Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Mech Ageing Dev ; 122(11): 1203-20, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11389933

ABSTRACT

Deficits in the motility of fibroblasts contribute to age-related impairment of wound healing. We analyzed 'young' fibroblasts from four healthy donors 22-30 years old and 'aged' fibroblasts from six healthy donors 81-92 years old for migratory ability on type I collagen, secretion of matrix metalloproteases (MMPs), attachment to matrices and, expression and function of integrin alpha2beta1. Cells from each donor were analyzed separately in each experiment. Whereas migration of young fibroblasts was uniformly robust, three aged lines migrated well and three migrated poorly. Synthesis of MMP1 and TIMP1, but not MMP2 or MMP9, was increased in the aged fibroblasts relative to the young fibroblast lines irrespective of their motility. All lines of young and aged fibroblasts attached to plastic or collagen with similar efficiency. Although young and aged fibroblasts expressed comparable levels of the alpha2 integrin; the lines of aged fibroblasts that were poor migrators exhibited a significant reduction in alpha2beta1 function relative to fibroblasts with normal migratory capacities. Moreover, the lines of aged fibroblasts that exhibited poor migration demonstrated a disordered actin cytoskeleton and a reduced ability to contract collagen gels. In conclusion, aged fibroblasts, unlike young fibroblasts, displayed variable migratory capacities. Deficient migration by specific lines of aged fibroblasts was not related to the capacity to attach, express alpha2 integrin, or secrete MMPs and TIMP1, but was characterized by disorganized cytoskeletal actin and reduced alpha2beta1 function.


Subject(s)
Actins/metabolism , Cell Movement/physiology , Cellular Senescence/physiology , Integrins/biosynthesis , Adult , Aged , Aged, 80 and over , Aging , Cells, Cultured , Collagen/metabolism , Cytoskeleton/metabolism , Cytoskeleton/physiology , Fibroblasts/cytology , Fibroblasts/metabolism , Fibroblasts/physiology , Gels , Humans , Integrins/physiology , Male , Matrix Metalloproteinases/biosynthesis , Receptors, Collagen , Skin/cytology , Skin Aging/physiology , Tissue Donors , Tissue Inhibitor of Metalloproteinase-1/biosynthesis
3.
J Cell Biochem ; 74(2): 152-67, 1999 Aug 01.
Article in English | MEDLINE | ID: mdl-10404386

ABSTRACT

Secreted protein acidic and rich in cysteine (SPARC) is a matricellular protein that inhibits cellular adhesion and proliferation. In this study, we report the detection of SPARC in the interphase nuclei of embryonic chicken cells in vivo. Differential partitioning of SPARC was also noted in the cytoplasm of these cells during discrete stages of M-phase: cells in metaphase and anaphase exhibited strong cytoplasmic immunoreactivity, whereas cells in telophase were devoid of labeling. Immunocytochemical analysis of embryonic chicken cells in vitro likewise showed the presence of SPARC in the nucleus. Furthermore, elution of soluble proteins and DNA from these cells indicated that SPARC might be a component of the nuclear matrix. We subsequently examined cultured bovine aortic endothelial cells, which initially appeared to express SPARC only in the cytoplasm. However, after elution of soluble proteins and chromatin, we also detected SPARC in the nuclear matrix of these cells. Embryonic chicken cells incubated with recombinant SPARC were seen to take up the protein and to translocate it to the nucleus progressively over a period of 17 h. These observations provide new information about SPARC, generally recognized as a secreted glycoprotein that mediates interactions between cells and components of the extracellular matrix. The evidence presented in this study indicates that SPARC might subserve analogous functions in the nuclear matrix.


Subject(s)
Cell Cycle , Nuclear Matrix/metabolism , Osteonectin/metabolism , Animals , Biological Transport , Blotting, Western , Cattle , Cells, Cultured , Chick Embryo , Immunohistochemistry , Recombinant Proteins/metabolism
4.
Microvasc Res ; 57(2): 118-33, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10049660

ABSTRACT

Interactions between migratory endothelial cells (ECs) and surrounding extracellular matrix (ECM) are of central importance to vascular growth. Here, we present a new model of EC migration and morphogenesis within three-dimensional ECM termed "radial invasion of matrix by aggregated cells" (RIMAC). In the RIMAC model, single aggregates of defined numbers of bovine aortic ECs were embedded within small, lenticular gels of type I collagen supported by annuli of nylon mesh. Culture of the gels in nutrient media resulted in quantifiable, reproducible, radial migration of ECs into the collagen. The angiogenic proteins basic fibroblast growth factor (bFGF) and vascular endothelial growth factor (VEGF) each stimulated migration of ECs in a concentration-dependent manner. In combination, bFGF and VEGF stimulated migration synergistically. In contrast, transforming growth factor-beta1 inhibited migration of ECs. Low concentrations (0.1-0.5 ng/ml) of VEGF induced ECs to form multicellular sprouts, some of which possessed lumen-like spaces. Mitomycin C, an inhibitor of cell proliferation, did not affect the migration of ECs into collagen induced by 0.5 ng/ml VEGF but moderately inhibited migration induced by 5 ng/ml VEGF. Increasing the density (concentration) of the collagen gel inhibited the migration of single ECs and increased the branching and anastomosis of multicellular sprouts. We conclude that the RIMAC model is a highly efficacious assay for the screening of potentially angiogenic and angiostatic compounds and, moreover, is advantageous for mechanistic studies of vascular morphogenesis.


Subject(s)
Endothelium, Vascular/cytology , Models, Cardiovascular , Neovascularization, Physiologic , Animals , Cattle , Cell Movement/drug effects , Clone Cells , Collagen , Endothelial Growth Factors/pharmacology , Endothelium, Vascular/drug effects , Endothelium, Vascular/growth & development , Extracellular Matrix/physiology , Fibroblast Growth Factor 2/pharmacology , Gels , Lymphokines/pharmacology , Mitomycin/pharmacology , Neovascularization, Physiologic/drug effects , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
5.
J Histochem Cytochem ; 46(9): 1043-49, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9705970

ABSTRACT

In adult mammals, growth of new vasculature from extant blood vessels (angiogenesis) is rare in the absence of pathology. However, nonpathogenic angiogenesis occurs in the cycling ovary when the avascular postovulatory follicle transforms into a highly vascularized corpus luteum (CL). To improve our understanding of molecular mechanisms that regulate nonpathogenic vascular growth, we characterized the expression of two secreted matricellular proteins associated with angiogenesis, SPARC and thrombospondin (TSP), in postovulatory preluteal follicles and CL of hormone-primed immature rats. By indirect immunofluorescence with specific antibodies, we found SPARC in the cytoplasOFFf granulosa cells and thecal cells of preluteal follicles, in connective tissue cells of the ovarian interstitium, and in the oocyte nucleus. Administration of a luteinizing stimulus (chorionic gonadotropin) increased the expression of SPARC in granulosa cells. TSP was prominent in the basement membranes of growing follicles. Many cells in the early vascularizing CL expressed both SPARC and TSP. Neovascularization of CL was accompanied by expression of SPARC in nascent vessels and concentration of TSP in central avascular areas. In mature CL, steroidogenic luteal cells expressed both SPARC and TSP. Luteal cells of regressing CL retained SPARC to a variable degree but did not express TSP. The observed changes in expression of SPARC and TSP during development of the CL support distinct roles for these matricellular proteins in nonpathological morphogenesis and angiogenesis.


Subject(s)
Corpus Luteum/metabolism , Neovascularization, Physiologic , Osteonectin/metabolism , Ovarian Follicle/metabolism , Thrombospondins/metabolism , Animals , Corpus Luteum/blood supply , Corpus Luteum/growth & development , Female , Fluorescent Antibody Technique, Indirect , Ovarian Follicle/blood supply , Ovarian Follicle/growth & development , Rats
6.
Microvasc Res ; 55(3): 260-70, 1998 May.
Article in English | MEDLINE | ID: mdl-9657926

ABSTRACT

Aging is accompanied by impaired angiogenesis and deficient expression of several angiogenic growth factors. To test the hypothesis that replacement of these factors would improve angiogenesis in aged animals, we cultured microvessels derived from the epididymal fat pad of aged and young mice ("aged" and "young" microvessels) in three-dimensional collagen gels for 2 weeks and measured their sprouting (formation of branch points) in response to fetal bovine serum (FBS), endothelial cell growth supplement (ECGS), and the specific growth factors transforming growth factor-beta1 (TGF-beta1), vascular endothelial growth factor (VEGF), insulin-like growth factor-1 (IGF-1), and basic fibroblast growth factor (bFGF). In the presence of culture medium with 1% FBS (Minimal medium), sprouting of aged microvessels was significantly less than sprouting of young microvessels. The addition of high levels of FBS and ECGS to Minimal medium enhanced the sprouting of microvessels from aged mice to a greater degree than that of young mice, such that the difference between the two age groups was no longer significant. Formation of branch points by aged microvessels was also significantly increased by Minimal medium supplemented with TGF-beta1, bFGF, IGF-1, or VEGF (listed in order of highest to lowest stimulation). Sprouts generated in the presence of VEGF possessed a particularly high percentage of endothelial cells. Mitomycin C did not diminish the degree of sprouting induced by TGF-beta1, VEGF, or IGF-1, a result indicating that early stages of angiogenesis, including formation of branch points, do not require cell division. From our findings in vitro, we propose that age-related deficiencies in angiogenesis in vivo are likely to be due, in part, to a decrease in angiogenic growth factors in the extracellular milieu.


Subject(s)
Adipose Tissue/blood supply , Aging/physiology , Endothelium, Vascular/physiology , Growth Substances/pharmacology , Aging/drug effects , Animals , Cell Division/drug effects , Cell Movement/drug effects , Collagen/pharmacology , Crosses, Genetic , Culture Techniques , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Gels , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Inbred DBA , Microcirculation/cytology , Microcirculation/drug effects , Microcirculation/physiology
7.
Acta Biotheor ; 44(3-4): 271-82, 1996 Nov.
Article in English | MEDLINE | ID: mdl-8953213

ABSTRACT

Endothelial cells, when cultured on gelled basement membrane matrix exert forces of tension through which they deform the matrix and at the same time they aggregate into clusters. The cells eventually form a network of cord-like structures connecting cell aggregates. In this network, almost all of the matrix has been pulled underneath the cell cords and cell clusters. This phenomenon has been proposed as a possible model for the growth and development of planar vascular systems in vitro. Our hypothesis is that the matrix is reorganized and the cellular networks form as a result of traction forces exerted by the cells on the matrix and the latter's elasticity. We construct and analyze a mathematical model based on this hypothesis and examine conditions necessary for the formation of the pattern. We show cell migration is not necessary for pattern formation and that isotropic, strain-stimulated traction is sufficient to form the observed patterns.


Subject(s)
Blood Vessels/cytology , Cell Aggregation/physiology , Cell Differentiation/physiology , Endothelium, Vascular/cytology , Models, Cardiovascular , Models, Theoretical , Animals , Biomechanical Phenomena , Cell Membrane/physiology , Collagen , Drug Combinations , Extracellular Matrix/physiology , Humans , In Vitro Techniques , Laminin , Proteoglycans
8.
Invest Ophthalmol Vis Sci ; 37(11): 2185-94, 1996 Oct.
Article in English | MEDLINE | ID: mdl-8843905

ABSTRACT

PURPOSE: To develop methods for the culture of microvascular endothelial cells (EC) from Macaca monkey retina and to investigate their propagation and survival in vitro. METHODS: Endothelial cells from capillary fragments were cultured on fibronectin-coated dishes in QB-58 serum-free medium containing 20 microliters/ml bovine retinal extract, 90 micrograms/ml heparin, 10% fetal bovine serum, and 10% monkey serum. Non-EC were removed manually. Endothelial cell-specific properties were assessed by endocytosis of acetylated low-density lipoprotein (ac-LDL) and by immunocytochemical staining. The response to growth factors was assayed by 3H-thymidine incorporation. The synthesis of matrix macromolecules was studied by metabolic labeling with 3H-proline and identification by sodium dodecyl sulfate-polyacrylamide gel electrophoresis-immunoblotting. RESULTS: Under these culture conditions, migrating cells emerged from capillary fragments after 1 to 2 days and formed large colonies by 1 week. Cells exhibited a mean doubling time of 44.5 hours during the first 3 to 5 days of culture and 23 hours at 6 to 8 days in culture, and they formed a confluent monolayer by 12 to 14 days. These cells demonstrated uptake of ac-LDL, expressed von Willebrand factor and the cell adhesion protein CD31, and did not contain smooth muscle alpha-actin. Before purification, 92% of the cells in primary cultures were identified as EC. The EC could be maintained in vitro for more than 1 month without the addition of growth factors; however, basic fibroblast growth factor and vascular endothelial growth factor each stimulated cell replication. Secreted extracellular proteins included fibronectin, collagen types I and IV, laminin, and SPARC (secreted protein, acidic, and rich in cysteine). CONCLUSIONS: This study is the first description of the culture and propagation of purified retinal EC from Macaca monkey, a widely accepted model for the human retina. These cultures will be highly relevant to studies of abnormal vascular disease in the human eye.


Subject(s)
Capillaries/cytology , Endothelium, Vascular/cytology , Endothelium, Vascular/physiology , Retinal Vessels/cytology , Acetylation , Animals , Blotting, Western , Capillaries/physiology , Cell Separation/methods , Cell Survival , Cells, Cultured , DNA/biosynthesis , Endocytosis/physiology , Extracellular Matrix Proteins/metabolism , Growth Substances/pharmacology , Immunohistochemistry , Lipoproteins, LDL/metabolism , Macaca fascicularis , Platelet Endothelial Cell Adhesion Molecule-1/biosynthesis , Retinal Vessels/physiology , Thymidine/metabolism , von Willebrand Factor/biosynthesis
9.
Dev Dyn ; 207(2): 171-83, 1996 Oct.
Article in English | MEDLINE | ID: mdl-8906420

ABSTRACT

The Mov-13 strain of mice was created by the insertion of the murine Moloney leukemia virus into the first intron of the alpha 1 (I) collagen gene. Consequently, Mov-13 embryos do not transcribe alpha 1 (I) collagen mRNA and lack type I collagen protein in the extracellular matrix (ECM). Homozygotes die within 12-14 days of embryonic development, in part from the rupture of large blood vessels, and also exhibit deficiencies in hematopoesis and assembly of the ECM (Lohler et al. [1984] Cell 38:597-607). Several matricellular proteins, proteoglycans, and growth factors bind to type I collagen, e.g., fibronectin, secreted protein acidic and rich in cysteine (SPARC), decorin, and transforming growth factor-beta. Here we investigate the expression and function of SPARC in the absence of type I collagen. We show that fibroblasts isolated from Mov-13 homozygous, heterozygous, and wild-type embryos transcribed and translated SPARC mRNA in vitro. However, accumulation of extracellular SPARC was severely affected in the tissues of Mov-13 homozygotes, whereas extracellular deposition of the secreted glycoproteins fibronectin and type III collagen was not altered. Since SPARC has been shown to be a regulator of cell shape, the functional consequences of the absence of extracellular SPARC were evaluated in collagen gel contraction assays. Fibroblasts isolated from homozygous Mov-13 mice did not contract native type I collagen gels as efficiently as fibroblasts from heterozygous littermates; however, addition of exogenous SPARC enhanced the contraction of collagen by homozygous Mov-13 fibroblasts. The stimulatory effect of SPARC was blocked by antibodies specific for the amino terminus of the protein. These results provide evidence that type I collagen is one of the major extracellular proteins that binds SPARC in vivo. Furthermore, the capacity of fibroblasts to contract ECM in vitro is enhanced by extracellular SPARC. We therefore propose that the remodeling of ECM by cells in vivo is regulated in part by a specific interaction between SPARC and type I collagen.


Subject(s)
Collagen/metabolism , Extracellular Matrix/metabolism , Fibroblasts/metabolism , Osteonectin/metabolism , Animals , Cells, Cultured , Collagen/genetics , Collagen Type I, alpha 1 Chain , Female , Fibroblasts/cytology , Male , Mice , Osteonectin/genetics , RNA, Messenger
10.
Mech Ageing Dev ; 89(1): 21-43, 1996 Jul 31.
Article in English | MEDLINE | ID: mdl-8819104

ABSTRACT

Aged mice that have undergone long-term caloric-restriction (CR) have improved health and enhanced longevity in comparison to aged mice that are ad libitum-fed (AL). However, caloric-restriction does not benefit the impaired wound healing of aged mice. To test the hypothesis that CR mice have the capacity for enhanced wound repair, but require a short-term period of additional nutrient intake to show this advantage, we assessed wound healing in CR mice that had been refed (RF) an ad libitum diet for 4 weeks prior to wounding. Two strains of AL young (Y AL) (4-6 months), AL middle-aged (M AL) (15-17 months), and three different, matched cohorts of old mice (O) (30-33 months): O AL, O CR, and O RF were studied. Two full-thickness 4 mm diameter punch biopsy skin wounds were created on the dorsum of each mouse. Animals were sacrificed and wounds were harvested at 1,2,3,5, and 7 days post-wounding. Repair of wounds was slower in O AL and O CR mice compared to Y AL and M AL animals. In contrast, the O RF mice healed similarly to that of the Y AL and M AL mice, as assessed by measures of wound area and histologic criteria. O RF mice demonstrated enhanced synthesis of type I collagen mRNA in comparison to O AL and O CR mice. A greater number of endothelial cells and fibroblasts at the wound edge of the O RF mice exhibited replication in vivo as measured by uptake of BrdU. O RF mice had higher levels of insulin-like binding protein 3 (IGFBP-3). Furthermore, fibroblasts derived from the explant of the punch biopsy of O CR mouse skin revealed enhanced proliferation and contraction in vitro, in comparison to fibroblasts from the O AL mice. In conclusion, O RF mice demonstrate an enhanced capacity to undergo wound repair in comparison to O AL mice. This effect appears to be mediated, in part, by enhanced cell proliferation, contraction, and collagen biosynthesis. In addition, short-term refeeding induced an increase in the serum level of IGFBP-3, the major binding protein for IGF-1. These data confirm that cells from O CR animals have a preserved proliferative, biosynthetic, and contractile capacity, but that an adequate source of nutrients is necessary to demonstrate this advantage in wound healing.


Subject(s)
Aging/physiology , Wound Healing/physiology , Animals , Cell Division/physiology , Collagen/genetics , Insulin-Like Growth Factor Binding Proteins/metabolism , Insulin-Like Growth Factor I/metabolism , Male , Mice , Mice, Inbred Strains , RNA, Messenger/biosynthesis
11.
J Cell Biochem ; 60(2): 185-97, 1996 Feb.
Article in English | MEDLINE | ID: mdl-8655629

ABSTRACT

The formation of microvascular sprouts during angiogenesis requires that endothelial cells move through an extracellular matrix. Endothelial cells that migrate in vitro generate forces of traction that compress (i.e., contract) and reorganize vicinial extracellular matrix, a process that might be important for angiogenic invasion and morphogenesis in vivo. To study potential relationships between traction and angiogenesis, we have measured the contraction of fibrillar type I collagen gels by endothelial cells in vitro. We found that the capacity of bovine aortic endothelial (BAE) cells to remodel type I collagen was similar to that of human dermal fibroblasts--a cell type that generates high levels of traction. Contraction of collagen by BAE cells was stimulated by fetal bovine serum, human plasma-derived serum, bovine serum albumin, and the angiogenic factors phorbol myristate acetate and basic fibroblast growth factor (bFGF). In contrast, fibronectin and immunoglobulin from bovine serum, several nonserum proteins, and polyvinyl pyrrolidone (a nonproteinaceous substitute for albumin in artificial plasma) were not stimulatory. Contraction of collagen by BAE cells was diminished by an inhibitor of metalloproteinases (1,10-phenanthroline) at concentrations that were not obviously cytotoxic. Zymography of proteins secreted by BAE cells that had contracted collagen gels revealed matrix metalloproteinase 2. Subconfluent BAE cells that were migratory and proliferating were more effective contractors of collagen than were quiescent, confluent cells of the same strain. Moreover, bovine capillary endothelial cells contracted collagen gels to a greater degree than was seen with BAE cells. Collectively, our observations indicate that traction-driven reorganization of fibrillar type I collagen by endothelial cells is sensitive to different mediators, some of which, e.g., bFGF, are known regulators of angiogenesis in vivo.


Subject(s)
Cell Movement/physiology , Collagen/ultrastructure , Endothelium, Vascular/cytology , Extracellular Matrix/physiology , Neovascularization, Physiologic , Animals , Biomechanical Phenomena , Cattle , Cells, Cultured , Endothelium, Vascular/drug effects , Enzyme Inhibitors/metabolism , Gels , Humans , Metalloendopeptidases/antagonists & inhibitors , Tetradecanoylphorbol Acetate/pharmacology
12.
Arch Biochem Biophys ; 325(1): 8-19, 1996 Jan 01.
Article in English | MEDLINE | ID: mdl-8554346

ABSTRACT

Human SPARC has been cloned by the polymerase chain reaction from an endothelial cell cDNA library and expressed in Escherichia coli as a biologically active protein. Transcriptional expression of the insert cDNA was dependent on the activation of the T7 RNA polymerase promoter by isopropylgalactopyranoside. Two forms of recombinant SPARC (rSPARC) protein were recovered from BL21 (DE3) E. coli after transformation with the plasmid pSPARCwt: a soluble, monomeric form of rSPARC and an insoluble, aggregated form sequestered in inclusion bodies. The isolation of the soluble form of rSPARC was accomplished by anion-exchange, nickel-chelate affinity, and gel filtration chromatographies. The isolated protein was an intact, full-length polypeptide of 293 amino acids by the following criteria: N-terminal amino acid sequence, reaction with anti-SPARC immunoglobulins specific for N-terminal and C-terminal sequences, and interaction of the C-terminal histidine tag of rSPARC with a nickel-chelate affinity resin. Circular dichroism and intrinsic fluorescence spectroscopy indicated that the conformation of rSPARC was dependent on interaction with Ca2- ions. The recombinant protein inhibited cell spreading and bound specifically to bovine aortic endothelial cells. Levels of bacterial endotoxin (< 18 pg/microgram rSPARC) present in rSPARC preparations were below the threshold that affects the behavior of these endothelial cells. These conformational and biological properties of rSPARC are consistent with previously described characteristics of the native protein. The purification of biologically active rSPARC, as well as mutated forms of the protein, will provide sufficient quantities of protein for the determination of structure/function relationships.


Subject(s)
Escherichia coli/genetics , Gene Expression , Osteonectin/genetics , Amino Acid Sequence , Animals , Base Sequence , Calcium/pharmacology , Cattle , Cloning, Molecular , DNA, Complementary/chemistry , DNA, Complementary/genetics , Endothelium, Vascular/metabolism , Humans , Molecular Sequence Data , Osteonectin/chemistry , Osteonectin/metabolism , Plasmids , Polymerase Chain Reaction , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Spectrometry, Fluorescence , Transformation, Bacterial
13.
Am J Pathol ; 147(4): 873-83, 1995 Oct.
Article in English | MEDLINE | ID: mdl-7573362

ABSTRACT

In response to an angiogenic stimulus, ECs initiate programs of gene expression that result in the quantitative alteration of gene products within nuclear, cytoplasmic, cell surface, and extracellular compartments. During the formation of new microvasculature, patterns of molecular expression among individual ECs must direct the creation of complex, multicellular morphologies in two and three dimensions. Studies in vitro indicate that cell-generated forces of tension can organize ECM into structures that direct the behavior of single cells (via influences on cellular elongation, alignment, and migration) and that provide positional information for the creation of multicellular patterns. Significantly, the formation of organized matrical structures is controlled by gene products (of ECs or other cell types that populate the ECM) that influence the balance between the forces of cellular tension and the viscoelastic resistance of the ECM. Regulation of relevant genes could be accomplished by soluble molecular signals (eg, growth factors) and/or solid-state signals arising from specific arrangements of cytoskeletal structure that, in turn, are a function of the equilibrium between cellular tension and matrical resistance. Within cells, information for the construction of complex organelles is encoded in the shapes of the constituent molecules. Similarly, the creation of complex vascular architecture must be mediated by molecular shapes, a fact that is readily apparent in simple receptor-ligand interactions such as the binding of growth factors to ECs or the attachment of ECs to one another. However, between molecules and morphology also exists a set of multilayered, interactive, multimolecular systems that establish vascular form at unicellular and multicellular levels. Characterization of these systems is an elusive target that resides at the frontier of vascular biology; the identification of models in vitro that accurately reproduce macroscale events of vascular morphogenesis should advance considerably our understanding of vascular development and lead to an elucidation of its regulation in vivo.


Subject(s)
Extracellular Matrix/physiology , Neovascularization, Physiologic , Animals , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/physiology , Humans , Microcirculation , Models, Cardiovascular , Traction
14.
In Vitro Cell Dev Biol Anim ; 31(2): 120-31, 1995 Feb.
Article in English | MEDLINE | ID: mdl-7537585

ABSTRACT

Selected strains of vascular endothelial cells, grown as confluent monolayers on tissue culture plastic, generate flat networks of cellular cords that resemble beds of capillaries--a phenomenon referred to as "spontaneous angiogenesis in vitro". We have studied spontaneous angiogenic activity by a clonal population (clone A) of bovine aortic endothelial cells to identify processes that mediate the development of cellular networks. Confluent cultures of clone A endothelial cells synthesized type I collagen, a portion of which was incorporated into narrow, extracellular cables that formed a planar network beneath the cellular monolayer. The collagenous cables acted as a template for the development of cellular networks: flattened, polygonal cells of the monolayer that were in direct contact with the cables acquired spindle shapes, associated to form cellular cords, and became elevated above the monolayer. Networks of cables and cellular cords did not form in a strain of bovine aortic endothelial cells that did not synthesize type I collagen, or when traction forces generated by clone A endothelial cells were inhibited with cytochalasin D. In a model of cable development, tension applied by a confluent monolayer of endothelial cells reorganized a sheetlike substrate of malleable type I collagen into a network of cables via the formation and radial enlargement of perforations through the collagen sheet. Our results point to a general involvement of extracellular matrix templates in two-dimensional (planar) models of vascular development in vitro. For several reasons, planar models simulate invasive angiogenesis poorly. In contrast, planar models might offer insights into the growth and development of planar vascular systems in vivo.


Subject(s)
Blood Vessels/growth & development , Collagen/physiology , Endothelium, Vascular/cytology , Neovascularization, Pathologic , Animals , Cattle , Cells, Cultured , Endothelium, Vascular/metabolism , Endothelium, Vascular/ultrastructure , Extracellular Matrix/ultrastructure , Immunohistochemistry , Microscopy, Electron, Scanning , Models, Biological
15.
J Hypertens Suppl ; 12(10): S145-52, 1994 Dec.
Article in English | MEDLINE | ID: mdl-7539492

ABSTRACT

DEFINITION: Angiogenesis is broadly defined as the growth of new capillaries from extant vessels and constitutes a major part of developmental morphogenesis, response to injury and pathogenesis. Two regulatory pathways are proposed by which angiogenesis is thought to proceed. PROLIFERATIVE PATHWAY: The proliferative pathway depends on various cytokines and other factors that both stimulate and inhibit the proliferation of endothelial cells. One of these components, secreted protein acidic and rich in cysteine (SPARC), might function at several levels to control the progression of neovessels. Proteolysis of this component (e.g. by plasmin) results in the release of peptides containing the sequence Gly-His-Lys, which are angiogenic in vitro and in vivo. At later stages of angiogenesis when endothelial cell proliferation ceases, the intact protein is proposed to exert its known inhibitory effect on cell cycle progression. MORPHOGENETIC PATHWAY: The morphogenetic pathway depends on the synthesis and assembly of fibrillar type I collagen, which can be used as a template for endothelial cell migration and lumen formation. Endothelial cells interact with substrates of type I collagen and form networks based on the establishment of traction centers. These planar cellular networks, in some respects, resemble developing vasculature in vivo. CONCLUSION: An understanding of how proliferation and morphogenesis are controlled during vascular growth is likely to reconcile several models with respect to the factors that regulate this dynamic process.


Subject(s)
Extracellular Matrix/physiology , Neovascularization, Pathologic , Animals , Cell Division/physiology , Collagen/physiology , Endothelium, Vascular/cytology , Membrane Glycoproteins/physiology
16.
J Cell Physiol ; 158(1): 169-79, 1994 Jan.
Article in English | MEDLINE | ID: mdl-8263022

ABSTRACT

Fibroblasts have a major role in the synthesis and reorganization of extracellular matrix that occur during wound repair. An impaired biosynthetic or functional response of these cells to stimulation by growth factors might contribute to the delayed wound healing noted in aging. We, therefore, compared the responses of dermal fibroblasts from young and elderly individuals (26, 29, 65, 89, 90, and 92 years of age) to transforming growth factor-beta 1 (TGF-beta 1) with respect to: (1) the synthesis of type I collagen and SPARC (two extracellular matrix proteins that are highly expressed by dermal fibroblasts during the remodeling phase of wound repair) and (2) the contraction of collagen gels, and in vitro assay of wound contraction. With the exception of one young donor, all cultures exposed for 44 hours to 10 ng/ml TGF-beta 1 exhibited a 1.6- to 5.5-fold increase in the levels of secreted type I collagen and SPARC, relative to untreated cultures, and exhibited a 2.0- to 6.2-fold increase in the amounts of the corresponding mRNAs. Moreover, the dose-response to TGF-beta 1 (0.1-10 ng/ml), as determined by synthesis of type I collagen and SPARC mRNA, was as vigorous in cells from aged donors as in cells from a young donor. In assays of collagen gel contraction, fibroblasts from all donors were stimulated to a similar degree by 10 ng/ml TGF-beta 1. In conclusion, cells from both young and aged donors exhibited similar biosynthetic and contractile properties with exposure to TGF-beta 1. It therefore appears that the impaired wound healing noted in the aged does not result from a failure of their dermal fibroblasts to respond to this cytokine.


Subject(s)
Aging/metabolism , Collagen/metabolism , Collagen/physiology , Osteonectin/metabolism , Skin/metabolism , Transforming Growth Factor beta/pharmacology , Adult , Aged , Aged, 80 and over , Collagen/genetics , DNA/biosynthesis , Fibroblasts/metabolism , Gels , Humans , Male , Osteonectin/genetics , RNA, Messenger/metabolism , Skin/cytology
17.
Lab Invest ; 66(5): 536-47, 1992 May.
Article in English | MEDLINE | ID: mdl-1374138

ABSTRACT

Vascular endothelial cells that are cultured on layers of gelled basement membrane matrix organize rapidly into networks of cords or tubelike structures. Although this phenomenon is a potential model for angiogenesis in vivo, we questioned whether basement membrane matrix directs the differentiation of endothelial cells in a specific manner. In this study, we have examined factors that influence the formation of cellular networks in vitro in an attempt to define a basic mechanism for this process. We found that endothelial cells, fibroblasts, smooth muscle cells, and cells of the murine Leydig cell line TM3 formed networks on basement membrane matrix in much the same fashion. Light and electron microscopy, combined with time-lapse videomicroscopy, revealed that cells organized on a tesselated network of aligned basement membrane matrix that was generated by tension forces of cellular traction. Cellular elongation and progressive motility across the surface of the gel were restricted to tracks of aligned matrix and did not occur until the tracks appeared. The formation of cellular networks on basement membrane matrix was inhibited by reducing the thickness of the matrix, by including native type I collagen in the matrix, or by disrupting cytoskeletal microfilaments and microtubules. Cell division was not required for network formation. Bovine aortic endothelial cells that formed networks did not simultaneously transcribe mRNA for type I collagen, a protein synthesized by endothelial cells that form tubes spontaneously in vitro. Moreover, levels of mRNA for fibronectin and SPARC (Secreted Protein that is Acidic and Rich in Cysteine) in network-forming cells were similar to levels seen in endothelial cells that did not form networks. Endothelial cells and TM3 cells that were plated on highly malleable gels of native type I collagen also formed cords and aligned matrix fibers into linear tracks that resembled those generated on basement membrane matrix, although the structures were not as well-defined. Our observations suggest that the mechanochemical properties of extracellular matrices are able to translate the forces of cellular traction into templates that direct the formation of complex cellular patterns.


Subject(s)
Basement Membrane/physiology , Cell Communication/physiology , Extracellular Matrix/physiology , Animals , Cattle , Cell Adhesion/physiology , Cell Differentiation/physiology , Cell Movement/physiology , Cells, Cultured , Culture Media , Endothelium, Vascular/cytology , Endothelium, Vascular/physiology , Fibroblasts/physiology , Humans , Leydig Cells/physiology , Male , Mice , Muscle, Smooth/cytology , Muscle, Smooth/physiology , Neovascularization, Pathologic/physiopathology
18.
Biol Reprod ; 44(1): 157-70, 1991 Jan.
Article in English | MEDLINE | ID: mdl-2015346

ABSTRACT

Interactions between Leydig cells and the extracellular matrix (ECM) within the interstitial compartment of the mammalian testis have not been characterized. We have examined the influence of ECM on adult mouse Leydig cells by culturing the cells on different ECM substrates. Leydig cells adhere weakly to hydrated gels of type I collagen (including those supplemented with collagen types IV, V, or VIII), or to air-dried films of collagen types I, V, or VIII. In contrast, the cells attach firmly to substrates of purified type IV collagen, fibronectin, or laminin. Leydig cells also attach rapidly and adhere strongly to gelled basement membrane matrix derived from the murine Englebreth-Holm-Swarm sarcoma (Matrigel). Leydig cells assume spherical shapes and form aggregates on thick (1.5-mm) layers of Matrigel; however, on thin (0.1-mm) layers, networks of cell clusters linked by cords of elongated cells are formed within 48 h. Similar networks are formed on thick layers of Matrigel that are supplemented with type I collagen. On substrates with high ratios of collagen I to Matrigel or on untreated tissue culture plastic, Leydig cells flatten and do not aggregate. On substrates that induce rounded shapes, proliferation is inhibited and the cells maintain the steroidogenic enzyme 3 beta-hydroxysteroid dehydrogenase for as long as 2 wk. Under conditions where Leydig cells are flattened, they divide and cease expressing the enzyme. Proliferating Leydig cells also exhibit elevated levels of mRNA for SPARC (Secreted Protein, Acidic and Rich in Cysteine), a Ca2(+)-binding glycoprotein associated with changes in cell shape that accompany morphogenesis and tissue remodeling. Our results indicate that the shape, association, proliferation, and expression of gene products by Leydig cells can be significantly affected in vitro by altering the composition of the extracellular substratum.


Subject(s)
Extracellular Matrix/physiology , Leydig Cells/cytology , Animals , Cell Adhesion , Cell Division , DNA/biosynthesis , DNA/genetics , Gene Expression , In Vitro Techniques , Leydig Cells/physiology , Male , Mice
19.
Placenta ; 11(2): 167-80, 1990.
Article in English | MEDLINE | ID: mdl-2188252

ABSTRACT

Monoclonal antibody MA21 recognized a 44kDa plasma membrane protein on F9 teratocarcinoma cells, trophectoderm of mouse peri-implantation-stage blastocyst and ectoplacental cone cells of 5 day postcoitum implanted blastocyst (Vernon, Linnemeyer and Hamilton, 1989). We show here that this antigen is expressed by trophoblast cells of the maturing placenta. Immunohistochemical assays of early and mature placental tissue sections, indirect immunofluorescence labelling of placental cultures and blastocyst outgrowths in vitro, and immunoprecipitation of 35S-labelled NP-40 extracts of placental cultures indicate the presence of a plasma membrane-associated antigen with the same characteristics as MA21 antigen of peri-implantation embryos and F9 teratocarcinoma cells. In sections of placentae, antigen-positive cells are always situated in a thin layer between trophoblastic giant cells and maternal tissue. In cultures of postimplantation stage embryos, attached trophoblast cells express MA21 antigen initially, but following transformation to the giant cell state, antigen is no longer expressed. These results indicate the presence of a plasma membrane protein antigen associated with a distinct population of cells believed to be trophoblast. We believe that these cells are the foremost trophoblast cells opposing maternal decidua and that they may give rise to secondary trophoblastic giant cells.


Subject(s)
Placenta/immunology , Trophoblasts/immunology , Animals , Antibodies, Monoclonal/immunology , Antigen-Antibody Reactions , Antigens, Surface , Female , Fluorescent Antibody Technique , Gene Expression , Immunohistochemistry , Mice , Mice, Inbred Strains , Microscopy , Placenta/cytology , Precipitin Tests
20.
J Cell Biol ; 109(1): 341-56, 1989 Jul.
Article in English | MEDLINE | ID: mdl-2745554

ABSTRACT

SPARC (Secreted Protein Acidic and Rich in Cysteine) is a Ca+2-binding glycoprotein that is differentially associated with morphogenesis, remodeling, cellular migration, and proliferation. We show here that exogenous SPARC, added to cells in culture, was associated with profound changes in cell shape, caused rapid, partial detachment of a confluent monolayer, and inhibited spreading of newly plated cells. Bovine aortic endothelial cells, exposed to 2-40 micrograms SPARC/ml per 2 x 10(6) cells, exhibited a rounded morphology in a dose-dependent manner but remained attached to plastic or collagen-coated surfaces. These round cells synthesized protein, uniformly excluded trypan blue, and grew in aggregates after replating in media without SPARC. SPARC caused rounding of bovine endothelial cells, fibroblasts, and smooth muscle cells; however, the cell lines F9, PYS-2, and 3T3 were not affected. The activity of native SPARC was inhibited by heat denaturation and prior incubation with anti-SPARC IgG. The effect of SPARC on endothelial cells appeared to be independent of the rounding phenomenon produced by the peptide GRGDSP. Immunofluorescence localization of SPARC on endothelial cells showed preferential distribution at the leading edges of membranous extensions. SPARC bound Ca+2 in both amino- and carboxyl-terminal (EF-hand) domains and required this cation for maintenance of native structure. Solid-phase binding assays indicated a preferential affinity of native SPARC for several proteins comprising the extracellular matrix, including types III and V collagen, and thrombospondin. This binding was saturable, Ca+2 dependent, and inhibited by anti-SPARC IgG. Endothelial cells also failed to spread on a substrate of native type III collagen complexed with SPARC. We propose that SPARC is an extracellular modulator of Ca+2 and cation-sensitive proteins or proteinases, which facilitates changes in cellular shape and disengagement of cells from the extracellular matrix.


Subject(s)
Calcium/physiology , Carrier Proteins/physiology , Endothelium, Vascular/cytology , Extracellular Matrix/physiology , Animals , Carrier Proteins/ultrastructure , Cattle , Cell Adhesion , Cell Division , Cell Movement , Cells, Cultured , Collagen/metabolism , In Vitro Techniques , Osteonectin , Protein Binding
SELECTION OF CITATIONS
SEARCH DETAIL
...