1
|
Wang H, Chen X, Meng X, Cao Y, Han S, Liu K, Zhao X, Zhao X, Zhang X. The pathogenic mechanism of syndactyly type V identified in a Hoxd13Q50R knock-in mice. Bone Res 2024; 12:21. [PMID: 38561387 PMCID: PMC10984994 DOI: 10.1038/s41413-024-00322-y] [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: 04/24/2023] [Revised: 01/30/2024] [Accepted: 02/19/2024] [Indexed: 04/04/2024] Open
Abstract
Syndactyly type V (SDTY5) is an autosomal dominant extremity malformation characterized by fusion of the fourth and fifth metacarpals. In the previous publication, we first identified a heterozygous missense mutation Q50R in homeobox domain (HD) of HOXD13 in a large Chinese family with SDTY5. In order to substantiate the pathogenicity of the variant and elucidate the underlying pathogenic mechanism causing limb malformation, transcription-activator-like effector nucleases (TALEN) was employed to generate a Hoxd13Q50R mutant mouse. The mutant mice exhibited obvious limb malformations including slight brachydactyly and partial syndactyly between digits 2-4 in the heterozygotes, and severe syndactyly, brachydactyly and polydactyly in homozygotes. Focusing on BMP2 and SHH/GREM1/AER-FGF epithelial mesenchymal (e-m) feedback, a crucial signal pathway for limb development, we found the ectopically expressed Shh, Grem1 and Fgf8 and down-regulated Bmp2 in the embryonic limb bud at E10.5 to E12.5. A transcriptome sequencing analysis was conducted on limb buds (LBs) at E11.5, revealing 31 genes that exhibited notable disparities in mRNA level between the Hoxd13Q50R homozygotes and the wild-type. These genes are known to be involved in various processes such as limb development, cell proliferation, migration, and apoptosis. Our findings indicate that the ectopic expression of Shh and Fgf8, in conjunction with the down-regulation of Bmp2, results in a failure of patterning along both the anterior-posterior and proximal-distal axes, as well as a decrease in interdigital programmed cell death (PCD). This cascade ultimately leads to the development of syndactyly and brachydactyly in heterozygous mice, and severe limb malformations in homozygous mice. These findings suggest that abnormal expression of SHH, FGF8, and BMP2 induced by HOXD13Q50R may be responsible for the manifestation of human SDTY5.
Collapse
Affiliation(s)
- Han Wang
- McKusick-Zhang Center for Genetic Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Department of Medical Genetics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
- Department of Orthopedics, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| | - Xiumin Chen
- McKusick-Zhang Center for Genetic Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Department of Medical Genetics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
| | - Xiaolu Meng
- McKusick-Zhang Center for Genetic Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Department of Medical Genetics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
| | - Yixuan Cao
- McKusick-Zhang Center for Genetic Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Department of Medical Genetics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
| | - Shirui Han
- McKusick-Zhang Center for Genetic Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Department of Medical Genetics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
| | - Keqiang Liu
- McKusick-Zhang Center for Genetic Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Department of Medical Genetics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
| | - Ximeng Zhao
- McKusick-Zhang Center for Genetic Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Department of Medical Genetics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
| | - Xiuli Zhao
- McKusick-Zhang Center for Genetic Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Department of Medical Genetics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China.
| | - Xue Zhang
- McKusick-Zhang Center for Genetic Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Department of Medical Genetics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China.
| |
Collapse
|
2
|
Bardhan S, Bhargava N, Dighe S, Vats N, Naganathan SR. Emergence of a left-right symmetric body plan in vertebrate embryos. Curr Top Dev Biol 2024; 159:310-342. [PMID: 38729680 DOI: 10.1016/bs.ctdb.2024.01.003] [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: 05/12/2024]
Abstract
External bilateral symmetry is a prevalent feature in vertebrates, which emerges during early embryonic development. To begin with, vertebrate embryos are largely radially symmetric before transitioning to bilaterally symmetry, after which, morphogenesis of various bilateral tissues (e.g somites, otic vesicle, limb bud), and structures (e.g palate, jaw) ensue. While a significant amount of work has probed the mechanisms behind symmetry breaking in the left-right axis leading to asymmetric positioning of internal organs, little is known about how bilateral tissues emerge at the same time with the same shape and size and at the same position on the two sides of the embryo. By discussing emergence of symmetry in many bilateral tissues and structures across vertebrate model systems, we highlight that understanding symmetry establishment is largely an open field, which will provide deep insights into fundamental problems in developmental biology for decades to come.
Collapse
Affiliation(s)
- Siddhartha Bardhan
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Nandini Bhargava
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Swarali Dighe
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Neha Vats
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Sundar Ram Naganathan
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India.
| |
Collapse
|
3
|
Bahrami M, Darabi S, Roozbahany NA, Abbaszadeh HA, Moghadasali R. Great potential of renal progenitor cells in kidney: From the development to clinic. Exp Cell Res 2024; 434:113875. [PMID: 38092345 DOI: 10.1016/j.yexcr.2023.113875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 12/02/2023] [Accepted: 12/03/2023] [Indexed: 12/23/2023]
Abstract
The mammalian renal organ represents a pinnacle of complexity, housing functional filtering units known as nephrons. During embryogenesis, the depletion of niches containing renal progenitor cells (RPCs) and the subsequent incapacity of adult kidneys to generate new nephrons have prompted the formulation of protocols aimed at isolating residual RPCs from mature kidneys and inducing their generation from diverse cell sources, notably pluripotent stem cells. Recent strides in the realm of regenerative medicine and the repair of tissues using stem cells have unveiled critical signaling pathways essential for the maintenance and generation of human RPCs in vitro. These findings have ushered in a new era for exploring novel strategies for renal protection. The present investigation delves into potential transcription factors and signaling cascades implicated in the realm of renal progenitor cells, focusing on their protection and differentiation. The discourse herein elucidates contemporary research endeavors dedicated to the acquisition of progenitor cells, offering crucial insights into the developmental mechanisms of these cells within the renal milieu and paving the way for the formulation of innovative treatment modalities.
Collapse
Affiliation(s)
- Maryam Bahrami
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Laser Applications in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shahram Darabi
- Cellular and Molecular Research Center, Research Institute for Non-Communicable Diseases, Qazvin University of Medical Sciences, Qazvin, Iran
| | | | - Hojjat Allah Abbaszadeh
- Laser Applications in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Reza Moghadasali
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| |
Collapse
|
4
|
Wang S, Tanaka Y, Xu Y, Takeda S, Hirokawa N. KIF3B promotes a PI3K signaling gradient causing changes in a Shh protein gradient and suppressing polydactyly in mice. Dev Cell 2022; 57:2273-2289.e11. [DOI: 10.1016/j.devcel.2022.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 07/27/2022] [Accepted: 09/13/2022] [Indexed: 11/03/2022]
|
5
|
Sader F, Roy S. Tgf-β superfamily and limb regeneration: Tgf-β to start and Bmp to end. Dev Dyn 2021; 251:973-987. [PMID: 34096672 DOI: 10.1002/dvdy.379] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 05/31/2021] [Accepted: 06/02/2021] [Indexed: 12/19/2022] Open
Abstract
Axolotls represent a popular model to study how nature solved the problem of regenerating lost appendages in tetrapods. Our work over many years focused on trying to understand how these animals can achieve such a feat and not end up with a scarred up stump. The Tgf-β superfamily represents an interesting family to target since they are involved in wound healing in adults and pattern formation during development. This family is large and comprises Tgf-β, Bmps, activins and GDFs. In this review, we present work from us and others on Tgf-β & Bmps and highlight interesting observations between these two sub-families. Tgf-β is important for the preparation phase of regeneration and Bmps for the redevelopment phase and they do not overlap with one another. We present novel data showing that the Tgf-β non-canonical pathway is also not active during redevelopment. Finally, we propose a molecular model to explain how Tgf-β and Bmps maintain distinct windows of expression during regeneration in axolotls.
Collapse
Affiliation(s)
- Fadi Sader
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Université de Montréal, Montréal, Québec, Canada
| | - Stéphane Roy
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Université de Montréal, Montréal, Québec, Canada.,Department of Stomatology, Faculty of Dentistry, Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
6
|
Shimizu M, Tachikawa S, Saitoh N, Nakazono K, Yu-Jung L, Suga M, Ohnuma K. Thalidomide affects limb formation and multiple myeloma related genes in human induced pluripotent stem cells and their mesoderm differentiation. Biochem Biophys Rep 2021; 26:100978. [PMID: 33763605 PMCID: PMC7973312 DOI: 10.1016/j.bbrep.2021.100978] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 03/02/2021] [Accepted: 03/03/2021] [Indexed: 12/28/2022] Open
Abstract
Although thalidomide is highly teratogenic, it has been prescribed for treating multiple myeloma and Hansen's disease. However, its mechanism of action is not fully understood. Here, we employed a reverse transcription quantitative PCR array to measure the expression of 84 genes in human induced pluripotent stem cells (hiPSCs) and their mesodermal differentiation. Thalidomide altered the expression of undifferentiated marker genes in both cell types. Thalidomide affected more genes in the mesoderm than in the hiPSCs. Ectoderm genes were upregulated but mesendoderm genes were downregulated by thalidomide during mesoderm induction, suggesting that thalidomide altered mesoderm differentiation. We found that FABP7 (fatty acid binding protein 7) was dramatically downregulated in the hiPSCs. FABP is related to retinoic acid, which is important signaling for limb formation. Moreover, thalidomide altered the expression of the genes involved in TGF-β signaling, limb formation, and multiple myeloma, which are related to thalidomide-induced malformations and medication. In summary, iPSCs can serve as useful tools to elucidate the mechanisms underlying thalidomide malformations in vitro. Thalidomide downregulated FABP7, a fatty acid binding protein (FABP) cording gene. FABP is related to retinoic acid, which is important signaling for limb formation. Thalidomide treatment affected the expression of limb formation related genes. Thalidomide treatment affected 5 genes related to multiple myeloma. Thalidomide upregulated ectoderm but downregulated mesendoderm markers in mesoderm.
Collapse
Affiliation(s)
- Maho Shimizu
- Department of Bioengineering, Nagaoka University of Technology, 1603-1 Kamitomioka, Nagaoka, Niigata, 940-2188, Japan
| | - Saoko Tachikawa
- Department of Bioengineering, Nagaoka University of Technology, 1603-1 Kamitomioka, Nagaoka, Niigata, 940-2188, Japan
| | - Nagatsuki Saitoh
- Department of Bioengineering, Nagaoka University of Technology, 1603-1 Kamitomioka, Nagaoka, Niigata, 940-2188, Japan
| | - Kohei Nakazono
- Department of Bioengineering, Nagaoka University of Technology, 1603-1 Kamitomioka, Nagaoka, Niigata, 940-2188, Japan
| | - Liu Yu-Jung
- Laboratory of Stem Cell Cultures, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8 Saito-Asagi, Osaka, Ibaraki, 567-0085, Japan
| | - Mika Suga
- Laboratory of Stem Cell Cultures, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8 Saito-Asagi, Osaka, Ibaraki, 567-0085, Japan
| | - Kiyoshi Ohnuma
- Department of Bioengineering, Nagaoka University of Technology, 1603-1 Kamitomioka, Nagaoka, Niigata, 940-2188, Japan.,Department of Science of Technology Innovation, Nagaoka University of Technology, 1603-1 Kamitomioka, Nagaoka, Niigata, 940-2188, Japan
| |
Collapse
|
7
|
Truong BT, Artinger KB. The power of zebrafish models for understanding the co-occurrence of craniofacial and limb disorders. Genesis 2021; 59:e23407. [PMID: 33393730 PMCID: PMC8153179 DOI: 10.1002/dvg.23407] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 12/23/2020] [Accepted: 12/24/2020] [Indexed: 12/30/2022]
Abstract
Craniofacial and limb defects are two of the most common congenital anomalies in the general population. Interestingly, these defects are not mutually exclusive. Many patients with craniofacial phenotypes, such as orofacial clefting and craniosynostosis, also present with limb defects, including polydactyly, syndactyly, brachydactyly, or ectrodactyly. The gene regulatory networks governing craniofacial and limb development initially seem distinct from one another, and yet these birth defects frequently occur together. Both developmental processes are highly conserved among vertebrates, and zebrafish have emerged as an advantageous model due to their high fecundity, relative ease of genetic manipulation, and transparency during development. Here we summarize studies that have used zebrafish models to study human syndromes that present with both craniofacial and limb phenotypes. We discuss the highly conserved processes of craniofacial and limb/fin development and describe recent zebrafish studies that have explored the function of genes associated with human syndromes with phenotypes in both structures. We attempt to identify commonalities between the two to help explain why craniofacial and limb anomalies often occur together.
Collapse
Affiliation(s)
- Brittany T. Truong
- Human Medical Genetics & Genomics Graduate Program, University of Colorado Denver Anschutz Medical Campus, Aurora, CO
- Department of Craniofacial Biology, University of Colorado Denver Anschutz Medical Campus, Aurora, CO
| | - Kristin Bruk Artinger
- Department of Craniofacial Biology, University of Colorado Denver Anschutz Medical Campus, Aurora, CO
| |
Collapse
|
8
|
Bernheim S, Meilhac SM. Mesoderm patterning by a dynamic gradient of retinoic acid signalling. Philos Trans R Soc Lond B Biol Sci 2020; 375:20190556. [PMID: 32829679 PMCID: PMC7482219 DOI: 10.1098/rstb.2019.0556] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/11/2020] [Indexed: 12/15/2022] Open
Abstract
Retinoic acid (RA), derived from vitamin A, is a major teratogen, clinically recognized in 1983. Identification of its natural presence in the embryo and dissection of its molecular mechanism of action became possible in the animal model with the advent of molecular biology, starting with the cloning of its nuclear receptor. In normal development, the dose of RA is tightly controlled to regulate organ formation. Its production depends on enzymes, which have a dynamic expression profile during embryonic development. As a small molecule, it diffuses rapidly and acts as a morphogen. Here, we review advances in deciphering how endogenously produced RA provides positional information to cells. We compare three mesodermal tissues, the limb, the somites and the heart, and discuss how RA signalling regulates antero-posterior and left-right patterning. A common principle is the establishment of its spatio-temporal dynamics by positive and negative feedback mechanisms and by antagonistic signalling by FGF. However, the response is cell-specific, pointing to the existence of cofactors and effectors, which are as yet incompletely characterized. This article is part of a discussion meeting issue 'Contemporary morphogenesis'.
Collapse
Affiliation(s)
- Ségolène Bernheim
- Imagine-Institut Pasteur, Laboratory of Heart Morphogenesis, 75015 Paris, France
- INSERM UMR1163, 75015 Paris, France
- Université de Paris, Paris, France
| | - Sigolène M. Meilhac
- Imagine-Institut Pasteur, Laboratory of Heart Morphogenesis, 75015 Paris, France
- INSERM UMR1163, 75015 Paris, France
- Université de Paris, Paris, France
| |
Collapse
|
9
|
Son C, Choi MS, Park JC. Different Responsiveness of Alveolar Bone and Long Bone to Epithelial-Mesenchymal Interaction-Related Factor. JBMR Plus 2020; 4:e10382. [PMID: 32803111 PMCID: PMC7422712 DOI: 10.1002/jbm4.10382] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 06/02/2020] [Indexed: 12/29/2022] Open
Abstract
Alveolar bone is both morphologically and functionally different from other bones of the axial or peripheral skeleton. Because of its sensitive nature to external stimuli including mechanical stress, bone loss stimuli, and medication-related osteonecrosis of the jaw, alveolar bone rendering is seen as an important factor in various dental surgical processes. Although multiple studies have validated the response of long bone to various factors, how alveolar bone responds to functional stimuli still needs further clarification. To examine the characteristics of bone in vitro, we isolated cells from alveolar, femur, and tibia bone tissue. Although primary cultured mouse alveolar bone-derived cells (mABDCs) and mouse long bone-derived cells (mLBDCs) exhibited similar osteoblastic characteristics, morphology, and proliferation rates, both showed distinct expression of neural crest (NC) and epithelial-mesenchymal interaction (EMI)-related genes. Furthermore, they showed significantly different mineralization rates. RNA sequencing data demonstrated distinct transcriptome profiles of alveolar bone and long bone. Osteogenic, NC-, and EMI-related genes showed distinct expression between mABDCs and mLBDCs. When the gene expression patterns during osteogenic differentiation were analyzed, excluding several osteogenic genes, NC- and EMI-related genes showed different expression patterns. Among EMI-related proteins, BMP4 elevated the expression levels of osteogenic genes, Msx2, Dlx5, and Bmp2 the most, more noticeably in mABDCs than in mLBDCs during osteogenic differentiation. In in vivo models, the BMP4-treated mABDC group showed massive bone formation and maturation as opposed to its counterpart. Bone sialoprotein expression was also validated in calcified tissues. Overall, our data suggest that alveolar bone and long bone have different responsiveness to EMI by distinct gene regulation. In particular, BMP4 has critical bone formation effects on alveolar bone, but not on long bone. © 2020 The Authors. JBMR Plus published by Wiley Periodicals, Inc. on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Chul Son
- Department of Oral Histology and Developmental Biology, School of Dentistry Seoul National University Seoul South Korea
| | - Moon Sil Choi
- Department of Dental Hygiene Songwon University Gwangju South Korea
| | - Joo-Cheol Park
- Department of Oral Histology and Developmental Biology, School of Dentistry Seoul National University Seoul South Korea
| |
Collapse
|
10
|
Suzuki T, Nakano M, Komatsu M, Takahashi J, Kato H, Nakamura Y. ZMAT2, a newly-identified potential disease-causing gene in congenital radioulnar synostosis, modulates BMP signaling. Bone 2020; 136:115349. [PMID: 32247068 DOI: 10.1016/j.bone.2020.115349] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 03/29/2020] [Accepted: 03/29/2020] [Indexed: 01/11/2023]
Abstract
Congenital radioulnar synostosis (RUS) is a rare skeletal disorder that is characterized by fusion of the radius and ulna. As the etiology of RUS is largely unknown, its treatment options are currently limited. A de novo missense mutation in the zinc finger matrin-type 2 (ZMAT2) gene was newly identified in a 5-year-old boy with RUS using whole-exome sequencing. Herein, we sought to further explore the function of zmat2 in zebrafish. Whole-mount in situ hybridization revealed site-specific expression of zzmat2 in the pectoral fins (equivalent to human upper limbs) and craniofacial regions, while immunohistochemistry showed the expression of zZmat2 in the pectoral fins and heart region. Gene knockdown produced defects in the pectoral fins and dorso-ventral patterning. zzmat2 knockdown also caused embryo dorsalization, a phenotype consistent with reduced/insufficient bone morphogenetic protein (BMP) signaling. These abnormalities were partially rescued by zbmp2b RNA overexpression and fully rescued by simultaneous overexpression of wild-type zzmat2. Importantly, the overexpression of mutant zzmat2 corresponding to the newly-identified mutation did not fully rescue the dorso-ventral patterning defects. The above findings indicate that ZMAT2 regulates skeletal development via the BMP signaling pathway, and its mutation may lead to a loss or reduction in biological activity. Thus, the newly identified ZMAT2 mutation potentially plays a causal role in RUS through deregulation of BMP signaling.
Collapse
Affiliation(s)
- Takako Suzuki
- Department of Orthopaedic Surgery, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan; Department of Human Nutrition, Faculty of Human Nutrition, Tokyo Kasei Gakuin University, 22 Sanban-cho, Chiyoda-ku, Tokyo 102-8341, Japan
| | - Masaki Nakano
- Department of Orthopaedic Surgery, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan
| | - Masatoshi Komatsu
- Department of Orthopaedic Surgery, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan
| | - Jun Takahashi
- Department of Orthopaedic Surgery, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan
| | - Hiroyuki Kato
- Department of Orthopaedic Surgery, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan
| | - Yukio Nakamura
- Department of Orthopaedic Surgery, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan.
| |
Collapse
|
11
|
Dreher SI, Fischer J, Walker T, Diederichs S, Richter W. Significance of MEF2C and RUNX3 Regulation for Endochondral Differentiation of Human Mesenchymal Progenitor Cells. Front Cell Dev Biol 2020; 8:81. [PMID: 32195247 PMCID: PMC7064729 DOI: 10.3389/fcell.2020.00081] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 01/29/2020] [Indexed: 12/14/2022] Open
Abstract
Guiding progenitor cell development between chondral versus endochondral pathways is still an unachieved task of cartilage neogenesis, and human mesenchymal progenitor cell (MPC) chondrogenesis is considered as a valuable model to better understand hypertrophic development of chondrocytes. Transcription factors Runx2, Runx3, and Mef2c play prominent roles for chondrocyte hypertrophy during mouse development, but little is known on the importance of these key fate-determining factors for endochondral development of human MPCs. The aim of this study was to unravel the regulation of RUNX2, RUNX3, and MEF2C during MPC chondrogenesis, the pathways driving their expression, and the downstream hypertrophic targets affected by their regulation. RUNX2, RUNX3, and MEF2C gene expression was differentially regulated during chondrogenesis of MPCs, but remained low and unregulated when non-hypertrophic articular chondrocytes were differentiated under the same conditions. RUNX3 and MEF2C mRNA and protein levels rose in parallel to hypertrophic marker upregulation, but surprisingly, RUNX2 gene expression changed only by trend and RUNX2 protein remained undetectable. While RUNX3 expression was driven by TGF-β and BMP signaling, MEF2C responded to WNT-, BMP-, and Hedgehog-pathway inhibition. MEF2C but not RUNX3 levels correlated significantly with COL10A1, IHH, and IBSP gene expression when hypertrophy was attenuated. IBSP was a downstream target of RUNX3 and MEF2C but not RUNX2 in SAOS-2 cells, underlining the capacity of RUNX3 and MEF2C to stimulate osteogenic marker expression in human cells. Conclusively, RUNX3 and MEF2C appeared more important than RUNX2 for human endochondral MPC chondrogenesis. Pathways altering the speed of chondrogenesis (FGF, TGF-β, BMP) affected RUNX2 or RUNX3, while pathways changing hypertrophy (WNT, PTHrP/HH) regulated mainly MEF2C. Taken together, reduction of MEF2C levels is a new goal to shift human cartilage neogenesis toward the chondral pathway.
Collapse
Affiliation(s)
- Simon I Dreher
- Research Center for Experimental Orthopaedics, Heidelberg University Hospital, Heidelberg, Germany
| | - Jennifer Fischer
- Research Center for Experimental Orthopaedics, Heidelberg University Hospital, Heidelberg, Germany
| | - Tilman Walker
- Clinic for Orthopaedics and Trauma Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Solvig Diederichs
- Research Center for Experimental Orthopaedics, Heidelberg University Hospital, Heidelberg, Germany
| | - Wiltrud Richter
- Research Center for Experimental Orthopaedics, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
12
|
dos Santos ÍGD, de Oliveira Mendes TA, Silva GAB, Reis AMS, Monteiro-Vitorello CB, Schaker PDC, Herai RH, Fabotti ABC, Coutinho LL, Jorge EC. Didelphis albiventris: an overview of unprecedented transcriptome sequencing of the white-eared opossum. BMC Genomics 2019; 20:866. [PMID: 31730444 PMCID: PMC6858782 DOI: 10.1186/s12864-019-6240-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Accepted: 10/29/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The white-eared opossum (Didelphis albiventris) is widely distributed throughout Brazil and South America. It has been used as an animal model for studying different scientific questions ranging from the restoration of degraded green areas to medical aspects of Chagas disease, leishmaniasis and resistance against snake venom. As a marsupial, D. albiventris can also contribute to the understanding of the molecular mechanisms that govern the different stages of organogenesis. Opossum joeys are born after only 13 days, and the final stages of organogenesis occur when the neonates are inside the pouch, depending on lactation. As neither the genome of this opossum species nor its transcriptome has been completely sequenced, the use of D. albiventris as an animal model is limited. In this work, we sequenced the D. albiventris transcriptome by RNA-seq to obtain the first catalogue of differentially expressed (DE) genes and gene ontology (GO) annotations during the neonatal stages of marsupial development. RESULTS The D. albiventris transcriptome was obtained from whole neonates harvested at birth (P0), at 5 days of age (P5) and at 10 days of age (P10). The de novo assembly of these transcripts generated 85,338 transcripts. Approximately 30% of these transcripts could be mapped against the amino acid sequences of M. domestica, the evolutionarily closest relative of D. albiventris to be sequenced thus far. Among the expressed transcripts, 2077 were found to be DE between P0 and P5, 13,780 between P0 and P10, and 1453 between P5 and P10. The enriched GO terms were mainly related to the immune system, blood tissue development and differentiation, vision, hearing, digestion, the CNS and limb development. CONCLUSIONS The elucidation of opossum transcriptomes provides an out-group for better understanding the distinct characteristics associated with the evolution of mammalian species. This study provides the first transcriptome sequences and catalogue of genes for a marsupial species at different neonatal stages, allowing the study of the mechanisms involved in organogenesis.
Collapse
Affiliation(s)
- Íria Gabriela Dias dos Santos
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais Brazil
| | | | - Gerluza Aparecida Borges Silva
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais Brazil
| | - Amanda Maria Sena Reis
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais Brazil
| | | | - Patricia Dayane Carvalho Schaker
- Departamento de Genética, Escola Superior de Agricultura Luiz de Queiroz, Universidade de São Paulo, Piracicaba, São Paulo Brazil
| | - Roberto Hirochi Herai
- Graduate Program in Health Sciences, School of Medicine, Pontifícia Universidade Católica do Paraná (PUCPR), Curitiba, Paraná, Brazil
| | | | - Luiz Lehmann Coutinho
- Departamento de Zootecnia, Escola Superior de Agricultura Luiz de Queiroz, Universidade de São Paulo, Piracicaba, São Paulo Brazil
| | - Erika Cristina Jorge
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais Brazil
| |
Collapse
|
13
|
Regulatory roles of epithelial-mesenchymal interaction (EMI) during early and androgen dependent external genitalia development. Differentiation 2019; 110:29-35. [PMID: 31590136 DOI: 10.1016/j.diff.2019.08.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Revised: 08/23/2019] [Accepted: 08/26/2019] [Indexed: 02/07/2023]
Abstract
Development of external genitalia (ExG) has been a topic of long mystery in the field of organogenesis research. Early stage male and female of mouse embryos develop a common genital tubercle (GT) in the perineum whose outgrowth extends distally from the posterior cloacal regions. Concomitant with GT outgrowth, the cloaca is divided into urogenital sinus and anorectum by urorectal septum (URS) internally. The outgrowth of the GT is associated with the formation of endodermal epithelial urethral plate (UP) attached to the ventral epidermis of the GT. Such a common developmental phase is observed until around embryonic day 15.5 (E15.5) morphologically in mouse embryogenesis. Various growth factor genes, such as Fibroblast growth factor (Fgf) and Wnt genes are expressed and function during GT formation. Since the discovery of key growth factor signals and several regulatory molecules, elucidation of their functions has been achieved utilizing mouse developmental models, conditional gene knockout mouse and in vitro culture. Analyses on the phenotypes of such mouse models have revealed that several growth factor families play fundamental roles in ExG organogenesis based on the epithelial-mesenchymal interaction (EMI). More recently, EMI between developing urethral epithelia and its bilateral mesenchyme of later stages is also reported during subsequent stage of androgen-dependent male-type urethral formation in the mouse embryo. Mafb, belonging to AP-1 family and a key androgen-responsive mesenchymal gene, is identified and starts to be expressed around E14.5 when masculinization of the urethra is initiated. Mesenchymal cell condensation and migration, which are regulated by nonmuscle myosin, are shown to be essential process for masculinization. Hence, studies on EMI at various embryonic stages are important not only for early but also for subsequent masculinization of the urethra. In this review, a dynamic mode of EMI for both early and late phases of ExG development is discussed.
Collapse
|
14
|
UHRF genes regulate programmed interdigital tissue regression and chondrogenesis in the embryonic limb. Cell Death Dis 2019; 10:347. [PMID: 31024001 PMCID: PMC6484032 DOI: 10.1038/s41419-019-1575-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 03/26/2019] [Accepted: 04/04/2019] [Indexed: 12/12/2022]
Abstract
The primordium of the limb contains a number of progenitors far superior to those necessary to form the skeletal components of this appendage. During the course of development, precursors that do not follow the skeletogenic program are removed by cell senescence and apoptosis. The formation of the digits provides the most representative example of embryonic remodeling via cell degeneration. In the hand/foot regions of the embryonic vertebrate limb (autopod), the interdigital tissue and the zones of interphalangeal joint formation undergo massive degeneration that accounts for jointed and free digit morphology. Developmental senescence and caspase-dependent apoptosis are considered responsible for these remodeling processes. Our study uncovers a new upstream level of regulation of remodeling by the epigenetic regulators Uhrf1 and Uhrf2 genes. These genes are spatially and temporally expressed in the pre-apoptotic regions. UHRF1 and UHRF2 showed a nuclear localization associated with foci of methylated cytosine. Interestingly, nuclear labeling increased in cells progressing through the stages of degeneration prior to TUNEL positivity. Functional analysis in cultured limb skeletal progenitors via the overexpression of either UHRF1 or UHRF2 inhibited chondrogenesis and induced cell senescence and apoptosis accompanied with changes in global and regional DNA methylation. Uhrfs modulated canonical cell differentiation factors, such as Sox9 and Scleraxis, promoted apoptosis via up-regulation of Bak1, and induced cell senescence, by arresting progenitors at the S phase and upregulating the expression of p21. Expression of Uhrf genes in vivo was positively modulated by FGF signaling. In the micromass culture assay Uhrf1 was down-regulated as the progenitors lost stemness and differentiated into cartilage. Together, our findings emphasize the importance of tuning the balance between cell differentiation and cell stemness as a central step in the initiation of the so-called “embryonic programmed cell death” and suggest that the structural organization of the chromatin, via epigenetic modifications, may be a precocious and critical factor in these regulatory events.
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
|
Bloomquist RF, Fowler TE, Sylvester JB, Miro RJ, Streelman JT. A compendium of developmental gene expression in Lake Malawi cichlid fishes. BMC DEVELOPMENTAL BIOLOGY 2017; 17:3. [PMID: 28158974 PMCID: PMC5291978 DOI: 10.1186/s12861-017-0146-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 01/26/2017] [Indexed: 12/24/2022]
Abstract
BACKGROUND Lake Malawi cichlids represent one of a growing number of vertebrate models used to uncover the genetic and developmental basis of trait diversity. Rapid evolutionary radiation has resulted in species that share similar genomes but differ markedly in phenotypes including brains and behavior, nuptial coloration and the craniofacial skeleton. Research has begun to identify the genes, as well as the molecular and developmental pathways that underlie trait divergence. RESULTS We assemble a compendium of gene expression for Lake Malawi cichlids, across pharyngula (the phylotypic stage) and larval stages of development, encompassing hundreds of gene transcripts. We chart patterns of expression in Bone morphogenetic protein (BMP), Fibroblast growth factor (FGF), Hedgehog (Hh), Notch and Wingless (Wnt) signaling pathways, as well as genes involved in neurogenesis, calcium and endocrine signaling, stem cell biology, and numerous homeobox (Hox) factors-in three planes using whole-mount in situ hybridization. Because of low sequence divergence across the Malawi cichlid assemblage, the probes we employ are broadly applicable in hundreds of species. We tabulate gene expression across general tissue domains, and highlight examples of unexpected expression patterns. CONCLUSIONS On the heels of recently published genomes, this compendium of developmental gene expression in Lake Malawi cichlids provides a valuable resource for those interested in the relationship between evolution and development.
Collapse
Affiliation(s)
- R F Bloomquist
- Georgia Institute of Technology, School of Biological Sciences and Institute for Bioengineering and Bioscience, Atlanta, GA, USA.,Medical College of Georgia, School of Dentistry, Augusta, GA, USA
| | - T E Fowler
- Georgia Institute of Technology, School of Biological Sciences and Institute for Bioengineering and Bioscience, Atlanta, GA, USA
| | - J B Sylvester
- Georgia Institute of Technology, School of Biological Sciences and Institute for Bioengineering and Bioscience, Atlanta, GA, USA
| | - R J Miro
- Georgia Institute of Technology, School of Biological Sciences and Institute for Bioengineering and Bioscience, Atlanta, GA, USA
| | - J T Streelman
- Georgia Institute of Technology, School of Biological Sciences and Institute for Bioengineering and Bioscience, Atlanta, GA, USA.
| |
Collapse
|
17
|
Suzuki K, Matsumaru D, Matsushita S, Murashima A, Ludwig M, Reutter H, Yamada G. Epispadias and the associated embryopathies: genetic and developmental basis. Clin Genet 2016; 91:247-253. [PMID: 27649475 DOI: 10.1111/cge.12871] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 09/13/2016] [Accepted: 09/16/2016] [Indexed: 12/25/2022]
Abstract
The abnormalities in the urogenital organs are frequently observed as human developmental diseases. Among such diseases, the defects in the upper part of external genitalia are rather rare named epispadias. The cleft in the dorsal part of external genitalia often reaches to the urethra. In general, the urogenital abnormalities accompany defects in the adjacent tissues and organs. The ventral body wall and bladder can also be affected in the patients with dorsal defects of the external genitalia. Therefore, such multiple malformations are often classified as bladder exstrophy and epispadias complex (BEEC). Because of the lower frequency of such birth defects and their early embryonic development, animal models are required to analyze the pathogenic mechanisms and the functions of responsible genes. Mutant mouse analyses on various signal cascades for external genitalia and body wall development are increasingly performed. The genetic interactions between growth factors such as bone morphogenetic proteins (Bmp) and transcription factors such as Msx1/2 and Isl1 have been suggested to play roles for such organogenesis. The significance of epithelial-mesenchymal interaction (EMI) is suggested during development. In this review, we describe on such local interactions and developmental regulators. We also introduce some mutant mouse models displaying external genitalia-body wall abnormalities.
Collapse
Affiliation(s)
- K Suzuki
- Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University (WMU), Wakayama, Japan
| | - D Matsumaru
- Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University (WMU), Wakayama, Japan
| | - S Matsushita
- Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University (WMU), Wakayama, Japan
| | - A Murashima
- Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University (WMU), Wakayama, Japan.,Division of Human Embryology, Department of Anatomy, Iwate Medical University, Yahaba, Japan
| | - M Ludwig
- Department of Clinical Chemistry and Clinical Pharmacology, University Hospital of Bonn, Bonn, Germany
| | - H Reutter
- Institute of Human Genetics, University Hospital of Bonn, Bonn, Germany.,Department of Neonatology and Pediatric Intensive Care, University Hospital of Bonn, Bonn, Germany
| | - G Yamada
- Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University (WMU), Wakayama, Japan
| |
Collapse
|
18
|
Hayashi K, Ikari T, Sugiyama G, Sugiura T, Ohyama Y, Kumamaru W, Shirasuna K, Mori Y. Involvement of the T-box transcription factor Brachyury in early-stage embryonic mouse salivary gland. Biochem Biophys Res Commun 2016; 477:814-819. [PMID: 27369076 DOI: 10.1016/j.bbrc.2016.06.140] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2016] [Accepted: 06/27/2016] [Indexed: 10/21/2022]
Abstract
The mouse submandibular gland (SMG) is important organ for embryonic development, and branching morphogenesis is regulated by many molecules containing transcription factors. Real-time reverse transcriptase polymerase chain reaction revealed that the expression of Brachyury increased in the SMG and peaked between E12.5-E13.5, concomitant with the early stage of branching morphogenesis. The expression of Brachyury in SMG rudiments between E12.5-E13.5 was confirmed by western blotting. In addition, fibronectin and Btbd7 (regulated by fibronectin), which are both essential for cleft formation, were expressed strongly during the same period. The Sox2 and Wnt3a, which regulate cell growth, were also expressed strongly during E12.5-E13.5. On the other hand, cleft formation and branching morphogenesis was suppressed by knockdown of Brachyury gene, suggesting that Brachyury plays a central role in regulating cell growth and cleft formation in early-stage embryonic mouse salivary gland development.
Collapse
Affiliation(s)
- Kouhei Hayashi
- Section of Oral and Maxillofacial Surgery, Division of Maxillofacial Diagnostic and Surgical Science, Faculty of Dental Science, Kyushu University, Japan.
| | - Tatsuya Ikari
- Section of Oral and Maxillofacial Surgery, Division of Maxillofacial Diagnostic and Surgical Science, Faculty of Dental Science, Kyushu University, Japan.
| | - Goro Sugiyama
- Section of Oral and Maxillofacial Surgery, Division of Maxillofacial Diagnostic and Surgical Science, Faculty of Dental Science, Kyushu University, Japan.
| | - Tsuyoshi Sugiura
- Maxillofacial Diagnostic and Surgical Sciences, Department of Oral and Maxillofacial Rehabilitation, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima 890-8520, Japan.
| | - Yukiko Ohyama
- Section of Oral and Maxillofacial Surgery, Division of Maxillofacial Diagnostic and Surgical Science, Faculty of Dental Science, Kyushu University, Japan.
| | - Wataru Kumamaru
- Section of Oral and Maxillofacial Surgery, Division of Maxillofacial Diagnostic and Surgical Science, Faculty of Dental Science, Kyushu University, Japan.
| | - Kanemitsu Shirasuna
- Section of Oral and Maxillofacial Surgery, Division of Maxillofacial Diagnostic and Surgical Science, Faculty of Dental Science, Kyushu University, Japan.
| | - Yoshihide Mori
- Section of Oral and Maxillofacial Surgery, Division of Maxillofacial Diagnostic and Surgical Science, Faculty of Dental Science, Kyushu University, Japan.
| |
Collapse
|
19
|
Wang J, Yu T, Wang Z, Ohte S, Yao RE, Zheng Z, Geng J, Cai H, Ge Y, Li Y, Xu Y, Zhang Q, Gusella JF, Fu Q, Pregizer S, Rosen V, Shen Y. A New Subtype of Multiple Synostoses Syndrome Is Caused by a Mutation in GDF6 That Decreases Its Sensitivity to Noggin and Enhances Its Potency as a BMP Signal. J Bone Miner Res 2016; 31:882-9. [PMID: 26643732 PMCID: PMC5268166 DOI: 10.1002/jbmr.2761] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Revised: 11/19/2015] [Accepted: 12/05/2015] [Indexed: 12/23/2022]
Abstract
Growth and differentiation factors (GDFs) are secreted signaling molecules within the BMP family that have critical roles in joint morphogenesis during skeletal development in mice and humans. Using genetic data obtained from a six-generation Chinese family, we identified a missense variant in GDF6 (NP_001001557.1; p.Y444N) that fully segregates with a novel autosomal dominant synostoses (SYNS) phenotype, which we designate as SYNS4. Affected individuals display bilateral wrist and ankle deformities at birth and progressive conductive deafness after age 40 years. We find that the Y444N variant affects a highly conserved residue of GDF6 in a region critical for binding of GDF6 to its receptor(s) and to the BMP antagonist NOG, and show that this mutant GDF6 is a more potent stimulator of the canonical BMP signaling pathway compared with wild-type GDF6. Further, we determine that the enhanced BMP activity exhibited by mutant GDF6 is attributable to resistance to NOG-mediated antagonism. Collectively, our findings indicate that increased BMP signaling owing to a GDF6 gain-of-function mutation is responsible for loss of joint formation and profound functional impairment in patients with SYNS4. More broadly, our study highlights the delicate balance of BMP signaling required for proper joint morphogenesis and reinforces the critical role of BMP signaling in skeletal development.
Collapse
Affiliation(s)
- Jian Wang
- Department of Laboratory Medicine, Shanghai Children’s Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, PR China
| | - Tingting Yu
- Department of Laboratory Medicine, Shanghai Children’s Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, PR China
| | - Zhigang Wang
- Department of Pediatric Orthopedics, Shanghai Children’s Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, PR China
| | - Satoshi Ohte
- Developmental Biology, Harvard School of Dental Medicine, Boston, MA, USA
| | - Ru-en Yao
- Department of Laboratory Medicine, Shanghai Children’s Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, PR China
| | - Zhaojing Zheng
- Department of Laboratory Medicine, Shanghai Children’s Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, PR China
| | - Juan Geng
- Department of Laboratory Medicine, Shanghai Children’s Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, PR China
| | - Haiqing Cai
- Department of Pediatric Orthopedics, Shanghai Children’s Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, PR China
| | - Yihua Ge
- Department of Pediatric Orthopedics, Shanghai Children’s Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, PR China
| | - Yuchan Li
- Department of Pediatric Orthopedics, Shanghai Children’s Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, PR China
| | - Yunlan Xu
- Department of Pediatric Orthopedics, Shanghai Children’s Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, PR China
| | - Qinghua Zhang
- State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, PR China
| | - James F Gusella
- Molecular Neurogenetics Unit and Center for Human Genetic Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Qihua Fu
- Department of Laboratory Medicine, Shanghai Children’s Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, PR China
| | - Steven Pregizer
- Developmental Biology, Harvard School of Dental Medicine, Boston, MA, USA
| | - Vicki Rosen
- Developmental Biology, Harvard School of Dental Medicine, Boston, MA, USA
| | - Yiping Shen
- Department of Laboratory Medicine, Shanghai Children’s Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, PR China
- Department of Laboratory Medicine, Boston Children’s Hospital, Boston, MA, USA
- Claritas Genomics, Cambridge, MA, USA
| |
Collapse
|
20
|
Yamada A, Futagi M, Fukumoto E, Saito K, Yoshizaki K, Ishikawa M, Arakaki M, Hino R, Sugawara Y, Ishikawa M, Naruse M, Miyazaki K, Nakamura T, Fukumoto S. Connexin 43 Is Necessary for Salivary Gland Branching Morphogenesis and FGF10-induced ERK1/2 Phosphorylation. J Biol Chem 2015; 291:904-12. [PMID: 26565022 DOI: 10.1074/jbc.m115.674663] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Indexed: 11/06/2022] Open
Abstract
Cell-cell interaction via the gap junction regulates cell growth and differentiation, leading to formation of organs of appropriate size and quality. To determine the role of connexin43 in salivary gland development, we analyzed its expression in developing submandibular glands (SMGs). Connexin43 (Cx43) was found to be expressed in salivary gland epithelium. In ex vivo organ cultures of SMGs, addition of the gap junctional inhibitors 18α-glycyrrhetinic acid (18α-GA) and oleamide inhibited SMG branching morphogenesis, suggesting that gap junctional communication contributes to salivary gland development. In Cx43(-/-) salivary glands, submandibular and sublingual gland size was reduced as compared with those from heterozygotes. The expression of Pdgfa, Pdgfb, Fgf7, and Fgf10, which induced branching of SMGs in Cx43(-/-) samples, were not changed as compared with those from heterozygotes. Furthermore, the blocking peptide for the hemichannel and gap junction channel showed inhibition of terminal bud branching. FGF10 induced branching morphogenesis, while it did not rescue the Cx43(-/-) phenotype, thus Cx43 may regulate FGF10 signaling during salivary gland development. FGF10 is expressed in salivary gland mesenchyme and regulates epithelial proliferation, and was shown to induce ERK1/2 phosphorylation in salivary epithelial cells, while ERK1/2 phosphorylation in HSY cells was dramatically inhibited by 18α-GA, a Cx43 peptide or siRNA. On the other hand, PDGF-AA and PDGF-BB separately induced ERK1/2 phosphorylation in primary cultured salivary mesenchymal cells regardless of the presence of 18α-GA. Together, our results suggest that Cx43 regulates FGF10-induced ERK1/2 phosphorylation in salivary epithelium but not in mesenchyme during the process of SMG branching morphogenesis.
Collapse
Affiliation(s)
- Aya Yamada
- From the Division of Pediatric Dentistry, Department of Oral Health and Development Sciences
| | - Masaharu Futagi
- From the Division of Pediatric Dentistry, Department of Oral Health and Development Sciences
| | - Emiko Fukumoto
- From the Division of Pediatric Dentistry, Department of Oral Health and Development Sciences
| | - Kan Saito
- From the Division of Pediatric Dentistry, Department of Oral Health and Development Sciences
| | - Keigo Yoshizaki
- Division of Orthodontics, Division of Oral Health, Growth and Development, Faculty of Dental Science, Kyushu University, Fukuoka 812-8582, Japan
| | - Masaki Ishikawa
- Operative Dentistry, Department of Restorative Dentistry Tohoku University Graduate School of Dentistry, Sendai 980-8575, Japan and
| | - Makiko Arakaki
- From the Division of Pediatric Dentistry, Department of Oral Health and Development Sciences
| | - Ryoko Hino
- From the Division of Pediatric Dentistry, Department of Oral Health and Development Sciences
| | - Yu Sugawara
- From the Division of Pediatric Dentistry, Department of Oral Health and Development Sciences
| | - Momoko Ishikawa
- From the Division of Pediatric Dentistry, Department of Oral Health and Development Sciences
| | - Masahiro Naruse
- From the Division of Pediatric Dentistry, Department of Oral Health and Development Sciences
| | - Kanako Miyazaki
- Division of Orthodontics, Division of Oral Health, Growth and Development, Faculty of Dental Science, Kyushu University, Fukuoka 812-8582, Japan
| | - Takashi Nakamura
- From the Division of Pediatric Dentistry, Department of Oral Health and Development Sciences
| | - Satoshi Fukumoto
- From the Division of Pediatric Dentistry, Department of Oral Health and Development Sciences,
| |
Collapse
|
21
|
Boswell BA, Musil LS. Synergistic interaction between the fibroblast growth factor and bone morphogenetic protein signaling pathways in lens cells. Mol Biol Cell 2015; 26:2561-72. [PMID: 25947138 PMCID: PMC4571308 DOI: 10.1091/mbc.e15-02-0117] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 05/01/2015] [Indexed: 12/12/2022] Open
Abstract
Relatively little is known about how receptor tyrosine kinase ligands can positively cooperate with BMP signaling. Primary cultures of lens cells were used to reveal an unprecedented type of cross-talk between the canonical FGF and BMP signaling pathways that regulates lens cell differentiation and intercellular coupling. Fibroblast growth factors (FGFs) play a central role in two processes essential for lens transparency—fiber cell differentiation and gap junction–mediated intercellular communication (GJIC). Using serum-free primary cultures of chick lens epithelial cells (DCDMLs), we investigated how the FGF and bone morphogenetic protein (BMP) signaling pathways positively cooperate to regulate lens development and function. We found that culturing DCDMLs for 6 d with the BMP blocker noggin inhibits the canonical FGF-to-ERK pathway upstream of FRS2 activation and also prevents FGF from stimulating FRS2- and ERK-independent gene expression, indicating that BMP signaling is required at the level of FGF receptors. Other experiments revealed a second type of BMP/FGF interaction by which FGF promotes expression of BMP target genes as well as of BMP4. Together these studies reveal a novel mode of cooperation between the FGF and BMP pathways in which BMP keeps lens cells in an optimally FGF-responsive state and, reciprocally, FGF enhances BMP-mediated gene expression. This interaction provides a mechanistic explanation for why disruption of either FGF or BMP signaling in the lens leads to defects in lens development and function.
Collapse
Affiliation(s)
- Bruce A Boswell
- Department of Biochemistry and Molecular Biology, Oregon Health and Science University, Portland, OR 97239
| | - Linda S Musil
- Department of Biochemistry and Molecular Biology, Oregon Health and Science University, Portland, OR 97239 )
| |
Collapse
|
22
|
Oh HK, Lee HS, Lee JH, Oh SH, Lim JY, Ahn S, Kang SB. Coadministration of basic fibroblast growth factor-loaded polycaprolactone beads and autologous myoblasts in a dog model of fecal incontinence. Int J Colorectal Dis 2015; 30:549-57. [PMID: 25592048 DOI: 10.1007/s00384-015-2121-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/01/2015] [Indexed: 02/04/2023]
Abstract
PURPOSE Basic fibroblastic growth factor (bFGF), a member of the heparin-binding growth factor family, regulates muscle differentiation. We investigated whether coadministration of autologous myoblasts and bFGF-loaded polycaprolactone beads could improve sphincter recovery in a dog model of fecal incontinence (FI). METHODS FI was induced by resecting 25% of the posterior anal sphincter in ten mongrel dogs. One month later, the dogs were randomized to receive either PKH-26-labeled autologous myoblasts alone (M group, five dogs) or autologous myoblasts and bFGF-loaded polycaprolactone beads (MBG group, five dogs). The outcomes included anal manometry, compound muscle action potentials (CMAPs) of the pudendal nerve, and histology. RESULTS The increase in anal contractile pressure over 3 months was significantly greater in the MBG group (from 4.85 to 6.83 mmHg) than that in the M group (from 4.94 to 4.25 mmHg), with a coefficient for the difference in recovery rate of 2.672 (95% confidence interval [CI] 0.962 to 4.373, p = 0.002). The change in the CMAP amplitude was also significantly greater in the MBG group (from 0.59 to 1.56 mV) than that in the M group (from 0.81 to 0.67 mV) (coefficient 1.114, 95% CI 0.43 to 1.80, p = 0.001). Labeled cells were detected in 2/5 (40%) and 5/5 (100%) dogs in the M and MBG groups, respectively. CONCLUSION Coadministration of bFGF-loaded PCL beads and autologous myoblasts improved the recovery of sphincter function in a dog model of FI and had better outcomes than cell-based therapy alone.
Collapse
Affiliation(s)
- Heung-Kwon Oh
- Department of Surgery, Seoul National University College of Medicine, Seoul National University Bundang Hospital, 300 Gumi-dong Bundang-gu, Seongnam-si, Gyeonggi-do, 463-707, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
23
|
Handschuh K, Feenstra J, Koss M, Ferretti E, Risolino M, Zewdu R, Sahai MA, Bénazet JD, Peng XP, Depew MJ, Quintana L, Sharpe J, Wang B, Alcorn H, Rivi R, Butcher S, Manak JR, Vaccari T, Weinstein H, Anderson KV, Lacy E, Selleri L. ESCRT-II/Vps25 constrains digit number by endosome-mediated selective modulation of FGF-SHH signaling. Cell Rep 2014; 9:674-87. [PMID: 25373905 DOI: 10.1016/j.celrep.2014.09.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2013] [Revised: 08/06/2014] [Accepted: 09/09/2014] [Indexed: 10/24/2022] Open
Abstract
Sorting and degradation of receptors and associated signaling molecules maintain homeostasis of conserved signaling pathways during cell specification and tissue development. Yet, whether machineries that sort signaling proteins act preferentially on different receptors and ligands in different contexts remains mysterious. Here, we show that Vacuolar protein sorting 25, Vps25, a component of ESCRT-II (Endosomal Sorting Complex Required for Transport II), directs preferential endosome-mediated modulation of FGF signaling in limbs. By ENU-induced mutagenesis, we isolated a polydactylous mouse line carrying a hypomorphic mutation of Vps25 (Vps25(ENU)). Unlike Vps25-null embryos we generated, Vps25(ENU/ENU) mutants survive until late gestation. Their limbs display FGF signaling enhancement and consequent hyperactivation of the FGF-SHH feedback loop causing polydactyly, whereas WNT and BMP signaling remain unperturbed. Notably, Vps25(ENU/ENU) Mouse Embryonic Fibroblasts exhibit aberrant FGFR trafficking and degradation; however, SHH signaling is unperturbed. These studies establish that the ESCRT-II machinery selectively limits FGF signaling in vertebrate skeletal patterning.
Collapse
Affiliation(s)
- Karen Handschuh
- Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, NY 10065, USA
| | - Jennifer Feenstra
- Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, NY 10065, USA
| | - Matthew Koss
- Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, NY 10065, USA
| | - Elisabetta Ferretti
- Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, NY 10065, USA
| | - Maurizio Risolino
- Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, NY 10065, USA
| | - Rediet Zewdu
- Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, NY 10065, USA
| | - Michelle A Sahai
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, NY 10065, USA
| | - Jean-Denis Bénazet
- Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, NY 10065, USA
| | - Xiao P Peng
- Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, NY 10065, USA
| | - Michael J Depew
- Department of Craniofacial Development, King's College London, Guy's Hospital, London Bridge, London SE1 9RT, UK; Department of Othopaedic Surgery, UCSF, San Francisco, CA 94110, USA
| | - Laura Quintana
- Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, NY 10065, USA; Centre for Genomic Regulation (CRG), 08003 Barcelona, Spain
| | - James Sharpe
- Centre for Genomic Regulation (CRG), 08003 Barcelona, Spain; Universitat Pompeu Fabra (UPF), 08003 Barcelona, Spain; Institucio Catalana de Recerca i Estudis Avancats (ICREA), 08010 Barcelona, Spain
| | - Baolin Wang
- Department of Genetic Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Heather Alcorn
- Developmental Biology Program, Sloan-Kettering Institute, New York, NY 10065, USA
| | - Roberta Rivi
- Developmental Biology Program, Sloan-Kettering Institute, New York, NY 10065, USA
| | - Stephen Butcher
- Departments of Biology and Pediatrics, University of Iowa, Iowa City, IA 52242, USA
| | - J Robert Manak
- Departments of Biology and Pediatrics, University of Iowa, Iowa City, IA 52242, USA
| | - Thomas Vaccari
- IFOM-FIRC Institute of Molecular Oncology, 20139 Milan, Italy
| | - Harel Weinstein
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, NY 10065, USA
| | - Kathryn V Anderson
- Developmental Biology Program, Sloan-Kettering Institute, New York, NY 10065, USA
| | - Elizabeth Lacy
- Developmental Biology Program, Sloan-Kettering Institute, New York, NY 10065, USA
| | - Licia Selleri
- Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, NY 10065, USA.
| |
Collapse
|
24
|
WT1 controls antagonistic FGF and BMP-pSMAD pathways in early renal progenitors. Nat Commun 2014; 5:4444. [DOI: 10.1038/ncomms5444] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Accepted: 06/18/2014] [Indexed: 01/28/2023] Open
|
25
|
Fibroblast growth factor 4 is required but not sufficient for the astrocyte dedifferentiation. Mol Neurobiol 2014; 50:997-1012. [PMID: 24510312 DOI: 10.1007/s12035-014-8649-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2013] [Accepted: 01/20/2014] [Indexed: 01/17/2023]
Abstract
Our recent studies demonstrated that mature astrocytes from spinal cord can be reprogrammed in vitro and in vivo to generate neural stem/progenitor cells (NSPCs) following treatment with conditioned medium collected from mechanically injured astrocytes. However, little is known regarding the molecular mechanisms underlying the reprogramming of astrocytes. Here, we show that fibroblast growth factor 4 (FGF4) exerts a critical role in synergistically converting astrocytes into NSPCs that can express multiple neural stem cell markers (nestin and CD133) and are capable of both self-renewal and differentiation into neurons and glia. Lack of FGF4 signals fails to elicit the dedifferentiation of astrocytes towards NSPCs, displaying a substantially lower efficiency in the reprogramming of astrocytes and a slower transition through fate-determined state. These astrocyte-derived NSPCs displayed relatively poor self-renewal and multipotency. More importantly, further investigation suggested that FGF4 is a key molecule necessary for activating PI3K/Akt/p21 signaling cascades, as well as their downstream effectors responsible for directing cell reprogramming towards NSPCs. Collectively, these findings provide a molecular basis for astrocyte dedifferentiation into NSPCs after central nervous system (CNS) injury and imply that FGF4 may be a clinically applicable molecule for in situ neural repair in the CNS disorders.
Collapse
|
26
|
Handorf AM, Li WJ. Induction of mesenchymal stem cell chondrogenesis through sequential administration of growth factors within specific temporal windows. J Cell Physiol 2013; 229:162-71. [PMID: 23996894 DOI: 10.1002/jcp.24428] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Accepted: 07/02/2013] [Indexed: 11/12/2022]
Abstract
Human mesenchymal stem cells (hMSCs) are capable of differentiating into chondrocyte-like cells but fail to produce the quality or quantity of cartilage matrix compared to articular chondrocytes using current differentiation protocols. In this study, we aim to improve the chondrogenic differentiation of hMSCs through the sequential administration of multiple growth factors (GFs). We began by looking at differentiating hMSCs' cell surface GF receptor expression every 3 days throughout differentiation using flow cytometry and found that not only was receptor expression dynamic throughout differentiation, but ligand sensitivity was positively correlated with receptor expression, suggesting that differentiating hMSCs may have varying GF requirements depending on their stage of differentiation. We then constructed GF sequences by administering several prochondrogenic GFs singly every 3 days throughout differentiation and assaying the expression of a variety of cartilage-related genes using qPCR. The resulting chondrocytic phenotype of sequentially induced hMSCs was then compared to that of hMSCs induced under standard culture conditions using qPCR, dimethylmethylene blue assay, and histology. We found that while the initial GF sequence was unable to improve hMSC chondrogenesis, withdrawal of GF treatment at Day 9 of differentiation in pellet culture vastly improved the success of differentiation beyond that induced by TGFβ1 alone. Additional modifications allowed us to further improve chondrogenesis to levels comparable to that obtained by co-administration of TGFβ1 and BMP7 throughout differentiation. Taken together, we demonstrated the ability to improve the chondrocytic phenotype of differentiated hMSCs through the sequential administration of multiple GFs.
Collapse
Affiliation(s)
- Andrew M Handorf
- Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, Wisconsin; Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin
| | | |
Collapse
|
27
|
Yang T, Bassuk AG, Fritzsch B. Prickle1 stunts limb growth through alteration of cell polarity and gene expression. Dev Dyn 2013; 242:1293-306. [PMID: 23913870 DOI: 10.1002/dvdy.24025] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Revised: 06/25/2013] [Accepted: 07/21/2013] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND Wnt/PCP signaling plays a critical role in multiple developmental processes, including limb development. Wnt5a, a ligand of the PCP pathway, signals through the Ror2/Vangl2 or the Vangl2/Ryk complex to regulate limb development along the proximal-distal axis in mice. Based on the interaction between Van Gogh and Prickle in Drosophila, we hypothesized the vertebrate Prickle1 has a similar function as Vangl2 in limb development. RESULTS We show Prickle1 is expressed in the skeletal condensates that will differentiate into chondrocytes and later form bones. Disrupted Prickle1 function in Prickle1(C251X/C251X) mouse mutants alters expression of genes such as Bmp4, Fgf8, Vangl2, and Wnt5a. These expression changes correlate with shorter and wider bones in the limbs and loss of one phalangeal segment in digits 2-5 of Prickle1C251X mutants. These growth defects along the proximal-distal axis are also associated with increased cell death in the growing digit tip, reduced cell death in the interdigital membrane, and disrupted chondrocyte polarity. CONCLUSIONS We suggest Prickle1 is part of the Wnt5a/PCP signaling, regulating cell polarity and affecting expression of multiple factors to stunt limb growth through altered patterns of gene expression, including the PCP genes Wnt5a and Vangl2.
Collapse
Affiliation(s)
- Tian Yang
- Department of Biology, University of Iowa, Iowa City, Iowa
| | | | | |
Collapse
|
28
|
Sheth R, Grégoire D, Dumouchel A, Scotti M, Pham JMT, Nemec S, Bastida MF, Ros MA, Kmita M. Decoupling the function of Hox and Shh in developing limb reveals multiple inputs of Hox genes on limb growth. Development 2013; 140:2130-8. [PMID: 23633510 DOI: 10.1242/dev.089409] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Limb development relies on an exquisite coordination between growth and patterning, but the underlying mechanisms remain elusive. Anterior-posterior and proximal-distal specification initiates in early limb bud concomitantly with the proliferative expansion of limb cells. Previous studies have shown that limb bud growth initially relies on fibroblast growth factors (FGFs) produced in the apical ectodermal ridge (AER-FGFs), the maintenance of which relies on a positive-feedback loop involving sonic hedgehog (Shh) and the BMP antagonist gremlin 1 (Grem1). The positive cross-regulation between Shh and the HoxA and HoxD clustered genes identified an indirect effect of Hox genes on the maintenance of AER-FGFs but the respective function of Shh and Hox genes in this process remains unknown. Here, by uncoupling Hox and Shh function, we show that HoxA and HoxD genes are required for proper AER-FGFs expression, independently of their function in controlling Shh expression. In addition, we provide evidence that the Hox-dependent control of AER-FGF expression is achieved through the regulation of key mesenchymal signals, namely Grem1 and Fgf10, ensuring proper epithelial-mesenchymal interactions. Notably, HoxA and HoxD genes contribute to both the initial activation of Grem1 and the subsequent anterior expansion of its expression domain. We propose that the intricate interactions between Hox genes and the FGF and Shh signaling pathways act as a molecular network that ensures proper limb bud growth and patterning, probably contributing to the coordination of these two processes.
Collapse
Affiliation(s)
- Rushikesh Sheth
- Laboratory of Genetics and Development, Institut de Recherches Cliniques de Montréal (IRCM, Québec, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Badugu A, Kraemer C, Germann P, Menshykau D, Iber D. Digit patterning during limb development as a result of the BMP-receptor interaction. Sci Rep 2012; 2:991. [PMID: 23251777 PMCID: PMC3524521 DOI: 10.1038/srep00991] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2012] [Accepted: 11/30/2012] [Indexed: 01/07/2023] Open
Abstract
Turing models have been proposed to explain the emergence of digits during limb development. However, so far the molecular components that would give rise to Turing patterns are elusive. We have recently shown that a particular type of receptor-ligand interaction can give rise to Schnakenberg-type Turing patterns, which reproduce patterning during lung and kidney branching morphogenesis. Recent knockout experiments have identified Smad4 as a key protein in digit patterning. We show here that the BMP-receptor interaction meets the conditions for a Schnakenberg-type Turing pattern, and that the resulting model reproduces available wildtype and mutant data on the expression patterns of BMP, its receptor, and Fgfs in the apical ectodermal ridge (AER) when solved on a realistic 2D domain that we extracted from limb bud images of E11.5 mouse embryos. We propose that receptor-ligand-based mechanisms serve as a molecular basis for the emergence of Turing patterns in many developing tissues.
Collapse
Affiliation(s)
- Amarendra Badugu
- Department for Biosystems Science and Engineering (D-BSSE) , ETH Zurich, Basel, Switzerland
| | | | | | | | | |
Collapse
|
30
|
Vanuytsel T, Senger S, Fasano A, Shea-Donohue T. Major signaling pathways in intestinal stem cells. Biochim Biophys Acta Gen Subj 2012; 1830:2410-26. [PMID: 22922290 DOI: 10.1016/j.bbagen.2012.08.006] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2012] [Revised: 07/05/2012] [Accepted: 08/07/2012] [Indexed: 12/12/2022]
Abstract
BACKGROUND The discovery of markers to identify the intestinal stem cell population and the generation of powerful transgenic mouse models to study stem cell physiology have led to seminal discoveries in stem cell biology. SCOPE OF REVIEW In this review we give an overview of the current knowledge in the field of intestinal stem cells (ISCs) highlighting the most recent progress on markers defining the ISC population and pathways governing intestinal stem cell maintenance and differentiation. Furthermore we review their interaction with other stem cell related pathways. Finally we give an overview of alteration of these pathways in human inflammatory gastrointestinal diseases. MAJOR CONCLUSIONS We highlight the complex network of interactions occurring among different pathways and put in perspective the many layers of regulation that occur in maintaining the intestinal homeostasis. GENERAL SIGNIFICANCE Understanding the involvement of ISCs in inflammatory diseases can potentially lead to new therapeutic approaches to treat inflammatory GI pathologies such as IBD and celiac disease and could reveal the molecular mechanisms leading to the pathogenesis of dysplasia and cancer in inflammatory chronic conditions. This article is part of a Special Issue entitled Biochemistry of Stem Cells.
Collapse
Affiliation(s)
- Tim Vanuytsel
- Mucosal Biology Research Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | | | | | | |
Collapse
|
31
|
Barrow J. Wnt/planar cell polarity signaling: an important mechanism to coordinate growth and patterning in the limb. Organogenesis 2012; 7:260-6. [PMID: 22198433 DOI: 10.4161/org.7.4.19049] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The limb is one of the premier models for studying how a simple embryonic anlage develops into complex three-dimensional form. One of the key issues in the limb field has been to determine how the limb becomes patterned along its proximal (shoulder/hip) to distal (digits) axis. For decades it has been known that the apical ectodermal ridge (AER) plays a crucial role in distal outgrowth and patterning of the vertebrate embryonic limb. Most studies have explored the relationship between the AER and the progressive assignment of cell fates to mesenchyme along the proximal to distal (PD) axis. Comparatively few, however, have examined the additional role of the AER to regulate distal outgrowth of the limb and how this growth may also influence pattern along the PD axis. Here, I will review key studies that explore the role of growth in limb development. In particular, I will focus on a recent flurry of papers that examine the role of the Wnt/planar cell polarity (PCP) pathway in regulating directed growth of the limb mesenchyme. Finally, I will discuss a potential mechanism that relates the AER to the Wnt/PCP pathway and how directed growth can play a role in shaping the limb along the PD axis.
Collapse
Affiliation(s)
- Jeffery Barrow
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, UT, USA.
| |
Collapse
|
32
|
Sun Y, Hu J, Zhou L, Pollard SM, Smith A. Interplay between FGF2 and BMP controls the self-renewal, dormancy and differentiation of rat neural stem cells. J Cell Sci 2011; 124:1867-77. [PMID: 21558414 PMCID: PMC3096055 DOI: 10.1242/jcs.085506] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/27/2011] [Indexed: 12/29/2022] Open
Abstract
Mouse and human central nervous system progenitor cells can be propagated extensively ex vivo as stem cell lines. For the rat, however, in vitro expansion has proven to be problematic owing to proliferation arrest and differentiation. Here, we analyse the establishment, in adherent culture, of undifferentiated tripotent neural stem (NS) cell lines derived from rat foetal brain and spinal cord. Rat NS cells invariably undergo growth arrest and apparent differentiation after several passages; however, conditioned medium from proliferating cultures can overcome this block, enabling continuous propagation of undifferentiated rat NS cells. We found that dormancy is induced by autocrine production of bone morphogenetic proteins (BMPs). Accordingly, the BMP antagonist noggin can replace conditioned medium to sustain continuous self-renewal. Noggin can also induce dormant cells to re-enter the cell cycle, upon which they reacquire neurogenic potential. We further show that fibroblast growth factor 2 (FGF2) is required to suppress terminal astrocytic differentiation and maintain stem cell potency during dormancy. These findings highlight an extrinsic regulatory network, comprising BMPs, BMP antagonists and FGF2 signals, that governs the proliferation, dormancy and differentiation of rat NS cells and which can be manipulated to enable long-term clonogenic self-renewal.
Collapse
Affiliation(s)
- Yirui Sun
- Wellcome Trust Centre for Stem Cell Research and Department of Biochemistry, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK
- Department of Neurosurgery, Shanghai Huashan Hospital, Fu Dan University, Shanghai 200040, People's Republic of China
- Shanghai No. 6 Hospital, Jiaotong University, Shanghai 200233, People's Republic of China
| | - Jin Hu
- Department of Neurosurgery, Shanghai Huashan Hospital, Fu Dan University, Shanghai 200040, People's Republic of China
| | - Liangfu Zhou
- Department of Neurosurgery, Shanghai Huashan Hospital, Fu Dan University, Shanghai 200040, People's Republic of China
| | - Steven M. Pollard
- Wellcome Trust Centre for Stem Cell Research and Department of Biochemistry, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK
| | - Austin Smith
- Wellcome Trust Centre for Stem Cell Research and Department of Biochemistry, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK
| |
Collapse
|
33
|
Holocephalan embryos provide evidence for gill arch appendage reduction and opercular evolution in cartilaginous fishes. Proc Natl Acad Sci U S A 2011; 108:1507-12. [PMID: 21220324 DOI: 10.1073/pnas.1012968108] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Chondrichthyans possess endoskeletal appendages called branchial rays that extend laterally from their hyoid and gill-bearing (branchial) arches. Branchial ray outgrowth, like tetrapod limb outgrowth, is maintained by Sonic hedgehog (Shh) signaling. In limbs, distal endoskeletal elements fail to form in the absence of normal Shh signaling, whereas shortened duration of Shh expression correlates with distal endoskeletal reduction in naturally variable populations. Chondrichthyans also exhibit natural variation with respect to branchial ray distribution--elasmobranchs (sharks and batoids) possess a series of ray-supported septa on their hyoid and gill arches, whereas holocephalans (chimaeras) possess a single hyoid arch ray-supported operculum. Here we show that the elongate hyoid rays of the holocephalan Callorhinchus milii grow in association with sustained Shh expression within an opercular epithelial fold, whereas Shh is only transiently expressed in the gill arches. Coincident with this transient Shh expression, branchial ray outgrowth is initiated in C. milii but is not maintained, yielding previously unrecognized vestigial gill arch branchial rays. This is in contrast to the condition seen in sharks, where sustained Shh expression corresponds to the presence of fully formed branchial rays on the hyoid and gill arches. Considered in light of current hypotheses of chondrichthyan phylogeny, our data suggest that the holocephalan operculum evolved in concert with gill arch appendage reduction by attenuation of Shh-mediated branchial ray outgrowth, and that chondrichthyan branchial rays and tetrapod limbs exhibit parallel developmental mechanisms of evolutionary reduction.
Collapse
|
34
|
Abstract
Fibroblast growth factor (FGF) signalling has been implicated during several phases of early embryogenesis, including the patterning of the embryonic axes, the induction and/or maintenance of several cell lineages and the coordination of morphogenetic movements. Here, we summarise our current understanding of the regulation and roles of FGF signalling during early vertebrate development.
Collapse
Affiliation(s)
- Karel Dorey
- The Healing Foundation Centre, Michael Smith Building, Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester, M13 9PT, UK
| | - Enrique Amaya
- The Healing Foundation Centre, Michael Smith Building, Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester, M13 9PT, UK
| |
Collapse
|
35
|
Estrogens attenuate oxidative stress and the differentiation and apoptosis of osteoblasts by DNA-binding-independent actions of the ERalpha. J Bone Miner Res 2010; 25:769-81. [PMID: 19821774 PMCID: PMC3153331 DOI: 10.1359/jbmr.091017] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Estrogens diminish oxidative stress in bone and bone marrow, attenuate the generation of osteoblasts, and decrease the prevalence of mature osteoblast apoptosis. We have searched for the molecular mechanism of these effects using as tools a mouse model bearing an estrogen receptor alpha (ERalpha) knock-in mutation that prevents binding to DNA (ERalpha(NERKI/-)) and several osteoblast progenitor cell models expressing the wild-type ERalpha or the ERalpha(NERKI/-). We report that the ability of estrogens to diminish the generation of reactive oxygen species, stimulate the activity of glutathione reductase, and decrease the phosphorylation of p66(shc), as well as osteoblastogenesis and osteoblast number and apoptosis, were fully preserved in ERalpha(NERKI/-) mice, indicating that the DNA-binding function of the ERalpha is dispensable for all these effects. Consistent with the attenuation of osteoblastogenesis in this animal model, 17beta-estradiol attenuated bone morphogenetic protein 2 (BMP-2)-induced gene transcription and osteoblast commitment and differentiation in murine and human osteoblastic cell lines. Moreover, 17beta-estradiol attenuated BMP-2-induced differentiation of primary cultures of calvaria- or bone marrow-derived osteoblastic cells from ERalpha(NERKI/-) mice as effectively as in cells from wild-type littermates. The inhibitory effect of the hormone on BMP-2 signaling resulted from an ERalpha-mediated activation of ERKs and the phosphorylation of Smad1 at the linker region of the protein, which leads to proteasomal degradation. These results illustrate that the effects of estrogens on oxidative stress and the birth and death of osteoblasts do not require the binding of ERalpha to DNA response elements, but instead they result from the activation of cytoplasmic kinases.
Collapse
|
36
|
Guimond JC, Lévesque M, Michaud PL, Berdugo J, Finnson K, Philip A, Roy S. BMP-2 functions independently of SHH signaling and triggers cell condensation and apoptosis in regenerating axolotl limbs. BMC DEVELOPMENTAL BIOLOGY 2010; 10:15. [PMID: 20152028 PMCID: PMC2829471 DOI: 10.1186/1471-213x-10-15] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2009] [Accepted: 02/12/2010] [Indexed: 11/20/2022]
Abstract
Background Axolotls have the unique ability, among vertebrates, to perfectly regenerate complex body parts, such as limbs, after amputation. In addition, axolotls pattern developing and regenerating autopods from the anterior to posterior axis instead of posterior to anterior like all tetrapods studied to date. Sonic hedgehog is important in establishing this anterior-posterior axis of limbs in all tetrapods including axolotls. Interestingly, its expression is conserved (to the posterior side of limb buds and blastemas) in axolotl limbs as in other tetrapods. It has been suggested that BMP-2 may be the secondary mediator of sonic hedgehog, although there is mounting evidence to the contrary in mice. Since BMP-2 expression is on the anterior portion of developing and regenerating limbs prior to digit patterning, opposite to the expression of sonic hedgehog, we examined whether BMP-2 expression was dependent on sonic hedgehog signaling and whether it affects patterning of the autopod during regeneration. Results The expression of BMP-2 and SOX-9 in developing and regenerating axolotl limbs corresponded to the first digits forming in the anterior portion of the autopods. The inhibition of sonic hedgehog signaling with cyclopamine caused hypomorphic limbs (during development and regeneration) but did not affect the expression of BMP-2 and SOX-9. Overexpression of BMP-2 in regenerating limbs caused a loss of digits. Overexpression of Noggin (BMP inhibitor) in regenerating limbs also resulted in a loss of digits. Histological analysis indicated that the loss due to BMP-2 overexpression was the result of increased cell condensation and apoptosis while the loss caused by Noggin was due to a decrease in cell division. Conclusion The expression of BMP-2 and its target SOX-9 was independent of sonic hedgehog signaling in developing and regenerating limbs. Their expression correlated with chondrogenesis and the appearance of skeletal elements has described in other tetrapods. Overexpression of BMP-2 did not cause the formation of extra digits, which is consistent with the hypothesis that it is not the secondary signal of sonic hedgehog. However, it did cause the formation of hypomorphic limbs as a result of increased cellular condensation and apoptosis. Taken together, these results suggest that BMP-2 does not have a direct role in patterning regenerating limbs but may be important to trigger condensation prior to ossification and to mediate apoptosis.
Collapse
Affiliation(s)
- Jean-Charles Guimond
- Department of Stomatology, Faculty of Dentistry, Université de Montréal, Montreal (QC) Canada.
| | | | | | | | | | | | | |
Collapse
|
37
|
Butterfield NC, McGlinn E, Wicking C. The molecular regulation of vertebrate limb patterning. Curr Top Dev Biol 2010; 90:319-41. [PMID: 20691854 DOI: 10.1016/s0070-2153(10)90009-4] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The limb has long been considered a paradigm for organogenesis because of its simplicity and ease of manipulation. However, it has become increasingly clear that the processes required to produce a perfectly formed limb involve complex molecular interactions across all three axes of limb development. Old models have evolved with acquisition of molecular knowledge, and in more recent times mathematical modeling approaches have been invoked to explain the precise spatio-temporal regulation of gene networks that coordinate limb patterning and outgrowth. This review focuses on recent advances in our understanding of vertebrate limb development, highlighting the signaling interactions required to lay down the pattern on which the processes of differentiation will act to ultimately produce the final limb.
Collapse
Affiliation(s)
- Natalie C Butterfield
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | | | | |
Collapse
|
38
|
Cartilage engineering from mesenchymal stem cells. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2010; 123:163-200. [PMID: 20535603 DOI: 10.1007/10_2010_67] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Mesenchymal progenitor cells known as multipotent mesenchymal stromal cells or mesenchymal stem cells (MSC) have been isolated from various tissues. Since they are able to differentiate along the mesenchymal lineages of cartilage and bone, they are regarded as promising sources for the treatment of skeletal defects. Tissue regeneration in the adult organism and in vitro engineering of tissues is hypothesized to follow the principles of embryogenesis. The embryonic development of the skeleton has been studied extensively with respect to the regulatory mechanisms governing morphogenesis, differentiation, and tissue formation. Various concepts have been designed for engineering tissues in vitro based on these developmental principles, most of them involving regulatory molecules such as growth factors or cytokines known to be the key regulators in developmental processes. Growth factors most commonly used for in vitro cultivation of cartilage tissue belong to the fibroblast growth factor (FGF) family, the transforming growth factor-beta (TGF-β) super-family, and the insulin-like growth factor (IGF) family. In this chapter, in vivo actions of members of these growth factors described in the literature are compared with in vitro concepts of cartilage engineering making use of these growth factors.
Collapse
|
39
|
Kosaka N, Sakamoto H, Terada M, Ochiya T. Pleiotropic function of FGF-4: its role in development and stem cells. Dev Dyn 2009; 238:265-76. [PMID: 18792115 DOI: 10.1002/dvdy.21699] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Fibroblast growth factors (FGFs) were initially recognized as fibroblast-specific growth factor, and it is now apparent that these growth factors regulate multiple biological functions. The diversity of FGFs function is paralleled by the emerging diversity of interactions between FGF ligands and their receptors. FGF-4 is a member of the FGF superfamily and is a mitogen exhibiting strong action on numerous different cell types. It plays a role in various stages of development and morphogenesis, as well as in a variety of biological processes. Recent studies reveal the molecular mechanisms of FGF-4 gene regulation in mammalian cells, which is involved in the developmental process. Furthermore, FGF-4 also acts on the regulation of proliferation and differentiation in embryonic stem cells and tissue stem cells. In this review, we focus on the diverse biological functions of FGF-4 in the developmental process and also discuss its putative roles in stem cell biology.
Collapse
Affiliation(s)
- Nobuyoshi Kosaka
- Section for Studies on Metastasis, National Cancer Center Research Institute, Chuo-ku, Tokyo, Japan
| | | | | | | |
Collapse
|
40
|
Satoh A, James MA, Gardiner DM. The role of nerve signaling in limb genesis and agenesis during axolotl limb regeneration. J Bone Joint Surg Am 2009; 91 Suppl 4:90-8. [PMID: 19571075 DOI: 10.2106/jbjs.i.00159] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Affiliation(s)
- Akira Satoh
- Okayama University, RCIS, Okayama-city, Okayama 700-8530, Japan
| | | | | |
Collapse
|
41
|
Abstract
Congenital hand differences affect approximately 1 in 500 persons and present unique diagnostic and classification challenges. The embryology of the hand and upper extremity is discussed, including the molecular basis for patterning of the upper limb. The Swanson/International Federation of Societies for Surgery of the Hand classification system is described, along with a discussion of the difficulties in classifying congenital hand differences based on morphology or etiology.
Collapse
Affiliation(s)
- Douglas M Sammer
- Department of Surgery, Washington University, St. Louis, MO 63110, USA
| | | |
Collapse
|
42
|
Satoh A, Bryant SV, Gardiner DM. Regulation of dermal fibroblast dedifferentiation and redifferentiation during wound healing and limb regeneration in the Axolotl. Dev Growth Differ 2008; 50:743-54. [DOI: 10.1111/j.1440-169x.2008.01072.x] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
43
|
Achieving bilateral symmetry during vertebrate limb development. Semin Cell Dev Biol 2008; 20:479-84. [PMID: 19027866 DOI: 10.1016/j.semcdb.2008.10.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2008] [Accepted: 10/24/2008] [Indexed: 02/06/2023]
Abstract
While the various internal organs of vertebrates display many obvious left-right asymmetries in their location and/or morphology, external features exhibit a high degree of bilateral symmetry. How this external bilateral symmetry is established during development is largely unknown. In this review, we explore several mechanisms, in place during development, that regulate the final size of the limb. These mechanisms rely on the presence of positive signaling feedback loops during limb bud growth. Through the activity of these signaling loops and their eventual breakdown when the limb bud has reached a certain size, bilateral symmetry can be achieved.
Collapse
|
44
|
In vitro whole-organ imaging: 4D quantification of growing mouse limb buds. Nat Methods 2008; 5:609-12. [DOI: 10.1038/nmeth.1219] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2007] [Accepted: 05/13/2008] [Indexed: 11/08/2022]
|
45
|
Yu K, Ornitz DM. FGF signaling regulates mesenchymal differentiation and skeletal patterning along the limb bud proximodistal axis. Development 2008; 135:483-91. [DOI: 10.1242/dev.013268] [Citation(s) in RCA: 103] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Fibroblast growth factors (FGFs) are signals from the apical ectodermal ridge (AER) that are essential for limb pattern formation along the proximodistal (PD) axis. However, how patterning along the PD axis is regulated by AER-FGF signals remains controversial. To further explore the molecular mechanism of FGF functions during limb development, we conditionally inactivated fgf receptor 2 (Fgfr2) in the mouse AER to terminate all AER functions; for comparison, we inactivated both Fgfr1 and Fgfr2 in limb mesenchyme to block mesenchymal AER-FGF signaling. We also re-examined published data in which Fgf4 and Fgf8 were inactivated in the AER. We conclude that limb skeletal phenotypes resulting from loss of AER-FGF signals cannot simply be a consequence of excessive mesenchymal cell death, as suggested by previous studies, but also must be a consequence of reduced mesenchymal proliferation and a failure of mesenchymal differentiation, which occur following loss of both Fgf4 and Fgf8. We further conclude that chondrogenic primordia formation,marked by initial Sox9 expression in limb mesenchyme, is an essential component of the PD patterning process and that a key role for AER-FGF signaling is to facilitate SOX9 function and to ensure progressive establishment of chondrogenic primordia along the PD axis.
Collapse
Affiliation(s)
- Kai Yu
- Department of Developmental Biology, 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
|
46
|
Mayshar Y, Rom E, Chumakov I, Kronman A, Yayon A, Benvenisty N. Fibroblast growth factor 4 and its novel splice isoform have opposing effects on the maintenance of human embryonic stem cell self-renewal. Stem Cells 2008; 26:767-74. [PMID: 18192227 DOI: 10.1634/stemcells.2007-1037] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Human embryonic stem cells (HESCs) are unique in their capacity to self-renew while remaining pluripotent. This undifferentiated state must be actively maintained by secreted factors. To identify autocrine factors that may support HESC growth, we have taken a global genetic approach. Microarray analysis identified fibroblast growth factor 4 (FGF4) as a prime candidate for autocrine signaling. Furthermore, the addition of recombinant FGF4 to HESCs supports their proliferation. We show that FGF4 is produced by multiple undifferentiated HESC lines, along with a novel fibroblast growth factor 4 splice isoform (FGF4si) that codes for the amino-terminal half of FGF4. Strikingly, although FGF4 supports the undifferentiated growth of HESCs, FGF4si effectively counters its effect. Furthermore, we show that FGF4si is an antagonist of FGF4, shutting down FGF4-induced Erk1/2 phosphorylation. Expression analysis shows that both isoforms are expressed in HESCs and early differentiated cells. However, whereas FGF4 ceases to be expressed in mature differentiated cells, FGF4si continues to be expressed after cell differentiation. Targeted knockdown of FGF4 using small interfering RNA increased differentiation of HESCs, demonstrating the importance of endogenous FGF4 signaling in maintaining their pluripotency. Taken together, these results suggest a growth-promoting role for FGF4 in HESCs and a putative feedback inhibition mechanism by a novel FGF4 splice isoform that may serve to promote differentiation at later stages of development.
Collapse
Affiliation(s)
- Yoav Mayshar
- Department of Genetics, The Life Sciences Institute, The Hebrew University, Jerusalem 91904, Israel
| | | | | | | | | | | |
Collapse
|
47
|
Reza HM, Takahashi Y, Yasuda K. Stage-dependent expression of Pax6 in optic vesicle/cup regulates patterning genes through signaling molecules. Differentiation 2007; 75:726-36. [PMID: 17381541 DOI: 10.1111/j.1432-0436.2007.00168.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Dorso-ventral and proximo-distal axis formation of the optic cup is apparent from early stages of development. Pax6 is initially detectable in the optic vesicle and later shows a distal-high and proximal-low gradient of expression in the retina. To determine the early role of Pax6 in pattern formation of the optic cup, we expressed Pax6 ectopically in the optic vesicle of stages 9-10 chick embryos by in ovo electroporation, which resulted in a small eye-like phenotype. The signaling molecule fibroblast growth factor (FGF)8, which appears to be restricted to the central retina, was increased, whereas bone morphogenetic protein (BMP)4 and Tbx5, two dorsal markers, were down-regulated in Pax6-electroporated eye. Pax6 overexpression also decreased the expression of the ventral marker Vax. Electroporation with a dominant-negative form of Pax6 resulted in a decrease in FGF8 expression, but BMP4 expression was unaffected initially while it was diminished later. Our data suggest a new role for Pax6 in regulating FGF8 and BMP4 expression during pattern formation of the optic cup, and that a Pax6-regulated balance between FGF8 and BMP4 is critical for retinogenesis.
Collapse
Affiliation(s)
- Hasan Mahmud Reza
- Graduate School of Biological Sciences, Nara Institute of Science and Technology, 8916-5 Takayama, Ikoma 630-0101, Japan
| | | | | |
Collapse
|
48
|
Abstract
Bone morphogenetic proteins (BMPs) are multifunctional growth factors belonging to the transforming growth factor beta (TGFbeta) multigene family. Current evidence indicates that they may play different and even antagonistic roles at different stages of limb development. Refined studies of their function in these processes have been impeded in the mouse due to the early lethality of null mutants for several BMP ligands and their receptors. Recently, however, these questions have benefited from the very powerful Cre-loxP technology. In this review, I intend to summarize what has been learned from this conditional mutagenesis approach in the mouse limb, focusing on Bmp2, Bmp4 and Bmp7 while restricting my analysis to the initial phases of limb formation and patterning. Two major aspects are discussed, the role of BMPs in dorsal-ventral polarization of the limb bud, together with their relation to apical ectodermal ridge (AER) induction, and their role in controlling digit number and identity. Particular attention is paid to the methodology, its power and its limits.
Collapse
Affiliation(s)
- Benoît Robert
- Institut Pasteur, Unité de Recherche Associée 2578 Centre National de la Recherche Scientifique, 25 Rue du Dr Roux, 75724 Paris Cedex 15, France.
| |
Collapse
|
49
|
Freitas R, Zhang G, Cohn MJ. Biphasic Hoxd gene expression in shark paired fins reveals an ancient origin of the distal limb domain. PLoS One 2007; 2:e754. [PMID: 17710153 PMCID: PMC1937022 DOI: 10.1371/journal.pone.0000754] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2007] [Accepted: 07/24/2007] [Indexed: 01/10/2023] Open
Abstract
The evolutionary transition of fins to limbs involved development of a new suite of distal skeletal structures, the digits. During tetrapod limb development, genes at the 5' end of the HoxD cluster are expressed in two spatiotemporally distinct phases. In the first phase, Hoxd9-13 are activated sequentially and form nested domains along the anteroposterior axis of the limb. This initial phase patterns the limb from its proximal limit to the middle of the forearm. Later in development, a second wave of transcription results in 5' HoxD gene expression along the distal end of the limb bud, which regulates formation of digits. Studies of zebrafish fins showed that the second phase of Hox expression does not occur, leading to the idea that the origin of digits was driven by addition of the distal Hox expression domain in the earliest tetrapods. Here we test this hypothesis by investigating Hoxd gene expression during paired fin development in the shark Scyliorhinus canicula, a member of the most basal lineage of jawed vertebrates. We report that at early stages, 5'Hoxd genes are expressed in anteroposteriorly nested patterns, consistent with the initial wave of Hoxd transcription in teleost and tetrapod paired appendages. Unexpectedly, a second phase of expression occurs at later stages of shark fin development, in which Hoxd12 and Hoxd13 are re-expressed along the distal margin of the fin buds. This second phase is similar to that observed in tetrapod limbs. The results indicate that a second, distal phase of Hoxd gene expression is not uniquely associated with tetrapod digit development, but is more likely a plesiomorphic condition present the common ancestor of chondrichthyans and osteichthyans. We propose that a temporal extension, rather than de novo activation, of Hoxd expression in the distal part of the fin may have led to the evolution of digits.
Collapse
Affiliation(s)
- Renata Freitas
- Department of Zoology, Cancer/Genetics Research Complex, University of Florida, Gainesville, Florida, United Sates of America
| | - GuangJun Zhang
- Department of Zoology, Cancer/Genetics Research Complex, University of Florida, Gainesville, Florida, United Sates of America
| | - Martin J. Cohn
- Department of Zoology, Cancer/Genetics Research Complex, University of Florida, Gainesville, Florida, United Sates of America
- Department of Anatomy and Cell Biology, Cancer/Genetics Research Complex, University of Florida, Gainesville, Florida, United States of America
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|
50
|
Pajni-Underwood S, Wilson CP, Elder C, Mishina Y, Lewandoski M. BMP signals control limb bud interdigital programmed cell death by regulating FGF signaling. Development 2007; 134:2359-68. [PMID: 17537800 DOI: 10.1242/dev.001677] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
In vertebrate limbs that lack webbing, the embryonic interdigit region is removed by programmed cell death (PCD). Established models suggest that bone morphogenetic proteins (BMPs) directly trigger such PCD, although no direct genetic evidence exists for this. Alternatively, BMPs might indirectly affect PCD by regulating fibroblast growth factors (FGFs), which act as cell survival factors. Here, we inactivated the mouse BMP receptor gene Bmpr1a specifically in the limb bud apical ectodermal ridge (AER), a source of FGF activity. Early inactivation completely prevents AER formation. However, inactivation after limb bud initiation causes an upregulation of two AER-FGFs, Fgf4 and Fgf8, and a loss of interdigital PCD leading to webbed limbs. To determine whether excess FGF signaling inhibits interdigit PCD in these Bmpr1a mutant limbs, we performed double and triple AER-specific inactivations of Bmpr1a, Fgf4 and Fgf8. Webbing persists in AER-specific inactivations of Bmpr1a and Fgf8 owing to elevated Fgf4 expression. Inactivation of Bmpr1a, Fgf8 and one copy of Fgf4 eliminates webbing. We conclude that during normal embryogenesis, BMP signaling to the AER indirectly regulates interdigit PCD by regulating AER-FGFs, which act as survival factors for the interdigit mesenchyme.
Collapse
Affiliation(s)
- Sangeeta Pajni-Underwood
- Laboratory of Cancer and Developmental Biology National Institutes of Health, Frederick, MD 21702, USA
| | | | | | | | | |
Collapse
|