1
|
Leung CLK, Karunakaran S, Atser MG, Innala L, Hu X, Viau V, Johnson JD, Clee SM. Analysis of a genetic region affecting mouse body weight. Physiol Genomics 2023; 55:132-146. [PMID: 36717164 PMCID: PMC10042608 DOI: 10.1152/physiolgenomics.00137.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Genetic factors affect an individual's risk of developing obesity, but in most cases each genetic variant has a small effect. Discovery of genes that regulate obesity may provide clues about its underlying biological processes and point to new ways the disease can be treated. Preclinical animal models facilitate genetic discovery in obesity because environmental factors can be better controlled compared with the human population. We studied inbred mouse strains to identify novel genes affecting obesity and glucose metabolism. BTBR T+ Itpr3tf/J (BTBR) mice are fatter and more glucose intolerant than C57BL/6J (B6) mice. Prior genetic studies of these strains identified an obesity locus on chromosome 2. Using congenic mice, we found that obesity was affected by a ∼316 kb region, with only two known genes, pyruvate dehydrogenase kinase 1 (Pdk1) and integrin α 6 (Itga6). Both genes had mutations affecting their amino acid sequence and reducing mRNA levels. Both genes have known functions that could modulate obesity, lipid metabolism, insulin secretion, and/or glucose homeostasis. We hypothesized that genetic variation in or near Pdk1 or Itga6 causing reduced Pdk1 and Itga6 expression would promote obesity and impaired glucose tolerance. We used knockout mice lacking Pdk1 or Itga6 fed an obesigenic diet to test this hypothesis. Under the conditions we studied, we were unable to detect an individual contribution of either Pdk1 or Itga6 to body weight. During our studies, with conditions outside our control, we were unable to reproduce some of our previous body weight data. However, we identified a previously unknown role for Pdk1 in cardiac cholesterol metabolism providing the basis for future investigations. The studies described in this paper highlight the importance and the challenge using physiological outcomes to study obesity genes in mice.
Collapse
Affiliation(s)
- Connie L K Leung
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Subashini Karunakaran
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Michael G Atser
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Leyla Innala
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Xiaoke Hu
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Victor Viau
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - James D Johnson
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Susanne M Clee
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
2
|
The cellular dynamics of neural tube formation. Biochem Soc Trans 2023; 51:343-352. [PMID: 36794768 PMCID: PMC9987952 DOI: 10.1042/bst20220871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/23/2023] [Accepted: 01/31/2023] [Indexed: 02/17/2023]
Abstract
The vertebrate brain and spinal cord arise from a common precursor, the neural tube, which forms very early during embryonic development. To shape the forming neural tube, changes in cellular architecture must be tightly co-ordinated in space and time. Live imaging of different animal models has provided valuable insights into the cellular dynamics driving neural tube formation. The most well-characterised morphogenetic processes underlying this transformation are convergent extension and apical constriction, which elongate and bend the neural plate. Recent work has focused on understanding how these two processes are spatiotemporally integrated from the tissue- to the subcellular scale. Various mechanisms of neural tube closure have also been visualised, yielding a growing understanding of how cellular movements, junctional remodelling and interactions with the extracellular matrix promote fusion and zippering of the neural tube. Additionally, live imaging has also now revealed a mechanical role for apoptosis in neural plate bending, and how cell intercalation forms the lumen of the secondary neural tube. Here, we highlight the latest research on the cellular dynamics underlying neural tube formation and provide some perspectives for the future.
Collapse
|
3
|
Kuriyama S, Tanaka M. Characteristic tetraspanin expression patterns mark various tissues during early Xenopus development. Dev Growth Differ 2023; 65:109-119. [PMID: 36606534 DOI: 10.1111/dgd.12836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 12/21/2022] [Accepted: 12/26/2022] [Indexed: 01/07/2023]
Abstract
The tetraspanins (Tspans) constitute a family of cell surface proteins with four transmembrane domains. Tspans have been found on the plasma membrane and on exosomes of various organelles. Reports on the function of Tspans during the early development of Xenopus have mainly focused on the expression of uroplakins in gametes. Although the roles of extracellular vesicles (EVs) including exosomes have been actively analyzed in cancer research, the contribution of EVs to early development is not well understood. This is because the diffusivity of EVs is not compatible with a very strict developmental process. In this study, we analyzed members of the Tspan family in early development of Xenopus. Expression was prominent in specific organs such as the notochord, eye, cranial neural crest cells (CNCs), trunk neural crest cells, placodes, and somites. We overexpressed several combinations of Tspans in CNCs in vitro and in vivo. Changing the partner changed the distribution of fluorescent-labeled Tspans. Therefore, it is suggested that expression of multiple Tspans in a particular tissue might produce heterogeneity of intercellular communication, which has not yet been recognized.
Collapse
Affiliation(s)
- Sei Kuriyama
- Department of Molecular Medicine and Biochemistry, Akita University, Akita, Japan
| | - Masamitsu Tanaka
- Department of Molecular Medicine and Biochemistry, Akita University, Akita, Japan
| |
Collapse
|
4
|
Nikolopoulou E, Galea GL, Rolo A, Greene NDE, Copp AJ. Neural tube closure: cellular, molecular and biomechanical mechanisms. Development 2017; 144:552-566. [PMID: 28196803 DOI: 10.1242/dev.145904] [Citation(s) in RCA: 320] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Neural tube closure has been studied for many decades, across a range of vertebrates, as a paradigm of embryonic morphogenesis. Neurulation is of particular interest in view of the severe congenital malformations - 'neural tube defects' - that result when closure fails. The process of neural tube closure is complex and involves cellular events such as convergent extension, apical constriction and interkinetic nuclear migration, as well as precise molecular control via the non-canonical Wnt/planar cell polarity pathway, Shh/BMP signalling, and the transcription factors Grhl2/3, Pax3, Cdx2 and Zic2. More recently, biomechanical inputs into neural tube morphogenesis have also been identified. Here, we review these cellular, molecular and biomechanical mechanisms involved in neural tube closure, based on studies of various vertebrate species, focusing on the most recent advances in the field.
Collapse
Affiliation(s)
- Evanthia Nikolopoulou
- Newlife Birth Defects Research Centre, Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, UK
| | - Gabriel L Galea
- Newlife Birth Defects Research Centre, Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, UK
| | - Ana Rolo
- Newlife Birth Defects Research Centre, Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, UK
| | - Nicholas D E Greene
- Newlife Birth Defects Research Centre, Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, UK
| | - Andrew J Copp
- Newlife Birth Defects Research Centre, Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, UK
| |
Collapse
|
5
|
Basu S, Rajakaruna S, De Arcangelis A, Zhang L, Georges-Labouesse E, Menko AS. α6 integrin transactivates insulin-like growth factor receptor-1 (IGF-1R) to regulate caspase-3-mediated lens epithelial cell differentiation initiation. J Biol Chem 2013; 289:3842-55. [PMID: 24381169 DOI: 10.1074/jbc.m113.515254] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The canonical mitochondrial death pathway was first discovered for its role in signaling apoptosis. It has since been found to have a requisite function in differentiation initiation in many cell types including the lens through low level activation of the caspase-3 protease. The ability of this pathway to function as a molecular switch in lens differentiation depends on the concurrent induction of survival molecules in the Bcl-2 and IAP families, induced downstream of an IGF-1R/NFκB coordinate survival signal, to regulate caspase-3 activity. Here we investigated whether α6 integrin signals upstream to this IGF-1R-mediated survival-linked differentiation signal. Our findings show that IGF-1R is recruited to and activated specifically in α6 integrin receptor signaling complexes in the lens equatorial region, where lens epithelial cells initiate their differentiation program. In studies with both α6 integrin knock-out mice lenses and primary lens cell cultures following α6 integrin siRNA knockdown, we show that IGF-1R activation is dependent on α6 integrin and that this transactivation requires Src kinase activity. In addition, without α6 integrin, activation and expression of NFκB was diminished, and expression of Bcl-2 and IAP family members were down-regulated, resulting in high levels of caspase-3 activation. As a result, a number of hallmarks of lens differentiation failed to be induced; including nuclear translocation of Prox1 in the differentiation initiation zone and apoptosis was promoted. We conclude that α6 integrin is an essential upstream regulator of the IGF-1R survival pathway that regulates the activity level of caspase-3 for it to signal differentiation initiation of lens epithelial cells.
Collapse
Affiliation(s)
- Subhasree Basu
- From the Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, Pennsylvania 19107 and
| | | | | | | | | | | |
Collapse
|
6
|
Edlund AF, Davidson LA, Keller RE. Cell segregation, mixing, and tissue pattern in the spinal cord of the Xenopus laevis neurula. Dev Dyn 2013; 242:1134-46. [PMID: 23813905 PMCID: PMC4104979 DOI: 10.1002/dvdy.24004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Accepted: 06/05/2013] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND During Xenopus laevis neurulation, neural ectodermal cells of the spinal cord are patterned at the same time that they intercalate mediolaterally and radially, moving within and between two cell layers. Curious if these rearrangements disrupt early cell identities, we lineage-traced cells in each layer from neural plate stages to the closed neural tube, and used in situ hybridization to assay gene expression in the moving cells. RESULTS Our biotin and fluorescent labeling of deep and superficial cells reveals that mediolateral intercalation does not disrupt cell cohorts; in other words, it is conservative. However, outside the midline notoplate, later radial intercalation does displace superficial cells dorsoventrally, radically disrupting cell cohorts. The tube roof is composed almost exclusively of superficial cells, including some displaced from ventral positions; gene expression in these displaced cells must now be surveyed further. Superficial cells also flank the tube's floor, which is, itself, almost exclusively composed of deep cells. CONCLUSIONS Our data provide: (1) a fate map of superficial- and deep-cell positions within the Xenopus neural tube, (2) the paths taken to these positions, and (3) preliminary evidence of re-patterning in cells carried out of one environment and into another, during neural morphogenesis.
Collapse
Affiliation(s)
- Anna F Edlund
- Department of Biology, University of Virginia, Charlottesville, Virginia
| | | | | |
Collapse
|
7
|
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] [Scholar 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
|
8
|
Wang R, Liu X, Küster-Schöck E, Fagotto F. Proteomic analysis of differences in ectoderm and mesoderm membranes by DiGE. J Proteome Res 2012; 11:4575-93. [PMID: 22852788 DOI: 10.1021/pr300379m] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Ectoderm and mesoderm can be considered as prototypes for epithelial and mesenchymal cell types. These two embryonic tissues display clear differences in adhesive and motility properties, which are phenomenologically well characterized but remain largely unexplored at the molecular level. Because the key downstream regulations must occur at the plasma membrane and in the underlying actin cortical structures, we have set out to compare the protein content of membrane fractions from Xenopus ectoderm and mesoderm tissues using 2-dimensional difference gel electrophoresis (DiGE). We have thus identified several proteins that are enriched in one or the other tissues, including regulators of the cytoskeleton and of cell signaling. This study represents to our knowledge the first attempt to use proteomics specifically targeted to the membrane-cortex compartment of embryonic tissues. The identified components should help unraveling a variety of tissue-specific functions in the embryo.
Collapse
Affiliation(s)
- Renee Wang
- Department of Biology, McGill University, Montreal, Canada
| | | | | | | |
Collapse
|
9
|
Morita H, Kajiura-Kobayashi H, Takagi C, Yamamoto TS, Nonaka S, Ueno N. Cell movements of the deep layer of non-neural ectoderm underlie complete neural tube closure in Xenopus. Development 2012; 139:1417-26. [PMID: 22378637 DOI: 10.1242/dev.073239] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
In developing vertebrates, the neural tube forms from a sheet of neural ectoderm by complex cell movements and morphogenesis. Convergent extension movements and the apical constriction along with apical-basal elongation of cells in the neural ectoderm are thought to be essential for the neural tube closure (NTC) process. In addition, it is known that non-neural ectoderm also plays a crucial role in this process, as the neural tube fails to close in the absence of this tissue in chick and axolotl. However, the cellular and molecular mechanisms by which it functions in NTC are as yet unclear. We demonstrate here that the non-neural superficial epithelium moves in the direction of tensile forces applied along the dorsal-ventral axis during NTC. We found that this force is partly attributable to the deep layer of non-neural ectoderm cells, which moved collectively towards the dorsal midline along with the superficial layer. Moreover, inhibition of this movement by deleting integrin β1 function resulted in incomplete NTC. Furthermore, we demonstrated that other proposed mechanisms, such as oriented cell division, cell rearrangement and cell-shape changes have no or only minor roles in the non-neural movement. This study is the first to demonstrate dorsally oriented deep-cell migration in non-neural ectoderm, and suggests that a global reorganization of embryo tissues is involved in NTC.
Collapse
Affiliation(s)
- Hitoshi Morita
- Division of Morphogenesis, National Institute for Basic Biology, Nishigonaka 38, Myodaiji, Okazaki, Aichi, Japan
| | | | | | | | | | | |
Collapse
|
10
|
Mukhopadhyay P, Brock G, Webb C, Pisano MM, Greene RM. Strain-specific modifier genes governing craniofacial phenotypes. ACTA ACUST UNITED AC 2012; 94:162-75. [PMID: 22371338 DOI: 10.1002/bdra.22890] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2011] [Revised: 12/12/2011] [Accepted: 12/13/2011] [Indexed: 01/06/2023]
Abstract
BACKGROUND The presence of strain-specific modifier genes is known to modulate the phenotype and pathophysiology of mice harboring genetically engineered mutations. Thus, identification of genetic modifier genes is requisite to understanding control of phenotypic expression. c-Ski is a transcriptional regulator. Ski(-/-) mice on a C57BL6J (B6) background exhibit facial clefting, while Ski(-/-) mice on a 129P3 (129) background present with exencephaly. METHODS In the present study, oligonucleotide-based gene expression profiling was used to identify potential strain-specific modifier gene candidates present in wild type mice of B6 and 129 genetic backgrounds. Changes in gene expression were verified by TaqMan quantitative real-time PCR. RESULTS Steady-state levels of 89 genes demonstrated a significantly higher level of expression, and those of 68 genes demonstrated a significantly lower level of expression in the developing neural tubes from embryonic day (E) 8.5, B6 embryos when compared to expression levels in neural tubes derived from E 8.5, 129 embryos. CONCLUSIONS Based on the results from the current comparative microarray study, and taking into consideration a number of relevant published reports, several potential strain-specific gene candidates, likely to modify the craniofacial phenotypes in various knockout mouse models have been identified.
Collapse
Affiliation(s)
- Partha Mukhopadhyay
- University of Louisville Birth Defects Center, Department of Molecular, Cellular and Craniofacial Biology, ULSD, University of Louisville, Louisville, Kentucky 40292, USA
| | | | | | | | | |
Collapse
|
11
|
Alfandari D, Cousin H, Marsden M. Mechanism of Xenopus cranial neural crest cell migration. Cell Adh Migr 2010; 4:553-60. [PMID: 20505318 DOI: 10.4161/cam.4.4.12202] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
This review focuses on recent advances in the field of cranial neural crest cell migration in Xenopus laevis with specific emphasis on cell adhesion and the regulation of cell migration. Our goal is to combine the understanding of cell adhesion to the extracellular matrix with the regulation of cell-cell adhesion and the involvement of the planar cell polarity signaling-pathway in guiding the migration of cranial neural crest cells during embryogenesis.
Collapse
Affiliation(s)
- Dominque Alfandari
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, USA.
| | | | | |
Collapse
|
12
|
Duband JL. Diversity in the molecular and cellular strategies of epithelium-to-mesenchyme transitions: Insights from the neural crest. Cell Adh Migr 2010; 4:458-82. [PMID: 20559020 DOI: 10.4161/cam.4.3.12501] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Although epithelial to mesenchymal transitions (EMT) are often viewed as a unique event, they are characterized by a great diversity of cellular processes resulting in strikingly different outcomes. They may be complete or partial, massive or progressive, and lead to the complete disruption of the epithelium or leave it intact. Although the molecular and cellular mechanisms of EMT are being elucidated owing chiefly from studies on transformed epithelial cell lines cultured in vitro or from cancer cells, the basis of the diversity of EMT processes remains poorly understood. Clues can be collected from EMT occuring during embryonic development and which affect equally tissues of ectodermal, endodermal or mesodermal origins. Here, based on our current knowledge of the diversity of processes underlying EMT of neural crest cells in the vertebrate embryo, we propose that the time course and extent of EMT do not depend merely on the identity of the EMT transcriptional regulators and their cellular effectors but rather on the combination of molecular players recruited and on the possible coordination of EMT with other cellular processes.
Collapse
|
13
|
Sroka IC, Anderson TA, McDaniel KM, Nagle RB, Gretzer MB, Cress AE. The laminin binding integrin alpha6beta1 in prostate cancer perineural invasion. J Cell Physiol 2010; 224:283-8. [PMID: 20432448 DOI: 10.1002/jcp.22149] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Metastasizing prostate tumor cells invade along nerves innervating the encapsulated human prostate gland in a process known as perineural invasion. The extracellular matrix laminin class of proteins line the neural route and tumor cells escaping from the gland express the laminin binding integrin alpha6beta1 as a prominent cell surface receptor. Integrin alpha6beta1 promotes aggressive disease and supports prostate tumor cell metastasis to bone. Laminins and their integrin receptors are necessary for the development and maintenance of the peripheral nervous system, indicating the potential role for integrin receptors in directing prostate tumor cell invasion on nerves during perineural invasion.
Collapse
Affiliation(s)
- Isis C Sroka
- Department of Pharmacology, The University of Arizona, Tucson, Arizona, USA
| | | | | | | | | | | |
Collapse
|
14
|
|
15
|
Rozario T, DeSimone DW. The extracellular matrix in development and morphogenesis: a dynamic view. Dev Biol 2010; 341:126-40. [PMID: 19854168 PMCID: PMC2854274 DOI: 10.1016/j.ydbio.2009.10.026] [Citation(s) in RCA: 932] [Impact Index Per Article: 62.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2009] [Revised: 10/16/2009] [Accepted: 10/17/2009] [Indexed: 02/06/2023]
Abstract
The extracellular matrix (ECM) is synthesized and secreted by embryonic cells beginning at the earliest stages of development. Our understanding of ECM composition, structure and function has grown considerably in the last several decades and this knowledge has revealed that the extracellular microenvironment is critically important for cell growth, survival, differentiation and morphogenesis. ECM and the cellular receptors that interact with it mediate both physical linkages with the cytoskeleton and the bidirectional flow of information between the extracellular and intracellular compartments. This review considers the range of cell and tissue functions attributed to ECM molecules and summarizes recent findings specific to key developmental processes. The importance of ECM as a dynamic repository for growth factors is highlighted along with more recent studies implicating the 3-dimensional organization and physical properties of the ECM as it relates to cell signaling and the regulation of morphogenetic cell behaviors. Embryonic cell and tissue generated forces and mechanical signals arising from ECM adhesion represent emerging areas of interest in this field.
Collapse
Affiliation(s)
- Tania Rozario
- Department of Cell Biology and the Morphogenesis and Regenerative Medicine Institute, University of Virginia, PO Box 800732, School of Medicine, Charlottesville, VA 22908, USA
| | | |
Collapse
|
16
|
Abstract
Integrin-mediated adhesion is as ancient as multicellularity, but it was not always as complex as it is in humans. Here, I examine the extent of conservation of 192 adhesome proteins across the genomes of nine model organisms spanning one and a half billion years of evolution. The work reveals that Rho GTPases, lipid- and serine/threonine-kinases, and phosphatases existed before integrins, but tyrosine phosphorylation developed concomitant with integrins. The expansion of specific functional groups such as GAPs, GEFs, adaptors, and receptors is demonstrated, along with the expansion of specific protein domains, such as SH3, PH, SH2, CH, and LIM. Expansion is due to gene duplication and creation of families of paralogues. Apparently, these paralogues share few partners and create new sets of interactions, thus increasing specificity and the repertoire of integrin-mediated signaling. Interestingly, the average number of interactions positively correlates with the evolutionary age of proteins. While shedding light on the evolution of adhesome complexity, this analysis also highlights the relevance and creates a framework for studying integrin-mediated adhesion in simpler model organisms.
Collapse
Affiliation(s)
- Ronen Zaidel-Bar
- Department of Zoology, University of Wisconsin-Madison, Madison, WI 53706, USA.
| |
Collapse
|
17
|
Thomas JT, Canelos P, Luyten FP, Moos M. Xenopus SMOC-1 Inhibits bone morphogenetic protein signaling downstream of receptor binding and is essential for postgastrulation development in Xenopus. J Biol Chem 2009; 284:18994-9005. [PMID: 19414592 PMCID: PMC2707235 DOI: 10.1074/jbc.m807759200] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2008] [Revised: 02/26/2009] [Indexed: 11/06/2022] Open
Abstract
The bone morphogenetic protein (BMP) family of signaling molecules and their antagonists are involved in patterning of the body axis and numerous aspects of organogenesis. Classical biochemical purification and protein sequencing of highly purified fractions containing potent bone forming activity from bovine cartilage identified several BMPs together with a number of other proteins. One such protein was SMOC-2 (secreted modular calcium-binding protein-2), classified as belonging to the BM-40 family of modular extracellular proteins. Data regarding the biological function of SMOC-2 and closely related SMOC-1 remain limited, and their expression or function during embryological development is unknown. We therefore isolated the Xenopus ortholog of human SMOC-1 (XSMOC-1) and explored its function in Xenopus embryos. In gain-of-function assays, XSMOC-1 acted similarly to a BMP antagonist. However, in contrast to known extracellular ligand-binding BMP antagonists, such as noggin, SMOC antagonizes BMP activity in the presence of a constitutively active BMP receptor, indicating a mechanism of action downstream of the receptor. We provide several lines of evidence to suggest that SMOC acts downstream of the BMP receptor via MAPK-mediated phosphorylation of the Smad linker region. Loss-of-function studies, using antisense morpholino oligonucleotides, revealed XSMOC-1 to be essential for postgastrulation development. The catastrophic developmental failure observed following XSMOC knockdown resembles that observed following simultaneous depletion of three ligand-binding BMP antagonists described in prior studies. These findings provide a direct link between the extracellular matrix-associated protein SMOC and a signaling pathway of general importance in anatomic patterning and cell or tissue fate specification.
Collapse
Affiliation(s)
- J. Terrig Thomas
- From the Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, United States Food and Drug Administration, Bethesda, Maryland 20892 and
| | - Paola Canelos
- From the Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, United States Food and Drug Administration, Bethesda, Maryland 20892 and
| | - Frank P. Luyten
- the Laboratory for Skeletal Development and Joint Disorders, Division of Rheumatology, Department of Musculoskeletal Sciences, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Malcolm Moos
- From the Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, United States Food and Drug Administration, Bethesda, Maryland 20892 and
| |
Collapse
|
18
|
Bello V, Sirour C, Moreau N, Denker E, Darribère T. A function for dystroglycan in pronephros development in Xenopus laevis. Dev Biol 2008; 317:106-20. [DOI: 10.1016/j.ydbio.2008.02.024] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2007] [Revised: 02/01/2008] [Accepted: 02/05/2008] [Indexed: 11/27/2022]
|
19
|
Demetriou MC, Stylianou P, Andreou M, Yiannikouri O, Tsaprailis G, Cress AE, Skourides P. Spatially and temporally regulated alpha6 integrin cleavage during Xenopus laevis development. Biochem Biophys Res Commun 2007; 366:779-85. [PMID: 18083114 DOI: 10.1016/j.bbrc.2007.12.040] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2007] [Accepted: 12/04/2007] [Indexed: 10/22/2022]
Abstract
The alpha6 integrin is essential for early nervous system development in Xenopus laevis. We have previously reported a uPA cleaved form of integrin alpha6 (alpha6p), in invasive human prostate cancer tissue, whose presence correlates with increased migration and invasive capacity. We now report that alpha6 is cleaved during the normal development of Xenopus in a spatially and temporally controlled manner. In addition, unlike normal mammalian tissues, which lack alpha6p, the major form of the alpha6 integrin present in adult Xenopus is alpha6p. The protease responsible for the cleavage in mammals, uPA, is not involved in the cleavage of Xenopus alpha6. Finally, overexpression of a mammalian alpha6 mutant which cannot be cleaved leads to developmental abnormalities suggesting a potential role for the cleavage in development.
Collapse
Affiliation(s)
- Manolis C Demetriou
- Department of Biological Sciences, The University of Cyprus, Kallipoleos 75, 1678 Nicosia, Cyprus
| | | | | | | | | | | | | |
Collapse
|
20
|
Strachan LR, Condic ML. Neural crest motility on fibronectin is regulated by integrin activation. Exp Cell Res 2007; 314:441-52. [PMID: 18036522 DOI: 10.1016/j.yexcr.2007.10.016] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2007] [Revised: 10/10/2007] [Accepted: 10/25/2007] [Indexed: 10/25/2022]
Abstract
Cell migration is essential for proper development of numerous structures derived from embryonic neural crest cells (NCCs). Although recent work has shown that receptor recycling plays an important role in NCC motility on laminin, the molecular mechanisms regulating NCC motility on fibronectin remain unclear. One mechanism by which cells regulate motility is by modulating the affinity of integrin receptors. Here, we provide evidence that cranial and trunk NCCs rely on functional regulation of integrins to migrate efficiently on fibronectin (FN) in vitro. For NCCs cultured on fibronectin, velocity decreases after Mn2+ application (a treatment that activates all surface integrins) while velocity on laminin (LM) is not affected. The distribution of activated integrin beta 1 receptors on the surface of NCCs is also substratum-dependent. Integrin activation affects cranial and trunk NCCs differently when cultured on different concentrations of FN substrata; only cranial NCCs slow in a FN concentration-dependent manner. Furthermore, Mn2+ treatment alters the distribution and number of activated integrin beta 1 receptors on the surface of cranial and trunk NCCs in different ways. We provide a hypothesis whereby a combination of activated surface integrin levels and the degree to which those receptors are clustered determines NCC motility on fibronectin.
Collapse
Affiliation(s)
- L R Strachan
- Department of Neurobiology and Anatomy, University of Utah, School of Medicine, 20 North 1900 East, Salt Lake City, UT 84132-3401, USA
| | | |
Collapse
|
21
|
Etchevers HC, Amiel J, Lyonnet S. Molecular bases of human neurocristopathies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2007; 589:213-34. [PMID: 17076285 DOI: 10.1007/978-0-387-46954-6_14] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Heather C Etchevers
- Département de Génétique Médicale, INSERM U393, Hôpital Necker - Enfants Malades, 149 rue de Sèvres, 75743 Paris 15, France.
| | | | | |
Collapse
|
22
|
Davies D. Temporal and spatial regulation of alpha6 integrin expression during the development of the cochlear-vestibular ganglion. J Comp Neurol 2007; 502:673-82. [PMID: 17436285 DOI: 10.1002/cne.21302] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The neurons of the cochlear-vestibular ganglion (CVG) that innervate the sensory hair cells of the inner ear are derived from the otic epithelium early in development. Neuroblasts detach from neighboring cells, migrate into the mesenchyme where they coalesce to form the ganglion complex, then send processes back into the epithelium. Cell migration and neuronal process formation involve changes in cellular interactions with other cells and proteins in the extracellular matrix that are orchestrated by cell surface-expressed adhesion molecules, including the integrins. I studied the expression pattern of the alpha6 integrin subunit during the early development of the CVG using immunohistochemistry and reverse-transcriptase polymerase chain reaction (RT-PCR) in murine tissue sections, otocyst, and ganglion explants. At embryonic day (E)10.5 alpha6 integrin was expressed in the otic epithelium but not in migrating neuroblasts. Importantly, the loss of alpha6 was associated with exit from the epithelium, not neuronal determination, revealing differentiation cues acutely associated with the cellular environment. Markers of glial and neuronal phenotype showed that alpha6-expressing cells present in the CVG at this stage were glia of neural crest origin. By E12.5 alpha6 expression in the ganglion increased alongside the elaboration of neuronal processes. Immunohistochemistry applied to otocyst cultures in the absence of glia revealed that neuronal processes remained alpha6-negative at this developmental stage and confirmed that alpha6 was expressed by closely apposed glia. The spatiotemporal modulation of alpha6 expression suggests changing roles for this integrin during the early development of inner ear innervation.
Collapse
Affiliation(s)
- Dawn Davies
- Department of Physiology, University of Bristol, School of Medical Sciences, University Walk, Bristol, BS8 1TD, UK.
| |
Collapse
|
23
|
Witt M, Hummel T. Vomeronasal versus olfactory epithelium: is there a cellular basis for human vomeronasal perception? INTERNATIONAL REVIEW OF CYTOLOGY 2006; 248:209-59. [PMID: 16487792 DOI: 10.1016/s0074-7696(06)48004-9] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The vomeronasal organ (VNO) constitutes an accessory olfactory organ that receives chemical stimuli, pheromones, which elicit behavioral, reproductive, or neuroendocrine responses among individuals of the same species. In many macrosmatic animals, the morphological substrate constitutes a separate organ system consisting of a vomeronasal duct (ductus vomeronasalis, VND), equipped with chemosensory cells, and a vomeronasal nerve (nervus vomeronasalis, VNN) conducting information into the accessory olfactory bulb (AOB) in the central nervous system (CNS). Recent data require that the long-accepted dual functionality of a main olfactory system and the VNO be reexamined, since all species without a VNO are nevertheless sexually active, and species possessing a VNO also can sense other than "vomeronasal" stimuli via the vomeronasal epithelium (VNE). The human case constitutes a borderline situation, as its embryonic VNO anlage exerts a developmental track common to most macrosmatics, but later typical structures such as the VNN, AOB, and probably most of the chemoreceptor cells within the still existent VND are lost. This review also presents recent information on the VND including immunohistochemical expression of neuronal markers, intermediate filaments, lectins, integrins, caveolin, CD44, and aquaporins. Further, we will address the issue of human pheromone candidates.
Collapse
Affiliation(s)
- Martin Witt
- Department of Anatomy, University of Technology Dresden, Dresden, Germany
| | | |
Collapse
|
24
|
Abstract
The PCP signaling cascade controls polarized cell behaviors in various organisms. New evidence suggests that this signaling cascade also controls the deposition of extracellular matrix during vertebrate gastrulation.
Collapse
Affiliation(s)
- John B Wallingford
- Section of Molecular Cell and Developmental Biology & Institute for Cellular and Molecular Biology, University of Texas, Austin 78712,
| |
Collapse
|
25
|
Tucker RP. Antisense knockdown of the beta1 integrin subunit in the chicken embryo results in abnormal neural crest cell development. Int J Biochem Cell Biol 2004; 36:1135-9. [PMID: 15094128 DOI: 10.1016/j.biocel.2004.01.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2003] [Revised: 01/06/2004] [Accepted: 01/08/2004] [Indexed: 10/26/2022]
Abstract
Neural crest cells escape the neural tube by undergoing an epithelial to mesenchymal transition (EMT). This is followed by extensive migration along specific pathways that are lined with extracellular matrix (ECM). In this study, we have examined the roles of matrix receptors containing beta1 integrin subunits in neural crest cell morphogenesis using antisense morpholino oligos electroporated in ovo into avian neural crest cell precursors. Our results show that reduced levels of expression of beta1 integrin subunits in the dorsal neural tube results in an abnormal epithelial to mesenchymal transition. In approximately half of the experimental embryos, however, some neural crest cells filled with beta1 antisense are able to escape the neural tube and migrate ventrally, indicating that grossly normal migration of trunk neural crest cells can take place after beta1 integrin expression is reduced. This study shows the potential of this novel method for investigating the roles of genes that are required for the survival of early mouse embryos in later development events.
Collapse
Affiliation(s)
- Richard P Tucker
- Department of Cell Biology and Human Anatomy, School of Medicine, University of California at Davis, 1 Shields Avenue, Davis, CA 95616, USA.
| |
Collapse
|
26
|
Alfandari D, Cousin H, Gaultier A, Hoffstrom BG, DeSimone DW. Integrin alpha5beta1 supports the migration of Xenopus cranial neural crest on fibronectin. Dev Biol 2003; 260:449-64. [PMID: 12921745 DOI: 10.1016/s0012-1606(03)00277-x] [Citation(s) in RCA: 102] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
During early embryonic development, cranial neural crest cells emerge from the developing mid- and hindbrain. While numerous studies have focused on integrin involvement in trunk neural crest cell migration, comparatively little is known about mechanisms of cranial neural crest cell migration. We show that fibronectin, but not laminin, vitronectin, or type I collagen can support cranial neural crest cell migration and segmentation in vitro. These behaviors require both the RGD and "synergy" sites located within the central cell-binding domain of fibronectin. While these two sites are sufficient for cranial neural crest cell migration, we find that the second Heparin-binding domain of fibronectin can provide additional support for cranial neural crest cell migration in vitro. Finally, using a function blocking monoclonal antibody, we show that cranial neural crest cell migration on fibronectin requires the integrin alpha5beta1.
Collapse
Affiliation(s)
- Dominique Alfandari
- Department of Cell Biology, University of Virginia Health Sciences System, Box 800732, School of Medicine, Charlottesville, VA 22908, USA.
| | | | | | | | | |
Collapse
|
27
|
Barreto G, Reintsch W, Kaufmann C, Dreyer C. The function of Xenopus germ cell nuclear factor (xGCNF) in morphogenetic movements during neurulation. Dev Biol 2003; 257:329-42. [PMID: 12729562 DOI: 10.1016/s0012-1606(03)00109-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The germ cell nuclear factor (GCNF, NR6A1) is a nuclear orphan receptor first described in the mouse testis and subsequently identified as an essential transcription factor in vertebrate embryogenesis. Here, we analyze the phenotype of Xenopus embryos after depletion of embryonic GCNF (xEmGCNF) protein using a specific morpholino antisense oligonucleotide. Morphological defects after xEmGCNF knockdown became obvious from neurulation onward. Among the abnormalities observed, defective formation of the neural tube and a short and curved main body axis were the most remarkable traits. Histological analysis, lineage tracing of injected blastomeres, and Keller sandwich explants revealed that xEmGCNF function is required for different patterns of cell intercalation during neurulation and consequently for the sequence of morphogenetic movements leading to formation of the neural tube. Further characterization of the phenotype at the molecular level showed an abnormal distribution of the extracellular matrix protein fibronectin and a reduction in the expression level of the integrin subunits alpha5 and alpha6, the limiting components of the laminin and fibronectin receptors, respectively. We propose integrin-mediated cell-matrix interaction as a process that requires xEmGCNF function and provides, in concert with cadherins-mediated cell-cell interactions, a molecular basis for morphogenetic cell movements during neurulation.
Collapse
Affiliation(s)
- Guillermo Barreto
- Max-Planck-Institut für Entwicklungsbiologie, Spemannstrasse 35/V, D-72076 Tübingen, Germany.
| | | | | | | |
Collapse
|
28
|
Ezin AM, Skoglund P, Keller R. The midline (notochord and notoplate) patterns the cell motility underlying convergence and extension of the Xenopus neural plate. Dev Biol 2003; 256:100-14. [PMID: 12654295 DOI: 10.1016/s0012-1606(02)00130-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
We investigated the role of the dorsal midline structures, the notochord and notoplate, in patterning the cell motilities that underlie convergent extension of the Xenopus neural plate. In explants of deep neural plate with underlying dorsal mesoderm, lateral neural plate cells show a monopolar, medially directed protrusive activity. In contrast, neural plate explants lacking the underlying dorsal mesoderm show a bipolar, mediolaterally directed protrusive activity. Here, we report that "midlineless" explants consisting of the deep neural plate and underlying somitic mesoderm, but lacking a midline, show bipolar, mediolaterally oriented protrusive activity. Adding an ectopic midline to the lateral edge of these explants restores the monopolar protrusive activity over the entire extent of the midlineless explant. Monopolarized cells near the ectopic midline orient toward it, whereas those located near the original, removed midline orient toward this midline. This behavior can be explained by two signals emanating from the midline. We postulate that one signal polarizes neural plate deep cells and is labile and short-lived and that the second signal orients any polarized cells toward the midline and is persistent.
Collapse
Affiliation(s)
- Akouavi M Ezin
- Department of Biology, University of Virginia, Charlottesville, VA 22903, USA.
| | | | | |
Collapse
|
29
|
Drawbridge J, Meighan CM, Lumpkins R, Kite ME. Pronephric duct extension in amphibian embryos: migration and other mechanisms. Dev Dyn 2003; 226:1-11. [PMID: 12508219 DOI: 10.1002/dvdy.10205] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Initiation of excretory system development in all vertebrates requires (1) delamination of the pronephric and pronephric duct rudiments from intermediate mesoderm at the ventral border of anterior somites, and (2) extension of the pronephric duct to the cloaca. Pronephric duct extension is the central event in nephric system development; the pronephric duct differentiates into the tubule that carries nephric filtrate out of the body and induces terminal differentiation of adult kidneys. Early studies concluded that pronephric ducts formed by means of in situ segregation of pronephric duct tissue from lateral mesoderm ventral to the forming somites; more recent studies highlight caudal migration of the pronephric duct as the major morphogenetic mechanism. The purpose of this review is to provide the historical background on studies of the mechanisms of amphibian pronephric duct extension, to review evidence showing that different amphibians perform pronephric duct morphogenesis in different ways, and to suggest future studies that may help illuminate the molecular basis of the mechanisms that have evolved in amphibians to extend the pronephric duct to the cloaca.
Collapse
Affiliation(s)
- Julie Drawbridge
- Department of Biology, Rider University, Lawrenceville, New Jersey 08648, USA.
| | | | | | | |
Collapse
|
30
|
Colognato H, Baron W, Avellana-Adalid V, Relvas JB, Baron-Van Evercooren A, Georges-Labouesse E, ffrench-Constant C. CNS integrins switch growth factor signalling to promote target-dependent survival. Nat Cell Biol 2002; 4:833-41. [PMID: 12379866 DOI: 10.1038/ncb865] [Citation(s) in RCA: 208] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2002] [Revised: 08/02/2002] [Accepted: 08/09/2002] [Indexed: 01/15/2023]
Abstract
Depending on the stage of development, a growth factor can mediate cell proliferation, survival or differentiation. The interaction of cell-surface integrins with extracellular matrix ligands can regulate growth factor responses and thus may influence the effect mediated by the growth factor. Here we show, by using mice lacking the alpha(6) integrin receptor for laminins, that myelin-forming oligodendrocytes activate an integrin-regulated switch in survival signalling when they contact axonal laminins. This switch alters survival signalling mediated by neuregulin from dependence on the phosphatidylinositol-3-OH kinase (PI(3)K) pathway to dependence on the mitogen-activated kinase pathway. The consequent enhanced survival provides a mechanism for target-dependent selection during development of the central nervous system. This integrin-regulated switch reverses the capacity of neuregulin to inhibit the differentiation of precursors, thereby explaining how neuregulin subsequently promotes differentiation and survival in myelinating oligodendrocytes. Our results provide a general mechanism by which growth factors can exert apparently contradictory effects at different stages of development in individual cell lineages.
Collapse
Affiliation(s)
- Holly Colognato
- Department of Medical Genetics and Center for Brain Repair, University of Cambridge, Cambridge CB2 2PY, UK
| | | | | | | | | | | | | |
Collapse
|
31
|
Walker JL, Zhang L, Zhou J, Woolkalis MJ, Menko AS. Role for alpha 6 integrin during lens development: Evidence for signaling through IGF-1R and ERK. Dev Dyn 2002; 223:273-84. [PMID: 11836791 DOI: 10.1002/dvdy.10050] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
We show that alpha 6 integrin function was required for normal lens cell differentiation by using an antisense construct to suppress alpha 6 integrin expression. To elucidate the mechanism by which this integrin functions in the regulation of the lens cell differentiation process, we determined the molecular composition of alpha 6 integrin signaling complexes at distinct stages of differentiation in vivo. Because both alpha 6 integrin and insulin-like growth factor-1 (IGF-1) have been implicated in signaling lens cell differentiation, we examined the possibility that they formed a signaling complex in the embryonic lens. Coprecipitation analysis revealed that alpha 6 integrin/IGF-1 receptor complexes were present and that their association was greatest in the equatorial zone, the region of the embryonic lens in which lens cells proliferate and then initiate their differentiation. These results provide in vivo support for the formation of integrin/growth factor receptor signaling complexes. We also found that extracellular signal-regulated kinase (ERK), a downstream effector of both integrin and growth factor receptor signaling pathways, was associated with the alpha 6 integrin signaling complexes in the embryonic lens. This result was supported by our findings that activated ERK, in addition to its nuclear location, localized to lens cell membranes in specific regions of cell-matrix and cell-cell contact. A connection between integrin ligand engagement and ERK activation was shown in vitro after lens cell attachment to laminin. These results demonstrate that alpha 6 integrin function is required for the early stages of lens cell differentiation most likely through its association with the IGF-1 receptor and the activation of ERK.
Collapse
Affiliation(s)
- Janice L Walker
- Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | | | | | | | | |
Collapse
|
32
|
Abstract
Cellular interactions with laminin are important for numerous morphogenetic events. In Xenopus, the first of these is neurulation. The integrin alpha6 subunit mediates an attachment of the cells of the neural plate to the underlying basal lamina. A disruption of this interaction results in embryos that fail to neurulate (T. E. Lallier et al., 1996, Development 122, 2539-2554). Here we provide evidence supporting the specificity of this phenomenon and characterize developmental events as either disrupted or unaffected by a perturbation of alpha6 integrin expression. First, reduction of alpha6 integrin expression does not halt mitotic division throughout the embryo, indicating that the neural defects observed are not simply a global perturbation of all developmental processes. Second, a gene associated with dorsal mesoderm formation, brachyury, is expressed normally in alpha6 integrin-perturbed embryos. Third, the expression of BMP4, noggin, chordin, and follistatin, all of which are critical for neural induction, are at near normal levels. In addition, several genes expressed shortly after neural induction (N-CAM, nrp1, and Xanf1) are not perturbed in nonneurulating embryos. Interestingly, expression of one neural-specific gene (synaptobrevin), which is normally detectable late in neurulation, is abolished in these alpha6 integrin-perturbed embryos. Furthermore, the spatial expression of several transcripts is expanded in alpha6 integrin-perturbed embryos (orthodenticle and engrailed). Taken together, these data indicate that while alpha6 integrin-mediated interactions with laminin are required for neurulation, they are not required for the initial processes of neural induction. However, these cell-extracellular matrix interactions appear to be important in later inductive events and rostrocaudal patterning of the neural tube.
Collapse
Affiliation(s)
- T E Lallier
- Department of Cell Biology and Anatomy, Louisiana State University Medical Center School of Dentistry, 1100 Florida Avenue, New Orleans, Louisiana, 70119, USA.
| | | |
Collapse
|
33
|
Abstract
The laminins are a family of glycoproteins that provide an integral part of the structural scaffolding of basement membranes in almost every animal tissue. Each laminin is a heterotrimer assembled from alpha, beta, and gamma chain subunits, secreted and incorporated into cell-associated extracellular matrices. The laminins can self-assemble, bind to other matrix macromolecules, and have unique and shared cell interactions mediated by integrins, dystroglycan, and other receptors. Through these interactions, laminins critically contribute to cell differentiation, cell shape and movement, maintenance of tissue phenotypes, and promotion of tissue survival. Recent advances in the characterization of genetic disruptions in humans, mice, nematodes and flies have revealed developmental roles for the different laminin subunits in diverse cell types, affecting differentiation from blastocyst formation to the post-natal period. These genetic defects have challenged some of the previous concepts about basement membranes and have shed new light on the diversity and complexity of laminin functions as well as established the molecular basis of several human diseases.
Collapse
Affiliation(s)
- H Colognato
- Department of Pathology & Laboratory Medicine, Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA
| | | |
Collapse
|
34
|
|
35
|
Carl TF, Dufton C, Hanken J, Klymkowsky MW. Inhibition of neural crest migration in Xenopus using antisense slug RNA. Dev Biol 1999; 213:101-15. [PMID: 10452849 DOI: 10.1006/dbio.1999.9320] [Citation(s) in RCA: 116] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Based primarily on studies in the chick, it has been assumed that the zinc finger transcription factor Slug is required for neural crest migration. In the mouse, however, Slug is not expressed in the premigratory neural crest, which forms normally in Slug -/- animals. To study the role of Slug in Xenopus laevis, we used the injection of XSlug antisense RNA and tissue transplantation. Injection of Slug antisense RNA did not suppress the early expression of the related gene XSnail, but led to reduced expression of both XSlug and XSnail in later stage embryos, whereas the expression of another neural crest marker, XTwist, was not affected. Down-regulation of XSlug and XSnail was associated with the inhibition of neural crest cell migration and the reduction or loss of many neural crest derivatives. In particular, the formation of rostral cartilages was often highly aberrant, whereas the posterior cartilages were less frequently affected. The effects of Slug antisense RNA on neural crest migration and cartilage formation were rescued by the injection of either XSlug or XSnail mRNA. These studies indicate that XSlug is required for neural crest migration, that XSlug and XSnail may be functionally redundant, and that both genes are required to maintain each other's expression in the neural crest development of xenopus laevis.
Collapse
Affiliation(s)
- T F Carl
- Environmental, Population, and Organismic Biology, University of Colorado at Boulder, Boulder, Colorado 80309, USA
| | | | | | | |
Collapse
|
36
|
Carroll T, Wallingford J, Seufert D, Vize PD. Molecular regulation of pronephric development. Curr Top Dev Biol 1999; 44:67-100. [PMID: 9891877 DOI: 10.1016/s0070-2153(08)60467-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- T Carroll
- Department of Zoology, University of Texas, Austin 78712, USA
| | | | | | | |
Collapse
|
37
|
Cai H, Krätzschmar J, Alfandari D, Hunnicutt G, Blobel CP. Neural crest-specific and general expression of distinct metalloprotease-disintegrins in early Xenopus laevis development. Dev Biol 1998; 204:508-24. [PMID: 9882486 DOI: 10.1006/dbio.1998.9017] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Metalloprotease-disintegrins are a family of membrane-anchored glycoproteins that have been implicated in diverse cellular processes, including fertilization and myoblast fusion, release of TNFalpha from the plasma membrane, and neurogenesis. Here we report the cloning of cDNAs encoding three full-length (xMDC9, xMDC11b, and xMDC13), and one partial (xMDC11a) metalloprotease-disintegrin from the amphibian Xenopus laevis, and the analysis of their expression during early X. laevis development and in adult tissues. The most notable finding was the highly localized and specific expression pattern of xmdc11a at the tailbud stage in the cranial neural crest and in a subset of neural tube cells in the trunk region. In contrast, expression of the closely related xmdc11b was not detectable during the early stages of X. laevis development, and remained low in the adult tissues examined here. Distinct expression patterns were also observed for two other highly related X. laevis genes, xmdc13 and adam13 (Alfandari et al., 1997). While adam13 is expressed in the somitic mesoderm and in neural crest cells, but not in adult testis, xmdc13 expression is low and ubiquitous in the developing embryo, but is clearly present in adult testis. Finally, xmdc9, the putative orthologue of human and mouse mdc9, was found at all stages of development, and in all tissues examined, suggesting a function that may be utilized by most or all cells. The noteworthy features of these four xmdc genes and the implications of their distinct spatial and temporal expression patterns are discussed.
Collapse
Affiliation(s)
- H Cai
- Cellular Biochemistry and Biophysics Program, Sloan-Kettering Institute, Memorial Sloan-Kettering Cancer Center, New York, New York, 10021, USA
| | | | | | | | | |
Collapse
|
38
|
Whittaker CA, Desimone DW. Molecular cloning and developmental expression of the Xenopus homolog of integrin alpha 4. Ann N Y Acad Sci 1998; 857:56-73. [PMID: 9917832 DOI: 10.1111/j.1749-6632.1998.tb10107.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Integrin receptors containing an alpha 4 subunit mediate cell-cell adhesion by binding to VCAM and MadCAM-1 in addition to supporting cell-extracellular matrix (ECM) adhesion by binding to the alternatively spliced V-region of fibronectin (FN). Studies in chick and mouse embryos have implicated these integrins in neural crest migration, myotube formation, heart development, and placentation. Because integrin-FN adhesive interactions have been shown to play essential roles in mammalian development, studies were initiated of integrin alpha 4 in amphibian embryos, which are better suited to experimental analyses of the earliest stages of embryogenesis. Here, the cDNA cloning and pattern of expression of the Xenopus laevis homolog of integrin alpha 4 is reported. Xenopus alpha 4 is 55% identical at the amino-acid level to both its human and mouse counterparts, including conservation of an alpha 4-specific protease cleavage site, 11 potential N-linked glycosylation sites, and 24 cysteine residues. In situ hybridization analysis reveals that transcripts encoding alpha 4 are expressed in epidermis and the branchial arches. Although alpha 4 transcripts can be detected as early as gastrulation, the protein is observed only after tailbud stages of development and is spatially restricted to the epidermis and gills of tadpole stage embryos. From these data it is concluded that Xenopus integrin alpha 4 has structural features in common with other vertebrate alpha 4 homologs, but is detected in a more restricted tissue distribution during development than alpha 4 in other species.
Collapse
Affiliation(s)
- C A Whittaker
- Department of Cell Biology, University of Virginia, Charlottesville 22908, USA
| | | |
Collapse
|
39
|
Georges-Labouesse E, Mark M, Messaddeq N, Gansmüller A. Essential role of alpha 6 integrins in cortical and retinal lamination. Curr Biol 1998; 8:983-6. [PMID: 9742403 DOI: 10.1016/s0960-9822(98)70402-6] [Citation(s) in RCA: 251] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Extracellular matrix (ECM) is believed to play important roles in many aspects of nervous system development [1]. The laminins are ECM glycoproteins expressed in neural tissues and are potent stimulators of neurite outgrowth in vitro [1-3]. Genetic approaches using Drosophila and Caenorhabditis elegans have demonstrated a role for laminin and a laminin receptor in vivo in axon pathfinding and fasciculation, respectively [4,5]. In higher organisms, however, the role of laminins in the development of the nervous system is poorly understood. Integrins alpha 6 beta 1 and alpha 6 beta 4 are major laminin receptors. A role for the alpha 6 integrin in neurulation has been reported in amphibians [6]. We previously described mice lacking integrin alpha 6; these mice died at birth with severe skin blistering [7]. Detailed analyses of integrin alpha 6-/- mice reported here revealed abnormalities in the laminar organization of the developing cerebral cortex and retina. Ectopic neuroblastic outgrowths were found on the brain surface and in the vitreous body in the eye. Alterations of laminin deposition were found in mutant brains. Thus, this study provides evidence for an essential role of integrin-laminin interactions in the proper development of the nervous system. These observations are particularly significant given the recent report that human patients suffering from epidermolysis bullosa can carry mutations in ITGA6, the gene encoding the alpha 6 integrin chain [8,9].
Collapse
Affiliation(s)
- E Georges-Labouesse
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM/ULP, C.U. de Strasbourg, France.
| | | | | | | |
Collapse
|
40
|
Jacques TS, Relvas JB, Nishimura S, Pytela R, Edwards GM, Streuli CH, ffrench-Constant C. Neural precursor cell chain migration and division are regulated through different beta1 integrins. Development 1998; 125:3167-77. [PMID: 9671589 DOI: 10.1242/dev.125.16.3167] [Citation(s) in RCA: 196] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Proliferation and tangential migration of neural precursor cells are essential determinants of CNS development. We have established cell culture models of both these processes using neural precursor cells grown as neurospheres. The pattern of migration that we observe in these cells is homotypic and occurs in the absence of a glial or neuronal scaffold, and is therefore equivalent to that previously described as chain migration. To determine the role of integrins in proliferation and migration, we have analysed the expression pattern of integrins on neurosphere cells and then performed blocking peptide and antibody experiments. Neurosphere cells express five major integrins, alpha5 beta1, alpha 6Abeta1, alphav beta1, alphav beta5 and alpha vbeta8 and, in addition, express low levels of alpha 6Bbeta1. Chain migration is inhibited by blocking the alpha 6beta1 integrin. Proliferation, by contrast, is inhibited by blocking the other beta1 integrins, alphav beta1 and alpha5 beta1. These results show that integrins are important regulators of neural precursor cell behaviour, with distinct beta1 integrins regulating proliferation and migration. They also demonstrate a novel role for the alpha6 beta1 integrin in the cell-cell interactions underlying homotypic chain migration.
Collapse
Affiliation(s)
- T S Jacques
- Wellcome/CRC Institute of Developmental Biology and Cancer, Tennis Court Road, Cambridge, CB2 1QR and Department of Medical Genetics, University of Cambridge, Cambridge, UK
| | | | | | | | | | | | | |
Collapse
|
41
|
Skalski M, Alfandari D, Darribère T. A key function for alphav containing integrins in mesodermal cell migration during Pleurodeles waltl gastrulation. Dev Biol 1998; 195:158-73. [PMID: 9520332 DOI: 10.1006/dbio.1997.8838] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
During cleavage of Pleurodeles waltl amphibian embryos, inner cells of the blastocoel roof (presumptive ectodermal and mesodermal cells) organize a fibrillar extracellular matrix (ECM) containing fibronectin on their basal surface by a beta1-integrin-dependent process. This matrix is used as a migratory substrate by mesodermal cells during gastrulation. While alpha5beta1 integrin is expressed on both ectodermal and mesodermal cell surface, we have shown previously that alphav containing integrins are essentially restricted to the surface of mesodermal cells (Alfandari, D., Whittaker, C. A., DeSimone, D. W., and Darribère, T., Dev. Biol. 170, 249-261, 1995). To investigate the function of alphav integrins during gastrulation, we have generated a function blocking antibody directed against the extracellular domain of the Pleurodeles integrin alphav subunit. The antibody did not prevent fibronectin fibril formation, whereas an antibody against the alpha5beta1 integrin did. When injected into the blastocoel, the antibody against integrin alphav subunit perturbed gastrulation and further development in a stage-dependent manner. Developmental defects were correlated to an abnormal positioning of the mesoderm layer. In vitro, the antibody blocked spreading of mesodermal cell to fibronectin or blastocoel roof ECM but not their attachment. In contrast, the antibody directed against the alpha5beta1 integrin inhibited both cell attachment and spreading to the same substrates. We propose that the alpha5beta1 integrin is required for fibronectin assembly into fibrils and mesodermal cell attachment to the blastocoel roof ECM, while the alphav containing integrins are necessary for cell spreading, and possibly migration, on this complex network.
Collapse
Affiliation(s)
- M Skalski
- Equipe Adhesion et Migration Cellulaires, Université P. et M. Curie, CNRS UMR 7622, 9 Quai Saint-Bernard, Paris Cedex 05, 75252, France
| | | | | |
Collapse
|
42
|
Abstract
The inner ear is a complex sensory organ responsible for balance and sound detection in vertebrates. It originates from a transient embryonic structure, the otic vesicle, that contains all of the information to develop autonomously into the mature inner ear. We review here the development of the otic vesicle, bringing together classical embryological experiments and recent genetic and molecular data. The specification of the prospective ectoderm and its commitment to the otic fate are very early events and can be related to the expression of genes with restricted expression domains. A combinatorial gene expression model for placode specification and diversification, based on classical embryological evidence and gene expression patterns, is discussed. The formation of the otic vesicle is dependent on inducing signals from endoderm, mesoderm and neuroectoderm. Ear induction consists of a sequence of discrete instructions from those tissues that confer its final identity on the otic field, rather than a single all-or-none process. The important role of the neural tube in otic development is highlighted by the abnormalities observed in mouse mutants for the Hoxa1, kreisler and fgf3 genes and those reported in retinoic acid-deficient quails. Still, the nature of the relation between the neural tube and otic development remains unclear. Gene targeting experiments in the mouse have provided evidence for genes potentially involved in regional and cell-fate specification in the inner ear. The disruption of the mouse Brn3.1 gene identifies the first mutation affecting sensory hair-cell specification, and mutants for Pax2 and Nkx5.1 genes show their requirement for the development of specific regions of the otic vesicle. Several growth-factors contribute to the patterned cell proliferation of the otic vesicle. Among these, IGF-I and FGF-2 are expressed in the otic vesicle and may act in an autocrine manner. Finally, little is known about early mechanisms involved in guiding ear innervation. However, targeted disruption of genes coding for neurotrophins and Trk receptors have shown that once synaptic contacts are established, they depend on specific trophic interactions that involve these two gene families. The accessibility of new cellular and molecular approaches are opening new perspectives in vertebrate development and are also starting to be applied to ear development. This will allow this classical and attractive model system to see a rapid progress in the near future.
Collapse
Affiliation(s)
- M Torres
- Departamento de Inmunologiá y Oncologiá, Centro Nacional de Biotecnología CSIC, Cantoblanco, E-28049, Madrid, Spain
| | | |
Collapse
|
43
|
Sung U, O'Rear JJ, Yurchenco PD. Localization of heparin binding activity in recombinant laminin G domain. EUROPEAN JOURNAL OF BIOCHEMISTRY 1997; 250:138-43. [PMID: 9432001 DOI: 10.1111/j.1432-1033.1997.00138.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Basement membrane laminin (laminin-1) is a multidomain glycoprotein that interacts with itself, heparin and cells. The interaction with heparin/heparan sulfate proteglycans is thought to be important for the architectural formation of basement membranes and adhesion to cells. The major heparin binding site has been known to reside in the long arm globular domain (G domain). The G domain is in turn subdivided into five subdomains (G1-G5). In order to localize the heparin binding regions further, recombinant G domains (rG and rG5) were expressed in Sf9 insect cells using baculovirus expression vector. By the limited proteolysis of recombinant G domains followed by either heparin affinity HPLC or overlay with radiolabeled heparin, the relative affinity of each subdomain to heparin was assigned as G1>G2 = G4>G5>G3, such that G1 bound strongly and G3 not at all. Since the activity in G1-G3 is cryptic in intact laminin long arm [Sung, U., O'Rear, J. J. & Yurchenco, P. D. (1993) J. Cell Biol. 123, 1255-1268], the active heparin binding site of G domain appears to be located in G4 and proximal G5.
Collapse
Affiliation(s)
- U Sung
- Department of Pathology, Robert Wood Johnson Medical School and University of Medicine and Dentistry of New Jersey, Piscataway, USA
| | | | | |
Collapse
|
44
|
Meng F, Whittaker CA, Ransom DG, DeSimone DW. Cloning and characterization of cDNAs encoding the integrin alpha2 and alpha3 subunits from Xenopus laevis. Mech Dev 1997; 67:141-55. [PMID: 9392512 DOI: 10.1016/s0925-4773(97)00108-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Integrins containing the alpha2 and alpha3 subunits associate with the beta1 subunit to form distinct receptors with partially overlapping adhesive specificities. We report the cloning and sequence of cDNAs that encode the Xenopus orthologues of integrins alpha2 and alpha3 and the expression of these subunits during embryogenesis. Integrin alpha2 and alpha3 mRNAs are first expressed in the dorsal mesoderm and developing notochord at gastrulation. We also show that alpha3 mRNAs are expressed in the entire marginal zone of gastrulae dorsalized with LiCl but that this localization is lost in embryos ventralized by ultraviolet light. Immunoblots reveal that the alpha3 protein is expressed throughout early development, however, the alpha2 protein is not detected until late tailbud stages. Injection of full-length alpha3 transcripts into the animal poles of fertilized eggs results in embryonic defects in paraxial mesoderm attributed to the failure of somites to form segments. Injection of the alpha3 transcripts into the vegetal pole and overexpression of a 5'-truncated alpha3 control construct have no apparent affect on development or somite formation. These data suggest that normal position-specific expression of integrins is important in maintaining the proper organization of tissues during early amphibian morphogenesis.
Collapse
Affiliation(s)
- F Meng
- Department of Cell Biology, School of Medicine, University of Virginia, Charlottesville 22908, USA
| | | | | | | |
Collapse
|
45
|
Shilling FM, Krätzschmar J, Cai H, Weskamp G, Gayko U, Leibow J, Myles DG, Nuccitelli R, Blobel CP. Identification of metalloprotease/disintegrins in Xenopus laevis testis with a potential role in fertilization. Dev Biol 1997; 186:155-64. [PMID: 9205136 DOI: 10.1006/dbio.1997.8586] [Citation(s) in RCA: 52] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Proteins containing a membrane-anchored metalloprotease domain, a disintegrin domain, and a cysteine-rich region (MDC proteins) are thought to play an important role in mammalian fertilization, as well as in somatic cell-cell interactions. We have identified PCR sequence tags encoding the disintegrin domain of five distinct MDC proteins from Xenopus laevis testis cDNA. Four of these sequence tags (xMDC9, xMDC11.1, xMDC11.2, and xMDC13) showed strong similarity to known mammalian MDC proteins, whereas the fifth (xMDC16) apparently represents a novel family member. Northern blot analysis revealed that the mRNA for xMDC16 was only expressed in testis, and not in heart, muscle, liver, ovaries, or eggs, whereas the mRNAs corresponding to the four other PCR products were expressed in testis and in some or all somatic tissues tested. The xMDC16 protein sequence, as predicted from the full-length cDNA, contains a metalloprotease domain with the active-site sequence HEXXH, a disintegrin domain, a cysteine-rich region, an EGF repeat, a transmembrane domain, and a short cytoplasmic tail. To study a potential role for these xMDC proteins in fertilization, peptides corresponding to the predicted integrin-binding domain of each protein were tested for their ability to inhibit X. laevis fertilization. Cyclic and linear xMDC16 peptides inhibited fertilization in a concentration-dependent manner, whereas xMDC16 peptides that were scrambled or had certain amino acid replacements in the predicted integrin-binding domain did not affect fertilization. Cyclic and linear xMDC9 peptides and linear xMDC13 peptides also inhibited fertilization similarly to xMDC16 peptides, whereas peptides corresponding to the predicted integrin-binding site of xMDC11.1 and xMDC11.2 did not. These results are discussed in the context of a model in which multiple MDC protein-receptor interactions are necessary for fertilization to occur.
Collapse
Affiliation(s)
- F M Shilling
- Section of Molecular and Cellular Biology, University of California, Davis 95616, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Alfandari D, Wolfsberg TG, White JM, DeSimone DW. ADAM 13: a novel ADAM expressed in somitic mesoderm and neural crest cells during Xenopus laevis development. Dev Biol 1997; 182:314-30. [PMID: 9070330 DOI: 10.1006/dbio.1996.8458] [Citation(s) in RCA: 93] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Embryonic development involves a series of cell adhesive interactions that provide mechanical and instructive information required for morphogenesis. The ADAMs family of membrane-anchored proteins, containing a disintegrin and metalloprotease domain, is well suited for participating in such developmental events. They encode not only a potential adhesive function, through an integrin-binding disintegrin domain, but also a potential antiadhesive function, through a zinc-dependent metalloprotease domain. In order to investigate the role of ADAMs in early development we cloned a cDNA encoding a novel member of the ADAM family from a Xenopus laevis neurula stage library. We call this cDNA, and the 915-amino-acid protein it encodes, ADAM 13, X-ADAM 13 RNA is expressed during embryogenesis from the midblastula stage through tadpole stage 45. X-ADAM 13 is localized to somitic mesoderm and cranial neural crest cells during gastrulation, neurulation, and in tail bud stages. Sequence analyses of the X-ADAM 13 metalloprotease and disintegrin domains indicate that the protein is likely to be involved in both proteolytic and cell-adhesive functions. The X-ADAM 13 sequence is most closely related to that of mouse meltrin alpha, which is implicated in myoblast fusion. Our data suggest that X-ADAM 13 may be involved in neural crest cell adhesion and migration as well as myoblast differentiation.
Collapse
Affiliation(s)
- D Alfandari
- Department of Cell Biology, Health Sciences Center, University of Virginia, Charlottesville 22908, USA
| | | | | | | |
Collapse
|