Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Nature ; 618(7965): 543-549, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37225983

ABSTRACT

The development of paired appendages was a key innovation during evolution and facilitated the aquatic to terrestrial transition of vertebrates. Largely derived from the lateral plate mesoderm (LPM), one hypothesis for the evolution of paired fins invokes derivation from unpaired median fins via a pair of lateral fin folds located between pectoral and pelvic fin territories1. Whilst unpaired and paired fins exhibit similar structural and molecular characteristics, no definitive evidence exists for paired lateral fin folds in larvae or adults of any extant or extinct species. As unpaired fin core components are regarded as exclusively derived from paraxial mesoderm, any transition presumes both co-option of a fin developmental programme to the LPM and bilateral duplication2. Here, we identify that the larval zebrafish unpaired pre-anal fin fold (PAFF) is derived from the LPM and thus may represent a developmental intermediate between median and paired fins. We trace the contribution of LPM to the PAFF in both cyclostomes and gnathostomes, supporting the notion that this is an ancient trait of vertebrates. Finally, we observe that the PAFF can be bifurcated by increasing bone morphogenetic protein signalling, generating LPM-derived paired fin folds. Our work provides evidence that lateral fin folds may have existed as embryonic anlage for elaboration to paired fins.


Subject(s)
Animal Fins , Biological Evolution , Mesoderm , Zebrafish , Animals , Animal Fins/anatomy & histology , Animal Fins/embryology , Animal Fins/growth & development , Larva/anatomy & histology , Larva/growth & development , Mesoderm/anatomy & histology , Mesoderm/embryology , Mesoderm/growth & development , Zebrafish/anatomy & histology , Zebrafish/embryology , Zebrafish/growth & development , Bone Morphogenetic Proteins/metabolism
2.
EMBO Rep ; 23(8): e54464, 2022 08 03.
Article in English | MEDLINE | ID: mdl-35679135

ABSTRACT

Immigration of mesenchymal cells into the growing fin and limb buds drives distal outgrowth, with subsequent tensile forces between these cells essential for fin and limb morphogenesis. Morphogens derived from the apical domain of the fin, orientate limb mesenchyme cell polarity, migration, division and adhesion. The zebrafish mutant stomp displays defects in fin morphogenesis including blister formation and associated loss of orientation and adhesion of immigrating fin mesenchyme cells. Positional cloning of stomp identifies a mutation in the gene encoding the axon guidance ligand, Slit3. We provide evidence that Slit ligands derived from immigrating mesenchyme act via Robo receptors at the apical ectodermal ridge (AER) to promote release of sphingosine-1-phosphate (S1P). S1P subsequently diffuses back to the mesenchyme to promote their polarisation, orientation, positioning and adhesion to the interstitial matrix of the fin fold. We thus demonstrate the coordination of the Slit-Robo and S1P signalling pathways in fin fold morphogenesis. Our work introduces a mechanism regulating the orientation, positioning and adhesion of its constituent cells.


Subject(s)
Gene Expression Regulation, Developmental , Zebrafish , Animals , Intracellular Signaling Peptides and Proteins/genetics , Lysophospholipids , Mesoderm/metabolism , Sphingosine/analogs & derivatives , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
3.
PLoS One ; 16(11): e0260372, 2021.
Article in English | MEDLINE | ID: mdl-34847198

ABSTRACT

Collective cell migration is essential for embryonic development and homeostatic processes. During zebrafish development, the posterior lateral line primordium (pLLP) navigates along the embryo flank by collective cell migration. The chemokine receptors, Cxcr4b and Cxcr7b, as well as their cognate ligand, Cxcl12a, are essential for this process. We corroborate that knockdown of the zebrafish cd9 tetraspanin orthologue, cd9b, results in mild pLL abnormalities. Through generation of CRISPR and TALEN mutants, we show that cd9a and cd9b function partially redundantly in pLLP migration, which is delayed in the cd9b single and cd9a; cd9b double mutants. This delay led to a transient reduction in neuromast numbers. Loss of both Cd9a and Cd9b sensitized embryos to reduced Cxcr4b and Cxcl12a levels. Together these results provide evidence that Cd9 modulates collective cell migration of the pLLP during zebrafish development. One interpretation of these observations is that Cd9 contributes to more effective chemokine signalling.


Subject(s)
Cell Movement , Chemokine CXCL12/metabolism , Receptors, CXCR4/metabolism , Signal Transduction , Tetraspanin 29/metabolism , Zebrafish Proteins/metabolism , Zebrafish/embryology , Animals , Animals, Genetically Modified/embryology , Animals, Genetically Modified/genetics , Chemokine CXCL12/genetics , Gene Knockdown Techniques , Receptors, CXCR4/genetics , Tetraspanin 29/genetics , Zebrafish/genetics , Zebrafish Proteins/genetics
4.
Elife ; 102021 06 24.
Article in English | MEDLINE | ID: mdl-34165081

ABSTRACT

Epithelial tissues are primed to respond to insults by activating epithelial cell motility and rapid inflammation. Such responses are also elicited upon overexpression of the membrane-bound protease, Matriptase, or mutation of its inhibitor, Hai1. Unrestricted Matriptase activity also predisposes to carcinoma. How Matriptase leads to these cellular outcomes is unknown. We demonstrate that zebrafish hai1a mutants show increased H2O2, NfκB signalling, and IP3R -mediated calcium flashes, and that these promote inflammation, but do not generate epithelial cell motility. In contrast, inhibition of the Gq subunit in hai1a mutants rescues both the inflammation and epithelial phenotypes, with the latter recapitulated by the DAG analogue, PMA. We demonstrate that hai1a has elevated MAPK pathway activity, inhibition of which rescues the epidermal defects. Finally, we identify RSK kinases as MAPK targets disrupting adherens junctions in hai1a mutants. Our work maps novel signalling cascades mediating the potent effects of Matriptase on epithelia, with implications for tissue damage response and carcinoma progression.


Cancer occurs when normal processes in the cell become corrupted or unregulated. Many proteins can contribute, including one enzyme called Matriptase that cuts other proteins at specific sites. Matriptase activity is tightly controlled by a protein called Hai1. In mice and zebrafish, when Hai1 cannot adequately control Matriptase activity, invasive cancers with severe inflammation develop. However, it is unclear how unregulated Matriptase leads to both inflammation and cancer invasion. One outcome of Matriptase activity is removal of proteins called Cadherins from the cell surface. These proteins have a role in cell adhesion: they act like glue to stick cells together. Without them, cells can dissociate from a tissue and move away, a critical step in cancer cells invading other organs. However, it is unknown exactly how Matriptase triggers the removal of Cadherins from the cell surface to promote invasion. Previous work has shown that Matriptase switches on a receptor called Proteinase-activated receptor 2, or Par2 for short, which is known to activate many enzymes, including one called phospholipase C. When activated, this enzyme releases two signals into the cell: a sugar called inositol triphosphate, IP3; and a lipid or fat called diacylglycerol, DAG. It is possible that these two signals have a role to play in how Matriptase removes Cadherins from the cell surface. To find out, Ma et al. mapped the effects of Matriptase in zebrafish lacking the Hai1 protein. This revealed that Matriptase increases IP3 and DAG levels, which initiate both inflammation and invasion. IP3 promotes inflammation by switching on pro-inflammatory signals inside the cell such as the chemical hydrogen peroxide. At the same time, DAG promotes cell invasion by activating a well-known cancer signalling pathway called MAPK. This pathway activates a protein called RSK. Ma et al. show that this protein is required to remove Cadherins from the surface of cells, thus connecting Matriptase's activation of phospholipase C with its role in disrupting cell adhesion. An increase in the ratio of Matriptase to HAI-1 (the human equivalent of Hai1) is present in many cancers. For this reason, the signal cascades described by Ma et al. may be of interest in developing treatments for these cancers. Understanding how these signals work together could lead to more direct targeted anti-cancer approaches in the future.


Subject(s)
GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Serine Endopeptidases/metabolism , Animals , Animals, Genetically Modified , Calcium/metabolism , Calcium Signaling , DNA/genetics , Embryo, Nonmammalian , Enzyme Activation , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , Gene Expression Regulation, Enzymologic/drug effects , Hydrogen Peroxide , Inflammation/metabolism , Mutation , Neutrophils/physiology , Peptides, Cyclic , Polymerase Chain Reaction , RNA/genetics , Serine Endopeptidases/genetics , Zebrafish
5.
Dev Cell ; 43(5): 577-587.e5, 2017 12 04.
Article in English | MEDLINE | ID: mdl-29173819

ABSTRACT

Cell migration is essential for morphogenesis, organ formation, and homeostasis, with relevance for clinical conditions. The migration of primordial germ cells (PGCs) is a useful model for studying this process in the context of the developing embryo. Zebrafish PGC migration depends on the formation of cellular protrusions in form of blebs, a type of protrusion found in various cell types. Here we report on the mechanisms allowing the inflation of the membrane during bleb formation. We show that the rapid expansion of the protrusion depends on membrane invaginations that are localized preferentially at the cell front. The formation of these invaginations requires the function of Cdc42, and their unfolding allows bleb inflation and dynamic cell-shape changes performed by migrating cells. Inhibiting the formation and release of the invaginations strongly interfered with bleb formation, cell motility, and the ability of the cells to reach their target.


Subject(s)
Cell Membrane/metabolism , Cell Movement/physiology , Cell Shape/physiology , Germ Cells/cytology , Zebrafish , Actins/metabolism , Animals , Cell Membrane Structures/metabolism , Cell Surface Extensions/metabolism , Germ Cells/metabolism , Zebrafish/metabolism
6.
Development ; 139(16): 2897-902, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22791893

ABSTRACT

A crucial regulator of Cxcl12 is the decoy receptor Cxcr7, which controls the level of the chemokine in the tissue. The molecular mechanisms that enable Cxcr7 to function as an efficient molecular sink are not known. Using zebrafish primordial germ cells as a model, we identify a novel role for ß-arrestins in controlling the intracellular trafficking of Cxcr7. ß-arrestins facilitate the recycling of Cxcr7 from late endosomal compartments back to the plasma membrane, whereas the internalized ligand undergoes lysosomal degradation. ß-arrestins thus function in regulating chemokine gradient formation, allowing responding cells to discriminate between alternative migration targets in vivo.


Subject(s)
Arrestins/metabolism , Receptors, CXCR/metabolism , Zebrafish Proteins/metabolism , Animals , Animals, Genetically Modified , Arrestins/antagonists & inhibitors , Arrestins/genetics , Cell Movement/physiology , Chemokine CXCL12/genetics , Chemokine CXCL12/metabolism , Endosomes/metabolism , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Germ Cells/cytology , Germ Cells/metabolism , Receptors, CXCR/genetics , Tissue Distribution , Zebrafish/embryology , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/antagonists & inhibitors , Zebrafish Proteins/genetics , beta-Arrestins
7.
PLoS One ; 5(2): e9175, 2010 Feb 11.
Article in English | MEDLINE | ID: mdl-20161793

ABSTRACT

BACKGROUND: CXCR7 (RDC1), the recently discovered second receptor for CXCL12, is phylogenetically closely related to chemokine receptors, but fails to couple to G-proteins and to induce typical chemokine receptor mediated cellular responses. The function of CXCR7 is controversial. Some studies suggest a signaling activity in mammalian cells and zebrafish embryos, while others indicate a decoy activity in fish. Here we investigated the two propositions in human tissues. METHODOLOGY/PRINCIPAL FINDINGS: We provide evidence and mechanistic insight that CXCR7 acts as specific scavenger for CXCL12 and CXCL11 mediating effective ligand internalization and targeting of the chemokine cargo for degradation. Consistently, CXCR7 continuously cycles between the plasma membrane and intracellular compartments in the absence and presence of ligand, both in mammalian cells and in zebrafish. In accordance with the proposed activity as a scavenger receptor CXCR7-dependent chemokine degradation does not become saturated with increasing ligand concentrations. Active CXCL12 sequestration by CXCR7 is demonstrated in adult mouse heart valves and human umbilical vein endothelium. CONCLUSIONS/SIGNIFICANCE: The finding that CXCR7 specifically scavenges CXCL12 suggests a critical function of the receptor in modulating the activity of the ubiquitously expressed CXCR4 in development and tumor formation. Scavenger activity of CXCR7 might also be important for the fine tuning of the mobility of hematopoietic cells in the bone marrow and lymphoid organs.


Subject(s)
Chemokine CXCL11/metabolism , Chemokine CXCL12/metabolism , Receptors, CXCR/physiology , Animals , Cell Line , Cell Line, Tumor , Cell Membrane/metabolism , Cells, Cultured , Chemokine CXCL11/genetics , Chemokine CXCL12/genetics , Embryo, Nonmammalian/metabolism , Endocytosis/physiology , Endothelial Cells/cytology , Endothelial Cells/metabolism , Female , Flow Cytometry , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Ligands , Male , Mice , Mice, Inbred BALB C , Microscopy, Confocal , Myocardium/metabolism , Receptors, CXCR/genetics , Receptors, CXCR/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transfection , Zebrafish
8.
Development ; 136(8): 1223-9, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19304885

ABSTRACT

Chemokines and their receptors were discovered about twenty years ago as mediators of leukocyte traffic. Over the past decade, functional studies of these molecules have revealed their importance for cell migration processes during embryogenesis, which, in addition to providing mechanistic insights into embryonic development, could complement information about chemokine function in the immune system. Here, we review the roles of the chemokine stromal cell-derived factor 1 (SDF-1/CXCL12) and its receptor CXCR4 during zebrafish and mouse embryonic development, and discuss their function in regulating the interactions of cells with their extracellular environment, in directing their migration, and in maintaining their location.


Subject(s)
Cell Movement , Chemokines/metabolism , Embryo, Nonmammalian/metabolism , Signal Transduction , Zebrafish/embryology , Zebrafish/metabolism , Animals , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/embryology , Humans
9.
Cell ; 132(3): 463-73, 2008 Feb 08.
Article in English | MEDLINE | ID: mdl-18267076

ABSTRACT

Primordial germ cell (PGC) migration in zebrafish is directed by the chemokine SDF-1a that activates its receptor CXCR4b. Little is known about the molecular mechanisms controlling the distribution of this chemoattractant in vivo. We demonstrate that the activity of a second SDF-1/CXCL12 receptor, CXCR7, is crucial for proper migration of PGCs toward their targets. We show that CXCR7 functions primarily in the somatic environment rather than within the migrating cells. In CXCR7 knocked-down embryos, the PGCs exhibit a phenotype that signifies defects in SDF-1a gradient formation as the cells fail to polarize effectively and to migrate toward their targets. Indeed, somatic cells expressing CXCR7 show enhanced internalization of the chemokine suggesting that CXCR7 acts as a sink for SDF-1a, thus allowing the dynamic changes in the transcription of sdf-1a to be mirrored by similar dynamics at the protein level.


Subject(s)
Cell Movement , Chemokine CXCL12/metabolism , Germ Cells/cytology , Receptors, CXCR/metabolism , Zebrafish Proteins/metabolism , Zebrafish/embryology , Animals , Animals, Genetically Modified , Cell Polarity , Embryo, Nonmammalian/cytology , Gene Expression Regulation, Developmental , Receptors, CXCR/genetics , Zebrafish Proteins/genetics
10.
Cell Adh Migr ; 2(2): 69-70, 2008.
Article in English | MEDLINE | ID: mdl-19262101

ABSTRACT

Primordial Germ Cell (PGC) migration in zebrafish is guided by SDF-1a. Binding of this chemokine to its receptor CXCR4b activates downstream signalling cascades leading to cell polarization and directed migration towards the attractant source. Despite the detailed information available concerning the role of SDF-1 in guiding the PGCs to their targets, little was known regarding the molecular mechanisms controlling the distribution of SDF-1a within the tissue. We have recently shown that the activity of a second SDF-1/CXCL12 receptor, CXCR7 is crucial for proper migration of PGCs. Although CXCR4 and CXCR7 are structurally related and serve as receptors for the same ligand, they appear to serve very different functions during PGC migration. Here we discuss a model according to which CXCR4b translates the polarized distribution of SDF-1 into directed PGC migration, while CXCR7 acts as a high-affinity decoy receptor and facilitates the migration of PGCs by shaping the distribution of the chemokine in the environment.


Subject(s)
Cell Movement , Germ Cells/cytology , Germ Cells/metabolism , Receptors, CXCR/metabolism , Zebrafish/metabolism , Animals , Chemokine CXCL12/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...