Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 153
Filter
1.
Cancer Res ; 61(7): 2857-61, 2001 Apr 01.
Article in English | MEDLINE | ID: mdl-11306458

ABSTRACT

Data presented in this report indicate short-term in vitro treatment of nonmetastatic MCF-7 breast carcinoma cells with the chemotherapeutic agents-, Adriamycin and/or 5-fluoro-2'-deoxyuridine (FUdR), induced changes in the expressed phenotype. Cells treated sequentially with Adriamycin and FUdR expressed a metastatic phenotype. The results also show short-term exposure of MCF-7 cells to either Adriamycin or FUdR rapidly increases, in a dose-dependent manner, the release of the angiogenic cytokine, interleukin-8(IL-8), which is released at consistently higher levels in metastatic cell lines. Cell populations surviving a single treatment with either one or both of these chemotherapeutic agents continue to stably release IL-8. Survivors of sequential treatment with Adriamycin and FUdR (MCF-7 A/F) release the most IL-8 and express the greatest phenotypic variance from the parental, MCF-7 cells. Parental MCF-7 cells and MCF-7 A/F cells both form primary tumors when used in an orthotopic tumor model; however, the MCF-7 A/F tumors have a more rapid initial growth phase in situ and give rise to spontaneous lung metastases within 10 weeks. A cell line that is established from lung metastases releases more IL-8, has a higher cloning efficiency, and forms looser colonies in monolayer than do their parental cells. These experiments indicate the in vitro exposure of tumor cells to chemotherapeutic agents either selects more aggressive cells or enhances the metastatic potential of the surviving cells.


Subject(s)
Antibiotics, Antineoplastic/adverse effects , Antimetabolites, Antineoplastic/adverse effects , Breast Neoplasms/pathology , Doxorubicin/adverse effects , Floxuridine/adverse effects , Animals , Breast Neoplasms/metabolism , Cell Division/drug effects , Cell Survival/physiology , Disease Progression , Female , Humans , Interleukin-8/metabolism , Mice , Mice, Nude , Neoplasm Metastasis , Phenotype , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/pathology
2.
J Biol Chem ; 276(22): 18786-94, 2001 Jun 01.
Article in English | MEDLINE | ID: mdl-11278606

ABSTRACT

Tumor cell adhesion and proteolysis of the extracellular matrix proteins surrounding the cells are tightly linked processes in tumor invasion. In this study, we sought to identify components of the cell surface of a vertical growth phase melanoma cell line, WM1341D, that mediate invasive cellular behavior. We determined by antisense inhibition that melanoma chondroitin sulfate proteoglycan (MCSP) and membrane-type 3 matrix metalloproteinase (MT3-MMP) expressed on WM1341D are required for invasion of type I collagen and degradation of type I gelatin. MT3-MMP co-immunoprecipitated with MCSP in WM1341D melanoma cells cultured on type I collagen or laminin. The association between MT3-MMP and MCSP was largely disrupted by removing chondroitin sulfate glycosaminoglycan (CS) from the cell surface, suggesting CS could mediate the association between the two cell surface core proteins. Recombinant MT3-MMP and MT3-MMP from whole cell lysates of WM1341D cells were specifically eluted from CS- conjugated affinity columns. The results indicate that MT3-MMP possesses the potential to promote melanoma invasion and proteolysis and that the formation of a complex between MT3-MMP and MCSP may be a crucial step in activating these processes.


Subject(s)
Chondroitin Sulfate Proteoglycans/metabolism , Collagen/metabolism , Matrix Metalloproteinases/metabolism , Melanoma/metabolism , Metalloendopeptidases/metabolism , Animals , Cell Adhesion , Cell Line , Cell Movement , Chromatography, Affinity , Flow Cytometry , Gelatin/metabolism , Humans , Matrix Metalloproteinase 16 , Matrix Metalloproteinases, Membrane-Associated , Oligonucleotides, Antisense/metabolism , Plasmids/metabolism , Precipitin Tests , RNA, Messenger/metabolism , Rats , Sepharose/metabolism , Tumor Cells, Cultured
3.
J Biol Chem ; 276(21): 17949-57, 2001 May 25.
Article in English | MEDLINE | ID: mdl-11278609

ABSTRACT

Bone marrow is the primary site of metastasis in patients with advanced stage prostate cancer. Prostate carcinoma cells metastasizing to bone must initially adhere to endothelial cells in the bone marrow sinusoids. In this report, we have modeled that interaction in vitro using two bone marrow endothelial cell (BMEC) lines and four prostate adenocarcinoma cell lines to investigate the adhesion mechanism. Highly metastatic PC3 and PC3M-LN4 cells were found to adhere rapidly and specifically (70-90%) to BMEC-1 and trHBMEC bone marrow endothelial cells, but not to human umbilical vein endothelial cells (15-25%). Specific adhesion to BMEC-1 and trHBMEC was dependent upon the presence of a hyaluronan (HA) pericellular matrix assembled on the prostate carcinoma cells. DU145 and LNCaP cells were only weakly adherent and retained no cell surface HA. Maximal BMEC adhesion and HA encapsulation were associated with high levels of HA synthesis by the prostate carcinoma cells. Up-regulation of HA synthase isoforms Has2 and Has3 relative to levels expressed by normal prostate corresponded to elevated HA synthesis and avid BMEC adhesion. These results support a model in which tumor cells with up-regulated HA synthase expression assemble a cell surface hyaluronan matrix that promotes adhesion to bone marrow endothelial cells. This interaction could contribute to preferential bone metastasis by prostate carcinoma cells.


Subject(s)
Biomarkers, Tumor , Bone Marrow Cells/pathology , Glucuronosyltransferase/metabolism , Glycosyltransferases , Hyaluronic Acid/metabolism , Membrane Proteins , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Transferases , Xenopus Proteins , Cell Adhesion , Endothelium/pathology , Humans , Hyaluronan Synthases , Male , Neoplasm Metastasis , Tumor Cells, Cultured , Up-Regulation
4.
Am J Pathol ; 158(2): 639-46, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11159200

ABSTRACT

This study shows a strong correlation between the metastatic potentials of breast carcinoma cell lines and their ectopic expression of interleukin-8 (IL-8). Correlations exist for both constitutive and induced levels of IL-8 released. A correlation was also observed between cell morphology, metastatic potential, and IL-8 profile. Metastatic lines are fusiform in appearance, whereas, nonmetastatic lines are epithelioid. The metastatic potential of two breast carcinoma lines was examined using an orthotopic model of spontaneous metastasis. Metastatic cells formed rapidly growing, poorly differentiated primary tumors that metastasized. Nonmetastatic cells formed rapidly growing differentiated primary tumors that did not produce detectable metastases. Comparison of IL-8 expression by the parental cells and cell cultures developed from primary and metastatic tumors, demonstrates that IL-8 released by cultured cells from the primary tumor is higher than that of the parental cells, and IL-8 released by cultured cells derived from the metastatic lung tumors is greater than that released by cultured cells derived from the primary tumor. These data demonstrate a strong correlation between the metastatic phenotype of a cell and its IL-8 expression, suggesting a role for IL-8 in promoting the metastatic potential of breast tumor cells.


Subject(s)
Breast Neoplasms/metabolism , Interleukin-8/metabolism , Neoplasm Metastasis/pathology , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Enzyme-Linked Immunosorbent Assay , Female , Gene Expression Regulation, Neoplastic , Humans , Interleukin-8/genetics , Interleukin-8/physiology , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Nude , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transplantation, Heterologous , Tumor Cells, Cultured
5.
Neuroscience ; 100(3): 521-30, 2000.
Article in English | MEDLINE | ID: mdl-11098115

ABSTRACT

A major obstacle in neural transplantation is a severe loss of neurons in grafts soon after implantation. In the present study, we have investigated whether the systemic administration of synthetic fibronectin peptide V can increase the survival of neural grafts. Synthetic fibronectin peptide V is derived from the 33,000 mol. wt carboxyl-terminal heparin-binding domain of fibronectin. Previous studies have shown that these polypeptides possess anti-inflammatory properties. However, it is currently unknown whether this peptide has anti-apoptotic properties. Dissociated neural grafts were prepared from the ventral mesencephalon of pregnant Sprague-Dawley rats and were stereotaxically injected as a cell suspension into the striatum of adult Sprague-Dawley rats. A group of recipient rats received i.v. injections of peptide V (5mg/kg, dissolved in saline) at 24 and 4h prior to transplantation, at the time of transplantation, and 24, 48 and 72h post-transplantation. Saline-treated rats served as controls. The rats were killed at two, four and 42 days post-grafting and the brain tissue was immunologically processed for tyrosine-hydroxylase, major histocompatibility complex class I and class II antigens, complement receptor type 3 and leukocyte common antigen immunocytochemistry, and terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling assay. We found a significant increase (approximately twofold) in the number of dopamine neurons in the grafts for the peptide-treated group at four and 42 days compared with the controls. In contrast, there was no significant difference in the patterns of inflammation using different immunocytochemical markers in the two different groups. The levels of expression for these markers, however, were reduced over time. Interestingly, the number of apoptotic cells in the graft areas was significantly smaller in the peptide-treated group than in the control group two days after grafting. The results demonstrate that the systemic administration of synthetic fibronectin peptide V can dramatically increase the survival of nigral grafts in the brain and substantially reduce the number of apoptotic cells in the graft site, suggesting that this peptide may exert a beneficial effect on survival of nigral grafts through an anti-apoptotic mechanism.


Subject(s)
Corpus Striatum/surgery , Fetal Tissue Transplantation , Fibronectins/chemical synthesis , Graft Survival/drug effects , Substantia Nigra/embryology , Animals , Apoptosis , DNA Nucleotidylexotransferase/physiology , Histocompatibility Antigens Class I/metabolism , Histocompatibility Antigens Class II/metabolism , Immunohistochemistry , In Situ Nick-End Labeling , Leukocyte Common Antigens/metabolism , Macrophage-1 Antigen/metabolism , Male , Rats , Rats, Sprague-Dawley , Substantia Nigra/enzymology , Tyrosine 3-Monooxygenase/metabolism
6.
Nat Cell Biol ; 1(8): 507-13, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10587647

ABSTRACT

Melanoma chondroitin sulphate proteoglycan (MCSP) is a cell-surface antigen that has been implicated in the growth and invasion of melanoma tumours. Although this antigen is expressed early in melanoma progression, its biological function is unknown. MCSP can stimulate the integrin-alpha4 beta1-mediated adhesion and spreading of melanoma cells. Here we show that stimulated MCSP recruits tyrosine-phosphorylated p130 cas, an adaptor protein important in tumour cell motility and invasion. MCSP stimulation also results in a pronounced activation and recruitment of the Rho-family GTPase Cdc42. MCSP-induced spreading of melanoma cells is dependent upon active Cdc42, a Cdc42-associated tyrosine kinase (Ack-1) and tyrosine phosphorylation of p130cas. Furthermore, vectors inhibiting Ack-1 or Cdc42 expression and/or function abrogate MCSP-induced tyrosine phosphorylation and recruitment of p130cas. Our findings indicate that MCSP may modify tumour growth or invasion by a unique signal-transduction pathway that links Cdc42 activation to downstream tyrosine phosphorylation and subsequent cytoskeletal reorganization.


Subject(s)
Chondroitin Sulfate Proteoglycans/metabolism , Melanoma/metabolism , Melanoma/pathology , Phosphoproteins/metabolism , Protein-Tyrosine Kinases/metabolism , Proteins , cdc42 GTP-Binding Protein/metabolism , Cell Movement , Crk-Associated Substrate Protein , Enzyme Activation , Humans , Integrin alpha4beta1 , Integrins/metabolism , Melanoma/genetics , Mutation/genetics , Neoplasm Invasiveness , Phosphorylation , Phosphotyrosine/metabolism , Precipitin Tests , Protein Binding , Protein-Tyrosine Kinases/genetics , Receptors, Lymphocyte Homing/metabolism , Retinoblastoma-Like Protein p130 , Signal Transduction , Transfection , Tumor Cells, Cultured , cdc42 GTP-Binding Protein/genetics
7.
Prostate ; 41(2): 78-88, 1999 Oct 01.
Article in English | MEDLINE | ID: mdl-10477904

ABSTRACT

BACKGROUND: Metastasis of prostate carcinoma requires invasion through the basement membrane, a thin extracellular matrix that underlies the epithelial cells, which must be breached by tumor cells invading into surrounding tissue. The CXC-chemokines, which have been shown to promote the migration of neutrophils and carcinoma cells, are candidates to influence prostate carcinoma-cell invasion. METHODS: CXC-chemokines were examined for the ability to stimulate prostate cell line PC3 invasion in vitro through a reconstituted basement membrane and long-term migration and short-term adhesion to laminin, a major component of the basement membrane. RESULTS: PC3 cells responded to IL-8 and GROalpha with a 1. 6-2-fold increase in invasion through reconstituted basement membrane. A corresponding 2-3-fold increase in chemotaxis toward IL-8 and GROa was seen on laminin. Anti-CXCR2 antibody inhibited IL-8-stimulated migration. Expression levels of the beta(1) integrins were not changed by IL-8, and alpha(6beta1) integrin was used for both stimulated and baseline migration. In addition to the increases in migration and invasion, 2-6-fold transient increases in adhesion on laminin were seen with both IL-8 and GROalpha. CONCLUSIONS: These results suggest that the CXC-chemokines stimulate migration and invasion in part by altering the activation state of the beta(1) integrins. The CXC-chemokines act on prostate carcinoma cells through the CXCR2 receptor to promote behavior important for metastasis, and as such may be important in prostate carcinoma progression and metastasis.


Subject(s)
Chemokines, CXC/pharmacology , Chemotaxis , Neoplasm Invasiveness/physiopathology , Neoplasm Metastasis/physiopathology , Prostatic Neoplasms/pathology , Amino Acid Sequence , Basement Membrane , Cell Adhesion , Cell Movement/physiology , Humans , Interleukin-8/genetics , Interleukin-8/pharmacology , Laminin/physiology , Male , Molecular Sequence Data , Prostatic Neoplasms/physiopathology , Tumor Cells, Cultured
8.
J Biol Chem ; 273(10): 5955-62, 1998 Mar 06.
Article in English | MEDLINE | ID: mdl-9488735

ABSTRACT

We have previously reported that alpha4beta1 (but not alpha5beta1) integrin-mediated melanoma cell adhesion is inhibited by removal of cell surface chondroitin sulfate glycosaminoglycan (CSGAG), suggesting that melanoma chondroitin sulfate proteoglycan plays a role in modulating the adhesive function of alpha4beta1 integrin. In the current study, we demonstrated that alpha4beta1 integrin binds to CSGAG. We have identified a peptide from within alpha4 integrin termed SG1 (KKEKDIMKKTI) that binds to cell surface melanoma chondroitin sulfate proteoglycan, indicating that SG1 represents a CSGAG binding site within the alpha4 integrin subunit. Soluble SG1 inhibits alpha4beta1 integrin-mediated human melanoma cell adhesion to CS1. Polyclonal antibody generated against the peptide inhibits melanoma cell adhesion to CS1, and the inhibition is reversed by Mn2+ and an activating monoclonal antibody anti-beta1 (8A2). Additionally, pretreatment of cells with anti-SG1 IgG inhibits the expression of the monoclonal antibody 15/7 epitope in the presence of soluble CS1 peptide, suggesting that anti-SG1 IgG prevents ligand binding by alpha4beta1 integrin. These results demonstrate that alpha4beta1 integrin interacts directly with CSGAG through SG1 site, and that this site can affect the ligand binding properties of the integrin.


Subject(s)
Cell Adhesion/physiology , Chondroitin Sulfates/physiology , Integrins/chemistry , Melanoma/metabolism , Proteoglycans/physiology , Receptors, Lymphocyte Homing/chemistry , Antibodies/immunology , Antibodies/pharmacology , Binding Sites/physiology , Cell Adhesion/drug effects , Humans , Integrin alpha4beta1 , Integrins/physiology , Manganese/pharmacology , Peptide Fragments/chemistry , Protein Binding/physiology , Receptors, Lymphocyte Homing/physiology , Tumor Cells, Cultured
10.
J Med Chem ; 40(19): 3077-84, 1997 Sep 12.
Article in English | MEDLINE | ID: mdl-9301671

ABSTRACT

Integrin-mediated tumor cell adhesion to type IV collagen is believed to play a role in the invasion of basement membrane proteins and the subsequent metastatic process. The cellular protein CAR (cell adhesion regulator) has been proposed to influence integrin-mediated binding to extracellular matrix proteins, including basement membrane (type IV) collagen. Three analogs of the CAR138-142 have been tested for activity. The first contains the 138-142 sequence (CAR138-142, Val-Glu-Ile-Leu-Tyr-NH2), the second contains the 138-142 sequence with a phosphorylated Tyr [pCAR138-142, Val-Glu-Ile-Leu-Tyr(PO3H2)-NH2], and the third contains the reversed 138-142 sequence (rCAR138-142, Tyr-Leu-Ile-Glu-Val-NH2). When added extracellularly, none of the analogs had a significant affect on cell adhesion to type IV collagen. Using a novel reversible cell permeabilization method, we found that intracellular incorporation of both CAR138-142 and pCAR138-142 resulted in inhibition of cell adhesion in a dose-dependent fashion. The IC50 values were approximately 90 and approximately 10 microM for CAR138-142 and pCAR138-142, respectively. Intracellular incorporation of the rCAR138-142 peptide had no affect on cell adhesion. Fluorescence microscopy of a fluorescein-labeled CAR138-142 peptide revealed that the reversible permeabilization procedure resulted in the peptides crossing the cell membrane. Affinity chromatography of melanoma cell lysates with pCAR138-142 or rCAR138-142 attached to a solid support of magnetic beads suggested that one protein was bound uniquely by pCAR138-142. Immunoprecipitation analysis identified vinculin, a protein associated with the actin cytoskeleton, as the protein specifically bound by pCAR138-142. Immunoprecipitation with pp125FAK- or beta 1-integrin-derived mAbs gave negative results. Our study suggests that a possible therapeutic approach for inhibition of melanoma cell adhesion adhesion to extracellular matrix proteins is the use of CAR peptide analogs intracellularly.


Subject(s)
Cell Adhesion Molecules/chemistry , Cell Adhesion/drug effects , Collagen , Peptide Fragments/chemical synthesis , Peptide Fragments/pharmacology , ATPases Associated with Diverse Cellular Activities , Amino Acid Sequence , Basement Membrane , Cell Adhesion/physiology , Cell Membrane Permeability , Humans , Kinetics , Melanoma , Metalloendopeptidases , Neoplasm Metastasis , Oligopeptides/chemical synthesis , Oligopeptides/chemistry , Oligopeptides/pharmacology , Peptide Fragments/chemistry , Structure-Activity Relationship , Tumor Cells, Cultured
11.
J Cereb Blood Flow Metab ; 17(6): 605-11, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9236717

ABSTRACT

Postischemic cerebral inflammation has been reported to contribute to ischemic brain damage. During inflammation, constituents of the extracellular matrix such as fibronectin and laminin are recognized by certain integrins or proteoglycans and play an important role in the cell adhesion process. The purpose of this study was to evaluate the efficacy of peptides derived from laminin on leukocyte accumulation, infarct size, and neurological outcome in rats subjected to 1 h of cerebral ischemia and 48 h of reperfusion. Forty-four animals were included in this study: transient ischemia without treatment (Group I), treatment with TG-1 peptide (Group II), GD-1 peptide (Group III), and GD-6 peptide (Group IV). Group II showed a significant reduction of the leukocyte accumulation (p < 0.001) and infarct size (p = 0.015) when compared with Group I. The neurological grade of Group II was also significantly better than in Group I at 48 h after reperfusion (p = 0.012). Based on these data, which are the first to explore the therapeutic potential of this peptide in cerebral ischemia, laminin peptide may offer a novel therapeutic approach to allaying injury in ischemic stroke.


Subject(s)
Ischemic Attack, Transient/therapy , Laminin/pharmacology , Leukocyte Count/drug effects , Peptide Fragments/pharmacology , Amino Acid Sequence , Animals , Cerebral Cortex/enzymology , Inflammation/therapy , Ischemic Attack, Transient/pathology , Ischemic Attack, Transient/physiopathology , Male , Peptide Fragments/genetics , Peroxidase/drug effects , Rats , Rats, Sprague-Dawley
12.
Neurosurgery ; 40(3): 557-63; discussion 563-4, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9055296

ABSTRACT

OBJECTIVE: Activated polymorphonuclear leukocytes (PMNs) seem to be directly involved in potentiating ischemic brain injury. Recent work in our laboratory demonstrated that synthetic fibronectin peptides significantly inhibit PMN accumulation in ischemic tissue, reduce the size of infarction, and reduce neurological dysfunction after transient focal cerebral ischemia in rats. The purpose of this study was to examine any dose-related effects (Experiment 1) and the optimal timing of the administration (Experiment 2) of synthetic fibronectin peptide V (FN-C/H-V) to further substantiate the role of the peptide in ameliorating cerebral ischemic damage. METHODS: Fifty-six animals were included in the study. We evaluated the efficacy of FN-C/H-V on PMN accumulation in ischemic tissue, infarct size, and neurological outcomes in rats subjected to 1 hour of cerebral ischemia and 48 hours of reperfusion. RESULTS: In Experiment 1, the animals receiving FN-C/H-V at a dose of 10 to 15 mg/kg of body weight per injection showed significant reduction of PMN accumulation, reduction of infarct size, and improvement of neurological outcomes at 48 hours after reperfusion compared to untreated animals (P < 0.05). In Experiment 2, the animals receiving FN-C/H-V within 3 hours after reperfusion also showed significantly better results than untreated animals (P < 0.05). Despite the treatment delay, the administration of FN-C/H-V inhibited PMN accumulation after reperfusion but did not reduce the size of infarction when administered 6 hours after reperfusion. CONCLUSION: These data suggest that relatively late postischemic administration of FN-C/H-V is effective in brain protection after ischemia/reperfusion.


Subject(s)
Fibronectins/pharmacology , Ischemic Attack, Transient/immunology , Leukocyte Adherence Inhibition Test , Peptide Fragments/pharmacology , Animals , Brain Damage, Chronic/immunology , Cerebral Infarction/immunology , Dose-Response Relationship, Drug , Leukocyte Count/drug effects , Leukocytes/drug effects , Leukocytes/immunology , Male , Neurologic Examination/drug effects , Peroxidase/metabolism , Rats , Rats, Sprague-Dawley , Reperfusion Injury/immunology
13.
Biochemistry ; 36(49): 15404-10, 1997 Dec 09.
Article in English | MEDLINE | ID: mdl-9398270

ABSTRACT

Type IV collagen promotes integrin-mediated cell adhesion, spreading, and motility. Several regions within the triple-helical domain of type IV collagen have been identified as tumor cellular recognition sites. Among these regions, the alpha1(IV)531-543 sequence, designated L-Hep-III, promotes integrin-mediated tumor cell adhesion and directly binds to the alpha3beta1 integrin [Miles, A. J., et al. (1994) J. Biol. Chem. 269, 30939-30945; Miles, A. J., et al. (1995) J. Biol. Chem. 270, 29047-29050]. We have presently compared the activities of the all-d enantiomeric peptide model of alpha1(IV)531-543, designated D-Hep-III, with L-Hep-III, for promoting the adhesion, spreading, and motility of metastatic melanoma and breast carcinoma cells. D-Hep-III was found to support melanoma and breast carcinoma cell adhesion, spreading, and motility in a dose-dependent fashion similar to that of L-Hep-III. The adhesions of melanoma and breast carcinoma cells to both type IV collagen and fibronectin were effectively inhibited by L-Hep-III and D-Hep-III. Melanoma cell invasion of the basement membrane was also inhibited by D-Hep-III. Characterization of the cell surface receptor for D-Hep-III was acheived via cell adhesion assays and affinity chromatography using monoclonal antibodies against integrin subunits. Immunoprecipitation analysis following EDTA elution from a D-Hep-III affinity column indicated that D-Hep-III binds to the alpha3beta1 integrin but not to the alpha2 or alpha6 integrin subunits. In summary, these studies demonstrate that an all-D model of the alpha1(IV)531-543 sequence mimics the biological activities of the all-L peptide. D-Hep-III is the first all-D peptide that has been shown to promote tumor cell adhesion, spreading, and migration, inhibit tumor cell adhesion and migration on type IV collagen and invasion of the basement membrane, and bind directly to an integrin. Due to the resistance to proteolysis, all-D receptor-binding peptides such as D-Hep-III have great potential for in vivo studies and as therapeutic agents.


Subject(s)
Breast Neoplasms/metabolism , Collagen/chemistry , Integrins/metabolism , Melanoma/metabolism , Peptide Fragments/metabolism , Antibodies, Monoclonal/immunology , Breast Neoplasms/pathology , Cell Adhesion , Cell Movement , Cell Separation , Chromatography, Affinity , Humans , Integrin alpha3beta1 , Integrins/immunology , Melanoma/pathology , Models, Chemical , Peptide Fragments/chemistry , Protein Binding , Tumor Cells, Cultured
14.
J Cereb Blood Flow Metab ; 16(6): 1120-5, 1996 Nov.
Article in English | MEDLINE | ID: mdl-8898683

ABSTRACT

Leukocytes play an important role in the development of ischemia/reperfusion injury. Recent work in our laboratory has demonstrated that a mixture of synthetic fibronectin peptides to leukocyte adhesion molecules reduces ischemic brain damage after transient focal cerebral ischemia. The purpose of this study was to evaluate the efficacy of the individual peptides on leukocyte accumulation, infarct size, and neurological outcome in rats subjected to 1 h of cerebral ischemia and 48 h of reperfusion. Thirty-five animals were divided into five groups: transient ischemia without treatment (Group I), treatment with arginyl-glycyl-aspartic acid (RGD) peptide (Group II), connecting segment (CS)-1 peptide (Group III), fibronectin (FN)-C/H-V peptide (Group IV), and scrambled FN-C/H-V peptide (Group V). Groups III and IV showed a significant decrease in the degree of leukocyte infiltration in the lesion and in the infarct size (p < 0.05) when compared to Groups I, II, and V. The neurological grade of Groups III and IV was significantly better than in Groups I, II, and V at 48 h after reperfusion (p < 0.01). Thus, in addition to demonstrating the potential efficacy of synthetic peptides as therapeutic agents for ischemia-reperfusion, these results also offer new insights into the mechanisms of leukocyte arrest and recruitment in ischemia/reperfusion injury.


Subject(s)
Brain Ischemia/drug therapy , Cell Adhesion Molecules/metabolism , Fibronectins/pharmacology , Leukocytes/pathology , Peptides/pharmacology , Animals , Brain Ischemia/pathology , Cell Adhesion/drug effects , Fibronectins/chemistry , Leukocytes/drug effects , Male , Neurons/drug effects , Neurons/pathology , Peptides/chemistry , Rats , Rats, Sprague-Dawley
15.
J Biomed Mater Res ; 32(2): 237-42, 1996 Oct.
Article in English | MEDLINE | ID: mdl-8884501

ABSTRACT

Based on the premise of achieving blood compatibility through mimicking the chemical constitutents of the biologically insert surface of the unactivated platelet membrane, a process was developed that entails the covalent grafting of modified phosphatidylcholine molecules to materials including silica, polypropylene, and polytetrafluoroethylene (PTFE) polymer films. These materials were characterized using x-ray photoelectron spectroscopy (XPS) and contactangle measurements. The phosphatidylcholine-containing materials (PC materials) were used as substrates in the plateletadhesion assays and were subjected to enzymatic degradation evaluation. Phosphatidylcholine-grafted silica materials do not support platelet adhesion. In addition the number of adherent platelets correlate with the amount of grafted phospholipid present, as indicated by the phosphorus/ carbon ratio obtained by XPS analysis. Platelet adhesion to phosphatidylcholine-grafted polypropylene and PTFE was inhibited 80% and 90%, respectively, when compared with platelet adhesion to unmodified polypropylene and PTFE.


Subject(s)
Biocompatible Materials , Blood Platelets/physiology , Phosphatidylcholines , Platelet Adhesiveness , Polypropylenes , Polytetrafluoroethylene , Silicon Dioxide , Cell Membrane/physiology , Humans , In Vitro Techniques , Spectrometry, X-Ray Emission , Type C Phospholipases
16.
J Neurosurg ; 85(1): 125-30, 1996 Jul.
Article in English | MEDLINE | ID: mdl-8683261

ABSTRACT

Leukocytes play an important role in the development of ischemia-reperfusion injury. This study was conducted to ascertain whether synthetic peptides corresponding to the cell- and heparin-binding sequences of fibronectin that disturb leukocyte adhesion molecules were effective in neuronal protection after transient focal cerebral ischemia in rats. The authors evaluated the efficacy of peptides on infarction size, leukocyte infiltration in the ischemic tissue, and neurological outcome in rats subjected to 1 hour of cerebral ischemia and 48 hours of reperfusion. Twenty-one animals were divided into three groups: transient ischemia without treatment (Group I), transient ischemia with administration of vehicle (Group II), and transient ischemia with administration of fibronectin peptides (Group III). The mean myeloperoxidase activity (U/g wet wt) in the ischemic area was as follows: Group I, 0.19% +/- 0.05; Group II, 0.21% +/- 0.03; and Group III, 0.08% +/- 0.02. The mean size of the infarction as a percentage of the total hemispheric volume was as follows: Group I, 38.35% +/- 1.34%; Group II, 39.21% +/- 2.42%; and Group III, 25.81% +/- 4.87%. Group III showed a significant decrease in myeloperoxidase activity in the lesion and the infarction size was smaller when compared to Groups I and II (p < 0.05). The neurological grade in Group III was significantly better than in Groups I and II at 48 hours after reperfusion (p < 0.01). This study is the first to explore the therapeutic potential of synthetic fibronectin peptides in brain protection after transient focal ischemia, and the results also serve as a basis for studies of important cellular and molecular events that contribute to tissue damage.


Subject(s)
Brain Ischemia/metabolism , Cerebral Arteries/metabolism , Fibronectins/biosynthesis , Leukocytes/metabolism , Animals , Male , Rats , Rats, Sprague-Dawley , Time Factors
17.
Semin Cancer Biol ; 7(3): 155-62, 1996 Jun.
Article in English | MEDLINE | ID: mdl-8773301

ABSTRACT

Tumor cell invasion and metastasis is highly dependent on dynamic changes in the adhesion and migration of transformed and malignant cells. As with normal cell adhesion, the adhesion of tumor cells influences their cytoskeletal organization, activation of signal transduction pathways within the cell, and nuclear events leading to changes in mRNA transcription and protein synthesis. Furthermore, as tumor cells invade the circulation, they adhere to activated endothelial cells at sites within the vasculature during arrest and extravasation. Studies in the area of tumor cell adhesion and migration have demonstrated that the recognition of extracellular matrix ligands, or adhesion promoting ligands expressed on neighboring cells (i.e. counter-receptors), involves complex molecular recognition mechanisms. The complexity arises, in part, from the multiple recognition sites that are present within adhesion promoting ligands. Some of these structures within ECM components act by binding integrins, whereas others bind additional receptors such as cell surface proteoglycans. In this sense, adhesion promoting ligands may be considered as informational arrays, that function to modulate cell phenotype by engaging specific combinations of adhesion receptors on the cell surface. Understanding the mechanism(s) by which these receptor 'cluster' modify cell adhesion, motility and growth may lead to novel therapeutic strategies to control tumor cell invasion and metastasis formation. This review will highlight the role that cell surface chondroitin sulfate proteoglycans may play in modulating tumor cell adhesion, migration and invasion, with an emphasis on the relationship between cell surface chondroitin sulfate proteoglycans and integrins.


Subject(s)
Cell Adhesion/physiology , Cell Movement/physiology , Chondroitin/biosynthesis , Proteoglycans/biosynthesis , Humans
18.
J Biol Chem ; 271(21): 12234-40, 1996 May 24.
Article in English | MEDLINE | ID: mdl-8647820

ABSTRACT

The dissection of the activities mediated by type I collagen requires an approach by which the influence of triple-helical conformation can be evaluated. The alpha 1 beta 1 and alpha 2 beta 1 integrin binding sites within type I collagen are dependent upon triple-helical conformation and contained within residues 14-822 from alpha 1(I). Seven alpha 1(I)-derived triple-helical peptides (THPs) were synthesized based on charge clustering (alpha 1(I)256-270, alpha 1(I)385-396, alpha 1(I)406-417, alpha 1(I)415-423, alpha 1(I)448-456, alpha 1(I)496-507, and alpha 1(I)526-537). Three additional THPs were synthesized (alpha 1(I)85-96, alpha 1(I)433-441, and alpha 1(I)772-786) based on previously described or proposed activities (Kleinman, H. K., McGoodwin, E.B., Martin, G. R., Klebe, R. J., Fietzek, P. P., and Wooley, D. E. (1978) J. Biol. Chem. 253, 5642-5646; Staatz, W. D., Foik, K. F., Zutter, M. M., Adams, S. P., Rodriquez, B. A., and Santoro, S. A. (1991) J. Biol. Chem. 266, 7363-7367; San Antonio, J. D., Lander, A. D., Karnovsky, M. J., and Slayter, H. S. (1994) J. Cell Biol. 125, 1179-1188). Of the ten THPs, alpha 1(I)772-786 THP had the greatest activity, with half-maximal normal dermal fibroblast adhesion occurring at a peptide concentration of 1.6 microM. Triple-helicity was essential for activity of this sequence, as the non-triple-helical peptide analog (alpha 1(I)772-786 SSP) exhibited considerably lower levels of cell adhesion promotion even at peptide concentrations as high as 100 microM. Within the sequence itself, residues 784-786 (Gly-Leu-Hyp) were important for cellular recognition, as the alpha 1(I)772-783 THP had greatly reduced cell adhesion activity compared with alpha 1(I)772-786 THP. Preliminary studies indicate that the beta 1 integrin subunit mediates fibroblast adhesion to alpha 1(I)772-786 THP. The identification of fibroblast integrin binding sites within type I collagen may have important implications for understanding collagen metabolism.


Subject(s)
Cell Adhesion/drug effects , Collagen/metabolism , Peptides/pharmacology , Amino Acid Sequence , Collagen/chemistry , Fibroblasts/cytology , Fibroblasts/drug effects , Humans , Integrin alpha1beta1 , Integrins/metabolism , Molecular Sequence Data , Protein Conformation , Receptors, Collagen
19.
J Surg Res ; 61(2): 367-72, 1996 Mar.
Article in English | MEDLINE | ID: mdl-8656610

ABSTRACT

Lovastatin, a 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitor, has been shown to inhibit the in vitro and in vivo growth of a number of different cell lines. However, the mechanism by which lovastatin exerts its effect is not clear. In this experiment, we investigated the effect of lovastatin on the incorporation of [3H]thymidine by Hepatoma Tissue Culture-4 (HTC-4) and Lewis Lung Carcinoma L-1 (LLC-L1) tumor cells. Tumor cells were grown under standard conditions and treated with four different concentrations of lovastatin. Cell growth was evaluated by daily hemacytometer cell counts. On Day 4, the plates were pulsed with 10 microCi [3H]thymidine. After 24 hr, the plates were harvested and [3H]thymidine incorporation was measured by scintillation counting. Lovastatin inhibited both HTC-4 and LLC-L1 cell growth in a dose-dependent manner. At the highest lovastatin dose, both LLC-L1 and HTC-4 cell growth was slowed to less than 15% of control. Remarkably, however, both cell lines showed a paradoxical, dose related, increase in [3H]thymidine uptake. Cell cycle analysis using flow cytometry was performed on Day 5 in the LLC-L1 cell line. As the lovastatin concentration increased, a lower percentage of cells was found in the G1 phase of the cell cycle and a higher percentage of cells was found in the S and G2 phases. These findings suggest that these tumor cells are undergoing brisk DNA repair or that lovastatin is more effectively blocking cell division than cellular DNA replication.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors , Lovastatin/pharmacology , Thymidine/metabolism , Animals , Cell Division/drug effects , DNA/biosynthesis , Tumor Cells, Cultured
20.
J Biol Chem ; 270(49): 29047-50, 1995 Dec 08.
Article in English | MEDLINE | ID: mdl-7493922

ABSTRACT

Tumor cell adhesion to the triple-helical domain of basement membrane (type IV) collagen occurs at several different regions. Cellular recognition of the sequence spanning alpha 1(IV)531-543 has been proposed to be independent of triple-helical conformation (Miles, A. J., Skubitz, A. P. N., Furcht, L. T., and Fields, G. B. (1994) J. Biol. Chem. 269, 30939-30945). In the present study, integrin interactions with a peptide analog of the alpha 1(IV)-531-543 sequence have been analyzed. Tumor cell adhesion (melanoma, ovarian carcinoma) to the alpha 1(IV)531-543 chemically synthesized peptide was inhibited by a monoclonal antibody against the alpha 3 integrin subunit, and to a lesser extent by monoclonal antibodies against the beta 1 and alpha 2 integrin subunits. An anti-alpha 5 monoclonal antibody and normal mouse IgG were ineffective as inhibitors of tumor cell adhesion to the peptide. Two cell surface proteins of 120 and 150 kDa bound to an alpha 1(IV)531-543 peptide affinity column and were eluted with 20 mM EDTA. When the eluted proteins were incubated with monoclonal antibodies against either the alpha 3 or beta 1 integrin subunit, proteins corresponding in molecular weight to alpha 3 and beta 1 integrin subunits were precipitated. No proteins were immunoprecipated with monoclonal antibodies against the alpha 2 or alpha 5 integrin subunits. Thus, the alpha 3 beta 1 integrin from two tumor cell types has been shown to bind directly to the alpha 1 (IV)531-543 peptide. The alpha 1(IV)531-543 peptide is the first collagen-like sequence that has been shown to bind the alpha 3 beta 1 integrin.


Subject(s)
Collagen/metabolism , Integrins/metabolism , Membrane Proteins/metabolism , Peptide Fragments/metabolism , Amino Acid Sequence , Animals , Basement Membrane/chemistry , Binding Sites , Cell Adhesion , Collagen/chemistry , Humans , Integrin alpha3beta1 , Mice , Molecular Sequence Data , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL