1
|
Alhashmi M, Gremida AME, Maharana SK, Antonaci M, Kerr A, Fu S, Lunn S, Turner DA, Al-Maslamani NA, Liu K, Meschis MM, Sutherland H, Wilson P, Clegg P, Wheeler GN, van 't Hof RJ, Bou-Gharios G, Yamamoto K. Skeletal progenitor LRP1 deficiency causes severe and persistent skeletal defects with Wnt pathway dysregulation. Bone Res 2025; 13:17. [PMID: 39865089 PMCID: PMC11770177 DOI: 10.1038/s41413-024-00393-x] [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] [Academic Contribution Register] [Received: 03/29/2024] [Revised: 09/26/2024] [Accepted: 11/13/2024] [Indexed: 01/28/2025] Open
Abstract
Low-density lipoprotein receptor-related protein 1 (LRP1) is a multifunctional endocytic receptor whose dysfunction is linked to developmental dysplasia of the hip, osteoporosis and osteoarthritis. Our work addresses the critical question of how these skeletal pathologies emerge. Here, we show the abundant expression of LRP1 in skeletal progenitor cells at mouse embryonic stage E10.5 and onwards, especially in the perichondrium, the stem cell layer surrounding developing limbs essential for bone formation. Lrp1 deficiency in these stem cells causes joint fusion, malformation of cartilage/bone template and markedly delayed or lack of primary ossification. These abnormalities, which resemble phenotypes associated with Wnt signalling pathways, result in severe and persistent skeletal defects including a severe deficit in hip joint and patella, and markedly deformed and low-density long bones leading to dwarfism and impaired mobility. Mechanistically, we show that LRP1 regulates core non-canonical Wnt/planar cell polarity (PCP) components that may explain the malformation of long bones. LRP1 directly binds to Wnt5a, facilitates its cell-association and endocytic degradation and recycling. In the developing limbs, LRP1 partially colocalises with Wnt5a and its deficiency alters abundance and distribution of Wnt5a and Vangl2. Finally, using Xenopus as a model system, we show the regulatory role for LRP1 in Wnt/PCP signalling. We propose that in skeletal progenitors, LRP1 plays a critical role in formation and maturity of multiple bones and joints by regulating Wnt signalling, providing novel insights into the fundamental processes of morphogenesis and the emergence of skeletal pathologies.
Collapse
Affiliation(s)
- Mohammad Alhashmi
- Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, UK
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Abdulrahman M E Gremida
- Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, UK
| | - Santosh K Maharana
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, Norfolk, UK
| | - Marco Antonaci
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, Norfolk, UK
| | - Amy Kerr
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, Norfolk, UK
| | - Shijian Fu
- Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, UK
| | - Sharna Lunn
- Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, UK
| | - David A Turner
- Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, UK
| | - Noor A Al-Maslamani
- Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, UK
| | - Ke Liu
- Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, UK
| | - Maria M Meschis
- Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, UK
| | - Hazel Sutherland
- Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, UK
| | - Peter Wilson
- Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, UK
| | - Peter Clegg
- Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, UK
| | - Grant N Wheeler
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, Norfolk, UK
| | - Robert J van 't Hof
- Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, UK
- VANTHOF SCIENTIFIC, Torun, Poland
| | - George Bou-Gharios
- Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, UK
| | - Kazuhiro Yamamoto
- Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, UK.
| |
Collapse
|
2
|
Shitikov AD, Parshina EA, Zaraisky AG, Tereshina MB. An improved method for whole-mount in situ hybridization in regenerating tails of Xenopus laevis tadpoles. Front Cell Dev Biol 2024; 12:1487644. [PMID: 39717842 PMCID: PMC11663889 DOI: 10.3389/fcell.2024.1487644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/28/2024] [Accepted: 11/14/2024] [Indexed: 12/25/2024] Open
Abstract
Whole-mount in situ hybridization (WISH) is a widely used method that supports the concept of "seeing is believing" by enabling the visualization of gene expression patterns in whole-mount multicellular samples or sections. This technique is essential in the study of epimorphic regeneration in cold-blooded vertebrates, where complex three-dimensional organs such as tails, limbs, and eyes are completely restored after loss. The tadpoles of the frog X. laevis serve as a convenient model for studying regeneration, as they can regenerate their tails within a week after amputation. Modern high-throughput sequencing methods have identified various cell populations involved in the regeneration process and determined the repertoire of genes activated during this time. Specifically, a population of reparative myeloid cells expressing mmp9 as a marker gene has been shown to be crucial for the initial stages of tail regeneration in X. laevis tadpoles. The validation of these data and further examination using WISH offers the advantage of providing detailed information on the spatial and temporal dynamics of target gene expression levels. However, detecting mRNA by WISH can be challenging when mRNA levels are very low, transcripts are localized in hard-to-access areas, or tissue samples are prone to background staining, as is the case with X. laevis regenerating tail samples. Here, we describe additional treatments for regenerating tail samples that minimize background staining and enhance the visualization of cells containing target RNA through in situ hybridization. Using an optimized WISH protocol on X. laevis tadpole tail regenerates, we obtained novel data on the mmp9 expression pattern during the first day post-amputation at the regeneration-competent stage 40 and the regeneration-incompetent stage 47 (refractory period). The significant differences in the expression patterns indicate that mmp9 activity is positively correlated with regeneration competence.
Collapse
Affiliation(s)
- A. D. Shitikov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - E. A. Parshina
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - A. G. Zaraisky
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Department of Regenerative Medicine, Pirogov Russian National Research Medical University, Moscow, Russia
| | - M. B. Tereshina
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Department of Regenerative Medicine, Pirogov Russian National Research Medical University, Moscow, Russia
| |
Collapse
|
3
|
Wang H, Xie L, Guo H, Li L, Chen S, Fan Y, Tian J, Xu L, Kong X, Xuan A. m 1A demethylase Alkbh3 regulates neurogenesis through m 1A demethylation of Mmp15 mRNA. Cell Biosci 2024; 14:92. [PMID: 39004750 PMCID: PMC11246583 DOI: 10.1186/s13578-024-01275-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/12/2024] [Accepted: 07/10/2024] [Indexed: 07/16/2024] Open
Abstract
BACKGROUND N1-Methyladenosine (m1A) is an abundant modification of transcripts regulating mRNA structure and translation efficiency. However, the characteristics and biological functions of mRNA m1A modification in adult hippocampal neurogenesis remain enigmatic. RESULTS We found that m1A demethylase Alkbh3 was dramatically enriched in neurons and neuronal genesis. Functionally, depletion of Alkbh3 in neural stem cells (NSCs) significantly decreased m1A modification, neuronal differentiation and proliferation coupling with increasing gliogenesis, whereas overexpressing Alkbh3 facilitated neuronal differentiation and proliferation. Mechanistically, the m1A demethylation of Mmp15 mRNA by Alkbh3 improved its RNA stability and translational efficacy, which promoted neurogenesis. Therapeutically, the silencing of Alkbh3 reduced hippocampal neurogenesis and impaired spatial memory in the adult mice. CONCLUSIONS We reveal a novel function of m1A demethylation on Mmp15 mRNA in Alkbh3-mediated neurogenesis, which shed light on advancing Alkbh3 regulation of neurogenesis as a novel neurotherapeutic strategy.
Collapse
Affiliation(s)
- Huan Wang
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Linjie Xie
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Haomin Guo
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Lishi Li
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Shuwei Chen
- School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Ye Fan
- Scientific Research Center of Guangzhou Medical University, Guangzhou, 511436, China
| | - Jingyuan Tian
- Scientific Research Center of Guangzhou Medical University, Guangzhou, 511436, China
| | - Liping Xu
- Scientific Research Center of Guangzhou Medical University, Guangzhou, 511436, China.
- The Affiliated TCM Hospital of Guangzhou Medical University, Guangzhou, 510645, China.
| | - Xuejian Kong
- Qingyuan People's Hospital, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, 511518, China.
| | - Aiguo Xuan
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China.
- School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China.
- Scientific Research Center of Guangzhou Medical University, Guangzhou, 511436, China.
- Qingyuan People's Hospital, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, 511518, China.
| |
Collapse
|
4
|
de Oliveira SG, Kotowski N, Sampaio-Filho HR, Aguiar FHB, Dávila AMR, Jardim R. Metalloproteinases in Restorative Dentistry: An In Silico Study toward an Ideal Animal Model. Biomedicines 2023; 11:3042. [PMID: 38002041 PMCID: PMC10669239 DOI: 10.3390/biomedicines11113042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/09/2023] [Revised: 09/02/2023] [Accepted: 09/13/2023] [Indexed: 11/26/2023] Open
Abstract
In dentistry, various animal models are used to evaluate adhesive systems, dental caries and periodontal diseases. Metalloproteinases (MMPs) are enzymes that degrade collagen in the dentin matrix and are categorized in over 20 different classes. Collagenases and gelatinases are intrinsic constituents of the human dentin organic matrix fibrillar network and are the most abundant MMPs in this tissue. Understanding such enzymes' action on dentin is important in the development of approaches that could reduce dentin degradation and provide restorative procedures with extended longevity. This in silico study is based on dentistry's most used animal models and intends to search for the most suitable, evolutionarily close to Homo sapiens. We were able to retrieve 176,077 mammalian MMP sequences from the UniProt database. These sequences were manually curated through a three-step process. After such, the remaining 3178 sequences were aligned in a multifasta file and phylogenetically reconstructed using the maximum likelihood method. Our study inferred that the animal models most evolutionarily related to Homo sapiens were Orcytolagus cuniculus (MMP-1 and MMP-8), Canis lupus (MMP-13), Rattus norvegicus (MMP-2) and Orcytolagus cuniculus (MMP-9). Further research will be needed for the biological validation of our findings.
Collapse
Affiliation(s)
- Simone Gomes de Oliveira
- Piracicaba School of Dentistry, Campinas State University, Piracicaba 13414-903, SP, Brazil
- School of Dentistry, State University of Rio de Janeiro, Rio de Janeiro 20551-030, RJ, Brazil
| | - Nelson Kotowski
- Computational and Systems Biology Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, RJ, Brazil; (N.K.); (A.M.R.D.)
| | | | | | - Alberto Martín Rivera Dávila
- Computational and Systems Biology Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, RJ, Brazil; (N.K.); (A.M.R.D.)
| | - Rodrigo Jardim
- Computational and Systems Biology Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, RJ, Brazil; (N.K.); (A.M.R.D.)
| |
Collapse
|
5
|
Godden AM, Antonaci M, Wheeler GN. An Efficient CRISPR-Cas9 Method to Knock Out MiRNA Expression in Xenopus Tropicalis. Methods Mol Biol 2023; 2630:231-241. [PMID: 36689186 DOI: 10.1007/978-1-0716-2982-6_15] [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] [Academic Contribution Register] [Indexed: 01/24/2023]
Abstract
In recent years CRISPR-Cas9 knockouts (KO) have become increasingly utilized to study gene function. MicroRNAs (miRNAs) are short noncoding RNAs, 20-25 nucleotides long, which affect gene expression through posttranscriptional repression. As miRNAs are so small and due to the limitations of known PAM sequences, it is difficult to design CRISPR sgRNAs that reproducibly lead to a KO. We have therefore developed a novel approach using two guide RNAs to effectively "drop out" a miRNA. Validation of efficient CRISPR miRNA KO and phenotype analysis included use of q-RT-PCR and Sanger sequencing. To show specificity of the phenotype, we provide a protocol to use miRNA mimics to rescue the KO phenotype.
Collapse
Affiliation(s)
- Alice M Godden
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, UK
| | - Marco Antonaci
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, UK
| | - Grant N Wheeler
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, UK.
| |
Collapse
|
6
|
Godden AM, Antonaci M, Ward NJ, van der Lee M, Abu-Daya A, Guille M, Wheeler GN. An efficient miRNA knockout approach using CRISPR-Cas9 in Xenopus. Dev Biol 2022; 483:66-75. [PMID: 34968443 PMCID: PMC8865746 DOI: 10.1016/j.ydbio.2021.12.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/30/2021] [Revised: 12/15/2021] [Accepted: 12/23/2021] [Indexed: 12/14/2022]
Abstract
In recent years CRISPR-Cas9 knockouts (KO) have become increasingly ultilised to study gene function. MicroRNAs (miRNAs) are short non-coding RNAs, 20-22 nucleotides long, which affect gene expression through post-transcriptional repression. We previously identified miRNAs-196a and -219 as implicated in the development of Xenopus neural crest (NC). The NC is a multipotent stem-cell population, specified during early neurulation. Following EMT, NC cells migrate to various points in the developing embryo where they give rise to a number of tissues including parts of the peripheral nervous system, pigment cells and craniofacial skeleton. Dysregulation of NC development results in many diseases grouped under the term neurocristopathies. As miRNAs are so small, it is difficult to design CRISPR sgRNAs that reproducibly lead to a KO. We have therefore designed a novel approach using two guide RNAs to effectively 'drop out' a miRNA. We have knocked out miR-196a and miR-219 and compared the results to morpholino knockdowns (KD) of the same miRNAs. Validation of efficient CRISPR miRNA KO and phenotype analysis included use of whole-mount in situ hybridization of key NC and neural plate border markers such as Pax3, Xhe2, Sox10 and Snail2, q-RT-PCR and Sanger sequencing. To show specificity we have also rescued the knockout phenotype using miRNA mimics. MiRNA-219 and miR-196a KO's both show loss of NC, altered neural plate and hatching gland phenotypes. Tadpoles show gross craniofacial and pigment phenotypes.
Collapse
Affiliation(s)
- Alice M Godden
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, United Kingdom
| | - Marco Antonaci
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, United Kingdom
| | - Nicole J Ward
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, United Kingdom
| | - Michael van der Lee
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, United Kingdom
| | - Anita Abu-Daya
- King Henry Building, King Henry I St, Portsmouth, PO1 2DY, United Kingdom
| | - Matthew Guille
- King Henry Building, King Henry I St, Portsmouth, PO1 2DY, United Kingdom
| | - Grant N Wheeler
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, United Kingdom.
| |
Collapse
|
7
|
Pentagna N, Pinheiro da Costa T, Soares Dos Santos Cardoso F, Martins de Almeida F, Blanco Martinez AM, Abreu JG, Levin M, Carneiro K. Epigenetic control of myeloid cells behavior by Histone Deacetylase activity (HDAC) during tissue and organ regeneration in Xenopus laevis. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2021; 114:103840. [PMID: 32858087 DOI: 10.1016/j.dci.2020.103840] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 05/09/2020] [Revised: 08/18/2020] [Accepted: 08/18/2020] [Indexed: 06/11/2023]
Abstract
In the present work we have focused on the Histone Deacetylase (HDAC) control of myeloid cells behavior during Xenopus tail regeneration. Here we show that myeloid differentiation is crucial to modulate the regenerative ability of Xenopus tadpoles in a HDAC activity-dependent fashion. HDAC activity inhibition during the first wave of myeloid differentiation disrupted myeloid cells dynamics in the regenerative bud as well the mRNA expression pattern of myeloid markers, such as LURP, MPOX, Spib and mmp7. We also functionally bridge the spatial and temporal dynamics of lipid droplets, the main platform of lipid mediators synthesis in myeloid cells during the inflammatory response, and the regenerative ability of Xenopus tadpoles. In addition, we showed that 15-LOX activity is necessary during tail regeneration. Taken together our results support a role for the epigenetic control of myeloid behavior during tissue and organ regeneration, which may positively impact translational approaches for regenerative medicine.
Collapse
Affiliation(s)
- Nathalia Pentagna
- Instituto de Ciências Biomédicas, Universidade Federal Do Rio de Janeiro, Av. Carlos Chagas Filho 373 Bloco F Sala F2-01, Rio de Janeiro, 21941-902, Brazil; Programa de Pós-graduação Em Medicina (Anatomia Patológica), Faculdade de Medicina, Universidade Federal Do Rio de Janeiro, R. Prof. Rodolpho Paulo Rocco, 255, Rio de Janeiro, 21941-590, Brazil.
| | - Thayse Pinheiro da Costa
- Instituto de Ciências Biomédicas, Universidade Federal Do Rio de Janeiro, Av. Carlos Chagas Filho 373 Bloco F Sala F2-01, Rio de Janeiro, 21941-902, Brazil
| | - Fellipe Soares Dos Santos Cardoso
- Programa de Pós-graduação Em Medicina (Anatomia Patológica), Faculdade de Medicina, Universidade Federal Do Rio de Janeiro, R. Prof. Rodolpho Paulo Rocco, 255, Rio de Janeiro, 21941-590, Brazil.
| | - Fernanda Martins de Almeida
- Instituto de Ciências Biomédicas, Universidade Federal Do Rio de Janeiro, Av. Carlos Chagas Filho 373 Bloco F Sala F2-01, Rio de Janeiro, 21941-902, Brazil; Programa de Pós-graduação Em Medicina (Anatomia Patológica), Faculdade de Medicina, Universidade Federal Do Rio de Janeiro, R. Prof. Rodolpho Paulo Rocco, 255, Rio de Janeiro, 21941-590, Brazil.
| | - Ana Maria Blanco Martinez
- Programa de Pós-graduação Em Medicina (Anatomia Patológica), Faculdade de Medicina, Universidade Federal Do Rio de Janeiro, R. Prof. Rodolpho Paulo Rocco, 255, Rio de Janeiro, 21941-590, Brazil.
| | - José Garcia Abreu
- Instituto de Ciências Biomédicas, Universidade Federal Do Rio de Janeiro, Av. Carlos Chagas Filho 373 Bloco F Sala F2-01, Rio de Janeiro, 21941-902, Brazil.
| | - Michael Levin
- Allen Discovery Center, Tufts University, School of Arts and Science, Department of Biology, Suite, 4600, Medford, MA, United States.
| | - Katia Carneiro
- Instituto de Ciências Biomédicas, Universidade Federal Do Rio de Janeiro, Av. Carlos Chagas Filho 373 Bloco F Sala F2-01, Rio de Janeiro, 21941-902, Brazil; Programa de Pós-graduação Em Medicina (Anatomia Patológica), Faculdade de Medicina, Universidade Federal Do Rio de Janeiro, R. Prof. Rodolpho Paulo Rocco, 255, Rio de Janeiro, 21941-590, Brazil.
| |
Collapse
|
8
|
Gouignard N, Theveneau E, Saint-Jeannet JP. Dynamic expression of MMP28 during cranial morphogenesis. Philos Trans R Soc Lond B Biol Sci 2020; 375:20190559. [PMID: 32829678 DOI: 10.1098/rstb.2019.0559] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/22/2022] Open
Abstract
Matrix metalloproteinases (MMPs) are a large family of proteases comprising 24 members in vertebrates. They are well known for their extracellular matrix remodelling activity. MMP28 is the latest member of the family to be discovered. It is a secreted MMP involved in wound healing, immune system maturation, cell survival and migration. MMP28 is also expressed during embryogenesis in human and mouse. Here, we describe the detailed expression profile of MMP28 in Xenopus laevis embryos. We show that MMP28 is expressed maternally and accumulates at neurula and tail bud stages specifically in the cranial placode territories adjacent to migrating neural crest cells. As a secreted MMP, MMP28 may be required in neural crest-placode interactions. This article is part of a discussion meeting issue 'Contemporary morphogenesis'.
Collapse
Affiliation(s)
- Nadege Gouignard
- Centre de Biologie du Développement (CBD), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, France.,Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA
| | - Eric Theveneau
- Centre de Biologie du Développement (CBD), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Jean-Pierre Saint-Jeannet
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA
| |
Collapse
|
9
|
Tagliatela AC, Hempstead SC, Hibshman PS, Hockenberry MA, Brighton HE, Pecot CV, Bear JE. Coronin 1C inhibits melanoma metastasis through regulation of MT1-MMP-containing extracellular vesicle secretion. Sci Rep 2020; 10:11958. [PMID: 32686704 PMCID: PMC7371684 DOI: 10.1038/s41598-020-67465-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/21/2020] [Accepted: 06/05/2020] [Indexed: 02/06/2023] Open
Abstract
Coronin 1C is overexpressed in multiple tumors, leading to the widely held view that this gene drives tumor progression, but this hypothesis has not been rigorously tested in melanoma. Here, we combined a conditional knockout of Coronin 1C with a genetically engineered mouse model of PTEN/BRAF-driven melanoma. Loss of Coronin 1C in this model increases both primary tumor growth rates and distant metastases. Coronin 1C-null cells isolated from this model are more invasive in vitro and produce more metastatic lesions in orthotopic transplants than Coronin 1C-reexpressing cells due to the shedding of extracellular vesicles (EVs) containing MT1-MMP. Interestingly, these vesicles contain melanosome markers suggesting a melanoma-specific mechanism of EV release, regulated by Coronin 1C, that contributes to the high rates of metastasis in melanoma.
Collapse
Affiliation(s)
- Alicia C Tagliatela
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Stephanie C Hempstead
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Priya S Hibshman
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Max A Hockenberry
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Hailey E Brighton
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Chad V Pecot
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Division of Hematology and Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - James E Bear
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
10
|
Leonard CE, Taneyhill LA. The road best traveled: Neural crest migration upon the extracellular matrix. Semin Cell Dev Biol 2020; 100:177-185. [PMID: 31727473 PMCID: PMC7071992 DOI: 10.1016/j.semcdb.2019.10.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/05/2019] [Revised: 09/29/2019] [Accepted: 10/30/2019] [Indexed: 12/22/2022]
Abstract
Neural crest cells have the extraordinary task of building much of the vertebrate body plan, including the craniofacial cartilage and skeleton, melanocytes, portions of the heart, and the peripheral nervous system. To execute these developmental programs, stationary premigratory neural crest cells first acquire the capacity to migrate through an extensive process known as the epithelial-to-mesenchymal transition. Once motile, neural crest cells must traverse a complex environment consisting of other cells and the protein-rich extracellular matrix in order to get to their final destinations. Herein, we will highlight some of the main molecular machinery that allow neural crest cells to first exit the neuroepithelium and then later successfully navigate this intricate in vivo milieu. Collectively, these extracellular and intracellular factors mediate the appropriate migration of neural crest cells and allow for the proper development of the vertebrate embryo.
Collapse
Affiliation(s)
- Carrie E Leonard
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD 20742 USA.
| | - Lisa A Taneyhill
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD 20742 USA.
| |
Collapse
|
11
|
An in vivo brain-bacteria interface: the developing brain as a key regulator of innate immunity. NPJ Regen Med 2020; 5:2. [PMID: 32047653 PMCID: PMC7000827 DOI: 10.1038/s41536-020-0087-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/04/2019] [Accepted: 12/20/2019] [Indexed: 01/11/2023] Open
Abstract
Infections have numerous effects on the brain. However, possible roles of the brain in protecting against infection, and the developmental origin and role of brain signaling in immune response, are largely unknown. We exploited a unique Xenopus embryonic model to reveal control of innate immune response to pathogenic E. coli by the developing brain. Using survival assays, morphological analysis of innate immune cells and apoptosis, and RNA-seq, we analyzed combinations of infection, brain removal, and tail-regenerative response. Without a brain, survival of embryos injected with bacteria decreased significantly. The protective effect of the developing brain was mediated by decrease of the infection-induced damage and of apoptosis, and increase of macrophage migration, as well as suppression of the transcriptional consequences of the infection, all of which decrease susceptibility to pathogen. Functional and pharmacological assays implicated dopamine signaling in the bacteria–brain–immune crosstalk. Our data establish a model that reveals the very early brain to be a central player in innate immunity, identify the developmental origins of brain–immune interactions, and suggest several targets for immune therapies.
Collapse
|
12
|
Abstract
Background The study of the mechanisms controlling wound healing is an attractive area within the field of biology, with it having a potentially significant impact on the health sector given the current medical burden associated with healing in the elderly population. Healing is a complex process and includes many steps that are regulated by coding and noncoding RNAs, proteins and other molecules. Nitric oxide (NO) is one of these small molecule regulators and its function has already been associated with inflammation and angiogenesis during adult healing. Results Our results showed that NO is also an essential component during embryonic scarless healing and acts via a previously unknown mechanism. NO is mainly produced during the early phase of healing and it is crucial for the expression of genes associated with healing. However, we also observed a late phase of healing, which occurs for several hours after wound closure and takes place under the epidermis and includes tissue remodelling that is dependent on NO. We also found that the NO is associated with multiple cellular metabolic pathways, in particularly the glucose metabolism pathway. This is particular noteworthy as the use of NO donors have already been found to be beneficial for the treatment of chronic healing defects (including those associated with diabetes) and it is possible that its mechanism of action follows those observed during embryonic wound healing. Conclusions Our study describes a new role of NO during healing, which may potentially translate to improved therapeutic treatments, especially for individual suffering with problematic healing.
Collapse
|
13
|
Saide K, Sherwood V, Wheeler GN. Paracetamol-induced liver injury modelled in Xenopus laevis embryos. Toxicol Lett 2019; 302:83-91. [DOI: 10.1016/j.toxlet.2018.09.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/10/2018] [Revised: 09/19/2018] [Accepted: 09/28/2018] [Indexed: 01/25/2023]
|
14
|
Desanlis I, Felstead HL, Edwards DR, Wheeler GN. ADAMTS9, a member of the ADAMTS family, in Xenopus development. Gene Expr Patterns 2018; 29:72-81. [PMID: 29935379 PMCID: PMC6119763 DOI: 10.1016/j.gep.2018.06.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/25/2018] [Accepted: 06/19/2018] [Indexed: 11/09/2022]
Abstract
Extracellular matrix (ECM) remodeling by metalloproteinases is crucial during development. The ADAMTS (A Disintegrin and Metalloproteinase with Thrombospondin type I motifs) enzymes are secreted, multi-domain matrix-associated zinc metalloendopeptidases that have diverse roles in tissue morphogenesis and patho-physiological remodeling. The human family includes 19 members. In this study we identified the 19 members of the ADAMTS family in Xenopus laevis and Xenopus tropicalis. Gene identification and a phylogenetic study revealed strong conservation of the ADAMTS family and contributed to a better annotation of the Xenopus genomes. Expression of the entire ADAMTS family was studied from early stages to tadpole stages of Xenopus, and detailed analysis of ADAMTS9 revealed expression in many structures during organogenesis such as neural crest (NC) derivative tissues, the pronephros and the pancreas. Versican, a matrix component substrate of ADAMTS9 shows a similar expression pattern suggesting a role of ADAMTS9 in the remodeling of the ECM in these structures by degradation of versican.
Collapse
Affiliation(s)
- Ines Desanlis
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | - Hannah L Felstead
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | - Dylan R Edwards
- Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | - Grant N Wheeler
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK.
| |
Collapse
|
15
|
Neurocristopathies: New insights 150 years after the neural crest discovery. Dev Biol 2018; 444 Suppl 1:S110-S143. [PMID: 29802835 DOI: 10.1016/j.ydbio.2018.05.013] [Citation(s) in RCA: 114] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/29/2017] [Revised: 05/16/2018] [Accepted: 05/17/2018] [Indexed: 12/12/2022]
Abstract
The neural crest (NC) is a transient, multipotent and migratory cell population that generates an astonishingly diverse array of cell types during vertebrate development. These cells, which originate from the ectoderm in a region lateral to the neural plate in the neural fold, give rise to neurons, glia, melanocytes, chondrocytes, smooth muscle cells, odontoblasts and neuroendocrine cells, among others. Neurocristopathies (NCP) are a class of pathologies occurring in vertebrates, especially in humans that result from the abnormal specification, migration, differentiation or death of neural crest cells during embryonic development. Various pigment, skin, thyroid and hearing disorders, craniofacial and heart abnormalities, malfunctions of the digestive tract and tumors can also be considered as neurocristopathies. In this review we revisit the current classification and propose a new way to classify NCP based on the embryonic origin of the affected tissues, on recent findings regarding the molecular mechanisms that drive NC formation, and on the increased complexity of current molecular embryology techniques.
Collapse
|
16
|
Gouignard N, Andrieu C, Theveneau E. Neural crest delamination and migration: Looking forward to the next 150 years. Genesis 2018; 56:e23107. [PMID: 29675839 DOI: 10.1002/dvg.23107] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/16/2018] [Revised: 03/27/2018] [Accepted: 03/28/2018] [Indexed: 12/24/2022]
Abstract
Neural crest (NC) cells were described for the first time in 1868 by Wilhelm His. Since then, this amazing population of migratory stem cells has been intensively studied. It took a century to fully unravel their incredible abilities to contribute to nearly every organ of the body. Yet, our understanding of the cell and molecular mechanisms controlling their migration is far from complete. In this review, we summarize the current knowledge on epithelial-mesenchymal transition and collective behavior of NC cells and propose further stops at which the NC train might be calling in the near future.
Collapse
Affiliation(s)
- Nadège Gouignard
- Centre de Biologie du Développement, Centre de Biologie Intégrative, Université de Toulouse, CNRS, UPS, France
| | - Cyril Andrieu
- Centre de Biologie du Développement, Centre de Biologie Intégrative, Université de Toulouse, CNRS, UPS, France
| | - Eric Theveneau
- Centre de Biologie du Développement, Centre de Biologie Intégrative, Université de Toulouse, CNRS, UPS, France
| |
Collapse
|
17
|
Abstract
Wnt signalling regulates cardiogenesis during specification of heart tissue and the morphogenetic movements necessary to form the linear heart. Wnt11-mediated non-canonical signalling promotes early cardiac development whilst Wnt11-R, which is expressed later, also signals through the non-canonical pathway to promote heart development. It is unclear which Frizzled proteins mediate these interactions. Frizzled-7 (fzd7) is expressed during gastrulation in the mesodermal cells fated to become heart, and then in the primary heart field. This expression is complementary to the expression of wnt11 and wnt11-R. We further show co-localisation of fzd7 with other early- and late-heart-specific markers using double in situ hybridisation. We have used loss of function analysis to determine the role of fzd7 during heart development. Morpholino antisense oligonucleotide-mediated knockdown of Fzd7 results in effects on heart development, similar to that caused by Wnt11 loss of function. Surprisingly, overexpression of dominant-negative Fzd7 cysteine rich domain (Fzd7 CRD) results in a cardia bifida phenotype, similar to the loss of wnt11-R phenotype. Overexpression of Fzd7 and activation of non-canonical wnt signalling can rescue the effect of Fzd7 CRD. We propose that Fzd7 has an important role during Xenopus heart development. Summary: Wnt signalling has been shown to be important in heart development. Here, we demonstrate that the wnt receptor fzd7 is required in mediating these Wnt signals.
Collapse
Affiliation(s)
- Muhammad Abu-Elmagd
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, P.O. Box 80216 Jeddah 21589, Kingdom of Saudi Arabia.,School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | - Joanna Mulvaney
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | - Grant N Wheeler
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| |
Collapse
|
18
|
Abstract
Collective cell migration is the coordinated movement emerging from the interaction of at least two cells. In multicellular organisms, collective cell migration is ubiquitous. During development, embryonic cells often travel in numbers, whereas in adults, epithelial cells close wounds collectively. There is often a division of labour and two categories of cells have been proposed: leaders and followers. These two terms imply that followers are subordinated to leaders whose proposed broad range of actions significantly biases the direction of the group of cells towards a specific target. These two terms are also tied to topology. Leaders are at the front while followers are located behind them. Here, we review recent work on some of the main experimental models for collective cell migration, concluding that leader-follower terminology may not be the most appropriate. It appears that not all collectively migrating groups are driven by cells located at the front. Moreover, the qualities that define leaders (pathfinding, traction forces and matrix remodelling) are not specific to front cells. These observations indicate that the terms leaders and followers are not suited to every case. We think that it would be more accurate to dissociate the function of a cell from its position in the group. The position of cells can be precisely defined with respect to the direction of movement by purely topological terms such as "front" or "rear" cells. In addition, we propose the more ample and strictly functional definition of "steering cells" which are able to determine the directionality of movement for the entire group. In this context, a leader cell represents only a specific case in which a steering cell is positioned at the front of the group.
Collapse
Affiliation(s)
- Eric Theveneau
- Centre de Biologie du Développement (CBD), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, France
| | - Claudia Linker
- Randall Division of Cell & Molecular Biophysics, King's College London, London, UK
| |
Collapse
|
19
|
Paré JF, Martyniuk CJ, Levin M. Bioelectric regulation of innate immune system function in regenerating and intact Xenopus laevis. NPJ Regen Med 2017; 2:15. [PMID: 29302351 PMCID: PMC5677984 DOI: 10.1038/s41536-017-0019-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/19/2016] [Revised: 02/27/2017] [Accepted: 04/02/2017] [Indexed: 02/07/2023] Open
Abstract
Two key inputs that regulate regeneration are the function of the immune system, and spatial gradients of transmembrane potential (Vmem). Endogenous bioelectric signaling in somatic tissues during regenerative patterning is beginning to be understood, but its role in the context of immune response has never been investigated. Here, we show that Vmem levels modulate innate immunity activity in Xenopus laevis embryos. We developed an assay in which X. laevis embryos are infected with a uropathogenic microorganism, in the presence or absence of reagents that modify Vmem, prior to the ontogenesis of the adaptive immune system. General depolarization of the organism's Vmem by pharmacological or molecular genetic (ion channel misexpression) methods increased resistance to infection, while hyperpolarization made the embryos more susceptible to death by infection. Hyperpolarized specimens harbored a higher load of infectious microorganisms when compared to controls. We identified two mechanisms by which Vmem mediates immune function: serotonergic signaling involving melanocytes and an increase in the number of primitive myeloid cells. Bioinformatics analysis of genes whose transcription is altered by depolarization revealed a number of immune system targets consistent with mammalian data. Remarkably, amputation of the tail bud potentiates systemic resistance to infection by increasing the number of peripheral myeloid cells, revealing an interplay of regenerative response, innate immunity, and bioelectric regulation. Our study identifies bioelectricity as a new mechanism by which innate immune response can be regulated in the context of infection or regeneration. Vmem modulation using drugs already approved for human use could be exploited to improve resistance to infections in clinical settings.
Collapse
Affiliation(s)
- Jean-François Paré
- Biology Department, and Allen Discovery Center at Tufts, Tufts University, Medford, MA USA
| | - Christopher J. Martyniuk
- Center for Environmental and Human Toxicology and Department of Physiological Sciences, University of Florida Genetics Institute, College of Veterinary Medicine, University of Florida, Gainesville, FL USA
| | - Michael Levin
- Biology Department, and Allen Discovery Center at Tufts, Tufts University, Medford, MA USA
| |
Collapse
|
20
|
Naitoh H, Suganuma Y, Ueda Y, Sato T, Hiramuki Y, Fujisawa-Sehara A, Taketani S, Araki M. Upregulation of matrix metalloproteinase triggers transdifferentiation of retinal pigmented epithelial cells in Xenopus laevis: A Link between inflammatory response and regeneration. Dev Neurobiol 2017; 77:1086-1100. [PMID: 28371543 DOI: 10.1002/dneu.22497] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/01/2017] [Revised: 03/23/2017] [Accepted: 03/23/2017] [Indexed: 11/09/2022]
Abstract
In adult Xenopus eyes, when the whole retina is removed, retinal pigmented epithelial (RPE) cells become activated to be retinal stem cells and regenerate the whole retina. In the present study, using a tissue culture model, it was examined whether upregulation of matrix metalloproteinases (Mmps) triggers retinal regeneration. Soon after retinal removal, Xmmp9 and Xmmp18 were strongly upregulated in the tissues of the RPE and the choroid. In the culture, Mmp expression in the RPE cells corresponded with their migration from the choroid. A potent MMP inhibitor, 1,10-PNTL, suppressed RPE cell migration, proliferation, and formation of an epithelial structure in vitro. The mechanism involved in upregulation of Mmps was further investigated. After retinal removal, inflammatory cytokine genes, IL-1β and TNF-α, were upregulated both in vivo and in vitro. When the inflammation inhibitors dexamethasone or Withaferin A were applied in vitro, RPE cell migration was severely affected, suppressing transdifferentiation. These results demonstrate that Mmps play a pivotal role in retinal regeneration, and suggest that inflammatory cytokines trigger Mmp upregulation, indicating a direct link between the inflammatory reaction and retinal regeneration. © 2017 Wiley Periodicals, Inc. Develop Neurobiol 77: 1086-1100, 2017.
Collapse
Affiliation(s)
- Hanako Naitoh
- Developmental Neurobiology Laboratory, Department of Biological Sciences, Nara Women's University, Nara, 630-8506, Japan
| | - Yukari Suganuma
- Developmental Neurobiology Laboratory, Department of Biological Sciences, Nara Women's University, Nara, 630-8506, Japan
| | - Yoko Ueda
- Developmental Neurobiology Laboratory, Department of Biological Sciences, Nara Women's University, Nara, 630-8506, Japan
| | - Takahiko Sato
- Department of Growth Regulation, Institute of Frontier Medical Sciences, Kyoto University, 606-8585, Japan
| | - Yosuke Hiramuki
- Department of Growth Regulation, Institute of Frontier Medical Sciences, Kyoto University, 606-8585, Japan
| | - Atsuko Fujisawa-Sehara
- Department of Growth Regulation, Institute of Frontier Medical Sciences, Kyoto University, 606-8585, Japan
| | - Shigeru Taketani
- Department of Biotechnology, Kyoto Institute of Technology, Kyoto, 606-8585, Japan
| | - Masasuke Araki
- Developmental Neurobiology Laboratory, Department of Biological Sciences, Nara Women's University, Nara, 630-8506, Japan.,Unit of Neural Development and Regeneration, Department of Biology, Nara Medical University, Nara, 634-8521, Japan
| |
Collapse
|
21
|
Taneyhill LA, Schiffmacher AT. Should I stay or should I go? Cadherin function and regulation in the neural crest. Genesis 2017; 55. [PMID: 28253541 DOI: 10.1002/dvg.23028] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/13/2016] [Revised: 02/21/2017] [Accepted: 02/27/2017] [Indexed: 12/20/2022]
Abstract
Our increasing comprehension of neural crest cell development has reciprocally advanced our understanding of cadherin expression, regulation, and function. As a transient population of multipotent stem cells that significantly contribute to the vertebrate body plan, neural crest cells undergo a variety of transformative processes and exhibit many cellular behaviors, including epithelial-to-mesenchymal transition (EMT), motility, collective cell migration, and differentiation. Multiple studies have elucidated regulatory and mechanistic details of specific cadherins during neural crest cell development in a highly contextual manner. Collectively, these results reveal that gradual changes within neural crest cells are accompanied by often times subtle, yet important, alterations in cadherin expression and function. The primary focus of this review is to coalesce recent data on cadherins in neural crest cells, from their specification to their emergence as motile cells soon after EMT, and to highlight the complexities of cadherin expression beyond our current perceptions, including the hypothesis that the neural crest EMT is a transition involving a predominantly singular cadherin switch. Further advancements in genetic approaches and molecular techniques will provide greater opportunities to integrate data from various model systems in order to distinguish unique or overlapping functions of cadherins expressed at any point throughout the ontogeny of the neural crest.
Collapse
Affiliation(s)
- Lisa A Taneyhill
- Department of Animal and Avian Sciences, University of Maryland, College Park, Maryland, 20742
| | - Andrew T Schiffmacher
- Department of Animal and Avian Sciences, University of Maryland, College Park, Maryland, 20742
| |
Collapse
|
22
|
Thakur V, Bedogni B. The membrane tethered matrix metalloproteinase MT1-MMP at the forefront of melanoma cell invasion and metastasis. Pharmacol Res 2016; 111:17-22. [DOI: 10.1016/j.phrs.2016.05.019] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 04/17/2016] [Revised: 05/13/2016] [Accepted: 05/17/2016] [Indexed: 01/18/2023]
|
23
|
Hatch VL, Marin-Barba M, Moxon S, Ford CT, Ward NJ, Tomlinson ML, Desanlis I, Hendry AE, Hontelez S, van Kruijsbergen I, Veenstra GJC, Münsterberg AE, Wheeler GN. The positive transcriptional elongation factor (P-TEFb) is required for neural crest specification. Dev Biol 2016; 416:361-72. [PMID: 27343897 DOI: 10.1016/j.ydbio.2016.06.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/12/2015] [Revised: 05/06/2016] [Accepted: 06/08/2016] [Indexed: 12/31/2022]
Abstract
Regulation of gene expression at the level of transcriptional elongation has been shown to be important in stem cells and tumour cells, but its role in the whole animal is only now being fully explored. Neural crest cells (NCCs) are a multipotent population of cells that migrate during early development from the dorsal neural tube throughout the embryo where they differentiate into a variety of cell types including pigment cells, cranio-facial skeleton and sensory neurons. Specification of NCCs is both spatially and temporally regulated during embryonic development. Here we show that components of the transcriptional elongation regulatory machinery, CDK9 and CYCLINT1 of the P-TEFb complex, are required to regulate neural crest specification. In particular, we show that expression of the proto-oncogene c-Myc and c-Myc responsive genes are affected. Our data suggest that P-TEFb is crucial to drive expression of c-Myc, which acts as a 'gate-keeper' for the correct temporal and spatial development of the neural crest.
Collapse
Affiliation(s)
- Victoria L Hatch
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, UK
| | - Marta Marin-Barba
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, UK
| | - Simon Moxon
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, UK
| | - Christopher T Ford
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, UK
| | - Nicole J Ward
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, UK
| | - Matthew L Tomlinson
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, UK
| | - Ines Desanlis
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, UK
| | - Adam E Hendry
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, UK
| | - Saartje Hontelez
- Radboud University, Department of Molecular Developmental Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Ila van Kruijsbergen
- Radboud University, Department of Molecular Developmental Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Gert Jan C Veenstra
- Radboud University, Department of Molecular Developmental Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Andrea E Münsterberg
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, UK
| | - Grant N Wheeler
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, UK.
| |
Collapse
|
24
|
Macrì S, Simula L, Pellarin I, Pegoraro S, Onorati M, Sgarra R, Manfioletti G, Vignali R. Hmga2 is required for neural crest cell specification in Xenopus laevis. Dev Biol 2016; 411:25-37. [PMID: 26806704 DOI: 10.1016/j.ydbio.2016.01.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/07/2015] [Revised: 12/23/2015] [Accepted: 01/20/2016] [Indexed: 12/12/2022]
Abstract
HMGA proteins are small nuclear proteins that bind DNA by conserved AT-hook motifs, modify chromatin architecture and assist in gene expression. Two HMGAs (HMGA1 and HMGA2), encoded by distinct genes, exist in mammals and are highly expressed during embryogenesis or reactivated in tumour progression. We here addressed the in vivo role of Xenopus hmga2 in the neural crest cells (NCCs). We show that hmga2 is required for normal NCC specification and development. hmga2 knockdown leads to severe disruption of major skeletal derivatives of anterior NCCs. We show that, within the NCC genetic network, hmga2 acts downstream of msx1, and is required for msx1, pax3 and snail2 activities, thus participating at different levels of the network. Because of hmga2 early effects in NCC specification, the subsequent epithelial-mesenchymal transition (EMT) and migration of NCCs towards the branchial pouches are also compromised. Strictly paralleling results on embryos, interfering with Hmga2 in a breast cancer cell model for EMT leads to molecular effects largely consistent with those observed on NCCs. These data indicate that Hmga2 is recruited in key molecular events that are shared by both NCCs and tumour cells.
Collapse
Affiliation(s)
- Simone Macrì
- Dipartimento di Biologia, Unità di Biologia Cellulare e dello Sviluppo, Università degli Studi di Pisa, SS12 Abetone e Brennero 4, 56127 Pisa, Italy
| | - Luca Simula
- Dipartimento di Biologia, Unità di Biologia Cellulare e dello Sviluppo, Università degli Studi di Pisa, SS12 Abetone e Brennero 4, 56127 Pisa, Italy
| | - Ilenia Pellarin
- Dipartimento di Scienze della Vita, Università degli Studi di Trieste, via Giorgieri 5, 34127 Trieste, Italy
| | - Silvia Pegoraro
- Dipartimento di Scienze della Vita, Università degli Studi di Trieste, via Giorgieri 5, 34127 Trieste, Italy
| | - Marco Onorati
- Dipartimento di Biologia, Unità di Biologia Cellulare e dello Sviluppo, Università degli Studi di Pisa, SS12 Abetone e Brennero 4, 56127 Pisa, Italy
| | - Riccardo Sgarra
- Dipartimento di Scienze della Vita, Università degli Studi di Trieste, via Giorgieri 5, 34127 Trieste, Italy
| | - Guidalberto Manfioletti
- Dipartimento di Scienze della Vita, Università degli Studi di Trieste, via Giorgieri 5, 34127 Trieste, Italy
| | - Robert Vignali
- Dipartimento di Biologia, Unità di Biologia Cellulare e dello Sviluppo, Università degli Studi di Pisa, SS12 Abetone e Brennero 4, 56127 Pisa, Italy.
| |
Collapse
|
25
|
Agricola ZN, Jagpal AK, Allbee AW, Prewitt AR, Shifley ET, Rankin SA, Zorn AM, Kenny AP. Identification of genes expressed in the migrating primitive myeloid lineage of Xenopus laevis. Dev Dyn 2015; 245:47-55. [PMID: 26264370 DOI: 10.1002/dvdy.24314] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/06/2015] [Revised: 06/23/2015] [Accepted: 07/13/2015] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND During primitive hematopoiesis in Xenopus, cebpa and spib expressing myeloid cells emerge from the anterior ventral blood island. Primitive myeloid cells migrate throughout the embryo and are critical for immunity, healing, and development. Although definitive hematopoiesis has been studied extensively, molecular mechanisms leading to the migration of primitive myelocytes remain poorly understood. We hypothesized these cells have specific extracellular matrix modifying and cell motility gene expression. RESULTS In situ hybridization screens of transcripts expressed in Xenopus foregut mesendoderm at stage 23 identified seven genes with restricted expression in primitive myeloid cells: destrin; coronin actin binding protein, 1a; formin-like 1; ADAM metallopeptidase domain 28; cathepsin S; tissue inhibitor of metalloproteinase-1; and protein tyrosine phosphatase nonreceptor 6. A detailed in situ hybridization analysis revealed these genes are initially expressed in the aVBI but become dispersed throughout the embryo as the primitive myeloid cells become migratory, similar to known myeloid markers. Morpholino-mediated loss-of-function and mRNA-mediated gain-of-function studies revealed the identified genes are downstream of Spib.a and Cebpa, key transcriptional regulators of the myeloid lineage. CONCLUSIONS We have identified genes specifically expressed in migratory primitive myeloid progenitors, providing tools to study how different gene networks operate in these primitive myelocytes during development and immunity.
Collapse
Affiliation(s)
- Zachary N Agricola
- Perinatal Institute, Cincinnati Children's Hospital Research Foundation and Department of Pediatrics College of Medicine, University of Cincinnati, Cincinnati, Ohio.,Division of Neonatology, Cincinnati Children's Hospital Research Foundation and Department of Pediatrics College of Medicine, University of Cincinnati, Cincinnati, Ohio
| | - Amrita K Jagpal
- Perinatal Institute, Cincinnati Children's Hospital Research Foundation and Department of Pediatrics College of Medicine, University of Cincinnati, Cincinnati, Ohio.,Division of Neonatology, Cincinnati Children's Hospital Research Foundation and Department of Pediatrics College of Medicine, University of Cincinnati, Cincinnati, Ohio
| | - Andrew W Allbee
- Perinatal Institute, Cincinnati Children's Hospital Research Foundation and Department of Pediatrics College of Medicine, University of Cincinnati, Cincinnati, Ohio.,Division of Neonatology, Cincinnati Children's Hospital Research Foundation and Department of Pediatrics College of Medicine, University of Cincinnati, Cincinnati, Ohio
| | - Allison R Prewitt
- Perinatal Institute, Cincinnati Children's Hospital Research Foundation and Department of Pediatrics College of Medicine, University of Cincinnati, Cincinnati, Ohio.,Division of Neonatology, Cincinnati Children's Hospital Research Foundation and Department of Pediatrics College of Medicine, University of Cincinnati, Cincinnati, Ohio
| | - Emily T Shifley
- Perinatal Institute, Cincinnati Children's Hospital Research Foundation and Department of Pediatrics College of Medicine, University of Cincinnati, Cincinnati, Ohio.,Division of Developmental Biology, Cincinnati Children's Hospital Research Foundation and Department of Pediatrics College of Medicine, University of Cincinnati, Cincinnati, Ohio
| | - Scott A Rankin
- Perinatal Institute, Cincinnati Children's Hospital Research Foundation and Department of Pediatrics College of Medicine, University of Cincinnati, Cincinnati, Ohio.,Division of Developmental Biology, Cincinnati Children's Hospital Research Foundation and Department of Pediatrics College of Medicine, University of Cincinnati, Cincinnati, Ohio
| | - Aaron M Zorn
- Perinatal Institute, Cincinnati Children's Hospital Research Foundation and Department of Pediatrics College of Medicine, University of Cincinnati, Cincinnati, Ohio.,Division of Developmental Biology, Cincinnati Children's Hospital Research Foundation and Department of Pediatrics College of Medicine, University of Cincinnati, Cincinnati, Ohio
| | - Alan P Kenny
- Perinatal Institute, Cincinnati Children's Hospital Research Foundation and Department of Pediatrics College of Medicine, University of Cincinnati, Cincinnati, Ohio.,Division of Neonatology, Cincinnati Children's Hospital Research Foundation and Department of Pediatrics College of Medicine, University of Cincinnati, Cincinnati, Ohio
| |
Collapse
|
26
|
Ahmed A, Ward NJ, Moxon S, Lopez-Gomollon S, Viaut C, Tomlinson ML, Patrushev I, Gilchrist MJ, Dalmay T, Dotlic D, Münsterberg AE, Wheeler GN. A Database of microRNA Expression Patterns in Xenopus laevis. PLoS One 2015; 10:e0138313. [PMID: 26506012 PMCID: PMC4624429 DOI: 10.1371/journal.pone.0138313] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/07/2015] [Accepted: 08/28/2015] [Indexed: 12/31/2022] Open
Abstract
MicroRNAs (miRNAs) are short, non-coding RNAs around 22 nucleotides long. They inhibit gene expression either by translational repression or by causing the degradation of the mRNAs they bind to. Many are highly conserved amongst diverse organisms and have restricted spatio-temporal expression patterns during embryonic development where they are thought to be involved in generating accuracy of developmental timing and in supporting cell fate decisions and tissue identity. We determined the expression patterns of 180 miRNAs in Xenopus laevis embryos using LNA oligonucleotides. In addition we carried out small RNA-seq on different stages of early Xenopus development, identified 44 miRNAs belonging to 29 new families and characterized the expression of 5 of these. Our analyses identified miRNA expression in many organs of the developing embryo. In particular a large number were expressed in neural tissue and in the somites. Surprisingly none of the miRNAs we have looked at show expression in the heart. Our results have been made freely available as a resource in both XenMARK and Xenbase.
Collapse
Affiliation(s)
- Ayisha Ahmed
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, United Kingdom
| | - Nicole J. Ward
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, United Kingdom
| | - Simon Moxon
- The Genome Analysis Centre (TGAC), Norwich Research Park, Norwich, NR4 7UH, United Kingdom
| | - Sara Lopez-Gomollon
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, United Kingdom
| | - Camille Viaut
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, United Kingdom
| | - Matthew L. Tomlinson
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, United Kingdom
| | - Ilya Patrushev
- The Francis Crick Institute, Mill Hill Laboratory, The Ridgeway, Mill Hill, London, NW7 1AA, United Kingdom
| | - Michael J. Gilchrist
- The Francis Crick Institute, Mill Hill Laboratory, The Ridgeway, Mill Hill, London, NW7 1AA, United Kingdom
| | - Tamas Dalmay
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, United Kingdom
| | - Dario Dotlic
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, United Kingdom
| | - Andrea E. Münsterberg
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, United Kingdom
| | - Grant N. Wheeler
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, United Kingdom
- * E-mail:
| |
Collapse
|
27
|
Abstract
Some organisms have a remarkable ability to heal wounds without scars and to regenerate complex tissues following injury. By gaining a more complete understanding of the biological mechanisms that promote scar-free healing and tissue regeneration, it is hoped that novel treatments that can enhance the healing and regenerative capacity of human patients can be found. In the present article, we briefly examine the genetic, molecular and cellular mechanisms underlying the regeneration of the Xenopus tadpole tail.
Collapse
|
28
|
Ma J, Tang X, Wong P, Jacobs B, Borden EC, Bedogni B. Noncanonical activation of Notch1 protein by membrane type 1 matrix metalloproteinase (MT1-MMP) controls melanoma cell proliferation. J Biol Chem 2014; 289:8442-9. [PMID: 24492617 DOI: 10.1074/jbc.m113.516039] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/06/2023] Open
Abstract
Notch1 is an evolutionarily conserved signaling molecule required for stem cell maintenance that is inappropriately reactivated in several cancers. We have previously shown that melanomas reactivate Notch1 and require its function for growth and survival. However, no Notch1-activating mutations have been observed in melanoma, suggesting the involvement of other activating mechanisms. Notch1 activation requires two cleavage steps: first by a protease and then by γ-secretase, which releases the active intracellular domain (Notch1(NIC)). Interestingly, although ADAM10 and -17 are generally accepted as the proteases responsible of Notch1 cleavage, here we show that MT1-MMP, a membrane-tethered matrix metalloproteinase involved in the pathogenesis of a number of tumors, is a novel protease required for the cleavage of Notch1 in melanoma cells. We find that active Notch1 and MT1-MMP expression correlate significantly in over 70% of melanoma tumors and 80% of melanoma cell lines, whereas such correlation does not exist between Notch1(NIC) and ADAM10 or -17. Modulation of MT1-MMP expression in melanoma cells affects Notch1 cleavage, whereas MT1-MMP expression in ADAM10/17 double knock-out fibroblasts restores the processing of Notch1, indicating that MT1-MMP is sufficient to promote Notch1 activation independently of the canonical proteases. Importantly, we find that MT1-MMP interacts with Notch1 at the cell membrane, supporting a potential direct cleavage mechanism of MT1-MMP on Notch1, and that MT1-MMP-dependent activation of Notch1 sustains melanoma cell growth. Together, the data highlight a novel mechanism of activation of Notch1 in melanoma cells and identify Notch1 as a new MT1-MMP substrate that plays important biological roles in melanoma.
Collapse
Affiliation(s)
- Jun Ma
- From the Department of Biochemistry, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106 and
| | | | | | | | | | | |
Collapse
|
29
|
Shaverdashvili K, Wong P, Ma J, Zhang K, Osman I, Bedogni B. MT1-MMP modulates melanoma cell dissemination and metastasis through activation of MMP2 and RAC1. Pigment Cell Melanoma Res 2014; 27:287-96. [PMID: 24387669 DOI: 10.1111/pcmr.12201] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/26/2013] [Accepted: 12/02/2013] [Indexed: 12/11/2022]
Abstract
Metastatic melanoma remains the deadliest of all skin cancers with a survival rate at five years of less than 15%. MT1-MMP is a membrane-associated matrix metalloproteinase that controls pericellular proteolysis and is an important, invasion-promoting, pro-tumorigenic MMP in cancer. We show that deregulation of MT1-MMP expression happens as early as the transition from nevus to primary melanoma and continues to increase during melanoma progression. Furthermore, MT1-MMP expression is associated with poor melanoma patient outcome, underscoring a pivotal role of MT1-MMP in melanoma pathogenesis. We demonstrate that MT1-MMP is directly required for melanoma cells to metastasize, as cells deprived of MT1-MMP fail to form distant metastasis in an orthotopic mouse melanoma model. We show that MT1-MMP affects cell invasion by activating its target MMP2. Importantly, we demonstrate, for the first time, that activation of MMP2 by MT1-MMP is required to sustain RAC1 activity and promote MT1-MMP-dependent cell motility. These data highlight a novel MT1-MMP/MMP2/RAC1 signaling axis in melanoma that may represent an intriguing molecular target for the treatment of invasive melanoma.
Collapse
Affiliation(s)
- Khvaramze Shaverdashvili
- Department of Biochemistry, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | | | | | | | | | | |
Collapse
|
30
|
Christian L, Bahudhanapati H, Wei S. Extracellular metalloproteinases in neural crest development and craniofacial morphogenesis. Crit Rev Biochem Mol Biol 2013; 48:544-60. [PMID: 24066766 DOI: 10.3109/10409238.2013.838203] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 02/06/2023]
Abstract
The neural crest (NC) is a population of migratory stem/progenitor cells that is found in early vertebrate embryos. NC cells are induced during gastrulation, and later migrate to multiple destinations and contribute to many types of cells and tissues, such as craniofacial structures, cardiac tissues, pigment cells and the peripheral nervous system. Recently, accumulating evidence suggests that many extracellular metalloproteinases, including matrix metalloproteinases (MMPs), a disintegrin and metalloproteinases (ADAMs), and ADAMs with thrombospondin motifs (ADAMTSs), play important roles in various stages of NC development. Interference with metalloproteinase functions often causes defects in craniofacial structures, as well as in other cells and tissues that are contributed by NC cells, in humans and other vertebrates. In this review, we summarize the current state of the field concerning the roles of these three families of metalloproteinases in NC development and related tissue morphogenesis, with a special emphasis on craniofacial morphogenesis.
Collapse
Affiliation(s)
- Laura Christian
- Department of Biology, West Virginia University , Morgantown, WV , USA
| | | | | |
Collapse
|
31
|
Patterson RA, Cavanaugh AM, Cantemir V, Brauer PR, Reedy MV. MT2-MMP expression during early avian morphogenesis. Anat Rec (Hoboken) 2012; 296:64-70. [PMID: 23161772 DOI: 10.1002/ar.22618] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/08/2012] [Accepted: 09/20/2012] [Indexed: 12/18/2022]
Abstract
Membrane-type 2 matrix metalloproteinase (MT2-MMP; also called MMP15) is a membrane-bound protease that degrades extracellular matrix and activates proMMPs such as proMMP-2. MMP-2 expression in avian embryos is well documented, but it is not clear how proMMP-2 is activated during avian embryogenesis. Herein, we report that MT2-MMP mRNA is expressed in several tissues including the neural folds and epidermal ectoderm, intermediate mesoderm, pharyngeal arches, limb buds, and dermis. Several, but not all, of these tissues are known to express MMP-2. These observations suggest MT2-MMP may play a role during embryonic development not only through its own proteolytic activity but also by activating proMMP-2.
Collapse
|
32
|
McKeown SJ, Wallace AS, Anderson RB. Expression and function of cell adhesion molecules during neural crest migration. Dev Biol 2012; 373:244-57. [PMID: 23123967 DOI: 10.1016/j.ydbio.2012.10.028] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/08/2012] [Revised: 10/18/2012] [Accepted: 10/25/2012] [Indexed: 01/13/2023]
Abstract
Neural crest cells are highly migratory cells that give rise to many derivatives including peripheral ganglia, craniofacial structures and melanocytes. Neural crest cells migrate along defined pathways to their target sites, interacting with each other and their environment as they migrate. Cell adhesion molecules are critical during this process. In this review we discuss the expression and function of cell adhesion molecules during the process of neural crest migration, in particular cadherins, integrins, members of the immunoglobulin superfamily of cell adhesion molecules, and the proteolytic enzymes that cleave these cell adhesion molecules. The expression and function of these cell adhesion molecules and proteases are compared across neural crest emigrating from different axial levels, and across different species of vertebrates.
Collapse
Affiliation(s)
- Sonja J McKeown
- Department of Anatomy and Neuroscience, University of Melbourne, 3010 VIC, Australia.
| | | | | |
Collapse
|
33
|
Monsonego-Ornan E, Kosonovsky J, Bar A, Roth L, Fraggi-Rankis V, Simsa S, Kohl A, Sela-Donenfeld D. Matrix metalloproteinase 9/gelatinase B is required for neural crest cell migration. Dev Biol 2012; 364:162-77. [DOI: 10.1016/j.ydbio.2012.01.028] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/25/2011] [Revised: 01/30/2012] [Accepted: 01/31/2012] [Indexed: 11/27/2022]
|
34
|
Regulation of PP2A activity by Mid1 controls cranial neural crest speed and gangliogenesis. Mech Dev 2012; 128:560-76. [DOI: 10.1016/j.mod.2012.01.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/12/2011] [Revised: 01/05/2012] [Accepted: 01/11/2012] [Indexed: 12/22/2022]
|
35
|
Love NR, Chen Y, Bonev B, Gilchrist MJ, Fairclough L, Lea R, Mohun TJ, Paredes R, Zeef LAH, Amaya E. Genome-wide analysis of gene expression during Xenopus tropicalis tadpole tail regeneration. BMC DEVELOPMENTAL BIOLOGY 2011; 11:70. [PMID: 22085734 PMCID: PMC3247858 DOI: 10.1186/1471-213x-11-70] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Academic Contribution Register] [Received: 08/21/2011] [Accepted: 11/15/2011] [Indexed: 01/08/2023]
Abstract
Background The molecular mechanisms governing vertebrate appendage regeneration remain poorly understood. Uncovering these mechanisms may lead to novel therapies aimed at alleviating human disfigurement and visible loss of function following injury. Here, we explore tadpole tail regeneration in Xenopus tropicalis, a diploid frog with a sequenced genome. Results We found that, like the traditionally used Xenopus laevis, the Xenopus tropicalis tadpole has the capacity to regenerate its tail following amputation, including its spinal cord, muscle, and major blood vessels. We examined gene expression using the Xenopus tropicalis Affymetrix genome array during three phases of regeneration, uncovering more than 1,000 genes that are significantly modulated during tail regeneration. Target validation, using RT-qPCR followed by gene ontology (GO) analysis, revealed a dynamic regulation of genes involved in the inflammatory response, intracellular metabolism, and energy regulation. Meta-analyses of the array data and validation by RT-qPCR and in situ hybridization uncovered a subset of genes upregulated during the early and intermediate phases of regeneration that are involved in the generation of NADP/H, suggesting that these pathways may be important for proper tail regeneration. Conclusions The Xenopus tropicalis tadpole is a powerful model to elucidate the genetic mechanisms of vertebrate appendage regeneration. We have produced a novel and substantial microarray data set examining gene expression during vertebrate appendage regeneration.
Collapse
Affiliation(s)
- Nick R Love
- Faculty of Life Sciences, University of Manchester, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Theveneau E, Mayor R. Collective cell migration of the cephalic neural crest: The art of integrating information. Genesis 2011; 49:164-76. [DOI: 10.1002/dvg.20700] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/05/2010] [Revised: 11/30/2010] [Accepted: 12/04/2010] [Indexed: 02/03/2023]
|
37
|
Malloch EL, Perry KJ, Fukui L, Johnson VR, Wever J, Beck CW, King MW, Henry JJ. Gene expression profiles of lens regeneration and development in Xenopus laevis. Dev Dyn 2009; 238:2340-56. [PMID: 19681139 PMCID: PMC2773617 DOI: 10.1002/dvdy.21998] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/17/2023] Open
Abstract
Seven hundred and thirty-four unique genes were recovered from a cDNA library enriched for genes up-regulated during the process of lens regeneration in the frog Xenopus laevis. The sequences represent transcription factors, proteins involved in RNA synthesis/processing, components of prominent cell signaling pathways, genes involved in protein processing, transport, and degradation (e.g., the ubiquitin/proteasome pathway), matrix metalloproteases (MMPs), as well as many other proteins. The findings implicate specific signal transduction pathways in the process of lens regeneration, including the FGF, TGF-beta, MAPK, Retinoic acid, Wnt, and hedgehog signaling pathways, which are known to play important roles in eye/lens development and regeneration in various systems. In situ hybridization revealed that the majority of genes recovered are expressed during embryogenesis, including in eye tissues. Several novel genes specifically expressed in lenses were identified. The suite of genes was compared to those up-regulated in other regenerating tissues/organisms, and a small degree of overlap was detected.
Collapse
Affiliation(s)
- Erica L. Malloch
- University of Illinois, Department of Cell & Developmental Biology, 601 S. Goodwin Ave. Urbana, IL 61801
| | - Kimberly J. Perry
- University of Illinois, Department of Cell & Developmental Biology, 601 S. Goodwin Ave. Urbana, IL 61801
| | - Lisa Fukui
- University of Illinois, Department of Cell & Developmental Biology, 601 S. Goodwin Ave. Urbana, IL 61801
| | - Verity R. Johnson
- University of Illinois, Department of Cell & Developmental Biology, 601 S. Goodwin Ave. Urbana, IL 61801
| | - Jason Wever
- University of Illinois, Department of Cell & Developmental Biology, 601 S. Goodwin Ave. Urbana, IL 61801
| | - Caroline W. Beck
- University of Otago, Department of Zoology, 340 Great King Street, Dunedin, New Zealand
| | - Michael W. King
- Indiana University School of Medicine and Center for Regenerative Biology and Medicine, Terre Haute, IN 47809
| | - Jonathan J. Henry
- University of Illinois, Department of Cell & Developmental Biology, 601 S. Goodwin Ave. Urbana, IL 61801
| |
Collapse
|
38
|
Garcia-Morales C, Liu CH, Abu-Elmagd M, Hajihosseini MK, Wheeler GN. Frizzled-10 promotes sensory neuron development in Xenopus embryos. Dev Biol 2009; 335:143-55. [PMID: 19716814 DOI: 10.1016/j.ydbio.2009.08.021] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/12/2009] [Revised: 08/20/2009] [Accepted: 08/21/2009] [Indexed: 12/23/2022]
Abstract
Formation of the vertebrate nervous system requires coordinated cell-cell interactions, intracellular signalling events, gene transcription, and morphogenetic cell movements. Wnt signalling has been involved in regulating a wide variety of biological processes such as embryonic patterning, cell proliferation, cell polarity, motility, and the specification of cell fate. Wnt ligands associate with their receptors, members of the frizzled family (Fz). In Xenopus, five members of the frizzled family are expressed in the early nervous system. We have investigated the role of Xenopus frizzled-10 (Fz10) in neural development. We show that Fz10 is expressed in the dorsal neural ectoderm and neural folds in the region where primary sensory neurons develop. Fz10 mediates canonical Wnt signalling and interacts with Wnt1 and Wnt8 but not Wnt3a as shown in synergy assays. We find that Fz10 is required for the late stages of sensory neuron differentiation. Overexpression of Fz10 in Xenopus leads to an increase in the number of sensory neurons. Loss of Fz10 function using morpholinos inhibits the development of sensory neurons in Xenopus at later stages of neurogenesis and this can be rescued by co-injection of modified Fz10B and beta-catenin. In mouse P19 cells induced by retinoic acid to undergo neural differentiation, overexpression of Xenopus Fz10 leads to an increase in the number of neurons generated while siRNA knockdown of endogenous mouse Fz10 inhibits neurogenesis. Thus we propose Fz10 mediates Wnt1 signalling to determine sensory neural differentiation in Xenopus in vivo and in mouse cell culture.
Collapse
|
39
|
Tomlinson ML, Guan P, Morris RJ, Fidock MD, Rejzek M, Garcia-Morales C, Field RA, Wheeler GN. A chemical genomic approach identifies matrix metalloproteinases as playing an essential and specific role in Xenopus melanophore migration. ACTA ACUST UNITED AC 2009; 16:93-104. [PMID: 19171309 DOI: 10.1016/j.chembiol.2008.12.005] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/04/2008] [Revised: 12/10/2008] [Accepted: 12/12/2008] [Indexed: 01/27/2023]
Abstract
To dissect the function of matrix metalloproteinases (MMPs) involved in cellular migration in vivo, we undertook both a forward chemical genomic screen and a functional approach to discover modulators of melanophore (pigment cell) migration in Xenopus laevis. We identified the 8-quinolinol derivative NSC 84093 as affecting melanophore migration in the developing embryo and have shown it to act as a MMP inhibitor. Potential targets of NSC 84093 investigated include MMP-14 and MMP-2. MMP-14 is expressed in migrating neural crest cells from which melanophores are derived. MMP-2 is expressed at the relevant time of development and in a pattern that suggests it contributes to melanophore migration. Morpholino-mediated knockdown of both MMPs demonstrates they play a key role in melanophore migration and partially phenocopy the effect of NSC 84093.
Collapse
|
40
|
Fu L, Das B, Mathew S, Shi YB. Genome-wide identification of Xenopus matrix metalloproteinases: conservation and unique duplications in amphibians. BMC Genomics 2009; 10:81. [PMID: 19222855 PMCID: PMC2656525 DOI: 10.1186/1471-2164-10-81] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/29/2008] [Accepted: 02/17/2009] [Indexed: 01/17/2023] Open
Abstract
Background Matrix metalloproteinases (MMPs) are members of the superfamily of Zn2+ dependent extracellular or membrane-bound endopeptidases which have been implicated to play critical roles in vertebrate development and human pathogenesis. A number of MMP genes have been found to be upregulated in some or all organs during frog metamorphosis, suggesting that different MMPs may have different functions in various organs/tissues. The recent advances in EST (expressed sequence tag) sequencing and the completion of the genome of Xenopus (X.) tropicalis prompted us to systematically analyze the existence of MMPs in the Xenopus genome. Results We examined X. laevis and X. tropicalis ESTs and genomic sequences for MMPs and obtained likely homologs for 20 out of the 25 MMPs known in higher vertebrates. Four of the five missing MMPs, i.e. MMPs 8, 10, 12 and 27, were all encoded on human Chromosome 11 and the other missing MMP, MMP22 (a chicken MMP), was also absent in human genome. In addition, we identified several novel MMPs which appears to be derived from unique duplications over evolution, are present in the genomes of both Xenopus species. Conclusion We identified the homologs of most of the mammalian MMPs in Xenopus and discovered a number of novel MMPs. Our results suggest that MMP genes undergo dynamic changes over evolution. It will be of interest in the future to investigate whether MMP expression and functions during vertebrate development are conserved. The sequence information reported here should facilitate such an endeavor in the near future.
Collapse
Affiliation(s)
- Liezhen Fu
- Section on Molecular Morphogenesis, PCRM, NICHD, NIH, Bethesda, MD 20892, USA.
| | | | | | | |
Collapse
|
41
|
Slater BJ, Liu KJ, Kwan MD, Quarto N, Longaker MT. Cranial osteogenesis and suture morphology in Xenopus laevis: a unique model system for studying craniofacial development. PLoS One 2009; 4:e3914. [PMID: 19156194 PMCID: PMC2615207 DOI: 10.1371/journal.pone.0003914] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/21/2008] [Accepted: 11/13/2008] [Indexed: 11/18/2022] Open
Abstract
Background The tremendous diversity in vertebrate skull formation illustrates the range of forms and functions generated by varying genetic programs. Understanding the molecular basis for this variety may provide us with insights into mechanisms underlying human craniofacial anomalies. In this study, we provide evidence that the anuran Xenopus laevis can be developed as a simplified model system for the study of cranial ossification and suture patterning. The head structures of Xenopus undergo dramatic remodelling during metamorphosis; as a result, tadpole morphology differs greatly from the adult bony skull. Because of the extended larval period in Xenopus, the molecular basis of these alterations has not been well studied. Methodology/Principal Findings We examined late larval, metamorphosing, and post-metamorphosis froglet stages in intact and sectioned animals. Using micro-computed tomography (μCT) and tissue staining of the frontoparietal bone and surrounding cartilage, we observed that bone formation initiates from lateral ossification centers, proceeding from posterior-to-anterior. Histological analyses revealed midline abutting and posterior overlapping sutures. To determine the mechanisms underlying the large-scale cranial changes, we examined proliferation, apoptosis, and proteinase activity during remodelling of the skull roof. We found that tissue turnover during metamorphosis could be accounted for by abundant matrix metalloproteinase (MMP) activity, at least in part by MMP-1 and -13. Conclusion A better understanding of the dramatic transformation from cartilaginous head structures to bony skull during Xenopus metamorphosis may provide insights into tissue remodelling and regeneration in other systems. Our studies provide some new molecular insights into this process.
Collapse
Affiliation(s)
- Bethany J. Slater
- Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Hagey Laboratory for Pediatric Regenerative Medicine, Stanford, California, United States of America
| | - Karen J. Liu
- Department of Craniofacial Development, King's College London, London, United Kingdom
- * E-mail: (KJL); (MTL)
| | - Matthew D. Kwan
- Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Hagey Laboratory for Pediatric Regenerative Medicine, Stanford, California, United States of America
| | - Natalina Quarto
- Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Hagey Laboratory for Pediatric Regenerative Medicine, Stanford, California, United States of America
- Department of Structural and Functional Biology, University of Naples Federico II Complesso M. S. Angelo, Napoli, Italy
| | - Michael T. Longaker
- Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Hagey Laboratory for Pediatric Regenerative Medicine, Stanford, California, United States of America
- Department of Surgery, Stanford University School of Medicine, Hagey Laboratory for Pediatric Regenerative Medicine, Stanford, California, United States of America
- * E-mail: (KJL); (MTL)
| |
Collapse
|
42
|
Tomlinson ML, Rejzek M, Fidock M, Field RA, Wheeler GN. Chemical genomics identifies compounds affecting Xenopus laevis pigment cell development. MOLECULAR BIOSYSTEMS 2009; 5:376-84. [DOI: 10.1039/b818695b] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 11/21/2022]
|
43
|
Tomlinson ML, Garcia-Morales C, Abu-Elmagd M, Wheeler GN. Three matrix metalloproteinases are required in vivo for macrophage migration during embryonic development. Mech Dev 2008; 125:1059-70. [PMID: 18684398 DOI: 10.1016/j.mod.2008.07.005] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/24/2008] [Revised: 07/08/2008] [Accepted: 07/09/2008] [Indexed: 11/19/2022]
Abstract
Macrophages are essential in development, repair and pathology of a variety of tissues via their roles in tissue remodelling, wound healing and inflammation. These biological functions are also associated with a number of human diseases, for example tumour associated macrophages have well defined functions in cancer progression. Xenopus embryonic macrophages arise from a haematopoietic stem cell population by direct differentiation and act as the main mechanism of host defence, before lymphoid cells and a circulatory system have developed. This function is conserved in mouse and human development. Macrophages express a number of matrix metalloproteinases (MMPs), which are central to their function. MMPs are a large family of zinc-dependent endoproteases with multiple roles in extracellular matrix remodelling and the modulation of signalling pathways. We have previously shown MMP-7 to be expressed by Xenopus embryonic macrophages. Here we investigate the role of MMP-7 and two other MMPs (MMP-18 and MMP-9) that are also expressed in the migrating macrophages. Using morpholino (MO) mediated knockdown of each of the MMPs we demonstrate that they are necessary for normal macrophage migration in vivo. The loss-of-function effect can be rescued using the specific MMPs, altered to be resistant to morpholinos but not by overexpression of the other MMPs. Double and triple morpholino knockdowns further suggest that these MMPs act combinatorily to promote embryonic macrophage migration. Thus, our results imply that these three MMPs have distinct functions, which together are crucial to mediate macrophage migration in the developing embryo. This demonstrates conclusively that MMPs are required for normal macrophage cell migration in the whole organism.
Collapse
Affiliation(s)
- Matthew L Tomlinson
- School of Biological Sciences, University of East Anglia, Norwich NR4 7TJ, UK
| | | | | | | |
Collapse
|
44
|
Abstract
Vertebrate blood formation occurs in 2 spatially and temporally distinct waves, so-called primitive and definitive hematopoiesis. Although definitive hematopoiesis has been extensively studied, the development of primitive myeloid blood has received far less attention. In Xenopus, primitive myeloid cells originate in the anterior ventral blood islands, the equivalent of the mammalian yolk sac, and migrate out to colonize the embryo. Using fluorescence time-lapse video microscopy, we recorded the migratory behavior of primitive myeloid cells from their birth. We show that these cells are the first blood cells to differentiate in the embryo and that they are efficiently recruited to embryonic wounds, well before the establishment of a functional vasculature. Furthermore, we isolated spib, an ETS transcription factor, specifically expressed in primitive myeloid precursors. Using spib antisense morpholino knockdown experiments, we show that spib is required for myeloid specification, and, in its absence, primitive myeloid cells retain hemangioblast-like characteristics and fail to migrate. Thus, we conclude that spib sits at the top of the known genetic hierarchy that leads to the specification of primitive myeloid cells in amphibians.
Collapse
|
45
|
Abstract
The neural crest (NC) cells have been called the 'explorers of the embryos' because they migrate all over the embryo where they differentiate into a variety of diverse kinds of cells. In this work, we analyse the role of different molecules controlling the migration of NC cells. First, we describe the strong similarity between the process of NC migration and metastasis in tumour cells. The epithelial-mesenchymal transition process that both kinds of cells undergo is controlled by the same molecular machinery, including cadherins, connexins, Snail and Twist genes and matrix metalloproteases. Second, we analysed the molecular signals that control the patterned migration of the cephalic and trunk NC cells. Most of the factors described so far, such as Eph/ephrins, semaphorins/neuropilins and Slit/Robo, are negative signals that prohibit the migration of NC cells into target areas of the embryo. Finally, we analyse how the direction of migration is controlled by regulation of cell polarity and how the planar cell polarity or non-canonical Wnt signalling is involved in this process.
Collapse
Affiliation(s)
- Sei Kuriyama
- Department of Anatomy and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | | |
Collapse
|
46
|
Abstract
Matrix metalloproteinases (MMPs) were discovered because of their role in amphibian metamorphosis, yet they have attracted more attention because of their roles in disease. Despite intensive scrutiny in vitro, in cell culture and in animal models, the normal physiological roles of these extracellular proteases have been elusive. Recent studies in mice and flies point to essential roles of MMPs as mediators of change and physical adaptation in tissues, whether developmentally regulated, environmentally induced or disease associated.
Collapse
Affiliation(s)
- Andrea Page-McCaw
- Department of Biology and Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York 12180, USA
| | - Andrew J. Ewald
- Department of Anatomy and Program in Biomedical Sciences, University of California, San Francisco, California 94143-0452, USA
| | - Zena Werb
- Department of Anatomy and Program in Biomedical Sciences, University of California, San Francisco, California 94143-0452, USA
| |
Collapse
|
47
|
Greenlee KJ, Werb Z, Kheradmand F. Matrix metalloproteinases in lung: multiple, multifarious, and multifaceted. Physiol Rev 2007; 87:69-98. [PMID: 17237343 PMCID: PMC2656382 DOI: 10.1152/physrev.00022.2006] [Citation(s) in RCA: 334] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 02/06/2023] Open
Abstract
The matrix metalloproteinases (MMPs), a family of 25 secreted and cell surface-bound neutral proteinases, process a large array of extracellular and cell surface proteins under normal and pathological conditions. MMPs play critical roles in lung organogenesis, but their expression, for the most part, is downregulated after generation of the alveoli. Our knowledge about the resurgence of the MMPs that occurs in most inflammatory diseases of the lung is rapidly expanding. Although not all members of the MMP family are found within the lung tissue, many are upregulated during the acute and chronic phases of these diseases. Furthermore, potential MMP targets in the lung include all structural proteins in the extracellular matrix (ECM), cell adhesion molecules, growth factors, cytokines, and chemokines. However, what is less known is the role of MMP proteolysis in modulating the function of these substrates in vivo. Because of their multiplicity and substantial substrate overlap, MMPs are thought to have redundant functions. However, as we explore in this review, such redundancy most likely evolved as a necessary compensatory mechanism given the critical regulatory importance of MMPs. While inhibition of MMPs has been proposed as a therapeutic option in a variety of inflammatory lung conditions, a complete understanding of the biology of these complex enzymes is needed before we can reasonably consider them as therapeutic targets.
Collapse
Affiliation(s)
- Kendra J Greenlee
- Departments of Medicine and Immunology, Baylor College of Medicine, Houston, Texas, USA
| | | | | |
Collapse
|
48
|
Gourdet C, Iribarren C, Morin V, Bustos P, Puchi M, Imschenetzky M. Nuclear cysteine-protease involved in male chromatin remodeling after fertilization is ubiquitously distributed during sea urchin development. J Cell Biochem 2007; 101:1-8. [PMID: 17340626 DOI: 10.1002/jcb.21056] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/08/2022]
Abstract
Previously we have identified a cysteine-protease involved in male chromatin remodeling which segregates into the nuclei of the two blastomeres at the first cleavage division. Here we have investigated the fate of this protease during early embryogenesis by immunodetecting this protein with antibodies elicited against its N-terminal sequence. As shown in this report, the major 60 kDa active form of this protease was found to be present in the extracts of chromosomal proteins obtained from all developmental stages analyzed. In morula and gastrula the 70 kDa inactive precursor, which corresponds to the major form of the zymogen found in unfertilized eggs, was detected. In plutei larvas, the major 60 kDa form of this enzyme was found together with a higher molecular weight precursor (90 kDa) which is consistent with the less abundant zymogen primarily detected in unfertilized eggs. As reported here, either the active protease or its zymogens were visualized in most of the embryonic territories indicating that this enzyme lacks a specific pattern of spatial-temporal developmental segregation. Taken together our results indicate that this protease persists in the embryo and is ubiquitously distributed up to larval stages of development, either as an active enzyme and/or as an inactive precursor. These results suggest that this enzyme may display yet unknown functions during embryonic development that complement its role in male chromatin remodeling after fertilization.
Collapse
Affiliation(s)
- Claudia Gourdet
- Department of Biochemistry and Molecular Biology, Universidad de Concepción, Casilla 160-C, Concepción, Chile
| | | | | | | | | | | |
Collapse
|
49
|
Abu-Elmagd M, Garcia-Morales C, Wheeler GN. Frizzled7 mediates canonical Wnt signaling in neural crest induction. Dev Biol 2006; 298:285-98. [PMID: 16928367 DOI: 10.1016/j.ydbio.2006.06.037] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/27/2006] [Revised: 06/21/2006] [Accepted: 06/22/2006] [Indexed: 11/30/2022]
Abstract
The neural crest is a multipotent cell population that migrates from the dorsal edge of the neural tube to various parts of the embryo where it differentiates into a remarkable variety of different cell types. Initial induction of neural crest is mediated by a combination of BMP, Wnt, FGF, Retinoic acid and Notch/Delta signaling. The two-signal model for neural crest induction suggests that BMP signaling induces the competence to become neural crest. The second signal involves Wnt acting through the canonical pathway and leads to expression of neural crest markers such as slug. Wnt signals from the neural plate, non-neural ectoderm and paraxial mesoderm have all been suggested to play a role in neural crest induction. We show that Xenopus frizzled7 (Xfz7) is expressed in the dorsal ectoderm including early neural crest progenitors and is a key mediator of the Wnt inductive signal. We demonstrate that Xfz7 expression is induced in response to a BMP antagonist, noggin, and that Xfz7 can induce neural crest specific genes in noggin-treated ectodermal explants (animal caps). Morpholino-mediated or dominant negative inhibition of Xfz7 inhibits Wnt induced Xslug expression in the animal cap assay and in the whole embryo leading to a loss of neural crest derived pigment cells. Full-length Xfz7 rescues the morpholino-induced phenotype, as does activated beta-catenin, suggesting that Xfz7 is signaling through the canonical pathway. We therefore demonstrate that Xfz7 is regulated by BMP antagonism and is required for neural crest induction by Wnt in the developing vertebrate embryo.
Collapse
Affiliation(s)
- Muhammad Abu-Elmagd
- School of Biological Sciences, University of East Anglia, Norwich NR4 7TJ, UK
| | | | | |
Collapse
|
50
|
Hasebe T, Hartman R, Matsuda H, Shi YB. Spatial and temporal expression profiles suggest the involvement of gelatinase A and membrane type 1 matrix metalloproteinase in amphibian metamorphosis. Cell Tissue Res 2006; 324:105-16. [PMID: 16418836 DOI: 10.1007/s00441-005-0099-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/29/2005] [Revised: 10/04/2005] [Accepted: 10/18/2005] [Indexed: 10/25/2022]
Abstract
The matrix metalloproteinases (MMPs) are a family of proteases capable of degrading various components of the extracellular matrix (ECM). Among them, the membrane type MMP-1 (MT1-MMP) has been shown to participate in the activation of MMP gelatinase A (GelA), suggesting that they may function together in development and pathogenesis. Here, we have investigated the spatiotemporal expression profiles of Xenopus laevis MT1-MMP and GelA genes during thyroid-hormone-dependent metamorphosis. We have focused our studies on two organs: (1) the intestine, which undergoes first the degeneration of the tadpole epithelium through apoptosis and then the development of adult epithelium and other tissues, and (2) the tail, which completely resorbs through programmed cell death. We show that both MT1-MMP and GelA are upregulated in the intestine and tail when both organs undergo metamorphosis. Within the organs, MT1-MMP and GelA are coexpressed in the connective tissues during both natural and thyroid-hormone-induced metamorphosis. In addition, MT1-MMP (but not GelA) is also expressed in the longitudinal muscle cells of the metamorphosing intestine. These results suggest that MT1-MMP and GelA function together in the ECM degradation or remodeling associated with metamorphosis and that MT1-MMP has additional GelA-independent roles in the development of adult longitudinal muscle in the intestine.
Collapse
Affiliation(s)
- Takashi Hasebe
- Section on Molecular Morphogenesis, Laboratory of Gene Regulation and Development, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | |
Collapse
|