Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 121
Filter
Add more filters

Publication year range
1.
Biol Lett ; 18(10): 20220359, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36259234

ABSTRACT

Hosts may limit exposure to pathogens through changes in behaviour, such as avoiding infected individuals or contaminated areas. Here, we tested for a behavioural response to ranavirus infection in juvenile wood frogs (Rana sylvatica) because the majority of dispersal between populations occurs during this life stage. We hypothesized that if infections are transmissible and detectable at this life stage, then susceptibles would display avoidance behaviours when introduced to an infected conspecific. Despite no apparent signs of infection, we observed a greater distance between susceptible-infected pairs, compared to pairs of either two infected or two susceptible animals. Further, distances between susceptible-infected pairs were positively related to the infection intensity of the focal exposed frog, suggesting the cue to avoid infected conspecifics may become more detectable with more intense infections. Although we did not quantify whether the transmission was affected by their distancing, our findings suggest that juvenile frogs have the potential to reduce terrestrial transmission of ranaviruses through avoidance behaviours.


Subject(s)
DNA Virus Infections , Ranavirus , Animals , Avoidance Learning , Ranidae , Amphibians , Anura
2.
J Cell Biol ; 111(6 Pt 2): 3065-76, 1990 Dec.
Article in English | MEDLINE | ID: mdl-2269665

ABSTRACT

Using synthetic peptides, we have identified two distinct regions of the glycoprotein SPARC (Secreted Protein Acidic and Rich in Cysteine) (osteonectin/BM-40) that inhibit cell spreading. One of these sites also contributes to the affinity of SPARC for extracellular matrix components. Peptides representing subregions of SPARC were synthesized and antipeptide antibodies were produced. Immunoglobulin fractions of sera recognizing an NH2-terminal peptide (designated 1.1) blocked SPARC-mediated anti-spreading activity. Furthermore, when peptides were added to newly plated endothelial cells or fibroblasts, peptide 1.1 and a peptide corresponding to the COOH terminal EF-hand domain (designated 4.2) inhibited cell spreading in a dose-dependent manner. These peptides exhibited anti-spreading activity at concentrations from 0.1 to 1 mM. The ability of peptides 1.1 and 4.2 to modulate cell shape was augmented by an inhibitor of protein synthesis and was blocked by specific antipeptide immunoglobulins. In addition to blocking cell spreading, peptide 4.2 competed for binding of [125I]SPARC and exhibited differential affinity for extracellular matrix molecules in solid-phase binding assays. The binding of peptide 4.2 to matrix components was Ca+(+)-dependent and displayed specificities similar to those of native SPARC. These studies demonstrate that both anti-spreading activity and affinity for collagens are functions of unique regions within the SPARC amino acid sequence. The finding that two separate regions of the SPARC protein contribute to its anti-spreading activity lead us to propose that multiple regions of the protein act in concert to regulate the interactions of cells with their extracellular matrix.


Subject(s)
Cell Adhesion/physiology , Cell Movement/physiology , Osteonectin/physiology , Amino Acid Sequence , Animals , Antibodies , Binding, Competitive , Calcium/metabolism , Cells, Cultured , Collagen/metabolism , Mice , Molecular Sequence Data , Osteonectin/chemistry , Peptide Fragments/chemical synthesis , Peptide Fragments/metabolism , Structure-Activity Relationship
3.
J Cell Biol ; 121(6): 1433-44, 1993 Jun.
Article in English | MEDLINE | ID: mdl-8509459

ABSTRACT

SPARC (osteonectin/BM40) is a secreted protein that modifies the interaction of cells with extracellular matrix (ECM). When we added SPARC to cultured rabbit synovial fibroblasts and analyzed the secreted proteins, we observed an increase in the expression of three metalloproteinases--collagenase, stromelysin, and the 92-kD gelatinase--that together can degrade both interstitial and basement membrane matrices. We further characterized the regulation of one of these metalloproteinases, collagenase, and showed that both collagenase mRNA and protein are upregulated in fibroblasts treated with SPARC. Experiments with synthetic SPARC peptides indicated that a region in the neutral alpha-helical domain III of the SPARC molecule, which previously had no described function, was involved in the regulation of collagenase expression by SPARC. A sequence in the carboxyl-terminal Ca(2+)-binding domain IV exhibited similar activity, but to a lesser extent. SPARC induced collagenase expression in cells plated on collagen types I, II, III, and V, and vitronectin, but not on collagen type IV. SPARC also increased collagenase expression in fibroblasts plated on ECM produced by smooth muscle cells, but not in fibroblasts plated on a basement membrane-like ECM from Engelbreth-Holm-Swarm sarcoma. Collagenase was induced within 4 h in cells treated with phorbol diesters or plated on fibronectin fragments, but was induced after 8 h in cells treated with SPARC. A number of proteins were transiently secreted by SPARC-treated cells within 6 h of treatment. Conditioned medium that was harvested from cultures 7 h after the addition of SPARC, and depleted of residual SPARC, induced collagenase expression in untreated fibroblasts; thus, part of the regulation of collagenase expression by SPARC appears to be indirect and proceeds through a secreted intermediate. Because the interactions of cells with ECM play an important role in regulation of cell behavior and tissue morphogenesis, these results suggest that molecules like SPARC are important in modulating tissue remodeling and cell-ECM interactions.


Subject(s)
Extracellular Matrix/physiology , Metalloendopeptidases/biosynthesis , Osteonectin/physiology , Amino Acid Sequence , Animals , Cells, Cultured , Collagenases/biosynthesis , Fibroblasts/enzymology , Fibronectins/physiology , Mice , Molecular Sequence Data , Peptide Fragments , Rabbits , Synovial Membrane/enzymology , Time Factors , Tissue Plasminogen Activator/physiology
4.
J Cell Biol ; 107(2): 721-30, 1988 Aug.
Article in English | MEDLINE | ID: mdl-3047147

ABSTRACT

A pepsin-resistant triple helical domain (chain 50,000 Mr) of type VIII collagen was isolated from bovine corneal Descemet's membrane and used as an immunogen for the production of mAbs. An antibody was selected for biochemical and tissue immunofluorescence studies which reacted both with Descemet's membrane and with type VIII collagen 50,000-Mr polypeptides by competition ELISA and immunoblotting. This antibody exhibited no crossreactivity with collagen types I-VI by competition ELISA. The mAb specifically precipitated a high molecular mass component of type VIII collagen (EC2, of chain 125,000 Mr) from the culture medium of subconfluent bovine corneal endothelial cells metabolically labeled for 24 h. In contrast, confluent cells in the presence of FCS and isotope for 7 d secreted a collagenous component of chain 60,000 Mr that did not react with the anti-type VIII collagen IgG. Type VIII collagen therefore appears to be synthesized as a discontinuous triple helical molecule with a predominant chain 125,000 Mr by subconfluent, proliferating cells in culture. Immunofluorescence studies with the mAb showed that type VIII collagen was deposited as fibrils in the extracellular matrix of corneal endothelial cells. In the fetal calf, type VIII collagen was absent from basement membranes and was found in a limited number of tissues. In addition to the linear staining pattern observed in the Descemet's membrane, type VIII collagen was found in highly fibrillar arrays in the ocular sclera, in the meninges surrounding brain, spinal cord, and optic nerve, and in periosteum and perichondrium. Fine fibrils were evident in the white matter of spinal cord, whereas a more generalized staining was apparent in the matrices of cartilage and bone. Despite attempts to unmask the epitope, type VIII collagen was not found in aorta, kidney, lung, liver, skin, and ligament. We conclude that this unusual collagen is a component of certain specialized extracellular matrices, several of which are derived from the neural crest.


Subject(s)
Collagen/analysis , Cornea/analysis , Extracellular Matrix/analysis , Animals , Antibodies, Monoclonal/immunology , Cattle , Cells, Cultured , Collagen/immunology , Cornea/immunology , Cornea/ultrastructure , Cross Reactions , Culture Media , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , Hybridomas , Immunoassay , Precipitin Tests
5.
J Cell Biol ; 125(4): 917-28, 1994 May.
Article in English | MEDLINE | ID: mdl-7514607

ABSTRACT

To delineate potential angiogenic roles of platelet-derived growth factor (PDGF), we have investigated PDGF and its receptors on bovine aortic endothelial cells that exhibit spontaneous angiogenesis in vitro (angiogenic endothelial cells). Initiation of cord/tube formation by angiogenic endothelial cells required bovine or human serum. Neutralization of PDGF-BB in human serum with a monoclonal anti-PDGF-BB antibody reduced cord/tube formation by 37 +/- 10%, whereas neutralizing anti-PDGF-AA and an IgG isotype-matched control antibody had no effect. DNA synthesis in response to PDGF-BB increased as the cords and tubes developed; furthermore, PDGF-BB induced the incorporation of BrdU in the nuclei of cells associated with these structures. PDGF beta-receptor (PDGF-beta) mRNA increased concomitantly with cord/tube formation, and PDGFR-beta were specifically localized by immunocytochemistry to developing and mature cords and tubes. However, PDGFR-beta transcripts and protein were undetectable in nonangiogenic endothelial cells, and PDGF alpha-receptor mRNA was not expressed in either endothelial cell strain. In contrast to nonangiogenic endothelial cells, angiogenic endothelial cells did not express the PDGF B-chain, the required ligand for the PDGFR-beta. We conclude that (a) PDGF-BB can contribute to angiogenesis in vitro, (b) PDGFR-beta are specific for cord/tube-forming endothelial cells and mediate endothelial proliferation and cord/tube formation, and (c) in angiogenic and nonangiogenic endothelial cells, the expression of PDGFR-beta and PDGF B-chain is inversely correlated. We therefore suggest that paracrine PDGF might amplify angiogenesis via direct action on endothelially expressed PDGFR-beta.


Subject(s)
Endothelium, Vascular/cytology , Neovascularization, Pathologic/metabolism , Platelet-Derived Growth Factor/metabolism , Receptors, Platelet-Derived Growth Factor/metabolism , Animals , Becaplermin , Cattle , Cell Division , Cells, Cultured , Culture Media , Culture Media, Serum-Free , DNA/biosynthesis , Endothelium, Vascular/metabolism , Humans , Neovascularization, Pathologic/etiology , Phenotype , Proto-Oncogene Proteins c-sis , Recombinant Proteins
6.
J Cell Biol ; 125(4): 929-43, 1994 May.
Article in English | MEDLINE | ID: mdl-7514608

ABSTRACT

SPARC is a transiently expressed extracellular matrix-binding protein that alters cell shape and regulates endothelial cell proliferation in vitro. In this study, we show that SPARC mRNA and protein are synthesized by endothelial cells during angiogenesis in vivo. SPARC and peptides derived from a cationic region of the protein (amino acids 113-130) stimulated the formation of endothelial cords in vitro; moreover, these peptides stimulated angiogenesis in vivo. Mapping of the active domain demonstrated that the sequence KGHK was responsible for most of the angiogenic activity; substitution of the His residue decreased the effect. We found that proteolysis of SPARC provided a source of KGHK, GHK, and longer peptides that contained these sequences. Although the Cu(2+)-GHK complex had been identified as a mitogen/morphogen in normal human plasma, we found KGHK and longer peptides to be potent stimulators of angiogenesis. SPARC113-130 and KGHK were shown to bind Cu2+ with high affinity; however, previous incubation with Cu2+ was not required for the stimulatory activity. Since a peptide from a second cationic region of SPARC (SPARC54-73) also bound Cu2+ but had no effect on angiogenesis, the angiogenic activity appeared to be sequence specific and independent of bound Cu2+. Thus, specific degradation of SPARC, a matrix-associated protein expressed by endothelial cells during vascular remodeling, releases a bioactive peptide or peptides, containing the sequence (K)GHK, that could regulate angiogenesis in vivo.


Subject(s)
Carrier Proteins/metabolism , Copper/metabolism , Neovascularization, Pathologic/metabolism , Osteonectin/metabolism , Amino Acid Sequence , Animals , Binding Sites , Cattle , Cells, Cultured , Endopeptidases/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Extracellular Space/enzymology , Female , Fibrinolysin/metabolism , Male , Mice , Molecular Sequence Data , Neovascularization, Pathologic/etiology , Peptide Fragments/chemical synthesis , Peptide Fragments/metabolism , Trypsin/metabolism
7.
J Cell Biol ; 125(3): 695-704, 1994 May.
Article in English | MEDLINE | ID: mdl-8175887

ABSTRACT

Sequences within the first intron of the alpha 1(I) collagen gene have been implicated in the regulation of expression of alpha 1(I) collagen-reporter gene constructs in cultured cells. However, the physiological significance of these intronic elements has not been established. We have used in situ hybridization to examine whether a cell-specific pattern of expression of human alpha 1(I) collagen-human growth hormone minigenes exists in transgenic mice. Our results indicate that transgenes which contained 2,300 bp of promoter/5' flanking sequence and an intact first intron were well expressed by fibroblasts in dermis and fascia, whereas transgenes lacking the intronic sequence, +292 to +1440, were not expressed in dermis and poorly expressed in fascia. Analysis of transgene expression in cultured fibroblasts obtained from dermal explants of transgenic animals confirmed the requirement for these intronic sequences in the regulation of the alpha 1(I) collagen gene. In contrast, transgenes with or without the intronic deletion were expressed equally well in tendon and bone, in a manner comparable to the endogenous mouse alpha 1(I) collagen gene, and expression of neither transgene was detected in skeletal muscle or perichondrium. These data support a model in which cis-acting elements in the first intron, and their cognate DNA-binding proteins, mediate transcription of the alpha 1(I) collagen gene in some cells, such as dermal fibroblasts, but not in tendon cells or osteoblasts. Moreover, regions of the gene not included in the sequence, -2300 to +1440, appear to be required for transcription in tissues such as skeletal muscle and perichondrium.


Subject(s)
Collagen/genetics , Growth Hormone/genetics , Animals , Female , Fibroblasts/physiology , Gene Expression Regulation , In Situ Hybridization , Introns , Male , Mice , Mice, Transgenic/genetics , Osteoblasts/physiology , Promoter Regions, Genetic , RNA, Messenger/genetics , Skin Physiological Phenomena , Tendons/physiology , Tissue Distribution
8.
Leukemia ; 21(9): 1931-6, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17625608

ABSTRACT

The commonly deleted region (CDR) for the 5q- syndrome has been identified as a 1.5-megabase interval on human chromosome 5q32. We studied, by real-time reverse-transcription (RT)-PCR, the expression of 33 genes within the CDR that are known to be expressed in CD34+ hematopoietic stem cells. Genes in the 5q- samples that showed the most pronounced decrease in expression compared to non-5q- samples were: solute carrier family 36, member 1 (SLC36A1; 89% downregulated), Ras-GTPase-activating protein SH3 domain-binding (G3BP; 79%), antioxidant protein 1 (ATOX1; 76%), colony-stimulating factor-1 receptor precursor (CSF1R; 76%), ribosomal protein S14 (RPS14; 74%), platelet-derived growth factor receptor-beta (PDGFRB; 73%), Nef-associated factor 1 (TNIP1; 72%), secreted protein, acidic and rich in cysteine (SPARC; 71%), annexin VI (ANAX6; 69%), NSDT (66%) and TIGD (60%). We further studied the hematopoietic system in SPARC-null mice. These mice showed significantly lower platelet counts compared to wild-type animals (P=0.008). Although hemoglobin, hematocrit and mean corpuscular volume (MCV) were lower in mice lacking SPARC, differences were not statistically significant. SPARC-null mice showed a significantly impaired ability to form erythroid burst-forming units (BFU-E). However, no significant differences were found in the formation of erythroid colony-forming units (CFU-E), granulocyte/monocyte colony-forming units (CFU-GM) or megakaryocyte colony-forming units (CFU-Mk) in these animals. We conclude that many of the genes within the CDR associated with the 5q- syndrome exhibit significantly decreased expression and that SPARC, as a potential tumor suppressor gene, may play a role in the pathogenesis of this disease.


Subject(s)
Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/pathology , Osteonectin/genetics , Thrombocytopenia/genetics , Thrombocytopenia/pathology , Animals , Bone Marrow Cells/cytology , Chromosome Deletion , Erythrocyte Count , Erythroid Cells/cytology , Flow Cytometry , Gene Expression Profiling , Genes, Tumor Suppressor , HL-60 Cells , Hematopoiesis/genetics , Humans , Megakaryocytes/cytology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Platelet Count , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells
9.
J Clin Invest ; 97(2): 403-12, 1996 Jan 15.
Article in English | MEDLINE | ID: mdl-8567961

ABSTRACT

Thrombospondin-1 (TSP1), a multifunctional extracellular matrix glycoprotein, has been shown to suppress the angiogenic response in vivo and in vitro. We hypothesized that TSP1 might play a role in the inhibition of capillary morphogenesis during the endometrial cycle and examined its expression in 46 human endometrial specimens. Our results show that the expression of TSP1 in the endometrium is (a) cycle-dependent, (b) associated with periods of low capillary growth, and (c) regulated by progesterone. TSP1 protein was identified in the basement membrane of capillaries of the functional endometrium during the secretory phase. Abundant expression of TSP1 mRNA in the secretory phase was also detected by in situ hybridization, in contrast to the low levels seen in the proliferative phase. These findings were confirmed by Northern analysis of proliferative and secretory endometrium. Transcripts for TSP1 were observed predominantly in stromal cells, but signal was also detected in some endothelial and smooth muscle cells. Since the proliferation of endometrial tissue is regulated by steroid hormones, we tested the effects of estrogen and progesterone on TSP1 expression by stromal cells isolated from human endometrium. We found that levels of TSP1 mRNA and protein were increased after incubation with progesterone. Maximal stimulation of mRNA was observed after 8 h of treatment with 10-50 microM progesterone, and the effect was suppressed by the progesterone antagonist RU-486. Induction by progesterone was cell-specific and equivalent to the stimulation mediated by PDGF. Finally, the levels of TSP1 present in progesterone-stimulated cultures were sufficient to inhibit the migration of endothelial cells in vitro; this effect was nullified by anti-TSP antibodies. We therefore propose that the production of TSP1 at later stages of the endometrial cycle is linked to the inhibition of vessel formation and that TSP1 expression is progesterone-dependent in this tissue.


Subject(s)
Endometrium/physiology , Membrane Glycoproteins/genetics , Progesterone/physiology , Adult , Aged , Base Sequence , Basement Membrane/metabolism , Cells, Cultured , DNA Primers/chemistry , Extracellular Matrix Proteins/genetics , Female , Gene Expression , Hormone Antagonists/pharmacology , Humans , In Situ Hybridization , Middle Aged , Mifepristone/pharmacology , Molecular Sequence Data , Neovascularization, Physiologic/drug effects , Proliferating Cell Nuclear Antigen/genetics , RNA, Messenger/genetics , Thrombospondins
10.
Nat Biotechnol ; 15(6): 581-5, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9181583

ABSTRACT

Human SPARC (secreted protein acidic and rich in cysteine), an extracellular matrix protein containing 14 cysteine residues, was found to partition equally between soluble and insoluble cellular fractions when overexpressed in the Escherichia coli cytoplasm. While the growth temperature and medium pH had little effect on inclusion body formation, co-overproduction of the dnaKJ operon, but not of the groE operon, suppressed aggregation at the expense of intracellular accumulation. Although both forms of the protein were fully reduced in wild-type cells, 70% to 85% of soluble and insoluble SPARC could be converted into oxidized species in a thioredoxin reductase (trxB) null mutant following incubation on ice. Approximately 15% to 20% of SPARC exhibited the electrophoretic mobility of the biologically active protein. Overproduction of the dnaKJ operon in trxB cells decreased the formation of disulfide-bonded SPARC multimers in the aggregated material but not in its soluble counterpart. Our results suggest that the activity responsible for disulfide bond formation in trxB mutants acts at the post-translational level and is able to freely diffuse within inclusion bodies.


Subject(s)
Escherichia coli/metabolism , Osteonectin/metabolism , Amino Acid Sequence , Cloning, Molecular/methods , Cysteine , Cytoplasm/metabolism , Disulfides/analysis , Escherichia coli/genetics , Fermentation , Humans , Operon , Osteonectin/biosynthesis , Oxidation-Reduction , Plasmids , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
11.
Mol Biol Cell ; 10(5): 1569-79, 1999 May.
Article in English | MEDLINE | ID: mdl-10233163

ABSTRACT

SPARC (secreted protein acidic and rich in cysteine)/BM 40/osteonectin is a matricellular protein shown to function as a counteradhesive factor that induces cell rounding and as an inhibitor of cell proliferation. These activities have been defined in cell culture, in which interpretation has been complicated by the presence of endogenous SPARC. We therefore sought to determine whether cell shape and proliferation would be affected by the absence of SPARC. Mesangial cells, fibroblasts, and aortic smooth muscle cells were isolated from SPARC-null and age-matched, wild-type mice. In contrast to wild-type cells, SPARC-null mesangial cells exhibited a flat morphology and an altered actin cytoskeleton. In addition, vinculin-containing focal adhesions were distributed over the center of SPARC-null cells, whereas in wild-type cells, the number of focal adhesions was reduced, and these structures were restricted largely to the cell periphery. Although the SPARC-null fibroblasts did not display overt differences in cell morphology, the cells responded to exogenous recombinant SPARC by rounding up in a manner similar to that of wild-type fibroblasts. Thus, the expression of endogenous SPARC is not required for the response of cells to SPARC. Additionally, SPARC-null mesangial cells, fibroblasts, and smooth muscle cells proliferated faster than their respective wild-type counterparts. Null cells also showed a greater sensitivity to the inhibition of cell cycle progression by the addition of recombinant SPARC. The increased proliferation rate of SPARC-null cells appeared to be mediated, at least in part, by an increase in the cell cycle regulatory protein cyclin A. We conclude that the expression of SPARC influences the cellular architecture of mesangial cells and that SPARC plays a role in the regulation of cell cycle in mesangial cells, fibroblasts, and smooth muscle cells.


Subject(s)
Mesoderm/cytology , Osteonectin/genetics , Animals , Cell Cycle/genetics , Cell Division , Cell Size , Fibroblasts/cytology , Kidney/cytology , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Mutant Strains , Muscle, Smooth, Vascular/cytology , Osteonectin/metabolism
12.
Mol Biol Cell ; 6(3): 327-43, 1995 Mar.
Article in English | MEDLINE | ID: mdl-7612967

ABSTRACT

SPARC is a secreted glycoprotein that has been shown to disrupt focal adhesions and to regulate the proliferation of endothelial cells in vitro. Moreover, peptides resulting from the proteolysis of SPARC exhibit angiogenic activity. Here we describe the temporal synthesis, turnover, and angiogenic potential of SPARC in the chicken chorioallantoic membrane. Confocal immunofluorescence microscopy revealed specific expression of SPARC protein in endothelial cells, and significantly higher levels of SPARC were observed in smaller newly formed blood vessels in comparison to larger, developmentally older vessels. SPARC mRNA was detected at the earliest stages of chorioallantoic membrane morphogenesis and reached maximal levels at day 13 of embryonic development. Interestingly, steady-state levels of SPARC mRNA did not correlate directly with protein accumulation; moreover, the protein appeared to undergo limited degradation during days 10-15. Incubation of [125I]-SPARC with chorioallantoic membranes of different developmental ages confirmed that extracellular proteolysis occurred during days 9-15, but not at later stages (e.g., days 17-21). Comparison of peptides produced by incubation with chorioallantoic membranes with those generated by plasmin showed an identical pattern of proteolysis. Plasmin activity was present throughout development, and in situ zymography identified sites of plasminogen activator activity that corresponded to areas exhibiting high levels of SPARC expression. Synthetic peptides from a plasmin-sensitive region of SPARC, between amino acids 113-130, stimulated angiogenesis in the chorioallantoic membrane in a dose-dependent manner; in contrast, intact SPARC was inactive in similar assays. We have shown that SPARC is expressed in endothelial cells of newly formed blood vessels in a manner that is both temporally and spatially restricted. Between days 9 and 15 of chorioallantoic membrane development, the protein undergoes proteolytic cleavage that is mediated, in part, by plasmin. SPARC peptides released specifically by plasmin induce angiogenesis in vivo. We therefore propose that SPARC acts as an intrinsic regulator of angiogenesis in vivo.


Subject(s)
Allantois/growth & development , Chorion/growth & development , Endopeptidases/metabolism , Gene Expression Regulation, Developmental , Osteonectin/biosynthesis , Amino Acid Sequence , Animals , Capillaries/embryology , Cell Adhesion , Chick Embryo , Endothelium, Vascular/embryology , Endothelium, Vascular/metabolism , Extracellular Space/enzymology , Fibrinolysin/metabolism , Microscopy, Confocal , Molecular Sequence Data , Morphogenesis , Osteonectin/genetics , Peptide Fragments/pharmacology
14.
Cancer Res ; 58(23): 5529-36, 1998 Dec 01.
Article in English | MEDLINE | ID: mdl-9850090

ABSTRACT

Activation of the matrix metalloproteinase 2 (MMP-2) has been shown to play a major role in the proteolysis of extracellular matrix (ECM) associated with tumor invasion. Although the precise mechanism of this activation remains elusive, levels of the membrane type 1-MMP (MT1-MMP) at the cell surface and of the tissue inhibitor of MMP-2 (TIMP-2) appear to be two important determinants. Induction of MMP-2 activation in cells cultivated on collagen type I gels indicated that the ECM is important in the regulation of this process. In this study, we show that SPARC/osteonectin, a small ECM-associated matricellular glycoprotein, can induce MMP-2 activation in two invasive breast cancer cell lines (MDA-MB-231 and BT549) but not in a noninvasive counterpart (MCF-7), which lacks MT1-MMP. Using a set of peptides from different regions of SPARC, we found that peptide 1.1 (corresponding to the NH2-terminal region of the protein) contained the activity that induced MMP-2 activation. Despite the requirement for MT1-MMP, seen in MCF-7 cells transfected with MT1-MMP, the activation of MMP-2 by SPARC peptide 1.1 was not associated with increased steady-state levels of MT1-MMP mRNA or protein in either MT1-MMP-transfected MCF-7 cells or constitutively expressing MDA-MB-231 and BT549 cells. We did, however, detect decreased levels of TIMP-2 protein in the media of cells incubated with peptide 1.1 or recombinant SPARC; thus, the induction of MMP-2 activation by SPARC might be due in part to a diminution of TIMP-2 protein. We conclude that SPARC, and specifically its NH2-terminal domain, regulates the activation of MMP-2 at the cell surface and is therefore likely to contribute to the proteolytic pathways associated with tumor invasion.


Subject(s)
Breast Neoplasms/enzymology , Gelatinases/metabolism , Metalloendopeptidases/metabolism , Osteonectin/physiology , Amino Acid Sequence , Antibodies/pharmacology , Binding Sites , Collagen/pharmacology , Enzyme Activation , Humans , Integrins/immunology , Matrix Metalloproteinase 2 , Molecular Sequence Data , Neoplasm Invasiveness , Osteonectin/biosynthesis , Osteonectin/genetics , Peptide Fragments/pharmacology , Tissue Inhibitor of Metalloproteinase-2/metabolism , Transfection , Tumor Cells, Cultured
15.
Trends Cardiovasc Med ; 11(1): 32-7, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11413050

ABSTRACT

In the last decade, numerous studies have emphasized the important functions that matricellular proteins subserve during angiogenesis, wound healing, and the maintenance of organ and tissue integrity. Matricellular proteins are defined as a group of secreted regulatory macromolecules that are not structural components of the extracellular matrix (ECM) but rather mediate interactions between the ECM and cells. One of these matricellular proteins, termed SPARC (secreted protein acidic and rich in cysteine), is produced during the process of wound healing and is prominent in several types of injury. An excessive deposition of glomerular matrix and an elevated proliferation of certain glomerular cells characterize a variety of kidney diseases. The proliferation of these cells is associated typically with the remodeling process that occurs after kidney injury, and is, at least in part, modulated by the altered expression of ECM, various growth factors, and the elevated production of matricellular proteins (e.g., SPARC). The secretion of one or more of the matricellular proteins can lead to expansion of the glomerular basement membrane, infiltration of immunocompetent cells, and, in some cases, to a reversal of the pathological condition. However, these proteins can also contribute collectively to renal fibrosis, glomerulosclerosis, glomerulonephritis, and the eventual loss of renal function. The purpose of this review is to evaluate the multiple functions of SPARC in the kidney glomerulus under normal and pathological conditions.


Subject(s)
Kidney Glomerulus/physiology , Osteonectin/physiology , Animals , Cell Division , Extracellular Matrix Proteins/metabolism , Extracellular Matrix Proteins/physiology , Glomerular Mesangium/cytology , Glomerular Mesangium/metabolism , Growth Substances/metabolism , Humans , Kidney Diseases/physiopathology , Kidney Glomerulus/metabolism , Nephritis/physiopathology , Osteonectin/chemistry , Osteonectin/metabolism
16.
Int J Biochem Cell Biol ; 28(9): 1031-43, 1996 Sep.
Article in English | MEDLINE | ID: mdl-8930126

ABSTRACT

SPARC (secreted protein acidic and rich in cysteine, also known as osteonectin and BM-40) belongs to a group of secreted macromolecules that modulate cellular interactions with the extracellular matrix. During vertebrate embryogenesis, as well as in tissues undergoing remodeling and repair, the expression pattern of SPARC is consistent with a fundamental role for this protein in tissue morphogenesis and cellular differentiation. Human SPARC was cloned by the polymerase chain reaction from an endothelial cell cDNA library and was expressed in Escherichia coli as a biologically active protein. Two forms of recombinant SPARC (rSPARC) were recovered from BL21(DE3) cells after transformation with the plasmid pSPARCwt: a soluble, monomeric form that is biologically active (Bassuk et al., 1996, Archiv. Biochem. Biophys. 325, 8-19), and an insoluble form sequestered in inclusion bodies. Aggregated rSPARC was unfolded by urea treatment, purified by nickel-chelate affinity chromatography, and renatured by gradual removal of the denaturant. Proper isomerization of the disulfide bonds was achieved in the presence of a glutathione redox couple. After final purification by high resolution gel filtration chromatography, a monomeric form of rSPARC displaying biological activity was obtained. The recombinant protein inhibited the spreading and synthesis of DNA by endothelial cells, two properties characteristic of the native protein. We conclude that the information for the correct folding of rSPARC resides in the primary structure of the protein, and suggest that post-translational modifications are required neither for folding nor for biological activity.


Subject(s)
Disulfides/chemistry , Osteonectin/chemistry , Animals , Cattle , Chromatography, Affinity , Circular Dichroism , Electrophoresis, Polyacrylamide Gel , Escherichia coli , Glutathione/analogs & derivatives , Glutathione/pharmacology , Glutathione Disulfide , Humans , Inclusion Bodies/chemistry , Isomerism , Molecular Weight , Recombinant Proteins/chemistry , Solubility
17.
Matrix Biol ; 19(7): 569-80, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11102747

ABSTRACT

SPARC is a multifunctional glycoprotein that belongs to the matricellular group of proteins. It modulates cellular interaction with the extracellular matrix (ECM) by its binding to structural matrix proteins, such as collagen and vitronectin, and by its abrogation of focal adhesions, features contributing to a counteradhesive effect on cells. SPARC inhibits cellular proliferation by an arrest of cells in the G1 phase of the cell cycle. It also regulates the activity of growth factors, such as platelet-derived growth factor (PDGF), fibroblast growth factor (FGF)-2, and vascular endothelial growth factor (VEGF). The expression of SPARC in adult animals is limited largely to remodeling tissue, such as bone, gut mucosa, and healing wounds, and it is prominent in tumors and in disorders associated with fibrosis. The crystal structure of two of the three domains of the protein has revealed a novel follistatin-like module and an extracellular calcium-binding (EC) module containing two EF-hand motifs. The follistatin-like module and the EC module are shared by at least four other proteins that comprise a family of SPARC-related genes. Targeted disruption of the SPARC locus in mice has shown that SPARC is important for lens transparency, as SPARC-null mice develop cataracts shortly after birth. SPARC is a prototypical matricellular protein that functions to regulate cell-matrix interactions and thereby influences many important physiological and pathological processes.


Subject(s)
Extracellular Matrix Proteins/metabolism , Extracellular Matrix/metabolism , Osteonectin/metabolism , Animals , Extracellular Matrix Proteins/chemistry , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/physiology , Gene Expression Regulation , Growth Substances/metabolism , Humans , Osteonectin/chemistry , Osteonectin/genetics , Osteonectin/physiology , Protein Structure, Secondary , Receptors, Cell Surface/metabolism , Signal Transduction
18.
Matrix Biol ; 19(8): 816-27, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11223341

ABSTRACT

SPARC is a multifunctional glycoprotein that belongs to the matricellular group of proteins. It modulates cellular interaction with the extracellular matrix (ECM) by its binding to structural matrix proteins, such as collagen and vitronectin, and by its abrogation of focal adhesions, features contributing to a counteradhesive effect on cells. SPARC inhibits cellular proliferation by an arrest of cells in the G1 phase of the cell cycle. It also regulates the activity of growth factors, such as platelet-derived growth factor (PDGF), fibroblast growth factor (FGF)-2, and vascular endothelial growth factor (VEGF). The expression of SPARC in adult animals is limited largely to remodeling tissue, such as bone, gut mucosa, and healing wounds, and it is prominent in tumors and in disorders associated with fibrosis. The crystal structure of two of the three domains of the protein has revealed a novel follistatin-like module and an extracellular calcium-binding (EC) module containing two EF-hand motifs. The follistatin-like module and the EC module are shared by at least four other proteins that comprise a family of SPARC-related genes. Targeted disruption of the SPARC locus in mice has shown that SPARC is important for lens transparency, as SPARC-null mice develop cataracts shortly after birth. SPARC is a prototypical matricellular protein that functions to regulate cell-matrix interactions and thereby influences many important physiological and pathological processes.


Subject(s)
Osteonectin/physiology , Animals , Extracellular Matrix/metabolism , Extracellular Matrix/physiology , Gene Expression Regulation , Growth Substances/metabolism , Humans , Osteonectin/chemistry , Osteonectin/genetics , Osteonectin/metabolism , Receptors, Cell Surface/metabolism , Signal Transduction
19.
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
20.
J Histochem Cytochem ; 47(12): 1495-506, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10567433

ABSTRACT

SPARC (secreted protein, acidic and rich in cysteine) is a unique matricellular glycoprotein that is expressed by many different types of cells and is associated with development, remodeling, cell turnover, and tissue repair. Its principal functions in vitro are counteradhesion and antiproliferation, which proceed via different signaling pathways. SPARC consists of three domains, each of which has independent activity and unique properties. The extracellular calcium binding module and the follistatin-like module have been recently crystallized. Specific interactions between SPARC and growth factors, extracellular matrix proteins, and cell surface proteins contribute to the diverse activities described for SPARC in vivo and in vitro. The location of SPARC in the nuclear matrix of certain proliferating cells, but only in the cytosol of postmitotic neurons, indicates potential functions of SPARC as a nuclear protein, which might be involved in the regulation of cell cycle progression and mitosis. High levels of SPARC have been found in adult eye, and SPARC-null mice exhibit cataracts at 1-2 months of age. This animal model provides an excellent opportunity to confirm and explore some of the properties of SPARC, to investigate cataractogenesis, and to study SPARC-related family proteins, e.g., SC1/hevin, a counteradhesive matricellular protein that might functionally compensate for SPARC in certain tissues.(J Histochem Cytochem 47:1495-1505, 1999)


Subject(s)
Nuclear Matrix/metabolism , Osteonectin/physiology , Animals , Calcium/metabolism , Cataract/genetics , Cataract/pathology , Cell Adhesion , Cell Division , Cell Nucleus/metabolism , Cytokines/metabolism , Extracellular Matrix Proteins/metabolism , Humans , Mice , Mice, Knockout , Osteonectin/chemistry , Osteonectin/genetics , Osteonectin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL