38. [PMID: 27499877 PMCID: PMC4857750 DOI: 10.1002/reg2.49] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Revised: 10/16/2015] [Accepted: 10/16/2015] [Indexed: 01/08/2023]
Abstract
A functional joint requires integration of multiple tissues: the apposing skeletal elements should form an interlocking structure, and muscles should insert into skeletal tissues via tendons across the joint. Whereas newts can regenerate functional joints after amputation, Xenopus laevis regenerates a cartilaginous rod without joints, a "spike." Previously we reported that the reintegration mechanism between the remaining and regenerated tissues has a significant effect on regenerating joint morphogenesis during elbow joint regeneration in newt. Based on this insight into the importance of reintegration, we amputated frogs' limbs at the elbow joint and found that frogs could regenerate a functional elbow joint between the remaining tissues and regenerated spike. During regeneration, the regenerating cartilage was partially connected to the remaining articular cartilage to reform the interlocking structure of the elbow joint at the proximal end of the spike. Furthermore, the muscles of the remaining part inserted into the regenerated spike cartilage via tendons. This study might open up an avenue for analyzing molecular and cellular mechanisms of joint regeneration using Xenopus.
Collapse
Affiliation(s)
- Rio Tsutsumi
- Department of Biophysics, Graduate School of Science Kyoto University Kyoto Japan
| | - Shigehito Yamada
- Human Health Science, Graduate School of Medicine Kyoto University Kyoto Japan; Congenital Anomaly Research Center, Graduate School of Medicine Kyoto University Kyoto Japan
| | - Kiyokazu Agata
- Department of Biophysics, Graduate School of Science Kyoto University Kyoto Japan
| |
Collapse
|
27
|
Lu Y, Bradley JS, Siegel PB, Yang N, Johnson SE. Selection for divergent body size alters rates of embryonic skeletal muscle formation and muscle gene expression patterns. Dev Growth Differ 2015; 57:614-24. [PMID: 26660844 DOI: 10.1111/dgd.12250] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 10/05/2015] [Accepted: 10/13/2015] [Indexed: 11/28/2022]
Abstract
The impact of divergent selection for body size on embryogenesis is poorly understood. The objective of this experiment was to document skeletal muscle development during embryogenesis in two lines of chickens that display divergent growth as adults. Results reveal that after 54 generations of opposing selection from a common founder population, the embryos from the low weight select (LWS) line develop more rapidly during early embryogenesis than those from the high weight select (HWS) line. Muscle formation during the late embryonic period is more rapid and extensive in the HWS embryo than in the LWS contemporary. Isolated muscle progenitors from embryonic day 10 HWS embryos proliferated more rapidly, forming fibers sooner with a larger size than the LWS cells. The limited myogenic capacity of the LWS progenitor cells is not attributed to altered patterns of expression of Pax7, Pax3 or the myogenic regulatory factor genes. Members of the fibroblast growth factor family are potent mitogens and inhibitors of myoblast differentiation. Transcript abundance of FGF2 and FGF4 was measured in cultures of HWS and LWS progenitors as a function of time. The pattern of expression of FGF4 was similar between HWS and LWS with a large increase between days 1 and 3 followed by a reduction at day 5 of culture. Expression of FGF2 in LWS muscle cells did not change while a significant reduction in FGF2 expression was observed by day 5 in the HWS. Our results indicate that divergent selection for postnatal growth has altered embryonic development.
Collapse
Affiliation(s)
- Yue Lu
- National Engineering Laboratory for Animal Breeding and MOA Key Laboratory of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China.,Department of Animal and Poultry Sciences, Virginia Polytechnic and State University, Blacksburg, Virginia, 24061, USA
| | - Jennifer S Bradley
- Department of Animal and Poultry Sciences, Virginia Polytechnic and State University, Blacksburg, Virginia, 24061, USA
| | - Paul B Siegel
- Department of Animal and Poultry Sciences, Virginia Polytechnic and State University, Blacksburg, Virginia, 24061, USA
| | - Ning Yang
- National Engineering Laboratory for Animal Breeding and MOA Key Laboratory of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Sally E Johnson
- Department of Animal and Poultry Sciences, Virginia Polytechnic and State University, Blacksburg, Virginia, 24061, USA
| |
Collapse
|
28
|
Abstract
The tongue and mandible have common origins. They arise simultaneously from the mandibular arch and are coordinated in their development and growth, which is evident from several clinical conditions such as Pierre Robin sequence. Here, we review in detail the molecular networks controlling both mandible and tongue development. We also discuss their mechanical relationship and evolution as well as the potential for stem cell-based therapies for disorders affecting these organs.
Collapse
Affiliation(s)
- Carolina Parada
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, California, USA.
| | - Yang Chai
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, California, USA.
| |
Collapse
|
29
|
Gaut L, Duprez D. Tendon development and diseases. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2015; 5:5-23. [PMID: 26256998 DOI: 10.1002/wdev.201] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Revised: 06/15/2015] [Accepted: 06/20/2015] [Indexed: 12/22/2022]
Abstract
Tendon is a uniaxial connective tissue component of the musculoskeletal system. Tendon is involved in force transmission between muscle and bone. Tendon injury is very common and debilitating but tendon repair remains a clinical challenge for orthopedic medicine. In vertebrates, tendon is mainly composed of type I collagen fibrils, displaying a parallel organization along the tendon axis. The tendon-specific spatial organization of type I collagen provides the mechanical properties for tendon function. In contrast to other components of the musculoskeletal system, tendon biology is poorly understood. An important goal in tendon biology is to understand the mechanisms involved in the production and assembly of type I collagen fibrils during development, postnatal formation, and healing processes in order to design new therapies for tendon repair. In this review we highlight the current understanding of the molecular and mechanical signals known to be involved in tenogenesis during development, and how development provides insights into tendon healing processes. WIREs Dev Biol 2016, 5:5-23. doi: 10.1002/wdev.201 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Ludovic Gaut
- CNRS UMR 7622, IBPS-Developmental Biology Laboratory, Paris, France.,Sorbonne Universités, UPMC Univ Paris 06, IBPS-Developmental Biology Laboratory, Paris, France.,Inserm U1156, Paris, France
| | - Delphine Duprez
- CNRS UMR 7622, IBPS-Developmental Biology Laboratory, Paris, France.,Sorbonne Universités, UPMC Univ Paris 06, IBPS-Developmental Biology Laboratory, Paris, France.,Inserm U1156, Paris, France
| |
Collapse
|
30
|
Huang AH, Lu HH, Schweitzer R. Molecular regulation of tendon cell fate during development. J Orthop Res 2015; 33:800-12. [PMID: 25664867 DOI: 10.1002/jor.22834] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 01/16/2015] [Indexed: 02/04/2023]
Abstract
Although there have been several advances identifying novel mediators of tendon induction, differentiation, and patterning, much of the basic landscape of tendon biology from developmental stages onward remain almost completely undefined. During the New Frontiers in Tendon Research meeting, a group of developmental biologists with expertise across musculoskeletal disciplines identified key challenges for the tendon development field. The tools generated and the molecular regulators identified in developmental research have enhanced mechanistic studies in tendon injury and repair, both by defining benchmarks for success, as well as informing regenerative strategies. To address the needs of the orthopedic research community, this review will therefore focus on three key areas in tendon development that may have critical implications for the fields of tendon repair/regeneration and tendon tissue engineering, including functional markers of tendon cell identity, signaling regulators of tendon induction and differentiation, and in vitro culture models for tendon cell differentiation. Our goal is to provide a useful list of the currently known molecular players and their function in tendon differentiation within the context of development.
Collapse
Affiliation(s)
- Alice H Huang
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY
| | | | | |
Collapse
|
31
|
Zelzer E, Blitz E, Killian ML, Thomopoulos S. Tendon-to-bone attachment: from development to maturity. ACTA ACUST UNITED AC 2015; 102:101-12. [PMID: 24677726 DOI: 10.1002/bdrc.21056] [Citation(s) in RCA: 120] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Accepted: 01/14/2014] [Indexed: 12/13/2022]
Abstract
The attachment between tendon and bone occurs across a complex transitional tissue that minimizes stress concentrations and allows for load transfer between muscles and skeleton. This unique tissue cannot be reconstructed following injury, leading to high incidence of recurrent failure and stressing the need for new clinical approaches. This review describes the current understanding of the development and function of the attachment site between tendon and bone. The embryonic attachment unit, namely, the tip of the tendon and the bone eminence into which it is inserted, was recently shown to develop modularly from a unique population of Sox9- and Scx-positive cells, which are distinct from tendon fibroblasts and chondrocytes. The fate and differentiation of these cells is regulated by transforming growth factor beta and bone morphogenetic protein signaling, respectively. Muscle loads are then necessary for the tissue to mature and mineralize. Mineralization of the attachment unit, which occurs postnatally at most sites, is largely controlled by an Indian hedgehog/parathyroid hormone-related protein feedback loop. A number of fundamental questions regarding the development of this remarkable attachment system require further study. These relate to the signaling mechanism that facilitates the formation of an interface with a gradient of cellular and extracellular phenotypes, as well as to the interactions between tendon and bone at the point of attachment.
Collapse
Affiliation(s)
- Elazar Zelzer
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | | | | | | |
Collapse
|
32
|
Abu-Elmagd M, Goljanek Whysall K, Wheeler G, Münsterberg A. Sprouty2 mediated tuning of signalling is essential for somite myogenesis. BMC Med Genomics 2015; 8 Suppl 1:S8. [PMID: 25783674 PMCID: PMC4315326 DOI: 10.1186/1755-8794-8-s1-s8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Background Negative regulators of signal transduction cascades play critical roles in controlling different aspects of normal embryonic development. Sprouty2 (Spry2) negatively regulates receptor tyrosine kinases (RTK) and FGF signalling and is important in differentiation, cell migration and proliferation. In vertebrate embryos, Spry2 is expressed in paraxial mesoderm and in forming somites. Expression is maintained in the myotome until late stages of somite differentiation. However, its role and mode of action during somite myogenesis is still unclear. Results Here, we analysed chick Spry2 expression and showed that it overlaps with that of myogenic regulatory factors MyoD and Mgn. Targeted mis-expression of Spry2 led to inhibition of myogenesis, whilst its C-terminal domain led to an increased number of myogenic cells by stimulating cell proliferation. Conclusions Spry2 is expressed in somite myotomes and its expression overlaps with myogenic regulatory factors. Overexpression and dominant-negative interference showed that Spry2 plays a crucial role in regulating chick myogenesis by fine tuning of FGF signaling through a negative feedback loop. We also propose that mir-23, mir-27 and mir-128 could be part of the negative feedback loop mechanism. Our analysis is the first to shed some light on in vivo Spry2 function during chick somite myogenesis.
Collapse
|
33
|
Abstract
Tendon is a crucial component of the musculoskeletal system. Tendons connect muscle to bone and transmit forces to produce motion. Chronic and acute tendon injuries are very common and result in considerable pain and disability. The management of tendon injuries remains a challenge for clinicians. Effective treatments for tendon injuries are lacking because the understanding of tendon biology lags behind that of the other components of the musculoskeletal system. Animal and cellular models have been developed to study tendon-cell differentiation and tendon repair following injury. These studies have highlighted specific growth factors and transcription factors involved in tenogenesis during developmental and repair processes. Mechanical factors also seem to be essential for tendon development, homeostasis and repair. Mechanical signals are transduced via molecular signalling pathways that trigger adaptive responses in the tendon. Understanding the links between the mechanical and biological parameters involved in tendon development, homeostasis and repair is prerequisite for the identification of effective treatments for chronic and acute tendon injuries.
Collapse
Affiliation(s)
- Geoffroy Nourissat
- Service de chirurgie orthopédique et traumatologique, INSERM UMR_S938, DHU i2B, Assistance Publique-Hopitaux de Paris, Hôpital Saint-Antoine, 184 rue du Faubourg Saint-Antoine, Paris 75012, France
| | - Francis Berenbaum
- Service de rhumatologie, INSERM UMR_S938, DHU i2B, Assistance Publique-Hopitaux de Paris, Hôpital Saint-Antoine, 184 rue du Faubourg Saint-Antoine, Paris 75012, France
| | - Delphine Duprez
- Centre national de la recherche scientifique UMR 7622, IBPS Developmental Biology Laboratory, F-75005, Paris 5005, France
| |
Collapse
|
34
|
Specific pattern of cell cycle during limb fetal myogenesis. Dev Biol 2014; 392:308-23. [DOI: 10.1016/j.ydbio.2014.05.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Revised: 03/24/2014] [Accepted: 05/21/2014] [Indexed: 01/20/2023]
|
35
|
Iwata JI, Suzuki A, Pelikan RC, Ho TV, Chai Y. Noncanonical transforming growth factor β (TGFβ) signaling in cranial neural crest cells causes tongue muscle developmental defects. J Biol Chem 2013; 288:29760-70. [PMID: 23950180 DOI: 10.1074/jbc.m113.493551] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Microglossia is a congenital birth defect in humans and adversely impacts quality of life. In vertebrates, tongue muscle derives from the cranial mesoderm, whereas tendons and connective tissues in the craniofacial region originate from cranial neural crest (CNC) cells. Loss of transforming growth factor β (TGFβ) type II receptor in CNC cells in mice (Tgfbr2(fl/fl);Wnt1-Cre) causes microglossia due to a failure of cell-cell communication between cranial mesoderm and CNC cells during tongue development. However, it is still unclear how TGFβ signaling in CNC cells regulates the fate of mesoderm-derived myoblasts during tongue development. Here we show that activation of the cytoplasmic and nuclear tyrosine kinase 1 (ABL1) cascade in Tgfbr2(fl/fl);Wnt1-Cre mice results in a failure of CNC-derived cell differentiation followed by a disruption of TGFβ-mediated induction of growth factors and reduction of myogenic cell proliferation and differentiation activities. Among the affected growth factors, the addition of fibroblast growth factor 4 (FGF4) and neutralizing antibody for follistatin (FST; an antagonist of bone morphogenetic protein (BMP)) could most efficiently restore cell proliferation, differentiation, and organization of muscle cells in the tongue of Tgfbr2(fl/fl);Wnt1-Cre mice. Thus, our data indicate that CNC-derived fibroblasts regulate the fate of mesoderm-derived myoblasts through TGFβ-mediated regulation of FGF and BMP signaling during tongue development.
Collapse
Affiliation(s)
- Jun-ichi Iwata
- From the Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, California 90033
| | | | | | | | | |
Collapse
|
36
|
Jiang Z, Price CA. Differential actions of fibroblast growth factors on intracellular pathways and target gene expression in bovine ovarian granulosa cells. Reproduction 2012; 144:625-32. [PMID: 22956519 DOI: 10.1530/rep-12-0199] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Several fibroblast growth factors (FGFs), including FGF1, FGF4 and FGF10, alter ovarian granulosa cell function. These ligands exhibit different patterns of receptor activation, and their mechanisms of action on granulosa cells remain unknown. The objective of this study was to identify the major pathways and target genes activated by FGF1, FGF4 and FGF10 in primary oestrogenic granulosa cells cultured under serum-free conditions. FGF1 and FGF4 increased levels of mRNA encoding Sprouty family members, SPRY2 and SPRY4, and the orphan nuclear receptors NR4A1 and NR4A3. Both FGF1 and FGF4 decreased levels of mRNA encoding SPRY3 and the pro-apoptotic factor BAX. FGF1 but not FGF4 stimulated expression of the cell cycle regulator, GADD45B. In contrast, FGF10 altered the expression of none of these genes. Western blot demonstrated that FGF4 activated ERK1/2 and Akt signalling rapidly and transiently, whereas FGF10 elicited a modest and delayed activation of ERK1/2. These data show that FGF1 and FGF4 activate typical FGF signalling pathways in granulosa cells, whereas FGF10 activates atypical pathways.
Collapse
Affiliation(s)
- Zhongliang Jiang
- College of Animal Science and Technology, Northwestern A&F University, Yangling, ShaanXi, China
| | | |
Collapse
|
37
|
Han D, Zhao H, Parada C, Hacia JG, Bringas P, Chai Y. A TGFβ-Smad4-Fgf6 signaling cascade controls myogenic differentiation and myoblast fusion during tongue development. Development 2012; 139:1640-50. [PMID: 22438570 DOI: 10.1242/dev.076653] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The tongue is a muscular organ and plays a crucial role in speech, deglutition and taste. Despite the important physiological functions of the tongue, little is known about the regulatory mechanisms of tongue muscle development. TGFβ family members play important roles in regulating myogenesis, but the functional significance of Smad-dependent TGFβ signaling in regulating tongue skeletal muscle development remains unclear. In this study, we have investigated Smad4-mediated TGFβ signaling in the development of occipital somite-derived myogenic progenitors during tongue morphogenesis through tissue-specific inactivation of Smad4 (using Myf5-Cre;Smad4(flox/flox) mice). During the initiation of tongue development, cranial neural crest (CNC) cells occupy the tongue buds before myogenic progenitors migrate into the tongue primordium, suggesting that CNC cells play an instructive role in guiding tongue muscle development. Moreover, ablation of Smad4 results in defects in myogenic terminal differentiation and myoblast fusion. Despite compromised muscle differentiation, tendon formation appears unaffected in the tongue of Myf5-Cre;Smad4(flox/flox) mice, suggesting that the differentiation and maintenance of CNC-derived tendon cells are independent of Smad4-mediated signaling in myogenic cells in the tongue. Furthermore, loss of Smad4 results in a significant reduction in expression of several members of the FGF family, including Fgf6 and Fgfr4. Exogenous Fgf6 partially rescues the tongue myoblast fusion defect of Myf5-Cre;Smad4(flox/flox) mice. Taken together, our study demonstrates that a TGFβ-Smad4-Fgf6 signaling cascade plays a crucial role in myogenic cell fate determination and lineage progression during tongue myogenesis.
Collapse
Affiliation(s)
- Dong Han
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA 90033, USA
| | | | | | | | | | | |
Collapse
|
38
|
Parada C, Han D, Chai Y. Molecular and cellular regulatory mechanisms of tongue myogenesis. J Dent Res 2012; 91:528-35. [PMID: 22219210 DOI: 10.1177/0022034511434055] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The tongue exerts crucial functions in our daily life. However, we know very little about the regulatory mechanisms of mammalian tongue development. In this review, we summarize recent findings of the molecular and cellular mechanisms that control tissue-tissue interactions during tongue morphogenesis. Specifically, cranial neural crest cells (CNCC) lead the initiation of tongue bud formation and contribute to the interstitial connective tissue, which ultimately compartmentalizes tongue muscles and serves as their attachments. Occipital somite-derived cells migrate into the tongue primordium and give rise to muscle cells in the tongue. The intimate relationship between CNCC- and mesoderm-derived cells, as well as growth and transcription factors that have been shown to be crucial for tongue myogenesis, clearly indicate that tissue-tissue interactions play an important role in regulating tongue morphogenesis.
Collapse
Affiliation(s)
- C Parada
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA 90033, USA
| | | | | |
Collapse
|
39
|
Nowick K, Fields C, Gernat T, Caetano-Anolles D, Kholina N, Stubbs L. Gain, loss and divergence in primate zinc-finger genes: a rich resource for evolution of gene regulatory differences between species. PLoS One 2011; 6:e21553. [PMID: 21738707 PMCID: PMC3126818 DOI: 10.1371/journal.pone.0021553] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2011] [Accepted: 06/01/2011] [Indexed: 12/14/2022] Open
Abstract
The molecular changes underlying major phenotypic differences between humans and other primates are not well understood, but alterations in gene regulation are likely to play a major role. Here we performed a thorough evolutionary analysis of the largest family of primate transcription factors, the Krüppel-type zinc finger (KZNF) gene family. We identified and curated gene and pseudogene models for KZNFs in three primate species, chimpanzee, orangutan and rhesus macaque, to allow for a comparison with the curated set of human KZNFs. We show that the recent evolutionary history of primate KZNFs has been complex, including many lineage-specific duplications and deletions. We found 213 species-specific KZNFs, among them 7 human-specific and 23 chimpanzee-specific genes. Two human-specific genes were validated experimentally. Ten genes have been lost in humans and 13 in chimpanzees, either through deletion or pseudogenization. We also identified 30 KZNF orthologs with human-specific and 42 with chimpanzee-specific sequence changes that are predicted to affect DNA binding properties of the proteins. Eleven of these genes show signatures of accelerated evolution, suggesting positive selection between humans and chimpanzees. During primate evolution the most extensive re-shaping of the KZNF repertoire, including most gene additions, pseudogenizations, and structural changes occurred within the subfamily homininae. Using zinc finger (ZNF) binding predictions, we suggest potential impact these changes have had on human gene regulatory networks. The large species differences in this family of TFs stands in stark contrast to the overall high conservation of primate genomes and potentially represents a potent driver of primate evolution.
Collapse
Affiliation(s)
- Katja Nowick
- Institute for Genomic Biology, University of Illinois, Urbana, Illinois, United States of America
- Department of Cell and Developmental Biology, University of Illinois, Urbana, Illinois, United States of America
| | - Christopher Fields
- Institute for Genomic Biology, University of Illinois, Urbana, Illinois, United States of America
| | - Tim Gernat
- Institute for Genomic Biology, University of Illinois, Urbana, Illinois, United States of America
| | - Derek Caetano-Anolles
- Institute for Genomic Biology, University of Illinois, Urbana, Illinois, United States of America
- Department of Cell and Developmental Biology, University of Illinois, Urbana, Illinois, United States of America
| | - Nadezda Kholina
- Institute for Genomic Biology, University of Illinois, Urbana, Illinois, United States of America
- Department of Cell and Developmental Biology, University of Illinois, Urbana, Illinois, United States of America
| | - Lisa Stubbs
- Institute for Genomic Biology, University of Illinois, Urbana, Illinois, United States of America
- Department of Cell and Developmental Biology, University of Illinois, Urbana, Illinois, United States of America
- * E-mail:
| |
Collapse
|
40
|
Hasson P. "Soft" tissue patterning: muscles and tendons of the limb take their form. Dev Dyn 2011; 240:1100-7. [PMID: 21438070 DOI: 10.1002/dvdy.22608] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/22/2011] [Indexed: 12/18/2022] Open
Abstract
The musculoskeletal system grants our bodies a vast range of movements. Because it is mainly composed of easily identifiable components, it serves as an ideal model to study patterning of the specific tissues that make up the organ. Surprisingly, although critical for the function of the musculoskeletal system, understanding of the embryonic processes that regulate muscle and tendon patterning is very limited. The recent identification of specific markers and the reagents stemming from them has revealed some of the molecular events regulating patterning of these soft tissues. This review will focus on some of the current work, with an emphasis on the roles of the muscle connective tissue, and discuss several key points that addressing them will advance our understanding of these patterning events.
Collapse
Affiliation(s)
- Peleg Hasson
- Department of Anatomy and Cell Biology, The Rappaport Family Institute for Research in the Medical Sciences, Technion-Israel Institute of Technology, Bat Galim, Haifa, Israel.
| |
Collapse
|
41
|
Moura RS, Coutinho-Borges JP, Pacheco AP, Damota PO, Correia-Pinto J. FGF signaling pathway in the developing chick lung: expression and inhibition studies. PLoS One 2011; 6:e17660. [PMID: 21412430 PMCID: PMC3055888 DOI: 10.1371/journal.pone.0017660] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2010] [Accepted: 02/07/2011] [Indexed: 12/02/2022] Open
Abstract
Background Fibroblast growth factors (FGF) are essential key players during embryonic development. Through their specific cognate receptors (FGFR) they activate intracellular cascades, finely regulated by modulators such as Sprouty. Several FGF ligands (FGF1, 2, 7, 9, 10 and 18) signaling through the four known FGFRs, have been implicated in lung morphogenesis. Although much is known about mammalian lung, so far, the avian model has not been explored for lung studies. Methodology/Principal Findings In this study we provide the first description of fgf10, fgfr1-4 and spry2 expression patterns in early stages of chick lung development by in situ hybridization and observe that they are expressed similarly to their mammalian counterparts. Furthermore, aiming to determine a role for FGF signaling in chick lung development, in vitro FGFR inhibition studies were performed. Lung explants treated with an FGF receptor antagonist (SU5402) presented an impairment of secondary branch formation after 48 h of culture; moreover, abnormal lung growth with a cystic appearance of secondary bronchi and reduction of the mesenchymal tissue was observed. Branching and morphometric analysis of lung explants confirmed that FGFR inhibition impaired branching morphogenesis and induced a significant reduction of the mesenchyme. Conclusions/Significance This work demonstrates that FGFRs are essential for the epithelial-mesenchymal interactions that determine epithelial branching and mesenchymal growth and validate the avian embryo as a good model for pulmonary studies, namely to explore the FGF pathway as a therapeutic target.
Collapse
Affiliation(s)
- Rute S Moura
- School of Health Sciences, Life and Health Sciences Research Institute (ICVS), University of Minho, Braga, Portugal.
| | | | | | | | | |
Collapse
|
42
|
Lejard V, Blais F, Guerquin MJ, Bonnet A, Bonnin MA, Havis E, Malbouyres M, Bidaud CB, Maro G, Gilardi-Hebenstreit P, Rossert J, Ruggiero F, Duprez D. EGR1 and EGR2 involvement in vertebrate tendon differentiation. J Biol Chem 2010; 286:5855-67. [PMID: 21173153 DOI: 10.1074/jbc.m110.153106] [Citation(s) in RCA: 158] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
The molecules involved in vertebrate tendon formation during development remain largely unknown. To date, only two DNA-binding proteins have been identified as being involved in vertebrate tendon formation, the basic helix-loop-helix transcription factor Scleraxis and, recently, the Mohawk homeobox gene. We investigated the involvement of the early growth response transcription factors Egr1 and Egr2 in vertebrate tendon formation. We established that Egr1 and Egr2 expression in tendon cells was correlated with the increase of collagen expression during tendon cell differentiation in embryonic limbs. Vertebrate tendon differentiation relies on a muscle-derived FGF (fibroblast growth factor) signal. FGF4 was able to activate the expression of Egr genes and that of the tendon-associated collagens in chick limbs. Egr gene misexpression experiments using the chick model allowed us to establish that either Egr gene has the ability to induce de novo expression of the reference tendon marker scleraxis, the main tendon collagen Col1a1, and other tendon-associated collagens Col3a1, Col5a1, Col12a1, and Col14a1. Mouse mutants for Egr1 or Egr2 displayed reduced amounts of Col1a1 transcripts and a decrease in the number of collagen fibrils in embryonic tendons. Moreover, EGR1 and EGR2 trans-activated the mouse Col1a1 proximal promoter and were recruited to the tendon regulatory regions of this promoter. These results identify EGRs as novel DNA-binding proteins involved in vertebrate tendon differentiation by regulating type I collagen production.
Collapse
Affiliation(s)
- Véronique Lejard
- Université Pierre et Marie Curie, CNRS UMR 7622, Paris 75005, France
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Schweitzer R, Zelzer E, Volk T. Connecting muscles to tendons: tendons and musculoskeletal development in flies and vertebrates. Development 2010; 137:2807-17. [PMID: 20699295 DOI: 10.1242/dev.047498] [Citation(s) in RCA: 178] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The formation of the musculoskeletal system represents an intricate process of tissue assembly involving heterotypic inductive interactions between tendons, muscles and cartilage. An essential component of all musculoskeletal systems is the anchoring of the force-generating muscles to the solid support of the organism: the skeleton in vertebrates and the exoskeleton in invertebrates. Here, we discuss recent findings that illuminate musculoskeletal assembly in the vertebrate embryo, findings that emphasize the reciprocal interactions between the forming tendons, muscle and cartilage tissues. We also compare these events with those of the corresponding system in the Drosophila embryo, highlighting distinct and common pathways that promote efficient locomotion while preserving the form of the organism.
Collapse
Affiliation(s)
- Ronen Schweitzer
- Shriners Hospital for Children, Research Division, Portland, OR 97239, USA.
| | | | | |
Collapse
|