Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Mol Biol Cell ; 12(10): 2975-86, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11598185

ABSTRACT

The urokinase receptor (uPAR) is linked to cellular migration through its capacity to promote pericellular proteolysis, regulate integrin function, and mediate cell signaling in response to urokinase (uPA) binding. The mechanisms for these activities remain incompletely defined, although uPAR was recently identified as a cis-acting ligand for the beta2 integrin CD11b/CD18 (Mac-1). Here we show that a major beta1 integrin partner for uPAR/uPA signaling is alpha3. In uPAR-transfected 293 cells uPAR complexed (>90%) with alpha3beta1 and antibodies to alpha3 blocked uPAR-dependent vitronectin (Vn) adhesion. Soluble uPAR bound to recombinant alpha3beta1 in a uPA-dependent manner (K(d) < 20 nM) and binding was blocked by a 17-mer alpha3beta1 integrin peptide (alpha325) homologous to the CD11b uPAR-binding site. uPAR colocalized with alpha3beta1 in MDA-MB-231 cells and uPA (1 nM) enhanced spreading and focal adhesion kinase phosphorylation on fibronectin (Fn) or collagen type I (Col) in a pertussis toxin- and alpha325-sensitive manner. A critical role of alpha3beta1 in uPA signaling was verified by studies of epithelial cells from alpha3-deficient mice. Thus, uPAR preferentially complexes with alpha3beta1, promoting direct (Vn) and indirect (Fn, Col) pathways of cell adhesion, the latter a heterotrimeric G protein-dependent mechanism of signaling between alpha3beta1 and other beta1 integrins.


Subject(s)
Cell Movement/physiology , Integrin beta1/metabolism , Integrins/metabolism , Receptors, Cell Surface/metabolism , Urokinase-Type Plasminogen Activator/metabolism , Binding Sites/physiology , CD18 Antigens , Cell Line , Cell Movement/drug effects , Collagen/metabolism , Fibronectins/metabolism , Heterotrimeric GTP-Binding Proteins/chemistry , Humans , Integrin alpha3beta1 , Integrins/chemistry , Integrins/genetics , Macrophage-1 Antigen/genetics , Macrophage-1 Antigen/metabolism , Precipitin Tests , Receptors, Urokinase Plasminogen Activator , Recombinant Proteins/metabolism , Sequence Homology , Signal Transduction , Tumor Cells, Cultured , Urokinase-Type Plasminogen Activator/pharmacology , src-Family Kinases/chemistry
2.
Dev Dyn ; 222(1): 26-39, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11507767

ABSTRACT

Epithelial-mesenchymal tissue interactions play a central role in vertebrate organogenesis, but the molecular mediators and mechanisms of these morphogenetic interactions are still not well characterized. We report here on the expression pattern of Wnt-2b during mouse organogenesis and on tests of its function in epithelial- mesenchymal interactions during kidney development. Wnt-2b is expressed in numerous developing organs in the mouse embryo, including the kidney, lung, salivary gland, gut, pancreas, adrenal gland, and genital tubercle. Additional sites of expression include the branchial arches and craniofacial placodes such as the eye and ear. The data suggest that the expression of Wnt-2b is associated with organs regulated by epithelial-mesenchymal interactions. It is typically localized in the capsular epithelium or peripheral mesenchymal cells of organ rudiments, e.g., the perinephric mesenchymal cells in the region of the presumptive renal stroma in the developing kidney at E11.5. Functional studies of the kidney demonstrate that cells expressing Wnt-2b are not capable of inducing tubule formation but instead stimulate ureter development. Incubation of isolated ureteric buds on such cells supports bud growth and branching. In addition, recombination of Wnt-2b-pretreated ureteric bud tissue with isolated nephrogenic mesenchyme results in a recovery of organogenesis and the expression of epithelial genes within the reconstituted organ explant. Lithium, a known activator of Wnt signaling (Hedgepeth et al. [1997] Dev Biol 185:82-91), is also sufficient to promote ureter branching in the reconstituted kidney in a comparable manner to Wnt-2b signaling, whereas Wnt-4, which induces tubules, neither supports the growth of a ureteric bud nor leads to reconstitution of the ureteric bud with the kidney mesenchyme. We conclude that Wnt-2b may act in the mouse kidney as an early mesenchymal signal controlling morphogenesis of epithelial tissue, and that the Wnt pathway may regulate ureter branching directly. In addition, Wnt signals in the kidney differ qualitatively and are specific to either the epithelial ureteric bud or the kidney mesenchyme.


Subject(s)
Glycoproteins/metabolism , Intercellular Signaling Peptides and Proteins , Kidney/embryology , Ureter/physiology , 3T3 Cells , Animals , Cloning, Molecular , Coculture Techniques , DNA, Complementary/metabolism , Glycoproteins/physiology , In Situ Hybridization , Kidney Tubules/embryology , Kidney Tubules/metabolism , Mice , Models, Biological , Protein Binding , Proto-Oncogene Proteins/metabolism , RNA/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Time Factors , Tissue Distribution , Wnt Proteins , Wnt4 Protein
3.
Dev Biol ; 233(2): 449-67, 2001 May 15.
Article in English | MEDLINE | ID: mdl-11336507

ABSTRACT

In the mammary gland, both laminin and integrins have been shown to be required for normal ductal morphogenesis during development in vivo, and for functional differentiation in culture models. Major integrin receptors for laminins in the mammary gland are alpha 3 beta 1, alpha 6 beta 1, and alpha 6 beta 4. However, the specific subunits that contribute to laminin-mediated mammary cell function and development have not been identified. In this study, we use a genetic approach to test the hypothesis that laminin-binding integrins are required for the function of the mammary gland in vivo. Rudiments of embryonic mammary gland were shown to develop in the absence of these integrin subunits. Postnatal development of the mammary gland was studied in integrin null tissue that had been transplanted into the mammary fat pads of syngeneic hosts. In mammary epithelium lacking alpha 6 integrin, the beta 4 subunit was not apparent and hemidesmosome formation was only rudimentary. However, despite this deficiency, normal ductal morphogenesis and branching of the mammary gland occurred and myoepithelial cells were distributed normally with respect to luminal cells. Mammary alveoli devoid of alpha 3 or alpha 6 integrin formed in pregnancy and were histologically and functionally identical to those in wild-type mammary gland. The tissue underwent full morphological differentiation, and the epithelial cells retained the ability to synthesize beta-casein. This work demonstrates that mammary tissue genetically lacking major laminin-binding integrin receptors is still able to develop and function.


Subject(s)
Antigens, CD/physiology , Integrins/physiology , Mammary Glands, Animal/embryology , Mammary Glands, Animal/growth & development , Animals , Antigens, CD/genetics , Basement Membrane/ultrastructure , Body Patterning/genetics , Body Patterning/physiology , Cell Differentiation , Epithelium/embryology , Epithelium/growth & development , Epithelium/metabolism , Female , Hemidesmosomes/ultrastructure , Integrin alpha3 , Integrin alpha6 , Integrin beta4 , Integrins/genetics , Laminin/metabolism , Mammary Glands, Animal/metabolism , Mammary Glands, Animal/transplantation , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron , Pregnancy , Transplantation, Isogeneic
4.
Dev Dyn ; 220(4): 337-49, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11307167

ABSTRACT

Mammalian submandibular gland (SMG) development leads to the establishment of highly organized secretory acinar and nonsecretory ductal epithelial cells. The ability of maturing salivary epithelial cells to attain their differentiated state has been shown to depend, in part, on interactions between extracellular matrix (ECM) proteins and their integrin receptors. In a search for key regulators of salivary cell lineage, we have studied alpha3beta1 integrin, a receptor for the basement membrane protein laminin, by characterizing embryonic day 18 (E18) SMGs isolated from mice carrying a targeted mutation in the alpha3 integrin gene. Transmission electron microscopy studies showed that the mutant SMGs exhibited an aberrant differentiation phenotype with defects in the apical-basal polarity axis and in the basement membrane. Based on immunohistochemistry and Western blot analyses, the alpha3beta1-deficient SMGs had altered expression and/or localization of several ECM and adhesive molecules, including laminin beta1, fibronectin, alpha5 integrin, and E-cadherin. These changes correlated with alterations in the activation state of Ras-extracellular signal-regulated kinase (ERK), as well as the expression and/or localization of Cdc42 and RhoA, two Rho GTPases that regulate the organization of the actin cytoskeleton. We conclude that alpha3beta1 is required for normal salivary cell differentiation and that its absence affects multiple components of adhesive complexes and their associated signalling pathways.


Subject(s)
Integrins/genetics , Integrins/physiology , Submandibular Gland/embryology , Actins/metabolism , Animals , Antigens, CD/metabolism , Blotting, Western , Cadherins/biosynthesis , Cadherins/metabolism , Cell Adhesion , Cell Differentiation , Cell Membrane/metabolism , Cytoskeleton/metabolism , Fibronectins/metabolism , Fluorescent Antibody Technique , Immunohistochemistry , Integrin alpha3beta1 , Integrin alpha5 , Keratins/metabolism , Laminin/metabolism , Mice , Microscopy, Confocal , Microscopy, Electron , Mitogen-Activated Protein Kinases/metabolism , Mutagenesis, Site-Directed , Phenotype , Signal Transduction , Submandibular Gland/ultrastructure , Time Factors , cdc42 GTP-Binding Protein/metabolism , rho GTP-Binding Proteins/metabolism , rhoA GTP-Binding Protein/metabolism
5.
Dev Biol ; 238(2): 289-302, 2001 Oct 15.
Article in English | MEDLINE | ID: mdl-11784011

ABSTRACT

Branching morphogenesis of the ureteric bud (UB) [induced by the metanephric mesenchyme (MM)] is necessary for normal kidney development. The role of integrins in this complex developmental process is not well understood. However, the recent advent of in vitro model systems to study branching of UB cells and isolated UB tissue makes possible a more detailed analysis of the integrins involved. We detected integrin subunits alpha3, alpha6, beta1, and beta4 in both the UB and cells derived from the early UB. Blocking the function of each of these integrin subunits individually markedly inhibited branching morphogenesis in cell culture models. However, inhibiting individual integrin function with blocking antibodies in whole kidney and isolated UB culture only partially inhibited UB branching morphogenesis, suggesting that, in these more complex in vitro systems, multiple integrins are involved in the branching program. In whole organ and isolated bud culture, marked retardation of UB branching was observed only when both alpha3 and alpha6 integrin subunits were inhibited. The alpha6 integrin subunit can be expressed as both alpha6beta1 and alpha6beta4, and both of these beta subunits are important for UB branching morphogenesis in both cell and organ culture. Furthermore, laminin-5, a common ligand for integrins alpha3beta1 and alpha6beta4, was detected in the developing UB and shown to be required for normal UB branching morphogenesis in whole embryonic kidney organ culture as well as isolated UB culture. Together, these data from UB cell culture, organ culture, and isolated UB culture models indicate that both integrin alpha3 and alpha6 subunits play a direct role in UB branching morphogenesis, as opposed to being modulators of the inductive effects of mesenchyme on UB development. Furthermore the data are consistent with a role for laminin-5, acting through its alpha3beta1 and/or alpha6beta4 integrin receptors, in UB branching during nephrogenesis. These data may help to partially explain the renal phenotype seen in integrin alpha3 and alpha3/alpha6 subunit-deficient animals.


Subject(s)
Cell Adhesion Molecules/metabolism , Kidney/embryology , Laminin/metabolism , Ureter/embryology , Urethra/embryology , Animals , Antigens, CD/biosynthesis , Cell Adhesion Molecules/biosynthesis , Cells, Cultured , Flow Cytometry , Gene Expression Regulation, Developmental , Immunohistochemistry , Integrin alpha3 , Integrin alpha6 , Integrin alpha6beta1 , Integrins/biosynthesis , Integrins/metabolism , Kidney Tubules/embryology , Lectins/metabolism , Ligands , Mice , Mice, Knockout , Microscopy, Confocal , Organ Culture Techniques , Phenotype , Precipitin Tests , Protein Binding , Rats , Reverse Transcriptase Polymerase Chain Reaction , Kalinin
6.
Curr Opin Cell Biol ; 12(5): 548-53, 2000 Oct.
Article in English | MEDLINE | ID: mdl-10978888

ABSTRACT

alpha3beta1 integrin is a laminin receptor with apparently diverse functions. In epithelial cells it acts as a receptor for the basement membrane, whereas in neuronal and possibly tumor cells it mediates migration. Interactions of alpha3beta1 integrin with tetraspanin proteins may provide clues to how it transduces signals that affect cell behavior.


Subject(s)
Integrins/physiology , Animals , Cell Adhesion/physiology , Cell Movement/physiology , Humans , Integrin alpha3beta1 , Signal Transduction
7.
Neuron ; 27(1): 33-44, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10939329

ABSTRACT

Mice that are mutant for Reelin or Dab1, or doubly mutant for the VLDL receptor (VLDLR) and ApoE receptor 2 (ApoER2), show disorders of cerebral cortical lamination. How Reelin and its receptors regulate laminar organization of cerebral cortex is unknown. We show that Reelin inhibits migration of cortical neurons and enables detachment of neurons from radial glia. Recombinant and native Reelin associate with alpha3beta1 integrin, which regulates neuron-glia interactions and is required to achieve proper laminar organization. The effect of Reelin on cortical neuronal migration in vitro and in vivo depends on interactions between Reelin and alpha3beta1 integrin. Absence of alpha3beta1 leads to a reduction of Dab1, a signaling protein acting downstream of Reelin. Thus, Reelin may arrest neuronal migration and promote normal cortical lamination by binding alpha3beta1 integrin and modulating integrin-mediated cellular adhesion.


Subject(s)
Cell Adhesion Molecules, Neuronal/metabolism , Cell Adhesion Molecules, Neuronal/physiology , Extracellular Matrix Proteins/metabolism , Extracellular Matrix Proteins/physiology , Integrins/metabolism , Neurons/physiology , Adaptor Proteins, Signal Transducing , Animals , Antibodies, Monoclonal , Cell Movement/physiology , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Humans , Immunohistochemistry , Integrin alpha3beta1 , Integrins/genetics , Mice , Mice, Neurologic Mutants , Nerve Tissue Proteins/biosynthesis , Precipitin Tests , Receptors, Cell Surface/metabolism , Reelin Protein , Serine Endopeptidases
8.
J Cell Sci ; 113 ( Pt 17): 3051-62, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10934043

ABSTRACT

Continuous regeneration and homeostasis of the stratified epidermis requires coordinated regulation of cell proliferation, cell differentiation, and cell survival. Integrin-mediated cell adhesion to the extracellular matrix has important roles in regulating each of these processes. Integrins alpha3beta1 and alpha6beta4 are both receptors on epidermal keratinocytes for the basement membrane protein laminin-5, the major ligand for epidermal adhesion in mature skin. Ablation in mice of either alpha3beta1 or alpha6beta4, through null mutation of the gene encoding the alpha3, alpha6, or beta4 integrin subunit, results in epidermal blistering of varying severity. Our previous studies showed that, despite blistering, differentiation and stratification of the epidermis appeared essentially normal in mice that lacked either alpha3beta1 or alpha6beta4. However, these studies did not definitively address the specific developmental importance of each integrin, since they may have overlapping and/or compensatory functions. Given the individual importance of alpha3beta1 or alpha6beta4 in maintaining the dermo-epidermal junction in mature skin, we sought to determine the importance of these integrins for embryonic skin development and epidermal morphogenesis. In the current study, we analyzed skin development in mutant embryos that completely lack both integrins alpha3beta1 and alpha6beta4. Although alpha3beta1/alpha6beta4-deficient embryos displayed epidermal blistering by stage E15.5 of development, they also retained regions of extensive epidermal adhesion to the basement membrane through stage E16.5, indicating alternative adhesion mechanisms. Apoptosis was induced in detached epidermis of alpha3beta1/alpha6beta4-deficient embryos, exemplifying vividly the importance of epithelial attachment to the basement membrane for cell survival. However, apoptotic cells were completely absent from attached epidermis of alpha3beta1/alpha6beta4-deficient embryos, showing that epithelial adhesion that occurred independently of alpha3beta1 and alpha6beta4 also protected cells from apoptosis. Remarkably, in the absence of the known laminin-5 binding integrins (alpha3beta1, alpha6beta4, and alpha6beta1), keratinocytes retained the capacity to proliferate in the epidermis, and epidermal stratification and skin morphogenesis appeared normal prior to blister formation. These findings show that while alpha3beta1 and alpha6beta4 are both required for integrity of the dermo-epidermal junction, neither one is essential for epidermal morphogenesis during skin development.


Subject(s)
Antigens, Surface/physiology , Cell Adhesion Molecules/metabolism , Epidermis/embryology , Integrins/physiology , Morphogenesis , Skin/embryology , Animals , Antigens, Surface/genetics , Apoptosis , Basement Membrane/anatomy & histology , Cell Adhesion , Cell Division , Cell Survival , Epidermis/anatomy & histology , Fluorescent Antibody Technique , Homeostasis , Integrin alpha3beta1 , Integrin alpha6beta4 , Integrins/genetics , Keratinocytes/physiology , Mice , Mice, Knockout , Models, Biological , Skin/anatomy & histology , Kalinin
9.
Am J Physiol Renal Physiol ; 279(2): F233-42, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10919841

ABSTRACT

Integrins are heterodimeric cell surface receptors that mediate heterophilic cell-cell interactions and interactions between cells and the extracellular matrix (Hynes RO. Cell 69: 11-25, 1991). As such, they are involved in morphogenetic processes during development, as well as in the maintenance of normal tissue architecture in fully developed organs. Integrins are now recognized to be a large family of receptors, and several different integrins have been demonstrated as being expressed in the developing and adult kidney (Korhonen M, Ylkanne J, Laitinen L, and Virtanen I. Development 122: 3537-3547, 1996; Rahilly MA and Fleming S. J Pathol 167: 327-334, 1992). This review will summarize present knowledge about integrin expression in the developing, normal, and diseased kidney and attempt to provide a hypothetical framework for understanding integrin function in the urogenital system. Since the last time this area was reviewed (Hamerski DA and Santoro S. Curr Opin Nephrol Hypertens 8: 9-14, 1999), there have been significant publications on the roles of integrins in kidney development and disease. At present, there are many more questions than answers, and integrins present an area where many novel and exciting findings will emerge in the coming years.


Subject(s)
Integrins/physiology , Kidney/embryology , Kidney/growth & development , Aging/physiology , Animals , Embryo, Mammalian/physiology , Embryonic and Fetal Development , Humans , Integrins/metabolism , Kidney Diseases/physiopathology
10.
J Cell Sci ; 113 ( Pt 16): 2909-21, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10910775

ABSTRACT

Remodeling of the extracellular matrix during tissue development, wound repair and tumor cell invasion depends on the coordinated regulation of cell adhesion receptors, matrix proteins and enzymes that proteolyse the extracellular matrix. Integrin alpha3beta1 is a major receptor on epidermal keratinocytes for laminin-5 in the cutaneous basement membrane and is required for normal basement membrane organization during skin development. alpha3beta1 is also expressed at high levels in the majority of adherent transformed cells and in most tumors, and it could have similar roles in extracellular matrix remodeling during tumorigenesis and cell invasion. In the present study, we show that alpha3beta1 expression is required in immortalized mouse keratinocytes (MK) for the production of the matrix metalloproteinase MMP-9/gelatinase B, an MMP that is coexpressed with alpha3beta1 in epithelial cell carcinomas and during wound healing, and contributes to the invasive potential of some tumor cells. MMP-9 was expressed in MK cells derived from wild-type mice, but not in MK cells derived from alpha3-null mice. Reconstitution of alpha3beta1 expression in alpha3-null MK cells through transfection with the alpha3 subunit restored MMP-9 secretion, indicating an alpha3beta1-dependent pathway for MMP-9 production. alpha3beta1-dependent expression of MMP-9 was associated with the immortalized phenotype, since nonimmortalized, primary keratinocytes required soluble growth factors, but not alpha3beta1, for efficient expression of MMP-9. Our results suggest that an alpha3beta1-independent pathway(s) for MMP-9 production is suppressed in keratinocytes immortalized with large T antigen, and that an alpha3beta1-dependent pathway is required for sustained production of MMP-9 in the absence of other pathways.


Subject(s)
Antigens, Polyomavirus Transforming/genetics , Integrins/metabolism , Keratinocytes/enzymology , Keratinocytes/metabolism , Matrix Metalloproteinase 9/metabolism , Actins/metabolism , Animals , Cell Adhesion Molecules/metabolism , Cell Adhesion Molecules/pharmacology , Cell Division/drug effects , Cell Division/physiology , Cell Line, Transformed , Cytoskeleton/metabolism , Extracellular Matrix/enzymology , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Integrin alpha3beta1 , Keratinocytes/cytology , Matrix Metalloproteinase 9/genetics , Mice , Signal Transduction/physiology , Skin/cytology , Kalinin
11.
Nature ; 403(6772): 909-13, 2000 Feb 24.
Article in English | MEDLINE | ID: mdl-10706291

ABSTRACT

During mammalian embryonic development, the ovaries and testes develop from somatic cells of the urogenital ridges as indifferent gonads, harbouring primordial germ cells that have migrated there. After sex determination of the gonads, the testes produce testosterone and anti-Mullerian hormone which mediate male sexual differentiation, and the female developmental pathway ensues in their absence. Here we show that transcripts of the LIM homeobox gene Lhx9 are present in urogenital ridges of mice at embryonic day 9.5; later they localize to the interstitial region as morphological differentiation occurs. In mice lacking Lhx9 function, germ cells migrate normally, but somatic cells of the genital ridge fail to proliferate and a discrete gonad fails to form. In the absence of testosterone and anti-Mullerian hormone, genetically male mice are phenotypically female. The expression of steroidogenic factor 1 (Sf1), a nuclear receptor essential for gonadogenesis, is reduced to minimal levels in the Lhx9-deficient genital ridge, indicating that Lhx9 may lie upstream of Sf1 in a developmental cascade. Unlike mice lacking other genes that mediate early stages of gonadogenesis, Lhx9 mutants do not exhibit additional major developmental defects. Thus, LHX9 mutations may underlie certain forms of isolated gonadal agenesis in humans.


Subject(s)
Genes, Homeobox , Gonads/embryology , Homeodomain Proteins/genetics , Animals , Chromosome Mapping , Female , Gene Deletion , Gonadal Dysgenesis/genetics , Gonads/abnormalities , LIM-Homeodomain Proteins , Male , Mice , Mice, Inbred C57BL , Sex Determination Processes , Sex Differentiation/genetics , Steroidogenic Factor 1 , Testis/embryology , Transcription Factors/genetics , Transcription Factors/physiology
12.
Dev Biol ; 215(2): 264-77, 1999 Nov 15.
Article in English | MEDLINE | ID: mdl-10545236

ABSTRACT

Mouse embryos deficient in fibronectin (FN-null) die at E8.5 with mesodermal defects. Eight integrin heterodimers alpha3beta1, alpha4beta1, alpha5beta1, alpha8beta1, alphavbeta1, alphavbeta3, alphavbeta6, and alphaIIbbeta3 can bind to FN. However, embryos deficient in each of these integrins exhibit less severe defects than do FN-null embryos, raising questions as to which integrin(s) are the key FN receptors for these early FN-dependent processes. alpha5beta1 is believed to be the key receptor and alpha5-null embryos display mesodermal defects similar to, although less severe than, those of FN-null. Here we report that the alpha5-null mutation exhibits a more severe phenotype on a 129Sv (129) than on a C57BL/6 (B6) background, as does the FN-null mutation. While alpha5-null/B6 embryos develop normal headfolds, alpha5-null/129 embryos have headfold defects similar to those of FN-null. The differences between FN-null and alpha5-null embryos, however, cannot be attributed to genetic background. FN-null embryos never form somites, whereas in alpha5-null/129 embryos the somites do condense but fail to epithelialize. Second, we examined double mutants carrying all possible pairwise combinations of null mutations in alpha3, alpha4, and alpha5 integrin genes. There was no evidence for any synergy between paired mutations, suggesting that these integrin genes do not have overlapping functions during early embryonic development. Finally, we examined double-mutant embryos deficient in both alpha5 and alphav integrin genes. These double-mutant embryos have an amniotic defect similar to that of FN-null embryos, but die even earlier with a defect in gastrulation. These studies thus revealed a gradation in the severity of defects in the mutations alpha5(-/-); alphav(-/-) > FN(-/-) (129) > FN(-/-) (B6) > alpha5(-/-) (129) > alpha5(-/-) (B6), and in each step in this series there is a certain degree of phenotypic overlap, suggesting that the defects arising from these mutations may result from disruptions of the same embryonic process.


Subject(s)
Integrins/physiology , Mesoderm/physiology , Receptors, Fibronectin/physiology , Animals , Antigens, CD/physiology , Female , Fibronectins/analysis , Integrin alpha3 , Integrin alpha4 , Integrin alpha5 , Mice , Phenotype , Pregnancy
13.
Development ; 126(17): 3957-68, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10433923

ABSTRACT

Integrins alpha6beta1 and alpha6beta4 are cell surface receptors for laminins. Integrin alpha6-null mice die at birth with severe skin blistering and defects in the cerebral cortex and in the retina. Integrin alpha3beta1 can associate with laminins and other ligands. Integrin alpha3-null mice also die at birth, with kidney and lung defects at late stages of development, and moderate skin blistering. To investigate possible overlapping functions between alpha3 and alpha6 integrins, we analyzed the phenotype of compound alpha3-/-/alpha6-/- mutant embryos. Double homozygous mutant embryos were growth-retarded and displayed several developmental defects not observed in the single mutant animals. First, limb abnormalities characterized by an absence of digit separation and the fusion of preskeletal elements were observed. Further analyses indicated a defect in the apical ectodermal ridge, an essential limb organizing center. In the double mutant, the ridge appeared flattened, and ridge cells did not show a columnar morphology. A strong reduction in ridge cell proliferation and alterations of the basal lamina underlying the ectoderm were observed. These results suggest that alpha3 and alpha6 integrins are required for the organization or compaction of presumptive apical ectodermal ridge cells into a distinct differentiated structure. Additional defects were present: an absence of neural tube closure, bilateral lung hypoplasia, and several abnormalities in the urogenital tract. Finally, an aggravation of brain and eye lamination defects was observed. The presence of novel phenotypes in double mutant embryos demonstrates the synergism between alpha3 and alpha6 integrins and their essential roles in multiple processes during embryogenesis.


Subject(s)
Antigens, CD/physiology , Embryonic and Fetal Development/physiology , Integrins/physiology , Abnormalities, Multiple/embryology , Abnormalities, Multiple/genetics , Animals , Antigens, CD/genetics , Base Sequence , Basement Membrane/embryology , Cell Death , Cell Division , DNA Primers/genetics , Ectoderm/cytology , Ectoderm/metabolism , Embryonic and Fetal Development/genetics , Extremities/embryology , Female , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/physiology , In Situ Hybridization , Integrin alpha3 , Integrin alpha6 , Integrins/genetics , Limb Deformities, Congenital/genetics , Male , Mice , Mice, Knockout , Phenotype , Pregnancy
14.
J Cell Sci ; 112 ( Pt 17): 2925-35, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10444387

ABSTRACT

Epithelial cell morphology and cytoskeletal organization are determined by interactions, with both adjacent cells and the extracellular matrix, which are mediated by integrins and cadherins. Little is known, however, of the relative contributions of integrins and cadherins to maintaining the sub-cortical cytoskeleton characteristic of epithelial cells. Since most studies that utilize integrin-blocking antibodies result in a loss of both cell-cell adhesion and sub-cortical cytoskeletal organization, it has been difficult to distinguish whether integrins and cadherins both mediate cytoskeletal assembly in epithelial cells. Therefore, cells derived from kidney collecting ducts of (alpha)3(beta)1 integrin-deficient mice were used to examine the role of integrins in epithelial cell morphology and cytoskeletal organization. In primary cell culture, (alpha)3(beta)1 integrin-deficient kidney collecting duct cells maintain cadherin-mediated cell-cell adhesions but fail to form the sub-cortical cytoskeleton that is characteristic of epithelial cells, and instead assemble actin stress fibers. Moreover, the cell-cell junctions in mutant cells were irregular, rather than being uniformly oriented perpendicular to the culture substrate. These results demonstrated that integrins have an primary and essential function in establishing and maintaining the sub-cortical cytoskeleton that is characteristic of epithelial cells. To further study the role of (alpha)3(beta)1 integrin in establishing and maintaining cytoskeletal organization in tubular epithelial cells, we derived immortalized cell lines from wild-type and (alpha)3(beta)1 integrin-deficient kidney collecting ducts that duplicated the cytoskeletal and cadherin organization observed in primary cells. E-cadherin and (alpha)- and (beta)-catenin were complexed together in equal amounts in membranes of wild-type and (alpha)3(beta)1 integrin-deficient cells. However, association of the cadherin:catenin complex with (alpha)-actinin was greatly decreased in mutant cells, indicating that integrin-mediated assembly of the sub-cortical cytoskeleton is essential for subsequent association of the cytoskeleton with the cadherin:catenin complex. These results present direct evidence for integrin:cadherin cross-regulation in which cadherin function is dependent on the presence of an integrin.


Subject(s)
Cytoskeletal Proteins/analysis , Cytoskeleton/ultrastructure , Epithelial Cells/ultrastructure , Integrins/physiology , Trans-Activators , Animals , Antigens, CD/genetics , Cadherins/analysis , Cell Line, Transformed , DNA, Complementary/genetics , Desmoplakins , Epithelial Cells/metabolism , Humans , Integrin alpha3 , Integrin alpha3beta1 , Integrins/deficiency , Integrins/genetics , Intercellular Junctions/metabolism , Intercellular Junctions/ultrastructure , Kidney Tubules, Collecting/cytology , Kidney Tubules, Collecting/metabolism , Laminin/biosynthesis , Macromolecular Substances , Mice , Mice, Knockout , Mice, Transgenic , Ureter/cytology , Ureter/metabolism , alpha Catenin , beta Catenin
15.
Dev Genet ; 24(3-4): 252-62, 1999.
Article in English | MEDLINE | ID: mdl-10322633

ABSTRACT

The early development of the metanephric kidney is characterized by the induced differentiation of mesenchymal cells into a stem cell population that undergoes a mesenchymal to epithelial transformation in response to stimuli from the ureteric bud. The Wilms' tumor suppressor gene, Wt1, is required for mesenchymal cells to complete this developmental program. In the absence of WT1, a prospective metanephric mesenchyme appears, but becomes apoptotic, and outgrowth of the ureteric bud from the Wolffian duct does not occur. Therefore, the examination of Wt1 -/- embryos allows the determination of those markers of early metanephric differentiation that do not require the ureteric bud or WT1 for their expression. Here, we demonstrate that several markers, including Pax-2, Six-2, and GDNF, were present as RNAs in the metanephric mesenchyme of Wt1 -/- embryos. These findings demonstrate that the metanephric mesenchyme in mutant embryos has begun to differentiate towards the nephrogenic lineage, and that this early differentiation does not require either WT1 or the presence of the ureteric bud. To determine whether WT1 functions other than to induce expression of factors that stimulate ureteric bud outgrowth, Wt1 -/- metanephric mesenchymes were recombined with wild-type ureteric buds in organ culture, but this failed to rescue tubulogenesis. However, the Wolffian duct from Wt1 -/- embryos was a competent inducer of wild-type metanephric mesenchyme.


Subject(s)
Genes, Wilms Tumor , Kidney/embryology , Ureter/embryology , Animals , Cell Differentiation , Gene Expression Regulation, Developmental , In Situ Hybridization , Kidney/cytology , Kidney/metabolism , Mesoderm/cytology , Mesoderm/metabolism , Mice , Mice, Knockout , Organ Culture Techniques , Ureter/cytology , Ureter/metabolism
16.
Development ; 126(9): 1845-57, 1999 May.
Article in English | MEDLINE | ID: mdl-10101119

ABSTRACT

The Wilms' Tumour gene WT1 has important functions during development. Knock-out mice were shown to have defects in the urogenital system and to die at embryonic day E13.5, probably due to heart failure. Using a lacZ reporter gene inserted into a YAC construct, we demonstrate that WT1 is expressed in the early proepicardium, the epicardium and the subepicardial mesenchymal cells (SEMC). Lack of WT1 leads to severe defects in the epicardial layer and a concomitant absence of SEMCs, which explains the pericardial bleeding and subsequent embryonic death observed in Wt1 null embryos. We further show that a human-derived WT1 YAC construct is able to completely rescue heart defects, but only partially rescues defects in the urogenital system. Analysis of the observed hypoplastic kidneys demonstrate a continuous requirement for WT1 during nephrogenesis, in particular, in the formation of mature glomeruli. Finally, we show that the development of adrenal glands is also severely affected in partially rescued embryos. These data demonstrate a variety of new functions for WT1 and suggest a general requirement for this protein in the formation of organs derived from the intermediate mesoderm.


Subject(s)
Adrenal Glands/embryology , DNA-Binding Proteins/physiology , Gene Expression Regulation, Developmental , Genes, Wilms Tumor , Nephrons/embryology , Pericardium/embryology , Transcription Factors/physiology , Animals , Chromosomes, Artificial, Yeast , DNA-Binding Proteins/genetics , Embryonic and Fetal Development , Fetal Heart/physiology , Genes, Reporter , Genetic Complementation Test , Mesoderm/physiology , Mice , Mice, Knockout , Transcription Factors/genetics , WT1 Proteins , beta-Galactosidase/genetics
17.
Mol Reprod Dev ; 52(4): 366-75, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10092116

ABSTRACT

The Wt1 gene, originally identified as a tumor suppressor gene associated with Wilms' tumors, encodes a zinc finger containing transcription factor expressed during gonadal and kidney development. Although Wt1 appears to be required for gonadal and kidney development, no reproductive defects were observed in outbred females heterozygous for a targeted mutation in Wt1. In contrast, no litters were obtained from Wt1 +/- females on a strain 129/Sv inbred genetic background. Ovaries were smaller in Wt1 +/- 129/Sv mice and produced fewer ova, but transplanted Wt1 +/- ovaries from 129/Sv females were able to support successful pregnancies. The inability of Wt1 +/- 129/Sv females to produce successful implantations after ovulation and fertilization appeared to be due to the failure of one-cell embryos to undergo mitosis, such that they were lost in the oviduct before reaching the uterus. Approximately 50% of Wt1 +/- females generated from a backcross of Wt1 +/- 129/Sv:C57BI/6 F1 hybrids to 129/Sv were fertile, indicating the presence of a Wt1 modifier gene that affects survival of the preimplantation embryo. Neither levels of WT1 protein nor the ratio of WT1 spice forms were significantly altered in Wt1 +/- reproductive organs, suggesting that this modifier effect acts downstream of WT1. Wt1 is therefore among a small subset of genes required for survival of the pre-implantation embryo, and appears to function non-autonomously.


Subject(s)
DNA-Binding Proteins/physiology , Embryonic and Fetal Development/genetics , Fallopian Tubes/physiology , Genes, Wilms Tumor , Ovary/physiology , Transcription Factors/physiology , Animals , Crosses, Genetic , DNA-Binding Proteins/genetics , Female , Fertilization in Vitro , Gene Transfer Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Ovary/abnormalities , Ovary/transplantation , Pregnancy , Progesterone/blood , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/genetics , Uterus/physiology , WT1 Proteins , Zinc Fingers
18.
Neuron ; 22(2): 277-89, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10069334

ABSTRACT

Changes in specific cell-cell recognition and adhesion interactions between neurons and radial glial cells regulate neuronal migration as well as the establishment of distinct layers in the developing cerebral cortex. Here, we show that alpha3beta1 integrin is necessary for neuron-glial recognition during neuronal migration and that alpha(v) integrins provide optimal levels of the basic neuron-glial adhesion needed to maintain neuronal migration on radial glial fibers. A gliophilic-to-neurophilic switch in the adhesive preference of developing cortical neurons occurs following the loss of alpha3beta1 integrin function. Furthermore, the targeted mutation of the alpha3 integrin gene results in abnormal layering of the cerebral cortex. These results suggest that alpha3beta1 and alpha(v) integrins regulate distinct aspects of neuronal migration and neuron-glial interactions during corticogenesis.


Subject(s)
Antigens, CD/physiology , Cerebral Cortex/embryology , Integrins/physiology , Neurons/physiology , Animals , Antigens, CD/genetics , Cell Aggregation/physiology , Cell Movement/physiology , Cerebral Cortex/abnormalities , Cerebral Cortex/cytology , Embryo, Mammalian/cytology , Embryo, Mammalian/physiology , Embryonic and Fetal Development/physiology , Integrin alpha3 , Integrin alpha3beta1 , Integrin alphaV , Integrins/genetics , Mice , Mutation/physiology , Neuroglia/physiology
19.
Development ; 126(8): 1655-64, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10079228

ABSTRACT

Primordial germ cells are the founder cells of the gametes. They are set aside at the initial stages of gastrulation in mammals, become embedded in the hind-gut endoderm, then actively migrate to the sites of gonad formation. The molecular basis of this migration is poorly understood. Here we sought to determine if members of the integrin family of cell surface receptors are required for primordial germ cell migration, as integrins have been implicated in the migration of several other motile cell types. We have established a line of mice which express green fluorescent protein in germline cells that has enabled us to efficiently purify primordial germ cells at different stages by flow cytometry. We have catalogued the spectrum of integrin subunit expression by primordial germ cells during and after migration, using flow cytometry, immunocytochemistry and RT-PCR. Through analysis of integrin beta1(-/-)-->wild-type chimeras, we show that embryonic cells lacking beta1 integrins can enter the germline. However, integrin beta1(-/-) primordial germ cells do not colonize the gonad efficiently. Embryos with targeted deletion of integrin subunit alpha3, alpha6, or alphaV show no major defects in primordial germ cell migration. These results demonstrate a role for beta1-containing integrins in the development of the germline, although an equivalent role for * integrin subunit(s) has yet to be established.


Subject(s)
Cell Movement , Integrin beta1/physiology , Animals , Germ Cells , Gonads , Green Fluorescent Proteins , Integrin beta1/genetics , Luminescent Proteins , Mice , Mice, Knockout , Mice, Transgenic , Reverse Transcriptase Polymerase Chain Reaction
20.
J Neurosci ; 18(23): 9703-15, 1998 Dec 01.
Article in English | MEDLINE | ID: mdl-9822731

ABSTRACT

Embryonic retinal neurons lose the ability to extend neurites on laminin-1 (LN-1) with increasing developmental age yet still do so on other laminin isoforms. However, after treatment of LN-1 with antibodies to "short-arm" regions or removal of the short arms proteolytically, LN-1 supports attachment and extension of neurites even by late embryonic retinal neurons. We have mapped a domain for antibody-mediated "activation" of LN-1 to the N-terminal end of the alpha1 chain. Furthermore, we show that the primary receptors used in the retinal neuron response to "activated" LN-1 are integrins alpha3 beta1 and alpha6 beta1; these are the same receptors used by these neurons for outgrowth on other LN isoforms. Interestingly, alpha3 beta1 is preferentially involved in neurite outgrowth, whereas alpha6beta1 preferentially mediates attachment and spreading. However, in cultures from alpha3 integrin-deficient mice, alpha6 beta1 mediates retinal ganglion cell neurite outgrowth and compensates for the absence of alpha3 beta1. Finally, we show that key features of the retinal neuron response to LN-1 also characterize neurons of the hippocampus, thalamus, and cerebral cortex; these include poor response to untreated LN-1, extensive neurite outgrowth on antibody-activated LN-1 or on fragment E8, and dependence of this response on integrin alpha6 beta1 and at least one other long arm-binding beta1 integrin. These data suggest that regulation of LN-1 function via the process of activation could have important consequences for axonal regeneration. Curiously, the data also imply that the mechanism of laminin activation involves enhanced function at sites that cannot be considered cryptic.


Subject(s)
Laminin/immunology , Neurites/chemistry , Receptors, Laminin/chemistry , Receptors, Laminin/immunology , Animals , Axons/chemistry , Chick Embryo , Cricetinae , Epitope Mapping , Female , Integrin alpha3beta1 , Integrins/chemistry , Integrins/genetics , Integrins/immunology , Laminin/metabolism , Mice , Mutagenesis/physiology , Pregnancy , Protein Structure, Tertiary , Rats , Rats, Sprague-Dawley , Receptors, Collagen , Receptors, Laminin/genetics , Retina/chemistry , Retina/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...