1
|
Oliveira Filho HS, Duarte JLC, Paranhos GF, Santos KMOL, Oliveira RL, Vasconcelos IFF, De Araújo JL. Polymelia and concurrent intestinal duplication and cloacal atresia in a broiler chick: case report and literature review. J Vet Diagn Invest 2024; 36:859-863. [PMID: 39267415 PMCID: PMC11529053 DOI: 10.1177/10406387241273909] [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] [Indexed: 09/17/2024] Open
Abstract
Congenital malformations are a highly diverse group of conditions reported in both humans and animals, characterized by defects in morphogenesis observed at birth. Although most cases are idiopathic, genetic and environmental factors may be involved. The frequency of such conditions varies with species, geographic regions, and the specific malformation involved. In polymelia, supernumerary limbs are attached to different parts of the body. Gastrointestinal duplications are described less frequently and can be associated with polymelia. Cloacal atresia is among the least-reported malformations in avian species, described only once in a kiwi. Here we describe a case with these 3 malformations in a single broiler chick (Gallus gallus domesticus) and provide a literature review about the occurrence of these malformations in birds. The 3-d-old chick also had an unidentified structure projecting from the pygostyle region. We performed clinical, radiographic, and postmortem examinations. The intestinal duplication was identified only during the postmortem evaluation. Detailed descriptions of avian congenital malformations are scarce. Although similar cases have been reported, we retrieved no cases of concurrent polymelia, intestinal duplication, and cloacal atresia in broiler chickens in our literature search, suggesting that the simultaneous occurrence of these conditions has not been reported previously in this species.
Collapse
Affiliation(s)
- Hodias S. Oliveira Filho
- Department of Veterinary Sciences, Universidade Federal da Paraíba (UFPB), Areia, Paraiba, Brazil
| | - José L. C. Duarte
- Department of Veterinary Sciences, Universidade Federal da Paraíba (UFPB), Areia, Paraiba, Brazil
| | - Gabriel F. Paranhos
- Department of Veterinary Sciences, Universidade Federal da Paraíba (UFPB), Areia, Paraiba, Brazil
| | - Kin M. O. L. Santos
- Department of Veterinary Sciences, Universidade Federal da Paraíba (UFPB), Areia, Paraiba, Brazil
| | - Rafael L. Oliveira
- Department of Veterinary Sciences, Universidade Federal da Paraíba (UFPB), Areia, Paraiba, Brazil
| | - Igor F. F. Vasconcelos
- Department of Veterinary Sciences, Universidade Federal da Paraíba (UFPB), Areia, Paraiba, Brazil
| | - Jeann L. De Araújo
- Department of Veterinary Sciences, Universidade Federal da Paraíba (UFPB), Areia, Paraiba, Brazil
| |
Collapse
|
2
|
Quadri N, Upadhyai P. Primary cilia in skeletal development and disease. Exp Cell Res 2023; 431:113751. [PMID: 37574037 DOI: 10.1016/j.yexcr.2023.113751] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 08/09/2023] [Accepted: 08/11/2023] [Indexed: 08/15/2023]
Abstract
Primary cilia are non-motile, microtubule-based sensory organelle present in most vertebrate cells with a fundamental role in the modulation of organismal development, morphogenesis, and repair. Here we focus on the role of primary cilia in embryonic and postnatal skeletal development. We examine evidence supporting its involvement in physiochemical and developmental signaling that regulates proliferation, patterning, differentiation and homeostasis of osteoblasts, chondrocytes, and their progenitor cells in the skeleton. We discuss how signaling effectors in mechanotransduction and bone development, such as Hedgehog, Wnt, Fibroblast growth factor and second messenger pathways operate at least in part at the primary cilium. The relevance of primary cilia in bone formation and maintenance is underscored by a growing list of rare genetic skeletal ciliopathies. We collate these findings and summarize the current understanding of molecular factors and mechanisms governing primary ciliogenesis and ciliary function in skeletal development and disease.
Collapse
Affiliation(s)
- Neha Quadri
- Department of Medical Genetics, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India
| | - Priyanka Upadhyai
- Department of Medical Genetics, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India.
| |
Collapse
|
3
|
Sedas Perez S, McQueen C, Stainton H, Pickering J, Chinnaiya K, Saiz-Lopez P, Placzek M, Ros MA, Towers M. Fgf signalling triggers an intrinsic mesodermal timer that determines the duration of limb patterning. Nat Commun 2023; 14:5841. [PMID: 37730682 PMCID: PMC10511490 DOI: 10.1038/s41467-023-41457-6] [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: 02/22/2023] [Accepted: 09/05/2023] [Indexed: 09/22/2023] Open
Abstract
Complex signalling between the apical ectodermal ridge (AER - a thickening of the distal epithelium) and the mesoderm controls limb patterning along the proximo-distal axis (humerus to digits). However, the essential in vivo requirement for AER-Fgf signalling makes it difficult to understand the exact roles that it fulfils. To overcome this barrier, we developed an amenable ex vivo chick wing tissue explant system that faithfully replicates in vivo parameters. Using inhibition experiments and RNA-sequencing, we identify a transient role for Fgfs in triggering the distal patterning phase. Fgfs are then dispensable for the maintenance of an intrinsic mesodermal transcriptome, which controls proliferation/differentiation timing and the duration of patterning. We also uncover additional roles for Fgf signalling in maintaining AER-related gene expression and in suppressing myogenesis. We describe a simple logic for limb patterning duration, which is potentially applicable to other systems, including the main body axis.
Collapse
Affiliation(s)
- Sofia Sedas Perez
- School of Biosciences, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK
| | - Caitlin McQueen
- School of Biosciences, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK
- Chester Medical School, Chester, CH2 1BR, UK
| | - Holly Stainton
- School of Biosciences, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK
| | - Joseph Pickering
- School of Biosciences, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK
| | - Kavitha Chinnaiya
- School of Biosciences, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK
| | - Patricia Saiz-Lopez
- Instituto de Biomedicina y Biotecnología de Cantabria, IBBTEC (CSIC-Universidad de Cantabria), 39011, Santander, Spain
- Departamento de Anatomía y Biología Celular Facultad de Medicina, Universidad de Cantabria, 39011, Santander, Spain
| | - Marysia Placzek
- School of Biosciences, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK
| | - Maria A Ros
- Instituto de Biomedicina y Biotecnología de Cantabria, IBBTEC (CSIC-Universidad de Cantabria), 39011, Santander, Spain
- Departamento de Anatomía y Biología Celular Facultad de Medicina, Universidad de Cantabria, 39011, Santander, Spain
| | - Matthew Towers
- School of Biosciences, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK.
| |
Collapse
|
4
|
Derham JM, Kalsotra A. The discovery, function, and regulation of epithelial splicing regulatory proteins (ESRP) 1 and 2. Biochem Soc Trans 2023; 51:1097-1109. [PMID: 37314029 PMCID: PMC11298080 DOI: 10.1042/bst20221124] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 06/01/2023] [Accepted: 06/05/2023] [Indexed: 06/15/2023]
Abstract
Alternative splicing is a broad and evolutionarily conserved mechanism to diversify gene expression and functionality. The process relies on RNA binding proteins (RBPs) to recognize and bind target sequences in pre-mRNAs, which allows for the inclusion or skipping of various alternative exons. One recently discovered family of RBPs is the epithelial splicing regulatory proteins (ESRP) 1 and 2. Here, we discuss the structure and physiological function of the ESRPs in a variety of contexts. We emphasize the current understanding of their splicing activities, using the classic example of fibroblast growth factor receptor 2 mutually exclusive splicing. We also describe the mechanistic roles of ESRPs in coordinating the splicing and functional output of key signaling pathways that support the maintenance of, or shift between, epithelial and mesenchymal cell states. In particular, we highlight their functions in the development of mammalian limbs, the inner ear, and craniofacial structure while discussing the genetic and biochemical evidence that showcases their conserved roles in tissue regeneration, disease, and cancer pathogenesis.
Collapse
Affiliation(s)
- Jessica M. Derham
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Auinash Kalsotra
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, IL, USA
- Cancer Center @ Illinois, University of Illinois Urbana-Champaign, Urbana, IL, USA
- Carl R. Woese Institute of Genomic Biology, University of Illinois Urbana-Champaign, Urbana, IL, USA
| |
Collapse
|
5
|
Purushothaman S, Lopez Aviña BB, Seifert AW. Sonic hedgehog is Essential for Proximal-Distal Outgrowth of the Limb Bud in Salamanders. Front Cell Dev Biol 2022; 10:797352. [PMID: 35433673 PMCID: PMC9010949 DOI: 10.3389/fcell.2022.797352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 01/24/2022] [Indexed: 11/20/2022] Open
Abstract
The developing forelimb has been a foundational model to understand how specified progenitor cells integrate genetic information to produce the tetrapod limb bauplan. Although the reigning hypothesis is that all tetrapods develop limbs in a similar manner, recent work suggests that urodeles have evolved a derived mode of limb dvelopment. Here, we demonstrate through pharmacological and genetic inactivation of Sonic hedgehog (Shh) signaling in axolotls that Shh directs expansion and survival of limb progenitor cells in addition to patterning the limb across the proximodistal and antero-posterior axis. In contrast to inactivation of Shh in mouse or chick embryos where a humerus, radius, and single digit develop, Shh crispant axolotls completely lack forelimbs. In rescuing limb development by implanting SHH-N protein beads into the nascent limb field of Shh crispants, we show that the limb field is specified in the absence of Shh and that hedgehog pathway activation is required to initiate proximodistal outgrowth. When our results are examined alongside other derived aspects of salamander limb development and placed in a phylogenetic context, a new hypothesis emerges whereby the ability for cells at an amputation plane to activate morphogenesis and regenerate a limb may have evolved uniquely in urodeles.
Collapse
|
6
|
Chioni AM, Grose RP. Biological Significance and Targeting of the FGFR Axis in Cancer. Cancers (Basel) 2021; 13:5681. [PMID: 34830836 PMCID: PMC8616401 DOI: 10.3390/cancers13225681] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 11/08/2021] [Accepted: 11/11/2021] [Indexed: 12/15/2022] Open
Abstract
The pleiotropic effects of fibroblast growth factors (FGFs), the widespread expression of all seven signalling FGF receptors (FGFRs) throughout the body, and the dramatic phenotypes shown by many FGF/R knockout mice, highlight the diversity, complexity and functional importance of FGFR signalling. The FGF/R axis is critical during normal tissue development, homeostasis and repair. Therefore, it is not surprising that substantial evidence also pinpoints the involvement of aberrant FGFR signalling in disease, including tumourigenesis. FGFR aberrations in cancer include mutations, gene fusions, and amplifications as well as corrupted autocrine/paracrine loops. Indeed, many clinical trials on cancer are focusing on targeting the FGF/FGFR axis, using selective FGFR inhibitors, nonselective FGFR tyrosine kinase inhibitors, ligand traps, and monoclonal antibodies and some have already been approved for the treatment of cancer patients. The heterogeneous tumour microenvironment and complexity of FGFR signalling may be some of the factors responsible for the resistance or poor response to therapy with FGFR axis-directed therapeutic agents. In the present review we will focus on the structure and function of FGF(R)s, their common irregularities in cancer and the therapeutic value of targeting their function in cancer.
Collapse
Affiliation(s)
- Athina-Myrto Chioni
- School of Life Sciences Pharmacy and Chemistry, Kingston University, Penrhyn Road, Kingston upon Thames KT1 2EE, UK
| | - Richard P. Grose
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK;
| |
Collapse
|
7
|
Yamada D, Nakamura M, Takao T, Takihira S, Yoshida A, Kawai S, Miura A, Ming L, Yoshitomi H, Gozu M, Okamoto K, Hojo H, Kusaka N, Iwai R, Nakata E, Ozaki T, Toguchida J, Takarada T. Induction and expansion of human PRRX1 + limb-bud-like mesenchymal cells from pluripotent stem cells. Nat Biomed Eng 2021; 5:926-940. [PMID: 34373601 DOI: 10.1038/s41551-021-00778-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 07/01/2021] [Indexed: 02/05/2023]
Abstract
Current protocols for the differentiation of human pluripotent stem cells (hPSCs) into chondrocytes do not allow for the expansion of intermediate progenitors so as to prospectively assess their chondrogenic potential. Here we report a protocol that leverages PRRX1-tdTomato reporter hPSCs for the selective induction of expandable and ontogenetically defined PRRX1+ limb-bud-like mesenchymal cells under defined xeno-free conditions, and the prospective assessment of the cells' chondrogenic potential via the cell-surface markers CD90, CD140B and CD82. The cells, which proliferated stably and exhibited the potential to undergo chondrogenic differentiation, formed hyaline cartilaginous-like tissue commensurate to their PRRX1-expression levels. Moreover, we show that limb-bud-like mesenchymal cells derived from patient-derived induced hPSCs can be used to identify therapeutic candidates for type II collagenopathy and we developed a method to generate uniformly sized hyaline cartilaginous-like particles by plating the cells on culture dishes coated with spots of a zwitterionic polymer. PRRX1+ limb-bud-like mesenchymal cells could facilitate the mass production of chondrocytes and cartilaginous tissues for applications in drug screening and tissue engineering.
Collapse
Affiliation(s)
- Daisuke Yamada
- Department of Regenerative Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Masahiro Nakamura
- Precision Health, Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Tomoka Takao
- Department of Regenerative Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Shota Takihira
- Department of Regenerative Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan.,Department Orthopedic Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Aki Yoshida
- Department Orthopedic Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Shunsuke Kawai
- Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Akihiro Miura
- Department of Regenerative Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Lu Ming
- Department of Regenerative Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Hiroyuki Yoshitomi
- Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan.,Department of Regeneration Science and Engineering, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Mai Gozu
- Department of Regeneration Science and Engineering, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Kumi Okamoto
- Department of Regeneration Science and Engineering, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Hironori Hojo
- Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Naoyuki Kusaka
- Institute of Frontier Science and Technology, Okayama University of Science, Okayama, Japan
| | - Ryosuke Iwai
- Institute of Frontier Science and Technology, Okayama University of Science, Okayama, Japan
| | - Eiji Nakata
- Department Orthopedic Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Toshifumi Ozaki
- Department Orthopedic Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Junya Toguchida
- Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan.,Department of Regeneration Science and Engineering, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Takeshi Takarada
- Department of Regenerative Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan.
| |
Collapse
|
8
|
Czarkwiani A, Dylus DV, Carballo L, Oliveri P. FGF signalling plays similar roles in development and regeneration of the skeleton in the brittle star Amphiura filiformis. Development 2021; 148:dev180760. [PMID: 34042967 PMCID: PMC8180256 DOI: 10.1242/dev.180760] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 04/13/2021] [Indexed: 12/16/2022]
Abstract
Regeneration as an adult developmental process is in many aspects similar to embryonic development. Although many studies point out similarities and differences, no large-scale, direct and functional comparative analyses between development and regeneration of a specific cell type or structure in one animal exist. Here, we use the brittle star Amphiura filiformis to characterise the role of the FGF signalling pathway during skeletal development in embryos and arm regeneration. In both processes, we find ligands expressed in ectodermal cells that flank underlying skeletal mesenchymal cells, which express the receptors. Perturbation of FGF signalling showed inhibited skeleton formation in both embryogenesis and regeneration, without affecting other key developmental processes. Differential transcriptome analysis finds mostly differentiation genes rather than transcription factors to be downregulated in both contexts. Moreover, comparative gene analysis allowed us to discover brittle star-specific differentiation genes. In conclusion, our results show that the FGF pathway is crucial for skeletogenesis in the brittle star, as in other deuterostomes, and provide evidence for the re-deployment of a developmental gene regulatory module during regeneration.
Collapse
Affiliation(s)
- Anna Czarkwiani
- Department of Genetics, Evolution and Environment, University College London, London WC1E 6BT, UK
| | - David V. Dylus
- Department of Genetics, Evolution and Environment, University College London, London WC1E 6BT, UK
- Centre for Mathematics, Physics and Engineering in the Life Sciences and Experimental Biology, University College London, London WC1E 6BT, UK
| | - Luisana Carballo
- Department of Genetics, Evolution and Environment, University College London, London WC1E 6BT, UK
| | - Paola Oliveri
- Department of Genetics, Evolution and Environment, University College London, London WC1E 6BT, UK
- Centre for Life's Origin and Evolution (CLOE), University College London, London WC1E 6BT, UK
| |
Collapse
|
9
|
Otsuka T, Mengsteab PY, Laurencin CT. Control of mesenchymal cell fate via application of FGF-8b in vitro. Stem Cell Res 2021; 51:102155. [PMID: 33445073 PMCID: PMC8027992 DOI: 10.1016/j.scr.2021.102155] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 11/30/2020] [Accepted: 01/01/2021] [Indexed: 12/29/2022] Open
Abstract
In order to develop strategies to regenerate complex tissues in mammals, understanding the role of signaling in regeneration competent species and mammalian development is of critical importance. Fibroblast growth factor 8 (FGF-8) signaling has an essential role in limb morphogenesis and blastema outgrowth. Therefore, we aimed to study the effect of FGF-8b on the proliferation and differentiation of mesenchymal stem cells (MSCs), which have tremendous potential for therapeutic use of cell-based therapy. Rat adipose derived stem cells (ADSCs) and muscle progenitor cells (MPCs) were isolated and cultured in growth medium and various types of differentiation medium (osteogenic, chondrogenic, adipogenic, tenogenic, and myogenic medium) with or without FGF-8b supplementation. We found that FGF-8b induced robust proliferation regardless of culture medium. Genes related to limb development were upregulated in ADSCs by FGF-8b supplementation. Moreover, FGF-8b enhanced chondrogenic differentiation and suppressed adipogenic and tenogenic differentiation in ADSCs. Osteogenic differentiation was not affected by FGF-8b supplementation. FGF-8b was found to enhance myofiber formation in rat MPCs. Overall, this study provides foundational knowledge on the effect of FGF-8b in the proliferation and fate determination of MSCs and provides insight in its potential efficacy for musculoskeletal therapies.
Collapse
Affiliation(s)
- Takayoshi Otsuka
- Connecticut Convergence Institute for Translation in Regenerative Engineering, University of Connecticut Health, Farmington, CT 06030, USA; Raymond and Beverly Sackler Center for Biological, Physical and Engineering Sciences, University of Connecticut Health, CT 06030, USA; Department of Orthopedic Surgery, University of Connecticut Health, Farmington, CT 06030, USA
| | - Paulos Y Mengsteab
- Connecticut Convergence Institute for Translation in Regenerative Engineering, University of Connecticut Health, Farmington, CT 06030, USA; Raymond and Beverly Sackler Center for Biological, Physical and Engineering Sciences, University of Connecticut Health, CT 06030, USA; Department of Orthopedic Surgery, University of Connecticut Health, Farmington, CT 06030, USA; Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA
| | - Cato T Laurencin
- Connecticut Convergence Institute for Translation in Regenerative Engineering, University of Connecticut Health, Farmington, CT 06030, USA; Raymond and Beverly Sackler Center for Biological, Physical and Engineering Sciences, University of Connecticut Health, CT 06030, USA; Department of Orthopedic Surgery, University of Connecticut Health, Farmington, CT 06030, USA; Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA; Department of Materials Science and Engineering, University of Connecticut, Storrs, CT 06269, USA; Department of Chemical and Biomolecular Engineering, University of Connecticut, Storrs, CT 06269, USA.
| |
Collapse
|
10
|
p63 is a cereblon substrate involved in thalidomide teratogenicity. Nat Chem Biol 2019; 15:1077-1084. [DOI: 10.1038/s41589-019-0366-7] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 08/19/2019] [Indexed: 12/16/2022]
|
11
|
McKenzie J, Smith C, Karuppaiah K, Langberg J, Silva MJ, Ornitz DM. Osteocyte Death and Bone Overgrowth in Mice Lacking Fibroblast Growth Factor Receptors 1 and 2 in Mature Osteoblasts and Osteocytes. J Bone Miner Res 2019; 34:1660-1675. [PMID: 31206783 PMCID: PMC6744314 DOI: 10.1002/jbmr.3742] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 03/27/2019] [Accepted: 04/05/2019] [Indexed: 01/11/2023]
Abstract
Fibroblast growth factor (FGF) signaling pathways have well-established roles in skeletal development, with essential functions in both chondrogenesis and osteogenesis. In mice, previous conditional knockout studies suggested distinct roles for FGF receptor 1 (FGFR1) signaling at different stages of osteogenesis and a role for FGFR2 in osteoblast maturation. However, the potential for redundancy among FGFRs and the mechanisms and consequences of stage-specific osteoblast lineage regulation were not addressed. Here, we conditionally inactivate Fgfr1 and Fgfr2 in mature osteoblasts with an Osteocalcin (OC)-Cre or Dentin matrix protein 1 (Dmp1)-CreER driver. We find that young mice lacking both receptors or only FGFR1 are phenotypically normal. However, between 6 and 12 weeks of age, OC-Cre Fgfr1/Fgfr2 double- and Fgfr1 single-conditional knockout mice develop a high bone mass phenotype with increased periosteal apposition, increased and disorganized endocortical bone with increased porosity, and biomechanical properties that reflect increased bone mass but impaired material properties. Histopathological and gene expression analyses show that this phenotype is preceded by a striking loss of osteocytes and accompanied by activation of the Wnt/β-catenin signaling pathway. These data identify a role for FGFR1 signaling in mature osteoblasts/osteocytes that is directly or indirectly required for osteocyte survival and regulation of bone mass during postnatal bone growth. © 2019 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Jennifer McKenzie
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, USA.,Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Craig Smith
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Kannan Karuppaiah
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Joshua Langberg
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Matthew J Silva
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, USA.,Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - David M Ornitz
- Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, MO, USA.,Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
12
|
Tuzon CT, Rigueur D, Merrill AE. Nuclear Fibroblast Growth Factor Receptor Signaling in Skeletal Development and Disease. Curr Osteoporos Rep 2019; 17:138-146. [PMID: 30982184 PMCID: PMC8221190 DOI: 10.1007/s11914-019-00512-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW Fibroblast growth factor receptor (FGFR) signaling regulates proliferation and differentiation during development and homeostasis. While membrane-bound FGFRs play a central role in these processes, the function of nuclear FGFRs is also critical. Here, we highlight mechanisms for nuclear FGFR translocation and the effects of nuclear FGFRs on skeletal development and disease. RECENT FINDINGS Full-length FGFRs, internalized by endocytosis, enter the nucleus through β-importin-dependent mechanisms that recognize the nuclear localization signal within FGFs. Alternatively, soluble FGFR intracellular fragments undergo nuclear translocation following their proteolytic release from the membrane. FGFRs enter the nucleus during the cellular transition between proliferation and differentiation. Once nuclear, FGFRs interact with chromatin remodelers to alter the epigenetic state and transcription of their target genes. Dysregulation of nuclear FGFR is linked to the etiology of congenital skeletal disorders and neoplastic transformation. Revealing the activities of nuclear FGFR will advance our understanding of 20 congenital skeletal disorders caused by FGFR mutations, as well as FGFR-related cancers.
Collapse
Affiliation(s)
- Creighton T Tuzon
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, 90033, USA
| | - Diana Rigueur
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, 90033, USA
| | - Amy E Merrill
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, 90033, USA.
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA.
| |
Collapse
|
13
|
Reinhardt R, Gullotta F, Nusspaumer G, Ünal E, Ivanek R, Zuniga A, Zeller R. Molecular signatures identify immature mesenchymal progenitors in early mouse limb buds that respond differentially to morphogen signaling. Development 2019; 146:dev.173328. [PMID: 31076486 PMCID: PMC6550019 DOI: 10.1242/dev.173328] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 05/01/2019] [Indexed: 12/31/2022]
Abstract
The key molecular interactions governing vertebrate limb bud development are a paradigm for studying the mechanisms controlling progenitor cell proliferation and specification during vertebrate organogenesis. However, little is known about the cellular heterogeneity of the mesenchymal progenitors in early limb buds that ultimately contribute to the chondrogenic condensations prefiguring the skeleton. We combined flow cytometric and transcriptome analyses to identify the molecular signatures of several distinct mesenchymal progenitor cell populations present in early mouse forelimb buds. In particular, jagged 1 (JAG1)-positive cells located in the posterior-distal mesenchyme were identified as the most immature limb bud mesenchymal progenitors (LMPs), which crucially depend on SHH and FGF signaling in culture. The analysis of gremlin 1 (Grem1)-deficient forelimb buds showed that JAG1-expressing LMPs are protected from apoptosis by GREM1-mediated BMP antagonism. At the same stage, the osteo-chondrogenic progenitors (OCPs) located in the core mesenchyme are already actively responding to BMP signaling. This analysis sheds light on the cellular heterogeneity of the early mouse limb bud mesenchyme and on the distinct response of LMPs and OCPs to morphogen signaling.
Collapse
Affiliation(s)
- Robert Reinhardt
- Developmental Genetics, Department of Biomedicine, University of Basel, 4058 Basel, Switzerland
| | - Fabiana Gullotta
- Developmental Genetics, Department of Biomedicine, University of Basel, 4058 Basel, Switzerland
| | - Gretel Nusspaumer
- Developmental Genetics, Department of Biomedicine, University of Basel, 4058 Basel, Switzerland.,Development and Evolution, Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide, 41013 Sevilla, Spain
| | - Erkan Ünal
- Developmental Genetics, Department of Biomedicine, University of Basel, 4058 Basel, Switzerland.,Swiss Institute of Bioinformatics, 4058 Basel, Switzerland.,Bioinformatics Core Facility, Department of Biomedicine, University of Basel, 4056 Basel, Switzerland
| | - Robert Ivanek
- Swiss Institute of Bioinformatics, 4058 Basel, Switzerland.,Bioinformatics Core Facility, Department of Biomedicine, University of Basel, 4056 Basel, Switzerland
| | - Aimée Zuniga
- Developmental Genetics, Department of Biomedicine, University of Basel, 4058 Basel, Switzerland
| | - Rolf Zeller
- Developmental Genetics, Department of Biomedicine, University of Basel, 4058 Basel, Switzerland
| |
Collapse
|
14
|
Giffin JL, Gaitor D, Franz-Odendaal TA. The Forgotten Skeletogenic Condensations: A Comparison of Early Skeletal Development Amongst Vertebrates. J Dev Biol 2019; 7:jdb7010004. [PMID: 30717314 PMCID: PMC6473759 DOI: 10.3390/jdb7010004] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Revised: 01/24/2019] [Accepted: 01/30/2019] [Indexed: 01/13/2023] Open
Abstract
The development of a skeletogenic condensation is perhaps the most critical yet considerably overlooked stage of skeletogenesis. Described in this comprehensive review are the mechanisms that facilitate skeletogenic condensation formation, growth, and maintenance to allow for overt differentiation into a skeletal element. This review discusses the current knowledge of gene regulation and characterization of skeletogenic condensations in the chicken, mouse, zebrafish, and other developmental models. We limited our scope to condensations that give rise to the bones and cartilages of the vertebrate skeleton, with a particular focus on craniofacial and limb bud regions. While many of the skeletogenic processes are similar among vertebrate lineages, differences are apparent in the site and timing of the initial epithelial⁻mesenchymal interactions as well as in whether the condensation has an osteogenic or chondrogenic fate, both within and among species. Further comparative studies are needed to clarify and broaden the existing knowledge of this intricate phenomenon.
Collapse
Affiliation(s)
- Jennifer L Giffin
- Department of Biology, Mount Saint Vincent University, 166 Bedford Highway, Halifax, NS B3M 2J6, Canada.
| | - Danielle Gaitor
- Department of Biology, Mount Saint Vincent University, 166 Bedford Highway, Halifax, NS B3M 2J6, Canada.
- Department of Medical Neuroscience, Dalhousie University, 5850 College Street, Halifax, NS B3H 4R2, Canada.
| | - Tamara A Franz-Odendaal
- Department of Biology, Mount Saint Vincent University, 166 Bedford Highway, Halifax, NS B3M 2J6, Canada.
- Department of Medical Neuroscience, Dalhousie University, 5850 College Street, Halifax, NS B3H 4R2, Canada.
| |
Collapse
|
15
|
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
|
16
|
Jin L, Wu J, Bellusci S, Zhang JS. Fibroblast Growth Factor 10 and Vertebrate Limb Development. Front Genet 2019; 9:705. [PMID: 30687387 PMCID: PMC6338048 DOI: 10.3389/fgene.2018.00705] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Accepted: 12/14/2018] [Indexed: 12/11/2022] Open
Abstract
Early limb development requires fibroblast growth factor (Fgf)-mediated coordination between growth and patterning to ensure the proper formation of a functional organ. The apical ectodermal ridge (AER) is a domain of thickened epithelium located at the distal edge of the limb bud that coordinates outgrowth along the proximodistal axis. Considerable amount of work has been done to elucidate the cellular and molecular mechanisms underlying induction, maintenance and regression of the AER. Fgf10, a paracrine Fgf that elicits its biological responses by activating the fibroblast growth factor receptor 2b (Fgfr2b), is crucial for governing proximal distal outgrowth as well as patterning and acts upstream of the known AER marker Fgf8. A transgenic mouse line allowing doxycycline-based inducible and ubiquitous expression of a soluble form of Fgfr2b has been extensively used to identify the role of Fgfr2b ligands at different time points during development. Overexpression of soluble Fgfr2b (sFgfr2b) post-AER induction leads to irreversible loss of cellular β-catenin organization and decreased Fgf8 expression in the AER. A similar approach has been carried out pre-AER induction. The observed limb phenotype is similar to the severe proximal truncations observed in human babies exposed to thalidomide, which has been proposed to block the Fgf10-AER-Fgf8 feedback loop. Novel insights on the role of Fgf10 signaling in limb formation pre- and post-AER induction are summarized in this review and will be integrated with possible future investigations on the role of Fgf10 throughout limb development.
Collapse
Affiliation(s)
- Libo Jin
- Institute of Life Sciences, Wenzhou University-Wenzhou Medical University Collaborative Innovation Center for Biomedicine, Wenzhou, China
| | - Jin Wu
- Institute of Life Sciences, Wenzhou University-Wenzhou Medical University Collaborative Innovation Center for Biomedicine, Wenzhou, China
| | - Saverio Bellusci
- Institute of Life Sciences, Wenzhou University-Wenzhou Medical University Collaborative Innovation Center for Biomedicine, Wenzhou, China.,Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Excellence Cluster Cardio-Pulmonary System, Universities of Giessen and Marburg Lung Center, member of the German Center for Lung Research, Justus-Liebig-University Giessen, Giessen, Germany
| | - Jin-San Zhang
- Institute of Life Sciences, Wenzhou University-Wenzhou Medical University Collaborative Innovation Center for Biomedicine, Wenzhou, China.,Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
17
|
Abstract
Fibroblast growth factors (FGFs) and their receptors (FGFRs) are expressed throughout all stages of skeletal development. In the limb bud and in cranial mesenchyme, FGF signaling is important for formation of mesenchymal condensations that give rise to bone. Once skeletal elements are initiated and patterned, FGFs regulate both endochondral and intramembranous ossification programs. In this chapter, we review functions of the FGF signaling pathway during these critical stages of skeletogenesis, and explore skeletal malformations in humans that are caused by mutations in FGF signaling molecules.
Collapse
Affiliation(s)
- David M Ornitz
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, United States.
| | - Pierre J Marie
- UMR-1132 Inserm (Institut national de la Santé et de la Recherche Médicale) and University Paris Diderot, Sorbonne Paris Cité, Hôpital Lariboisière, Paris, France
| |
Collapse
|
18
|
Pickering J, Rich CA, Stainton H, Aceituno C, Chinnaiya K, Saiz-Lopez P, Ros MA, Towers M. An intrinsic cell cycle timer terminates limb bud outgrowth. eLife 2018; 7:37429. [PMID: 30175958 PMCID: PMC6143340 DOI: 10.7554/elife.37429] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 09/02/2018] [Indexed: 12/24/2022] Open
Abstract
The longstanding view of how proliferative outgrowth terminates following the patterning phase of limb development involves the breakdown of reciprocal extrinsic signalling between the distal mesenchyme and the overlying epithelium (e-m signalling). However, by grafting distal mesenchyme cells from late stage chick wing buds to the epithelial environment of younger wing buds, we show that this mechanism is not required. RNA sequencing reveals that distal mesenchyme cells complete proliferative outgrowth by an intrinsic cell cycle timer in the presence of e-m signalling. In this process, e-m signalling is required permissively to allow the intrinsic cell cycle timer to run its course. We provide evidence that a temporal switch from BMP antagonism to BMP signalling controls the intrinsic cell cycle timer during limb outgrowth. Our findings have general implications for other patterning systems in which extrinsic signals and intrinsic timers are integrated.
Collapse
Affiliation(s)
- Joseph Pickering
- Department of Biomedical Science, The Bateson Centre, University of Sheffield, Sheffield, United Kingdom
| | - Constance A Rich
- Department of Biomedical Science, The Bateson Centre, University of Sheffield, Sheffield, United Kingdom
| | - Holly Stainton
- Department of Biomedical Science, The Bateson Centre, University of Sheffield, Sheffield, United Kingdom
| | - Cristina Aceituno
- Instituto de Biomedicina y Biotecnología de Cantabria, IBBTEC (CSIC-Universidad de Cantabria), Santander, Spain
| | - Kavitha Chinnaiya
- Department of Biomedical Science, The Bateson Centre, University of Sheffield, Sheffield, United Kingdom
| | - Patricia Saiz-Lopez
- Instituto de Biomedicina y Biotecnología de Cantabria, IBBTEC (CSIC-Universidad de Cantabria), Santander, Spain
| | - Marian A Ros
- Instituto de Biomedicina y Biotecnología de Cantabria, IBBTEC (CSIC-Universidad de Cantabria), Santander, Spain.,Departamento de Anatomía y Biología Celular, Facultad de Medicina, Universidad de Cantabria, Santander, Spain
| | - Matthew Towers
- Department of Biomedical Science, The Bateson Centre, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
19
|
Gao B, Ajima R, Yang W, Li C, Song H, Anderson MJ, Liu RR, Lewandoski MB, Yamaguchi TP, Yang Y. Coordinated directional outgrowth and pattern formation by integration of Wnt5a and Fgf signaling in planar cell polarity. Development 2018; 145:dev.163824. [PMID: 29615464 DOI: 10.1242/dev.163824] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 03/19/2018] [Indexed: 12/28/2022]
Abstract
Embryonic morphogenesis of a complex organism requires proper regulation of patterning and directional growth. Planar cell polarity (PCP) signaling is emerging as a crucial evolutionarily conserved mechanism whereby directional information is conveyed. PCP is thought to be established by global cues, and recent studies have revealed an instructive role of a Wnt signaling gradient in epithelial tissues of both invertebrates and vertebrates. However, it remains unclear whether Wnt/PCP signaling is regulated in a coordinated manner with embryonic patterning during morphogenesis. Here, in mouse developing limbs, we find that apical ectoderm ridge-derived Fgfs required for limb patterning regulate PCP along the proximal-distal axis in a Wnt5a-dependent manner. We demonstrate with genetic evidence that the Wnt5a gradient acts as a global cue that is instructive in establishing PCP in the limb mesenchyme, and that Wnt5a also plays a permissive role to allow Fgf signaling to orient PCP. Our results indicate that limb morphogenesis is regulated by coordination of directional growth and patterning through integration of Wnt5a and Fgf signaling.
Collapse
Affiliation(s)
- Bo Gao
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China .,Developmental Genetics Section, Genetic Disease Research Branch, National Human Genome Research Institute, NIH, Bethesda, MD 20892, USA
| | - Rieko Ajima
- Cancer and Developmental Biology Laboratory, Center for Cancer Research, National Cancer Institute-Frederick, NIH, Frederick, MD 21702, USA
| | - Wei Yang
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Chunyu Li
- Developmental Genetics Section, Genetic Disease Research Branch, National Human Genome Research Institute, NIH, Bethesda, MD 20892, USA.,Department of Hand Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, China
| | - Hai Song
- Developmental Genetics Section, Genetic Disease Research Branch, National Human Genome Research Institute, NIH, Bethesda, MD 20892, USA
| | - Matthew J Anderson
- Cancer and Developmental Biology Laboratory, Center for Cancer Research, National Cancer Institute-Frederick, NIH, Frederick, MD 21702, USA
| | - Robert R Liu
- Developmental Genetics Section, Genetic Disease Research Branch, National Human Genome Research Institute, NIH, Bethesda, MD 20892, USA
| | - Mark B Lewandoski
- Cancer and Developmental Biology Laboratory, Center for Cancer Research, National Cancer Institute-Frederick, NIH, Frederick, MD 21702, USA
| | - Terry P Yamaguchi
- Cancer and Developmental Biology Laboratory, Center for Cancer Research, National Cancer Institute-Frederick, NIH, Frederick, MD 21702, USA
| | - Yingzi Yang
- Developmental Genetics Section, Genetic Disease Research Branch, National Human Genome Research Institute, NIH, Bethesda, MD 20892, USA .,Department of Developmental Biology, Harvard School of Dental Medicine, 188 Longwood Avenue, Boston, MA 02115, USA
| |
Collapse
|
20
|
Moore ER, Jacobs CR. The primary cilium as a signaling nexus for growth plate function and subsequent skeletal development. J Orthop Res 2018; 36:533-545. [PMID: 28901584 PMCID: PMC5839937 DOI: 10.1002/jor.23732] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 09/07/2017] [Indexed: 02/04/2023]
Abstract
The primary cilium is a solitary, antenna-like sensory organelle with many important roles in cartilage and bone development, maintenance, and function. The primary cilium's potential role as a signaling nexus in the growth plate makes it an attractive therapeutic target for diseases and disorders associated with bone development and maintenance. Many signaling pathways that are mediated by the cilium-such as Hh, Wnt, Ihh/PTHrP, TGFβ, BMP, FGF, and Notch-are also known to influence endochondral ossification, primarily by directing growth plate formation and chondrocyte behavior. Although a few studies have demonstrated that these signaling pathways can be directly tied to the primary cilium, many pathways have yet to be evaluated in context of the cilium. This review serves to bridge this knowledge gap in the literature, as well as discuss the cilium's importance in the growth plate's ability to sense and respond to chemical and mechanical stimuli. Furthermore, we explore the importance of using the appropriate mechanism to study the cilium in vivo and suggest IFT88 deletion is the best available technique. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 36:533-545, 2018.
Collapse
Affiliation(s)
- Emily R. Moore
- Department of Biomedical Engineering; Columbia University; 351 Engineering Terrace, Mail Code 8904, 1210 Amsterdam Avenue New York 10027 New York
| | - Christopher R. Jacobs
- Department of Biomedical Engineering; Columbia University; 351 Engineering Terrace, Mail Code 8904, 1210 Amsterdam Avenue New York 10027 New York
| |
Collapse
|
21
|
Garg A, Bansal M, Gotoh N, Feng GS, Zhong J, Wang F, Kariminejad A, Brooks S, Zhang X. Alx4 relays sequential FGF signaling to induce lacrimal gland morphogenesis. PLoS Genet 2017; 13:e1007047. [PMID: 29028795 PMCID: PMC5656309 DOI: 10.1371/journal.pgen.1007047] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 10/25/2017] [Accepted: 09/28/2017] [Indexed: 11/18/2022] Open
Abstract
The sequential use of signaling pathways is essential for the guidance of pluripotent progenitors into diverse cell fates. Here, we show that Shp2 exclusively mediates FGF but not PDGF signaling in the neural crest to control lacrimal gland development. In addition to preventing p53-independent apoptosis and promoting the migration of Sox10-expressing neural crests, Shp2 is also required for expression of the homeodomain transcription factor Alx4, which directly controls Fgf10 expression in the periocular mesenchyme that is necessary for lacrimal gland induction. We show that Alx4 binds an Fgf10 intronic element conserved in terrestrial but not aquatic animals, underlying the evolutionary emergence of the lacrimal gland system in response to an airy environment. Inactivation of ALX4/Alx4 causes lacrimal gland aplasia in both human and mouse. These results reveal a key role of Alx4 in mediating FGF-Shp2-FGF signaling in the neural crest for lacrimal gland development. The dry eye disease caused by lacrimal gland dysgenesis is one of the most common ocular ailments. In this study, we show that Shp2 mediates the sequential use of FGF signaling in lacrimal gland development. Our study identifies Alx4 as a novel target of Shp2 signaling and a causal gene for lacrimal gland aplasia in humans. Given this result, there may also be a potential role for Alx4 in guiding pluripotent stem cells to produce lacrimal gland tissue. Finally, our data reveals an Alx4-Fgf10 regulatory unit broadly conserved in the diverse array of terrestrial animals from humans to reptiles, but not in aquatic animals such as amphibians and fish, which sheds light on how the lacrimal gland arose as an evolutionary innovation of terrestrial animals to adapt to their newfound exposure to an airy environment.
Collapse
Affiliation(s)
- Ankur Garg
- Departments of Ophthalmology, Pathology and Cell Biology, Columbia University, New York, NY, United States of America
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, United States of America
| | - Mukesh Bansal
- PsychoGenics Inc., Tarrytown, NY, United States of America
| | - Noriko Gotoh
- Division of Cancer Cell Biology, Cancer Research Institute, Kanazawa University Kakuma-machi, Kanazawa city, Japan
| | - Gen-Sheng Feng
- Department of Pathology, School of Medicine, and Section of Molecular Biology, Division of Biological Sciences, University of California San Diego, La Jolla, CA, United States of America
| | - Jian Zhong
- Burke Medical Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, White Plains, NY, United States of America
| | - Fen Wang
- Center for Cancer Biology and Nutrition, Institute of Biosciences and Technology, Texas A&M, Houston, TX, United States of America
| | | | - Steven Brooks
- Departments of Ophthalmology, Pathology and Cell Biology, Columbia University, New York, NY, United States of America
| | - Xin Zhang
- Departments of Ophthalmology, Pathology and Cell Biology, Columbia University, New York, NY, United States of America
- * E-mail:
| |
Collapse
|
22
|
Hopyan S. Biophysical regulation of early limb bud morphogenesis. Dev Biol 2017; 429:429-433. [DOI: 10.1016/j.ydbio.2017.06.034] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 04/30/2017] [Accepted: 06/29/2017] [Indexed: 12/11/2022]
|
23
|
Abstract
Endochondral ossification is the fundamental process of skeletal development in vertebrates. Chondrocytes undergo sequential steps of differentiation, including mesenchymal condensation, proliferation, hypertrophy, and mineralization. These steps, which are required for the morphological and functional changes in differentiating chondrocytes, are strictly regulated by a complex transcriptional network. Biochemical and mice genetic studies identified chondrogenic transcription factors critical for endochondral ossification. The transcription factor sex-determining region Y (SRY)-box 9 (Sox9) is essential for early chondrogenesis, and impaired Sox9 function causes severe chondrodysplasia in humans and mice. In addition, recent genome-wide chromatin immunoprecipitation-sequencing studies revealed the precise regulatory mechanism of Sox9 during early chondrogenesis. Runt-related transcription factor 2 promotes chondrocyte hypertrophy and terminal differentiation. Interestingly, endoplasmic reticulum (ER) stress-related transcription factors have recently emerged as novel regulators of chondrocyte differentiation. Here we review the transcriptional mechanisms that regulate endochondral ossification, with a focus on Sox9.
Collapse
Affiliation(s)
- Kenji Hata
- Department of Molecular and Cellular Biochemistry, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Yoshifumi Takahata
- Department of Molecular and Cellular Biochemistry, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Tomohiko Murakami
- Department of Molecular and Cellular Biochemistry, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Riko Nishimura
- Department of Molecular and Cellular Biochemistry, Osaka University Graduate School of Dentistry, Osaka, Japan
| |
Collapse
|
24
|
Ungewitter E, Rotgers E, Bantukul T, Kawakami Y, Kissling GE, Yao HHC. From the Cover: Teratogenic Effects of in Utero Exposure to Di-(2-Ethylhexyl)-Phthalate (DEHP) in B6:129S4 Mice. Toxicol Sci 2017; 157:8-19. [PMID: 28123099 PMCID: PMC6074946 DOI: 10.1093/toxsci/kfx019] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Intrauterine exposure to phthalates is known to cause disorders of male reproductive function including androgen insufficiency, decreased fertility, and germ cell defects in rodents. In this study, we set out to investigate the effects of intrauterine exposure to di-(2-ethylhexyl)-phthalate (DEHP) on fetal development of the B6:129S4 mouse strain. Time-mated pregnant C57BL/6 dams were exposed to 0, 5, 250, or 500 mg/kg DEHP with corn oil as the vehicle via oral gavage from embryonic days (E)7 to 16. Survival and gross morphology of the pups were analyzed one day after the last treatment. Anogenital distance (AGD) and testicular cell functions were examined in male embryos to confirm the known effects of phthalate exposure. DEHP exposure significantly reduced the survival rate of fetuses in the 250 and 500 mg/kg dosage groups compared with the control and 5 mg/kg groups. Exposure to 250 and 500 mg/kg DEHP was teratogenic and induced exencephaly and limb malformations such as polydactyly in the B6:126S4 embryos. No gross malformations were observed in control or 5 mg/kg DEHP groups. In male embryos, exposure to both 5 and 250 mg/kg DEHP in utero was sufficient to induce the formation of multinucleated germ cells in the testes and widespread changes in mRNA expression of germ cell, interstitium and Sertoli cell-associated genes. These findings reveal that intrauterine DEHP exposure has a strong teratogenic, and lethal impact on the fetuses of B6:129S4 mouse strain.
Collapse
Affiliation(s)
| | | | | | - Yasuhiko Kawakami
- Gennetics, Cell Biology, and Development, University of Minnesota, Minneapolis, Minnesota 55455
| | - Grace E. Kissling
- Biostatistics & Computational Biology Branch, National Institute of Environmental Health Sciences Research Triangle Park, North Carolina 27709
| | | |
Collapse
|
25
|
Bryant DM, Sousounis K, Farkas JE, Bryant S, Thao N, Guzikowski AR, Monaghan JR, Levin M, Whited JL. Repeated removal of developing limb buds permanently reduces appendage size in the highly-regenerative axolotl. Dev Biol 2017; 424:1-9. [PMID: 28235582 PMCID: PMC5707178 DOI: 10.1016/j.ydbio.2017.02.013] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Revised: 02/01/2017] [Accepted: 02/20/2017] [Indexed: 12/30/2022]
Abstract
Matching appendage size to body size is fundamental to animal function. Generating an appropriately-sized appendage is a robust process executed during development which is also critical for regeneration. When challenged, larger animals are programmed to regenerate larger limbs than smaller animals within a single species. Understanding this process has important implications for regenerative medicine. To approach this complex question, models with altered appendage size:body size ratios are required. We hypothesized that repeatedly challenging axolotls to regrow limb buds would affect their developmental program resulting in altered target morphology. We discovered that after 10 months following this experimental procedure, limbs that developed were permanently miniaturized. This altered target morphology was preserved upon amputation and regeneration. Future experiments using this platform should provide critical information about how target limb size is encoded within limb progenitors.
Collapse
Affiliation(s)
- Donald M Bryant
- Harvard Medical School, the Harvard stem Cell Institute, and the Department of Orthopedic Surgery, Brigham & Women's Hospital, Cambridge, MA 02139, USA
| | - Konstantinos Sousounis
- Harvard Medical School, the Harvard stem Cell Institute, and the Department of Orthopedic Surgery, Brigham & Women's Hospital, Cambridge, MA 02139, USA; Allen Discovery Center at Tufts, Medford, MA 02155, USA
| | - Johanna E Farkas
- Department of Biology, Northeastern University, Boston, MA 02115, USA
| | - Sevara Bryant
- Harvard Medical School, the Harvard stem Cell Institute, and the Department of Orthopedic Surgery, Brigham & Women's Hospital, Cambridge, MA 02139, USA
| | - Neng Thao
- Harvard Medical School, the Harvard stem Cell Institute, and the Department of Orthopedic Surgery, Brigham & Women's Hospital, Cambridge, MA 02139, USA
| | - Anna R Guzikowski
- Harvard Medical School, the Harvard stem Cell Institute, and the Department of Orthopedic Surgery, Brigham & Women's Hospital, Cambridge, MA 02139, USA
| | - James R Monaghan
- Department of Biology, Northeastern University, Boston, MA 02115, USA
| | - Michael Levin
- Allen Discovery Center at Tufts, Medford, MA 02155, USA; Tufts Center for Regenerative and Developmental Biology, Medford, MA 02155, USA
| | - Jessica L Whited
- Harvard Medical School, the Harvard stem Cell Institute, and the Department of Orthopedic Surgery, Brigham & Women's Hospital, Cambridge, MA 02139, USA; Allen Discovery Center at Tufts, Medford, MA 02155, USA.
| |
Collapse
|
26
|
Mariani FV, Fernandez-Teran M, Ros MA. Ectoderm-mesoderm crosstalk in the embryonic limb: The role of fibroblast growth factor signaling. Dev Dyn 2017; 246:208-216. [PMID: 28002626 PMCID: PMC8262604 DOI: 10.1002/dvdy.24480] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 12/07/2016] [Accepted: 12/09/2016] [Indexed: 01/27/2023] Open
Abstract
In this commentary we focus on the function of FGFs during limb development and morphogenesis. Our goal is to understand, interpret and, when possible, reconcile the interesting findings and conflicting results that remain unexplained. For example, the cell death pattern observed after surgical removal of the AER versus genetic removal of the AER-Fgfs is strikingly different and the field is at an impasse with regard to an explanation. We also discuss the idea that AER function may involve signaling components in addition to the AER-FGFs and that signaling from the non-AER ectoderm may also have a significant contribution. We hope that a re-evaluation of current studies and a discussion of outstanding questions will motivate new experiments, especially considering the availability of new technologies, that will fuel further progress toward understanding the intricate ectoderm-to-mesoderm crosstalk during limb development. Developmental Dynamics 246:208-216, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Francesca V Mariani
- Department of Cell and Neurobiology, Broad CIRM Center for Regenerative Medicine & Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Marian Fernandez-Teran
- Departamento de Anatomía y Biología Celular, Facultad de Medicina, Universidad de Cantabria, 39011, Santander, Spain
| | - Maria A Ros
- Departamento de Anatomía y Biología Celular, Facultad de Medicina, Universidad de Cantabria, 39011, Santander, Spain
- Instituto de Biomedicina y Biotecnología de Cantabria, CSIC-SODERCAN-Universidad de Cantabria, 39011, Santander, Spain
| |
Collapse
|
27
|
Wang B, Wang W, Ni F. Classification of Congenital Deformities of Hands and Upper Limbs and Selection of Surgery Timing. Plast Reconstr Surg 2017. [DOI: 10.1007/978-981-10-5101-2_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
28
|
Takagi T, Seki A, Mochida J, Takayama S. Congenital Anomalies of the Extremities Occurring in Siblings. J Hand Surg Asian Pac Vol 2016; 21:49-53. [PMID: 27454502 DOI: 10.1142/s2424835516500065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
BACKGROUND When examining children with congenital anomalies of the extremities, it is not uncommon for parents to ask about the possibility of similar anomalies occurring in their next child. However, the inheritance of the disease in many congenital anomalies of the extremities has never been elucidated. METHODS In the present study we reviewed cases of their occurrence in siblings that we encountered in our department, and we investigated their characteristics. RESULTS The results did not reveal any disease specificity, but a tendency for bilateral cases and male cases (cases in brothers) to be more common was observed. CONCLUSIONS In recent years there have been reports of the discovery of causative genes in some congenital anomalies, but because cases of occurrence in siblings with no familial occurrence in the past are seen, there may be a variety of causative genes in many congenital anomalies. In the present study there were many male cases (cases in brothers) and many bilateral cases, and there appears to have been a strong possibility of familial occurrence in such cases, but there were also quite a few exceptions. It is necessary to bear the possibility of heredity in mind in every case and provide the parents with an explanation.
Collapse
Affiliation(s)
- Takehiko Takagi
- * Department of Orthopaedic Surgery, Surgical Science, Tokai University School of Medicine, Kanagawa, Japan.,† Department of Orthopaedic Surgery, National Center for Child Health and Development, Tokyo, Japan
| | - Atsuhito Seki
- † Department of Orthopaedic Surgery, National Center for Child Health and Development, Tokyo, Japan
| | - Joji Mochida
- * Department of Orthopaedic Surgery, Surgical Science, Tokai University School of Medicine, Kanagawa, Japan
| | - Shinichiro Takayama
- † Department of Orthopaedic Surgery, National Center for Child Health and Development, Tokyo, Japan
| |
Collapse
|
29
|
Dowling A, Doroba C, Maier JA, Cohen L, VandeBerg J, Sears KE. Cellular and molecular drivers of differential organ growth: insights from the limbs of Monodelphis domestica. Dev Genes Evol 2016; 226:235-43. [PMID: 27194412 DOI: 10.1007/s00427-016-0549-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 05/03/2016] [Indexed: 10/21/2022]
Abstract
A fundamental question in biology is "how is growth differentially regulated during development to produce organs of particular sizes?" We used a new model system for the study of differential organ growth, the limbs of the opossum (Monodelphis domestica), to investigate the cellular and molecular basis of differential organ growth in mammals. Opossum forelimbs grow much faster than hindlimbs, making opossum limbs an exceptional system with which to study differential growth. We first used the great differences in opossum forelimb and hindlimb growth to identify cellular processes and molecular signals that underlie differential limb growth. We then used organ culture and pharmacological addition of FGF ligands and inhibitors to test the role of the Fgf/Mitogen-activated protein kinases (MAPK) signaling pathway in driving these cellular processes. We found that molecular signals from within the limb drive differences in cell proliferation that contribute to the differential growth of the forelimb and hindlimbs of opossums. We also found that alterations in the Fgf/MAPK pathway can generate differences in cell proliferation that mirror those observed between wild-type forelimb and hindlimbs of opossums and that manipulation of Fgf/MAPK signaling affects downstream focal adhesion-extracellular matrix (FA-ECM) and Wnt signaling in opossum limbs. Taken together, these findings suggest that evolutionary changes in the Fgf/MAPK pathway could help drive the observed differences in cell behaviors and growth in opossum forelimb and hindlimbs.
Collapse
Affiliation(s)
- Anna Dowling
- School of Integrative Biology, University of Illinois, 505 S Goodwin Avenue, Urbana, IL, 61801, USA
| | - Carolyn Doroba
- School of Integrative Biology, University of Illinois, 505 S Goodwin Avenue, Urbana, IL, 61801, USA
| | - Jennifer A Maier
- School of Integrative Biology, University of Illinois, 505 S Goodwin Avenue, Urbana, IL, 61801, USA
| | - Lorna Cohen
- School of Integrative Biology, University of Illinois, 505 S Goodwin Avenue, Urbana, IL, 61801, USA
| | - John VandeBerg
- Department of Genetics, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Karen E Sears
- School of Integrative Biology, University of Illinois, 505 S Goodwin Avenue, Urbana, IL, 61801, USA.
- Institute for Genomic Biology, University of Illinois, 1206 W Gregory Drive, Urbana, IL, 61801, USA.
| |
Collapse
|
30
|
Czarkwiani A, Ferrario C, Dylus DV, Sugni M, Oliveri P. Skeletal regeneration in the brittle star Amphiura filiformis. Front Zool 2016; 13:18. [PMID: 27110269 PMCID: PMC4841056 DOI: 10.1186/s12983-016-0149-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 04/12/2016] [Indexed: 12/17/2022] Open
Abstract
Background Brittle stars regenerate their whole arms post-amputation. Amphiura filiformis can now be used for molecular characterization of arm regeneration due to the availability of transcriptomic data. Previous work showed that specific developmental transcription factors known to take part in echinoderm skeletogenesis are expressed during adult arm regeneration in A. filiformis; however, the process of skeleton formation remained poorly understood. Here, we present the results of an in-depth microscopic analysis of skeletal morphogenesis during regeneration, using calcein staining, EdU labeling and in situ hybridization. Results To better compare different samples, we propose a staging system for the early A. filiformis arm regeneration stages based on morphological landmarks identifiable in living animals and supported by histological analysis. We show that the calcified spicules forming the endoskeleton first appear very early during regeneration in the dermal layer of regenerates. These spicules then mature into complex skeletal elements of the differentiated arm during late regeneration. The mesenchymal cells in the dermal area express the skeletal marker genes Afi-c-lectin, Afi-p58b and Afi-p19; however, EdU labeling shows that these dermal cells do not proliferate. Conclusions A. filiformis arms regenerate through a consistent set of developmental stages using a distalization-intercalation mode, despite variability in regeneration rate. Skeletal elements form in a mesenchymal cell layer that does not proliferate and thus must be supplied from a different source. Our work provides the basis for future cellular and molecular studies of skeleton regeneration in brittle stars. Electronic supplementary material The online version of this article (doi:10.1186/s12983-016-0149-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Anna Czarkwiani
- Department of Genetics, Evolution and Environment, University College London, London, UK
| | - Cinzia Ferrario
- Department of Biosciences, University of Milan, Milan, Italy
| | - David Viktor Dylus
- Department of Genetics, Evolution and Environment, University College London, London, UK ; Centre for Mathematics, Physics and Engineering in the Life Sciences and Experimental Biology, University College London, London, UK ; Present address: Department of Ecology and Evolution & Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Michela Sugni
- Department of Biosciences, University of Milan, Milan, Italy
| | - Paola Oliveri
- Department of Genetics, Evolution and Environment, University College London, London, UK ; Research Department of Genetics, Evolution and Environment, University College London, Room 426, Darwin Building, Gower Street, London, WC1E 6BT UK
| |
Collapse
|
31
|
Karuppaiah K, Yu K, Lim J, Chen J, Smith C, Long F, Ornitz DM. FGF signaling in the osteoprogenitor lineage non-autonomously regulates postnatal chondrocyte proliferation and skeletal growth. Development 2016; 143:1811-22. [PMID: 27052727 DOI: 10.1242/dev.131722] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 03/18/2016] [Indexed: 12/22/2022]
Abstract
Fibroblast growth factor (FGF) signaling is important for skeletal development; however, cell-specific functions, redundancy and feedback mechanisms regulating bone growth are poorly understood. FGF receptors 1 and 2 (Fgfr1 and Fgfr2) are both expressed in the osteoprogenitor lineage. Double conditional knockout mice, in which both receptors were inactivated using an osteoprogenitor-specific Cre driver, appeared normal at birth; however, these mice showed severe postnatal growth defects that include an ∼50% reduction in body weight and bone mass, and impaired longitudinal bone growth. Histological analysis showed reduced cortical and trabecular bone, suggesting cell-autonomous functions of FGF signaling during postnatal bone formation. Surprisingly, the double conditional knockout mice also showed growth plate defects and an arrest in chondrocyte proliferation. We provide genetic evidence of a non-cell-autonomous feedback pathway regulating Fgf9, Fgf18 and Pthlh expression, which led to increased expression and signaling of Fgfr3 in growth plate chondrocytes and suppression of chondrocyte proliferation. These observations show that FGF signaling in the osteoprogenitor lineage is obligately coupled to chondrocyte proliferation and the regulation of longitudinal bone growth.
Collapse
Affiliation(s)
- Kannan Karuppaiah
- Department of Developmental Biology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Kai Yu
- Department of Developmental Biology, Washington University School of Medicine, St Louis, MO 63110, USA Division of Craniofacial Medicine, Department of Pediatrics, University of Washington and Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Joohyun Lim
- Departments of Orthopaedic Surgery and Medicine, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Jianquan Chen
- Departments of Orthopaedic Surgery and Medicine, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Craig Smith
- Department of Developmental Biology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Fanxin Long
- Departments of Orthopaedic Surgery and Medicine, Washington University School of Medicine, St Louis, MO 63110, USA
| | - David M Ornitz
- Department of Developmental Biology, Washington University School of Medicine, St Louis, MO 63110, USA
| |
Collapse
|
32
|
Abstract
The fibroblast growth factor (Fgf) family of ligands and receptor tyrosine kinases is required throughout embryonic and postnatal development and also regulates multiple homeostatic functions in the adult. Aberrant Fgf signaling causes many congenital disorders and underlies multiple forms of cancer. Understanding the mechanisms that govern Fgf signaling is therefore important to appreciate many aspects of Fgf biology and disease. Here we review the mechanisms of Fgf signaling by focusing on genetic strategies that enable in vivo analysis. These studies support an important role for Erk1/2 as a mediator of Fgf signaling in many biological processes but have also provided strong evidence for additional signaling pathways in transmitting Fgf signaling in vivo.
Collapse
Affiliation(s)
- J Richard Brewer
- Department of Developmental and Regenerative Biology, Tisch Cancer Institute, Icahn School of Medicine at Mt. Sinai, New York, New York 10029, USA
| | - Pierre Mazot
- Department of Developmental and Regenerative Biology, Tisch Cancer Institute, Icahn School of Medicine at Mt. Sinai, New York, New York 10029, USA
| | - Philippe Soriano
- Department of Developmental and Regenerative Biology, Tisch Cancer Institute, Icahn School of Medicine at Mt. Sinai, New York, New York 10029, USA
| |
Collapse
|
33
|
Hung IH, Schoenwolf GC, Lewandoski M, Ornitz DM. A combined series of Fgf9 and Fgf18 mutant alleles identifies unique and redundant roles in skeletal development. Dev Biol 2016; 411:72-84. [PMID: 26794256 PMCID: PMC4801039 DOI: 10.1016/j.ydbio.2016.01.008] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 01/14/2016] [Accepted: 01/15/2016] [Indexed: 01/14/2023]
Abstract
Fibroblast growth factor (FGF) signaling is a critical regulator of skeletal development. Fgf9 and Fgf18 are the only FGF ligands with identified functions in embryonic bone growth. Mice lacking Fgf9 or Fgf18 have distinct skeletal phenotypes; however, the extent of overlapping or redundant functions for these ligands and the stage-specific contributions of FGF signaling to chondrogenesis and osteogenesis are not known. To identify separate versus shared roles for FGF9 and FGF18, we generated a combined series of Fgf9 and Fgf18 null alleles. Analysis of embryos lacking alleles of Fgf9 and Fgf18 shows that both encoded ligands function redundantly to control all stages of skeletogenesis; however, they have variable potencies along the proximodistal limb axis, suggesting gradients of activity during formation of the appendicular skeleton. Congenital absence of both Fgf9 and Fgf18 results in a striking osteochondrodysplasia and revealed functions for FGF signaling in early proximal limb chondrogenesis. Additional defects were also noted in craniofacial bones, vertebrae, and ribs. Loss of alleles of Fgf9 and Fgf18 also affect the expression of genes encoding other key intrinsic skeletal regulators, including IHH, PTHLH (PTHrP), and RUNX2, revealing potential direct, indirect, and compensatory mechanisms to coordinate chondrogenesis and osteogenesis.
Collapse
Affiliation(s)
- Irene H Hung
- Department of Neurobiology and Anatomy, University of Utah School of Medicine, Salt Lake City, UT 84132, United States; Cancer and Developmental Biology Lab, National Cancer Institute, Frederick, MD 21701, United States; Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, United States.
| | - Gary C Schoenwolf
- Department of Neurobiology and Anatomy, University of Utah School of Medicine, Salt Lake City, UT 84132, United States
| | - Mark Lewandoski
- Cancer and Developmental Biology Lab, National Cancer Institute, Frederick, MD 21701, United States
| | - David M Ornitz
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, United States.
| |
Collapse
|
34
|
Getting a handle on embryo limb development: Molecular interactions driving limb outgrowth and patterning. Semin Cell Dev Biol 2016; 49:92-101. [DOI: 10.1016/j.semcdb.2015.01.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Revised: 01/13/2015] [Accepted: 01/14/2015] [Indexed: 11/21/2022]
|
35
|
FGF10: A multifunctional mesenchymal-epithelial signaling growth factor in development, health, and disease. Cytokine Growth Factor Rev 2015; 28:63-9. [PMID: 26559461 DOI: 10.1016/j.cytogfr.2015.10.001] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2015] [Accepted: 10/19/2015] [Indexed: 12/15/2022]
Abstract
The FGF family comprises 22 members with diverse functions in development and health. FGF10 specifically activates FGFR2b in a paracrine manner with heparan sulfate as a co-factor. FGF10and FGFR2b are preferentially expressed in the mesenchyme and epithelium, respectively. FGF10 is a mesenchymal signaling molecule in the epithelium. FGF10 knockout mice die shortly after birth due to the complete absence of lungs as well as fore- and hindlimbs. FGF10 is also essential for the development of multiple organs. The phenotypes of Fgf10 knockout mice are very similar to those of FGFR2b knockout mice, indicating that FGF10 acts as a ligand that is specific to FGFR2b in mouse multi-organ development. FGF10 also plays roles in epithelial-mesenchymal transition, the repair of tissue injury, and embryonic stem cell differentiation. In humans, FGF10 loss-of-function mutations result in inherited diseases including aplasia of lacrimal and salivary gland, lacrimo-auriculo-dento-digital syndrome, and chronic obstructive pulmonary disease. FGF10 is also involved in the oncogenicity of pancreatic and breast cancers. Single nucleotide polymorphisms in FGF10 are also potential risk factors for limb deficiencies, cleft lip and palate, and extreme myopia. These findings indicate that FGF10 is a crucial paracrine signal from the mesenchyme to epithelium for development, health, and disease.
Collapse
|
36
|
Goto M, Hojo M, Ando M, Kita A, Kitagawa M, Ohtsuka T, Kageyama R, Miyamoto S. Hes1 and Hes5 are required for differentiation of pituicytes and formation of the neurohypophysis in pituitary development. Brain Res 2015; 1625:206-17. [PMID: 26348989 DOI: 10.1016/j.brainres.2015.08.045] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Revised: 08/28/2015] [Accepted: 08/31/2015] [Indexed: 12/11/2022]
Abstract
The pituitary gland is a critical endocrine organ regulating diverse physiological functions, including homeostasis, metabolism, reproduction, and growth. It is composed of two distinct entities: the adenohypophysis, including the anterior and intermediate lobes, and the neurohypophysis known as the posterior lobe. The neurohypophysis is composed of pituicytes (glial cells) and axons projected from hypothalamic neurons. The adenohypophysis derives from Rathke's pouch, whereas the neurohypophysis derives from the infundibulum, an evagination of the ventral diencephalon. Molecular mechanisms of adenohypophysis development are much better understood, but little is known about mechanisms that regulate neurohypophysis development. Hes genes, known as Notch effectors, play a crucial role in specifying cellular fates during the development of various tissues and organs. Here, we report that the ventral diencephalon fails to evaginate resulting in complete loss of the posterior pituitary lobe in Hes1(-/-); Hes5(+/-) mutant embryos. In these mutant mice, progenitor cells are differentiated into neurons at the expense of pituicytes in the ventral diencephalon. In the developing neurohypophysis, the proliferative zone is located at the base of the infundibulum. Thus, Hes1 and Hes5 modulate not only maintenance of progenitor cells but also pituicyte versus neuron fate specification during neurohypophysis development.
Collapse
Affiliation(s)
- Masanori Goto
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan; Institute for Virus Research, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Masato Hojo
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan; Department of Neurosurgery, Shiga Medical Center for Adults, 5-4-30 Moriyama, Moriyama, Shiga 524-8524, Japan.
| | - Mitsushige Ando
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan; Institute for Virus Research, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Aya Kita
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan; Institute for Virus Research, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Masashi Kitagawa
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan; Institute for Virus Research, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Toshiyuki Ohtsuka
- Institute for Virus Research, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Ryoichiro Kageyama
- Institute for Virus Research, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Susumu Miyamoto
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| |
Collapse
|
37
|
Yu K, Karuppaiah K, Ornitz DM. Mesenchymal fibroblast growth factor receptor signaling regulates palatal shelf elevation during secondary palate formation. Dev Dyn 2015; 244:1427-38. [PMID: 26250517 DOI: 10.1002/dvdy.24319] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Revised: 06/17/2015] [Accepted: 07/26/2015] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Palatal shelf elevation is an essential morphogenetic process during secondary palate closure and failure or delay of palatal shelf elevation is a common cause of cleft palate, one of the most common birth defects in humans. Here, we studied the role of mesenchymal fibroblast growth factor receptor (FGFR) signaling during palate development by conditional inactivation of Fgfrs using a mesenchyme-specific Dermo1-Cre driver. RESULTS We showed that Fgfr1 is expressed throughout the palatal mesenchyme and Fgfr2 is expressed in the medial aspect of the posterior palatal mesenchyme overlapping with Fgfr1. Mesenchyme-specific disruption of Fgfr1 and Fgfr2 affected palatal shelf elevation and resulted in cleft palate. We further showed that both Fgfr1 and Fgfr2 are expressed in mesenchymal tissues of the mandibular process but display distinct expression patterns. Loss of mesenchymal FGFR signaling reduced mandibular ossification and lower jaw growth resulting in abnormal tongue insertion in the oral-nasal cavity. CONCLUSIONS We propose a model to explain how redundant Fgfr1 and Fgfr2 expression in the palatal and mandibular mesenchyme regulates shelf medial wall protrusion and growth of the mandible to coordinate the craniofacial tissue movements that are required for palatal shelf elevation.
Collapse
Affiliation(s)
- Kai Yu
- Division of Craniofacial Medicine, Department of Pediatrics, University of Washington and Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, Washington
| | - Kannan Karuppaiah
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri
| | - David M Ornitz
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
38
|
Abstract
Fibroblast growth factor (FGF) signaling pathways are essential regulators of vertebrate skeletal development. FGF signaling regulates development of the limb bud and formation of the mesenchymal condensation and has key roles in regulating chondrogenesis, osteogenesis, and bone and mineral homeostasis. This review updates our review on FGFs in skeletal development published in Genes & Development in 2002, examines progress made on understanding the functions of the FGF signaling pathway during critical stages of skeletogenesis, and explores the mechanisms by which mutations in FGF signaling molecules cause skeletal malformations in humans. Links between FGF signaling pathways and other interacting pathways that are critical for skeletal development and could be exploited to treat genetic diseases and repair bone are also explored.
Collapse
Affiliation(s)
- David M Ornitz
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Pierre J Marie
- UMR-1132, Institut National de la Santé et de la Recherche Médicale, Hopital Lariboisiere, 75475 Paris Cedex 10, France; Université Paris Diderot, Sorbonne Paris Cité, 75475 Paris Cedex 10, France
| |
Collapse
|
39
|
Uzkudun M, Marcon L, Sharpe J. Data-driven modelling of a gene regulatory network for cell fate decisions in the growing limb bud. Mol Syst Biol 2015; 11:815. [PMID: 26174932 PMCID: PMC4547844 DOI: 10.15252/msb.20145882] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Parameter optimization coupled with model selection is a convenient approach to infer gene regulatory networks from experimental gene expression data, but so far it has been limited to single cells or static tissues where growth is not significant. Here, we present a computational study in which we determine an optimal gene regulatory network from the spatiotemporal dynamics of gene expression patterns in a complex 2D growing tissue (non-isotropic and heterogeneous growth rates). We use this method to predict the regulatory mechanisms that underlie proximodistal (PD) patterning of the developing limb bud. First, we map the expression patterns of the PD markers Meis1, Hoxa11 and Hoxa13 into a dynamic description of the tissue movements that drive limb morphogenesis. Secondly, we use reverse-engineering to test how different gene regulatory networks can interpret the opposing gradients of fibroblast growth factors (FGF) and retinoic acid (RA) to pattern the PD markers. Finally, we validate and extend the best model against various previously published manipulative experiments, including exogenous application of RA, surgical removal of the FGF source and genetic ectopic expression of Meis1. Our approach identifies the most parsimonious gene regulatory network that can correctly pattern the PD markers downstream of FGF and RA. This network reveals a new model of PD regulation which we call the “crossover model”, because the proximal morphogen (RA) controls the distal boundary of Hoxa11, while conversely the distal morphogens (FGFs) control the proximal boundary.
Collapse
Affiliation(s)
- Manu Uzkudun
- EMBL-CRG Systems Biology Program Centre for Genomic Regulation (CRG) Universitat Pompeu Fabra (UPF), Barcelona Spain
| | - Luciano Marcon
- EMBL-CRG Systems Biology Program Centre for Genomic Regulation (CRG) Universitat Pompeu Fabra (UPF), Barcelona Spain
| | - James Sharpe
- EMBL-CRG Systems Biology Program Centre for Genomic Regulation (CRG) Universitat Pompeu Fabra (UPF), Barcelona Spain Institucio Catalana de Recerca i Estudis Avancats (ICREA), Barcelona, Spain
| |
Collapse
|
40
|
Harada M, Omori A, Nakahara C, Nakagata N, Akita K, Yamada G. Tissue-specific roles of FGF signaling in external genitalia development. Dev Dyn 2015; 244:759-73. [DOI: 10.1002/dvdy.24277] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Revised: 03/22/2015] [Accepted: 03/22/2015] [Indexed: 11/11/2022] Open
Affiliation(s)
- Masayo Harada
- Institute of Molecular Embryology and Genetics; Kumamoto University; Kumamoto Japan
- Department of Clinical Anatomy; Graduate School of Medical and Dental Sciences; Tokyo Medical and Dental University; Tokyo Japan
| | - Akiko Omori
- Institute of Molecular Embryology and Genetics; Kumamoto University; Kumamoto Japan
- Department of Developmental Genetics; Institute of Advanced Medicine; Wakayama Medical University; Wakayama Japan
| | - Chiaki Nakahara
- Institute of Molecular Embryology and Genetics; Kumamoto University; Kumamoto Japan
| | - Naomi Nakagata
- Division of Reproductive Engineering; Center for Animal Resources and Development, Kumamoto University; Kumamoto Japan
| | - Keiichi Akita
- Department of Clinical Anatomy; Graduate School of Medical and Dental Sciences; Tokyo Medical and Dental University; Tokyo Japan
| | - Gen Yamada
- Institute of Molecular Embryology and Genetics; Kumamoto University; Kumamoto Japan
- Department of Developmental Genetics; Institute of Advanced Medicine; Wakayama Medical University; Wakayama Japan
| |
Collapse
|
41
|
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: 118] [Impact Index Per Article: 11.8] [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
|
42
|
Ornitz DM, Itoh N. The Fibroblast Growth Factor signaling pathway. WILEY INTERDISCIPLINARY REVIEWS. DEVELOPMENTAL BIOLOGY 2015; 4:215-66. [PMID: 25772309 PMCID: PMC4393358 DOI: 10.1002/wdev.176] [Citation(s) in RCA: 1383] [Impact Index Per Article: 138.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Revised: 11/23/2014] [Accepted: 01/08/2015] [Indexed: 12/13/2022]
Abstract
The signaling component of the mammalian Fibroblast Growth Factor (FGF) family is comprised of eighteen secreted proteins that interact with four signaling tyrosine kinase FGF receptors (FGFRs). Interaction of FGF ligands with their signaling receptors is regulated by protein or proteoglycan cofactors and by extracellular binding proteins. Activated FGFRs phosphorylate specific tyrosine residues that mediate interaction with cytosolic adaptor proteins and the RAS-MAPK, PI3K-AKT, PLCγ, and STAT intracellular signaling pathways. Four structurally related intracellular non-signaling FGFs interact with and regulate the family of voltage gated sodium channels. Members of the FGF family function in the earliest stages of embryonic development and during organogenesis to maintain progenitor cells and mediate their growth, differentiation, survival, and patterning. FGFs also have roles in adult tissues where they mediate metabolic functions, tissue repair, and regeneration, often by reactivating developmental signaling pathways. Consistent with the presence of FGFs in almost all tissues and organs, aberrant activity of the pathway is associated with developmental defects that disrupt organogenesis, impair the response to injury, and result in metabolic disorders, and cancer. For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- David M Ornitz
- Department of Developmental Biology, Washington University School of MedicineSt. Louis, MO, USA
- *
Correspondence to:
| | - Nobuyuki Itoh
- Graduate School of Pharmaceutical Sciences, Kyoto UniversitySakyo, Kyoto, Japan
| |
Collapse
|
43
|
Restelli M, Molinari E, Marinari B, Conte D, Gnesutta N, Costanzo A, Merlo GR, Guerrini L. FGF8, c-Abl and p300 participate in a pathway that controls stability and function of the ΔNp63α protein. Hum Mol Genet 2015; 24:4185-97. [PMID: 25911675 PMCID: PMC4492388 DOI: 10.1093/hmg/ddv151] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 04/21/2015] [Indexed: 12/22/2022] Open
Abstract
The p63 transcription factor, homolog to the p53 tumor suppressor gene, plays a crucial role in epidermal and limb development, as its mutations are associated to human congenital syndromes characterized by skin, craniofacial and limb defects. While limb and skin-specific p63 transcriptional targets are being discovered, little is known of the post-translation modifications controlling ΔNp63α functions. Here we show that the p300 acetyl-transferase physically interacts in vivo with ΔNp63α and catalyzes its acetylation on lysine 193 (K193) inducing ΔNp63α stabilization and activating specific transcriptional functions. Furthermore we show that Fibroblast Growth Factor-8 (FGF8), a morphogenetic signaling molecule essential for embryonic limb development, increases the binding of ΔNp63α to the tyrosine kinase c-Abl as well as the levels of ΔNp63α acetylation. Notably, the natural mutant ΔNp63α-K193E, associated to the Split-Hand/Foot Malformation-IV syndrome, cannot be acetylated by this pathway. This mutant ΔNp63α protein displays promoter-specific loss of DNA binding activity and consequent altered expression of development-associated ΔNp63α target genes. Our results link FGF8, c-Abl and p300 in a regulatory pathway that controls ΔNp63α protein stability and transcriptional activity. Hence, limb malformation-causing p63 mutations, such as the K193E mutation, are likely to result in aberrant limb development via the combined action of altered protein stability and altered promoter occupancy.
Collapse
Affiliation(s)
- Michela Restelli
- Department of Biosciences, Università degli Studi di Milano, 20133 Milano, Italy
| | - Elisa Molinari
- Department of Biosciences, Università degli Studi di Milano, 20133 Milano, Italy
| | - Barbara Marinari
- Dermatology Unit, NESMOS Department, Università di Roma La Sapienza, I-00189 Rome, Italy and
| | - Daniele Conte
- Department of Molecular Biotechnologies and Health Sciences, Università di Torino, I-10126 Torino, Italy
| | - Nerina Gnesutta
- Department of Biosciences, Università degli Studi di Milano, 20133 Milano, Italy
| | - Antonio Costanzo
- Dermatology Unit, NESMOS Department, Università di Roma La Sapienza, I-00189 Rome, Italy and
| | - Giorgio Roberto Merlo
- Department of Molecular Biotechnologies and Health Sciences, Università di Torino, I-10126 Torino, Italy
| | - Luisa Guerrini
- Department of Biosciences, Università degli Studi di Milano, 20133 Milano, Italy,
| |
Collapse
|
44
|
Teven CM, Farina EM, Rivas J, Reid RR. Fibroblast growth factor (FGF) signaling in development and skeletal diseases. Genes Dis 2014; 1:199-213. [PMID: 25679016 PMCID: PMC4323088 DOI: 10.1016/j.gendis.2014.09.005] [Citation(s) in RCA: 163] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Fibroblast growth factors (FGF) and their receptors serve many functions in both the developing and adult organism. Humans contain 18 FGF ligands and four FGF receptors (FGFR). FGF ligands are polypeptide growth factors that regulate several developmental processes including cellular proliferation, differentiation, and migration, morphogenesis, and patterning. FGF-FGFR signaling is also critical to the developing axial and craniofacial skeleton. In particular, the signaling cascade has been implicated in intramembranous ossification of cranial bones as well as cranial suture homeostasis. In the adult, FGFs and FGFRs are crucial for tissue repair. FGF signaling generally follows one of three transduction pathways: RAS/MAP kinase, PI3/AKT, or PLCγ. Each pathway likely regulates specific cellular behaviors. Inappropriate expression of FGF and improper activation of FGFRs are associated with various pathologic conditions, unregulated cell growth, and tumorigenesis. Additionally, aberrant signaling has been implicated in many skeletal abnormalities including achondroplasia and craniosynostosis. The biology and mechanisms of the FGF family have been the subject of significant research over the past 30 years. Recently, work has focused on the therapeutic targeting and potential of FGF ligands and their associated receptors. The majority of FGF-related therapy is aimed at age-related disorders. Increased understanding of FGF signaling and biology may reveal additional therapeutic roles, both in utero and postnatally. This review discusses the role of FGF signaling in general physiologic and pathologic embryogenesis and further explores it within the context of skeletal development.
Collapse
Affiliation(s)
- Chad M Teven
- The Laboratory of Craniofacial Biology, Section of Plastic & Reconstructive Surgery, Department of Surgery, The University of Chicago Medical Center, 5841 South Maryland Avenue, MC 6035, Chicago, IL 60637, USA
| | - Evan M Farina
- The University of Chicago Pritzker School of Medicine, Chicago, IL 60637, USA
| | - Jane Rivas
- The University of Chicago Pritzker School of Medicine, Chicago, IL 60637, USA
| | - Russell R Reid
- The Laboratory of Craniofacial Biology, Section of Plastic & Reconstructive Surgery, Department of Surgery, The University of Chicago Medical Center, 5841 South Maryland Avenue, MC 6035, Chicago, IL 60637, USA
| |
Collapse
|
45
|
Sowińska-Seidler A, Piwecka M, Olech E, Socha M, Latos-Bieleńska A, Jamsheer A. Hyperosmia, ectrodactyly, mild intellectual disability, and other defects in a male patient with an X-linked partial microduplication and overexpression of the KAL1 gene. J Appl Genet 2014; 56:177-84. [PMID: 25339597 PMCID: PMC4412513 DOI: 10.1007/s13353-014-0252-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2014] [Revised: 10/07/2014] [Accepted: 10/08/2014] [Indexed: 12/16/2022]
Abstract
Loss-of-function mutations of the KAL1 gene are a known cause of Kallmann syndrome, a disorder characterized by the coexistence of hypogonadotropic hypogonadism and anosmia/hiposmia. On the other hand, neither complete nor partial duplications of KAL1 have been reported in the literature; thus, clinical symptoms associated with such alterations remain unknown. Ectrodactyly is a clinically and genetically heterogeneous abnormality presenting with hypoplasia of the central rays of the extremity, which, in around 68 % of cases, has unknown underlying molecular defect. In this paper, we report on a sporadic male patient manifesting hyperosmia and ectrodactyly accompanied by additional symptoms involving mild intellectual disability, unilateral hearing loss, genital anomalies, stocky build, and facial dysmorphism. Using a combination of high-resolution array comparative genomic hybridization (array CGH) and breakpoint analysis, we detected a hemizygous tandem duplication of 110,967 bp on Xp22.31, encompassing the promoter region and the first two exons of KAL1. In order to confirm pathogenicity of the duplication, we tested the level of KAL1 transcript in blood lymphocytes, showing 79 times higher expression in the proband compared to controls. We, therefore, hypothesize that olfactory hypersensitivity in our proband directly results from KAL1 overproduction. Additionally, a literature review allowed us to conclude that KAL1 protein at high levels may interfere with FGFR1 signaling activity, most probably indirectly giving rise to ectrodactyly, intellectual disability, and genital anomalies. Noteworthy, those symptoms overlap with Hartsfield syndrome caused by FGFR1 loss-of-function mutations. To conclude, our paper highlights the role of KAL1 in embryogenesis and provides data on the contribution of KAL1 overexpression to human pathology.
Collapse
Affiliation(s)
- Anna Sowińska-Seidler
- Department of Medical Genetics, Poznan University of Medical Sciences, Rokietnicka 8 Street, 60-806 Poznan, Poland
| | - Monika Piwecka
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego 12/14 Street, 61-704 Poznan, Poland
| | - Ewelina Olech
- Department of Medical Genetics, Poznan University of Medical Sciences, Rokietnicka 8 Street, 60-806 Poznan, Poland
| | - Magdalena Socha
- Department of Medical Genetics, Poznan University of Medical Sciences, Rokietnicka 8 Street, 60-806 Poznan, Poland
| | - Anna Latos-Bieleńska
- Department of Medical Genetics, Poznan University of Medical Sciences, Rokietnicka 8 Street, 60-806 Poznan, Poland
- NZOZ Center for Medical Genetics GENESIS, 4 Grudzieniec Street, 60-601 Poznan, Poland
| | - Aleksander Jamsheer
- Department of Medical Genetics, Poznan University of Medical Sciences, Rokietnicka 8 Street, 60-806 Poznan, Poland
- NZOZ Center for Medical Genetics GENESIS, 4 Grudzieniec Street, 60-601 Poznan, Poland
| |
Collapse
|
46
|
Sp6 and Sp8 transcription factors control AER formation and dorsal-ventral patterning in limb development. PLoS Genet 2014; 10:e1004468. [PMID: 25166858 PMCID: PMC4148220 DOI: 10.1371/journal.pgen.1004468] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Accepted: 05/14/2014] [Indexed: 12/27/2022] Open
Abstract
The formation and maintenance of the apical ectodermal ridge (AER) is critical for the outgrowth and patterning of the vertebrate limb. The induction of the AER is a complex process that relies on integrated interactions among the Fgf, Wnt, and Bmp signaling pathways that operate within the ectoderm and between the ectoderm and the mesoderm of the early limb bud. The transcription factors Sp6 and Sp8 are expressed in the limb ectoderm and AER during limb development. Sp6 mutant mice display a mild syndactyly phenotype while Sp8 mutants exhibit severe limb truncations. Both mutants show defects in AER maturation and in dorsal-ventral patterning. To gain further insights into the role Sp6 and Sp8 play in limb development, we have produced mice lacking both Sp6 and Sp8 activity in the limb ectoderm. Remarkably, the elimination or significant reduction in Sp6;Sp8 gene dosage leads to tetra-amelia; initial budding occurs, but neither Fgf8 nor En1 are activated. Mutants bearing a single functional allele of Sp8 (Sp6−/−;Sp8+/−) exhibit a split-hand/foot malformation phenotype with double dorsal digit tips probably due to an irregular and immature AER that is not maintained in the center of the bud and on the abnormal expansion of Wnt7a expression to the ventral ectoderm. Our data are compatible with Sp6 and Sp8 working together and in a dose-dependent manner as indispensable mediators of Wnt/βcatenin and Bmp signaling in the limb ectoderm. We suggest that the function of these factors links proximal-distal and dorsal-ventral patterning. In this report we examined the functional roles of Sp6 and Sp8 during limb development using compound loss-of-function mutants. Sp6 and Sp8, two members of the Sp gene family, are expressed in the limb bud ectoderm and function downstream of WNT/βcatenin signaling for Fgf8 induction. The analysis of the allelic series shows that the progressive reduction in the dose of Sp6 and Sp8 gene products leads to predictable morphology, from syndactyly, to split hand/foot malformation, oligodactyly, truncation and finally amelia, indicating that these two factors act in a complementary manner. The molecular characterization of the mutant limbs reveal that Sp6/Sp8 are required in a dose-dependent manner for Fgf8 and En1 induction, thereby placing them as an important link between the induction of the AER and the establishment of dorsal-ventral patterning during limb development.
Collapse
|
47
|
Xie Y, Zhou S, Chen H, Du X, Chen L. Recent research on the growth plate: Advances in fibroblast growth factor signaling in growth plate development and disorders. J Mol Endocrinol 2014; 53:T11-34. [PMID: 25114206 DOI: 10.1530/jme-14-0012] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Skeletons are formed through two distinct developmental actions, intramembranous ossification and endochondral ossification. During embryonic development, most bone is formed by endochondral ossification. The growth plate is the developmental center for endochondral ossification. Multiple signaling pathways participate in the regulation of endochondral ossification. Fibroblast growth factor (FGF)/FGF receptor (FGFR) signaling has been found to play a vital role in the development and maintenance of growth plates. Missense mutations in FGFs and FGFRs can cause multiple genetic skeletal diseases with disordered endochondral ossification. Clarifying the molecular mechanisms of FGFs/FGFRs signaling in skeletal development and genetic skeletal diseases will have implications for the development of therapies for FGF-signaling-related skeletal dysplasias and growth plate injuries. In this review, we summarize the recent advances in elucidating the role of FGFs/FGFRs signaling in growth plate development, genetic skeletal disorders, and the promising therapies for those genetic skeletal diseases resulting from FGFs/FGFRs dysfunction. Finally, we also examine the potential important research in this field in the future.
Collapse
Affiliation(s)
- Yangli Xie
- Department of Rehabilitation MedicineCenter of Bone Metabolism and Repair, Trauma Center, State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Siru Zhou
- Department of Rehabilitation MedicineCenter of Bone Metabolism and Repair, Trauma Center, State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Hangang Chen
- Department of Rehabilitation MedicineCenter of Bone Metabolism and Repair, Trauma Center, State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Xiaolan Du
- Department of Rehabilitation MedicineCenter of Bone Metabolism and Repair, Trauma Center, State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Lin Chen
- Department of Rehabilitation MedicineCenter of Bone Metabolism and Repair, Trauma Center, State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| |
Collapse
|
48
|
Al-Qattan MM. Central and ulnar cleft hands: a review of concurrent deformities in a series of 47 patients and their pathogenesis. J Hand Surg Eur Vol 2014; 39:510-9. [PMID: 23824219 DOI: 10.1177/1753193413496945] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Two main types of cleft hands have been described. The ulnar cleft hand deformity is very rare and is characterized by two constant features: a deep cleft radial to the little finger and hypoplasia of the ulnar digits. The pathogenesis of ulnar clefts is unknown. The second type is the central cleft hand deformity, which is characterized by a soft tissue/bone defect in the hand centrally. Patients with central clefts also have several concurrent deformities in the remaining digits. This paper reviews the clinical features of three cases with ulnar cleft hands and 44 cases of central cleft hands, with special emphasis on concurrent deformities. The author's hypothesis of pathogenesis for both types of clefts and their concurrent deformities is then offered.
Collapse
Affiliation(s)
- M M Al-Qattan
- Division of Plastic Surgery, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
49
|
Danopoulos S, Parsa S, Al Alam D, Tabatabai R, Baptista S, Tiozzo C, Carraro G, Wheeler M, Barreto G, Braun T, Li X, Hajihosseini MK, Bellusci S. Transient Inhibition of FGFR2b-ligands signaling leads to irreversible loss of cellular β-catenin organization and signaling in AER during mouse limb development. PLoS One 2013; 8:e76248. [PMID: 24167544 PMCID: PMC3805551 DOI: 10.1371/journal.pone.0076248] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Accepted: 08/21/2013] [Indexed: 01/30/2023] Open
Abstract
The vertebrate limbs develop through coordinated series of inductive, growth and patterning events. Fibroblast Growth Factor receptor 2b (FGFR2b) signaling controls the induction of the Apical Ectodermal Ridge (AER) but its putative roles in limb outgrowth and patterning, as well as in AER morphology and cell behavior have remained unclear. We have investigated these roles through graded and reversible expression of soluble dominant-negative FGFR2b molecules at various times during mouse limb development, using a doxycycline/transactivator/tet(O)-responsive system. Transient attenuation (≤24 hours) of FGFR2b-ligands signaling at E8.5, prior to limb bud induction, leads mostly to the loss or truncation of proximal skeletal elements with less severe impact on distal elements. Attenuation from E9.5 onwards, however, has an irreversible effect on the stability of the AER, resulting in a progressive loss of distal limb skeletal elements. The primary consequences of FGFR2b-ligands attenuation is a transient loss of cell adhesion and down-regulation of P63, β1-integrin and E-cadherin, and a permanent loss of cellular β-catenin organization and WNT signaling within the AER. Combined, these effects lead to the progressive transformation of the AER cells from pluristratified to squamous epithelial-like cells within 24 hours of doxycycline administration. These findings show that FGFR2b-ligands signaling has critical stage-specific roles in maintaining the AER during limb development.
Collapse
Affiliation(s)
- Soula Danopoulos
- Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
- Developmental Biology and Regenerative Medicine Program, Saban Research Institute of Childrens Hospital Los Angeles, Los Angeles, California, United States of America
| | - Sara Parsa
- Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
- Developmental Biology and Regenerative Medicine Program, Saban Research Institute of Childrens Hospital Los Angeles, Los Angeles, California, United States of America
- Life Science Division, Lawrence Berkeley National Laboratory, Berkeley, California, United States of America
| | - Denise Al Alam
- Developmental Biology and Regenerative Medicine Program, Saban Research Institute of Childrens Hospital Los Angeles, Los Angeles, California, United States of America
| | - Reza Tabatabai
- Developmental Biology and Regenerative Medicine Program, Saban Research Institute of Childrens Hospital Los Angeles, Los Angeles, California, United States of America
| | - Sheryl Baptista
- Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Caterina Tiozzo
- Developmental Biology and Regenerative Medicine Program, Saban Research Institute of Childrens Hospital Los Angeles, Los Angeles, California, United States of America
- Nassau University Medical Center, Pediatric Department, New York, New York, United States of America
| | - Gianni Carraro
- Department of Internal Medicine II, University of Giessen Lung Center and Member of the German Lung Research Center (DZL), Giessen, Germany
| | - Matthew Wheeler
- Departement of Cardiac Development and Remodelling, Max-Planck Institute for Heart and Lung Research and Member of the DZL, Bad Nauheim, Germany
| | - Guillermo Barreto
- Max-Planck-Institute for Heart and Lung Research, LOEWE Research Group Lung Cancer Epigenetic, Bad Nauheim, Germany
| | - Thomas Braun
- Departement of Cardiac Development and Remodelling, Max-Planck Institute for Heart and Lung Research and Member of the DZL, Bad Nauheim, Germany
| | - Xiaokun Li
- School of Pharmacy, Wenzhou Medical College, Wenzhou, China
| | - Mohammad K. Hajihosseini
- School of Biological Sciences, University of East Anglia (UEA), Norwich, Norfolk, United Kingdom
| | - Saverio Bellusci
- Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
- Developmental Biology and Regenerative Medicine Program, Saban Research Institute of Childrens Hospital Los Angeles, Los Angeles, California, United States of America
- Department of Internal Medicine II, University of Giessen Lung Center and Member of the German Lung Research Center (DZL), Giessen, Germany
- * E-mail:
| |
Collapse
|
50
|
Gao B, Yang Y. Planar cell polarity in vertebrate limb morphogenesis. Curr Opin Genet Dev 2013; 23:438-44. [PMID: 23747034 PMCID: PMC3759593 DOI: 10.1016/j.gde.2013.05.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Revised: 04/29/2013] [Accepted: 05/07/2013] [Indexed: 11/21/2022]
Abstract
Studies of the vertebrate limb development have contributed significantly to understanding the fundamental mechanisms underlying growth, patterning, and morphogenesis of a complex multicellular organism. In the limb, well-defined signaling centers interact to coordinate limb growth and patterning along the three axes. Recent analyses of live imaging and mathematical modeling have provided evidence that polarized cell behaviors governed by morphogen gradients play an important role in shaping the limb bud. Furthermore, the Wnt/planar cell polarity (PCP) pathway that controls uniformly polarized cell behaviors in a field of cells has emerged to be critical for directional morphogenesis in the developing limb. Directional information coded in the morphogen gradient may be interpreted by responding cells through regulating the activities of PCP components in a Wnt morphogen dose-dependent manner.
Collapse
Affiliation(s)
- Bo Gao
- National Human Genome Research Institute, Bethesda, MD 20892, United States
| | | |
Collapse
|