1
|
Matrigel is required for efficient differentiation of isolated, stem cell-derived otic vesicles into inner ear organoids. Stem Cell Res 2021; 53:102295. [PMID: 33773390 PMCID: PMC8360351 DOI: 10.1016/j.scr.2021.102295] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 02/09/2021] [Accepted: 03/12/2021] [Indexed: 01/07/2023] Open
Abstract
Inner ear organoids derived from pluripotent stem cells could be a useful model system to study development, disease, and regeneration. However, there is considerable heterogeneity in the size, morphology, and efficiency of organoid production using standard protocols. Greater control of the culture microenvironment could decrease heterogeneity and increase the yield of organoids. Animal-derived otic vesicles show some autonomy during development and can differentiate into cochlear and vestibular domains in mesenchyme-free ex vivo culture. Therefore, we investigated whether stem cell-derived otic vesicles can autonomously generate inner ear organoids. Isolated, stem cell-derived vesicles grew into cyst-like organoids with high efficiency, over 90%, when embedded in droplets of the basement membrane matrix Matrigel. Though nearly all vesicles within the aggregate were competent to mature into organoids, the efficiency of organoid production depended on the stage of vesicle isolation and required supplementation with Matrigel.
Collapse
|
2
|
Holt E, Stanton-Turcotte D, Iulianella A. Development of the Vertebrate Trunk Sensory System: Origins, Specification, Axon Guidance, and Central Connectivity. Neuroscience 2021; 458:229-243. [PMID: 33460728 DOI: 10.1016/j.neuroscience.2020.12.037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 12/09/2020] [Accepted: 12/31/2020] [Indexed: 12/26/2022]
Abstract
Crucial to an animal's movement through their environment and to the maintenance of their homeostatic physiology is the integration of sensory information. This is achieved by axons communicating from organs, muscle spindles and skin that connect to the sensory ganglia composing the peripheral nervous system (PNS), enabling organisms to collect an ever-constant flow of sensations and relay it to the spinal cord. The sensory system carries a wide spectrum of sensory modalities - from sharp pain to cool refreshing touch - traveling from the periphery to the spinal cord via the dorsal root ganglia (DRG). This review covers the origins and development of the DRG and the cells that populate it, and focuses on how sensory connectivity to the spinal cord is achieved by the diverse developmental and molecular processes that control axon guidance in the trunk sensory system. We also describe convergences and differences in sensory neuron formation among different vertebrate species to gain insight into underlying developmental mechanisms.
Collapse
Affiliation(s)
- Emily Holt
- Department of Medical Neuroscience, Faculty of Medicine, Dalhousie University, and Brain Repair Centre, Life Science Research Institute, 1348 Summer Street, Halifax, Nova Scotia B3H-4R2, Canada
| | - Danielle Stanton-Turcotte
- Department of Medical Neuroscience, Faculty of Medicine, Dalhousie University, and Brain Repair Centre, Life Science Research Institute, 1348 Summer Street, Halifax, Nova Scotia B3H-4R2, Canada
| | - Angelo Iulianella
- Department of Medical Neuroscience, Faculty of Medicine, Dalhousie University, and Brain Repair Centre, Life Science Research Institute, 1348 Summer Street, Halifax, Nova Scotia B3H-4R2, Canada.
| |
Collapse
|
3
|
Grocott T, Lozano-Velasco E, Mok GF, Münsterberg AE. The Pax6 master control gene initiates spontaneous retinal development via a self-organising Turing network. Development 2020; 147:dev185827. [PMID: 33214222 PMCID: PMC7774904 DOI: 10.1242/dev.185827] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 11/05/2020] [Indexed: 12/14/2022]
Abstract
Understanding how complex organ systems are assembled from simple embryonic tissues is a major challenge. Across the animal kingdom a great diversity of visual organs are initiated by a 'master control gene' called Pax6, which is both necessary and sufficient for eye development. Yet precisely how Pax6 achieves this deeply homologous function is poorly understood. Using the chick as a model organism, we show that vertebrate Pax6 interacts with a pair of morphogen-coding genes, Tgfb2 and Fst, to form a putative Turing network, which we have computationally modelled. Computer simulations suggest that this gene network is sufficient to spontaneously polarise the developing retina, establishing the first organisational axis of the eye and prefiguring its further development. Our findings reveal how retinal self-organisation may be initiated independently of the highly ordered tissue interactions that help to assemble the eye in vivo These results help to explain how stem cell aggregates spontaneously self-organise into functional eye-cups in vitro We anticipate these findings will help to underpin retinal organoid technology, which holds much promise as a platform for disease modelling, drug development and regenerative therapies.
Collapse
Affiliation(s)
- Timothy Grocott
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, UK
| | | | | | | |
Collapse
|
4
|
Lu J, Doyle AD, Shinsato Y, Wang S, Bodendorfer MA, Zheng M, Yamada KM. Basement Membrane Regulates Fibronectin Organization Using Sliding Focal Adhesions Driven by a Contractile Winch. Dev Cell 2020; 52:631-646.e4. [PMID: 32004443 DOI: 10.1016/j.devcel.2020.01.007] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 09/28/2019] [Accepted: 01/07/2020] [Indexed: 01/05/2023]
Abstract
We have discovered that basement membrane and its major components can induce rapid, strikingly robust fibronectin organization. In this new matrix assembly mechanism, α5β1 integrin-based focal adhesions slide actively on the underlying matrix toward the ventral cell center through the dynamic shortening of myosin IIA-associated actin stress fibers to drive rapid fibronectin fibrillogenesis distal to the adhesion. This mechanism contrasts with classical fibronectin assembly based on stable or fixed-position focal adhesions containing αVβ3 integrins plus α5β1 integrin translocation into proximal fibrillar adhesions. On basement membrane components, these sliding focal adhesions contain standard focal adhesion constituents but completely lack classical αVβ3 integrins. Instead, peripheral α3β1 or α2β1 adhesions mediate initial cell attachment but over time are switched to α5β1 integrin-based sliding focal adhesions to assemble fibronectin matrix. This basement-membrane-triggered mechanism produces rapid fibronectin fibrillogenesis, providing a mechanistic explanation for the well-known widespread accumulation of fibronectin at many organ basement membranes.
Collapse
Affiliation(s)
- Jiaoyang Lu
- School of Medicine, Shandong University, Jinan, Shandong 250012, China; Cell Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA; Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Department of Gastroenterology, Qilu Hospital, Jinan, Shandong 250012, China
| | - Andrew D Doyle
- Cell Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yoshinari Shinsato
- Cell Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Shaohe Wang
- Cell Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Molly A Bodendorfer
- Cell Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Minhua Zheng
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Kenneth M Yamada
- Cell Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
5
|
Puelles L, Tvrdik P, Martínez-de-la-torre M. The Postmigratory Alar Topography of Visceral Cranial Nerve Efferents Challenges the Classical Model of Hindbrain Columns. Anat Rec (Hoboken) 2018; 302:485-504. [DOI: 10.1002/ar.23830] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 12/07/2017] [Accepted: 12/08/2017] [Indexed: 12/21/2022]
Affiliation(s)
- Luis Puelles
- Department of Human Anatomy and Psychobiology and IMIB-Arrixaca Institute, School of Medicine; University of Murcia; Murcia 30071 Spain
| | - Petr Tvrdik
- Department of Neurosurgery-Physiology; University of Utah; Salt Lake City, Utah 84112
| | - Margaret Martínez-de-la-torre
- Department of Human Anatomy and Psychobiology and IMIB-Arrixaca Institute, School of Medicine; University of Murcia; Murcia 30071 Spain
| |
Collapse
|
6
|
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] [Scholar 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
|
7
|
de Almeida PG, Pinheiro GG, Nunes AM, Gonçalves AB, Thorsteinsdóttir S. Fibronectin assembly during early embryo development: A versatile communication system between cells and tissues. Dev Dyn 2016; 245:520-35. [PMID: 26845241 DOI: 10.1002/dvdy.24391] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Revised: 01/20/2016] [Accepted: 01/25/2016] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Fibronectin extracellular matrix is essential for embryogenesis. Its assembly is a cell-mediated process where secreted fibronectin dimers bind to integrin receptors on receiving cells, which actively assemble fibronectin into a fibrillar matrix. During development, paracrine communication between tissues is crucial for coordinating morphogenesis, typically being mediated by growth factors and their receptors. Recent reports of situations where fibronectin is produced by one tissue and assembled by another, with implications on tissue morphogenesis, suggest that fibronectin assembly may also be a paracrine communication event in certain contexts. RESULTS Here we addressed which tissues express fibronectin (Fn1) while also localizing assembled fibronectin matrix and determining the mRNA expression and/or protein distribution pattern of integrins α5 and αV, α chains of the major fibronectin assembly receptors, during early chick and mouse development. We found evidence supporting a paracrine system in fibronectin matrix assembly in several tissues, including immature mesenchymal tissues, components of central and peripheral nervous system and developing muscle. CONCLUSIONS Thus, similarly to growth factor signaling, fibronectin matrix assembly during early development can be both autocrine and paracrine. We therefore propose that it be considered a cell-cell communication event at the same level and significance as growth factor signaling during embryogenesis.
Collapse
Affiliation(s)
- Patrícia Gomes de Almeida
- Centre for Ecology, Evolution and Environmental Change (cE3c), Departamento de Biologia Animal, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
| | - Gonçalo G Pinheiro
- Centre for Ecology, Evolution and Environmental Change (cE3c), Departamento de Biologia Animal, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
| | - Andreia M Nunes
- Centre for Ecology, Evolution and Environmental Change (cE3c), Departamento de Biologia Animal, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
| | - André B Gonçalves
- Centre for Ecology, Evolution and Environmental Change (cE3c), Departamento de Biologia Animal, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
| | - Sólveig Thorsteinsdóttir
- Centre for Ecology, Evolution and Environmental Change (cE3c), Departamento de Biologia Animal, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
| |
Collapse
|
8
|
Interepithelial signaling with nephric duct is required for the formation of overlying coelomic epithelial cell sheet. Proc Natl Acad Sci U S A 2014; 111:6660-5. [PMID: 24753584 DOI: 10.1073/pnas.1316728111] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
In most organs of the body, epithelial tissues are supported by their own basement membrane and underlying stroma, the latter being regarded as a complex of amorphous cells, extracellular matrices, and soluble factors. We demonstrate here that an epithelial tube can serve as a component of stroma that supports the formation of epithelial cell sheet derived from a different origin. During development of the mesonephros in chicken embryos, the intermediate mesoderm (IMM), which contains the Wolffian duct (WD) and its associated tubules, is overlain by a sheet of epithelial cells derived from lateral plate (coelomic) mesoderm. We describe that in normal embryos, epitheliogenesis of IMM tubes and the adjacent coelomic cell sheet proceed in a coordinated manner. When the WD was surgically ablated, the overlying coelomic epithelium exhibited aberrant morphology accompanied by a punctated basement membrane. Furthermore, the WD-ablated coelomic epithelium became susceptible to latent external stress; electroporation of Rac1 resulted in epithelial-to-mesenchymal transitions (EMTs) within the coelomic epithelium. The distorted coelomic epithelium was rescued by implanting fibronectin-producing cells in place of the WD, suggesting that fibronectin provided by WD has an important role acting interepithelially. This notion was corroborated further by directly visualizing a translocation of EGFP-tagged fibronectin from fibronectin-producing to -receiving epithelia in vivo. Our findings provide a novel insight into interepithelial signaling that also might occur in adult tissues to protect against EMT and suggest a possible new target for anticancer therapeutic strategy.
Collapse
|
9
|
Borycki AG. The myotomal basement membrane: insight into laminin-111 function and its control by Sonic hedgehog signaling. Cell Adh Migr 2013; 7:72-81. [PMID: 23287393 DOI: 10.4161/cam.23411] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The importance of laminin-containing basement membranes (BM) for adult muscle function is well established, in particular due to the severe phenotype of congenital muscular dystrophies in patients with mutations disrupting the BM-muscle cell interaction. Developing muscles in the embryo are also dependent on an intact BM. However, the processes controlled by BM-muscle cell interactions in the embryo are only beginning to be elucidated. In this review, we focus on the myotomal BM to illustrate the critical role of laminin-111 in BM assembly and function at the surface of embryonic muscle cells. The myotomal BM provides also an interesting paradigm to study the complex interplay between laminins-containing BM and growth factor-mediated signaling and activity.
Collapse
|
10
|
Lessons from the embryonic neural stem cell niche for neural lineage differentiation of pluripotent stem cells. Stem Cell Rev Rep 2012; 8:813-29. [PMID: 22628111 PMCID: PMC3412081 DOI: 10.1007/s12015-012-9381-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Pluripotent stem cells offer an abundant and malleable source for the generation of differentiated cells for transplantation as well as for in vitro screens. Patterning and differentiation protocols have been developed to generate neural progeny from human embryonic or induced pluripotent stem cells. However, continued refinement is required to enhance efficiency and to prevent the generation of unwanted cell types. We summarize and interpret insights gained from studies of embryonic neuroepithelium. A multitude of factors including soluble molecules, interactions with the extracellular matrix and neighboring cells cooperate to control neural stem cell self-renewal versus differentiation. Applying these findings and concepts to human stem cell systems in vitro may yield more appropriately patterned cell types for biomedical applications.
Collapse
|
11
|
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
|
12
|
Rifes P, Thorsteinsdóttir S. Extracellular matrix assembly and 3D organization during paraxial mesoderm development in the chick embryo. Dev Biol 2012; 368:370-81. [DOI: 10.1016/j.ydbio.2012.06.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2012] [Revised: 06/01/2012] [Accepted: 06/02/2012] [Indexed: 10/28/2022]
|
13
|
Szabó A, Rupp PA, Rongish BJ, Little CD, Czirók A. Extracellular matrix fluctuations during early embryogenesis. Phys Biol 2011; 8:045006. [PMID: 21750366 DOI: 10.1088/1478-3975/8/4/045006] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Extracellular matrix (ECM) movements and rearrangements were studied in avian embryos during early stages of development. We show that the ECM moves as a composite material, whereby distinct molecular components as well as spatially separated layers exhibit similar displacements. Using scanning wide field and confocal microscopy we show that the velocity field of ECM displacement is smooth in space and that ECM movements are correlated even at locations separated by several hundred micrometers. Velocity vectors, however, strongly fluctuate in time. The autocorrelation time of the velocity fluctuations is less than a minute. Suppression of the fluctuations yields a persistent movement pattern that is shared among embryos at equivalent stages of development. The high resolution of the velocity fields allows a detailed spatio-temporal characterization of important morphogenetic processes, especially tissue dynamics surrounding the embryonic organizer (Hensen's node).
Collapse
Affiliation(s)
- A Szabó
- Department of Anatomy & Cell Biology, University of Kansas Medical Center, Kansas City, KS, USA. Department of Biological Physics, Eotvos University, Budapest, Hungary
| | | | | | | | | |
Collapse
|
14
|
Schwarzbauer JE, DeSimone DW. Fibronectins, their fibrillogenesis, and in vivo functions. Cold Spring Harb Perspect Biol 2011; 3:cshperspect.a005041. [PMID: 21576254 DOI: 10.1101/cshperspect.a005041] [Citation(s) in RCA: 299] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Fibronectin (FN) is a multidomain protein with the ability to bind simultaneously to cell surface receptors, collagen, proteoglycans, and other FN molecules. Many of these domains and interactions are also involved in the assembly of FN dimers into a multimeric fibrillar matrix. When, where, and how FN binds to its various partners must be controlled and coordinated during fibrillogenesis. Steps in the process of FN fibrillogenesis including FN self-association, receptor activities, and intracellular pathways have been under intense investigation for years. In this review, the domain organization of FN including the extra domains and variable region that are controlled by alternative splicing are described. We discuss how FN-FN and cell-FN interactions play essential roles in the initiation and progression of matrix assembly using complementary results from cell culture and embryonic model systems that have enhanced our understanding of this process.
Collapse
Affiliation(s)
- Jean E Schwarzbauer
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544, USA.
| | | |
Collapse
|
15
|
The extracellular matrix dimension of skeletal muscle development. Dev Biol 2011; 354:191-207. [PMID: 21420400 DOI: 10.1016/j.ydbio.2011.03.015] [Citation(s) in RCA: 104] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2011] [Revised: 03/10/2011] [Accepted: 03/11/2011] [Indexed: 12/25/2022]
Abstract
Cells anchor to substrates by binding to extracellular matrix (ECM). In addition to this anchoring function however, cell-ECM binding is a mechanism for cells to sense their surroundings and to communicate and coordinate behaviour amongst themselves. Several ECM molecules and their receptors play essential roles in muscle development and maintenance. Defects in these proteins are responsible for some of the most severe muscle dystrophies at every stage of life from neonates to adults. However, recent studies have also revealed a role of cell-ECM interactions at much earlier stages of development as skeletal muscle forms. Here we review which ECM molecules are present during the early phases of myogenesis, how myogenic cells interact with the ECM that surrounds them and the potential consequences of those interactions. We conclude that cell-ECM interactions play significant roles during all stages of skeletal muscle development in the embryo and suggest that this "extracellular matrix dimension" should be added to our conceptual network of factors contributing to skeletal myogenesis.
Collapse
|
16
|
Goody MF, Henry CA. Dynamic interactions between cells and their extracellular matrix mediate embryonic development. Mol Reprod Dev 2010; 77:475-88. [DOI: 10.1002/mrd.21157] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
17
|
Nagy N, Mwizerwa O, Yaniv K, Carmel L, Pieretti-Vanmarcke R, Weinstein BM, Goldstein AM. Endothelial cells promote migration and proliferation of enteric neural crest cells via beta1 integrin signaling. Dev Biol 2009; 330:263-72. [PMID: 19345201 DOI: 10.1016/j.ydbio.2009.03.025] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2008] [Revised: 03/24/2009] [Accepted: 03/26/2009] [Indexed: 10/21/2022]
Abstract
Enteric neural crest-derived cells (ENCCs) migrate along the intestine to form a highly organized network of ganglia that comprises the enteric nervous system (ENS). The signals driving the migration and patterning of these cells are largely unknown. Examining the spatiotemporal development of the intestinal neurovasculature in avian embryos, we find endothelial cells (ECs) present in the gut prior to the arrival of migrating ENCCs. These ECs are patterned in concentric rings that are predictive of the positioning of later arriving crest-derived cells, leading us to hypothesize that blood vessels may serve as a substrate to guide ENCC migration. Immunohistochemistry at multiple stages during ENS development reveals that ENCCs are positioned adjacent to vessels as they colonize the gut. A similar close anatomic relationship between vessels and enteric neurons was observed in zebrafish larvae. When EC development is inhibited in cultured avian intestine, ENCC migration is arrested and distal aganglionosis results, suggesting that ENCCs require the presence of vessels to colonize the gut. Neural tube and avian midgut were explanted onto a variety of substrates, including components of the extracellular matrix and various cell types, such as fibroblasts, smooth muscle cells, and endothelial cells. We find that crest-derived cells from both the neural tube and the midgut migrate avidly onto cultured endothelial cells. This EC-induced migration is inhibited by the presence of CSAT antibody, which blocks binding to beta1 integrins expressed on the surface of crest-derived cells. These results demonstrate that ECs provide a substrate for the migration of ENCCs via an interaction between beta1 integrins on the ENCC surface and extracellular matrix proteins expressed by the intestinal vasculature. These interactions may play an important role in guiding migration and patterning in the developing ENS.
Collapse
Affiliation(s)
- Nandor Nagy
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | | | | | | | | | | | | |
Collapse
|
18
|
Rifes P, Carvalho L, Lopes C, Andrade RP, Rodrigues G, Palmeirim I, Thorsteinsdóttir S. Redefining the role of ectoderm in somitogenesis: a player in the formation of the fibronectin matrix of presomitic mesoderm. Development 2007; 134:3155-65. [PMID: 17670788 DOI: 10.1242/dev.003665] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The absence of ectoderm impairs somite formation in cultured presomitic mesoderm (PSM) explants, suggesting that an ectoderm-derived signal is essential for somitogenesis. Here we show in chick that the standard enzymatic treatments used for explant isolation destroy the fibronectin matrix surrounding the anterior PSM, which fails to form somites when cultured for 6 hours. By contrast, explants isolated with collagenase retain their fibronectin matrix and form somites under identical culture conditions. The additional presence of ectoderm enhances somite formation, whereas endoderm has no effect. Furthermore, we show that pancreatin-isolated PSM explants cultured in fibronectin-supplemented medium, form significantly more somites than control explants. Interestingly, ectoderm is the major producer of fibronectin (Fn1) transcripts, whereas all but the anterior-most region of the PSM expresses the fibronectin assembly receptor, integrin alpha5 (Itga5). We thus propose that the ectoderm-derived fibronectin is assembled by mesodermal alpha5beta1 integrin on the surface of the PSM. Finally, we demonstrate that inhibition of fibronectin fibrillogenesis in explants with ectoderm abrogates somitogenesis. We conclude that a fibronectin matrix is essential for morphological somite formation and that a major, previously unrecognised role of ectoderm in somitogenesis is the synthesis of fibronectin.
Collapse
Affiliation(s)
- Pedro Rifes
- Departamento de Biologia Animal e Centro de Biologia Ambiental, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisboa, Portugal
| | | | | | | | | | | | | |
Collapse
|
19
|
Czirok A, Zamir EA, Filla MB, Little CD, Rongish BJ. Extracellular matrix macroassembly dynamics in early vertebrate embryos. Curr Top Dev Biol 2006; 73:237-58. [PMID: 16782461 DOI: 10.1016/s0070-2153(05)73008-8] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
This chapter focuses on the in vivo macroassembly dynamics of fibronectin and fibrillin-2--two prominent extracellular matrix (ECM) components, present in vertebrate embryos at the earliest stages of development. The ECM is an inherently dynamic structure with a well-defined position fate: ECM filaments are not only anchored to and move with established tissue boundaries, but are repositioned prior to the formation of new anatomical features. We distinguish two ECM filament relocation processes-each operating on different length scales. First, ECM filaments are moved by large-scale tissue motion, which rearranges major organ primordia within the embryo. The second type of motion, on the scale of the individual ECM filaments, is driven by local motility and protrusive activity of nearby cells. The motion decomposition is made practically possible by recent advances in microscopy and high-resolution particle image velocimetry algorithms. We demonstrate that both kinds of motion contribute substantially to the establishment of normal ECM structure, and both must be taken into account when attempting to understand ECM macroassembly during embryonic morphogenesis. The tissue-scale motion changes the local amount (density) and the tissue-level structure (e.g., orientation) of ECM fibers. Local reorganization includes filament assembly and the segregation of ECM into specific patterns. Local reorganization takes place most actively at Hensen's node and around the primitive streak. These regions are also sites of active cell migration, where fibrillin-2 and fibronectin are often colocalized in ECM globules, and new fibrillin-2 foci are deposited. During filament assembly, the globular patches of ECM are joined into larger linear structures in a hierarchical process: increasingly larger structures are created by the aggregation of smaller units. A future understanding of ECM assembly thus requires the study of the complex interactions between biochemical assembly steps, local cell action, and tissue motion.
Collapse
Affiliation(s)
- Andras Czirok
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, 66160, USA
| | | | | | | | | |
Collapse
|
20
|
Linask KK, Manisastry S, Han M. Cross talk between cell-cell and cell-matrix adhesion signaling pathways during heart organogenesis: implications for cardiac birth defects. MICROSCOPY AND MICROANALYSIS : THE OFFICIAL JOURNAL OF MICROSCOPY SOCIETY OF AMERICA, MICROBEAM ANALYSIS SOCIETY, MICROSCOPICAL SOCIETY OF CANADA 2005; 11:200-8. [PMID: 16060972 DOI: 10.1017/s1431927605050440] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2004] [Accepted: 10/27/2004] [Indexed: 05/03/2023]
Abstract
The anterior-posterior and dorsal-ventral progression of heart organogenesis is well illustrated by the patterning and activity of two members of different families of cell adhesion molecules: the calcium-dependent cadherins, specifically N-cadherin, and the extracellular matrix glycoproteins, fibronectin. N-cadherin by its binding to the intracellular molecule beta-catenin and fibronectin by its binding to integrins at focal adhesion sites, are involved in regulation of gene expression by their association with the cytoskeleton and through signal transduction pathways. The ventral precardiac mesoderm cells epithelialize and become stably committed by the activation of these cell-matrix and intracellular signaling transduction pathways. Cross talk between the adhesion signaling pathways initiates the characteristic phenotypic changes associated with cardiomyocyte differentiation: electrical activity and organization of myofibrils. The development of both organ form and function occurs within a short interval thereafter. Mutations in any of the interacting molecules, or environmental insults affecting either of these signaling pathways, can result in embryonic lethality or fetuses born with severe heart defects. As an example, we have defined that exposure of the embryo temporally to lithium during an early sensitive developmental period affects a canonical Wnt pathway leading to beta-catenin stabilization. Lithium exposure results in an anterior-posterior progression of severe cardiac defects.
Collapse
Affiliation(s)
- Kersti K Linask
- Department of Pediatrics, University of South Florida, College of Medicine, The Children's Research Institute, St. Petersburg, FL 33701, USA.
| | | | | |
Collapse
|
21
|
Bajanca F, Luz M, Duxson MJ, Thorsteinsdóttir S. Integrins in the mouse myotome: Developmental changes and differences between the epaxial and hypaxial lineage. Dev Dyn 2004; 231:402-15. [PMID: 15366018 DOI: 10.1002/dvdy.20136] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Integrins are cellular adhesion receptors that mediate signaling and play key roles in the development of multicellular organisms. However, their role in the cellular events leading to myotome formation is completely unknown. Here, we describe the expression patterns of the alpha1, alpha4, alpha5, alpha6, and alpha7 integrin subunits in the mouse myotome and correlate them with the expression of several differentiation markers. Our results indicate that these integrin subunits may be differentially involved in the various phases of myogenic determination and differentiation. A detailed characterization of the myogenic cell types expressing the alpha4 and alpha6 subunits showed a regionalization of the myotome and dermomyotome based on cell-adhesion properties. We conclude that alpha6beta1 may be an early marker of epaxial myogenic progenitor cells. In contrast, alpha4beta1 is up-regulated in the intercalated myotome after myocyte differentiation. Furthermore, alpha4beta1 is expressed in the hypaxial dermomyotome and is maintained by early hypaxial myogenic progenitor cells colonizing the myotome.
Collapse
Affiliation(s)
- Fernanda Bajanca
- Departamento de Biologia Animal, Centro de Biologia Ambiental, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
| | | | | | | |
Collapse
|
22
|
Jarov A, Williams KP, Ling LE, Koteliansky VE, Duband JL, Fournier-Thibault C. A dual role for Sonic hedgehog in regulating adhesion and differentiation of neuroepithelial cells. Dev Biol 2003; 261:520-36. [PMID: 14499657 DOI: 10.1016/s0012-1606(03)00351-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In vertebrates, the nervous system arises from a flat sheet of epithelial cells, the neural plate, that gradually transforms into a hollow neural tube. This process, called neurulation, involves sequential changes in cellular interactions that are precisely coordinated both spatially and temporally by the combined actions of morphogens. To gain further insight into the molecular events regulating cell adhesion during neurulation, we investigated whether the adhesive and migratory capacities of neuroepithelial cells might be modulated by Sonic hedgehog (Shh), a signaling molecule involved in the control of cell differentiation in the ventral neural tube. When deposited onto extracellular matrix components in vitro, neural plates explanted from avian embryos at early neurulation readily dispersed into monolayers of spread cells, thereby revealing their intrinsic ability to migrate. In the presence of Shh added in solution to the culture medium, the explants still exhibited the same propensity to disperse. In contrast, when Shh was immobilized to the substrate or produced by neuroepithelial cells themselves after transfection, neural plate explants failed to disperse and instead formed compact structures. Changes in the adhesive capacities of neuroepithelial cells caused by Shh could be accounted for by inactivation of surface beta1-integrins combined with an increase in N-cadherin-mediated cell adhesion. Furthermore, immobilized Shh promoted differentiation of neuroepithelial cells into motor neurons and floor plate cells with the same potency as soluble Shh. However, the effect of Shh on the neuroepithelial cell adhesion was discernible and apparently independent from its differentiation effect and was not mediated by the signaling cascade elicited by the Patched-Smoothened receptor and involving the Gli transcription factors. Thus, our experiments indicate that Shh is able to control sequentially adhesion and differentiation of neuroepithelial cells through different mechanisms, leading to a coordinated regulation of the various cell interactions essential for neural tube morphogenesis.
Collapse
Affiliation(s)
- Artem Jarov
- Laboratoire de Biologie du Développement, CNRS et Université Pierre et Marie Curie, 9 quai St.-Bernard, 75252 05, Paris Cedex, France.
| | | | | | | | | | | |
Collapse
|
23
|
Kalcheim C. Mechanisms of early neural crest development: from cell specification to migration. INTERNATIONAL REVIEW OF CYTOLOGY 2001; 200:143-96. [PMID: 10965468 DOI: 10.1016/s0074-7696(00)00004-8] [Citation(s) in RCA: 46] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
The neural crest is a group of embryonic progenitors that forms during the process of neurulation by interactions that take place between the prospective epidermis and the specified neuroectoderm. Although initially an integral part of the neuroepithelium, neural crest cells separate from the central nervous system primordium by a process of epitheliomesenchymal transition and become a motile cell population. These mesenchymal cells then migrate through stereotypic pathways, some of which are common and others unique to various vertebrate species. Furthermore, the availability of distinct migratory pathways also differs according to embryonic stage and axial level. Studies have begun to address the molecular basis of neural crest specification, delamination, and migration. The present review summarizes some major advances in our understanding of the nature of the intercellular interactions and the molecules that mediate them during early phases of neural crest ontogeny.
Collapse
Affiliation(s)
- C Kalcheim
- Department of Anatomy and Cell Biology, Hebrew University of Jerusalem, Hadassah Medical School, Israel
| |
Collapse
|
24
|
Bronner-Fraser M. Rostrocaudal differences within the somites confer segmental pattern to trunk neural crest migration. Curr Top Dev Biol 1999; 47:279-96. [PMID: 10595308 DOI: 10.1016/s0070-2153(08)60728-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
Affiliation(s)
- M Bronner-Fraser
- Division of Biology, California Institute of Technology, Pasadena 91125, USA
| |
Collapse
|
25
|
Beauvais-Jouneau A, Pla P, Bernex F, Dufour S, Salamero J, Fässler R, Panthier JJ, Thiery JP, Larue L. A novel model to study the dorsolateral migration of melanoblasts. Mech Dev 1999; 89:3-14. [PMID: 10559475 DOI: 10.1016/s0925-4773(99)00191-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Melanocytes derived from pluripotent neural crest cells migrate initially in the dorsolateral pathway between the ectoderm and dermomyotome. To understand the role of specific proteins involved in this cell migration, we looked for a cellular model that mimics the in vivo behavior of melanoblasts, and that allows functional studies of their migration. We report here that wild-type embryonic stem (ES) cells are able to follow the ventral and dorsolateral neural crest pathways after being grafted into chicken embryos. By contrast, a mutant ES cell line deficient for beta1 integrin subunits, proteins involved in cell-extracellular interactions, had a severely impaired migratory behavior. Interestingly, ES cells deficient for Kit, the tyrosine kinase receptor for the stem cell factor (SCF), behaved similarly to wild-type ES cells. Thus, grafting mouse ES cells into chicken embryos provides a new cellular system that allows both in vitro and in vivo studies of the molecular mechanisms controlling dorsolateral migration.
Collapse
Affiliation(s)
- A Beauvais-Jouneau
- Developmental Genetics of Melanocytes, UMR 146 CNRS-Institut Curie, Bat. 110, 91405, Orsay, France
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Kil SH, Krull CE, Cann G, Clegg D, Bronner-Fraser M. The alpha4 subunit of integrin is important for neural crest cell migration. Dev Biol 1998; 202:29-42. [PMID: 9758701 DOI: 10.1006/dbio.1998.8985] [Citation(s) in RCA: 95] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We identify the alpha4 subunit of integrin as a predominant integrin expressed by neural crest cells in both avian and murine embryos. Using degenerate primers, we obtained a PCR fragment of the chick integrin alpha4 subunit that was subsequently used to clone the full-length subunit with a predicted amino acid sequence 60% identical to human and mouse alpha4 subunits. In situ hybridization demonstrates that chick integrin alpha4 mRNA is expressed at high levels by migrating neural crest cells and neural crest-derived ganglia at both cranial and trunk levels. An antibody against the murine alpha4 subunit revealed similar distribution patterns in mouse to chick. In addition to neural crest cells, the integrin alpha4 subunit was later observed on the muscle masses of the limb, the apical ectodermal ridge, and the developing liver. To examine the functional role of the integrin alpha4 subunit in neural crest cell migration, we used an explant preparation that allows visualization of neural crest cells in their normal environment with or without perturbing reagents. In the presence of a blocking antibody against the mouse integrin alpha4 subunit, there was a profound abrogation of neural crest cell migration at trunk and hindbrain levels. Both the numbers of migrating neural crest cells and the total distance traversed were markedly reduced. Similarly, avian embryos injected with synthetic peptides that contain the integrin alpha4 binding site in fibronectin displayed abnormal neural crest cell migration. Our results suggest that the integrin alpha4 subunit is important for normal neural crest cell migration and may be one of the primary alpha subunits used for neural crest cell migration in vivo. Furthermore, the integrin alpha4 subunit represents a useful neural crest marker in the mouse.
Collapse
Affiliation(s)
- S H Kil
- Division of Biology 139-74, California Institute of Technology, Pasadena, California, 91125, USA
| | | | | | | | | |
Collapse
|
27
|
Abstract
In 1967, Okamoto et al suggested that the absence of ganglion cells in Hirschsprung's disease (HD) was attributable to failure of migration of neural crest cells. The earlier the arrest of migration, the longer the aganglionic segment. Since then, this hypothesis generally has been accepted. However, subsequent experiments using mouse models of intestinal aganglionosis indicate that nerve cells may reach the correct position but then fail to develop or survive. An alternative hypothesis has been proposed that the aganglionosis may be caused by failure of differentiation as a result of microenvironmental changes after the migration has occurred. Extracellular matrix proteins are recognized as important microenvironmental factors. It has been shown that enteric neurogenesis is dependent on extracellular matrices, which provide a migration pathway for neural crest-derived cells and promote the maturation of settled neural crest-derived cells. Altered distributions of extracellular matrices have been shown in human HD cases and murine HD models, suggesting the role of extracellular matrices in the pathogenesis of HD. Recent studies suggest that intestinal smooth muscle cells, target cells of enteric neurons, play an important role in guiding and influencing its own innervation. Normal maturation was inhibited in neurons cultured with smooth muscle cells of aganglionic colon in comparison to normal colon. Furthermore, it was demonstrated that levels of neurotrophic factors, crucial in the development and survival of enteric neurons, are decreased in circular muscle layers of aganglionic colon in comparison to normoganglionic colon. The smooth muscle cells of the aganglionic colon may represent an unfavorable microenvironment for neuronal development compared with the normally innervated region. Recently, markedly increased immunoreactivity of major histocompatibility complex (MHC) class II antigens and ICAM-1 was demonstrated in aganglionic bowel, suggesting the immunological mechanisms may be involved in the etiology of HD. Genetic factors have been implicated in the etiology of this condition because HD is known to occur in families and in association with some chromosomal abnormalities. Recent expansion of molecular genetics identified multiple susceptibility genes of HD, including the RET gene, the glial cell line-derived neurotrophic factor gene, the endothelin-B receptor gene, and endothelin-3 gene. Of these, inactivating mutations of the RET gene are the most frequent, occurring in 50% of familial and 15% to 20% of sporadic cases of HD. To date, despite extensive research, the exact etiology of this condition remains poorly understood. The present report describes the authors' current understanding of and recent progress in the etiology of HD.
Collapse
Affiliation(s)
- P Puri
- Children's Research Centre, Our Lady's Hospital for Sick Children, Dublin, Ireland
| | | | | |
Collapse
|
28
|
Sanders RJ, Mainiero F, Giancotti FG. The role of integrins in tumorigenesis and metastasis. Cancer Invest 1998; 16:329-44. [PMID: 9627681 DOI: 10.3109/07357909809084653] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Affiliation(s)
- R J Sanders
- Department of Pediatrics, New York University School of Medicine, New York, USA
| | | | | |
Collapse
|
29
|
Henderson DJ, Ybot-Gonzalez P, Copp AJ. Over-expression of the chondroitin sulphate proteoglycan versican is associated with defective neural crest migration in the Pax3 mutant mouse (splotch). Mech Dev 1997; 69:39-51. [PMID: 9486530 DOI: 10.1016/s0925-4773(97)00151-2] [Citation(s) in RCA: 75] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Splotch mice, which harbour mutations in the Pax3 gene, exhibit neural crest-related abnormalities including pigmentation defects, reduced or absent dorsal root ganglia and failure of cardiac outflow tract septation in homozygotes. Although splotch neural crest cells fail to colonise target tissues, they initiate migration in vivo and appear to migrate as well as wild type neural crest cells in vitro, suggesting that the neural crest abnormality in splotch may reside not in the neural crest cells themselves, but rather in the extracellular environment through which they migrate. We have examined the expression of genes encoding extracellular matrix molecules in Sp2H homozygous embryos and find a marked over-expression of transcripts for the chondroitin sulphate proteoglycan versican in the pathways of neural crest cell migration. Use of cadherin-6 expression as a marker for neural crest demonstrates a striking correlation between up-regulation of versican expression and absence of migrating neural crest cells, both in the mesenchyme lateral to the neural tube and in the lower branchial arches of Sp2H homozygotes. Pax3 and versican have mutually exclusive expression patterns in normal embryos whereas, in Sp2H homozygotes, versican is generally over-expressed with 'infilling' in regions that would normally express functional Pax3. Versican, like other chondroitin sulphate proteoglycans, is non-permissive for migration of neural crest cells in vitro, and we suggest that over-expression of this molecule leads to the arrest of neural crest cell migration in splotch embryos. Pax3 may serve to negatively regulate versican expression during normal development, thereby guiding neural crest cells into their pathways of migration.
Collapse
Affiliation(s)
- D J Henderson
- Neural Development Unit, Institute of Child Health, University College London, UK
| | | | | |
Collapse
|
30
|
Abstract
Development of the neural crest involves a remarkable feat of coordinated cell migration in which cells detach from the neural tube, take varying routes of migration through the embryonic tissues and then differentiate at the end of their journey to participate in the formation of a number of organ systems. In general, neural crest cells appear to migrate without the guidance of long-range physical or chemical cues, but rather they respond to heterogeneity in the extracellular matrix that forms their migration substrate. Molecules such as fibronectin and laminin act as permissive substrate components, encouraging neural crest cell attachment and spreading, whereas chondroitin sulphate proteoglycans are nonpermissive for migration. A balance between permissive and nonpermissive substrate components seems to be necessary to ensure successful migration, as indicated by a number of studies in mouse mutant systems where nonpermissive molecules are over-expressed, leading to inhibition of neural crest migration. The neural crest expresses cell surface receptors that permit interaction with the extracellular matrix and may also modify the matrix by secretion of proteases. Thus the principles that govern the complex migration of neural crest cells are beginning to emerge.
Collapse
Affiliation(s)
- DEBORAH J.
HENDERSON
- Neural Development Unit, Division of Cell and Molecular Biology, Institute of Child Health, London, UK
| | - ANDREW J.
COPP
- Neural Development Unit, Division of Cell and Molecular Biology, Institute of Child Health, London, UK
- Correspondence to Professor Andrew Copp, Neural Development Unit, Institute of Child Health, Guilford Street, London WC1N 1EH, UK. Tel: +44 0171 829 8893; fax: +44 0171 813 8494; e-mail:
| |
Collapse
|
31
|
Budinsky AC, Brodowicz T, Wiltschke C, Czerwenka K, Michl I, Krainer M, Zielinski CC. Decreased expression of ICAM-1 and its induction by tumor necrosis factor on breast-cancer cells in vitro. Int J Cancer 1997; 71:1086-90. [PMID: 9185715 DOI: 10.1002/(sici)1097-0215(19970611)71:6<1086::aid-ijc27>3.0.co;2-a] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
In order to study adhesion-molecule expression and its consequences for cellular recognition, the presence of adhesion molecules ICAM-1, VCAM-1, VLA-4, LFA-1, alpha, LFA-1 beta, LFA-3, beta1-integrin and beta3-integrin was studied on specimens from breast tissue by immunohistochemistry and on cells from breast cell lines propagated in vitro. Breast-cancer tissue and the breast-cancer cell lines MCF-7, SK-BR-3 and ZR-75-1 showed expression of ICAM-1 and VLA-4 significantly lower than that of benign breast cells or normal breast epithelium. Of various cytokines tested, including recombinant human (rh) interleukin-6 (IL-6), rh tumor necrosis factor alpha (TNF-alpha), interleukin 2 (IL-2), granulocyte/macrophage-colony-stimulating-factor (GM-CSF), interferon-alpha (IFN-alpha) and interferon-gamma (IFN-gamma), only TNF was able to re-induce expression of ICAM-1 on cells from MCF-7, SK-BR-3 and ZR-75-1. Further, the ability of either unstimulated or lymphokine-stimulated killer (LAK) cells to recognize and lyse native or TNF-stimulated breast-cancer cells was studied. Whereas neither unstimulated lymphocytes or LAK cells were able to lyse untreated breast-cancer cells deficient for ICAM-1 expression, pre-treatment of tumor cells with TNF led to increased tumor-cell lysis. Anti-ICAM-1 antibodies, and pre-treatment of tumor cells with anti-TNF-receptor antibodies, abrogated these findings, corroborating their specificity. We thus conclude that the defective expression of ICAM-1 in our model might constitute a mechanism by which breast-cancer cells escape immunologic recognition and lysis by appropriate effector cells.
Collapse
Affiliation(s)
- A C Budinsky
- Department of Internal Medicine I, University Hospital, Vienna, Austria
| | | | | | | | | | | | | |
Collapse
|
32
|
Newgreen DF, Kerr RS, Minichiello J, Warren N. Changes in cell adhesion and extracellular matrix molecules in spontaneous spinal neural tube defects in avian embryos. TERATOLOGY 1997; 55:195-207. [PMID: 9181673 DOI: 10.1002/(sici)1096-9926(199703)55:3<195::aid-tera4>3.0.co;2-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Quail embryos (embryonic days 2-2.5) with spontaneous neural tube defects (NTDs), along with age-matched normal embryos, were examined immunocytochemically for the extracellular matrix (ECM) molecules laminin, fibronectin, and chondroitin sulfate proteoglycan, the cell adhesion molecules (CAMs) E- and N-cadherin and neural CAM (NCAM), and the neural crest marker HNK-1. The embryos with NTDs were at the lower limit of the normal stage range and the affected region was about 25% shorter than in normal embryos. Open NTDs occurred in cervical and upper thoracic level, although often the ventral neural tube was morphologically normal. Widened, irregular but closed neural tubes (lower thoracic to sacral levels) showed disorganized mesenchyme-like cells centrally and often multiple lumens. Finger-like tabs projecting from the ectoderm over the neural tube also occurred at lower thoracic to sacral levels. In open NTDs, the E-cadherin-labeled epidermis was incomplete dorsally, and was continuous with the N-cadherin-labeled neural tissue, with a sharp demarcation between E- and N-cadherin-expressing regions, as in the early stages of normal primary neurulation. A sharp inverted peak of epidermis extended ventrally, closely applied to the side of the neural tissue. The intervening matrix labeled less intensely for chondroitin sulfate proteoglycan relative to laminin and fibronectin, in comparison to control embryos. In closed NTDs, the dorsal superficial cell layer (i.e., positionally epidermis) was not separated from the underlying neural tissue by a band of matrix as in control embryos. In addition, this layer expressed E-cadherin (as in normal embryos), but coexpressed N-cadherin and NCAM, which are not normally found here at this stage. This overlap region resembled the mid-dorsal tissue at earlier stages in normal secondary neurulation in the tail-bud. The tabs of tissue appeared to be localized hypertrophy of the epidermal and neural ectoderm, and also showed codistribution of E- and N-cadherin. In all these defects, matrix molecules occurred within (rather than around) the neural and epidermal epithelia. HNK-1-labeled neural crest cells were frequently absent in regions of NTDs, in contrast to control embryos. These results show that matrix and cell adhesion molecules are disturbed in spontaneous NTDs at the time of neurulation, and therefore could be involved in the generation of the defects by altering cell adhesion-dependent morphogenetic events.
Collapse
Affiliation(s)
- D F Newgreen
- Murdoch Institute, Royal Children's Hospital, Victoria, Australia.
| | | | | | | |
Collapse
|
33
|
Lentz SI, Miner JH, Sanes JR, Snider WD. Distribution of the ten known laminin chains in the pathways and targets of developing sensory axons. J Comp Neurol 1997; 378:547-61. [PMID: 9034910 DOI: 10.1002/(sici)1096-9861(19970224)378:4<547::aid-cne9>3.0.co;2-2] [Citation(s) in RCA: 53] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Laminins are heterotrimers of alpha, beta, and gamma chains. At present, five alpha, three beta, and two gamma chains have been described. The best characterized laminin (laminin 1 = alpha 1, beta 1, gamma 1) promotes neurite outgrowth from virtually all classes of developing neurons, implying that laminins may serve as axon guidance molecules in vivo. Moreover, different laminin trimers exert distinct effects on subsets of laminin-1-responsive cells, suggesting that isoform diversity may underlie some axonal choices in vivo. As a first step toward evaluating these hypotheses, we have documented the expression patterns of all 10-known laminin chains in the peripheral nervous system and spinal cord of the murine embryo. The alpha 2, alpha 4, beta 1, and gamma 1 chains are expressed in peripheral axonal pathways by embryonic day (E) 11.5, when sensory and motor axonal outgrowth is underway. Thus, laminins (but not laminin 1) may promote peripheral axonal outgrowth. By E 13.5, laminin chains are differentially expressed in the limb-bud, with prominent expression of alpha 2 and alpha 4 in muscle and of alpha 3 and alpha 5 in skin. This pattern raises the possibility that laminin isoform diversity contributes to the ability of cutaneous and muscle sensory axons to distinguish their targets. Later in development, some chains (e.g., alpha 2, alpha 4, and beta 1) are downregulated in peripheral nerve while others (e.g., gamma 1), continue to be expressed by Schwann cells into adulthood. In contrast to peripheral nerves and ganglia, laminin chains are expressed at low levels, if at all, in the developing spinal cord gray matter.
Collapse
Affiliation(s)
- S I Lentz
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | | | | | | |
Collapse
|
34
|
Abstract
Fibronectins (FNs) are essential for the proper development of embryonic mesenchymal tissues. A lacZ reporter gene has been fused to 4.9 kbp of DNA from the rat FN gene 5' flanking region, and this construct has been microinjected into fertilized mouse embryos to investigate the cis elements needed for the temporal and spatial regulation of FN in vivo. Histochemical staining of embryos for beta-galactosidase activity demonstrated that four independent lines shared a specific pattern of lacZ expression, reflecting the activity of the fibronectin sequences contained within the transgene. Specifically, somites stained positively for lacZ, but expression was spatially and temporally non-uniform, with higher levels in more caudal somites after a total of ca. 13 somite pairs had formed. This rostral-caudal gradient of lacZ expression in somites of embryos beyond this stage resembled the distribution of endogenous FN mRNA, as detected by whole mount in situ hybridization. The transgene was not expressed in the developing heart where endogenous FN mRNA was detected. Unexpectedly, highly localized staining was observed within the neural tube beginning at ca. E10-10.5, and two of the lines exhibited additional areas of staining due to the individual integration sites. Thus, the 4.9 kbp FN fragment appears to recapitulate closely the complex pattern of FN expression observed during somitogenesis. A smaller fragment of 0.9 kbp also directed lacZ expression in caudal somites at E9.5, suggesting that these sequences are sufficient to establish the spatio-temporal pattern.
Collapse
Affiliation(s)
- R A Perkinson
- Department of Medicine, Jefferson Medical College of Thomas Jefferson University, Philadelphia, Pennsylvania 19107, USA
| | | |
Collapse
|
35
|
Perris R, Brandenberger R, Chiquet M. Differential neural crest cell attachment and migration on avian laminin isoforms. Int J Dev Neurosci 1996; 14:297-314. [PMID: 8842806 DOI: 10.1016/0736-5748(96)00015-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
A number of laminin isoforms have recently been identified and proposed to exert different functions during embryonic development. In the present study, we describe the purification and partial characterization of several isoforms isolated from chick heart and gizzard, and provide data on the molecular mechanisms underlying the interaction of avian neural crest cells with these molecules in vitro. Laminins extracted from heart and gizzard tissues were separated by gel filtration and purified to homogeneity by sequential lectin and immunoaffinity chromatography by utilizing monoclonal antibodies directed against the avian alpha 2, beta 2 and gamma 1 laminin chains. The sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) banding pattern of the polypeptide complexes obtained and immunoblotting with polyclonal antisera allowed the identification of Laminin-2 (alpha 2 beta 1 gamma 1), Laminin-4 (alpha 2 beta 2 gamma 1), and laminins comprising the beta 1, beta 2 and gamma 1 chains associated with a shorter alpha chain which, in SDS-PAGE, co-migrate with the beta/gamma complex in the 200 kDa region. These latter laminins, which are here arbitrarily denoted Laminin-alpha x (heart tissue) and Laminin-G (gizzard tissue), are somewhat distinct in their apparent molecular weight, are differentially associated with nidogen, and appear as "T"-shaped particles similar to Laminin-6 and Laminin-7 when analyzed by transmission electron microscopy following rotary shadowing. In contrast, the avian Laminin-2 and Laminin-4 isoforms exhibit the characteristic cruciform shape described previously for their mammalian counterparts. Isolated neural crest cells differentially attached and migrated on these laminin isoforms, showing a clear preference for Laminin-G. Similarly to the EHS Laminin-1, neural crest cells recognized all avian isoforms through their alpha 1 beta 1 integrin, shown previously to be the primary laminin-binding receptor on these cells. Neural crest cell interaction with the avian laminins was dependent upon maintenance of the secondary and tertiary structure of the molecules, as shown by the marked reduction in cell attachment and migration upon disruption of the alpha-helical coiled-coil structure of their constituent chains. The results demonstrate that different laminin isoforms may be differentially involved in the regulation of neural crest cell migration and suggest that this regulation operates through interaction of the cells with a structurally conserved cell binding site recognized by the alpha 1 beta 1 integrin.
Collapse
Affiliation(s)
- R Perris
- Reference Center for Oncology, Division for Experimental Oncology 2, Aviano (PN), Italy
| | | | | |
Collapse
|
36
|
Kil SH, Bronner-Fraser M. Expression of the avian alpha 7-integrin in developing nervous system and myotome. Int J Dev Neurosci 1996; 14:181-90. [PMID: 8842797 DOI: 10.1016/0736-5748(96)00006-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Integrins are cell surface receptors for a variety of extracellular matrix molecules including fibronectin, laminin and collagens. Although their role in development is not completely understood, they are likely to have important functions in cell migration and axon guidance. To characterize the types of integrins expressed in the developing nervous system, we have used monoclonal antibodies against alpha 7- and alpha v-integrin subunits to examine the distribution of these subunits in the early chick embryo. Low levels of alpha 7 immunoreactivity were first observed in the neural tube and developing myotome of stage 17 embryos (E2.5). Although low levels of alpha 7 expression were associated with most neuroepithelial cells, distinct alpha 7 immunoreactivity was first detected in the ventrolateral portions of the neural tube at a stage corresponding to the time when the first neurons differentiate. Its distribution pattern overlapped with that of commissural neurons in the developing spinal cord. alpha 7 was also prominently localized to the motor neurons and their axons emanating from the neural tube. In addition, alpha 7 immunoreactivity was observed on a subpopulation of trunk neural crest cells migrating through the somitic sclerotome. At later stages, alpha 7 expression was observed in other nervous system structures such as the pigmented retinal epithelial cells. In addition to its distribution in the developing nervous system, alpha 7 immunoreactivity was associated with early myotomal cells shortly after myotome formation and its expression persisted throughout myotome development. In contrast to alpha 7, alpha v-integrin had a limited distribution in the nervous system, being expressed only at low levels in the neural tube. However, alpha v displayed prominent immunoreactivity in the myotome and in endothelial cells of the dorsal aorta. The results suggest that alpha 7-integrin is one of the prevalent integrin subunits on neurons and axons in the developing spinal cord.
Collapse
Affiliation(s)
- S H Kil
- Developmental Biology Center, University of California, Irvine 92717, USA
| | | |
Collapse
|
37
|
Veicsteinas A, Belleri M, Cinquetti A, Parolini S, Barbato G, Molinari Tosatti MP. Development of chicken embryos exposed to an intermittent horizontal sinusoidal 50 Hz magnetic field. Bioelectromagnetics 1996; 17:411-24. [PMID: 8915551 DOI: 10.1002/(sici)1521-186x(1996)17:5<411::aid-bem9>3.0.co;2-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The effects of intermittent exposure (2 h on/22 h off) to a 200 microT horizontal, sinusoidally oscillating (50 Hz) magnetic field were studied in 210 fertilized chicken eggs. Two hundred ten control eggs (sham-exposed) were incubated in the same chamber as the experimental eggs. Chick embryos were examined for developmental anomalies and maturity stage after 48 h of incubation. Immunohistochemical analysis of extracellular membrane components (laminin, fibronectin, and type IV collagen) were conducted on day 7 and histological examinations for malformations of brain, liver, and heart, on days 7, 12, and 18 of incubation. Furthermore, egg fertility and egg weights were evaluated on days 2, 7, 12, and 18. The investigation also measured the body weight of chickens for 90 days from hatching and included histological analysis of body organs. Each variable was investigated blind. Statistical comparison between exposed and sham-exposed values did not show significant differences in any of the variables investigated. Thus, it appears that the exposure of embryos to an intermittent 200 microT magnetic field at 50 Hz does not cause developmental anomalies, changes in maturity stage, alterations in distribution of extracellular membrane components, or malformations in the brain, liver, or heart. Moreover, there were no differences in body weight, morphology, or histology of central nervous system, liver, heart, or testis in 90-day-old chickens hatched from exposed in comparison to sham-exposed eggs.
Collapse
Affiliation(s)
- A Veicsteinas
- Department of Biomedical Sciences and Biotechnologies, University of Brescia, Italy
| | | | | | | | | | | |
Collapse
|
38
|
Krull CE, Collazo A, Fraser SE, Bronner-Fraser M. Segmental migration of trunk neural crest: time-lapse analysis reveals a role for PNA-binding molecules. Development 1995; 121:3733-43. [PMID: 8582285 DOI: 10.1242/dev.121.11.3733] [Citation(s) in RCA: 73] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Trunk neural crest cells migrate through the somites in a striking segmental fashion, entering the rostral but not caudal sclerotome, via cues intrinsic to the somites. Attempts to define the molecular bases of these cues have been hampered by the lack of an accessible assay system. To examine trunk neural crest migration over time and to perturb candidate guiding molecules, we have developed a novel explant preparation. Here, we demonstrate that trunk regions of the chicken embryo, placed in explant culture, continue to develop apparently normally for 2 days. Neural crest cells, recognized by prelabeling with DiI or by poststaining with the HNK-1 antibody, migrate in the somites of the explants in their typical segmental pattern. Furthermore, this paradigm allows us to follow trunk neural crest migration in situ for the first time using low-light-level videomicroscopy. The trajectories of individual neural crest cells were often complex, with cells migrating in an episodic mode encompassing forward, backward and lateral movements. Frequently, neural crest cells migrated in close-knit groups of 2–4 cells, moving at mean rates of migration of 10–14 microns/hour. Treatment of the explants with the lectin peanut agglutinin (PNA) both slowed the rate and altered the pattern of neural crest migration. Neural crest cells entered both the rostral and caudal halves of the sclerotome with mean rates of migration ranging from 6 to 13 microns/hour. These results suggest that peanut agglutinin-binding molecules are required for the segmental patterning of trunk neural crest migration. Because this approach permits neural crest migration to be both observed and perturbed, it offers the promise of more direct assays of the factors that influence neural crest development.
Collapse
Affiliation(s)
- C E Krull
- Developmental Biology Center, UC Irvine 92717, USA
| | | | | | | |
Collapse
|
39
|
Phillips GR, Edelman GM, Crossin KL. Separate cell binding sites within cytotactin/tenascin differentially promote neurite outgrowth. CELL ADHESION AND COMMUNICATION 1995; 3:257-71. [PMID: 8846026 DOI: 10.3109/15419069509081291] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Cytotactin/tenascin (CT/TN) is an extracellular matrix protein that binds to a variety of cell types and that influences neurite outgrowth. It has a multidomain structure with regions homologous to epidermal growth factor (EGF)-like repeats, fibronectin (FN) type II repeats, and the beta and gamma chains of fibrinogen (fg). The current study demonstrates that a fusion protein corresponding to the sixth fibronectin type III repeat in CT/TN (CTfn6) supported cell attachment and promoted an increase in the number of cells with neurites in both central and peripheral neurons in tissue culture. The third fibronectin type III repeat, CTfn3, like intact CT/TN, supported attachment of peripheral neurons but not of central neurons and, while it caused an increase in neurite length, it did not increase the number of cells that sprouted neurites. When CTfn3 and CTfn6 were combined, an increase in both the number of cells sprouting neurites and in neurite length was observed for peripheral neurons that resembled their response to intact CT/TN. Cell attachment to CTfn6 was inhibited in the presence of function-blocking antibodies against beta 1 integrins. In contrast, the interaction with CTfn3 was not inhibited by antibodies to beta 1 integrins, but was inhibited by RGD-containing peptides. The results suggest that cell binding to CT/TN involves two different sites within the molecule and occurs via different receptors which may be differentially expressed on different neuronal cell types. The location of these sites within the whole molecule in the context of other adhesive and counteradhesive domains may modulate their influence on cellular responses such as cell attachment and neurite outgrowth.
Collapse
Affiliation(s)
- G R Phillips
- Scripps Research Institute, Department of Neurobiology, La Jolla, CA 92037, USA
| | | | | |
Collapse
|
40
|
Müller U, Bossy B, Venstrom K, Reichardt LF. Integrin alpha 8 beta 1 promotes attachment, cell spreading, and neurite outgrowth on fibronectin. Mol Biol Cell 1995; 6:433-48. [PMID: 7626807 PMCID: PMC301202 DOI: 10.1091/mbc.6.4.433] [Citation(s) in RCA: 90] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
The integrin alpha 8 subunit, isolated by low stringency hybridization, is a novel integrin subunit that associates with beta 1. To identify ligands, we have prepared a function-blocking antiserum to the extracellular domain of alpha 8, and we have established by transfection K562 cell lines that stably express alpha 8 beta 1 heterodimers on the cell surface. We demonstrate here by cell adhesion and neurite outgrowth assays that alpha 8 beta 1 is a fibronectin receptor. Studies on fibronectin fragments using RGD peptides as inhibitors show that alpha 8 beta 1 binds to the RGD site of fibronectin. In contrast to the endogenous alpha 5 beta 1 fibronectin receptor in K562 cells, alpha 8 beta 1 not only promotes cell attachment but also extensive cell spreading, suggesting functional differences between the two receptors. In chick embryo fibroblasts, alpha 8 beta 1 is localized to focal adhesions. We conclude that alpha 8 beta 1 is a receptor for fibronectin and can promote attachment, cell spreading, and neurite outgrowth on fibronectin.
Collapse
Affiliation(s)
- U Müller
- Department of Physiology, University of California at San Francisco 94143-0724, USA
| | | | | | | |
Collapse
|
41
|
|
42
|
Abstract
The serum levels of beta 1 integrin (microgram/ml) were significantly higher in the patients with chronic persistent hepatitis (2.59 +/- 0.04), chronic active hepatitis (3.45 +/- 0.13), cirrhosis (4.77 +/- 0.30) and hepatocellular carcinoma (4.71 +/- 0.49) than in normal subjects (2.11 +/- 0.08). Serum levels of beta 3 integrin (microgram/ml) were significantly higher in the patients with chronic active hepatitis (10.48 +/- 1.22), liver cirrhosis (13.55 +/- 1.54) and hepatocellular carcinoma (14.1 +/- 1.77) when compared with normal subjects (5.51 +/- 0.52). A positive correlation was found between serum levels of beta 1 and beta 3 integrins (p < 0.001). A strong positive correlation was observed between serum levels of beta 1 integrin and histologic features, particularly in the degree of hepatic fibrosis, while no correlation was found between serum levels of beta 3 integrin and hepatic fibrosis. Immunohistochemical studies revealed that the beta 1 integrin was present on the plasma membranes of hepatocytes and sinusoidal lining cells in the normal liver, and was increased in fibrotic areas, and on the plasma membranes of hepatocytes and sinusoidal lining cells of the chronic liver disease. However, no positive staining for beta 3 integrin was observed in fibrotic area. The serum level of beta 1 integrin in patients with chronic liver diseases may therefore be a useful marker of hepatic fibrosis.
Collapse
Affiliation(s)
- M Yamauchi
- First Department of Internal Medicine, Jikei University School of Medicine, Tokyo, Japan
| | | | | | | |
Collapse
|
43
|
Abstract
The extracellular matrix supports the adhesion and migration of cells during morphogenesis and influences cell differentiation. Cell interactions with the extracellular matrix are mediated in large part by members of the integrin family of cell-surface receptors. Recent progress in this area has resulted in the identification of multiple integrins, many of which are expressed in position-specific patterns during vertebrate development. The contributions of these receptors to specific developmental events are now being investigated in a variety of systems using a combination of genetic, molecular and immunological approaches.
Collapse
Affiliation(s)
- D W DeSimone
- University of Virginia, Department of Cell Biology, School of Medicine, Charlottesville 22908
| |
Collapse
|
44
|
Delannet M, Martin F, Bossy B, Cheresh DA, Reichardt LF, Duband JL. Specific roles of the alpha V beta 1, alpha V beta 3 and alpha V beta 5 integrins in avian neural crest cell adhesion and migration on vitronectin. Development 1994; 120:2687-702. [PMID: 7525179 PMCID: PMC2710119 DOI: 10.1242/dev.120.9.2687] [Citation(s) in RCA: 120] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
To identify potentially important extracellular matrix adhesive molecules in neural crest cell migration, the possible role of vitronectin and its corresponding integrin receptors was examined in the adhesion and migration of avian neural crest cells in vitro. Adhesion and migration on vitronectin were comparable to those found on fibronectin and could be almost entirely abolished by antibodies against vitronectin and by RGD peptides. Immunoprecipitation and immunocytochemistry analyses revealed that neural crest cells expressed primarily the alpha V beta 1, alpha V beta 3 and alpha V beta 5 integrins as possible vitronectin receptors. Inhibition assays of cellular adhesion and migration with function-perturbing antibodies demonstrated that adhesion of neural crest cells to vitronectin was mediated essentially by one or more of the different alpha V integrins, with a possible preeminence of alpha V beta 1, whereas cell migration involved mostly the alpha V beta 3 and alpha V beta 5 integrins. Immunofluorescence labeling of cultured motile neural crest cells revealed that the alpha V integrins are differentially distributed on the cell surface. The beta 1 and alpha V subunits were both diffuse on the surface of cells and in focal adhesion sites in association with vinculin, talin and alpha-actinin, whereas the alpha V beta 3 and alpha V beta 5 integrins were essentially diffuse on the cell surface. Finally, vitronectin could be detected by immunoblotting and immunohistochemistry in the early embryo during the ontogeny of the neural crest. It was in particular closely associated with the surface of migrating neural crest cells. In conclusion, our study indicates that neural crest cells can adhere to and migrate on vitronectin in vitro by an RGD-dependent mechanism involving at least the alpha V beta 1, alpha V beta 3 and alpha V beta 5 integrins and that these integrins may have specific roles in the control of cell adhesion and migration.
Collapse
Affiliation(s)
- M Delannet
- Laboratoire de Biologie Cellulaire du Développement, Institut Jacques Monod, Université Paris, France
| | | | | | | | | | | |
Collapse
|
45
|
Stepp MA, Urry LA, Hynes RO. Expression of alpha 4 integrin mRNA and protein and fibronectin in the early chicken embryo. CELL ADHESION AND COMMUNICATION 1994; 2:359-75. [PMID: 7529637 DOI: 10.3109/15419069409014210] [Citation(s) in RCA: 47] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
alpha 4 integrins (alpha 4 beta 1 and alpha 4 beta 7) have been shown to mediate both cell-matrix adhesion to fibronectin and cell-cell adhesion to VCAM-1. These interactions have been suggested to contribute to hematopoiesis, lymphocyte homing, recruitment of inflammatory cells, neural crest cell migration and myogenesis. We report here the cloning of chicken alpha 4 cDNA and its use to define the patterns of expression of alpha 4 mRNA and protein in early chicken embryos (19-22 somite pairs), a stage at which neural crest cells can be examined at various points in their migration and somitic development and differentiation can also be observed at various stages. We observe widespread expression of both alpha 4 mRNA and protein, although the patterns of steady state expression do not conform precisely. Many neural crest cells contain significant levels of alpha 4 mRNA. Some neural crest cells express alpha 4 protein but its expression is transient and/or limited to a subset of these cells. alpha 4 is strongly expressed at both mRNA and protein levels by somitic cells and their derivatives in the sclerotome, dermatome and myotome and is also expressed in neural tube, otic placode, heart, gut endoderm and some other tissues. Comparison with the distributions of fibronectin shows that, although some alpha 4 expression occurs in locations consistent with a role in cell-matrix adhesion to fibronectin, alpha 4 is also expressed in other places where fibronectin is low or absent and a role for alpha 4 in cell-cell interactions appears more likely.
Collapse
Affiliation(s)
- M A Stepp
- Center for Cancer Research, Massachusetts Institute of Technology, Cambridge
| | | | | |
Collapse
|
46
|
Leblanc GG. Rostrocaudal differences in the expression of extracellular matrix proteins by avian neural crest cells in vitro. Cell Tissue Res 1994; 277:97-106. [PMID: 8055542 DOI: 10.1007/bf00303085] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Cranial and trunk neural crest cells developing in vitro differed in their patterns of expression of two major extracellular matrix proteins, fibronectin and collagen I. Cranial neural crest cells showed two distinct phases of fibronectin expression: the first occurred during the initial migration of cells from explants onto the culture dish; the second was associated with the differentiative period of in vitro development. Fibronectin-immunoreactive cells eventually represented one of the most abundant cell types in cranial cultures. Large numbers of procollagen I-immunoreactive cells also developed in cranial cultures, and procollagen I was colocalized with fibronectin in individual cranial neural crest cells. Neither fibronectin nor procollagen I immunoreactivities were seen in either neurons or melanocytes, consistent with the idea that the fibronectin-immunoreactive cells in cranial neural crest cultures are committed to the mesenchymal lineage. In contrast to cranial neural crest, trunk neural crest produced very few fibronectin-immunoreactive cells at any time in vitro. Trunk neutral crest also produced smaller proportions of procollagen I-immunoreactive cells than did cranial explants. Mitotic labelling experiments showed that the differing proportions of fibronectin- and procollagen I-immunoreactive cells in cranial versus trunk cultures did not result from differences in rates of cell proliferation.
Collapse
Affiliation(s)
- G G Leblanc
- Department of Biological Structure and Function, School of Dentistry, Oregon Health Sciences University, Portland 97201
| |
Collapse
|
47
|
Bhattacharyya A, Brackenbury R, Ratner N. Axons arrest the migration of Schwann cell precursors. Development 1994; 120:1411-20. [PMID: 8050352 DOI: 10.1242/dev.120.6.1411] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The neural crest gives rise to a variety of cell types including Schwann cells of the peripheral nervous system. Schwann cell precursors begin to differentiate early and migrate along specific pathways in the embryo before associating with nerve trunks. To determine whether motor axons direct the migration of Schwann cell precursors along specific pathways, we tested the effect of ablating the ventral half of the neural tube, which contains motor neuron cell bodies. The ventral neural tube was removed unilaterally from lumbar regions of chicken embryos at stage 17, when neural crest cells are just beginning to migrate and before motor axons have extended out of the neural tube. At several stages after ventral tube ablation, sections of the lumbar region of these embryos were stained with anti-acetylated tubulin to label developing axons, HNK-1 to label migrating neural crest cells and 1E8 to label Schwann cell precursors. In many embryos the ablation of motor neurons was incomplete. The staining patterns in these embryos support the idea that some Schwann cells are derived from the neural tube. In embryos with complete motor neuron ablation, at stage 18, HNK-1-positive neural crest cells had migrated to normal locations in both control and ablated sides of the embryo, suggesting that motor axons or the ventral neural tube are not required for proper migration of neural crest cells. However, by stage 19, cells that were positive for HNK-1 or 1E8 were no longer seen in the region of the ventral root, nor ventral to the ventral root region. Because Schwann cell precursors require neural-derived factors for their survival in vitro, we tested whether neural crest cells that migrate to the region of the ventral root in ventral neural tube-ablated embryos then die. Nile Blue staining for dead and dying cells in ventral neural tube-ablated embryos provided no evidence for cell death at stage 18. These results suggest that motor axons arrest the migration of Schwann cell precursors during neural crest migration.
Collapse
Affiliation(s)
- A Bhattacharyya
- Department of Anatomy and Cell Biology, University of Cincinnati College of Medicine, OH 45267-0521
| | | | | |
Collapse
|
48
|
Nakao T, Ishizawa A. Development of the spinal nerves in the mouse with special reference to innervation of the axial musculature. ANATOMY AND EMBRYOLOGY 1994; 189:115-38. [PMID: 8010411 DOI: 10.1007/bf00185771] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Development of the mouse spinal nerves was studied. On E11 (11th day of gestation), the primitive spinal nerve fascicle extended ventrally in the anterior half of the sclerotome. Spinal nerves in the forelimb region united with each other to form the primitive brachial plexus. Their terminal segment was covered by a peculiar cell mass. On E12, five primary branches developed along the primitive spinal nerve trunk. The ramus dorsalis was originally a cutaneous nerve, supplying two series of branches to the skin of the back. The medial series was derived from the dorsal ramus of C2-C8, and the lateral series from C8 and the more caudal dorsal rami. Nerves of the former series took the presegmental course through the intermyotomic space, while those of the latter the postsegmental course. The ramus cutaneous lateralis was a nerve that took the presegmental course to become cutaneous. The ramus intercostalis externus was a muscle branch whose distribution was restricted within the segment. The ramus anterior was a muscle branch from the end of the primitive spinal nerve trunk. The ramus visceralis connected a thoracic nerve with the para-aortic sympathetic cell cord. On E13-16 the ramus anterior secondarily gave off a cutaneous branch (ramus cutaneous anterior). The ramus intercostalis externus extended ventrally deep to the intercostalis externus muscle, crossing just caudal to the ramus cutaneous lateralis that secondarily gave off branches to the obliquus externus abdominis muscle.
Collapse
Affiliation(s)
- T Nakao
- Department of Anatomy, Akita University School of Medicine, Japan
| | | |
Collapse
|
49
|
Horacek MJ, Kawaguchi T, Terracio L. Adult adenohypophysial cells express beta 1 integrins and prefer laminin during cell-substratum adhesion. In Vitro Cell Dev Biol Anim 1994; 30A:35-40. [PMID: 7514937 DOI: 10.1007/bf02631416] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
beta 1 Integrins are a family of structurally related heterodimeric cell surface receptors that are involved in adhesion to molecules in the extracellular matrix (ECM) such as laminin (LN), fibronectin (FN), and collagen. These receptors are expressed by many cell types and mediate a variety of processes such as cell-matrix and cell-to-cell adhesion, cell migration, growth, and differentiation. The purpose of these studies was to identify and partially characterize beta 1 integrins on adenohypophyseal cells and to begin to elucidate their functional importance. Adenohypophyses were removed from adult male rats, dispersed using 0.25% trypsin, rinsed, and resuspended in a 1:1 mixture of Dulbecco's modified Eagle's medium and F12 medium containing 10% fetal bovine serum and antibiotics. Ten million cells were allowed to attach to each of five plastic culture dishes overnight. The next day, the adenohypophyseal cells were surface-labeled with 125I. The labeled cells were lysed and centrifuged. The supernatant was immunoprecipitated using preimmune IgGs (100 micrograms/ml) and was then incubated with a polyclonal antibody against the rat beta 1 family of integrins or with a variety of immune IgGs directed against the alpha subunit of the receptor (anti alpha 1, anti alpha 2, anti alpha 3, and anti alpha 5 antibodies). The receptors were then immunoprecipitated by addition of protein A-Sepharose or IgG1 Sepharose. After washing, the immunoprecipitates were subjected to sodium dodecyl sulfate polyacrylamide gel electrophoresis and autoradiography. Cultured adenohypophyseal cells expressed the beta 1 integrin subunit, which was associated with the alpha 1, alpha 2, alpha 3, and alpha 5 integrin subunits.(ABSTRACT TRUNCATED AT 250 WORDS)
Collapse
Affiliation(s)
- M J Horacek
- Department of Developmental Biology and Anatomy, University of South Carolina, School of Medicine, Columbia 29208
| | | | | |
Collapse
|
50
|
Abstract
Peripheral nerve cells, various endocrine and pigment cells and cranial connective tissue cells of vertebrates stem mainly from the embryonic neural crest. This originates with the central nervous system, but the crest cells detach from this tissue, via a decrease of cell-cell adhesion involving, particularly, a reduction of the adherens junction cell adhesive molecule A-CAM. This epithelio-mesenchymal transformation allows crest cells to migrate along pathways that are defined partly by the distribution of substrate adhesion molecules, the archetype being fibronectin, an extracellular matrix molecule recognized by integrin receptors on crest cells. Many other molecules, however, may act in the same way. In contrast, some molecules may define migration pathways by reducing adhesion; chondroitin sulfate proteoglycan is a candidate for this role. Pathway selection is most likely achieved by balanced combinations of molecules that promote and reduce adhesion. Cessation of migration, in the case of the nervous ganglia, correlated with re-expression of cell-cell adhesion molecules like A-CAM and others, consistent with an adhesive basis, although functional tests have not yet been performed. The development of the neural crest system provides a useful model that emphasizes the role of adhesion in morphogenesis.
Collapse
Affiliation(s)
- D F Newgreen
- Embryology Laboratory, Murdoch Institute, Parkville, Victoria, Australia
| | | |
Collapse
|