Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 92
Filter
Add more filters








Publication year range
1.
J Appl Physiol (1985) ; 98(2): 690-6, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15465888

ABSTRACT

The study was designed to determine whether beta1-integrin plays a role in mediating the acute skeletal response to mechanical unloading. Transgenic (TG) mice were generated to express a dominant negative form of beta1-integrin under the control of the osteocalcin promoter, which targets expression of the transgene to mature osteoblasts. At 63 days of age, wild-type (WT) and TG mice were subjected to hindlimb unloading by tail suspension for 1 wk. Pair-fed, normally loaded WT and TG mice served as age-matched controls. Bone samples from each mouse were processed for quantitative bone histomorphometry and biomechanical testing. The skeletal phenotype of TG mice was characterized by lower cancellous bone mass in the distal femoral metaphysis (-52%) and lumbar vertebral body (-20%), reduced curvature of the proximal tibia (-20%), and decreased bone strength (-20%) and stiffness (-23%) of the femoral diaphysis with relatively normal indexes of cancellous bone turnover. Hindlimb unloading for only 1 wk induced a 10% decline in tibial curvature and a 30% loss of cancellous bone in the distal femur due to a combination of increased bone resorption and decreased bone formation in both WT and TG mice. However, the strength and stiffness of the femoral diaphysis were unaffected by short-term hindlimb unloading in both genotypes. The observed increase in osteoclast surface was greater in unloaded TG mice (92%) than in unloaded WT mice (52%). Cancellous bone formation rate was decreased in unloaded WT (-29%) and TG (-15%) mice, but, in contrast to osteoclast surface, the genotype by loading interaction was not statistically significant. The results indicate that altered integrin function in mature osteoblasts may enhance the osteoclastic response to mechanical unloading but that it does not have a major effect on the development of cancellous osteopenia in mice during the early stages of hindlimb unloading.


Subject(s)
Bone Diseases, Metabolic/pathology , Bone Diseases, Metabolic/physiopathology , Bone Resorption/metabolism , Bone Resorption/pathology , Bone and Bones/pathology , Bone and Bones/physiopathology , Hindlimb Suspension/adverse effects , Integrin beta1/metabolism , Animals , Bone Diseases, Metabolic/genetics , Bone Resorption/etiology , Elasticity , Female , Hindlimb Suspension/methods , Integrin beta1/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Organ Size , Recombinant Proteins/metabolism
2.
Calcif Tissue Int ; 76(1): 39-49, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15477996

ABSTRACT

Skeletal modeling entails the deposition of large amounts of extracellular matrix (ECM) to form structures tailored to withstand increasing mechanical loads during rapid growth. Specific ECM molecules bind to integrin receptors on the cell surface, thereby triggering a cascade of signaling events that affect critical cell functions. To evaluate the role of integrins during skeletal growth, transgenic mice were engineered to express a function-perturbing fragment of beta1 integrin consisting of the transmembrane domain and cytoplasmic tail under the control of the osteocalcin promoter (TG mice). Thus, transgene expression was targeted to mature cells of the osteoblast lineage, and herein we show that cultured cells resembling osteocytes from 90-day-old TG mice display impaired adhesion to collagen I, a ligand for beta1 integrin. To determine the influence of beta1 integrin on bones that are responsible for providing structural support during periods of rapid growth, we examined the phenotype of the appendicular skeleton in TG mice compared to wild type (WT) mice. According to radiographs, bones from mice of both genotypes between 14 and 90 days of age appeared similar in gross structure and density, although proximal tibiae from 35-90 days old TG mice were less curved than those of WT mice (72-92% TG/WT). Although there were only mild and transient differences in absolute bone mass and strength, once normalized to body mass, the tibial dry mass (79.1% TG/WT females), ash mass (78.5% TG/WT females), and femoral strength in torsion (71.6% TG/WT females) were reduced in TG mice compared to WT mice at 90 days of age. Similar effects of genotype on bone mass and curvature were observed in 1-year-old retired breeders, indicating that these phenotypic differences between TG and WT mice were stable well into adulthood. Effects of genotype on histomorphometric indices of cancellous bone turnover were minimal and evident only transiently during growth, but when present they demonstrated differences in osteoblast rather than osteoclast parameters. Together, these results suggest that integrin signals generated during growth enhance the acquisition of a skeletal mass, structure, and strength to withstand the mechanical loads generated by weight-bearing.


Subject(s)
Bone and Bones/metabolism , Integrin beta1/metabolism , Mice, Transgenic/growth & development , Osteocytes/metabolism , Animals , Biomechanical Phenomena , Bone and Bones/diagnostic imaging , Cell Adhesion/physiology , Cells, Cultured , Collagen Type I , Extracellular Matrix/metabolism , Female , Femur/pathology , Femur/physiopathology , Gene Expression , Integrin beta1/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Organ Culture Techniques , Phenotype , RNA, Messenger/metabolism , Radiography , Tibia/diagnostic imaging , Tibia/pathology
3.
Neuron ; 31(3): 367-79, 2001 Aug 16.
Article in English | MEDLINE | ID: mdl-11516395

ABSTRACT

Mice that lack all beta1-class integrins in neurons and glia die prematurely after birth with severe brain malformations. Cortical hemispheres and cerebellar folia fuse, and cortical laminae are perturbed. These defects result from disorganization of the cortical marginal zone, where beta1-class integrins regulate glial endfeet anchorage, meningeal basement membrane remodeling, and formation of the Cajal-Retzius cell layer. Surprisingly, beta1-class integrins are not essential for neuron-glia interactions and neuronal migration during corticogenesis. The phenotype of the beta1-deficient mice resembles pathological changes observed in human cortical dysplasias, suggesting that defective integrin-mediated signal transduction contributes to the development of some of these diseases.


Subject(s)
Brain/abnormalities , Cerebellar Cortex/abnormalities , Cerebellar Cortex/embryology , Cerebral Cortex/abnormalities , Cerebral Cortex/embryology , Integrin beta1/physiology , Neurons/physiology , Animals , Cell Adhesion Molecules, Neuronal/analysis , Cells, Cultured , Cerebellar Cortex/pathology , Cerebral Cortex/pathology , Disease Models, Animal , Embryonic and Fetal Development , Extracellular Matrix/pathology , Extracellular Matrix/physiology , Extracellular Matrix Proteins/analysis , Integrin beta1/genetics , Intermediate Filament Proteins/genetics , Intermediate Filament Proteins/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Nerve Tissue Proteins/analysis , Nestin , Neuroglia/pathology , Neuroglia/physiology , Neurons/pathology , Neurons/ultrastructure , Reelin Protein , Serine Endopeptidases , Signal Transduction , beta-Galactosidase/genetics
4.
Mol Biol Cell ; 12(8): 2290-307, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11514617

ABSTRACT

Hypertrophic terminally differentiated cardiac myocytes show increased sarcomeric organization and altered gene expression. Previously, we established a role for the nonreceptor tyrosine kinase Src in signaling cardiac myocyte hypertrophy. Here we report evidence that p130Cas (Cas) and focal adhesion kinase (FAK) regulate this process. In neonatal cardiac myocytes, tyrosine phosphorylation of Cas and FAK increased upon endothelin (ET) stimulation. FAK, Cas, and paxillin were localized in sarcomeric Z-lines, suggesting that the Z-line is an important signaling locus in these cells. Cas, alone or in cooperation with Src, modulated basal and ET-stimulated atrial natriuretic peptide (ANP) gene promoter activity, a marker of cardiac hypertrophy. Expression of the C-terminal focal adhesion-targeting domain of FAK interfered with localization of endogenous FAK to Z-lines. Expression of the Cas-binding proline-rich region 1 of FAK hindered association of Cas with FAK and impaired the structural stability of sarcomeres. Collectively, these results suggest that interaction of Cas with FAK, together with their localization to Z-lines, is critical to assembly of sarcomeric units in cardiac myocytes in culture. Moreover, expression of the focal adhesion-targeting and/or the Cas-binding proline-rich regions of FAK inhibited ANP promoter activity and suppressed ET-induced ANP and brain natriuretic peptide gene expression. In summary, assembly of signaling complexes that include the focal adhesion proteins Cas, FAK, and paxillin at Z-lines in the cardiac myocyte may regulate, either directly or indirectly, both cytoskeletal organization and gene expression associated with cardiac myocyte hypertrophy.


Subject(s)
Cardiomegaly/metabolism , Gene Expression Regulation , Myocardium/cytology , Phosphoproteins/metabolism , Protein-Tyrosine Kinases/metabolism , Proteins , Sarcomeres/physiology , Animals , Atrial Natriuretic Factor/genetics , Cardiomegaly/genetics , Cell Fractionation , Cells, Cultured , Crk-Associated Substrate Protein , Culture Media, Serum-Free , Cytoskeletal Proteins/metabolism , Endothelins/metabolism , Endothelins/pharmacology , Focal Adhesion Kinase 1 , Focal Adhesion Protein-Tyrosine Kinases , Genes, Reporter , Immunoblotting , Immunohistochemistry , Microfilament Proteins/metabolism , Myocardium/metabolism , Myosin Heavy Chains/metabolism , Paxillin , Phosphoproteins/genetics , Phosphorylation , Promoter Regions, Genetic , Protein Structure, Tertiary , Protein-Tyrosine Kinases/genetics , Rats , Recombinant Fusion Proteins/metabolism , Retinoblastoma-Like Protein p130 , Sarcomeres/drug effects , Signal Transduction , Tensins , src-Family Kinases/metabolism
5.
Am J Pathol ; 159(1): 93-108, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11438458

ABSTRACT

During human pregnancy specialized placental cells of fetal origin, termed cytotrophoblasts, invade the uterus and its blood vessels. This tumor-like process anchors the conceptus to the mother and diverts the flow of uterine blood to the placenta. Previously, we showed that the expression of molecules with important functional roles, including a number of extracellular matrix integrin receptors, is precisely modulated during cytotrophoblast invasion in situ. Here we exploited this observation to study the role of the focal adhesion kinase (FAK), which transduces signals from the extracellular matrix and recruits additional signaling proteins to focal adhesions. Immunolocalization studies on tissue sections showed that FAK is expressed by cytotrophoblasts in all stages of differentiation. Because extracellular matrix-induced integrin clustering results in FAK (auto)phosphorylation on tyrosine 397 (Y397FAK), we also localized this form of the molecule. Immunolocalization experiments detected Y397FAK in a subset of cytotrophoblasts near the surface of the uterine wall. To assess the functional relevance of this observation, we used an adenovirus strategy to inhibit cytotrophoblast expression of FAK as the cells differentiated along the invasive pathway in vitro. Compared to control cells transduced with a wild-type virus, cytotrophoblasts that expressed antisense FAK exhibited a striking reduction in their ability to invade an extracellular matrix substrate. When cytotrophoblast differentiation was compromised (hypoxia in vitro, preeclampsia in vivo), Y397FAK levels associated with the plasma membrane were strikingly lower, although total FAK levels did not change. Together our results suggest that (auto)phosphorylation of Y397 on FAK is a critical component of the signaling pathway that mediates cytotrophoblast migration/invasion.


Subject(s)
Protein-Tyrosine Kinases/metabolism , Trophoblasts/physiology , Biomarkers , Cell Membrane/metabolism , Cell Movement/drug effects , Cell Movement/physiology , Chorionic Villi/drug effects , Chorionic Villi/physiology , Enzyme Activation , Female , Focal Adhesion Kinase 1 , Focal Adhesion Protein-Tyrosine Kinases , Humans , Hypoxia/enzymology , Hypoxia/physiopathology , Immunologic Techniques , In Vitro Techniques , Oligonucleotides, Antisense/pharmacology , Oxygen/pharmacology , Phosphorylation , Placenta/enzymology , Pre-Eclampsia/enzymology , Pre-Eclampsia/physiopathology , Pregnancy , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/genetics , Trophoblasts/drug effects , Trophoblasts/enzymology , Uterus/cytology , Uterus/physiology
6.
J Cell Biol ; 149(3): 741-54, 2000 May 01.
Article in English | MEDLINE | ID: mdl-10791986

ABSTRACT

Most transformed cells have lost anchorage and serum dependence for growth and survival. Previously, we established that when serum is absent, fibronectin survival signals transduced by focal adhesion kinase (FAK), suppress p53-regulated apoptosis in primary fibroblasts and endothelial cells (Ilic et al. 1998. J. Cell Biol. 143:547-560). The present goals are to identify survival sequences in FAK and signaling molecules downstream of FAK required for anchorage-dependent survival of primary fibroblasts. We report that binding of the SH3 domain of p130Cas to proline-rich region 1 of FAK is required to support survival of fibroblasts on fibronectin when serum is withdrawn. The FAK-p130Cas complex activates c-Jun NH2-terminal kinase (JNK) via a Ras/Rac1/Pak1/MAPK kinase 4 (MKK4) pathway. Activated (phospho-) JNK colocalizes with FAK in focal adhesions of fibroblasts cultured on fibronectin, which supports their survival, but not in fibroblasts cultured on collagen, which does not. Cells often survive in the absence of extracellular matrix if serum factors are provided. In that case, we confirm work of others that survival signals are transduced by FAK, phosphatidylinositol 3'-kinase (PI3-kinase), and Akt/protein kinase B (PKB). However, when serum is absent, PI3-kinase and Akt/PKB are not involved in the fibronectin-FAK-JNK survival pathway documented herein. Thus, survival signals from extracellular matrix and serum are transduced by FAK via two distinct pathways.


Subject(s)
Cell Adhesion , Extracellular Matrix/metabolism , Fibronectins/metabolism , Mitogen-Activated Protein Kinases/metabolism , Protein Serine-Threonine Kinases , Protein-Tyrosine Kinases/metabolism , Proteins , Animals , Cell Survival , Culture Media, Serum-Free , Fibroblasts , Focal Adhesion Protein-Tyrosine Kinases , Immunohistochemistry , JNK Mitogen-Activated Protein Kinases , Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins/metabolism , Protein Binding , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Rabbits , Retinoblastoma-Like Protein p130 , Signal Transduction , Transfection , src Homology Domains
7.
Nat Cell Biol ; 2(5): 249-56, 2000 May.
Article in English | MEDLINE | ID: mdl-10806474

ABSTRACT

Here we show that cells lacking focal adhesion kinase (FAK) are refractory to motility signals from platelet-derived and epidermal growth factors (PDGF and EGF respectively), and that stable re-expression of FAK rescues these defects. FAK associates with activated PDGF- and EGF-receptor (PDGFR and EGFR) signalling complexes, and expression of the band-4.1-like domain at the FAK amino terminus is sufficient to mediate an interaction with activated EGFR. However, efficient EGF-stimulated cell migration also requires FAK to be targeted, by its carboxy-terminal domain, to sites of integrin-receptor clustering. Although the kinase activity of FAK is not needed to promote PDGF- or EGF-stimulated cell motility, kinase-inactive FAK is transphosphorylated at the indispensable Src-kinase-binding site, FAK Y397, after EGF stimulation of cells. Our results establish that FAK is an important receptor-proximal link between growth-factor-receptor and integrin signalling pathways.


Subject(s)
Cell Movement/physiology , Integrins/metabolism , MAP Kinase Signaling System/physiology , Platelet-Derived Growth Factor/pharmacology , Protein-Tyrosine Kinases/metabolism , Cell Movement/drug effects , Cells, Cultured , Epidermal Growth Factor/pharmacology , ErbB Receptors/physiology , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/enzymology , Focal Adhesion Kinase 1 , Focal Adhesion Kinase 2 , Focal Adhesion Protein-Tyrosine Kinases , Humans , MAP Kinase Signaling System/drug effects , Mutagenesis/physiology , Protein Structure, Tertiary , Protein-Tyrosine Kinases/chemistry , Protein-Tyrosine Kinases/genetics , Receptors, Platelet-Derived Growth Factor/physiology , src-Family Kinases/metabolism
8.
Dev Biol ; 220(1): 2-15, 2000 Apr 01.
Article in English | MEDLINE | ID: mdl-10720426

ABSTRACT

To determine the role of integrins in mature osteoblasts in vivo, we expressed in transgenic mice a dominant-negative integrin subunit (beta1-DN) consisting of the beta1 subunit cytoplasmic and transmembrane domains, driven by the osteoblast-specific osteocalcin promoter. Immature osteoblasts isolated from transgenic animals differentiated normally in vitro until the osteocalcin promoter became active; thereafter they detached from the substratum, suggesting that beta1-DN was impairing adhesion in mature osteoblasts. Transgenic animals had reduced bone mass, with increased cortical porosity in long bones and thinner flat bones in the skull. At 35 days, the rate of bone formation was reduced in cortical bone, and the parietal bones were 45% thinner than in wild-type animals. Active osteoblasts were less polar and had larger areas of cytoplasm with intracellular stores of matrix molecules. Osteocyte lacunae appeared normal around the cell body but did not have normal canilicular structures. At 90 days, the parietal bone of transgenic males was of normal width, suggesting that the original defect in matrix deposition had been repaired or compensated for. In contrast, transgenic females still had decreased bone mass in the parietal bone at 90 days. The decreased bone mass in TG females was accompanied by increased staining for osteoclast activity, suggesting that there was a sex-specific defect in mature animals.


Subject(s)
Bone Development/physiology , Integrin beta1/physiology , Osteoblasts/physiology , Animals , Bone Development/genetics , Bone Resorption/genetics , Bone Resorption/pathology , Bone Resorption/physiopathology , Bone and Bones/abnormalities , Cell Adhesion , Extracellular Matrix/pathology , Female , Gene Expression Regulation, Developmental , In Vitro Techniques , Integrin beta1/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Transgenic , Models, Biological , Osteoblasts/pathology , Osteocalcin/genetics , Osteoclasts/pathology , Osteoclasts/physiology , Promoter Regions, Genetic , Signal Transduction
9.
Hum Genet ; 105(5): 480-8, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10598816

ABSTRACT

The autosomal beta1 integrin knockout mouse mutation was selected as a model to experimentally determine preimplantation diagnosis test reliability for autosomal gene deletions and duplications. In experiment 1, which analyzed 198 individually disaggregated single blastomeres, the observed test frequencies matched the mathematically predicted frequencies calculated from the independently derived values of 90% normal allele amplification, 92% mutant allele amplification, 4% alternate allele contamination, and 4% failure to transfer amplifiable target DNA into the PCR reaction mix. This experiment correctly predicted a normal embryonic phenotype in 143 (99.3%) of the 144 phenotypically normal autosomal recessive results. Experiment 2 compared single biopsied blastomere test results to test results on the remaining embryonic cells cultured 1 week until trophoblast outgrowth. Single biopsied blastomere analysis correctly predicted a normal autosomal recessive phenotype in 87 (98%) of the 89 embryos that would have been selected for implantation. Experiment 3 compared the PCR results of two biopsied blastomeres tested independently to the PCR result from the remaining cultured blastomeres to improve test reliability. Given that embryos would have been implanted only when two normal results were obtained, 17 of 17 phenotypically normal embryos would have been implanted from among the 44 embryos tested. These experiment 3 results are consistent with the mathematical prediction that about 99.9% of embryos implanted with two unaffected biopsied blastomere results would have had a phenotypically normal genotype.


Subject(s)
Aneuploidy , Integrin beta1/genetics , Models, Genetic , Mutation , Prenatal Diagnosis/methods , Animals , Base Sequence , Blastomeres/cytology , DNA Primers/genetics , Embryonic Development/genetics , Female , Genetic Diseases, Inborn/diagnosis , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/prevention & control , Genotype , Humans , Male , Mice , Mice, Knockout , Phenotype , Pregnancy
10.
J Cell Sci ; 112 ( Pt 22): 4067-78, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10547366

ABSTRACT

Integrin-mediated adhesion of monocytes and macrophages initiates a signal transduction pathway that leads to actin cytoskeletal reorganization, cell migration and immunologic activation. This signaling pathway is critically dependent on tyrosine kinases. To investigate the role of the Src-family of tyrosine kinases in integrin signal transduction, we have examined the adhesive properties of macrophages isolated from hck-/-fgr-/- double knockout mice which lack two of the three predominant Src-family kinases expressed in myeloid cells. Previous examination of polymorphonuclear leukocytes from these animals indicated that these kinases were critical in initiating the actin cytoskeletal rearrangements that lead to respiratory burst and granule secretion following integrin ligation. Double mutant peritoneal exudate macrophages demonstrated markedly reduced tyrosine phosphorylation responses compared to wild-type cells following plating on fibronectin, collagen or vitronectin-coated surfaces. Tyrosine phosphorylation of several actin-associated proteins (cortactin, paxillin, and tensin), as well as the Syk and Pyk2 tyrosine kinases, were all significantly reduced in double mutant cells. The subcellular localization of focal-adhesion associated proteins was also dramatically altered in mutant macrophages cultured on fibronectin-coated surfaces. In wild-type cells, filamentous actin, paxillin, and talin were concentrated along leading edges of the plasma membrane, suggesting that these proteins contribute to cellular polarization during migration in culture. Double mutant cells failed to show the polarized subcellular localization of these proteins. Likewise, double mutant macrophages failed to form normal filopodia under standard culture conditions. Together, these signaling and cytoskeletal defects may contribute to the reduced motility observed in in vitro assays. These data provide biochemical and morphological evidence that the Src-family kinases Hck and Fgr are required for normal integrin-mediated signal transduction in murine macrophages.


Subject(s)
Cell Movement/physiology , Cytoskeleton/metabolism , Integrins/physiology , Macrophages/metabolism , Protein-Tyrosine Kinases/deficiency , Proto-Oncogene Proteins/deficiency , Actins/metabolism , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Antigens, CD/chemistry , Antigens, CD/immunology , Bone Marrow Cells/cytology , Caseins/pharmacology , Cell Adhesion/drug effects , Cell Adhesion Molecules/metabolism , Cells, Cultured , Cross-Linking Reagents/pharmacology , Enzyme Activation , Enzyme Precursors/drug effects , Enzyme Precursors/metabolism , Extracellular Matrix/metabolism , Extracellular Matrix Proteins/pharmacology , Fibronectins/pharmacology , Focal Adhesion Kinase 2 , Integrin alpha5 , Integrin beta1/chemistry , Integrin beta1/immunology , Integrins/metabolism , Intracellular Signaling Peptides and Proteins , Macrophages/cytology , Macrophages/drug effects , Macrophages, Peritoneal/cytology , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/metabolism , Mice , Mice, Knockout , Phosphorylation/drug effects , Protein-Tyrosine Kinases/drug effects , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-hck , Pseudopodia , Signal Transduction , Syk Kinase , Tyrosine/metabolism , src-Family Kinases
11.
Bone ; 25(1): 95-6, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10423030

ABSTRACT

Previous work with cultured primary cells, from our group and from other laboratories, has shown that signals from extracellular matrix, transduced by integrins, play critical roles in regulating gene expression, tissue-specific differentiation, and survival of primary osteoblasts and fibroblasts. This summary will focus on our most recent work, dealing with the role of cell-extracellular matrix interactions and focal adhesion kinase in regulating cell survival in osteoblasts and fibroblasts, and the role of beta1 integrins in tissue organization and remodeling in bone.


Subject(s)
Bone Remodeling/physiology , Extracellular Matrix/metabolism , Integrins/metabolism , Osteoblasts/physiology , Cell Adhesion Molecules/metabolism , Cell Survival/physiology , Fibroblasts/physiology , Focal Adhesion Kinase 1 , Focal Adhesion Protein-Tyrosine Kinases , Genes, p53 , Humans , Protein-Tyrosine Kinases/metabolism
12.
J Bone Miner Res ; 14(7): 1075-83, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10404007

ABSTRACT

Studies in several cell types indicate that the actions of integrin receptors for extracellular matrix and receptors for growth factors are synergistic in regulating cellular differentiation and function. We studied the roles of the alpha1beta1 and alpha2beta1 integrin collagen receptors in regulating the differentiation of 2T3 osteoblastic cells in response to bone morphogenetic protein (BMP)-2. The immortalized 2T3 cell line was established from the calvaria of mice transgenic for a BMP-2 promoter driving SV40 T-antigen. These cells require exogenous BMP-2, as well as ascorbic acid and beta-glycerolphosphate, for expression of a mature osteoblast phenotype and formation of a mineralized matrix. To determine how integrin receptors for collagen-I affect BMP-2 signaling, function-perturbing anti-rat alpha1 and/or alpha2 integrin subunit, or anti-type I collagen (Col-I), antibodies were added to human recombinant (hr)BMP-2-treated 2T3 cultures at confluence (C0) or at 4 or 8 days postconfluence (C4, C8). After 4 days of exposure to the antibodies, cultures were assayed for alkaline phosphatase (ALP) mRNA levels and enzyme activity and for cAMP production in response to parathyroid hormone. Addition of anti-collagen-I or both anti-integrin-alpha1 and -alpha2 antibodies to C0 cultures blocked expression of these early osteoblast markers by more than 90%, and also blocked mineralization (0.5-1.8% control) of these cells. In all cases, adding anti-alpha1 or anti-alpha2 antibodies separately produced partial effects, while their combined effect approached that of anti-collagen-I. When antibodies were added to more differentiated 2T3 cells, the inhibitory effects decreased. 2T3 cells carrying constitutively active BMP receptor (caBMPR-IB) showed elevated ALP activity without hrBMP-2; this constitutive activity was also suppressed by alpha1 and alpha2 integrin antibodies and by anti-Col-I antibody. Together, our data suggest that a signal(s) from collagen integrin receptors regulates the response to BMP downstream of BMPR-IB and upstream of the regulation of ALP mRNA and other early markers of osteoblast differentiation.


Subject(s)
Bone Morphogenetic Proteins/pharmacology , Integrins/physiology , Osteoblasts/cytology , Transforming Growth Factor beta , Alkaline Phosphatase/metabolism , Animals , Antibodies, Blocking/pharmacology , Blotting, Northern , Bone Morphogenetic Protein 2 , Bone Morphogenetic Protein Receptors, Type I , Bone Morphogenetic Proteins/antagonists & inhibitors , Calcification, Physiologic/drug effects , Cell Differentiation/drug effects , Cell Line , Extracellular Matrix/metabolism , Humans , Integrin alpha1beta1 , Integrins/metabolism , Mice , Osteoblasts/drug effects , Osteoblasts/metabolism , Parathyroid Hormone/pharmacology , Protein Serine-Threonine Kinases/genetics , RNA, Messenger/metabolism , Rats , Receptors, Collagen , Receptors, Growth Factor/genetics , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/pharmacology , Transfection
13.
J Biol Chem ; 273(52): 35185-93, 1998 Dec 25.
Article in English | MEDLINE | ID: mdl-9857056

ABSTRACT

Activation of the atrial natriuretic peptide (ANP) gene is regarded as one of the earliest and most reliable markers of hypertrophy in the ventricular cardiac myocyte. We have examined the role of the nonreceptor tyrosine kinases in the signaling mechanism(s) leading to hypertrophy using human ANP gene promoter activity as a marker. Endothelin (ET), a well known hypertrophic agonist, increased activity of c-Src, c-Yes, and Fyn within minutes and promoted a selective redistribution of each of these kinases within the cell. Overexpression of c-Src effected a significant increase in activity of a cotransfected human ANP promoter-driven chloramphenicol acetyl transferase reporter, while expression of either c-Yes or Fyn was considerably less effective in this regard. ET-dependent stimulation of the human ANP gene promoter was partially inhibited by co-transfection with dominant negative Ras or dominant negative Src or Csk or by treatment with the potent Src family-selective tyrosine kinase inhibitor PP1, suggesting that the Src family kinases are involved in signaling ET-dependent activation of this promoter. Both ET- and Src-dependent activation of the ANP promoter required the presence of a CArG motif in a serum response element-like structure between -422 and -413 but did not appear to require assembly of a ternary complex for full activity. These findings support a role for Src in the activation of ANP gene expression and suggest that this kinase may contribute in an important way to the signaling mechanisms that activate hypertrophy in the cardiac myocyte.


Subject(s)
Atrial Natriuretic Factor/biosynthesis , Cardiomegaly/metabolism , Endothelins/metabolism , Proto-Oncogene Proteins pp60(c-src)/metabolism , Receptors, Endothelin/metabolism , Animals , Animals, Newborn , Atrial Natriuretic Factor/genetics , Biological Transport , CSK Tyrosine-Protein Kinase , Cell Compartmentation , Cells, Cultured , Humans , Myocardium/cytology , Phosphoprotein Phosphatases/metabolism , Promoter Regions, Genetic , Protein-Tyrosine Kinases , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-fyn , Proto-Oncogene Proteins c-yes , Rats , Signal Transduction , ras Proteins/genetics , ras Proteins/metabolism , src-Family Kinases/metabolism
14.
Curr Opin Cell Biol ; 10(5): 660-6, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9818178

ABSTRACT

During early development, a subset of fetal (placental) cytotrophoblasts exhibits tumor-like behavior and invades the uterus. To access a supply of maternal blood, they invade arterioles and form heterotypic interactions with, and replace, resident maternal endothelium, creating a hybrid uterine vasculature. Recently, it has become clear that invading cytotrophoblasts transform their adhesion receptor phenotype to resemble the endothelial cells they replace. Furthermore, they express vasculogenic factors and receptors. Is this a form of vasculogenesis?


Subject(s)
Cell Adhesion Molecules/biosynthesis , Endothelium, Vascular/cytology , Neovascularization, Physiologic , Trophoblasts/physiology , Uterus/blood supply , Cell Differentiation , Epithelial Cells/cytology , Female , Humans , Pregnancy
15.
J Cell Biol ; 143(2): 547-60, 1998 Oct 19.
Article in English | MEDLINE | ID: mdl-9786962

ABSTRACT

In many malignant cells, both the anchorage requirement for survival and the function of the p53 tumor suppressor gene are subverted. These effects are consistent with the hypothesis that survival signals from extracellular matrix (ECM) suppress a p53-regulated cell death pathway. We report that survival signals from fibronectin are transduced by the focal adhesion kinase (FAK). If FAK or the correct ECM is absent, cells enter apoptosis through a p53-dependent pathway activated by protein kinase C lambda/iota and cytosolic phospholipase A2. This pathway is suppressible by dominant-negative p53 and Bcl2 but not CrmA. Upon inactivation of p53, cells survive even if they lack matrix signals or FAK. This is the first report that p53 monitors survival signals from ECM/FAK in anchorage- dependent cells.


Subject(s)
Apoptosis/physiology , Cell Adhesion Molecules/metabolism , Extracellular Matrix/enzymology , Protein-Tyrosine Kinases/metabolism , Signal Transduction/physiology , Tumor Suppressor Protein p53/metabolism , Animals , Annexins/metabolism , Blood Proteins/pharmacology , Caspases/chemistry , Caspases/metabolism , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Culture Media, Serum-Free/pharmacology , Endothelium/cytology , Extracellular Matrix/chemistry , Fibroblasts/chemistry , Fibroblasts/cytology , Fibroblasts/enzymology , Fibronectins/metabolism , Focal Adhesion Protein-Tyrosine Kinases , Green Fluorescent Proteins , In Situ Nick-End Labeling , Indicators and Reagents , Isoenzymes/metabolism , Luminescent Proteins , Phospholipases A/metabolism , Phospholipases A2 , Protein Kinase C/metabolism , Protein Structure, Tertiary , Rabbits , Signal Transduction/drug effects , Synovial Membrane/cytology
16.
EMBO J ; 17(20): 5933-47, 1998 Oct 15.
Article in English | MEDLINE | ID: mdl-9774338

ABSTRACT

The focal adhesion kinase (FAK) protein-tyrosine kinase (PTK) links transmembrane integrin receptors to intracellular signaling pathways. We show that expression of the FAK-related PTK, Pyk2, is elevated in fibroblasts isolated from murine fak-/- embryos (FAK-) compared with cells from fak+/+ embryos (FAK+). Pyk2 was localized to perinuclear regions in both FAK+ and FAK- cells. Pyk2 tyrosine phosphorylation was enhanced by fibronectin (FN) stimulation of FAK- but not FAK+ cells. Increased Pyk2 tyrosine phosphorylation paralleled the time-course of Grb2 binding to Shc and activation of ERK2 in FAK- cells. Pyk2 in vitro autophosphorylation activity was not enhanced by FN plating of FAK- cells. However, Pyk2 associated with active Src-family PTKs after FN but not poly-L-lysine replating of the FAK- cells. Overexpression of both wild-type (WT) and kinase-inactive (Ala457), but not the autophosphorylation site mutant (Phe402) Pyk2, enhanced endogenous FN-stimulated c-Src in vitro kinase activity in FAK- cells, but only WT Pyk2 overexpression enhanced FN-stimulated activation of co-transfected ERK2. Interestingly, Pyk2 overexpression only weakly augmented FAK- cell migration to FN whereas transient FAK expression promoted FAK- cell migration to FN efficiently compared with FAK+ cells. Significantly, repression of endogenous Src-family PTK activity by p50(csk) overexpression inhibited FN-stimulated cell spreading, Pyk2 tyrosine phosphorylation, Grb2 binding to Shc, and ERK2 activation in the FAK- but not in FAK+ cells. These studies show that Pyk2 and Src-family PTKs combine to promote FN-stimulated signaling events to ERK2 in the absence of FAK, but that these signaling events are not sufficient to overcome the FAK- cell migration defects.


Subject(s)
Cell Adhesion Molecules/analysis , Cell Adhesion Molecules/physiology , Cell Movement/physiology , Fibronectins/metabolism , Protein-Tyrosine Kinases/analysis , Protein-Tyrosine Kinases/deficiency , Protein-Tyrosine Kinases/physiology , Signal Transduction/physiology , src-Family Kinases/physiology , Amino Acid Sequence , Animals , Calcium-Calmodulin-Dependent Protein Kinases/physiology , Cells, Cultured , Fibroblasts , Focal Adhesion Kinase 1 , Focal Adhesion Kinase 2 , Focal Adhesion Protein-Tyrosine Kinases , Humans , Mice , Mitogen-Activated Protein Kinase 1 , Molecular Sequence Data , Protein-Tyrosine Kinases/biosynthesis , Rabbits , Vinculin/analysis
17.
J Reprod Immunol ; 39(1-2): 197-213, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9786462

ABSTRACT

This review article focuses on the unique process by which the human placenta normally forms and how changes in this process can lead to serious pregnancy complications such as pre-eclampsia. One way to compare normal and pathologic pregnancies is to examine biopsy specimens of the placenta and placental bed for disease-associated morphological changes in cellular architecture. Our recent work has verified the decades-old observation that pre-eclampsia is associated with abnormally shallow placentation. We also discuss how these morphological observations prompted us to use a combination of in vitro modeling and in situ immunolocalization techniques to gain insights into the molecular bases of normal placentation and how these mechanisms go awry in pre-eclampsia.


Subject(s)
Endothelium, Vascular/cytology , Oxygen/pharmacology , Pre-Eclampsia/pathology , Trophoblasts/physiology , Cadherins/physiology , Cell Differentiation/drug effects , Female , Humans , Integrins/physiology , Placenta/pathology , Pregnancy , Vascular Cell Adhesion Molecule-1/physiology
18.
J Cell Sci ; 111 ( Pt 10): 1385-93, 1998 May.
Article in English | MEDLINE | ID: mdl-9570756

ABSTRACT

The skeletal extracellular matrix produced by osteoblasts contains the glycoprotein fibronectin, which regulates the adhesion, differentiation and function of various adherent cells. Interactions with fibronectin are required for osteoblast differentiation in vitro, since fibronectin antagonists added to cultures of immature fetal calvarial osteoblasts inhibit their progressive differentiation. To determine if fibronectin plays a unique role in fully differentiated osteoblasts, cultures that had already formed mineralized nodules in vitro were treated with fibronectin antagonists. Fibronectin antibodies caused >95% of the cells in the mature cultures to display characteristic features of apoptosis (nuclear condensation, apoptotic body formation, DNA laddering) within 24 hours. Cells appeared to acquire sensitivity to fibronectin antibody-induced apoptosis as a consequence of differentiation, since antibodies failed to kill immature cells and the first cells killed were those associated with mature nodules. Intact plasma fibronectin, as well as fragments corresponding to the amino-terminal, cell-binding, and carboxy-terminal domains of fibronectin, independently induced apoptosis of mature (day-13), but not immature (day-4), osteoblasts. Finally, transforming growth factor-beta1 partially protected cells from the apoptotic effects of fibronectin antagonists. Thus, in the course of maturation cultured osteoblasts switch from depending on fibronectin for differentiation to depending on fibronectin for survival. These data suggest that fibronectin, together with transforming growth factor-beta1, may affect bone formation, in part by regulating the survival of osteoblasts.


Subject(s)
Apoptosis/physiology , Fibronectins/metabolism , Osteoblasts/cytology , Animals , Antibodies/isolation & purification , Antibodies/pharmacology , Apoptosis/drug effects , Cell Differentiation/physiology , Cells, Cultured , Cellular Senescence/physiology , Fibronectins/immunology , Immunoglobulin G/isolation & purification , Immunoglobulin G/pharmacology , Microscopy, Electron , Osteoblasts/chemistry , Osteoblasts/ultrastructure , Rats , Skull/cytology , Staphylococcal Protein A , Transforming Growth Factor beta/pharmacology
19.
J Cell Sci ; 110 ( Pt 18): 2187-96, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9378768

ABSTRACT

We previously showed that anti-fibronectin antibodies or soluble fibronectin fragments containing the central cell-binding domain inhibit formation of mineralized nodules by fetal calvarial osteoblasts in vitro. These findings suggest a critical role for fibronectin in osteoblast differentiation and morphogenesis. In this study we tested the hypothesis that fibronectin's effects on osteogenesis are mediated via direct interactions with integrin receptors for fibronectin on osteoblasts. Immunocytochemical analysis identified the integrin fibronectin receptor alpha5ss1 in fetal rat calvarial tissue and in cultured osteoblasts at all stages of differentiation. Three other integrins, alpha3ss1, alpha8ss1 and alphavss3, which can bind fibronectin, as well as other matrix components, were also identified in tissue and at all stages of cell culture. Immunoprecipitation data showed that alpha5ss1 levels are constant throughout osteoblast differentiation whereas levels of alpha3ss1 and alpha8ss1 decline in mature mineralized cultures. To determine whether integrin fibronectin receptors are required for osteoblast formation of mineralized nodules, we examined the extent of nodule formation in the presence and absence of function-perturbing anti-integrin antibodies. The antibodies were present continuously in cultures beginning at confluence (day 3), and nodule formation was measured at days 10 and 20. An anti-alpha5 integrin subunit antibody reduced nodule formation to less than 5% of control values at both time points. Inhibition of nodule formation was reversible and did not affect cell attachment and viability. Function-perturbing antibodies against alpha3ss1 and alpha8ss1 also reduced nodule formation, to less than 20% of control values. In contrast, function-perturbing antibodies to alphavss3 and alphavss5 did not affect nodule formation, indicating that the inhibitions noted were indeed specific. To determine the effect of antibody treatment on gene expression, steady-state mRNA expression was examined and found to be suppressed for osteoblast markers alkaline phosphatase and osteocalcin. Together, these results indicate that direct osteoblast interactions with the extracellular matrix are mediated by a select group of integrin receptors that includes alpha5ss1, alpha3ss1 and alpha8ss1. We further conclude that the specific alpha5ss1 fibronectin receptor mediates critical interactions between osteoblasts and fibronectin required for both bone morphogenesis and osteoblast differentiation.


Subject(s)
Fibronectins/metabolism , Osteoblasts/chemistry , Osteoblasts/cytology , Receptors, Fibronectin/metabolism , Alkaline Phosphatase/genetics , Animals , Antibodies/pharmacology , Binding, Competitive/immunology , Calcification, Physiologic/physiology , Cell Differentiation/physiology , Extracellular Matrix/chemistry , Extracellular Matrix/metabolism , Fetus/cytology , Gene Expression Regulation, Enzymologic/physiology , Integrin alpha3beta1 , Integrins/immunology , Integrins/metabolism , Morphogenesis/physiology , Osteoblasts/enzymology , Osteocalcin/genetics , RNA, Messenger/analysis , Rats , Receptors, Fibronectin/immunology , Receptors, Laminin/immunology , Receptors, Laminin/metabolism , Receptors, Vitronectin/immunology , Receptors, Vitronectin/metabolism , Skull/cytology , Time Factors
20.
J Clin Invest ; 99(9): 2139-51, 1997 May 01.
Article in English | MEDLINE | ID: mdl-9151786

ABSTRACT

Establishment of the human placenta requires that fetal cytotrophoblast stem cells in anchoring chorionic villi become invasive. These cytotrophoblasts aggregate into cell columns and invade both the uterine interstitium and vasculature, anchoring the fetus to the mother and establishing blood flow to the placenta. Cytotrophoblasts colonizing spiral arterioles replace maternal endothelium as far as the first third of the myometrium. We show here that differentiating cytotrophoblasts transform their adhesion receptor phenotype so as to resemble the endothelial cells they replace. Cytotrophoblasts in cell columns show reduced E-cadherin staining and express VE-(endothelial) cadherin, platelet-endothelial adhesion molecule-1, vascular endothelial adhesion molecule-1, and alpha-4-integrins. Cytotrophoblasts in the uterine interstitium and maternal vasculature continue to express these receptors, and, like endothelial cells during angiogenesis, also stain for alphaVbeta3. In functional studies, alphaVbeta3 and VE-cadherin enhance, while E-cadherin restrains, cytotrophoblast invasiveness. Cytotrophoblasts expressing alpha4 integrins bound immobilized VCAM-1 in vitro, suggesting that this receptor-pair could mediate cytotrophoblast-endothelium or cytotrophoblast-cytotrophoblast interactions in vivo, during endovascular invasion. In the pregnancy disorder preeclampsia, in which endovascular invasion remains superficial, cytotrophoblasts fail to express most of these endothelial markers (Zhou et al., 1997. J. Clin. Invest. 99:2152-2164.), suggesting that this adhesion phenotype switch is required for successful endovascular invasion and normal placentation.


Subject(s)
Chorion/blood supply , Chorion/metabolism , Neovascularization, Physiologic , Trophoblasts/cytology , Antigens, CD , Cadherins/immunology , Cadherins/metabolism , Cell Adhesion , Cell Differentiation , Cells, Cultured , Chorion/cytology , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Female , Humans , Immunohistochemistry , Integrins/immunology , Integrins/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/immunology , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Pregnancy , Stem Cells/physiology , Uterus/blood supply , Uterus/metabolism , Vascular Cell Adhesion Molecule-1/immunology , Vascular Cell Adhesion Molecule-1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL