1
|
Oikonomou P, Calvary L, Cirne HC, Welch AE, Durel JF, Powell O, Kim K, Nerurkar NL. Application of tissue-scale tension to avian epithelia in vivo to study multiscale mechanics and inter-germ layer coupling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.04.588089. [PMID: 38617324 PMCID: PMC11014599 DOI: 10.1101/2024.04.04.588089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
As cross-disciplinary approaches drawing from physics and mechanics have increasingly influenced our understanding of morphogenesis, the tools available to measure and perturb physical aspects of embryonic development have expanded as well. However, it remains a challenge to measure mechanical properties and apply exogenous tissue-scale forces in vivo, particularly for epithelia. Exploiting the size and accessibility of the developing chick embryo, here we describe a simple technique to quantitatively apply exogenous forces on the order of ~1-100 μN to the endodermal epithelium. To demonstrate the utility of this approach, we performed a series of proof-of-concept experiments that reveal fundamental and unexpected mechanical behaviors in the early chick embryo, including mechanotype heterogeneity among cells of the midgut endoderm, complex non-cell autonomous effects of actin disruption, and a high degree of mechanical coupling between the endoderm and adjacent paraxial mesoderm. To illustrate the broader utility of this method, we determined that forces on the order of ~ 10 μN are sufficient to unzip the neural tube during primary neurulation. Together, these findings provide basic insights into the mechanics of embryonic epithelia in vivo in the early avian embryo, and provide a useful tool for future investigations of how morphogenesis is influenced by mechanical factors.
Collapse
Affiliation(s)
| | | | - Helena C. Cirne
- Department of Biomedical Engineering, Columbia University, New York NY 10027
| | - Andreas E. Welch
- Department of Biomedical Engineering, Columbia University, New York NY 10027
| | - John F. Durel
- Department of Biomedical Engineering, Columbia University, New York NY 10027
| | - Olivia Powell
- Department of Biomedical Engineering, Columbia University, New York NY 10027
| | - Kwantae Kim
- Department of Biomedical Engineering, Columbia University, New York NY 10027
| | - Nandan L. Nerurkar
- Department of Biomedical Engineering, Columbia University, New York NY 10027
| |
Collapse
|
2
|
Wopat S, Adhyapok P, Daga B, Crawford JM, Norman J, Bagwell J, Peskin B, Magre I, Fogerson SM, Levic DS, Di Talia S, Kiehart DP, Charbonneau P, Bagnat M. Notochord segmentation in zebrafish controlled by iterative mechanical signaling. Dev Cell 2024; 59:1860-1875.e5. [PMID: 38697108 PMCID: PMC11265980 DOI: 10.1016/j.devcel.2024.04.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 01/25/2024] [Accepted: 04/12/2024] [Indexed: 05/04/2024]
Abstract
In bony fishes, patterning of the vertebral column, or spine, is guided by a metameric blueprint established in the notochord sheath. Notochord segmentation begins days after somitogenesis concludes and can occur in its absence. However, somite patterning defects lead to imprecise notochord segmentation, suggesting that these processes are linked. Here, we identify that interactions between the notochord and the axial musculature ensure precise spatiotemporal segmentation of the zebrafish spine. We demonstrate that myoseptum-notochord linkages drive notochord segment initiation by locally deforming the notochord extracellular matrix and recruiting focal adhesion machinery at these contact points. Irregular somite patterning alters this mechanical signaling, causing non-sequential and dysmorphic notochord segmentation, leading to altered spine development. Using a model that captures myoseptum-notochord interactions, we find that a fixed spatial interval is critical for driving sequential segment initiation. Thus, mechanical coupling of axial tissues facilitates spatiotemporal spine patterning.
Collapse
Affiliation(s)
- Susan Wopat
- Department of Cell Biology, Duke University, Durham, NC 27710, USA
| | - Priyom Adhyapok
- Department of Cell Biology, Duke University, Durham, NC 27710, USA
| | - Bijoy Daga
- Department of Cell Biology, Duke University, Durham, NC 27710, USA
| | | | - James Norman
- Department of Cell Biology, Duke University, Durham, NC 27710, USA
| | - Jennifer Bagwell
- Department of Cell Biology, Duke University, Durham, NC 27710, USA
| | - Brianna Peskin
- Department of Cell Biology, Duke University, Durham, NC 27710, USA
| | - Indrasen Magre
- Department of Cell Biology, Duke University, Durham, NC 27710, USA
| | | | - Daniel S Levic
- Department of Cell Biology, Duke University, Durham, NC 27710, USA
| | - Stefano Di Talia
- Department of Cell Biology, Duke University, Durham, NC 27710, USA
| | | | - Patrick Charbonneau
- Department of Chemistry, Duke University, Durham, NC 27708, USA; Department of Physics, Duke University, Durham, NC 27708, USA.
| | - Michel Bagnat
- Department of Cell Biology, Duke University, Durham, NC 27710, USA.
| |
Collapse
|
3
|
Sung CY, Kadiyala U, Blanchard O, Yourston L, Walker D, Li L, Fu J, Yang Q. Substrate Rigidity Modulates Segmentation Clock Dynamics in Isolated Presomitic Mesoderm Cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.02.601712. [PMID: 39005461 PMCID: PMC11244955 DOI: 10.1101/2024.07.02.601712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
The segmentation clock, a genetic oscillator in the presomitic mesoderm (PSM), is known to be influenced by biochemical signals, yet its potential regulation by mechanical cues remains unclear. The complex PSM microenvironment has made it challenging to isolate the effects of mechanical perturbations on clock behavior. Here we investigated how mechanical stimuli affect clock oscillations by culturing zebrafish PSM cells on PDMS micropost arrays with tunable rigidities (0.6-1200 kPa). We observed an inverse sigmoidal relationship between surface rigidity and both the percentage of oscillating cells and the number of oscillation cycles, with a switching threshold between 3-6 kPa. The periods of oscillating cells showed a consistently broad distribution across rigidity changes. Moreover, these cells exhibited distinct biophysical properties, such as reduced motility, contractility, and sustained circularity. These findings highlight the crucial role of cell-substrate interactions in regulating segmentation clock behavior, providing insights into the mechanobiology of somitogenesis.
Collapse
Affiliation(s)
- Chun-Yen Sung
- Department of Biophysics, University of Michigan, Ann Arbor, MI 48109
| | - Usha Kadiyala
- Department of Biophysics, University of Michigan, Ann Arbor, MI 48109
| | - Owen Blanchard
- Department of Biophysics, University of Michigan, Ann Arbor, MI 48109
| | - Liam Yourston
- Department of Biophysics, University of Michigan, Ann Arbor, MI 48109
| | - Derek Walker
- Department of Physics, University of Michigan, Ann Arbor, MI 48109
| | - Linyuan Li
- Department of Biophysics, University of Michigan, Ann Arbor, MI 48109
| | - Jianping Fu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, 48109
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109
- Department of Cell & Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, 48109
| | - Qiong Yang
- Department of Biophysics, University of Michigan, Ann Arbor, MI 48109
- Department of Physics, University of Michigan, Ann Arbor, MI 48109
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109
- Department of Cell & Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, 48109
| |
Collapse
|
4
|
Alila-Fersi O, Tej A, Maalej M, Kharrat M, Boughamoura L, Chouchen J, Tlili A, Fakhfakh F. Mitochondrial genes modulate the phenotypic expression of congenital scoliosis syndrome caused by mutations in the TBXT gene. Gene 2024; 914:148388. [PMID: 38499212 DOI: 10.1016/j.gene.2024.148388] [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: 01/28/2024] [Revised: 02/29/2024] [Accepted: 03/15/2024] [Indexed: 03/20/2024]
Abstract
BACKGROUND Congenital scoliosis (CS) is a spinal disorder caused by genetic-congenital vertebral malformations and may be associated with other congenital defects or may occur alone. It is genetically heterogeneous and numerous genes contributing to this disease have been identified. In addition, CS has a wide range of phenotypic and genotypic variability, which has been explained by the intervention of genetic factors like modifiers and environment genes. The aim of the present study was to determine the possible cause of CS in a Tunisian patient and to examine the association between mtDNA mutations and mtDNA content and CS. METHODS Here we performed Whole-Exome Sequencing (WES) in a patient presenting clinical features suggestive of severe congenital scoliosis syndrome. Direct sequencing of the whole mitochondrial DNA (mtDNA) was also performed in addition to copy number quantification in the blood of the indexed case. In silico prediction tools, 3D modeling and molecular docking approaches were used. RESULTS The WES revealed the homozygous missense mutation c.512A > G (p.H171R) in the TBXT gene. Bioinformatic analysis demonstrated that the p.H171R variant was highly deleterious and caused the TBXT structure instability. Molecular docking revealed that the p.H171R mutation disrupted the monomer stability which seemed to be crucial for maintaining the stability of the homodimer and consequently to the destabilization of the homodimer-DNA complex. On the other hand, we hypothesized that mtDNA can be a modifier factor, so, the screening of the whole mtDNA showed a novel heteroplasmic m.10150T > A (p.M31K) variation in the MT-ND3 gene. Further, qPCR analyses of the patient's blood excluded mtDNA depletion. Bioinformatic investigation revealed that the p.M31K mutation in the ND3 protein was highly deleterious and may cause the ND3 protein structure destabilization and could disturb the interaction between complex I subunits. CONCLUSION We described the possible role of mtDNA genetics on the pathogenesis of congenital scoliosis by hypothesizing that the presence of the homozygous variant in TBXT accounts for the CS phenotype in our patient and the MT-ND3 gene may act as a modifier gene.
Collapse
Affiliation(s)
- Olfa Alila-Fersi
- Molecular Genetics and Functional Laboratory, Faculty of Science of Sfax, University of Sfax, Sfax 3000, Tunisia.
| | - Amel Tej
- Department of Pediatrics, University Hospital Farhat Hached, Sousse, Tunisia
| | - Marwa Maalej
- Molecular Genetics and Functional Laboratory, Faculty of Science of Sfax, University of Sfax, Sfax 3000, Tunisia
| | - Marwa Kharrat
- Molecular Genetics and Functional Laboratory, Faculty of Science of Sfax, University of Sfax, Sfax 3000, Tunisia
| | - Lamia Boughamoura
- Department of Pediatrics, University Hospital Farhat Hached, Sousse, Tunisia
| | - Jihen Chouchen
- Molecular Genetics and Stem Cell Research Laboratory, University of Sharjah, Sharjah, United Arab Emirates
| | - Abdelaziz Tlili
- Human Genetics and Stem Cell Research Group, Research Institute of Sciences and Engineering, University of Sharjah, Sharjah, United Arab Emirates
| | - Faiza Fakhfakh
- Molecular Genetics and Functional Laboratory, Faculty of Science of Sfax, University of Sfax, Sfax 3000, Tunisia.
| |
Collapse
|
5
|
Feng X, Ye Y, Zhang J, Zhang Y, Zhao S, Mak JCW, Otomo N, Zhao Z, Niu Y, Yonezawa Y, Li G, Lin M, Li X, Cheung PWH, Xu K, Takeda K, Wang S, Xie J, Kotani T, Choi VNT, Song YQ, Yang Y, Luk KDK, Lee KS, Li Z, Li PS, Leung CYH, Lin X, Wang X, Qiu G, Watanabe K, Wu Z, Posey JE, Ikegawa S, Lupski JR, Cheung JPY, Zhang TJ, Gao B, Wu N. Core planar cell polarity genes VANGL1 and VANGL2 in predisposition to congenital vertebral malformations. Proc Natl Acad Sci U S A 2024; 121:e2310283121. [PMID: 38669183 PMCID: PMC11067467 DOI: 10.1073/pnas.2310283121] [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: 06/20/2023] [Accepted: 03/11/2024] [Indexed: 04/28/2024] Open
Abstract
Congenital scoliosis (CS), affecting approximately 0.5 to 1 in 1,000 live births, is commonly caused by congenital vertebral malformations (CVMs) arising from aberrant somitogenesis or somite differentiation. While Wnt/ß-catenin signaling has been implicated in somite development, the function of Wnt/planar cell polarity (Wnt/PCP) signaling in this process remains unclear. Here, we investigated the role of Vangl1 and Vangl2 in vertebral development and found that their deletion causes vertebral anomalies resembling human CVMs. Analysis of exome sequencing data from multiethnic CS patients revealed a number of rare and deleterious variants in VANGL1 and VANGL2, many of which exhibited loss-of-function and dominant-negative effects. Zebrafish models confirmed the pathogenicity of these variants. Furthermore, we found that Vangl1 knock-in (p.R258H) mice exhibited vertebral malformations in a Vangl gene dose- and environment-dependent manner. Our findings highlight critical roles for PCP signaling in vertebral development and predisposition to CVMs in CS patients, providing insights into the molecular mechanisms underlying this disorder.
Collapse
Affiliation(s)
- Xin Feng
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
- Department of Orthopedic Surgery, State Key Laboratory of Complex Severe and Rare Diseases, all at Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing100730, China
- Key laboratory of big data for spinal deformities, Chinese Academy of Medical Sciences, Beijing100730, China
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Yongyu Ye
- Department of Orthopedic Surgery, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou510080, China
| | - Jianan Zhang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
- Department of Orthopaedics and Traumatology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Yuanqiang Zhang
- Department of Orthopaedic Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan250012, China
| | - Sen Zhao
- Department of Orthopedic Surgery, State Key Laboratory of Complex Severe and Rare Diseases, all at Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing100730, China
- Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing100730, China
| | - Judith C. W. Mak
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Nao Otomo
- Department of Orthopedic Surgery, Keio University School of Medicine, Tokyo160-8582, Japan
- Laboratory of Bone and Joint Diseases, RIKEN Center for Integrative Medical Sciences, Tokyo108-8639, Japan
| | - Zhengye Zhao
- Department of Orthopedic Surgery, State Key Laboratory of Complex Severe and Rare Diseases, all at Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing100730, China
- Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing100730, China
| | - Yuchen Niu
- Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing100730, China
- Department of Medical Research Center, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing100730, China
| | - Yoshiro Yonezawa
- Department of Orthopedic Surgery, Keio University School of Medicine, Tokyo160-8582, Japan
- Laboratory of Bone and Joint Diseases, RIKEN Center for Integrative Medical Sciences, Tokyo108-8639, Japan
| | - Guozhuang Li
- Department of Orthopedic Surgery, State Key Laboratory of Complex Severe and Rare Diseases, all at Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing100730, China
- Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing100730, China
| | - Mao Lin
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou310003, China
| | - Xiaoxin Li
- Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing100730, China
- Department of Medical Research Center, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing100730, China
| | - Prudence Wing Hang Cheung
- Department of Orthopaedics and Traumatology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Kexin Xu
- Department of Orthopedic Surgery, State Key Laboratory of Complex Severe and Rare Diseases, all at Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing100730, China
- Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing100730, China
| | - Kazuki Takeda
- Department of Orthopedic Surgery, Keio University School of Medicine, Tokyo160-8582, Japan
- Laboratory of Bone and Joint Diseases, RIKEN Center for Integrative Medical Sciences, Tokyo108-8639, Japan
| | - Shengru Wang
- Department of Orthopedic Surgery, State Key Laboratory of Complex Severe and Rare Diseases, all at Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing100730, China
| | - Junjie Xie
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Toshiaki Kotani
- Department of Orthopedic Surgery, Keio University School of Medicine, Tokyo160-8582, Japan
| | - Vanessa N. T. Choi
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - You-Qiang Song
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
- Department of Medicine, The University of Hong Kong-Shenzhen Hospital, Shenzhen518009, China
| | - Yang Yang
- Department of Orthopedic Surgery, State Key Laboratory of Complex Severe and Rare Diseases, all at Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing100730, China
- Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing100730, China
| | - Keith Dip Kei Luk
- Department of Orthopaedics and Traumatology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Kin Shing Lee
- Center for Comparative Medicine Research, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Ziquan Li
- Department of Orthopedic Surgery, State Key Laboratory of Complex Severe and Rare Diseases, all at Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing100730, China
- Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing100730, China
| | - Pik Shan Li
- Center for Comparative Medicine Research, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Connie Y. H. Leung
- Center for Comparative Medicine Research, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Xiaochen Lin
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Xiaolu Wang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
- Department of Orthopaedics and Traumatology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Guixing Qiu
- Department of Orthopedic Surgery, State Key Laboratory of Complex Severe and Rare Diseases, all at Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing100730, China
- Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing100730, China
| | | | - Kota Watanabe
- Department of Orthopedic Surgery, Keio University School of Medicine, Tokyo160-8582, Japan
| | | | - Zhihong Wu
- Department of Medical Research Center, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing100730, China
| | - Jennifer E. Posey
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston77030, TX
| | - Shiro Ikegawa
- Laboratory of Bone and Joint Diseases, RIKEN Center for Integrative Medical Sciences, Tokyo108-8639, Japan
| | - James R. Lupski
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston77030, TX
- Human Genome Sequencing Center, Baylor College of Medicine, Houston77030, TX
- Texas Children’s Hospital, Houston77030, TX
- Department of Pediatrics, Baylor College of Medicine, Houston77030, TX
| | - Jason Pui Yin Cheung
- Department of Orthopaedics and Traumatology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
- Department of Orthopedics and Traumatology, The University of Hong Kong-Shenzhen Hospital, Shenzhen518009, China
| | - Terry Jianguo Zhang
- Department of Orthopedic Surgery, State Key Laboratory of Complex Severe and Rare Diseases, all at Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing100730, China
- Key laboratory of big data for spinal deformities, Chinese Academy of Medical Sciences, Beijing100730, China
- Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing100730, China
| | - Bo Gao
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
- Department of Orthopedics and Traumatology, The University of Hong Kong-Shenzhen Hospital, Shenzhen518009, China
- Centre for Translational Stem Cell Biology, Hong Kong Special Administrative Region, China
- Key Laboratory of Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Nan Wu
- Department of Orthopedic Surgery, State Key Laboratory of Complex Severe and Rare Diseases, all at Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing100730, China
- Key laboratory of big data for spinal deformities, Chinese Academy of Medical Sciences, Beijing100730, China
- Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing100730, China
| |
Collapse
|
6
|
McMillen P, Levin M. Collective intelligence: A unifying concept for integrating biology across scales and substrates. Commun Biol 2024; 7:378. [PMID: 38548821 PMCID: PMC10978875 DOI: 10.1038/s42003-024-06037-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 03/11/2024] [Indexed: 04/01/2024] Open
Abstract
A defining feature of biology is the use of a multiscale architecture, ranging from molecular networks to cells, tissues, organs, whole bodies, and swarms. Crucially however, biology is not only nested structurally, but also functionally: each level is able to solve problems in distinct problem spaces, such as physiological, morphological, and behavioral state space. Percolating adaptive functionality from one level of competent subunits to a higher functional level of organization requires collective dynamics: multiple components must work together to achieve specific outcomes. Here we overview a number of biological examples at different scales which highlight the ability of cellular material to make decisions that implement cooperation toward specific homeodynamic endpoints, and implement collective intelligence by solving problems at the cell, tissue, and whole-organism levels. We explore the hypothesis that collective intelligence is not only the province of groups of animals, and that an important symmetry exists between the behavioral science of swarms and the competencies of cells and other biological systems at different scales. We then briefly outline the implications of this approach, and the possible impact of tools from the field of diverse intelligence for regenerative medicine and synthetic bioengineering.
Collapse
Affiliation(s)
- Patrick McMillen
- Department of Biology, Tufts University, Medford, MA, 02155, USA
- Allen Discovery Center at Tufts University, Medford, MA, 02155, USA
| | - Michael Levin
- Department of Biology, Tufts University, Medford, MA, 02155, USA.
- Allen Discovery Center at Tufts University, Medford, MA, 02155, USA.
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA.
| |
Collapse
|
7
|
Pappas MP, Kawakami H, Corcoran D, Chen KQ, Scott EP, Wong J, Gearhart MD, Nishinakamura R, Nakagawa Y, Kawakami Y. Sall4 regulates posterior trunk mesoderm development by promoting mesodermal gene expression and repressing neural genes in the mesoderm. Development 2024; 151:dev202649. [PMID: 38345319 PMCID: PMC10946440 DOI: 10.1242/dev.202649] [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: 12/28/2023] [Accepted: 01/30/2024] [Indexed: 03/05/2024]
Abstract
The trunk axial skeleton develops from paraxial mesoderm cells. Our recent study demonstrated that conditional knockout of the stem cell factor Sall4 in mice by TCre caused tail truncation and a disorganized axial skeleton posterior to the lumbar level. Based on this phenotype, we hypothesized that, in addition to the previously reported role of Sall4 in neuromesodermal progenitors, Sall4 is involved in the development of the paraxial mesoderm tissue. Analysis of gene expression and SALL4 binding suggests that Sall4 directly or indirectly regulates genes involved in presomitic mesoderm differentiation, somite formation and somite differentiation. Furthermore, ATAC-seq in TCre; Sall4 mutant posterior trunk mesoderm shows that Sall4 knockout reduces chromatin accessibility. We found that Sall4-dependent open chromatin status drives activation and repression of WNT signaling activators and repressors, respectively, to promote WNT signaling. Moreover, footprinting analysis of ATAC-seq data suggests that Sall4-dependent chromatin accessibility facilitates CTCF binding, which contributes to the repression of neural genes within the mesoderm. This study unveils multiple mechanisms by which Sall4 regulates paraxial mesoderm development by directing activation of mesodermal genes and repression of neural genes.
Collapse
Affiliation(s)
- Matthew P. Pappas
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN 55455, USA
| | - Hiroko Kawakami
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN 55455, USA
- Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA
- Developmental Biology Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - Dylan Corcoran
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN 55455, USA
| | - Katherine Q. Chen
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN 55455, USA
| | - Earl Parker Scott
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Julia Wong
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN 55455, USA
| | - Micah D. Gearhart
- Developmental Biology Center, University of Minnesota, Minneapolis, MN 55455, USA
- Department of Obstetrics, Gynecology and Women's Health, University of Minnesota, Minneapolis, MN 55455, USA
| | - Ryuichi Nishinakamura
- Department of Kidney Development, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811, Japan
| | - Yasushi Nakagawa
- Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA
- Developmental Biology Center, University of Minnesota, Minneapolis, MN 55455, USA
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Yasuhiko Kawakami
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN 55455, USA
- Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA
- Developmental Biology Center, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
8
|
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
|
9
|
Ramesh PS, Chu LF. Species-specific roles of the Notch ligands, receptors, and targets orchestrating the signaling landscape of the segmentation clock. Front Cell Dev Biol 2024; 11:1327227. [PMID: 38348091 PMCID: PMC10859470 DOI: 10.3389/fcell.2023.1327227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 12/20/2023] [Indexed: 02/15/2024] Open
Abstract
Somitogenesis is a hallmark feature of all vertebrates and some invertebrate species that involves the periodic formation of block-like structures called somites. Somites are transient embryonic segments that eventually establish the entire vertebral column. A highly conserved molecular oscillator called the segmentation clock underlies this periodic event and the pace of this clock regulates the pace of somite formation. Although conserved signaling pathways govern the clock in most vertebrates, the mechanisms underlying the species-specific divergence in various clock characteristics remain elusive. For example, the segmentation clock in classical model species such as zebrafish, chick, and mouse embryos tick with a periodicity of ∼30, ∼90, and ∼120 min respectively. This enables them to form the species-specific number of vertebrae during their overall timespan of somitogenesis. Here, we perform a systematic review of the species-specific features of the segmentation clock with a keen focus on mouse embryos. We perform this review using three different perspectives: Notch-responsive clock genes, ligand-receptor dynamics, and synchronization between neighboring oscillators. We further review reports that use non-classical model organisms and in vitro model systems that complement our current understanding of the segmentation clock. Our review highlights the importance of comparative developmental biology to further our understanding of this essential developmental process.
Collapse
Affiliation(s)
- Pranav S. Ramesh
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
- Reproductive Biology and Regenerative Medicine Research Group, University of Calgary, Calgary, AB, Canada
- Alberta Children’s Hospital Research Institute, Calgary, AB, Canada
| | - Li-Fang Chu
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
- Reproductive Biology and Regenerative Medicine Research Group, University of Calgary, Calgary, AB, Canada
- Alberta Children’s Hospital Research Institute, Calgary, AB, Canada
| |
Collapse
|
10
|
Shapiro F, Wang J, Flynn E, Wu JY. Pudgy mouse rib deformities emanate from abnormal paravertebral longitudinal cartilage/bone accumulations. Biol Open 2024; 13:bio060139. [PMID: 38252118 PMCID: PMC10840853 DOI: 10.1242/bio.060139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 11/03/2023] [Indexed: 01/23/2024] Open
Abstract
The pudgy (pu/pu) mouse, caused by a recessive mutation in the Notch family Delta like-3 gene (Dll3), has severe rib, vertebral body and intervertebral disc abnormalities. Using whole-mount preparations and serial histologic sections we demonstrate: 1) localized paravertebral longitudinal cartilage/bone accumulations (PVLC/BAs) invariably associated with branched, fused and asymmetrically spaced ribs that emanate from it laterally; 2) abnormal rib formation immediately adjacent to abnormal vertebral body and intervertebral disc formation in asymmetric right/left fashion; and 3) patterns of rib deformation that differ in each mouse. Normal BALB/c embryo and age-matched non-affected pu/+ mice assessments allow for pu/pu comparisons. The Dll3 Notch family gene is involved in normal somitogenesis via the segmentation clock mechanism. Although pathogenesis of rib deformation is initially triggered by the Dll3 gene mutation, these findings of abnormal asymmetric costo-vertebral region structure imply that differing patterns cannot be attributed to this single gene mutation alone. All findings implicate a dual mechanism of malformation: the Dll3 gene mutation leading to subtle timing differences in traveling oscillation waves of the segmentation clock and further subsequent misdirection of tissue formation by altered chemical reaction-diffusion and epigenetic landscape responses. PVLC/BAs appear as primary supramolecular structures underlying severe rib malformation associated both with time-sensitive segmentation clock mutations and subsequent reactions.
Collapse
Affiliation(s)
- Frederic Shapiro
- Department of Medicine/Endocrinology, Stanford University School of Medicine, Palo Alto CA 94305, USA
- Department of Bioengineering, Northeastern University, Boston MA 02115, USA
| | - Jamie Wang
- Department of Medicine/Endocrinology, Stanford University School of Medicine, Palo Alto CA 94305, USA
| | - Evelyn Flynn
- Orthopaedic Research Laboratory, Boston Children's Hospital, Boston MA 02115, USA
| | - Joy Y. Wu
- Department of Medicine/Endocrinology, Stanford University School of Medicine, Palo Alto CA 94305, USA
| |
Collapse
|
11
|
Marc S, Savici J, Sicoe B, Boldura OM, Paul C, Otavă G. Exencephaly-Anencephaly Sequence Associated with Maxillary Brachygnathia, Spinal Defects, and Palatoschisis in a Male Domestic Cat. Animals (Basel) 2023; 13:3882. [PMID: 38136919 PMCID: PMC10741185 DOI: 10.3390/ani13243882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 12/08/2023] [Accepted: 12/15/2023] [Indexed: 12/24/2023] Open
Abstract
Anencephaly, a severe neural tube defect characterized by the absence of major parts of the brain and skull, is a rare congenital disorder that has been observed in various species, including cats. Considering the uncommon appearance of anencephaly, this paper aims to present anencephaly in a stillborn male kitten from an accidental inbreeding using various paraclinical methods. Histological examination of tissue samples from the cranial region, where parts of the skull were absent, revealed the presence of atypical nerve tissue with neurons and glial cells organized in clusters, surrounded by an extracellular matrix and with an abundance of blood vessels, which are large, dilated, and filled with blood, not characteristic of nerve tissue structure. In CT scans, the caudal part of the frontal bone, the fronto-temporal limits, and the parietal bone were observed to be missing. CT also revealed that the dorsal tubercle of the atlas, the dorsal neural arch, and the spinal process of the C2-C7 bones were missing. In conclusion, the kitten was affected by multiple congenital malformations, a combination of exencephaly-anencephaly, maxillary brachygnathism, closed cranial spina bifida at the level of cervical vertebrae, kyphoscoliosis, palatoschisis, and partial intestinal atresia. The importance of employing imaging techniques cannot be overstated when it comes to the accurate diagnosis of neural tube defects.
Collapse
Affiliation(s)
- Simona Marc
- Faculty of Veterinary Medicine, University of Life Sciences “King Mihai I” from Timisoara, Calea Aradului 119, 300645 Timisoara, Romania; (S.M.); (J.S.); (B.S.); (O.M.B.); (G.O.)
| | - Jelena Savici
- Faculty of Veterinary Medicine, University of Life Sciences “King Mihai I” from Timisoara, Calea Aradului 119, 300645 Timisoara, Romania; (S.M.); (J.S.); (B.S.); (O.M.B.); (G.O.)
| | - Bogdan Sicoe
- Faculty of Veterinary Medicine, University of Life Sciences “King Mihai I” from Timisoara, Calea Aradului 119, 300645 Timisoara, Romania; (S.M.); (J.S.); (B.S.); (O.M.B.); (G.O.)
| | - Oana Maria Boldura
- Faculty of Veterinary Medicine, University of Life Sciences “King Mihai I” from Timisoara, Calea Aradului 119, 300645 Timisoara, Romania; (S.M.); (J.S.); (B.S.); (O.M.B.); (G.O.)
| | - Cristina Paul
- Department of Applied Chemistry and Engineering of Organic and Natural Compounds, Faculty of Industrial Chemistry and Environmental Engineering, Politehnica University Timisoara, Carol Telbisz 6, 300001 Timisoara, Romania
| | - Gabriel Otavă
- Faculty of Veterinary Medicine, University of Life Sciences “King Mihai I” from Timisoara, Calea Aradului 119, 300645 Timisoara, Romania; (S.M.); (J.S.); (B.S.); (O.M.B.); (G.O.)
| |
Collapse
|
12
|
Onai T, Adachi N, Urakubo H, Sugahara F, Aramaki T, Matsumoto M, Ohno N. Ultrastructure of the lamprey head mesoderm reveals evolution of the vertebrate head. iScience 2023; 26:108338. [PMID: 38187188 PMCID: PMC10767164 DOI: 10.1016/j.isci.2023.108338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 09/20/2023] [Accepted: 10/23/2023] [Indexed: 01/09/2024] Open
Abstract
The cranial muscle is a critical component in the vertebrate head for a predatory lifestyle. However, its evolutionary origin and possible segmental nature during embryogenesis have been controversial. In jawed vertebrates, the presence of pre-otic segments similar to trunk somites has been claimed based on developmental observations. However, evaluating such arguments has been hampered by the paucity of research on jawless vertebrates. Here, we discovered different cellular arrangements in the head mesoderm in lamprey embryos (Lethenteron camtschaticum) using serial block-face scanning electron and laser scanning microscopies. These cell populations were morphologically and molecularly different from somites. Furthermore, genetic comparison among deuterostomes revealed that mesodermal gene expression domains were segregated antero-posteriorly in vertebrates, whereas such segregation was not recognized in invertebrate deuterostome embryos. These findings indicate that the vertebrate head mesoderm evolved from the anteroposterior repatterning of an ancient mesoderm and developmentally diversified before the split of jawless and jawed vertebrates.
Collapse
Affiliation(s)
- Takayuki Onai
- Department of Anatomy, University of Fukui, School of Medical Sciences, 23-3, Matsuokashimoaizuki, Eiheiji, Yoshida, Fukui, Japan
- Life Science Innovation Center, University of Fukui, 23-3, Matsuokashimoaizuki, Eiheiji, Yoshida, Fukui, Japan
| | - Noritaka Adachi
- Aix-Marseille Université, IBDM, CNRS UMR 7288, Campus De Luminy Case 907, 13288 Marseille Cedex 9, France
| | - Hidetoshi Urakubo
- Section of Electron Microscopy, National Institute for Physiological Sciences, 38, Nishigonaka, Myodaiji, Okazaki, Aichi, Japan
| | - Fumiaki Sugahara
- Division of Biology, Hyogo Medical University, 1-1, Mukogawa, Nishinomiya, Hyogo, Japan
| | - Toshihiro Aramaki
- Graduate School of Frontier Biosciences, Osaka University, 1-1, Yamadaoka, Suita, Osaka, Japan
| | - Mami Matsumoto
- Section of Electron Microscopy, Supportive Center for Brain Research, National Institute for Physiological Sciences, 38, Nishigonaka, Myodaiji, Okazaki, Aichi, Japan
- Department of Developmental and Regenerative Biology, Nagoya City University Graduate School of Medical Sciences, 1, Kawasumi, Mizuho, Nagoya, Aichi, Japan
| | - Nobuhiko Ohno
- Department of Anatomy, Division of Histology and Cell Biology, School of Medicine, Jichi Medical University, 3311-1, Yakushiji, Shimotsuke, Tochigi, Japan
- Division of Ultrastructural Research, National Institute for Physiological Sciences, 38, Nishigonaka, Myodaiji, Okazaki, Aichi, Japan
| |
Collapse
|
13
|
Martini A, Sahd L, Rücklin M, Huysseune A, Hall BK, Boglione C, Witten PE. Deformity or variation? Phenotypic diversity in the zebrafish vertebral column. J Anat 2023; 243:960-981. [PMID: 37424444 PMCID: PMC10641053 DOI: 10.1111/joa.13926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 06/14/2023] [Accepted: 06/23/2023] [Indexed: 07/11/2023] Open
Abstract
Vertebral bodies are composed of two types of metameric elements, centra and arches, each of which is considered as a developmental module. Most parts of the teleost vertebral column have a one-to-one relationship between centra and arches, although, in all teleosts, this one-to-one relationship is lost in the caudal fin endoskeleton. Deviation from the one-to-one relationship occurs in most vertebrates, related to changes in the number of vertebral centra or to a change in the number of arches. In zebrafish, deviations also occur predominantly in the caudal region of the vertebral column. In-depth phenotypic analysis of wild-type zebrafish was performed using whole-mount stained samples, histological analyses and synchrotron radiation X-ray tomographic microscopy 3D reconstructions. Three deviant centra phenotypes were observed: (i) fusion of two vertebral centra, (ii) wedge-shaped hemivertebrae and (iii) centra with reduced length. Neural and haemal arches and their spines displayed bilateral and unilateral variations that resemble vertebral column phenotypes of stem-ward actinopterygians or other gnathostomes as well as pathological conditions in extant species. Whether it is possible to distinguish variations from pathological alterations and whether alterations resemble ancestral conditions is discussed in the context of centra and arch variations in other vertebrate groups and basal actinopterygian species.
Collapse
Affiliation(s)
- Arianna Martini
- Laboratory of Experimental Ecology and Aquaculture, Department of Biology, University of Rome Tor Vergata, Rome, Italy
- Research Group Evolutionary Developmental Biology, Biology Department, Ghent University, Ghent, Belgium
- PhD Program in Evolutionary Biology and Ecology, Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Lauren Sahd
- Research Group Evolutionary Developmental Biology, Biology Department, Ghent University, Ghent, Belgium
| | - Martin Rücklin
- Department of Vertebrate Evolution, Development and Ecology, Naturalis Biodiversity Center, Leiden, The Netherlands
| | - Ann Huysseune
- Research Group Evolutionary Developmental Biology, Biology Department, Ghent University, Ghent, Belgium
- Department of Zoology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Brian K Hall
- Department of Biology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Clara Boglione
- Laboratory of Experimental Ecology and Aquaculture, Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - P Eckhard Witten
- Research Group Evolutionary Developmental Biology, Biology Department, Ghent University, Ghent, Belgium
| |
Collapse
|
14
|
Abstract
Organismal development requires the reproducible unfolding of an ordered sequence of discrete steps (cell fate determination, migration, tissue folding, etc.) in both time and space. Here, we review the mechanisms that grant temporal specificity to developmental steps, including molecular clocks and timers. Individual timing mechanisms must be coordinated with each other to maintain the overall developmental sequence. However, phenotypic novelties can also arise through the modification of temporal patterns over the course of evolution. Two main types of variation in temporal patterning characterize interspecies differences in developmental time: allochrony, where the overall developmental sequence is either accelerated or slowed down while maintaining the relative duration of individual steps, and heterochrony, where the duration of specific developmental steps is altered relative to the rest. New advances in in vitro modeling of mammalian development using stem cells have recently enabled the revival of mechanistic studies of allochrony and heterochrony. In both cases, differences in the rate of basic cellular functions such as splicing, translation, protein degradation, and metabolism seem to underlie differences in developmental time. In the coming years, these studies should identify the genetic differences that drive divergence in developmental time between species.
Collapse
Affiliation(s)
- Margarete Diaz-Cuadros
- Department of Molecular Biology, Massachusetts General Hospital, Boston, Massachusetts, USA;
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, USA;
| | - Olivier Pourquié
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, USA;
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, USA
| |
Collapse
|
15
|
Ye Y, Zhang J, Feng X, Chen C, Chang Y, Qiu G, Wu Z, Zhang TJ, Gao B, Wu N. Exploring the association between congenital vertebral malformations and neural tube defects. J Med Genet 2023; 60:1146-1152. [PMID: 37775263 DOI: 10.1136/jmg-2023-109501] [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: 07/08/2023] [Accepted: 09/07/2023] [Indexed: 10/01/2023]
Abstract
Congenital vertebral malformations (CVMs) and neural tube defects (NTDs) are common birth defects affecting the spine and nervous system, respectively, due to defects in somitogenesis and neurulation. Somitogenesis and neurulation rely on factors secreted from neighbouring tissues and the integrity of the axial structure. Crucial signalling pathways like Wnt, Notch and planar cell polarity regulate somitogenesis and neurulation with significant crosstalk. While previous studies suggest an association between CVMs and NTDs, the exact mechanism underlying this relationship remains unclear. In this review, we explore embryonic development, signalling pathways and clinical phenotypes involved in the association between CVMs and NTDs. Moreover, we provide a summary of syndromes that exhibit occurrences of both CVMs and NTDs. We aim to provide insights into the potential mechanisms underlying the association between CVMs and NTDs, thereby facilitating clinical diagnosis and management of these anomalies.
Collapse
Affiliation(s)
- Yongyu Ye
- Department of Orthopedic Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Jianan Zhang
- Department of Orthopaedics and Traumatology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Xin Feng
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Chong Chen
- Department of Orthopedic Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Yunbing Chang
- Department of Orthopedic Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Guixing Qiu
- Department of Orthopedic Surgery, Key Laboratory of Big Data for Spinal Deformities, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, China
| | - Zhihong Wu
- Department of Orthopedic Surgery, Key Laboratory of Big Data for Spinal Deformities, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, China
| | - Terry Jianguo Zhang
- Department of Orthopedic Surgery, Key Laboratory of Big Data for Spinal Deformities, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, China
| | - Bo Gao
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
- Centre for Translational Stem Cell Biology, Hong Kong, China
| | - Nan Wu
- Department of Orthopedic Surgery, Key Laboratory of Big Data for Spinal Deformities, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, China
| |
Collapse
|
16
|
Kinney B, Sahu S, Stec N, Hills-Muckey K, Adams DW, Wang J, Jaremko M, Joshua-Tor L, Keil W, Hammell CM. A circadian-like gene network programs the timing and dosage of heterochronic miRNA transcription during C. elegans development. Dev Cell 2023; 58:2563-2579.e8. [PMID: 37643611 PMCID: PMC10840721 DOI: 10.1016/j.devcel.2023.08.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 07/10/2023] [Accepted: 08/02/2023] [Indexed: 08/31/2023]
Abstract
Development relies on the exquisite control of both the timing and the levels of gene expression to achieve robust developmental transitions. How cis- and trans-acting factors control both aspects simultaneously is unclear. We show that transcriptional pulses of the temporal patterning microRNA (miRNA) lin-4 are generated by two nuclear hormone receptors (NHRs) in C. elegans, NHR-85 and NHR-23, whose mammalian orthologs, Rev-Erb and ROR, function in the circadian clock. Although Rev-Erb and ROR antagonize each other to control once-daily transcription in mammals, NHR-85/NHR-23 heterodimers bind cooperatively to lin-4 regulatory elements to induce a single pulse of expression during each larval stage. Each pulse's timing, amplitude, and duration are dictated by the phased expression of these NHRs and the C. elegans Period ortholog, LIN-42, that binds to and represses NHR-85. Therefore, during nematode temporal patterning, an evolutionary rewiring of circadian clock components couples the timing of gene expression to the control of transcriptional dosage.
Collapse
Affiliation(s)
- Brian Kinney
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Shubham Sahu
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR168 Laboratoire Physico Chimie Curie, Paris 75005, France
| | - Natalia Stec
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | | | - Dexter W Adams
- Howard Hughes Medical Institute, W. M. Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA; Graduate Program in Genetics, Stony Brook University, Stony Brook, NY 11794, USA
| | - Jing Wang
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Matt Jaremko
- Howard Hughes Medical Institute, W. M. Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Leemor Joshua-Tor
- Howard Hughes Medical Institute, W. M. Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Wolfgang Keil
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR168 Laboratoire Physico Chimie Curie, Paris 75005, France.
| | | |
Collapse
|
17
|
Simsek MF, Özbudak EM. A design logic for sequential segmentation across organisms. FEBS J 2023; 290:5086-5093. [PMID: 37422856 PMCID: PMC10774455 DOI: 10.1111/febs.16899] [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: 04/19/2023] [Revised: 05/24/2023] [Accepted: 07/06/2023] [Indexed: 07/11/2023]
Abstract
Multitudes of organisms display metameric compartmentalization of their body plan. Segmentation of these compartments happens sequentially in diverse phyla. In several sequentially segmenting species, periodically active molecular clocks and signaling gradients have been found. The clocks are proposed to control the timing of segmentation, while the gradients are proposed to instruct the positions of segment boundaries. However, the identity of the clock and gradient molecules differs across species. Furthermore, sequential segmentation of a basal chordate, Amphioxus, continues at late stages when the small tail bud cell population cannot establish long-range signaling gradients. Thus, it remains to be explained how a conserved morphological trait (i.e., sequential segmentation) is achieved by using different molecules or molecules with different spatial profiles. Here, we first focus on sequential segmentation of somites in vertebrate embryos and then draw parallels with other species. Thereafter, we propose a candidate design principle that has the potential to answer this puzzling question.
Collapse
Affiliation(s)
- M Fethullah Simsek
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, OH, USA
| | - Ertuğrul M Özbudak
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, OH, USA
| |
Collapse
|
18
|
Rebello D, Wohler E, Erfani V, Li G, Aguilera AN, Santiago-Cornier A, Zhao S, Hwang SW, Steiner RD, Zhang TJ, Gurnett CA, Raggio C, Wu N, Sobreira N, Giampietro PF, Ciruna B. COL11A2 as a candidate gene for vertebral malformations and congenital scoliosis. Hum Mol Genet 2023; 32:2913-2928. [PMID: 37462524 PMCID: PMC10508038 DOI: 10.1093/hmg/ddad117] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 05/08/2023] [Accepted: 07/13/2023] [Indexed: 07/23/2023] Open
Abstract
Human vertebral malformations (VMs) have an estimated incidence of 1/2000 and are associated with significant health problems including congenital scoliosis (CS) and recurrent organ system malformation syndromes such as VACTERL (vertebral anomalies; anal abnormalities; cardiac abnormalities; tracheo-esophageal fistula; renal anomalies; limb anomalies). The genetic cause for the vast majority of VMs are unknown. In a CS/VM patient cohort, three COL11A2 variants (R130W, R1407L and R1413H) were identified in two patients with cervical VM. A third patient with a T9 hemivertebra and the R130W variant was identified from a separate study. These substitutions are predicted to be damaging to protein function, and R130 and R1407 residues are conserved in zebrafish Col11a2. To determine the role for COL11A2 in vertebral development, CRISPR/Cas9 was used to create a nonsense mutation (col11a2L642*) as well as a full gene locus deletion (col11a2del) in zebrafish. Both col11a2L642*/L642* and col11a2del/del mutant zebrafish exhibit vertebral fusions in the caudal spine, which form due to mineralization across intervertebral segments. To determine the functional consequence of VM-associated variants, we assayed their ability to suppress col11a2del VM phenotypes following transgenic expression within the developing spine. While wildtype col11a2 expression suppresses fusions in col11a2del/+ and col11a2del/del backgrounds, patient missense variant-bearing col11a2 failed to rescue the loss-of-function phenotype in these animals. These results highlight an essential role for COL11A2 in vertebral development and support a pathogenic role for two missense variants in CS.
Collapse
Affiliation(s)
- Denise Rebello
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada
- Department of Molecular Genetics, The University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Elizabeth Wohler
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Vida Erfani
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada
- Department of Molecular Genetics, The University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Guozhuang Li
- Department of Orthopedic Surgery, Key Laboratory of Big Data for Spinal Deformities, Beijing Key Laboratory for Genetic Research of Skeletal Deformity, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Alexya N Aguilera
- Department of Pediatrics, University of Illinois-Chicago, Chicago, IL 60612, USA
| | - Alberto Santiago-Cornier
- Genetic Section, San Jorge Children’s and Women’s Hospital, San Juan, Puerto Rico 00912, USA
- Department of Public Health, Ponce Health Sciences University, Ponce, Puerto Rico 00912, USA
| | - Sen Zhao
- Department of Orthopedic Surgery, Key Laboratory of Big Data for Spinal Deformities, Beijing Key Laboratory for Genetic Research of Skeletal Deformity, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Steven W Hwang
- Shriners Children’s-Philadelphia, Philadelphia, PA 19140, USA
| | - Robert D Steiner
- Department of Pediatrics, University of Wisconsin, Madison, WI 54449, USA
- Marshfield Clinic Health System, Marshfield, WI 54449, USA
| | - Terry Jianguo Zhang
- Department of Orthopedic Surgery, Key Laboratory of Big Data for Spinal Deformities, Beijing Key Laboratory for Genetic Research of Skeletal Deformity, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Christina A Gurnett
- Department of Neurology, Washington University in St. Louis, St. Louis, MO 63110, USA
| | | | - Nan Wu
- Department of Orthopedic Surgery, Key Laboratory of Big Data for Spinal Deformities, Beijing Key Laboratory for Genetic Research of Skeletal Deformity, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Nara Sobreira
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Philip F Giampietro
- Department of Pediatrics, University of Illinois-Chicago, Chicago, IL 60612, USA
| | - Brian Ciruna
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada
- Department of Molecular Genetics, The University of Toronto, Toronto, Ontario M5S 1A8, Canada
| |
Collapse
|
19
|
Encina A, Ligero M, Sánchez-Guerrero MJ, Rodríguez-Sainz de los Terreros A, Bartolomé E, Valera M. Phenotypic and Genetic Study of the Presence of Hair Whorls in Pura Raza Español Horses. Animals (Basel) 2023; 13:2943. [PMID: 37760344 PMCID: PMC10525084 DOI: 10.3390/ani13182943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 09/13/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
Hair whorls are a hereditary feature in horses that may be associated with temperament and coat color. Hair whorls are described as changes in the hair pattern and may take various forms, such as circular and linear whorls. We first carried out a frequency analysis of hair whorls (circular and linear). Next, a Generalized Non-Linear Model was computed to assess the significance of some potential influencing factors, and a genetic parameter estimation was performed. ENDOG software v4.8 was used to estimate the inbreeding coefficient of all the animals analyzed. It was more common to find horses with circular hair whorls than with linear whorls. The heritability ranges obtained were, in general, medium-high for both circular whorls (0.20 to 0.90) and linear whorls (0.44 to 0.84). High positive correlations were found on the between left and right positions, indicating a tendency to symmetry in certain locations. The laterality of hair whorls was also evidenced, with the biggest concentration on the left-hand side, particularly in gray horses, showing circular whorls below the central line of eyes, which has been associated in a previous paper with a calmer and more docile temperament.
Collapse
Affiliation(s)
- Ana Encina
- Real Asociación Nacional de Criadores de Caballos de Pura Raza Española (ANCCE), 41014 Sevilla, Spain;
| | - Manuel Ligero
- Departamento de Agronomía, Escuela Técnica Superior de Ingeniaría Agronómica, Universidad de Sevilla, Carretera de Utrera Km 1, 41013 Sevilla, Spain; (M.L.); (M.J.S.-G.); (E.B.); (M.V.)
| | - María José Sánchez-Guerrero
- Departamento de Agronomía, Escuela Técnica Superior de Ingeniaría Agronómica, Universidad de Sevilla, Carretera de Utrera Km 1, 41013 Sevilla, Spain; (M.L.); (M.J.S.-G.); (E.B.); (M.V.)
| | | | - Ester Bartolomé
- Departamento de Agronomía, Escuela Técnica Superior de Ingeniaría Agronómica, Universidad de Sevilla, Carretera de Utrera Km 1, 41013 Sevilla, Spain; (M.L.); (M.J.S.-G.); (E.B.); (M.V.)
| | - Mercedes Valera
- Departamento de Agronomía, Escuela Técnica Superior de Ingeniaría Agronómica, Universidad de Sevilla, Carretera de Utrera Km 1, 41013 Sevilla, Spain; (M.L.); (M.J.S.-G.); (E.B.); (M.V.)
| |
Collapse
|
20
|
Klumpe HE, Garcia-Ojalvo J, Elowitz MB, Antebi YE. The computational capabilities of many-to-many protein interaction networks. Cell Syst 2023; 14:430-446. [PMID: 37348461 PMCID: PMC10318606 DOI: 10.1016/j.cels.2023.05.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 04/14/2023] [Accepted: 05/11/2023] [Indexed: 06/24/2023]
Abstract
Many biological circuits comprise sets of protein variants that interact with one another in a many-to-many, or promiscuous, fashion. These architectures can provide powerful computational capabilities that are especially critical in multicellular organisms. Understanding the principles of biochemical computations in these circuits could allow more precise control of cellular behaviors. However, these systems are inherently difficult to analyze, due to their large number of interacting molecular components, partial redundancies, and cell context dependence. Here, we discuss recent experimental and theoretical advances that are beginning to reveal how promiscuous circuits compute, what roles those computations play in natural biological contexts, and how promiscuous architectures can be applied for the design of synthetic multicellular behaviors.
Collapse
Affiliation(s)
- Heidi E Klumpe
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA; Biological Design Center, Boston University, Boston, MA 02215, USA
| | - Jordi Garcia-Ojalvo
- Department of Medicine and Life Sciences, Pompeu Fabra University, 08003 Barcelona, Spain.
| | - Michael B Elowitz
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA.
| | - Yaron E Antebi
- Department of Molecular Genetics, Weizmann Institute of Science 76100, Rehovot, Israel.
| |
Collapse
|
21
|
Zheng Y, Shen P, Tong M, Li H, Ren C, Wu F, Li H, Yang H, Cai B, Du W, Zhao X, Yao S, Quan R. WISP2 downregulation inhibits the osteogenic differentiation of BMSCs in congenital scoliosis by regulating Wnt/β-catenin pathway. Biochim Biophys Acta Mol Basis Dis 2023:166783. [PMID: 37302424 DOI: 10.1016/j.bbadis.2023.166783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 05/09/2023] [Accepted: 06/05/2023] [Indexed: 06/13/2023]
Abstract
OBJECTIVES Bone marrow mesenchymal stem cells (BMSCs) are instrumental in bone development, metabolism, and marrow microenvironment homeostasis. Despite this, the relevant effects and mechanisms of BMSCs on congenital scoliosis (CS) remain undefined. Herein, it becomes our focus to reveal the corresponding effects and mechanisms implicated. METHODS BMSCs from CS patients (hereafter referred as CS-BMSCs) and healthy donors (NC-BMSCs) were observed and identified. Differentially expressed genes in BMSCs were analyzed utilizing scRNA-seq and RNA-seq profiles. The multi-differentiation potential of BMSCs following the transfection or infection was evaluated. The expression levels of factors related to osteogenic differentiation and Wnt/β-catenin pathway were further determined as appropriate. RESULTS A decreased osteogenic differentiation ability was shown in CS-BMSCs. Both the proportion of LEPR+ BMSCs and the expression level of WNT1-inducible-signaling pathway protein 2 (WISP2) were decreased in CS-BMSCs. WISP2 knockdown suppressed the osteogenic differentiation of NC-BMSCs, while WISP2 overexpression facilitated the osteogenesis of CS-BMSCs via acting on the Wnt/β-catenin pathway. CONCLUSIONS Our study collectively indicates WISP2 knockdown blocks the osteogenic differentiation of BMSCs in CS by regulating Wnt/β-catenin signaling, thus providing new insights into the aetiology of CS.
Collapse
Affiliation(s)
- Yang Zheng
- Zhejiang Chinese Medical University, Hangzhou, China; Department of Orthopedics Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Panyang Shen
- Department of Orthopedics Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Mengsha Tong
- Zhejiang Chinese Medical University, Hangzhou, China
| | - Hangchao Li
- Zhejiang Chinese Medical University, Hangzhou, China
| | - Conglin Ren
- Zhejiang Chinese Medical University, Hangzhou, China
| | - Fengqing Wu
- Zhejiang Chinese Medical University, Hangzhou, China
| | - Hanyu Li
- Zhejiang Chinese Medical University, Hangzhou, China
| | - Huan Yang
- Department of Biochemistry, Zhejiang University School of Medicine, Hangzhou, China
| | - Bingbing Cai
- Department of Orthopedics, Xiaoshan Traditional Chinese Medical Hospital, Hangzhou, China
| | - Weibin Du
- Department of Orthopedics, Xiaoshan Traditional Chinese Medical Hospital, Hangzhou, China
| | - Xing Zhao
- Department of Orthopedics Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Shasha Yao
- Department of Orthopedics Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Renfu Quan
- Zhejiang Chinese Medical University, Hangzhou, China; Department of Orthopedics, Xiaoshan Traditional Chinese Medical Hospital, Hangzhou, China; Research Institute of Orthopedics, The Affiliated Jiangnan Hospital of Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
22
|
Wu N, Liu L, Zhang Y, Wang L, Wang S, Zhao S, Li G, Yang Y, Lin G, Shen J, Wu Z, Qiu G, Zhang TJ. Retrospective Analysis of Associated Anomalies in 636 Patients with Operatively Treated Congenital Scoliosis. J Bone Joint Surg Am 2023; 105:537-548. [PMID: 37017616 DOI: 10.2106/jbjs.22.00277] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/06/2023]
Abstract
BACKGROUND Congenital scoliosis is frequently associated with anomalies in multiple organ systems. However, the prevalence and distribution of associated anomalies remain unclear, and there is a large amount of variation in data among different studies. METHODS Six hundred and thirty-six Chinese patients who had undergone scoliosis correction surgery at Peking Union Medical College Hospital from January 2012 to July 2019 were recruited, as a part of the Deciphering disorders Involving Scoliosis and COmorbidities (DISCO) study. The medical data for each subject were collected and analyzed. RESULTS The mean age (and standard deviation) at the time of presentation for scoliosis was 6.4 ± 6.3 years, and the mean Cobb angle of the major curve was 60.8° ± 26.5°. Intraspinal abnormalities were found in 186 (30.3%) of 614 patients, with diastematomyelia being the most common anomaly (59.1%; 110 of 186). The prevalence of intraspinal abnormalities was remarkably higher in patients with failure of segmentation and mixed deformities than in patients with failure of formation (p < 0.001). Patients with intraspinal anomalies showed more severe deformities, including larger Cobb angles of the major curve (p < 0.001). We also demonstrated that cardiac anomalies were associated with remarkably worse pulmonary function, i.e., lower forced expiratory volume in the first second (FEV1), forced vital capacity (FVC), and peak expiratory flow (PEF). Additionally, we identified associations among different concomitant malformations. We found that patients with musculoskeletal anomalies of types other than intraspinal and maxillofacial were 9.2 times more likely to have additional maxillofacial anomalies. CONCLUSIONS In our cohort, comorbidities associated with congenital scoliosis occurred at a rate of 55%. To our knowledge, our study is the first to show that patients with congenital scoliosis and cardiac anomalies have reduced pulmonary function, as demonstrated by lower FEV1, FVC, and PEF. Moreover, the potential associations among concomitant anomalies revealed the importance of a comprehensive preoperative evaluation scheme. LEVEL OF EVIDENCE Diagnostic Level III. See Instructions for Authors for a complete description of levels of evidence.
Collapse
Affiliation(s)
- Nan Wu
- Department of Orthopedic Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People's Republic of China
- Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, People's Republic of China
- Key Laboratory of Big Data for Spinal Deformities, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Lian Liu
- Department of Orthopedic Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People's Republic of China
- Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, People's Republic of China
- Key Laboratory of Big Data for Spinal Deformities, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
- Department of Emergency Surgery, Qilu Hospital of Shandong University, Jinan, Shandong Province, People's Republic of China
| | - Yuanqiang Zhang
- Department of Orthopedic Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People's Republic of China
- Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, People's Republic of China
- Key Laboratory of Big Data for Spinal Deformities, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
- Department of Orthopedic Surgery, Qilu Hospital of Shandong University, Jinan, Shandong Province, People's Republic of China
| | - Lianlei Wang
- Department of Orthopedic Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People's Republic of China
- Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, People's Republic of China
- Key Laboratory of Big Data for Spinal Deformities, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
- Department of Orthopedic Surgery, Qilu Hospital of Shandong University, Jinan, Shandong Province, People's Republic of China
| | - Shengru Wang
- Department of Orthopedic Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People's Republic of China
- Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, People's Republic of China
- Key Laboratory of Big Data for Spinal Deformities, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Sen Zhao
- Department of Orthopedic Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People's Republic of China
- Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, People's Republic of China
- Key Laboratory of Big Data for Spinal Deformities, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
- Graduate School of Peking Union Medical College, Beijing, People's Republic of China
| | - Guozhuang Li
- Department of Orthopedic Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People's Republic of China
- Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, People's Republic of China
- Key Laboratory of Big Data for Spinal Deformities, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
- Graduate School of Peking Union Medical College, Beijing, People's Republic of China
| | - Yang Yang
- Department of Orthopedic Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People's Republic of China
- Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, People's Republic of China
- Key Laboratory of Big Data for Spinal Deformities, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Guanfeng Lin
- Department of Orthopedic Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People's Republic of China
- Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, People's Republic of China
- Key Laboratory of Big Data for Spinal Deformities, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Jianxiong Shen
- Department of Orthopedic Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People's Republic of China
- Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, People's Republic of China
- Key Laboratory of Big Data for Spinal Deformities, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Zhihong Wu
- Department of Orthopedic Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People's Republic of China
- Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, People's Republic of China
- Key Laboratory of Big Data for Spinal Deformities, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Guixing Qiu
- Department of Orthopedic Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People's Republic of China
- Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, People's Republic of China
- Key Laboratory of Big Data for Spinal Deformities, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Terry Jianguo Zhang
- Department of Orthopedic Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People's Republic of China
- Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, People's Republic of China
- Key Laboratory of Big Data for Spinal Deformities, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| |
Collapse
|
23
|
Wopat S, Adhyapok P, Daga B, Crawford JM, Peskin B, Norman J, Bagwell J, Fogerson SM, Di Talia S, Kiehart DP, Charbonneau P, Bagnat M. Axial segmentation by iterative mechanical signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.27.534101. [PMID: 37034817 PMCID: PMC10081202 DOI: 10.1101/2023.03.27.534101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
In bony fishes, formation of the vertebral column, or spine, is guided by a metameric blueprint established in the epithelial sheath of the notochord. Generation of the notochord template begins days after somitogenesis and even occurs in the absence of somite segmentation. However, patterning defects in the somites lead to imprecise notochord segmentation, suggesting these processes are linked. Here, we reveal that spatial coordination between the notochord and the axial musculature is necessary to ensure segmentation of the zebrafish spine both in time and space. We find that the connective tissues that anchor the axial skeletal musculature, known as the myosepta in zebrafish, transmit spatial patterning cues necessary to initiate notochord segment formation, a critical pre-patterning step in spine morphogenesis. When an irregular pattern of muscle segments and myosepta interact with the notochord sheath, segments form non-sequentially, initiate at atypical locations, and eventually display altered morphology later in development. We determine that locations of myoseptum-notochord connections are hubs for mechanical signal transmission, which are characterized by localized sites of deformation of the extracellular matrix (ECM) layer encasing the notochord. The notochord sheath responds to the external mechanical changes by locally augmenting focal adhesion machinery to define the initiation site for segmentation. Using a coarse-grained mathematical model that captures the spatial patterns of myoseptum-notochord interactions, we find that a fixed-length scale of external cues is critical for driving sequential segment patterning in the notochord. Together, this work identifies a robust segmentation mechanism that hinges upon mechanical coupling of adjacent tissues to control patterning dynamics.
Collapse
Affiliation(s)
- Susan Wopat
- Department of Cell Biology, Duke University, Durham, North Carolina 27710, USA
- Present address: Department of Physics, University of California Santa Barbara, Santa Barbara, California 93106, USA
- Authors contributed equally to this work
| | - Priyom Adhyapok
- Department of Cell Biology, Duke University, Durham, North Carolina 27710, USA
- Authors contributed equally to this work
| | - Bijoy Daga
- Department of Cell Biology, Duke University, Durham, North Carolina 27710, USA
- Present address: Institute of Medical Sciences, Foresterhill Health Campus, University of Aberdeen, Aberdeen AB25 2ZD, UK
- Authors contributed equally to this work
| | - Janice M. Crawford
- Department of Biology, Duke University, Durham, North Carolina 27710, USA
| | - Brianna Peskin
- Department of Cell Biology, Duke University, Durham, North Carolina 27710, USA
| | - James Norman
- Department of Cell Biology, Duke University, Durham, North Carolina 27710, USA
| | - Jennifer Bagwell
- Department of Cell Biology, Duke University, Durham, North Carolina 27710, USA
| | | | - Stefano Di Talia
- Department of Cell Biology, Duke University, Durham, North Carolina 27710, USA
| | - Daniel P. Kiehart
- Department of Biology, Duke University, Durham, North Carolina 27710, USA
| | - Patrick Charbonneau
- Department of Chemistry, Duke University, Durham, North Carolina 27708, USA
- Department of Physics, Duke University, Durham, North Carolina 27708, USA
| | - Michel Bagnat
- Department of Cell Biology, Duke University, Durham, North Carolina 27710, USA
- Lead contact
| |
Collapse
|
24
|
Abstract
Notch signaling is a highly conserved signaling pathway that coordinates cellular differentiation during the development and homeostasis in numerous organs and tissues across metazoans. Activation of Notch signaling relies on direct contact between neighboring cells and mechanical pulling of the Notch receptors by the Notch ligands. Notch signaling is commonly used in developmental processes to coordinate the differentiation into distinct cell fates of neighboring cells. In this Development at a Glance article, we describe the current understanding of the Notch pathway activation and the different regulatory levels that control the pathway. We then describe several developmental processes where Notch is crucial for coordinating differentiation. These examples include processes that are largely based on lateral inhibition mechanisms giving rise to alternating patterns (e.g. SOP selection, hair cell in the inner ear and neural stem cell maintenance), as well as processes where Notch activity is oscillatory (e.g. somitogenesis and neurogenesis in mammals).
Collapse
Affiliation(s)
- Oren Gozlan
- School of Neurobiology, Biochemistry, and Biophysics, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - David Sprinzak
- School of Neurobiology, Biochemistry, and Biophysics, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| |
Collapse
|
25
|
Yaman YI, Ramanathan S. Controlling human organoid symmetry breaking reveals signaling gradients drive segmentation clock waves. Cell 2023; 186:513-527.e19. [PMID: 36657441 PMCID: PMC10025047 DOI: 10.1016/j.cell.2022.12.042] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 09/29/2022] [Accepted: 12/21/2022] [Indexed: 01/19/2023]
Abstract
Axial development of mammals involves coordinated morphogenetic events, including axial elongation, somitogenesis, and neural tube formation. To gain insight into the signals controlling the dynamics of human axial morphogenesis, we generated axially elongating organoids by inducing anteroposterior symmetry breaking of spatially coupled epithelial cysts derived from human pluripotent stem cells. Each organoid was composed of a neural tube flanked by presomitic mesoderm sequentially segmented into somites. Periodic activation of the somite differentiation gene MESP2 coincided in space and time with anteriorly traveling segmentation clock waves in the presomitic mesoderm of the organoids, recapitulating critical aspects of somitogenesis. Timed perturbations demonstrated that FGF and WNT signaling play distinct roles in axial elongation and somitogenesis, and that FGF signaling gradients drive segmentation clock waves. By generating and perturbing organoids that robustly recapitulate the architecture of multiple axial tissues in human embryos, this work offers a means to dissect mechanisms underlying human embryogenesis.
Collapse
Affiliation(s)
- Yusuf Ilker Yaman
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA.
| | - Sharad Ramanathan
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|
26
|
Uriu K, Morelli LG. Orchestration of tissue shape changes and gene expression patterns in development. Semin Cell Dev Biol 2023; 147:24-33. [PMID: 36631335 DOI: 10.1016/j.semcdb.2022.12.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/27/2022] [Accepted: 12/27/2022] [Indexed: 01/11/2023]
Abstract
In development, tissue shape changes and gene expression patterns give rise to morphogenesis. Understanding tissue shape changes requires the analysis of mechanical properties of the tissue such as tissue rigidity, cell influx from neighboring tissues, cell shape changes and cell proliferation. Local and global gene expression patterns can be influenced by neighbor exchange and tissue shape changes. Here we review recent studies on the mechanisms for tissue elongation and its influences on dynamic gene expression patterns by focusing on vertebrate somitogenesis. We first introduce mechanical and biochemical properties of the segmenting tissue that drive tissue elongation. Then, we discuss patterning in the presence of cell mixing, scaling of signaling gradients, and dynamic phase waves of rhythmic gene expression under tissue shape changes. We also highlight the importance of theoretical approaches to address the relation between tissue shape changes and patterning.
Collapse
Affiliation(s)
- Koichiro Uriu
- Graduate School of Natural Science and Technology, Kanazawa University, Kakuma-machi, Kanazawa 920-1192 Japan.
| | - Luis G Morelli
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)-CONICET-Partner Institute of the Max Planck Society, Polo Científico Tecnológico, Godoy Cruz 2390, C1425FQD, Buenos Aires, Argentina; Departamento de Física, FCEyN UBA, Ciudad Universitaria, 1428 Buenos Aires, Argentina.
| |
Collapse
|
27
|
Diaz-Cuadros M, Miettinen TP, Skinner OS, Sheedy D, Díaz-García CM, Gapon S, Hubaud A, Yellen G, Manalis SR, Oldham WM, Pourquié O. Metabolic regulation of species-specific developmental rates. Nature 2023; 613:550-557. [PMID: 36599986 PMCID: PMC9944513 DOI: 10.1038/s41586-022-05574-4] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 11/18/2022] [Indexed: 01/06/2023]
Abstract
Animals display substantial inter-species variation in the rate of embryonic development despite a broad conservation of the overall sequence of developmental events. Differences in biochemical reaction rates, including the rates of protein production and degradation, are thought to be responsible for species-specific rates of development1-3. However, the cause of differential biochemical reaction rates between species remains unknown. Here, using pluripotent stem cells, we have established an in vitro system that recapitulates the twofold difference in developmental rate between mouse and human embryos. This system provides a quantitative measure of developmental speed as revealed by the period of the segmentation clock, a molecular oscillator associated with the rhythmic production of vertebral precursors. Using this system, we show that mass-specific metabolic rates scale with the developmental rate and are therefore higher in mouse cells than in human cells. Reducing these metabolic rates by inhibiting the electron transport chain slowed down the segmentation clock by impairing the cellular NAD+/NADH redox balance and, further downstream, lowering the global rate of protein synthesis. Conversely, increasing the NAD+/NADH ratio in human cells by overexpression of the Lactobacillus brevis NADH oxidase LbNOX increased the translation rate and accelerated the segmentation clock. These findings represent a starting point for the manipulation of developmental rate, with multiple translational applications including accelerating the differentiation of human pluripotent stem cells for disease modelling and cell-based therapies.
Collapse
Affiliation(s)
- Margarete Diaz-Cuadros
- Department of Genetics, Harvard Medical School, Boston, MA, USA.
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA.
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA.
| | - Teemu P Miettinen
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Owen S Skinner
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
- Broad Institute, Cambridge, MA, USA
| | - Dylan Sheedy
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Carlos Manlio Díaz-García
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Svetlana Gapon
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Alexis Hubaud
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Gary Yellen
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Scott R Manalis
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - William M Oldham
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Olivier Pourquié
- Department of Genetics, Harvard Medical School, Boston, MA, USA.
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA.
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
28
|
Schnirman RE, Kuo SJ, Kelly RC, Yamaguchi TP. The role of Wnt signaling in the development of the epiblast and axial progenitors. Curr Top Dev Biol 2023; 153:145-180. [PMID: 36967193 DOI: 10.1016/bs.ctdb.2023.01.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
Abstract
Understanding how the body plan is established during embryogenesis remains a fundamental biological question. The Wnt/β-catenin signaling pathway plays a crucial and highly conserved role in body plan formation, functioning to polarize the primary anterior-posterior (AP) or head-to-tail body axis in most metazoans. In this chapter, we focus on the roles that the mammalian Wnt/β-catenin pathway plays to prepare the pluripotent epiblast for gastrulation, and to elicit the emergence of multipotent axial progenitors from the caudal epiblast. Interactions between Wnt and retinoic acid (RA), another powerful family of developmental signaling molecules, in axial progenitors will also be discussed. Gastrulation movements and somitogenesis result in the anterior displacement of the RA source (the rostral somites and lateral plate mesoderm (LPM)), from the posterior Wnt source (the primitive streak (PS)), leading to the establishment of antiparallel gradients of RA and Wnt that control the self-renewal and successive differentiation of neck, trunk and tail progenitors.
Collapse
Affiliation(s)
| | - Samuel J Kuo
- NCI-Frederick, NIH, Frederick, MD, United States
| | - Ryan C Kelly
- NCI-Frederick, NIH, Frederick, MD, United States
| | | |
Collapse
|
29
|
Wang M, Li Z, Zhao S, Zheng Z, Wang Y, Qiu G, Wu Z, Wu N, Zhang TJ, Cai S. The identification of PAX7 variants and a potential role of muscle development dysfunction in congenital scoliosis. CELL REGENERATION 2022; 11:16. [PMID: 35499749 PMCID: PMC9061922 DOI: 10.1186/s13619-022-00116-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Accepted: 03/16/2022] [Indexed: 11/17/2022]
|
30
|
Bearce EA, Irons ZH, O'Hara-Smith JR, Kuhns CJ, Fisher SI, Crow WE, Grimes DT. Urotensin II-related peptides, Urp1 and Urp2, control zebrafish spine morphology. eLife 2022; 11:e83883. [PMID: 36453722 PMCID: PMC9836392 DOI: 10.7554/elife.83883] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Accepted: 11/24/2022] [Indexed: 12/03/2022] Open
Abstract
The spine provides structure and support to the body, yet how it develops its characteristic morphology as the organism grows is little understood. This is underscored by the commonality of conditions in which the spine curves abnormally such as scoliosis, kyphosis, and lordosis. Understanding the origin of these spinal curves has been challenging in part due to the lack of appropriate animal models. Recently, zebrafish have emerged as promising tools with which to understand the origin of spinal curves. Using zebrafish, we demonstrate that the urotensin II-related peptides (URPs), Urp1 and Urp2, are essential for maintaining spine morphology. Urp1 and Urp2 are 10-amino acid cyclic peptides expressed by neurons lining the central canal of the spinal cord. Upon combined genetic loss of Urp1 and Urp2, adolescent-onset planar curves manifested in the caudal region of the spine. Highly similar curves were caused by mutation of Uts2r3, an URP receptor. Quantitative comparisons revealed that urotensin-associated curves were distinct from other zebrafish spinal curve mutants in curve position and direction. Last, we found that the Reissner fiber, a proteinaceous thread that sits in the central canal and has been implicated in the control of spine morphology, breaks down prior to curve formation in mutants with perturbed cilia motility but was unaffected by loss of Uts2r3. This suggests a Reissner fiber-independent mechanism of curvature in urotensin-deficient mutants. Overall, our results show that Urp1 and Urp2 control zebrafish spine morphology and establish new animal models of spine deformity.
Collapse
Affiliation(s)
- Elizabeth A Bearce
- Institute of Molecular Biology, Department of Biology, University of OregonEugeneUnited States
| | - Zoe H Irons
- Institute of Molecular Biology, Department of Biology, University of OregonEugeneUnited States
| | | | - Colin J Kuhns
- Institute of Molecular Biology, Department of Biology, University of OregonEugeneUnited States
| | - Sophie I Fisher
- Institute of Molecular Biology, Department of Biology, University of OregonEugeneUnited States
| | - William E Crow
- Institute of Molecular Biology, Department of Biology, University of OregonEugeneUnited States
| | - Daniel T Grimes
- Institute of Molecular Biology, Department of Biology, University of OregonEugeneUnited States
| |
Collapse
|
31
|
Sherratt E, Nash-Hahn T, Nankivell JH, Rasmussen AR, Hampton PM, Sanders KL. Macroevolution in axial morphospace: innovations accompanying the transition to marine environments in elapid snakes. ROYAL SOCIETY OPEN SCIENCE 2022; 9:221087. [PMID: 36569233 PMCID: PMC9768463 DOI: 10.1098/rsos.221087] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 11/29/2022] [Indexed: 06/17/2023]
Abstract
Sea snakes in the Hydrophis-Microcephalophis clade (Elapidae) show exceptional body shape variation along a continuum from similar forebody and hindbody girths, to dramatically reduced girths of the forebody relative to hindbody. The latter is associated with specializations on burrowing prey. This variation underpins high sympatric diversity and species richness and is not shared by other marine (or terrestrial) snakes. Here, we examined a hypothesis that macroevolutionary changes in axial development contribute to the propensity, at clade level, for body shape change. We quantified variation in the number and size of vertebrae in two body regions (pre- and post-apex of the heart) for approximately 94 terrestrial and marine elapids. We found Hydrophis-Microcephalophis exhibit increased rates of vertebral evolution in the pre- versus post-apex regions compared to all other Australasian elapids. Unlike other marine and terrestrial elapids, axial elongation in Hydrophis-Microcephalophis occurs via the preferential addition of vertebrae pre-heart apex, which is the region that undergoes concomitant shifts in vertebral number and size during transitions along the relative fore- to hindbody girth axis. We suggest that this macroevolutionary developmental change has potentially acted as a key innovation in Hydrophis-Microcephalophis by facilitating novel (especially burrowing) prey specializations that are not shared with other marine snakes.
Collapse
Affiliation(s)
- Emma Sherratt
- School of Biological Sciences, The University of Adelaide, Adelaide, South Australia 5005, Australia
- South Australian Museum, North Terrace, Adelaide, South Australia 5000, Australia
| | - Tamika Nash-Hahn
- School of Biological Sciences, The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - James H. Nankivell
- School of Biological Sciences, The University of Adelaide, Adelaide, South Australia 5005, Australia
- South Australian Museum, North Terrace, Adelaide, South Australia 5000, Australia
| | - Arne R. Rasmussen
- The Royal Danish Academy, Institute of Conservation, 1435 Copenhagen, Denmark
| | - Paul M. Hampton
- Department of Biology, Colorado Mesa University, Grand Junction, CO 81501, USA
| | - Kate L. Sanders
- School of Biological Sciences, The University of Adelaide, Adelaide, South Australia 5005, Australia
- South Australian Museum, North Terrace, Adelaide, South Australia 5000, Australia
| |
Collapse
|
32
|
Naznin RA, Haq MA, Sumi SA, Ahmad R, Haque M. A Semi-quantitative Evaluation of Out-to-Out Agenesis of Posterior Wall in a Dry Human Sacrum in Bangladesh. Cureus 2022; 14:e31163. [DOI: 10.7759/cureus.31163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/06/2022] [Indexed: 11/07/2022] Open
|
33
|
25 years of the segmentation clock gene. Nature 2022; 611:671-673. [PMID: 36352110 DOI: 10.1038/d41586-022-03562-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
34
|
Adler KA, De Nault DL, Cardoza CM, Womack M. Evolutionary rates and shape variation along the anuran vertebral column with attention to phylogeny, body size, and ecology. Evolution 2022; 76:2724-2738. [PMID: 36117276 DOI: 10.1111/evo.14614] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 07/31/2022] [Accepted: 08/08/2022] [Indexed: 01/22/2023]
Abstract
The vertebral column is critical to a vertebrate species' flexibility and skeletal support, making vertebrae a clear target for selection. Anurans (frogs and toads) have a unique, truncated vertebral column that appears constrained to provide axial rigidity for efficient jumping. However, no study has examined how presacral vertebrae shape varies among anuran species at the macroevolutionary scale nor how intrinsic (developmental and phylogenetic) and extrinsic (ecological) factors may have influenced vertebrae shape evolution. We used microCT scans and phylogenetic comparative methods to examine the vertebrae of hundreds of anuran species that vary in body size as well as adult and larval ecology. We found variation in shape and evolutionary rates among anuran vertebrae, dispelling any notion that trunk vertebrae evolve uniformly. We discovered the highest evolutionary rates in the cervical vertebrae and in the more caudal trunk vertebrae. We found little evidence for selection pressures related to adult or larval ecology affecting vertebrae evolution, but we did find body size was highly associated with vertebrae shape and microhabitat (mainly burrowing) affected those allometric relationships. Our results provide an interesting comparison to vertebrae evolution in other clades and a jumping-off point for studies of anuran vertebrae evolution and development.
Collapse
Affiliation(s)
- Katie A Adler
- Museum of Vertebrate Zoology, University of California, Berkeley, Berkeley, California, 94720
| | - Diego L De Nault
- Museum of Vertebrate Zoology, University of California, Berkeley, Berkeley, California, 94720
| | - Cassandra M Cardoza
- Museum of Vertebrate Zoology, University of California, Berkeley, Berkeley, California, 94720
| | - Molly Womack
- Museum of Vertebrate Zoology, University of California, Berkeley, Berkeley, California, 94720.,Department of Biology, Utah State University, Logan, Utah, 84322
| |
Collapse
|
35
|
Wu Y, Zhang HQ, Tang M, Guo C, Liu S, Li J, Wang Y, Xiao L, Yang G. Abnormal TNS3 gene methylation in patients with congenital scoliosis. BMC Musculoskelet Disord 2022; 23:797. [PMID: 35987623 PMCID: PMC9392296 DOI: 10.1186/s12891-022-05730-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 08/02/2022] [Indexed: 11/10/2022] Open
Abstract
Background Congenital scoliosis (CS) is a congenital deformity of the spine resulting from abnormal and asymmetrical development of vertebral bodies during pregnancy. However, the etiology and mechanism of CS remain unclear. Epigenetics is the study of heritable variations in gene expression outside of changes in nucleotide sequence. Among these, DNA methylation was described first and is the most characteristic and most stable epigenetic mechanism. Therefore, in this study, we aim to explore the association between genome methylation and CS which are not been studied before. Methods Two pairs of monozygotic twins were included, with each pair involving one individual with and one without CS. Agilent SureSelect XT Human Methyl-Sequencing was used for genome methylation sequencing. MethylTarget was used to detect methylation levels in target regions. Immunohistochemistry was performed to visualize expression of associated genes in candidate regions. Results A total of 75 differentially methylated regions were identified, including 24 with an increased methylation level and 51 with a decreased methylation level in the CS group. Nine of the differentially methylated regions were selected (TNS3, SEMAC3, GPR124, MEST, DLK1, SNTG1, PPIB, DEF8, and GRHL2). The results showed that the methylation level of the promoter region of TNS3 was 0.72 ± 0.08 in the CS group and 0.43 ± 0.06 in the control group (p = 0.00070 < 0.01). There was no significant difference in the degree of methylation of SEMAC3, GPR124, MEST, DLK1, SNTG1, PPIB, DEF8, or GRHL2 between the two groups. Immunohistochemistry showed significantly decreased TNS3 expression in the cartilage of the articular process in CS (CS: 0.011 ± 0.002; control: 0.018 ± 0.006, P = 0.003 < 0.01). Conclusion Compared with the control group, high-level methylation of the TNS3 promoter region and low TNS3 expression in the cartilage layer of the articular process characterize CS. Thus, DNA methylation and TNS3 may play important roles in the pathogenesis of CS. Supplementary Information The online version contains supplementary material available at 10.1186/s12891-022-05730-x.
Collapse
|
36
|
Naganathan SR, Popović M, Oates AC. Left-right symmetry of zebrafish embryos requires somite surface tension. Nature 2022; 605:516-521. [PMID: 35477753 DOI: 10.1038/s41586-022-04646-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 03/15/2022] [Indexed: 02/06/2023]
Abstract
The body axis of vertebrate embryos is periodically segmented into bilaterally symmetric pairs of somites1,2. The anteroposterior length of somites, their position and left-right symmetry are thought to be molecularly determined before somite morphogenesis3,4. Here we show that, in zebrafish embryos, initial somite anteroposterior lengths and positions are imprecise and, consequently, many somite pairs form left-right asymmetrically. Notably, these imprecisions are not left unchecked and we find that anteroposterior lengths adjust within an hour after somite formation, thereby increasing morphological symmetry. We find that anteroposterior length adjustments result entirely from changes in somite shape without change in somite volume, with changes in anteroposterior length being compensated by corresponding changes in mediolateral length. The anteroposterior adjustment mechanism is facilitated by somite surface tension, which we show by comparing in vivo experiments and in vitro single-somite explant cultures using a mechanical model. Length adjustment is inhibited by perturbation of molecules involved in surface tension, such as integrin and fibronectin. By contrast, the adjustment mechanism is unaffected by perturbations to the segmentation clock, therefore revealing a distinct process that influences morphological segment lengths. We propose that tissue surface tension provides a general mechanism to adjust shapes and ensure precision and symmetry of tissues in developing embryos.
Collapse
Affiliation(s)
- Sundar R Naganathan
- Institute of Bioengineering, École polytechnique fédérale de Lausanne, Lausanne, Switzerland.
| | - Marko Popović
- Institute of Physics, École polytechnique fédérale de Lausanne, Lausanne, Switzerland. .,Max Planck Institute for Physics of Complex Systems, Dresden, Germany. .,Center for Systems Biology Dresden, Dresden, Germany.
| | - Andrew C Oates
- Institute of Bioengineering, École polytechnique fédérale de Lausanne, Lausanne, Switzerland.
| |
Collapse
|
37
|
Notch signaling pathway: architecture, disease, and therapeutics. Signal Transduct Target Ther 2022; 7:95. [PMID: 35332121 PMCID: PMC8948217 DOI: 10.1038/s41392-022-00934-y] [Citation(s) in RCA: 441] [Impact Index Per Article: 147.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 02/16/2022] [Accepted: 02/16/2022] [Indexed: 02/07/2023] Open
Abstract
The NOTCH gene was identified approximately 110 years ago. Classical studies have revealed that NOTCH signaling is an evolutionarily conserved pathway. NOTCH receptors undergo three cleavages and translocate into the nucleus to regulate the transcription of target genes. NOTCH signaling deeply participates in the development and homeostasis of multiple tissues and organs, the aberration of which results in cancerous and noncancerous diseases. However, recent studies indicate that the outcomes of NOTCH signaling are changeable and highly dependent on context. In terms of cancers, NOTCH signaling can both promote and inhibit tumor development in various types of cancer. The overall performance of NOTCH-targeted therapies in clinical trials has failed to meet expectations. Additionally, NOTCH mutation has been proposed as a predictive biomarker for immune checkpoint blockade therapy in many cancers. Collectively, the NOTCH pathway needs to be integrally assessed with new perspectives to inspire discoveries and applications. In this review, we focus on both classical and the latest findings related to NOTCH signaling to illustrate the history, architecture, regulatory mechanisms, contributions to physiological development, related diseases, and therapeutic applications of the NOTCH pathway. The contributions of NOTCH signaling to the tumor immune microenvironment and cancer immunotherapy are also highlighted. We hope this review will help not only beginners but also experts to systematically and thoroughly understand the NOTCH signaling pathway.
Collapse
|
38
|
Developments in Congenital Scoliosis and Related Research from 1992 to 2021: A Thirty-Year Bibliometric Analysis. World Neurosurg 2022; 164:e24-e44. [PMID: 35248773 DOI: 10.1016/j.wneu.2022.02.117] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 02/25/2022] [Accepted: 02/26/2022] [Indexed: 11/21/2022]
Abstract
BACKGROUND Many studies, mainly original articles and reviews, have been reported on congenital scoliosis (CS), but there is a lack of bibliometric analyses. This study aimed to systematically analyze the developments and focuses in CS and related research fields. METHODS Data were retrieved from the Web of Science Core Collection database, and the top 100 most-cited studies were analyzed emphatically. The Web of Science Results Analysis and Citation Report was used to analyze different aspects of the literature. CiteSpace was used to analyze the cooperation network, reference co-citation, burst keywords, and burst citations. RESULTS The final analysis included 749 studies. CS and related research has been rapidly expanding. Several journals have published relevant studies and most-cited studies on this topic. Cooperation was noted among authors, institutions, and countries/regions in multiple instances. Surgical techniques (hemivertebra resection/posterior vertebral column resection/vertical expandable prosthetic titanium rib/double approach/grade 4 osteotomy) was one of the most common research focuses. In addition, research on genetics and molecular biology related to CS has become an emerging trend as a result of advances in basic science. CONCLUSIONS Over time, research on CS and in related fields has gained greater attention and has been expanding continuously, showing a trend toward globalization. We recommend that researchers focus on the progress of surgical techniques, advances in molecular biology and genetics, and characteristics of CS. The top clusters, most-cited articles, and references with the strongest burst citations should be studied further.
Collapse
|
39
|
FAM20C plays a critical role in the development of mouse vertebra. Spine J 2022; 22:337-348. [PMID: 34343663 DOI: 10.1016/j.spinee.2021.07.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 07/26/2021] [Accepted: 07/27/2021] [Indexed: 02/03/2023]
Abstract
BACKGROUND CONTEXT Family with sequence similarity 20-member C (FAM20C) is a protein kinase that is responsible for the phosphorylation of many secretory proteins; however, its roles in spine or vertebra development have not be studied. PURPOSE The aim of this investigation is to analyze the roles of FAM20C in vertebra development. STUDY DESIGN/SETTING A mouse study of the Fam20c gene using conditional knockout to assess the effects of its inactivation on vertebra development. METHODS By breeding Sox2-Cre mice with Fam20cflox/flox mice, Sox2-Cre;Fam20cflox/flox mice (abbreviated as cKO mice) are created. X-ray radiography, resin-casted scanning electron microscopy, Hematoxylin and Eosin staining, safranin O staining, Goldner's Masson trichrome staining, Von Kossa staining, tartrate-resistant alkaline phosphatase staining, immunohistochemistry staining, Western Immunoblotting and real-time PCR were employed to characterize the vertebrae of cKO mice compared to the normal control mice. RESULTS Inactivation of Fam20c in mice results in remarkable spine deformity, severe morphology and mineralization defects, altered levels of osteoblast differentiation markers, reduction of activity of the Wnt/β-catenin signaling pathway and reduced level of osteoclastogenesis in the vertebrae. CONCLUSIONS FAM20C plays an essential role in vertebral development; it may regulate vertebral formation through the Wnt/β-catenin signaling pathway. CLINICAL SIGNIFICANCE Mutations in the human FAM20C gene are associated with Raine syndrome. The findings of this study provide valuable clues for the clinical management of Raine syndrome regarding spine manifestations in patients.
Collapse
|
40
|
Pourquié O. A brief history of the segmentation clock. Dev Biol 2022; 485:24-36. [DOI: 10.1016/j.ydbio.2022.02.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 02/23/2022] [Indexed: 11/16/2022]
|
41
|
The canonical FGF-FGFR signaling system at the molecular level. POSTEP HIG MED DOSW 2022. [DOI: 10.2478/ahem-2021-0024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Extracellular signaling molecules, among them the fibroblast growth factors (FGFs), enable cells to communicate with neighboring cells. Such signaling molecules that receive and transmit a signal require specific tyrosine kinase receptors located at the cell surface (fibroblast growth factor receptors, FGFRs). The binding of a signaling molecule to its specific receptor results in receptor dimerization and conformational changes in the cytoplasmic part of the receptor. The conformational changes lead to trans-autophosphorylation of the tyrosine kinase domains of the receptors and subsequently to induction of several downstream signaling pathways and expression of appropriate genes. The signaling pathways activated by FGFs control and coordinate cell behaviors such as cell division, migration, differentiation, and cell death. FGFs and their transmembrane receptors are widely distributed in different tissues and participate in fundamental processes during embryonic, fetal, and adult human life. The human FGF/FGFR family comprises 22 ligands and 4 high affinity receptors. In addition, FGFs bind to low affinity receptors, heparan sulfate proteoglycans at the cell surface. The availability of appropriate ligand/receptor pair, combined with the co-receptor, initiates signaling. Inappropriate FGF/FGFR signaling can cause skeletal disorders, primarily dwarfism, craniofacial malformation syndromes, mood disorders, metabolic disorders, and Kallman syndrome. In addition, aberrations in FGF/FGFR signaling have already been reported in several types of malignant diseases. Knowledge about the molecular mechanisms of FGF/FGFR activation and signaling is necessary to understand the basis of these diseases.
Collapse
|
42
|
Dobreva MP, Camacho J, Abzhanov A. Time to synchronize our clocks: Connecting developmental mechanisms and evolutionary consequences of heterochrony. JOURNAL OF EXPERIMENTAL ZOOLOGY. PART B, MOLECULAR AND DEVELOPMENTAL EVOLUTION 2022; 338:87-106. [PMID: 34826199 DOI: 10.1002/jez.b.23103] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 09/27/2021] [Accepted: 10/12/2021] [Indexed: 06/13/2023]
Abstract
Heterochrony, defined as a change in the timing of developmental events altering the course of evolution, was first recognized by Ernst Haeckel in 1866. Haeckel's original definition was meant to explain the observed parallels between ontogeny and phylogeny, but the interpretation of his work became a source of controversy over time. Heterochrony took its modern meaning following the now classical work in the 1970-80s by Steven J. Gould, Pere Alberch, and co-workers. Predicted and described heterochronic scenarios emphasize the many ways in which developmental changes can influence evolution. However, while important examples of heterochrony detected with comparative morphological methods have multiplied, the more mechanistic understanding of this phenomenon lagged conspicuously behind. Considering the rapid progress in imaging and molecular tools available now for developmental biologists, this review aims to stress the need to take heterochrony research to the next level. It is time to synchronize the different levels of heterochrony research into a single analysis flow: from studies on organismal-level morphology to cells to molecules and genes, using complementary techniques. To illustrate how to achieve a more comprehensive understanding of phyletic morphological diversification associated with heterochrony, we discuss several recent case studies at various phylogenetic scales that combine morphological, cellular, and molecular analyses. Such a synergistic approach offers to more fully integrate phylogenetic and ontogenetic dimensions of the fascinating evolutionary phenomenon of heterochrony.
Collapse
Affiliation(s)
| | - Jasmin Camacho
- Stowers Institute for Medical Research, Kansas City, Missouri, USA
| | - Arkhat Abzhanov
- Department of Life Sciences, Imperial College London, Ascot, UK
- Department of Life Sciences, Natural History Museum, London, UK
| |
Collapse
|
43
|
Signalling dynamics in embryonic development. Biochem J 2021; 478:4045-4070. [PMID: 34871368 PMCID: PMC8718268 DOI: 10.1042/bcj20210043] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 11/10/2021] [Accepted: 11/15/2021] [Indexed: 02/08/2023]
Abstract
In multicellular organisms, cellular behaviour is tightly regulated to allow proper embryonic development and maintenance of adult tissue. A critical component in this control is the communication between cells via signalling pathways, as errors in intercellular communication can induce developmental defects or diseases such as cancer. It has become clear over the last years that signalling is not static but varies in activity over time. Feedback mechanisms present in every signalling pathway lead to diverse dynamic phenotypes, such as transient activation, signal ramping or oscillations, occurring in a cell type- and stage-dependent manner. In cells, such dynamics can exert various functions that allow organisms to develop in a robust and reproducible way. Here, we focus on Erk, Wnt and Notch signalling pathways, which are dynamic in several tissue types and organisms, including the periodic segmentation of vertebrate embryos, and are often dysregulated in cancer. We will discuss how biochemical processes influence their dynamics and how these impact on cellular behaviour within multicellular systems.
Collapse
|
44
|
Musser CG, Windsor RC, Wininger F. Corpectomy and spinal stabilization using a 3D-printed spine model and custom jigs to address severe spinal deformities from T9-11 and L2-4 in a 6-month-old German shepherd puppy. Clin Case Rep 2021; 9:e05229. [PMID: 34963810 PMCID: PMC8710705 DOI: 10.1002/ccr3.5229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 08/22/2021] [Accepted: 11/04/2021] [Indexed: 11/28/2022] Open
Abstract
This report describes surgical decompression and stabilization of 2 hemivertebrae in a German shepherd dog. Long-term clinical and imaging outcomes are documented. Spinal cord decompression via corpectomy improved neurological function and intrinsic spinal cord changes on MRI. The dog improved to have minimal paraparesis and an active lifestyle.
Collapse
Affiliation(s)
| | | | - Fred Wininger
- Charlotte Animal Referral and EmergencyCharlotteNorth CarolinaUSA
| |
Collapse
|
45
|
Abstract
Notch signalling is a well-conserved signalling pathway that regulates cell fate through cell-cell communication. A typical feature of Notch signalling is ‘lateral inhibition’, whereby two neighbouring cells of equivalent state of differentiation acquire different cell fates. Recently, mathematical and computational approaches have addressed the Notch dynamics in Drosophila neural development. Typical examples of lateral inhibition are observed in the specification of neural stem cells in the embryo and sensory organ precursors in the thorax. In eye disc development, Notch signalling cooperates with other signalling pathways to define the evenly spaced positioning of the photoreceptor cells. The interplay between Notch and epidermal growth factor receptor signalling regulates the timing of neural stem cell differentiation in the optic lobe. In this review, we summarize the theoretical studies that have been conducted to elucidate the Notch dynamics in these systems and discuss the advantages of combining mathematical models with biological experiments.
Collapse
Affiliation(s)
- Tetsuo Yasugi
- Mathematical Neuroscience Unit, Institute for Frontier Science Initiative, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Makoto Sato
- Mathematical Neuroscience Unit, Institute for Frontier Science Initiative, Kanazawa University, Kanazawa, Ishikawa, Japan.,Laboratory of Developmental Neurobiology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa, Japan
| |
Collapse
|
46
|
Diaz-Cuadros M, Pourquié O, El-Sherif E. Patterning with clocks and genetic cascades: Segmentation and regionalization of vertebrate versus insect body plans. PLoS Genet 2021; 17:e1009812. [PMID: 34648490 PMCID: PMC8516289 DOI: 10.1371/journal.pgen.1009812] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Oscillatory and sequential processes have been implicated in the spatial patterning of many embryonic tissues. For example, molecular clocks delimit segmental boundaries in vertebrates and insects and mediate lateral root formation in plants, whereas sequential gene activities are involved in the specification of regional identities of insect neuroblasts, vertebrate neural tube, vertebrate limb, and insect and vertebrate body axes. These processes take place in various tissues and organisms, and, hence, raise the question of what common themes and strategies they share. In this article, we review 2 processes that rely on the spatial regulation of periodic and sequential gene activities: segmentation and regionalization of the anterior-posterior (AP) axis of animal body plans. We study these processes in species that belong to 2 different phyla: vertebrates and insects. By contrasting 2 different processes (segmentation and regionalization) in species that belong to 2 distantly related phyla (arthropods and vertebrates), we elucidate the deep logic of patterning by oscillatory and sequential gene activities. Furthermore, in some of these organisms (e.g., the fruit fly Drosophila), a mode of AP patterning has evolved that seems not to overtly rely on oscillations or sequential gene activities, providing an opportunity to study the evolution of pattern formation mechanisms.
Collapse
Affiliation(s)
- Margarete Diaz-Cuadros
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Pathology, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
| | - Olivier Pourquié
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Pathology, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
- Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, United States of America
| | - Ezzat El-Sherif
- Division of Developmental Biology, Department of Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
47
|
Identification of Copy Number Variants in a Southern Chinese Cohort of Patients with Congenital Scoliosis. Genes (Basel) 2021; 12:genes12081213. [PMID: 34440387 PMCID: PMC8391542 DOI: 10.3390/genes12081213] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 08/01/2021] [Accepted: 08/03/2021] [Indexed: 12/15/2022] Open
Abstract
Congenital scoliosis (CS) is a lateral curvature of the spine resulting from congenital vertebral malformations (CVMs) and affects 0.5–1/1000 live births. The copy number variant (CNV) at chromosome 16p11.2 has been implicated in CVMs and recent studies identified a compound heterozygosity of 16p11.2 microdeletion and TBX6 variant/haplotype causing CS in multiple cohorts, which explains about 5–10% of the affected cases. Here, we studied the genetic etiology of CS by analyzing CNVs in a cohort of 67 patients with congenital hemivertebrae and 125 family controls. We employed both candidate gene and family-based approaches to filter CNVs called from whole exome sequencing data. This identified 12 CNVs in four scoliosis-associated genes (TBX6, NOTCH2, DSCAM, and SNTG1) as well as eight recessive and 64 novel rare CNVs in 15 additional genes. Some candidates, such as DHX40, NBPF20, RASA2, and MYSM1, have been found to be associated with syndromes with scoliosis or implicated in bone/spine development. In particular, the MYSM1 mutant mouse showed spinal deformities. Our findings suggest that, in addition to the 16p11.2 microdeletion, other CNVs are potentially important in predisposing to CS.
Collapse
|
48
|
Busby L, Steventon B. Tissue tectonics and the multi-scale regulation of developmental timing. Interface Focus 2021; 11:20200057. [PMID: 34055304 PMCID: PMC8086930 DOI: 10.1098/rsfs.2020.0057] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/22/2021] [Indexed: 12/17/2022] Open
Abstract
Development encompasses processes that occur at multiple length scales, including gene-regulatory interactions, cell movements and reorganization, cell signalling and growth. It is essential that the timing of events in all of these different processes is coordinated to generate well-patterned tissues and organs. However, how the timing of intrinsic cell state changes is coordinated with events occurring at the multi-tissue and whole-organism level is unknown. Here, we argue that an important mechanism that accounts for the integration of timing across levels of organization is provided by tissue tectonics, i.e. how morphogenetic events driving tissue shape changes result in the relative displacement of signalling and responding tissues and coordinate developmental timing across scales. In doing so, tissue tectonics provides a mechanism by which the cell specification events intrinsic to cells can be modulated by the temporal exposure to extracellular signals. This exposure is in turn regulated by higher-order properties of the embryo, such as their physical properties, rates of growth and the combination of dynamic cell behaviours, impacting tissue morphogenesis. Tissue tectonics creates a downward flow of information from higher to lower levels of biological organization, providing an instance of downward causation in development.
Collapse
Affiliation(s)
- Lara Busby
- Department of Genetics, University of Cambridge, Cambridge CB2 3EH, UK
| | | |
Collapse
|
49
|
Pantoja-Hernández J, Breña-Medina VF, Santillán M. Hybrid reaction-diffusion and clock-and-wavefront model for the arrest of oscillations in the somitogenesis segmentation clock. CHAOS (WOODBURY, N.Y.) 2021; 31:063107. [PMID: 34241318 DOI: 10.1063/5.0045460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 05/12/2021] [Indexed: 06/13/2023]
Abstract
The clock and wavefront paradigm is arguably the most widely accepted model for explaining the embryonic process of somitogenesis. According to this model, somitogenesis is based upon the interaction between a genetic oscillator, known as segmentation clock, and a differentiation wavefront, which provides the positional information indicating where each pair of somites is formed. Shortly after the clock and wavefront paradigm was introduced, Meinhardt presented a conceptually different mathematical model for morphogenesis in general, and somitogenesis in particular. Recently, Cotterell et al. [A local, self-organizing reaction-diffusion model can explain somite patterning in embryos, Cell Syst. 1, 257-269 (2015)] rediscovered an equivalent model by systematically enumerating and studying small networks performing segmentation. Cotterell et al. called it a progressive oscillatory reaction-diffusion (PORD) model. In the Meinhardt-PORD model, somitogenesis is driven by short-range interactions and the posterior movement of the front is a local, emergent phenomenon, which is not controlled by global positional information. With this model, it is possible to explain some experimental observations that are incompatible with the clock and wavefront model. However, the Meinhardt-PORD model has some important disadvantages of its own. Namely, it is quite sensitive to fluctuations and depends on very specific initial conditions (which are not biologically realistic). In this work, we propose an equivalent Meinhardt-PORD model and then amend it to couple it with a wavefront consisting of a receding morphogen gradient. By doing so, we get a hybrid model between the Meinhardt-PORD and the clock-and-wavefront ones, which overcomes most of the deficiencies of the two originating models.
Collapse
Affiliation(s)
- Jesús Pantoja-Hernández
- Centro de Investigación y de Estudios Avanzados del IPN, Unidad Monterrey, Vía del Conocimiento 201, Parque PIIT, 66628 Apodaca, NL, Mexico
| | | | - Moisés Santillán
- Centro de Investigación y de Estudios Avanzados del IPN, Unidad Monterrey, Vía del Conocimiento 201, Parque PIIT, 66628 Apodaca, NL, Mexico
| |
Collapse
|
50
|
Pitsidianaki I, Morgan J, Adams J, Campbell K. Mesenchymal-to-epithelial transitions require tissue-specific interactions with distinct laminins. THE JOURNAL OF CELL BIOLOGY 2021; 220:212200. [PMID: 34047771 PMCID: PMC8167899 DOI: 10.1083/jcb.202010154] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 03/29/2021] [Accepted: 05/10/2021] [Indexed: 01/06/2023]
Abstract
Mesenchymal-to-epithelial transition (MET) converts cells from migratory mesenchymal to polarized epithelial states. Despite its importance for both normal and pathological processes, very little is known about the regulation of MET in vivo. Here we exploit midgut morphogenesis in Drosophila melanogaster to investigate the mechanisms underlying MET. We show that down-regulation of the EMT transcription factor Serpent is required for MET, but not sufficient, as interactions with the surrounding mesoderm are also essential. We find that midgut MET relies on the secretion of specific laminins via the CopII secretory pathway from both mesoderm and midgut cells. We show that secretion of the laminin trimer containing the Wingblister α-subunit from the mesoderm is an upstream cue for midgut MET, leading to basal polarization of αPS1 integrin in midgut cells. Polarized αPS1 is required for the formation of a monolayered columnar epithelium and for the apical polarization of αPS3, Baz, and E-Cad. Secretion of a distinct LamininA-containing trimer from midgut cells is required to reinforce the localization of αPS1 basally, and αPS3 apically, for robust repolarization. Our data suggest that targeting these MET pathways, in conjunction with therapies preventing EMT, may present a two-pronged strategy toward blocking metastasis in cancer.
Collapse
Affiliation(s)
- Ioanna Pitsidianaki
- Department of Biomedical Science and Bateson Centre, The University of Sheffield, Sheffield, UK
| | - Jason Morgan
- Department of Biomedical Science and Bateson Centre, The University of Sheffield, Sheffield, UK
| | - Jamie Adams
- Department of Biomedical Science and Bateson Centre, The University of Sheffield, Sheffield, UK
| | - Kyra Campbell
- Department of Biomedical Science and Bateson Centre, The University of Sheffield, Sheffield, UK
| |
Collapse
|