151
|
Liu S, Schubert RL, Bugge TH, Leppla SH. Anthrax toxin: structures, functions and tumour targeting. Expert Opin Biol Ther 2003; 3:843-53. [PMID: 12880383 DOI: 10.1517/14712598.3.5.843] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Anthrax toxin, the major virulence factor of Bacillus anthracis, consists of three polypeptides: protective antigen (PrAg), lethal factor (LF) and oedema factor (EF). To intoxicate mammalian cells, PrAg binds to its cellular receptors and is subsequently activated via proteolysis, yielding a carboxyl-terminal fragment which coordinately assembles to form heptamers that bind and translocate LF and EF into the cytosol to exert their cytotoxic effects. Substantial progress has been made in recent years towards the characterisation of the structure and function of anthrax toxin, and this has greatly facilitated rational drug design of antianthrax agents. There is also emerging evidence that toxins can be manipulated for cancer therapy. LF can efficiently inactivate several mitogen-activated protein kinase kinases (MAPKKs) via cleavage of their amino-terminal sequences. Consequently, antitumour effects of wild type lethal toxin were observed after treatment of mitogen-activated protein kinase (MAPK)-dependent tumours such as human melanomas. Modification of the toxin's proteolytic activation site limits its cytotoxicity to certain cell types and creates a versatile method of treatment. One approach that has successfully achieved specific tumour targeting is the alteration of the furin cleavage of PrAg so that it is not activated by furin, but, alternatively, by proteases that are highly expressed by tumour tissues, including matrix metalloproteases and urokinase.
Collapse
Affiliation(s)
- Shihui Liu
- Microbial Pathogenesis Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| | | | | | | |
Collapse
|
152
|
Hjortland GO, Bjørnland K, Pettersen S, Garman-Vik SS, Emilsen E, Nesland JM, Fodstad O, Engebraaten O. Modulation of glioma cell invasion and motility by adenoviral gene transfer of PAI-1. Clin Exp Metastasis 2003; 20:301-9. [PMID: 12856717 DOI: 10.1023/a:1024040718238] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
A number of studies have emphasized the role of PAI-1 as an important regulator of tumor cell invasion and metastasis. The hallmark of primary tumors of the central nervous system and glioblastomas in particular is the diffuse invasion into the normal brain tissue. Since PAI-1 is expressed in such tumors, we studied the effect of adenoviral-mediated transfer of the PAI-1 gene in regulating the in vitro invasiveness of D54Mg glioma cells into Matrigel, and into fetal rat brain aggregates. Treatment of D54Mg cells with 50 MOI (multiplicity of infection) of the replication defective vector AdCMVPAI-1 increased PAI-1 expression 23-fold compared to control vectors, and the invasion through Matrigel was reduced by 67%. The motility of the cells was reduced by 58% compared to controls (indicating that inhibition of motility was the principal effect of PAI-1 in these cells). The ability of D54Mg tumor spheroids to invade fetal rat brain aggregates was not reduced by the PAI-1 gene transfer. The results show that overexpression of PAI-1 can inhibit glioma cell motility and invasion through extracellular matrix (ECM) components, like laminin and collagen, but does not inhibit tumor cell invasion in a three-dimensional invasion assay, simulating normal brain tissue having a different ECM and interstitial composition. The different results obtained in the two invasion assays reflect the complex biological effects of the uPA/PAI-1 system, and questions a simplistic view of PAI- I as an inhibitor of brain tumor invasion.
Collapse
Affiliation(s)
- G O Hjortland
- Department of Tumor Biology, Institute for Cancer Research, The Norwegian Radium Hospital, University of Oslo, Montebello, Oslo, Norway.
| | | | | | | | | | | | | | | |
Collapse
|
153
|
Ahmed N, Oliva K, Wang Y, Quinn M, Rice G. Downregulation of urokinase plasminogen activator receptor expression inhibits Erk signalling with concomitant suppression of invasiveness due to loss of uPAR-beta1 integrin complex in colon cancer cells. Br J Cancer 2003; 89:374-84. [PMID: 12865932 PMCID: PMC2394266 DOI: 10.1038/sj.bjc.6601098] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Cancer invasion is regulated by cell surface proteinases and adhesion molecules. Interaction between specific cell surface molecules such as urokinase plasminogen activator receptor (uPAR) and integrins is crucial for tumour invasion and metastasis. In this study, we examined whether uPAR and beta1 integrin form a functional complex to mediate signalling required for tumour invasion. We assessed the expression of uPAR/beta1 integrin complex, Erk signalling pathway, adhesion, uPA and matrix metalloproteinase (MMP) expression, migration/invasion and matrix degradation in a colon cancer cell line in which uPAR expression was modified. Antisense inhibition of the cell surface expression of uPAR by 50% in human colon carcinoma HCT116 cells (A/S) suppressed Erk-MAP kinase activity by two-fold. Urokinase plasminogen activator receptor antisense treatment of HCT116 cells was associated with a 1.3-fold inhibition of adhesion, approximately four-fold suppression of HMW-uPA secretion and inhibition of pro-MMP-9 secretion. At a functional level, uPAR antisense resulted in a four-fold decline in migration/invasion and abatement of plasmin-mediated matrix degradation. In empty vector-transfected cells (mock), uPA strongly elevated basal Erk activation. In contrast, in A/S cells, uPA induction of Erk activation was not observed. Urokinase plasminogen activator receptor associated with beta1 integrin in mock-transfected cells. Disruption of uPAR-beta1 integrin complex in mock-transfected cells with a specific peptide (P25) inhibited uPA-mediated Erk-MAP kinase pathway and inhibited migration/invasion and plasmin-dependent matrix degradation through suppression of pro-MMP-9/MMP-2 expression. This novel paradigm of uPAR-integrin signalling may afford opportunities for alternative therapeutic strategies for the treatment of cancer.
Collapse
Affiliation(s)
- N Ahmed
- Department of Obstetrics and Gynaecology, Gynaecological Cancer Research Centre, Royal Women's Hospital, University of Melbourne, Melbourne 3053, Australia.
| | | | | | | | | |
Collapse
|
154
|
Pierleoni C, Castellucci M, Kaufmann P, Lund LR, Schnack Nielsen B. Urokinase receptor is up-regulated in endothelial cells and macrophages associated with fibrinoid deposits in the human placenta. Placenta 2003; 24:677-85. [PMID: 12828926 DOI: 10.1016/s0143-4004(03)00082-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Clearance of fibrin deposits within the human placenta is an ongoing process during normal placental development. Plasminogen is a circulating fibrinolytic protease zymogen activated in situ by plasminogen activators. We have previously reported that the receptor for urokinase plasminogen activator (uPAR) is expressed by cells either covering or enmeshed within the perivillous fibrinoid deposits. Whereas these cells seemed likely to be trophoblasts, a definitive identification was lacking, and this question is central to the understanding of the cellular mechanisms directing fibrinolysis in the placenta. In this study we have performed immunohistochemical co-localization studies and found that the uPAR-positive cells covering fibrinoid deposits are immunoreactive for CD31 and vWF, indicating that they are actually endothelial cells. In addition, we found that perivillous fibrinoid deposits not covered with uPAR-positive endothelial cells were covered with platelets identified by integrin alpha(IIb)beta(3)-immunoreactivity. Also surprisingly, the uPAR-positive cells enmeshed within fibrinoid deposits express a cell specific marker indicating that they are macrophages. Both uPAR-positive cell populations also express uPA immunoreactivity. Taken together, the data suggest that both fibrinoid-covering endothelial cells and fibrinoid-enmeshed macrophages can participate in the clearance process of perivillous fibrinoid deposits formed in the human placenta.
Collapse
Affiliation(s)
- C Pierleoni
- Institute of Normal Human Morphology, Faculty of Medicine, University of Ancona, Italy
| | | | | | | | | |
Collapse
|
155
|
Pedersen TX, Leethanakul C, Patel V, Mitola D, Lund LR, Danø K, Johnsen M, Gutkind JS, Bugge TH. Laser capture microdissection-based in vivo genomic profiling of wound keratinocytes identifies similarities and differences to squamous cell carcinoma. Oncogene 2003; 22:3964-76. [PMID: 12813470 DOI: 10.1038/sj.onc.1206614] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Keratinocytes undergo a dramatic phenotypic conversion during reepithelialization of skin wounds to become hyperproliferative, migratory, and invasive. This transient healing response phenotypically resembles malignant transformation of keratinocytes during squamous cell carcinoma progression. Here we present the first analysis of global changes in keratinocyte gene expression during skin wound healing in vivo, and compare these changes to changes in gene expression during malignant conversion of keratinized epithelium. Laser capture microdissection was used to isolate RNA from wound keratinocytes from incisional mouse skin wounds and adjacent normal skin keratinocytes. Changes in gene expression were determined by comparative cDNA array analyses, and the approach was validated by in situ hybridization. The analyses identified 48 candidate genes not previously associated with wound reepithelialization. Furthermore, the analyses revealed that the phenotypic resemblance of wound keratinocytes to squamous cell carcinoma is mimicked at the level of gene expression, but notable differences between the two tissue-remodeling processes were also observed. The combination of laser capture microdissection and cDNA array analysis provides a powerful new tool to unravel the complex changes in gene expression that underlie physiological and pathological remodeling of keratinized epithelium.
Collapse
Affiliation(s)
- Tanja Xenia Pedersen
- Proteases and Tissue Remodeling Unit, Oral & Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Room 211, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
156
|
Takai N, Miyazaki T, Nishida M, Shang S, Nasu K, Miyakawa I. Clinical relevance of Elf-1 overexpression in endometrial carcinoma. Gynecol Oncol 2003; 89:408-13. [PMID: 12798703 DOI: 10.1016/s0090-8258(03)00131-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
OBJECTIVES Elf-1 is a member of the Ets transcription factor family that regulates the genes involved in cellular growth and differentiation. Enhanced expression of Elf-1 has been reported in prostate cancer, breast cancer, and osteosarcoma. METHODS To elucidate the involvement of Elf-1 in endometrial carcinogenesis, we analyzed serial frozen tissue sections from 31 patients with endometrial carcinoma and 20 patients with normal endometria for Elf-1 protein expression, using fluorescent immunohistochemistry. We analyzed the relationship between the percentages of Elf-1-stained cells and patient characteristics, including clinical stage, histological grade, presence of invasion to greater than one-half the myometrium, clinical outcome, and survival rate. RESULTS Elf-1 was weakly detected in some normal endometria in the proliferative phase (0-18.9%) and other normal endometria in the secretory phase (0-28.5%). There was, however, abundant Elf-1 immunoreactivity in the nucleus of the endometrial carcinoma cells along with a little cytoplasmic staining. Scoring on the basis of the percentage of nuclear-positive cells indicated that nuclear Elf-1 expression was significantly associated with PCNA-labeling index, clinical stage, histological grade, the presence of invasion to greater than one-half the myometrium, and clinical outcome (P < 0.01, respectively). Survival data were available for all patients and demonstrated that Elf-1 expression was significantly associated with poor prognosis (P < 0.01). CONCLUSIONS Our results demonstrate that Elf-1 expression in endometrial carcinoma correlates with the malignant potential of this tumor.
Collapse
Affiliation(s)
- Noriyuki Takai
- Department of Obstetrics and Gynecology, Oita Medical University, Oita 879-5593, Japan.
| | | | | | | | | | | |
Collapse
|
157
|
Elner SG, Elner VM, Kindzelskii AL, Horino K, Davis HR, Todd RF, Glagov S, Petty HR. Human RPE cell lysis of extracellular matrix: functional urokinase plasminogen activator receptor (uPAR), collagenase and elastase. Exp Eye Res 2003; 76:585-95. [PMID: 12697422 DOI: 10.1016/s0014-4835(03)00028-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Age-related macular degeneration (ARMD), proliferative vitreoretinopathy (PVR) and uveitis are characterized by RPE motility through the ECM of retinal lesions. The purpose of this study was to test the hypothesis that multiple proteolytic systems are functionally intact at the HRPE surface and peri-cellular region and that these activities are differentially modulated by IL-1beta. HRPE cells were evaluated: (1). as individual cells or cell extracts, (2). during migration across three-dimensional ECM-like layers and (3). in tissue sections. The urokirase plasminogen activator receptor (uPAR; CD87) was detected on HRPE cells as well as its functional activity. Although uPAR was associated with CD11b (CR3) on live resting cells, polarized migratory HRPE cells were found to dissociate uPAR from CR3; uPAR then translocated to anterior pole of the cell, where it enhanced PAI-1-inhibitable local proteolytic activity. The relative contribution of uPAR and collagenase in HRPE migration was evaluated using three-dimensional gelatin matrices. Interestingly, uPAR/uPA was found to play a key role in migration across these layers. IL-1 upregulated uPAR, collagenase, and elastase activities, suggesting that cytokines may affect the invasive program of HRPE cells in vivo. Immunohistochemistry for uPAR was performed in sections of human retina. Immunoreactive uPAR was present along the HRPE basolateral membrane in retinal sections and in sections of diseased retinal tissue at an enhanced level. Our results suggest that multiple proteolytic systems are present in association with HRPE and that the uPAR/uPA system may be particularly important.
Collapse
Affiliation(s)
- Susan G Elner
- Department of Ophthalmology, University of Michigan, Kellogg Eye Center, Ann Arbor, MI 48105, USA
| | | | | | | | | | | | | | | |
Collapse
|
158
|
Behrendt N, List K, Andreasen PA, Danø K. The pro-urokinase plasminogen-activation system in the presence of serpin-type inhibitors and the urokinase receptor: rescue of activity through reciprocal pro-enzyme activation. Biochem J 2003; 371:277-87. [PMID: 12534347 PMCID: PMC1223308 DOI: 10.1042/bj20021508] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2002] [Revised: 01/16/2003] [Accepted: 01/20/2003] [Indexed: 11/17/2022]
Abstract
The reciprocal pro-enzyme activation system of plasmin, urokinase-type plasminogen activator (uPA) and their respective zymogens is a potent mechanism in the generation of extracellular proteolytic activity. Plasminogen activator inhibitor type 1 (PAI-1) acts as a negative regulator. This system is complicated by a poorly understood intrinsic reactivity of the uPA pro-enzyme (pro-uPA) before proteolytic activation, directed against both plasminogen and PAI-1. We have studied the integrated activation mechanism under the repression of PAI-1 in a purified system. A covalent reaction between pro-uPA and PAI-1 was positively demonstrated but the reaction of PAI-1 with two-chain uPA was found to be at least 1000-fold faster. However, in spite of this very fast inhibition, two-chain uPA still became the dominant plasminogen activator when plasminogen was incubated with pro-uPA and PAI-1. The activity pattern observed under these conditions revealed an initial lag phase, followed by a continuous generation of minute amounts of active two-chain uPA, this uPA having a short lifetime before inhibition but still succeeding to generate new plasmin activity, thus preventing a complete inactivation of the feedback system. This property of the activation system was retained even in the simultaneous presence of PAI-1 and alpha(2)-antiplasmin. Addition of soluble uPA receptor to the system did not change the role of pro-uPA and the same pattern was observed when pro-uPA was bound to the uPA receptor on U937 cells. The present mechanism maintains the system at standby level and may be triggered to increased activity without the need for an external initiating event.
Collapse
Affiliation(s)
- Niels Behrendt
- Finsen Laboratory, Rigshospitalet, Strandboulevarden 49, Bldg. 7.2, DK-2100 Copenhagen Ø, Denmark.
| | | | | | | |
Collapse
|
159
|
Chen Z, Fan Z, McNeal JE, Nolley R, Caldwell MC, Mahadevappa M, Zhang Z, Warrington JA, Stamey TA. Hepsin and maspin are inversely expressed in laser capture microdissectioned prostate cancer. J Urol 2003; 169:1316-9. [PMID: 12629351 DOI: 10.1097/01.ju.0000050648.40164.0d] [Citation(s) in RCA: 62] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
PURPOSE Recent studies have shown that hepsin, a serine protease, is over expressed in prostate cancers, implicating hepsin activity in tumor invasion. Using microarray technology we have previously identified 22 genes that were up-regulated in high grade prostate cancers compared with benign prostatic hyperplasia. Of them hepsin was the most differentially over expressed. In the current report we compare hepsin to maspin (BD Transduction Laboratories, San Diego, California), a serine protease inhibitor (serpin), to measure the balance between levels of serine proteases and serpins, which are considered to be a critical determinant of net proteolytic activity. MATERIALS AND METHODS We combined the technique of laser capture microdissection with gene expression monitoring by micro-array analysis to investigate the gene expression profiles of prostate cells of different histological types. We also studied maspin immunohistochemically. RESULTS We observed that hepsin as well as 7 of 22 previously reported up-regulated genes demonstrated a pattern of increasing expression with increasing malignant phenotype. In contrast, the expression of maspin (a serpin) decreased with increasing malignancy of prostate cancers. Using immunohistochemistry we observed that maspin protein is expressed strongly in benign prostatic tissues and slightly in grade 3 prostate cancers, and is absent in grade 4/5 cancers. CONCLUSIONS We conclude that the increased ratio of hepsin-to-maspin may have an important role in prostate cancer progression and invasion.
Collapse
Affiliation(s)
- Zuxiong Chen
- Department of Urology, School of Medicine, Stanford University, Stanford, California 94305, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
160
|
Tavian D, Salvi A, De Petro G, Barlati S. Stable expression of antisense urokinase mRNA inhibits the proliferation and invasion of human hepatocellular carcinoma cells. Cancer Gene Ther 2003; 10:112-20. [PMID: 12536199 DOI: 10.1038/sj.cgt.7700533] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2002] [Indexed: 11/09/2022]
Abstract
Urokinase-type plasminogen activator (u-PA) plays a key role in malignant tumor behavior. We have previously shown that the expression of high levels of u-PA mRNA in human hepatocellular carcinoma (HCC) biopsies was inversely correlated with the survival of the patients. In order to evaluate the involvement of u-PA in the invasive and infiltrating properties of HCC cells, the SKHep1C3 cell line was stably transfected with an expression vector containing the 5' portion (257 bp) of u-PA cDNA in the antisense orientation. u-PA mRNA expression and its protein level and enzymatic activity were specifically inhibited in the antisense transfectants. A comparable inhibition of the u-PA receptor (u-PAR) mRNA and protein was also evidenced in the antisense transfected cells compared with the control ones. At the functional level, the SKHep1C3-AS cells showed a significant reduction in proliferation, Matrigel invasion, and motility assays compared to parental and vector-alone cells. These results indicate that u-PA is an essential factor in the growth and invasiveness of human hepatocarcinoma cells. Antisense u-PA strategy might be a potential approach to reduce tumor growth as well as the invasive capacity of the malignant cells in HCC.
Collapse
MESH Headings
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/therapy
- Cell Division/drug effects
- Cell Division/genetics
- Cell Movement/genetics
- Fluorescent Antibody Technique
- Genetic Therapy/methods
- Humans
- Immunoblotting
- Liver Neoplasms/genetics
- Liver Neoplasms/pathology
- Liver Neoplasms/therapy
- Neoplasm Invasiveness
- Polymerase Chain Reaction/methods
- RNA, Antisense/biosynthesis
- RNA, Antisense/genetics
- RNA, Antisense/pharmacology
- RNA, Messenger/biosynthesis
- RNA, Messenger/genetics
- RNA, Messenger/pharmacology
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/metabolism
- Receptors, Urokinase Plasminogen Activator
- Transfection
- Tumor Cells, Cultured
- Urokinase-Type Plasminogen Activator/antagonists & inhibitors
- Urokinase-Type Plasminogen Activator/genetics
- Urokinase-Type Plasminogen Activator/metabolism
Collapse
Affiliation(s)
- Daniela Tavian
- Division of Biology and Genetics, Department of Biomedical Sciences and Biotechnology, IDET Center of Excellence, University of Brescia, Brescia, Italy
| | | | | | | |
Collapse
|
161
|
Bertenshaw GP, Norcum MT, Bond JS. Structure of homo- and hetero-oligomeric meprin metalloproteases. Dimers, tetramers, and high molecular mass multimers. J Biol Chem 2003; 278:2522-32. [PMID: 12399461 DOI: 10.1074/jbc.m208808200] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Meprin A and B, metalloproteases consisting of evolutionarily related alpha and/or beta subunits, are membrane-bound and secreted enzymes expressed by kidney and intestinal epithelial cells, leukocytes, and cancer cells. Previous work established that the multidomain meprin subunits (each approximately 80 kDa) form disulfide-bridged homo- and heterodimers, and differ in substrate and peptide bond specificities. The work herein clearly demonstrates that meprin dimers differ markedly in their ability to oligomerize. Electrophoresis, light scattering, size exclusion chromatography, and electron microscopy were used to characterize quaternary structures of recombinant rat meprins. Meprin B, consisting of meprin beta subunits only, was dimeric under a wide range of conditions. By contrast, meprin alpha homodimers formed heterogeneous multimers (ring-, circle-, spiral-, and tube-like structures) containing up to 100 subunits, with molecular masses at protein peaks ranging from approximately 1.0 to 6.0 MDa. The size of the meprin alpha homo-oligomers was dependent on protein concentration, ionic strength, and activation state. Meprin alphabeta heterodimers tended to form tetramers but not higher oligomers. Thus, the presence of meprin beta, which has a transmembrane domain in vivo, restricts the oligomerization potential of meprin molecules and localizes meprins to the plasma membrane. By contrast, the propensity of secreted meprin alpha homodimers to self-associate concentrates proteolytic potential into high molecular mass multimers and thus allows for autocompartmentalization. The work indicates that different mechanisms exist to localize and concentrate the proteolytic activity of membrane-bound and secreted meprin metalloproteinases.
Collapse
Affiliation(s)
- Greg P Bertenshaw
- Department of Biochemistry and Molecular Biology, Pennsylvania State University College of Medicine, Hershey 17033, USA
| | | | | |
Collapse
|
162
|
Liu S, Aaronson H, Mitola DJ, Leppla SH, Bugge TH. Potent antitumor activity of a urokinase-activated engineered anthrax toxin. Proc Natl Acad Sci U S A 2003; 100:657-62. [PMID: 12525700 PMCID: PMC141052 DOI: 10.1073/pnas.0236849100] [Citation(s) in RCA: 97] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The acquisition of cell-surface urokinase plasminogen activator activity is a hallmark of malignancy. We generated an engineered anthrax toxin that is activated by cell-surface urokinase in vivo and displays limited toxicity to normal tissue but broad and potent tumoricidal activity. Native anthrax toxin protective antigen, when administered with a chimeric anthrax toxin lethal factor, Pseudomonas exotoxin fusion protein, was extremely toxic to mice, causing rapid and fatal organ damage. Replacing the furin activation sequence in anthrax toxin protective antigen with an artificial peptide sequence efficiently activated by urokinase greatly attenuated toxicity to mice. In addition, the mutation conferred cell-surface urokinase-dependent toxin activation in vivo, as determined by using a panel of plasminogen, plasminogen activator, plasminogen activator receptor, and plasminogen activator inhibitor-deficient mice. Surprisingly, toxin activation critically depended on both urokinase plasminogen activator receptor and plasminogen in vivo, showing that both proteins are essential cofactors for the generation of cell-surface urokinase. The engineered toxin displayed potent tumor cell cytotoxicity to a spectrum of transplanted tumors of diverse origin and could eradicate established solid tumors. This tumoricidal activity depended strictly on tumor cell-surface plasminogen activation. The data show that a simple change of protease activation specificity converts anthrax toxin from a highly lethal to a potent tumoricidal agent.
Collapse
Affiliation(s)
- Shihui Liu
- Oral Infection and Immunity Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | |
Collapse
|
163
|
Beneš P, Jurajda M, Žaloudík J, Izakovičová-Hollá L, Vácha J. C766T low-density lipoprotein receptor-related protein 1 (LRP1) gene polymorphism and susceptibility to breast cancer. Breast Cancer Res 2003; 5:R77-81. [PMID: 12793904 PMCID: PMC165006 DOI: 10.1186/bcr591] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2003] [Revised: 02/25/2003] [Accepted: 02/28/2003] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Low-density lipoprotein receptor-related protein 1 (LRP1) is a multifunctional endocytic receptor with an important role in regulating the activity of proteinases in extracellular matrix. Several studies have also described its role in intracellular signaling. Previous studies showed that the expression of LRP1 is related to invasiveness of cancer cells. However, recent data on LRP1 suggest that this receptor can also be involved in tumor establishment and progression. METHODS We investigated an association between the C766T polymorphism of the third exon of the LRP1 gene and breast cancer in a sample of women of Caucasian origin. Allele and genotype frequencies of this polymorphism were assessed in 164 women with breast cancer and in 183 age-compatible women without a history of any cancer disease. RESULTS An increase in LRP1 T allele frequency in subjects with breast cancer was observed compared with controls (0.21 versus 0.15, P = 0.01963). A significant excess of genotypes with the T allele (homozygotes plus heterozygotes) was also observed (odds ratio 1.743, 95% confidence interval 1.112-2.732). CONCLUSION The T allele of the C766T polymorphism in the LRP1 gene is associated with an increased risk of breast cancer development in women of Caucasian origin.
Collapse
Affiliation(s)
- Petr Beneš
- Department of Molecular Biology and Genetics, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Michal Jurajda
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Jan Žaloudík
- Masaryk Memorial Cancer Institute, Brno, Czech Republic
| | - Lydie Izakovičová-Hollá
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Jiří Vácha
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| |
Collapse
|
164
|
Silvestri I, Longanesi Cattani I, Franco P, Pirozzi G, Botti G, Stoppelli MP, Carriero MV. Engaged urokinase receptors enhance tumor breast cell migration and invasion by upregulating alpha(v)beta5 vitronectin receptor cell surface expression. Int J Cancer 2002; 102:562-71. [PMID: 12447996 DOI: 10.1002/ijc.10744] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
We have previously shown that urokinase receptor physically and functionally interacts with alpha(v)beta5 vitronectin receptor, leading to tumor breast cell migration and invasion. Here, the link between these 2 receptors was further investigated by analyzing the expression levels of urokinase receptor and alpha(v)beta5 integrin in 35 human breast carcinomas and 5 benign breast lesions. The occurrence of a positive correlation between urokinase receptor and alpha(v)beta5 protein levels in benign and malignant tumor specimens prompted us to investigate whether engaged urokinase receptors might modulate alpha(v)beta5 expression. Here, we report the receptor-dependent ability of catalytically inactive urokinase to upregulate the alpha(v) and beta5 chains in MDA-MB-231 and MCF-7 breast carcinoma cell lines in a time- and concentration-dependent manner. This effect is dependent on protein kinase C activity and requires new protein synthesis. Accordingly, the availability of assembled alpha(v)beta5 receptors on the cell surface increases upon urokinase treatment, as shown by immunoprecipitation and immunocytochemical analyses. Exposure to urokinase leads to enhanced tumor cell migration and invasion, which is prevented by the "phosphorylation-like" urokinase receptor antagonist His-uPA(138E/303E), the DNA-binding drug mithramycin, the protein kinase C inhibitor calphostin C and anti-alpha(v)beta5 antibodies. Finally, urokinase enables benign breast MCF-10A cells to cross Matrigel in a alpha(v)beta5- and urokinase receptor-dependent manner, indicating that urokinase controls a regulatory circuitry crucial to breast tumor progression.
Collapse
Affiliation(s)
- Immacolata Silvestri
- Department of Experimental Oncology, National Cancer Institute of Naples, Naples, Italy
| | | | | | | | | | | | | |
Collapse
|
165
|
Curino A, Mitola DJ, Aaronson H, McMahon GA, Raja K, Keegan AD, Lawrence DA, Bugge TH. Plasminogen promotes sarcoma growth and suppresses the accumulation of tumor-infiltrating macrophages. Oncogene 2002; 21:8830-42. [PMID: 12483535 DOI: 10.1038/sj.onc.1205951] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2002] [Revised: 07/31/2002] [Accepted: 08/07/2002] [Indexed: 11/09/2022]
Abstract
The specific functions of plasminogen, stromal plasminogen activator, stromal plasminogen activator receptor, and stromal plasminogen activator inhibitor in the progression of the murine soft tissue sarcoma, T241 were investigated. Negation of plasminogen to the tumor blunted the orthotopic growth of the sarcoma in syngeneic mice. The reduced tumor growth was associated with a dramatic increase in tumor-infiltrating F4/80-positive macrophages and a diminution of vessel density, but not with obvious differences in fibrin and collagen deposition, or invasiveness of the tumor. Ablation of plasminogen activation by the tumor stroma only modestly impaired the prolonged growth of the sarcoma, suggesting that tumor cell-produced plasminogen activator is sufficient to mediate productive plasminogen activation. Plasminogen facilitated sarcoma progression, angiogenesis, and suppression of macrophage infiltration in the absence of either stromal urokinase plasminogen activator receptor or stromal plasminogen activator inhibitor. These data demonstrate that tumor cell-produced plasminogen activator and host plasminogen cooperate to facilitate soft tissue sarcoma growth and suppress the accumulation of tumor-infiltrating macrophages.
Collapse
Affiliation(s)
- Alejandro Curino
- Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Bethesda, Maryland, MD 20892, USA
| | | | | | | | | | | | | | | |
Collapse
|
166
|
Abstract
While a genome-centric paradigm in human cancer development was useful for the understanding of some malignancies such as leukemias, causative molecular defects intrinsic to melanocytes have not been defined in the majority of human melanomas. Recent work, however, has shown that regulatory signals governing melanocytic cell growth and differentiation may originate from the surrounding host cells either directly through physical contact or indirectly through soluble factors and extracellular matrix molecules. In this review, we present experimental systems useful for dissecting melanoma-host interactions and highlight evidence that the tumor microenvironment contributes to the oncogenic process. Thus, melanomagenesis is not merely an act of a single outlaw but a conspiracy orchestrated by multiple partners in the neighborhood who come into play in a precise spatiotemporal order. Defining intercellular molecular dialogues in human skin promises to provide key information for the development of novel treatment strategies that target the functional unit of stroma and tumor.
Collapse
Affiliation(s)
- Mei-Yu Hsu
- The Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104-4283, USA
| | | | | |
Collapse
|
167
|
Netzel-Arnett S, Mitola DJ, Yamada SS, Chrysovergis K, Holmbeck K, Birkedal-Hansen H, Bugge TH. Collagen dissolution by keratinocytes requires cell surface plasminogen activation and matrix metalloproteinase activity. J Biol Chem 2002; 277:45154-61. [PMID: 12192005 DOI: 10.1074/jbc.m206354200] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Matrix metalloproteinase-14 is required for degradation of fibrillar collagen by mesenchymal cells. Here we show that keratinocytes use an alternative plasminogen and matrix metalloproteinase-13-dependent pathway for dissolution of collagen fibrils. Primary keratinocytes displayed an absolute requirement for serum to dissolve collagen. Dissolution of collagen was abolished in plasminogen-depleted serum and could be restored by the exogenous addition of plasminogen. Both plasminogen activator inhibitor-1 and tissue inhibitor of metalloproteinase blocked collagen dissolution, demonstrating the requirement of both plasminogen activation and matrix metalloproteinase activity for degradation. Cell surface plasmin activity was critical for the degradation process as aprotinin, but not alpha(2)-antiplasmin, prevented collagen dissolution. Keratinocytes with single deficiencies in either urokinase or tissue plasminogen activator retained the ability to dissolve collagen. However, collagen fibril dissolution was abolished in keratinocytes with a combined deficiency in both urokinase and tissue plasminogen activator. Combined, but not single, urokinase and tissue plasminogen activator deficiency also completely blocked the activation of the fibrillar collagenase, matrix metalloproteinase-13, by keratinocytes. The activation of matrix metalloproteinase-13 in normal keratinocytes was prevented by plasminogen activator inhibitor-1 and aprotinin but not by tissue inhibitor of metalloproteinase-1 and -2, suggesting that plasmin activates matrix metalloproteinase-13 directly. We propose that plasminogen activation facilitates keratinocyte-mediated collagen breakdown via the direct activation of matrix metalloproteinase-13 and possibly other fibrillar collagenases.
Collapse
Affiliation(s)
- Sarah Netzel-Arnett
- Matrix Metalloproteinase Unit, NIDCR, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | | | | | |
Collapse
|
168
|
Billottet C, Janji B, Thiery JP, Jouanneau J. Rapid tumor development and potent vascularization are independent events in carcinoma producing FGF-1 or FGF-2. Oncogene 2002; 21:8128-39. [PMID: 12444548 DOI: 10.1038/sj.onc.1205935] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2002] [Revised: 08/02/2002] [Accepted: 08/05/2002] [Indexed: 11/09/2022]
Abstract
FGF-1 and FGF-2 are pleiotropic growth factors for many cell types, operating through the activation of specific transmembrane FGF receptors (FGFRs). The role of these factors in tumor progression was investigated, with specific discrimination between their autocrine and non autocrine cellular activity. The rat bladder carcinoma NBT-II cells were engineered to produce FGF-1 or 18 kDa FGF-2 in the presence or absence of their specific receptor. Non-autocrine cells that produced FGF-1 or FGF-2 but lacked FGFRs were epithelial and reminiscent of the parental NBT-II cells. Whilst autocrine cells, which both constitutively produced and secreted the growth factor and expressed FGFRs, had a highly invasive mesenchymal phenotype. Correspondingly, the autocrine cells were highly tumorigenic in vivo compared to the parental and non-autocrine cells, which correlated with the increased production of uPAR and active uPA and increased in vitro invasive potential. Although all cells produced VEGF, only tumors derived from cells that produced FGF-1 or FGF-2 were highly vascularized, suggesting that these two growth factors could be involved in the angiogenic process by activating host endothelial cells. As a result of activation of the FGFR in autocrine cells, changes in cell morphology and an increase in the invasive and tumorigenic properties were observed, however no in vitro or in vivo differential functions between FGF-1 and FGF-2 could be identified in this system. In conclusion, our data demonstrates that rapid tumor development is not dependent upon increased tumor vascularization, suggesting that 'basal' angiogenesis, probably mediated by VEGF, is sufficient to support tumor growth.
Collapse
MESH Headings
- Animals
- Autocrine Communication
- Carcinoma/blood supply
- Carcinoma/genetics
- Carcinoma/metabolism
- Carcinoma/pathology
- Disease Progression
- Endothelial Growth Factors/metabolism
- Epithelial Cells/pathology
- Female
- Fibroblast Growth Factor 1/genetics
- Fibroblast Growth Factor 1/physiology
- Fibroblast Growth Factor 2/genetics
- Fibroblast Growth Factor 2/physiology
- Gene Expression Regulation, Neoplastic
- Intercellular Signaling Peptides and Proteins/metabolism
- Lymphokines/metabolism
- Matrix Metalloproteinases/biosynthesis
- Matrix Metalloproteinases/genetics
- Mesoderm
- Mice
- Mice, Nude
- Neoplasm Invasiveness
- Neoplasm Proteins/physiology
- Neoplasm Transplantation
- Neovascularization, Pathologic/genetics
- Neovascularization, Pathologic/physiopathology
- Phenotype
- Rats
- Receptor Protein-Tyrosine Kinases/genetics
- Receptor Protein-Tyrosine Kinases/physiology
- Receptor, Fibroblast Growth Factor, Type 1
- Receptor, Fibroblast Growth Factor, Type 2
- Receptors, Cell Surface/biosynthesis
- Receptors, Cell Surface/genetics
- Receptors, Fibroblast Growth Factor/genetics
- Receptors, Fibroblast Growth Factor/physiology
- Receptors, Urokinase Plasminogen Activator
- Recombinant Fusion Proteins/physiology
- Sequence Deletion
- Tissue Inhibitor of Metalloproteinase-2/biosynthesis
- Tissue Inhibitor of Metalloproteinase-2/genetics
- Transfection
- Tumor Cells, Cultured/metabolism
- Tumor Cells, Cultured/pathology
- Tumor Cells, Cultured/transplantation
- Urinary Bladder Neoplasms/blood supply
- Urinary Bladder Neoplasms/genetics
- Urinary Bladder Neoplasms/metabolism
- Urinary Bladder Neoplasms/pathology
- Urokinase-Type Plasminogen Activator/biosynthesis
- Urokinase-Type Plasminogen Activator/genetics
- Vascular Endothelial Growth Factor A
- Vascular Endothelial Growth Factors
Collapse
Affiliation(s)
- Clotilde Billottet
- Laboratory of Cell Morphogenesis and Tumor Progression, UMR 144 CNRS, Institut Curie, Section de recherche, 26 rue d'Ulm, 75248 Paris, cedex 05, France
| | | | | | | |
Collapse
|
169
|
Purcell WT, Rudek MA, Hidalgo M. Development of matrix metalloproteinase inhibitors in cancer therapy. Hematol Oncol Clin North Am 2002; 16:1189-227. [PMID: 12512389 DOI: 10.1016/s0889-8588(02)00044-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The matrix metalloproteinases represent an attractive target for cancer treatment, and a number of matrix metalloproteinase inhibitors are undergoing clinical trials. The results of these studies will establish whether any of these compounds are therapeutically useful. Independent of the conclusions from the first generation of studies, the field of matrix metalloproteinase inhibitors remains attractive for creative and innovative research. In the future, the development of novel, less toxic, and more effective matrix metalloproteinase inhibitors, and the combination of conventional agents with these novel anticancer agents will constitute the main focus of research efforts.
Collapse
Affiliation(s)
- W Thomas Purcell
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Division of Medical Oncology, Baltimore, MD 21231-2410, USA
| | | | | |
Collapse
|
170
|
Jensen MK, Riisbro R, de Nully Brown P, Brünner N, Hasselbalch HC. Elevated soluble urokinase plasminogen activator receptor in plasma from patients with idiopathic myelofibrosis or polycythaemia vera. Eur J Haematol 2002; 69:43-9. [PMID: 12270061 DOI: 10.1034/j.1600-0609.2002.01732.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Extracellular proteolytic enzymes of the urokinase-type plasminogen activator (uPA) system and the family of metalloproteases play a crucial role in the matrix degradation and tissue remodelling processes characteristic of malignant disorders. The receptor for urokinase plasminogen activator (uPAR) serves to localise and intensify the action of uPA and is expressed on the surface of malignant as well as tumour stromal cells including fibroblasts. A soluble form of uPAR (suPAR) cleaved from its glycosyl-phosphatidylinositol anchor is detected in plasma from healthy individuals and increased levels of suPAR have been found in advanced malignancy, suggesting that suPAR may be a marker of extensive tissue remodelling. In an attempt to clarify whether suPAR might be a marker for bone marrow tissue remodelling we measured plasma suPAR levels in a patient cohort comprising 17 with myelofibrosis (MF), 17 with polycythaemia vera (PV), 15 with essential thrombocythaemia (ET), one with a transitional myeloproliferative disorder evolving from PV and 30 controls. Compared with controls suPAR levels were significantly higher in the patients (P < 0.0001) (median 3.35 vs. 2.32 microg L(-1)). Moreover, in subgroup analyses including patients with MF, PV, and ET, respectively, suPAR levels differed significantly with the highest levels found in patients with MF and PV (MF vs. PV vs. ET; P = 0.0003). When comparing suPAR levels of the individual patient subgroups with controls, only suPAR levels of PV and MF patients were significantly increased (P < 0.0001). Sixty-five percent of patients with PV and MF (22/34) had suPAR plasma values that were above the mean +2 standard deviations (SD) of controls. The concentration of suPAR was significantly correlated to plasma lactate dehydrogenase, thrombomodulin, and complex of tPA:PAI-1 in the patients. There was no difference between patients and controls when comparing plasma uPA levels. Increased plasma suPAR levels in patients with chronic myeloproliferative disorders may reflect increased uPAR production in the bone marrow, leading to enhanced bone marrow remodelling.
Collapse
Affiliation(s)
- Morten Krogh Jensen
- Department of Haematology L, Righospilatet, University of Copenhagen, Denmark.
| | | | | | | | | |
Collapse
|
171
|
Han YP, Hughes MW, Nien YD, Garner WL. IL-8-stimulated expression of urokinase-type plasminogen activator in human skin and human epidermal cells. J Surg Res 2002; 106:328-34. [PMID: 12175988 DOI: 10.1006/jsre.2002.6482] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Extracellular matrix (ECM) remodeling is essential for normal development and tissue repair. Although many roles for extracellular proteinases in the breakdown of ECM have been established, the regulations of these proteinases in human tissue are not fully understood. Inflammatory cytokines have been implicated in the regulation of several matrix metalloproteinases. To determine whether these mediators have a similar effect on fibrinolysis and the remodeling of the fibrin provisional matrix, we examined the role of cytokines on the regulation of urokinase-type plasminogen activator (uPA) and tissue-type plasminogen activator (tPA) in human skin. In this report, we show that interleukin-8 (IL-8), but not other cytokines tested, is a potent inducer of the 38-kDa uPA in organ-cultured human skin. In addition, the uPA inhibitor, PAI-1, was not affected by IL-8. When primary epidermal human keratinocytes were treated with IL-8, 55-kDa pro-uPA was significantly induced in the conditioned medium. The mRNA expression of uPA in the keratinocytes was found to be constitutively elevated and was not affected by IL-8. To support such a notion, activation of the 5'-flanking promoter of the human uPA gene was measured using the CAT reporter assay. Consistent with the results of mRNA measurement, the promoter is constitutively active in keratinocytes and is not affected by IL-8. In contrast, the promoter construct is neither active in the dermal fibroblasts nor stimulated by the cytokine. This differential transactivation of uPA gene in these cells indicates that keratinocyte-specific factors may govern the basal expression of the gene. These results indicate a complex regulation of uPA expression in epidermal cells.
Collapse
Affiliation(s)
- Yuan-Ping Han
- Division of Plastic and Reconstructive Surgery, University of Southern California School of Medicine, Los Angeles 90033, USA
| | | | | | | |
Collapse
|
172
|
Seddighzadeh M, Steineck G, Larsson P, Wijkström H, Norming U, Onelöv E, Linder S. Expression of UPA and UPAR is associated with the clinical course of urinary bladder neoplasms. Int J Cancer 2002; 99:721-6. [PMID: 12115506 DOI: 10.1002/ijc.10426] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The expression of urokinase plasminogen activator (uPA) and its receptor (uPAR) mRNA was determined in 194 subjects with newly detected bladder neoplasms, selected from a larger population-based series. An association was found between uPA and uPAR expression (n = 172; Spearman r(s) = 0.60, p < 0.001). Both uPA and uPAR mRNA levels were higher in muscle invasive (T2+) tumors than in noninvasive mucosal tumors (Ta) or those invading submucosa (T1). The relative hazard ratios (RHRs) for cancer-specific death associated with elevated expression (95% CI), adjusted for age and gender in a Cox proportional hazard model, were 1.8 (1.0-3.3) for uPA (upper quartile cut-line), 2.2 (1.3-4.0) for uPAR (median quartile cut-line) and 2.5 (1.3-4.9) for uPA + uPAR. An RHR for metastatic disease of 4.0 (1.6-9.9) was observed for uPAR. Restricting the analyses to T2+ tumors, the corresponding figures were: 2.1 (1.1-3.9) for uPA, 1.6 (0.8-3.3) for uPAR and 2.5 (1.1-5.6) for both. We conclude that expression of uPA and uPAR is associated with the clinical behaviour of bladder neoplasms, possibly providing means for refined staging of muscle invasive tumors and target proteins for novel therapies.
Collapse
Affiliation(s)
- Maria Seddighzadeh
- Cancer Center Karolinska, Karolinska Institute and Hospital, Stockholm, Sweden
| | | | | | | | | | | | | |
Collapse
|
173
|
Bogdanov AA, Lin CP, Simonova M, Matuszewski L, Weissleder R. Cellular activation of the self-quenched fluorescent reporter probe in tumor microenvironment. Neoplasia 2002; 4:228-36. [PMID: 11988842 PMCID: PMC1531696 DOI: 10.1038/sj.neo.7900238] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2001] [Accepted: 07/09/2001] [Indexed: 11/09/2022]
Abstract
The effect of intralysosomal proteolysis of near-infrared fluorescent (NIRF) self-quenched macromolecular probe (PGC-Cy5.5) has been previously reported and used for tumor imaging. Here we demonstrate that proteolysis can be detected noninvasively in vivo at the cellular level. A codetection of GFP fluorescence (using two-photon excitation) and NIRF was performed in tumor-bearing animals injected with PGC-Cy5.5. In vivo microscopy of tumor cells in subdermal tissue layers (up to 160 microm) showed a strong Cy5.5 dequenching effect in GFP-negative cells. This observation was corroborated by flow cytometry, sorting, and reverse transcription polymerase chain reaction analysis of tumor-isolated cells. Both GFP-positive (81% total) and GFP-negative (19% total) populations contained Cy5.5-positive cells. The GFP-negative cells were confirmed to be host mouse cells by the absence of rat cathepsin mRNA signal. The subfraction of GFP-negative cells (2.5-3.0%) had seven times higher NIRF intensity than the majority of GFP-positive or GFP-negative cells (372 and 55 AU, respectively). Highly NIRF-positive, FP-negative cells were CD45- and MAC3-positive. Our results indicate that: 1) intracellular proteolysis can be imaged in vivo at the cellular level using cathepsin-sensitive probes; 2) tumor-recruited cells of hematopoetic origin participate most actively in uptake and degradation of long-circulating macromolecular probes.
Collapse
Affiliation(s)
- Alexei A Bogdanov
- Center for Molecular Imaging Research, Massachusetts General Hospital, Building 149, 13th Street, Charlestown, MA 02129, USA
| | | | | | | | | |
Collapse
|
174
|
Guo M, Mathieu PA, Linebaugh B, Sloane BF, Reiners JJ. Phorbol ester activation of a proteolytic cascade capable of activating latent transforming growth factor-betaL a process initiated by the exocytosis of cathepsin B. J Biol Chem 2002; 277:14829-37. [PMID: 11815600 DOI: 10.1074/jbc.m108180200] [Citation(s) in RCA: 67] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
12-O-Tetradecanoylphorbol-13-acetate (TPA) suppresses the proliferation of the human breast epithelial cell line MCF10A-Neo by initiating proteolytic processes that activate latent transforming growth factor (TGF)-beta in the serum used to supplement culture medium. Within 1 h of treatment, cultures accumulated an extracellular activity capable of cleaving a substrate for urokinase-type plasminogen activator (uPA) and tissue plasminogen activator (tPA). This activity was inhibited by plasminogen activator inhibitor-1 or antibodies to uPA but not tPA. Pro-uPA activation was preceded by dramatic changes in lysosome trafficking and the extracellular appearance of cathepsin B and beta-hexosaminidase but not cathepsins D or L. Co-treatment of cultures with the cathepsin B inhibitors CA-074 or Z-FA-FMK suppressed the cytostatic effects of TPA and activation of pro-uPA. In the absence of TPA, exogenously added cathepsin B activated pro-uPA and suppressed MCF10A-Neo proliferation. The cytostatic effects of both TPA and cathepsin B were suppressed in cells cultured in medium depleted of plasminogen/plasmin or supplemented with neutralizing TGF-beta antibody. Pretreatment with cycloheximide did not suppress the exocytosis of cathepsin B or the activation of pro-uPA. Hence, TPA activates signaling processes that trigger the exocytosis of a subpopulation of lysosomes/endosomes containing cathepsin B. Subsequently, extracellular cathepsin B initiates a proteolytic cascade involving uPA, plasminogen, and plasmin that activates serum-derived latent TGF-beta.
Collapse
Affiliation(s)
- Meng Guo
- Institute of Environmental Health Sciences, Wayne State University and the Department of Pharmacology, Wayne State University School of Medicine, Detroit, Michigan 48201, USA
| | | | | | | | | |
Collapse
|
175
|
Schnack Nielsen B, Rank F, Engelholm LH, Holm A, Danø K, Behrendt N. Urokinase receptor-associated protein (uPARAP) is expressed in connection with malignant as well as benign lesions of the human breast and occurs in specific populations of stromal cells. Int J Cancer 2002; 98:656-64. [PMID: 11920633 DOI: 10.1002/ijc.10227] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The urokinase-type plasminogen activator (uPA) and the uPA receptor (uPAR) are key components in the plasminogen activation system, serving to promote specific events of extracellular matrix degradation in connection with tissue remodeling and cancer invasion. We recently described a new uPAR-associated protein (uPARAP), an internalization receptor that interacts with the pro-uPA:uPAR complex. In our study, we generated a specific polyclonal peptide antibody against human uPARAP and used it for the localization of uPARAP in different breast lesions. The affinity-purified antibodies specifically recognized uPARAP in Western blotting and gave a strong signal in immunohistochemistry. The immunohistochemic localization pattern was found to be identical to that of uPARAP mRNA as determined in parallel by in situ hybridization. uPARAP expression was then studied in both benign and malignant breast lesions. Whereas the normal breast tissue was uPARAP-negative, all benign lesions and ductal carcinoma in situ lesions showed immunoreactivity in fibroblast-like cells and myoepithelial cells associated with the lesion. In invasive carcinoma, uPARAP immunoreactivity was limited to tumor-associated mesenchymal cells. Double immunofluorescence analysis of invasive ductal carcinoma using antibodies against specific cell markers showed that uPARAP was localized in myofibroblasts and macrophages. No malignant cells, no endothelial cells and no vascular smooth muscle cells showed uPARAP immunoreactivity. We conclude that expression of uPARAP is associated with the abnormal breast and that expression appears in myofibroblasts, macrophages and myoepithelium. We suggest that uPARAP is involved in the clearance of the uPA:uPAR complex as well as other possible ligands during benign and malignant tissue remodeling.
Collapse
MESH Headings
- Animals
- Antibody Formation
- Blotting, Western
- Breast/metabolism
- Breast Neoplasms/genetics
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Carcinoma in Situ/genetics
- Carcinoma in Situ/metabolism
- Carcinoma in Situ/pathology
- Carcinoma, Ductal, Breast/genetics
- Carcinoma, Ductal, Breast/metabolism
- Carcinoma, Ductal, Breast/pathology
- Cross-Linking Reagents
- Female
- Fluorescent Antibody Technique
- Humans
- In Situ Hybridization
- Mannose-Binding Lectins
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/metabolism
- Platelet Endothelial Cell Adhesion Molecule-1/metabolism
- RNA, Messenger/metabolism
- Rabbits
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/metabolism
- Stromal Cells/metabolism
- Stromal Cells/pathology
- Transcription, Genetic
- U937 Cells/metabolism
- U937 Cells/pathology
Collapse
|
176
|
Rijneveld AW, Levi M, Florquin S, Speelman P, Carmeliet P, van Der Poll T. Urokinase receptor is necessary for adequate host defense against pneumococcal pneumonia. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 168:3507-11. [PMID: 11907112 DOI: 10.4049/jimmunol.168.7.3507] [Citation(s) in RCA: 139] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Cell recruitment is a multistep process regulated by cytokines, chemokines, and growth factors. Previous work has indicated that the urokinase plasminogen activator receptor (uPAR) may also play a role in this mechanism, presumably by an interaction with the beta(2) integrin CD11b/CD18. Indeed, an essential role of uPAR in neutrophil recruitment during pulmonary infection has been demonstrated for beta(2) integrin-dependent respiratory pathogens. We investigated the role of uPAR and urokinase plasminogen activator (uPA) during pneumonia caused by a beta(2) integrin-independent respiratory pathogen, Streptococcus pneumoniae. uPAR-deficient (uPAR(-/-)), uPA-deficient (uPA(-/-)), and wild-type (Wt) mice were intranasally inoculated with 10(5) CFU S. pneumoniae. uPAR(-/-) mice showed reduced granulocyte accumulation in alveoli and lungs when compared with Wt mice, which was associated with more S. pneumoniae CFU in lungs, enhanced dissemination of the infection, and a reduced survival. In contrast, uPA(-/-) mice showed enhanced host defense, with more neutrophil influx and less pneumococci in the lungs compared with Wt mice. These data suggest that uPAR is necessary for adequate recruitment of neutrophils into the alveoli and lungs during pneumonia caused by S. pneumoniae, a pathogen eliciting a beta(2) integrin-independent inflammatory response. This function is even more pronounced when uPAR is unoccupied by uPA.
Collapse
MESH Headings
- Animals
- Bronchoalveolar Lavage Fluid/cytology
- Bronchoalveolar Lavage Fluid/immunology
- Bronchoalveolar Lavage Fluid/microbiology
- Cell Movement/genetics
- Cell Movement/immunology
- Chemokines/biosynthesis
- Colony Count, Microbial
- Cytokines/biosynthesis
- Lung/enzymology
- Lung/microbiology
- Lung/pathology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Pneumonia, Pneumococcal/immunology
- Pneumonia, Pneumococcal/microbiology
- Pneumonia, Pneumococcal/mortality
- Pneumonia, Pneumococcal/pathology
- Receptors, Cell Surface/deficiency
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/physiology
- Receptors, Urokinase Plasminogen Activator
- Streptococcus pneumoniae/growth & development
- Survival Analysis
- Urokinase-Type Plasminogen Activator/deficiency
- Urokinase-Type Plasminogen Activator/genetics
- Urokinase-Type Plasminogen Activator/metabolism
Collapse
Affiliation(s)
- Anita W Rijneveld
- Department of Experimental Internal Medicine, University of Amsterdam, Amsterdam, The Netherlands.
| | | | | | | | | | | |
Collapse
|
177
|
Herbst RS, Hidalgo M, Pierson AS, Holden SN, Bergen M, Eckhardt SG. Angiogenesis inhibitors in clinical development for lung cancer. Semin Oncol 2002; 29:66-77. [PMID: 11894016 DOI: 10.1053/sonc.2002.31527] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The use of tumor angiogenesis as a therapeutic target is based on extensive literature showing the dependence of tumors on the process of angiogenesis for growth, invasion, and metastasis. Seminal work performed by Folkman three decades ago determined that tumors beyond the size of approximately 2 mm require angiogenesis for subsequent growth and development. This basic hypothesis stimulated research in the field of angiogenesis and has resulted in the identification of factors that both enhance and inhibit this "angiogenic switch." The intent of this article is to present data on several angiogenesis inhibitors that are currently undergoing clinical evaluation in cancer patients. These agents may be particularly useful in the treatment of lung cancer, both as adjunctive therapy in early-stage or locally advanced disease, as well as in combination strategies with platinum-based therapy in metastatic disease. Although angiogenesis inhibitors have been in clinical trials for the past decade, there has been a shift in recent years towards the development of more mechanism-based and receptor-targeted agents. Interestingly, no antiangiogenic agent has been approved as such for use in cancer, perhaps because of the challenges involved in the clinical development of these novel agents. These include the potential requirement for long-term administration, difficulties in deriving biologically efficacious doses in early clinical trials, and the inability to use tumor regression as a primary endpoint in phase II trials.
Collapse
Affiliation(s)
- Roy S Herbst
- University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | | | | | | | | | | |
Collapse
|
178
|
Ruiter D, Bogenrieder T, Elder D, Herlyn M. Melanoma-stroma interactions: structural and functional aspects. Lancet Oncol 2002; 3:35-43. [PMID: 11905603 DOI: 10.1016/s1470-2045(01)00620-9] [Citation(s) in RCA: 169] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Cutaneous melanomas are notorious for their tendency to metastasise. Because the tumour microenvironment plays an important part in tumour development and progression, we review the structural and functional aspects of interactions between melanoma and the stroma. We emphasise fibrovascular patterns (both in uveal and cutaneous melanoma), cellular and extracellular composition of the stroma, and the molecules involved. Also, we discuss functional interactions, focusing on melanoma-fibroblast cross-talk by soluble factors and by direct cell-cell contact. On the basis of recent findings we propose that involvement of fibroblasts in melanoma-stromagenesis occurs through different stages: recruitment, activation, and conversion to myofibroblasts, or differentiation to fibrocytes. We reason that this involvement is topographically linked to different areas in and around the tumour, and hypothesise that stromal activation, as seen in tumor ulceration or immunological regression in melanoma, stimulates tumour progression.
Collapse
Affiliation(s)
- Dirk Ruiter
- The Wistar Institute, Philadelphia, PA, USA.
| | | | | | | |
Collapse
|
179
|
|
180
|
Elfman F, Bok R, Conn M, Shuman M, Cunha G. Urokinase plasminogen activator amino-terminal peptides inhibit development of the rat ventral prostate. Differentiation 2001; 69:108-20. [PMID: 11798065 DOI: 10.1046/j.1432-0436.2001.690205.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The plasma membrane urokinase plasminogen activator receptor (uPAR) localizes and enhances activation of pro-uPA. Active uPA, in turn, promotes increased degradation of the extracellular matrix (ECM) by activation of plasminogen. uPAR binds to ECM molecules and integrins, which can affect cellular adhesion, signal transduction, and gene regulation. The current study examines the expression and function of uPAR in developing rat ventral prostates (VPs). We report that newborn VPs express uPAR mRNA and protein. In addition, the function of uPAR-bound uPA during in vitro prostatic development was studied by adding recombinant peptide competitive inhibitors of uPA-uPAR binding. Newborn VP explants were cultured in serum-free media for one week with 10(-8) M testosterone plus chimeric peptides containing a human immunoglobulin G Fc domain and either human uPA amino acids 1-138 (hu-uPA 1-138) as a control or mouse uPA amino acids 1-138 (mo-uPA 1-138) or 1-48 (mo-uPA 1-48). Hu-uPA 1-138-treated VPs underwent normal ductal branching morphogenesis and tissue differentiation. In contrast, VPs treated with mo-uPA 1-138 or mo-uPA 1-48 displayed a dose-dependent perturbation of ductal branching. Differentiation of both epithelial and mesenchymal tissues was also impaired. Mo-uPA 1-48-treated VPs contained significantly more apoptotic cells. These observations suggest that disruption of uPA binding to uPAR results in a retardation of the development of newborn VPs.
Collapse
Affiliation(s)
- F Elfman
- Department of Anatomy, University of California, San Francisco 94143, USA
| | | | | | | | | |
Collapse
|
181
|
Seki T, Miyasu T, Noguchi T, Hamasaki A, Sasaki R, Ozawa Y, Okukita K, Declerck PJ, Ariga T. Reciprocal regulation of tissue-type and urokinase-type plasminogen activators in the differentiation of murine preadipocyte line 3T3-L1 and the hormonal regulation of fibrinolytic factors in the mature adipocytes. J Cell Physiol 2001; 189:72-8. [PMID: 11573206 DOI: 10.1002/jcp.1140] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Adipose tissue expresses a variety of genes including tumor necrosis factor alpha and type-1 plasminogen activator inhibitor (PAI-1); and these factors, produced by adipocytes, may be associated with the risk of coronary events in obesity. In this study, we characterized the production of fibrinolytic factors including tissue-type plasminogen activator (tPA), urokinase-type PA (uPA), and PAI-1 in the differentiation of preadipocytes, and examined the hormonal regulation of these fibrinolytic factors in mature adipocytes. Mouse 3T3-L1 preadipocytes were employed as a model of adipocytes. Adipocyte differentiation was induced by insulin, dexamethasone, and 3-isobutyl-1-methyl xanthine (IBMX). alpha-Glycerophosphate dehydrogenase (GPDH) activity and glucose transporter 4 (GLUT4) mRNA, indices for adipocyte maturation, were induced on Day 4, and gradually increased. GPDH activity reached its maximum level on Day 14. The level of tPA, a major PA in preadipocytes, dramatically decreased with differentiation. On the other hand, that of uPA reciprocally increased. PAI-1 production was also dramatically induced concomitant with differentiation. In mature adipocytes, uPA production was dominant (25 microg/ml/24 h vs. 0.8 microg/ml/24 h for tPA). Total PA activity in the mature adipocytes was reduced by insulin or dexamethasone, but not by glucagon. Insulin, IBMX, and dexamethasone significantly decreased both uPA and tPA production, and increased PAI-1 production. Glucagon had no effect on the production of these fibrinolytic factors. Our results reveal that uPA is one of the markers for the differentiation of 3T3-L1 cells and that insulin, IBMX, and dexamethasone are potent regulators of the fibrinolytic activity in differentiated 3T3-L1 cells, reciprocally affecting PA and PAI-1 levels in them.
Collapse
Affiliation(s)
- T Seki
- Department of Nutrition and Physiology, Nihon University College of Bioresource Sciences, Nihon University Graduate School of Applied Life Sciences, Kanagawa, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
182
|
Abstract
Cell migration over or through the extracellular matrix (ECM) is an integral feature of both physiological and pathological processes. Regulation of the changing cell-ECM interactions involved can be effected by proteolysis and requires strict spatial and temporal targeting of proteinase activity. The versatile use of different proteinase systems, with a variety of localisation mechanisms and cleavage targets, is being revealed by a plethora of studies using in vitro models. This mini review reflects the status of our knowledge of strategies for the localisation of proteolytic activity effected during cell migration.
Collapse
Affiliation(s)
- V Ellis
- School of Biological Sciences, University of East Anglia, Norwich NR4 7TJ, UK.
| | | |
Collapse
|
183
|
Andronicos NM, Ranson M. The topology of plasminogen binding and activation on the surface of human breast cancer cells. Br J Cancer 2001; 85:909-16. [PMID: 11556845 PMCID: PMC2375062 DOI: 10.1054/bjoc.2001.2022] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
The urokinase-dependent activation of plasminogen by breast cancer cells plays an important role in metastasis. We have previously shown that the metastatic breast cancer cell line MDA-MB-231 over-expresses urokinase and binds and efficiently activates plasminogen at the cell surface compared to non-metastatic cells. The aim of this study was to further characterise plasminogen binding and determine the topology of cell surface-bound plasminogen in terms of its potential for activation. The lysine-dependent binding of plasminogen at 4 degrees C to MDA-MB-231 cells was stable and resulted in an activation-susceptible conformation of plasminogen. Topologically, a fraction of bound plasminogen was co-localised with urokinase on the surfaces of MDA-MB-231 cells where it could be activated to plasmin. At 37 degrees C plasmin was rapidly lost from the cell surface. Apart from actin, other candidate plasminogen receptors were either not expressed or did not co-localise with plasminogen at the cell surface. Thus, based on co-localisation with urokinase, plasminogen binding is partitioned into two functional pools on the surface of MDA-MB-231 cells. In conclusion, these results shed further light on the functional organisation of the plasminogen activation cascade on the surface of a metastatic cancer cell.
Collapse
Affiliation(s)
- N M Andronicos
- Department of Biological Sciences, University of Wollongong, NSW Australia, 2522
| | | |
Collapse
|
184
|
Liu S, Bugge TH, Leppla SH. Targeting of tumor cells by cell surface urokinase plasminogen activator-dependent anthrax toxin. J Biol Chem 2001; 276:17976-84. [PMID: 11278833 DOI: 10.1074/jbc.m011085200] [Citation(s) in RCA: 131] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Urokinase plasminogen activator receptor (uPAR) binds pro-urokinase plasminogen activator (pro-uPA) and thereby localizes it near plasminogen, causing the generation of active uPA and plasmin on the cell surface. uPAR and uPA are overexpressed in a variety of human tumors and tumor cell lines, and expression of uPAR and uPA is highly correlated to tumor invasion and metastasis. To exploit these characteristics in the design of tumor cell-selective cytotoxins, we constructed mutated anthrax toxin-protective antigen (PrAg) proteins in which the furin cleavage site is replaced by sequences cleaved specifically by uPA. These uPA-targeted PrAg proteins were activated selectively on the surface of uPAR-expressing tumor cells in the presence of pro-uPA and plasminogen. The activated PrAg proteins caused internalization of a recombinant cytotoxin, FP59, consisting of anthrax toxin lethal factor residues 1-254 fused to the ADP-ribosylation domain of Pseudomonas exotoxin A, thereby killing the uPAR-expressing tumor cells. The activation and cytotoxicity of these uPA-targeted PrAg proteins were strictly dependent on the integrity of the tumor cell surface-associated plasminogen activation system. We also constructed a mutated PrAg protein that selectively killed tissue plasminogen activator-expressing cells. These mutated PrAg proteins may be useful as new therapeutic agents for cancer treatment.
Collapse
Affiliation(s)
- S Liu
- Oral Infection and Immunity Branch and Oral and Pharyngeal Cancer Branch, NIDCR, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | |
Collapse
|
185
|
Chappuis PO, Dieterich B, Sciretta V, Lohse C, Bonnefoi H, Remadi S, Sappino AP. Functional Evaluation of Plasmin Formation in Primary Breast Cancer. J Clin Oncol 2001; 19:2731-8. [PMID: 11352966 DOI: 10.1200/jco.2001.19.10.2731] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE: Plasmin generation is controlled by the plasminogen activators (PA)/plasmin system, which comprises proteases (urokinase-type PA [uPA] and tissue-type PA [tPA]) and antiproteases (PA inhibitors, PAI-1 and PAI-2). The tumoral content of uPA and PAI-1 has been shown to carry prognostic value in breast cancer; however, because most assays used so far have relied on immunometric determinations, we have explored the enzymatic activities governing plasmin formation in breast cancer specimens. PATIENTS AND METHODS: We applied semiquantitative histochemical zymography to 201 primary breast cancer tissue sections. Enzymatic activities were correlated with histopathologic parameters and clinical outcome. The median follow-up was 91 months. RESULTS: A wide range of PA-mediated catalytic activities was detected. The overall survival was significantly worse for patients with tumors showing tPA in the lowest quartile of activity (P = .003). The 5-year overall survival of patients with tPA activity in the lowest quartile was 58% compared with 81% for patients with tPA value in the other three quartiles. Tumor size, axillary lymph node metastasis, histologic grade, lymphovascular infiltration, TP53 mutation, and tPA activity were all major risk factors in univariate analysis. tPA activity was an independent prognostic factor in a multivariate Cox regression model, both in the whole population (relative risk = 0.5, 95% confidence interval, 0.3 to 0.9; P = .02) and in the node-negative subgroup (relative risk = 0.2, 95% confidence interval, 0.08 to 0.6; P = .004). CONCLUSION: By using a zymographic assay performed directly on primary tumor tissue sections, we demonstrate that reduced tPA-mediated plasmin production is an independent adverse prognostic factor in breast cancer.
Collapse
Affiliation(s)
- P O Chappuis
- Division of Oncology, Department of Medicine, Hôpitaux Universitaires, and Medical University Centre, Geneva, Switzerland.
| | | | | | | | | | | | | |
Collapse
|
186
|
Gavrilov D, Kenzior O, Evans M, Calaluce R, Folk WR. Expression of urokinase plasminogen activator and receptor in conjunction with the ets family and AP-1 complex transcription factors in high grade prostate cancers. Eur J Cancer 2001; 37:1033-40. [PMID: 11334730 DOI: 10.1016/s0959-8049(01)00077-6] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Expression of the urokinase plasminogen activator (uPA) and its receptor (uPAR) correlates with tumour cell invasiveness and helps to determine the prognosis of prostate and other cancers. The purpose of this study was to establish in prostate cancer, the ets family and AP-1 complex transcription factors that might activate the inducible AP-1 and AP-1/PEA3 elements of the uPA enhancer. uPA and uPAR were expressed preferentially in adenocarcinoma cells, but not the stroma of high grade prostate cancers. The ets family paralogues Fli-1 and Elf-1 were also highly expressed in adenocarcinoma cells of the majority of cancers, while Erg 1,2 and Ets-2 were expressed in a minority of cancers and Elk-1, PEA3 and PU.1 were minimally expressed. A minority of cancers expressed high levels of cytoplasmic and/or nuclear c-Jun and c-Fos transcription factors. We speculate as to the molecular basis for such expression.
Collapse
Affiliation(s)
- D Gavrilov
- Department of Biochemistry, 117 Scheitzer Hall, University of Missouri--Columbia, Columbia, MO 65211, USA
| | | | | | | | | |
Collapse
|
187
|
Oberst M, Anders J, Xie B, Singh B, Ossandon M, Johnson M, Dickson RB, Lin CY. Matriptase and HAI-1 are expressed by normal and malignant epithelial cells in vitro and in vivo. THE AMERICAN JOURNAL OF PATHOLOGY 2001; 158:1301-11. [PMID: 11290548 PMCID: PMC1891898 DOI: 10.1016/s0002-9440(10)64081-3] [Citation(s) in RCA: 153] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Matriptase and its cognate, Kunitz-type serine protease inhibitor, HAI-1, comprise a newly characterized extracellular matrix-degrading protease system that may function as an epithelial membrane activator for other proteases and latent growth factors. Both enzyme and inhibitor have been detected in breast cancer cells, immortalized mammary epithelial cells, and human milk, but not in cultured fibroblasts nor in fibrosarcoma cells. To test the hypothesis that this system is expressed by normal breast epithelium, invasive breast cancers, and other cancers of an epithelial origin (carcinomas) but not in cancers of a mesenchymal origin, we have expanded our expression analysis of matriptase and HAI-1 in vitro and in vivo. Matriptase and HAI-1 were detected at the protein and mRNA levels both in hormone-dependent and hormone-independent cultured breast cancer cells, and this expression correlated with the expression of the epithelial markers E-cadherin or ZO-1. However, none of the breast cancer cell lines tested that express the mesenchymal marker vimentin express matriptase or HAI-1, consistent with an epithelial-selective expression of this system. Expression of matriptase, as determined by Western blot analysis, was observed in primary human breast, gynecological, and colon carcinomas, but not in stromal-derived ovarian tumors and human sarcomas of various origins and histological grades. The epithelial-selective expression of matriptase and HAI-1 was further confirmed in human breast cancers by immunohistochemistry and in situ hybridization, where the expression of the protease and the inhibitor were found in the carcinoma cells and in surrounding normal breast epithelia. The expression of the matriptase/HAI-1 system by malignant epithelial cells in vivo suggests a possible role for this protease in multiple aspects of the pathophysiology of epithelial malignancy, including invasion and metastasis.
Collapse
Affiliation(s)
- M Oberst
- Lombardi Cancer Center, Georgetown University Medical Center, 3970 Reservoir Road NW, Washington, DC 20007, USA
| | | | | | | | | | | | | | | |
Collapse
|
188
|
Horino K, Kindezelskii AL, Elner VM, Hughes BA, Petty HR. Tumor cell invasion of model 3-dimensional matrices: demonstration of migratory pathways, collagen disruption, and intercellular cooperation. FASEB J 2001; 15:932-9. [PMID: 11292653 DOI: 10.1096/fj.00-0392com] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
We report a novel 3-dimensional model for visualizing tumor cell migration across a nylon mesh-supported gelatin matrix. To visualize migration across these model barriers, cell proteolytic activity of the pericellular matrix was detected using Bodipy-BSA (fluorescent upon proteolysis) and DQ collagen (fluorescent upon collagenase activity). For 3-dimensional image reconstruction, multiple optical images at sequential z axis positions were deconvoluted by computer analysis. Specificity was indicated using well-known inhibitors. Using these fluorescent proteolysis markers and imaging methods, we have directly demonstrated proteolytic and collagenolytic activity during tumor cell invasion. Moreover, it is possible to visualize migratory pathways followed by tumor cells during matrix invasion. Using cells of differing invasive potentials (uPAR-negative T-47D wild-type and uPAR-positive T-47D A2--1 cells), we show that the presence of the T-47D-A2--1 cells facilitates the entry of T-47D wild-type cells into the matrix. In some cases, wild-type cells follow T-47D A2--1 cells into the matrix whereas other T-47D-wild-type cells appear to enter without the direct intervention of T-47D A2--1 cells. Thus, we have developed a new 3-dimensional model of tumor cell invasion, demonstrated protein and collagen disruption, mapped the pathways followed by tumor cells during migration through an extracellular matrix, and illustrated cross-talk among tumor cell populations during invasion.
Collapse
Affiliation(s)
- K Horino
- Department of Biological Sciences, Wayne State University, Detroit, Michigan 48202, USA
| | | | | | | | | |
Collapse
|
189
|
Noël A, Albert V, Bajou K, Bisson C, Devy L, Frankenne F, Maquoi E, Masson V, Sounni NE, Foidart JM. New Functions of Stromal Proteases and Their Inhibitors in Tumor Progression. Surg Oncol Clin N Am 2001. [DOI: 10.1016/s1055-3207(18)30073-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
190
|
Horino K, Kindezelskii AL, Elner VM, Hughes BA, Petty HR. Tumor cell invasion of model 3‐dimensional matrices: demonstration of migratory pathways, collagen disruption, and intercellular cooperation. FASEB J 2001. [DOI: 10.1096/fsb2fj000392com] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Kei Horino
- Department of Biological SciencesWayne State UniversityDetroitMichigan48202USA
- Department of OphthalmologyUniversity of MichiganAnn ArborMichiganUSA
| | | | - Victor M. Elner
- Department of OphthalmologyUniversity of MichiganAnn ArborMichiganUSA
| | - Bret A. Hughes
- Department of OphthalmologyUniversity of MichiganAnn ArborMichiganUSA
| | - Howard R. Petty
- Department of Biological SciencesWayne State UniversityDetroitMichigan48202USA
| |
Collapse
|
191
|
Abstract
The role of proteases in general, and the matrix metalloproteinases in particular, in tumor invasion and metastasis is well established. However, the classic view that these enzymes simply provide a mechanism for the breakdown of connective tissue barriers has been challenged. This overview summarizes recent evidence to support the changing view of the role of matrix metalloproteinases in cancer progression. First we briefly review the central role of cell invasion in cancer progression and also the matrix metalloproteinase family members. We then focus on the emerging roles for these enzymes in cancer progression, including the role of matrix metalloproteinases in cell proliferation and release of growth factors, cell migration and in modification of the extracellular matrix to reveal cryptic sites that alter cell behaviour.
Collapse
Affiliation(s)
- W G Stetler-Stevenson
- Laboratory of Pathology, Division of Clinical Sciences, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | | |
Collapse
|
192
|
Radisky D, Hagios C, Bissell MJ. Tumors are unique organs defined by abnormal signaling and context. Semin Cancer Biol 2001; 11:87-95. [PMID: 11322828 DOI: 10.1006/scbi.2000.0360] [Citation(s) in RCA: 120] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Many cancer investigations have focussed on the eradication of the cancer cell itself and in doing so, overlook the inherent complexity and heterogeneity of solid tumors. Here, we argue that, in many cases, it is the altered communication within the tumor, rather than mutations per se, that is the defining characteristic of cancer. As a result, tumorigenesis can be indirectly initiated by environmental or inherited factors that affect the stromal cells. We propose that anticancer research might be more effective if aimed at eradicating the cause of abnormality rather than just treating the end result.
Collapse
Affiliation(s)
- D Radisky
- Life Science Division, Lawrence Berkeley National Laboratory, University of California, Berkeley, CA 94720, USA
| | | | | |
Collapse
|
193
|
Gall AL, Ruff M, Kannan R, Cuniasse P, Yiotakis A, Dive V, Rio MC, Basset P, Moras D. Crystal structure of the stromelysin-3 (MMP-11) catalytic domain complexed with a phosphinic inhibitor mimicking the transition-state. J Mol Biol 2001; 307:577-86. [PMID: 11254383 DOI: 10.1006/jmbi.2001.4493] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Stromelysin-3 (ST3) is a matrix metalloproteinase (MMP-11) whose proteolytic activity plays an important role in tumorigenicity enhancement. In breast cancer, ST3 is a bad prognosis marker: its expression is associated with a poor clinical outcome. This enzyme therefore represents an attractive therapeutic target. The topology of matrix metalloproteinases (MMPs) is remarkably well conserved, making the design of highly specific inhibitors difficult. The major difference between MMPs lies in the S(1)' subsite, a well-defined hydrophobic pocket of variable depth. The present crystal structure, the first 3D-structure of the ST3 catalytic domain in interaction with a phosphinic inhibitor mimicking a (d, l) peptide, clearly demonstrates that its S(1)' pocket corresponds to a tunnel running through the enzyme. This open channel is filled by the inhibitor P(1)' group which adopts a constrained conformation to fit this pocket, together with two water molecules interacting with the ST3-specific residue Gln215. These observations provide clues for the design of more specific inhibitors and show how ST3 can accommodate a phosphinic inhibitor mimicking a (d, l) peptide. The presence of a water molecule interacting with one oxygen atom of the inhibitor phosphinyl group and the proline residue of the Met-turn suggests how the intermediate formed during proteolysis may be stabilized. Furthermore, the hydrogen bond distance observed between the methyl of the phosphinic group and the carbonyl group of Ala182 mimics the interaction between this carbonyl group and the amide group of the cleaved peptidic bond. Our crystal structure provides a good model to study the MMPs mechanism of proteolysis.
Collapse
Affiliation(s)
- A L Gall
- Structural Biology and Genomics Laboratory, I.G.B.M.C., B.P. 163, F67404, Illkirch Cedex, France
| | | | | | | | | | | | | | | | | |
Collapse
|
194
|
Moriya Y, Niki T, Yamada T, Matsuno Y, Kondo H, Hirohashi S. Increased expression of laminin-5 and its prognostic significance in lung adenocarcinomas of small size. An immunohistochemical analysis of 102 cases. Cancer 2001; 91:1129-41. [PMID: 11267958 DOI: 10.1002/1097-0142(20010315)91:6<1129::aid-cncr1109>3.0.co;2-c] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
BACKGROUND Laminin-5 plays an important role in cell migration during tissue remodeling and tumor invasion. METHODS The authors studied the expression of laminin-5 immunohistochemically in 102 cases of small-sized lung adenocarcinoma (maximum dimension < or = 2 cm) using a monoclonal antibody against the laminin gamma2 chain, and they also investigated the associations of laminin-5 with clinicopathologic characteristics. Prognostic significance of increased laminin-5 expression was evaluated using the Kaplan-Meier method and the Cox proportional hazard model. RESULTS Overall, laminin-5 expression was observed in 82 cases (80.4%): 7 of 18 (38.9%) bronchioloalveolar carcinomas and 75 of 84 (89.3%) invasive adenocarcinomas. Laminin-5 was preferentially localized in the cytoplasm of tumor cells at the tumor-stromal interface, where budding or dissociation of cancer cells was frequently observed. Overexpression of laminin-5 (24 cases, 23.5%) was associated with vascular invasion (P = 0.021) and stromal fibroblastic reaction (P = 0.005) but not with nodal involvement, lymphatic invasion, or pleural invasion. Survival analysis revealed that overexpression of laminin-5 was associated with shorter patient survival (P = 0.0027 by log rank test). On multivariate analysis, overexpression of laminin-5 was an independent prognostic factor (P = 0.030), as were nodal involvement (P < 0.0001), vascular invasion (P = 0.047), and lymphatic invasion (P = 0.0047) in a whole cohort of patients. Moreover, when patients with Stage I (International Union Against Cancer [UICC] staging system) disease were considered in multivariate analysis, overexpression of laminin-5 was the only significant prognostic factor (P = 0.022), whereas vascular invasion had a marginally significant impact (P = 0.07) on patient survival. CONCLUSIONS The authors' results showed that laminin-5 is frequently expressed by cancer cells at the invasive front of lung adenocarcinoma. The study concluded that overexpression of laminin-5 may be a useful prognostic factor in patients with small-sized lung adenocarcinoma, especially in Stage I cases.
Collapse
Affiliation(s)
- Y Moriya
- Diagnostic Pathology, Clinical Laboratory Division, National Cancer Center Hospital, Tokyo, Japan
| | | | | | | | | | | |
Collapse
|
195
|
Rømer J, Pyke C, Lund LR, Ralfkiaer E, Danø K. Cancer cell expression of urokinase-type plasminogen activator receptor mRNA in squamous cell carcinomas of the skin. J Invest Dermatol 2001; 116:353-8. [PMID: 11231307 DOI: 10.1046/j.1523-1747.2001.01241.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
In this study we have used in situ hybridization with radiolabeled antisense RNA probes to examine the expression of mRNA for urokinase-type plasminogen activator and its receptor in histologic samples of squamous cell (n = 7) and basal cell (n = 7) carcinomas of the skin. Messenger RNA for both urokinase-type plasminogen activator and its receptor were expressed in all of the squamous cell carcinomas, but could not be detected in the basal cell carcinomas. In all of the seven squamous cell carcinomas a signal for urokinase-type plasminogen activator receptor mRNA was detected focally in well-differentiated cancer cells surrounding keratinized pearls, and in four specimens urokinase-type plasminogen activator receptor mRNA was in addition expressed by cancer cells at the edge of invasively growing strands of tumor. Urokinase-type plasminogen activator mRNA expression was found in virtually all the cancer cells of the squamous cell carcinomas, and importantly we found, by hybridizations for urokinase-type plasminogen activator and its receptor mRNA on adjacent sections of squamous cell carcinomas, that it was exactly the invading cancer cells that simultaneously expressed both these components required for plasmin-mediated proteolysis at the cell surface. We have previously shown that both urokinase-type plasminogen activator and its receptor mRNA are expressed by the leading-edge keratinocytes in regenerating epidermis during mouse skin wound healing, and that wound healing is impaired in mice made deficient in plasminogen by targeted gene disruption. We propose that there are similarities between the mechanisms of generation and regulation of extracellular proteolysis during skin re-epithelialization and squamous cell carcinoma invasion. The ability of the squamous carcinoma cells to mimic the "invasive" phenotype of re-epithelializing keratinocytes may be one of the factors that make squamous cell carcinomas more aggressive tumors than basal cell carcinomas.
Collapse
Affiliation(s)
- J Rømer
- The Finsen Laboratory, Rigshospitalet, Copenhagen, Denmark
| | | | | | | | | |
Collapse
|
196
|
Hidalgo M, Eckhardt SG. Development of matrix metalloproteinase inhibitors in cancer therapy. J Natl Cancer Inst 2001; 93:178-93. [PMID: 11158186 DOI: 10.1093/jnci/93.3.178] [Citation(s) in RCA: 560] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The matrix metalloproteinases (MMPs) are a family of zinc-dependent proteinases involved in the degradation of the extracellular matrix. The MMPs have been implicated in the processes of tumor growth, invasion, and metastasis; are frequently overexpressed in malignant tumors; and have been associated with an aggressive malignant phenotype and adverse prognosis in patients with cancer. A number of MMP inhibitors are being developed for the treatment of cancer. The most extensively studied class of MMP inhibitors includes collagen peptidomimetics and nonpeptidomimetic inhibitors of the MMP active site, tetracycline derivatives, and bisphosphonates. The hydroxamate peptidomimetic inhibitor batimastat and its orally bioavailable analogue marimastat, which bind covalently to the zinc atom at the MMP-active site, were the first MMP inhibitors to be studied in detail. Marimastat is currently being studied in randomized clinical trials. The nonpeptidic MMP inhibitors were synthesized in an attempt to improve the oral bioavailability and pharmaceutical properties of the peptidic inhibitors. Several members of this class of compounds are undergoing evaluation in phase III clinical trials. The tetracyclines and, particularly, the nonantibiotic chemically modified tetracyclines, interfere with several aspects of MMP expression and activation and inhibit tumor growth and metastases in preclinical models. A representative agent of this class, Col-3, is currently undergoing phase I clinical trials. The development of the MMP inhibitors, like that of other targeted and predominantly antiproliferative compounds, poses a challenge because the paradigms that have governed the design of clinical oncology trials may not be relevant to this new class of agents. The anticipated need for long-term administration of these drugs, together with their cytostatic mechanism of action, will require novel clinical trial design strategies.
Collapse
Affiliation(s)
- M Hidalgo
- Institute for Drug Development, The University of Texas Health Science Center at San Antonio, 78229, USA.
| | | |
Collapse
|
197
|
Coleman JL, Gebbia JA, Benach JL. Borrelia burgdorferi and other bacterial products induce expression and release of the urokinase receptor (CD87). JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 166:473-80. [PMID: 11123326 DOI: 10.4049/jimmunol.166.1.473] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The urokinase-type plasminogen activator receptor (uPAR, CD87) is a highly glycosylated 55- to 60-kDa protein anchored to the cell membrane through a glycosylphosphatidylinositol moiety that promotes the acquisition of plasmin on the surface of cells and subsequent cell movement and migration by binding urokinase-type plasminogen activator. uPAR also occurs in a soluble form in body fluids and tumor extracts, and both membrane and soluble uPAR are overexpressed in patients with tumors. uPAR may be a factor in inflammatory disorders as well. We investigated whether Borrelia burgdorferi could stimulate up-regulation of cell membrane uPAR in vitro. B. burgdorferi, purified native outer surface protein A, and a synthetic outer surface protein A hexalipopeptide stimulated human monocytes to up-regulate membrane uPAR as measured by immunofluorescence/FACS and Western blot. The presence of soluble uPAR in culture supernatants, measured by Ag capture ELISA, was also observed. LPS from Salmonella typhimurium and lipotechoic acid from Streptococcus pyogenes also induced the up-regulation of both membrane and soluble uPAR protein by monocytes. Up-regulation of uPAR was induced by conditioned medium from B. burgdorferi/monocyte cocultures. The up-regulation of uPAR by B. burgdorferi was concomitant with an increase in uPAR mRNA, indicating that synthesis was de novo. The expression and release of uPAR in response to B. burgdorferi and other bacterial components suggests a role in the pathogenesis of Lyme disease as well as in other bacterial infections.
Collapse
MESH Headings
- Animals
- Antigens, Surface/pharmacology
- Bacterial Outer Membrane Proteins/pharmacology
- Bacterial Vaccines
- Borrelia burgdorferi Group/growth & development
- Borrelia burgdorferi Group/immunology
- Borrelia burgdorferi Group/pathogenicity
- Cell Membrane/metabolism
- Cell Membrane/microbiology
- Cells, Cultured
- Culture Media, Conditioned/metabolism
- Culture Media, Conditioned/pharmacology
- Humans
- Lipopolysaccharides/immunology
- Lipopolysaccharides/pharmacology
- Lipoproteins
- Lyme Disease Vaccines/pharmacology
- Mice
- Mice, Inbred C3H
- Monocytes/immunology
- Monocytes/metabolism
- Monocytes/microbiology
- Plasminogen Activators/biosynthesis
- Plasminogen Activators/genetics
- Plasminogen Activators/metabolism
- Protein Isoforms/biosynthesis
- Protein Isoforms/metabolism
- RNA, Messenger/biosynthesis
- Receptors, Cell Surface/biosynthesis
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/metabolism
- Receptors, Urokinase Plasminogen Activator
- Salmonella typhimurium/immunology
- Solubility
- Streptococcus pyogenes/immunology
- Teichoic Acids/immunology
- Transcription, Genetic/immunology
- U937 Cells
- Up-Regulation/genetics
- Up-Regulation/immunology
Collapse
Affiliation(s)
- J L Coleman
- State of New York Department of Health, State University of New York, Stony Brook, NY 11794-5120, USA.
| | | | | |
Collapse
|
198
|
Lindberg P, Kinnby B, Lecander I, Lang NP, Matsson L. Increasing expression of tissue plasminogen activator and plasminogen activator inhibitor type 2 in dog gingival tissues with progressive inflammation. Arch Oral Biol 2001; 46:23-31. [PMID: 11163592 DOI: 10.1016/s0003-9969(00)00098-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Urokinase and tissue-type plasminogen activators (u--PA and t--PA) are serine proteases that convert plasminogen into plasmin, which degrades matrix proteins and activates metalloproteinases. The PAs are balanced by specific inhibitors (PAI--1 and PAI--2). Local production of t--PA and PAI--2 was recently demonstrated in human gingival tissues. The aim now was to investigate the production and localization of t--PA and PAI--2 in gingival tissues from dogs in three well-defined periodontal conditions; clinically healthy gingiva, chronic gingivitis and an initial stage of ligature-induced loss of attachment. At the start of the experiment the gingiva showed clear signs of inflammation. Clinically healthy gingiva were obtained after 21 days period of intense oral hygiene. Attachment loss was induced by placing rubber ligatures around the neck of some teeth. Biopsies were taken from areas representing the different conditions and prepared for in situ hybridization and immunohistochemistry. In clinically healthy gingiva both t--PA mRNA and antigen were expressed in a thin outer layer of the sulcular and junctional epithelia. No t--PA signals or staining were seen in connective tissue. Both mRNA signaling and immunostaining for t--PA were stronger in chronic gingivitis. In areas with loss of attachment, t--PA mRNA as well as antigen were found in the sulcular and junctional epithelia to a similar degree as in gingivitis. Occasionally the connective tissue was involved, especially in connection with vessels. PAI--2 mRNA was seen in a thin outer layer of the sulcular and junctional epithelia in clinically healthy gingiva, but no signals were seen in connective tissue. PAI--2 antigen was found primarily in the outer layer of the sulcular and junctional epithelia. Some cells in the connective tissue were stained. In gingivitis, PAI--2 signals were mainly found in the same locations, but more intense and extending towards the connective tissue. Immunostaining was seen in the outer half of the sulcular and junctional epithelia as well as in the upper part of the connective tissue, close to the sulcular epithelium. In sites with loss of attachment, PAI--2 mRNA was found throughout the sulcular and junctional epithelia, as was the antigen, which stained intensely. No PAI--2 mRNA was seen in connective tissue; the antigen was found scattered, especially near vessels. This study shows that the expression of both t--PA and PAI--2 increases with experimental gingival inflammation in the dog, and furthermore, the two techniques demonstrate a strong correlation between the topographical distribution of the site of protein synthesis and the tissue location of the antigens for both t--PA and PAI--2. The distribution correlates well with previous findings in humans.
Collapse
Affiliation(s)
- P Lindberg
- Center for Oral Health Sciences, Malmö University, S-214 21 Malmö, Sweden.
| | | | | | | | | |
Collapse
|
199
|
Lee SL, Dickson RB, Lin CY. Activation of hepatocyte growth factor and urokinase/plasminogen activator by matriptase, an epithelial membrane serine protease. J Biol Chem 2000; 275:36720-5. [PMID: 10962009 DOI: 10.1074/jbc.m007802200] [Citation(s) in RCA: 291] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Matriptase is an epithelial-derived, integral membrane serine protease. The enzyme was initially isolated from human breast cancer cells and has been implicated in breast cancer invasion and metastasis. In the current study, using active matriptase isolated from human milk, we demonstrate that matriptase is able to cleave various synthetic substrates with arginine or lysine as their P1 sites and prefers small side chain amino acids, such as Ala and Gly, at P2 sites. For the most reactive substrates, N-tert-butoxycarbonyl (N-t-Boc)-gamma-benzyl-Glu-Ala-Arg-7-amino-4-methylcoumarin (AMC) and N-t-Boc-Gln-Ala-Arg-AMC, the K(m) values were determined to be 3. 81 and 4.89 microm, respectively. We further demonstrated that matriptase can convert hepatocyte growth factor/scattering factor to its active form, which can induce scatter of Madin-Darby canine kidney epithelial cells and can activate c-Met tyrosine phosphorylation in A549 human lung carcinoma cells. In addition, we noted that matriptase can activate urokinase plasminogen activator but has no affect on plasminogen. These results suggest that matriptase could act as an epithelial, upstream membrane activator to recruit and activate stromal-derived downstream effectors important for extracellular matrix degradation and epithelial migration, two major events of tissue remodeling, cancer invasion, and metastasis.
Collapse
Affiliation(s)
- S L Lee
- Department of Oncology, Lombardi Cancer Center, Georgetown University, Medical Center, Washington, DC 20007, USA
| | | | | |
Collapse
|
200
|
Description of the plasminogen activating system in canine gingival crevicular fluid. ACTA ACUST UNITED AC 2000. [DOI: 10.1054/fipr.2000.0091] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|