1
|
Tune BXJ, Sim MS, Poh CL, Guad RM, Woon CK, Hazarika I, Das A, Gopinath SCB, Rajan M, Sekar M, Subramaniyan V, Fuloria NK, Fuloria S, Batumalaie K, Wu YS. Matrix Metalloproteinases in Chemoresistance: Regulatory Roles, Molecular Interactions, and Potential Inhibitors. JOURNAL OF ONCOLOGY 2022; 2022:3249766. [PMID: 35586209 PMCID: PMC9110224 DOI: 10.1155/2022/3249766] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 04/11/2022] [Accepted: 04/19/2022] [Indexed: 02/08/2023]
Abstract
Cancer is one of the major causes of death worldwide. Its treatments usually fail when the tumor has become malignant and metastasized. Metastasis is a key source of cancer recurrence, which often leads to resistance towards chemotherapeutic agents. Hence, most cancer-related deaths are linked to the occurrence of chemoresistance. Although chemoresistance can emerge through a multitude of mechanisms, chemoresistance and metastasis share a similar pathway, which is an epithelial-to-mesenchymal transition (EMT). Matrix metalloproteinases (MMPs), a class of zinc and calcium-chelated enzymes, are found to be key players in driving cancer migration and metastasis through EMT induction. The aim of this review is to discuss the regulatory roles and associated molecular mechanisms of specific MMPs in regulating chemoresistance, particularly EMT initiation and resistance to apoptosis. A brief presentation on their potential diagnostic and prognostic values was also deciphered. It also aimed to describe existing MMP inhibitors and the potential of utilizing other strategies to inhibit MMPs to reduce chemoresistance, such as upstream inhibition of MMP expressions and MMP-responsive nanomaterials to deliver drugs as well as epigenetic regulations. Hence, manipulation of MMP expression can be a powerful tool to aid in treating patients with chemo-resistant cancers. However, much still needs to be done to bring the solution from bench to bedside.
Collapse
Affiliation(s)
- Bernadette Xin Jie Tune
- Department of Pharmaceutical Life Sciences, Faculty of Pharmacy, Universiti Malaya, Kuala Lumpur 50603, Malaysia
| | - Maw Shin Sim
- Department of Pharmaceutical Life Sciences, Faculty of Pharmacy, Universiti Malaya, Kuala Lumpur 50603, Malaysia
| | - Chit Laa Poh
- Centre for Virus and Vaccine Research, School of Medical and Life Sciences, Sunway University, Selangor 47500, Malaysia
| | - Rhanye Mac Guad
- Department of Biomedical Science and Therapeutics, Faculty of Medicine and Health Science, Universiti Malaysia Sabah, Kota Kinabalu, 88400 Sabah, Malaysia
| | - Choy Ker Woon
- Department of Anatomy, Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh, 47000 Selangor, Malaysia
| | - Iswar Hazarika
- Department of Pharmacology, Girijananda Chowdhury Institute of Pharmaceutical Science, Guwahati 781017, India
| | - Anju Das
- Department of Pharmacology, Royal School of Pharmacy, Royal Global University, Guwahati 781035, India
| | - Subash C. B. Gopinath
- Faculty of Chemical Engineering Technology, Universiti Malaysia Perlis (UniMAP), Arau, 02600 Perlis, Malaysia
- Institute of Nano Electronic Engineering, Universiti Malaysia Perlis, Kangar, 01000 Perlis, Malaysia
| | - Mariappan Rajan
- Department of Natural Products Chemistry, School of Chemistry, Madurai Kamaraj University, Madurai 625021, India
| | - Mahendran Sekar
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Health Sciences, Royal College of Medicine Perak, Universiti Kuala Lumpur, Ipoh 30450, Perak, Malaysia
| | - Vetriselvan Subramaniyan
- Department of Pharmacology, School of Medicine, Faculty of Medicine, Bioscience and Nursing, MAHSA University, Selangor 42610, Malaysia
| | | | - Shivkanya Fuloria
- Faculty of Pharmacy, AIMST University, Semeling, Bedong, Kedah 08100, Malaysia
| | - Kalaivani Batumalaie
- Department of Biomedical Sciences, Faculty of Health Sciences, Asia Metropolitan University, 81750 Johor Bahru, Malaysia
| | - Yuan Seng Wu
- Centre for Virus and Vaccine Research, School of Medical and Life Sciences, Sunway University, Selangor 47500, Malaysia
- Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, Selangor 47500, Malaysia
| |
Collapse
|
2
|
Abstract
A compelling long-term goal of cancer biology is to understand the crucial players during tumorigenesis in order to develop new interventions. Here, we review how the four non-redundant tissue inhibitors of metalloproteinases (TIMPs) regulate the pericellular proteolysis of a vast range of matrix and cell surface proteins, generating simultaneous effects on tumour architecture and cell signalling. Experimental studies demonstrate the contribution of TIMPs to the majority of cancer hallmarks, and human cancers invariably show TIMP deregulation in the tumour or stroma. Of the four TIMPs, TIMP1 overexpression or TIMP3 silencing is consistently associated with cancer progression or poor patient prognosis. Future efforts will align mouse model systems with changes in TIMPs in patients, will delineate protease-independent TIMP function, will pinpoint therapeutic targets within the TIMP-metalloproteinase-substrate network and will use TIMPs in liquid biopsy samples as biomarkers for cancer prognosis.
Collapse
Affiliation(s)
- Hartland W Jackson
- Department of Medical Biophysics, University of Toronto, Princess Margaret Cancer Centre, TMDT 301-13, 101 College Street, Toronto, Ontario, M5G IL7 Canada
- Bodenmiller Laboratory, University of Zürich, Institute for Molecular Life Sciences, Winterthurstrasse 190, 8057 Zürich, Switzerland
| | - Virginie Defamie
- Department of Medical Biophysics, University of Toronto, Princess Margaret Cancer Centre, TMDT 301-13, 101 College Street, Toronto, Ontario, M5G IL7 Canada
| | - Paul Waterhouse
- Department of Medical Biophysics, University of Toronto, Princess Margaret Cancer Centre, TMDT 301-13, 101 College Street, Toronto, Ontario, M5G IL7 Canada
| | - Rama Khokha
- Department of Medical Biophysics, University of Toronto, Princess Margaret Cancer Centre, TMDT 301-13, 101 College Street, Toronto, Ontario, M5G IL7 Canada
| |
Collapse
|
3
|
Maral S, Acar M, Balcik OS, Uctepe E, Hatipoglu OF, Akdeniz D, Altun HU, Kosar A, Gunduz M, Gunduz E. Matrix Metalloproteinases 2 and 9 Polymorphism in Patients With Myeloproliferative Diseases: A STROBE-Compliant Observational Study. Medicine (Baltimore) 2015; 94:e732. [PMID: 25906101 PMCID: PMC4602695 DOI: 10.1097/md.0000000000000732] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2014] [Revised: 02/22/2015] [Accepted: 03/11/2015] [Indexed: 01/15/2023] Open
Abstract
Chronic myeloproliferative disorders such as polycythemia vera (PV), essential thrombocytosis (ET), and idiopathic myelofibrosis arise from clonal proliferation of neoplastic stem cells in the bone marrow. Matrix metalloproteinases (MMPs) are a family of zinc-dependent endopeptidases that have potential to degrade all types of extracellular matrix (ECM) and also play a role in remodeling of the ECM. It is known that MMPs play a role in bone marrow remodeling.The primary goal of our study is to explore the relationship between chronic myeloproliferative diseases and some of MMP gene polymorphisms. The demonstration of a relationship will help to understand whether these polymorphisms may be a potential early diagnosis marker of the diseases.Patients were selected from outpatient clinics of Turgut Ozal University Hospital, Ankara, Turkey, between December 2010 and May 2011. Twenty-eight patients that previously diagnosed and followed-up with PV, 17 with secondary polycythemia (SP), and 12 with ET were enrolled in the study, along with a control group of 22 healthy people.DNA was isolated from peripheral blood. Using polymerase chain reaction-restriction fragment length polymorphism method, MMP2 and MMP9 gene polymorphisms were analyzed with agarose gel electrophoresis. There was a statistically significant difference between the study groups and the control group in terms of Gln279Arg polymorphisms rates of MMP9. The highest MMP9 Gln279Arg polymorphism rate was observed in the ET group. But nobody from the control group had polymorphic MMP9. There was no statistically significant difference between the groups in terms of MMP2-735 C > T polymorphism rates.In conclusion, MMP9 gene Gln279Arg polymorphism was associated with ET, SP, and PV diseases. Hence, we believe that these gene polymorphisms may play a role in the mechanism of bone marrow fibrosis and may be a factor that increases the risk of thrombosis. Illumination of the molecular basis of the relationship between MMP-thrombosis and MMP-fibrosis provides a better understanding of the pathophysiology of PV and ET diseases and will allow new approaches to diagnosis and treatment.
Collapse
Affiliation(s)
- Senem Maral
- From the Department of Internal Medicine (SM, DA); Division of Hematology (OSB, AK); Department of Medical Genetics (MA, EU, OFH, MG, EG); Department of Medical Microbiology (HUA); and Department of Otolaryngology (MG), Turgut Özal University Faculty of Medicine, Ankara, Turkey
| | | | | | | | | | | | | | | | | | | |
Collapse
|
4
|
Jacob A, Prekeris R. The regulation of MMP targeting to invadopodia during cancer metastasis. Front Cell Dev Biol 2015; 3:4. [PMID: 25699257 PMCID: PMC4313772 DOI: 10.3389/fcell.2015.00004] [Citation(s) in RCA: 214] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 01/09/2015] [Indexed: 01/07/2023] Open
Abstract
The dissemination of cancer cells from the primary tumor to a distant site, known as metastasis, is the main cause of mortality in cancer patients. Metastasis is a very complex cellular process that involves many steps, including the breaching of the basement membrane (BM) to allow the movement of cells through tissues. The BM breach occurs via highly regulated and localized remodeling of the extracellular matrix (ECM), which is mediated by formation of structures, known as invadopodia, and targeted secretion of matrix metalloproteinases (MMPs). Recently, invadopodia have emerged as key cellular structures that regulate the metastasis of many cancers. Furthermore, targeting of various cytoskeletal modulators and MMPs has been shown to play a major role in regulating invadopodia function. Here, we highlight recent findings regarding the regulation of protein targeting during invadopodia formation and function.
Collapse
Affiliation(s)
- Abitha Jacob
- Department of Cell and Developmental Biology, School of Medicine, Anschutz Medical Campus, University of Colorado Denver Aurora, CO, USA
| | - Rytis Prekeris
- Department of Cell and Developmental Biology, School of Medicine, Anschutz Medical Campus, University of Colorado Denver Aurora, CO, USA
| |
Collapse
|
5
|
O'Kane D, Jackson MV, Kissenpfennig A, Spence S, Damkat-Thomas L, Tolland JP, Smyth AE, Denton CP, Stuart Elborn J, McAuley DF, O'Kane CM. SMAD inhibition attenuates epithelial to mesenchymal transition by primary keratinocytesin vitro. Exp Dermatol 2014; 23:497-503. [DOI: 10.1111/exd.12452] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/16/2014] [Indexed: 12/26/2022]
Affiliation(s)
- Donal O'Kane
- Centre For Infection and Immunity; Queen's University Belfast; Belfast UK
| | - Megan V. Jackson
- Centre For Infection and Immunity; Queen's University Belfast; Belfast UK
| | | | - Shaun Spence
- Centre For Infection and Immunity; Queen's University Belfast; Belfast UK
| | | | | | | | - Christopher P. Denton
- Centre for Rheumatology and Connective Tissue Disease; University College London; London UK
| | - J. Stuart Elborn
- Centre For Infection and Immunity; Queen's University Belfast; Belfast UK
| | - Daniel F. McAuley
- Centre For Infection and Immunity; Queen's University Belfast; Belfast UK
| | - Cecilia M. O'Kane
- Centre For Infection and Immunity; Queen's University Belfast; Belfast UK
| |
Collapse
|
6
|
Kim SK, Kim SY, Kim JH, Roh SA, Cho DH, Kim YS, Kim JC. A nineteen gene-based risk score classifier predicts prognosis of colorectal cancer patients. Mol Oncol 2014; 8:1653-66. [PMID: 25049118 DOI: 10.1016/j.molonc.2014.06.016] [Citation(s) in RCA: 123] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Accepted: 06/16/2014] [Indexed: 12/30/2022] Open
Abstract
Colorectal cancer (CRC) patients frequently experience disease recurrence and distant metastasis. This study aimed to identify prognostic indicators, including individual responses to chemotherapy, in CRC patients. RNA-seq data was generated using 54 samples (normal colon, primary CRC, and liver metastases) from 18 CRC patients and genes associated with CRC aggressiveness were identified. A risk score based on these genes was developed and validated in four independent CRC patient cohorts (n = 1063). Diverse statistical methods were applied to validate the risk scoring system, including a generalized linear model likelihood ratio test, Kaplan-Meier curves, a log-rank test, and the Cox model. TREM1 and CTGF were identified as two activated regulators associated with CRC aggressiveness. A risk score based on 19 genes regulated by TREM1 or CTGF activation (TCA19) was a significant prognostic indicator. In multivariate and subset analyses based on pathological staging, TCA19 was an independent risk factor (HR = 1.894, 95% CI = 1.227-2.809, P = 0.002). Subset stratification in stage III patients revealed that TCA19 had prognostic potential and identified patients who would benefit from adjuvant chemotherapy, regardless of age. The TCA19 predictor represents a novel diagnostic tool for identifying high-risk CRC patients and possibly predicting the response to adjuvant chemotherapy.
Collapse
Affiliation(s)
- Seon-Kyu Kim
- Medical Genomics Research Centre, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Korea
| | - Seon-Young Kim
- Medical Genomics Research Centre, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Korea
| | - Jeong-Hwan Kim
- Medical Genomics Research Centre, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Korea
| | - Seon Ae Roh
- Department of Surgery, University of Ulsan College of Medicine, Seoul, Korea; Department of Cancer Research, Institute of Innovative Cancer Research and Asan Institute for Life Sciences, Asan Medical Centre, Seoul, Korea
| | - Dong-Hyung Cho
- Department of Cancer Research, Institute of Innovative Cancer Research and Asan Institute for Life Sciences, Asan Medical Centre, Seoul, Korea; Graduate School of East-West Medical Science, Kyung Hee University, Gyeonggi-do, Korea
| | - Yong Sung Kim
- Medical Genomics Research Centre, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Korea; Department of Cancer Research, Institute of Innovative Cancer Research and Asan Institute for Life Sciences, Asan Medical Centre, Seoul, Korea.
| | - Jin Cheon Kim
- Department of Surgery, University of Ulsan College of Medicine, Seoul, Korea; Department of Cancer Research, Institute of Innovative Cancer Research and Asan Institute for Life Sciences, Asan Medical Centre, Seoul, Korea.
| |
Collapse
|
7
|
Zhao X, Qian Y, Zhou YL, Wang R, Wang Q, Li GJ. Pu-erh tea has in vitro anticancer activity in TCA8113 cells and preventive effects on buccal mucosa cancer in U14 cells injected mice in vivo. Nutr Cancer 2014; 66:1059-69. [PMID: 24945996 DOI: 10.1080/01635581.2014.916317] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Pu-erh tea is a functional tea production in China. The functional effects should be proved. The oral cancer preventive and antimetastatic effects of Pu-erh tea in vitro and in vivo have been studied respectively. Pu-erh tea showed an inhibitory effect on human tongue carcinoma TCA8113 cells proliferation tested by 3-(4,5-Dimethyl-2-Thiazolyl)-2,5-Diphenyltetrazolium Bromide assay and induced TCA8113 apoptosis shown anticancer effect. The antimetastatic effect of Pu-erh tea in TCA8113 cells was proved by the decreasing of matrix metalloproteinases (MMPs) and increasing of tissue inhibitors of metalloproteinases (TIMPs) mRNA transcription. In the animal experiments, the tumor volumes and lymph node metastasis rates of Pu-erh tea-treated mice were smaller than control mice. Pu-erh tea reduced the levels of the serum proinflammatory cytokines interleukin (IL)-6, IL-12, tumor necrosis factor-α, and interferon-γ to a greater extent compared with the control mice, and the levels of 200 μg/mL treatment was more close to the normal mice than 100 μg/mL treated mice. Pu-erh tea also significantly induced apoptosis in tissues of mice (P < 0.05) by upregulating Bax and downregulating Bcl-2. These results demonstrate Pu-erh tea has cancer preventive and anti-metastatic effects on buccal mucosa cancer, the higher concentration get better efficiency.
Collapse
Affiliation(s)
- Xin Zhao
- a Department of Biological and Chemical Engineering , Chongqing University of Education , Chongqing , People's Republic of China
| | | | | | | | | | | |
Collapse
|
8
|
Zhu K, Li G, Sun P, Wang R, Qian Y, Zhao X. In vitro and in vivo anti-cancer activities of Kuding tea ( Ilex kudingcha C.J. Tseng) against oral cancer. Exp Ther Med 2013; 7:709-715. [PMID: 24520272 PMCID: PMC3919927 DOI: 10.3892/etm.2013.1450] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2013] [Accepted: 12/05/2013] [Indexed: 12/23/2022] Open
Abstract
Kuding tea (Ilex kudingcha C.J. Tseng) is drunk widely in China. The in vitro anticancer effects of Kuding tea were evaluated in TCA8113 human tongue carcinoma cells using a 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyltetrazolium bromide (MTT) assay. At a concentration of 200 μg/ml, Kuding tea exhibited an inhibitory effect of 75% in TCA8113 cells, which was higher than that observed at concentrations of 100 and 50 μg/ml (41 and 10% inhibition, respectively). Reverse transcription-polymerase chain reaction (RT-PCR) and western blot analyses of the apoptosis, inflammation and metastasis genes and proteins in Kuding tea-treated cancer cells were performed. Kuding tea significantly induced apoptosis in TCA8113 cancer cells (P<0.05) by upregulating Bax, caspase-3 and caspase-9 expression, and downregulating Bcl-2 expression. Expression of the NF-κB, iNOS and COX-2 genes that are associated with inflammation was significantly downregulated by Kuding tea, which demonstrated its anti-inflammatory properties. Kuding tea also exerted an anti-metastatic effect on cancer cells. This was demonstrated by the decreased expression of matrix metalloproteases (MMPs) and the increased expression of tissue inhibitors of metalloproteinases (TIMPs), and confirmed by the inhibition of the metastasis of U14 squamous cell carcinoma cells in imprinting control region (ICR) mice. The ICR mouse buccal mucosa cancer model was established by injecting the mice with U14 cells. Following injection, the wound at the injection site was topically treated with Kuding tea. It was observed that the tumor volumes for the group treated with Kuding tea were smaller than those from the control mice. Analysis of the sections of buccal mucosa cancer tissue demonstrated that the buccal mucosa cancer degrees of the Kuding tea-treated mice were weaker than that in the control mice. Similar results were observed in the lesion sections of the cervical lymph nodes. Based on these results, Kuding tea exhibited successful in vitro anticancer effects in TCA8113 cells and in vivo buccal mucosa cancer preventive activity.
Collapse
Affiliation(s)
- Kai Zhu
- Department of Biological and Chemical Engineering, Chongqing University of Education, Chongqing 400067, P.R. China
| | - Guijie Li
- Department of Biological and Chemical Engineering, Chongqing University of Education, Chongqing 400067, P.R. China
| | - Peng Sun
- Department of Biological and Chemical Engineering, Chongqing University of Education, Chongqing 400067, P.R. China
| | - Rui Wang
- Department of Biological and Chemical Engineering, Chongqing University of Education, Chongqing 400067, P.R. China
| | - Yu Qian
- Department of Biological and Chemical Engineering, Chongqing University of Education, Chongqing 400067, P.R. China
| | - Xin Zhao
- Department of Biological and Chemical Engineering, Chongqing University of Education, Chongqing 400067, P.R. China
| |
Collapse
|
9
|
Voura EB, English JL, Yu HYE, Ho AT, Subarsky P, Hill RP, Hojilla CV, Khokha R. Proteolysis during tumor cell extravasation in vitro: metalloproteinase involvement across tumor cell types. PLoS One 2013; 8:e78413. [PMID: 24194929 PMCID: PMC3806793 DOI: 10.1371/journal.pone.0078413] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Accepted: 09/20/2013] [Indexed: 12/22/2022] Open
Abstract
To test if proteolysis is involved in tumor cell extravasation, we developed an in vitro model where tumor cells cross an endothelial monolayer cultured on a basement membrane. Using this model we classified the ability of the cells to transmigrate through the endothelial cell barrier onto the underlying matrix, and scored this invasion according to the stage of passage through the endothelium. Metalloproteinase inhibitors reduced tumor cell extravasation by at least 35%. Visualization of protease and cell adhesion molecules by confocal microscopy demonstrated the cell surface localization of MMP-2, MMP-9, MT1-MMP, furin, CD44 and αvβ3, during the process of transendothelial migration. By the addition of inhibitors and bio-modulators we assessed the functional requirement of the aforementioned molecules for efficient migration. Proteolytic digestion occurred at the cell-matrix interface and was most evident during the migratory stage. All of the inhibitors and biomodulators affected the transition of the tumor cells into the migratory stage, highlighting the most prevalent use of proteolysis at this particular step of tumor cell extravasation. These data suggest that a proteolytic interface operates at the tumor cell surface within the tumor-endothelial cell microenvironment.
Collapse
Affiliation(s)
- Evelyn B. Voura
- Ontario Cancer Institute, Princess Margaret Hospital, Toronto, Ontario, Canada
- Department of Biology, Dominican College, Orangeburg, New York, United States of America
| | - Jane L. English
- Ontario Cancer Institute, Princess Margaret Hospital, Toronto, Ontario, Canada
| | - Hoi-Ying E. Yu
- Ontario Cancer Institute, Princess Margaret Hospital, Toronto, Ontario, Canada
| | - Andrew T. Ho
- Ontario Cancer Institute, Princess Margaret Hospital, Toronto, Ontario, Canada
| | - Patrick Subarsky
- Ontario Cancer Institute, Princess Margaret Hospital, Toronto, Ontario, Canada
| | - Richard P. Hill
- Ontario Cancer Institute, Princess Margaret Hospital, Toronto, Ontario, Canada
| | - Carlo V. Hojilla
- Ontario Cancer Institute, Princess Margaret Hospital, Toronto, Ontario, Canada
| | - Rama Khokha
- Ontario Cancer Institute, Princess Margaret Hospital, Toronto, Ontario, Canada
- * E-mail:
| |
Collapse
|
10
|
Recombinant TIMP-1-GPI inhibits growth of fibrosarcoma and enhances tumor sensitivity to doxorubicin. Target Oncol 2013; 9:251-61. [PMID: 23934106 PMCID: PMC4156787 DOI: 10.1007/s11523-013-0294-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2013] [Accepted: 07/26/2013] [Indexed: 12/26/2022]
Abstract
Fibrosarcomas show a high incidence of recurrence and general resistance to apoptosis. Limiting tumor regrowth and increasing their sensitivity to chemotherapy and apoptosis represent key issues in developing more effective treatments of these tumors. Tissue inhibitor of metalloproteinase 1 (TIMP-1) broadly blocks matrix metalloproteinase (MMP) activity and can moderate tumor growth and metastasis. We previously described generation of a recombinant fusion protein linking TIMP-1 to glycosylphophatidylinositol (GPI) anchor (TIMP-1-GPI) that efficiently directs the inhibitor to cell surfaces. In the present report, we examined the effect of TIMP-1-GPI treatment on fibrosarcoma biology. Exogenously applied TIMP-1-GPI efficiently incorporated into surface membranes of human HT1080 fibrosarcoma cells. It inhibited their proliferation, migration, suppressed cancer cell clone formation, and enhanced apoptosis. Doxorubicin, the standard chemotherapeutic drug for fibrosarcoma, was tested alone or in combination with TIMP-1-GPI. In parallel, the influence of treatment on HT1080 side population cells (exhibiting tumor stem cell-like characteristics) was investigated using Hoechst 33342 staining. The sequential combination of TIMP-1-GPI and doxorubicin showed more than additive effects on apoptosis, while TIMP-1-GPI treatment alone effectively decreased “stem-cell like” side population cells of HT1080. TIMP-1-GPI treatment was validated using HT1080 fibrosarcoma murine xenografts. Growing tumors treated with repeated local injections of TIMP-1-GPI showed dramatically inhibited fibrosarcoma growth and reduced angiogenesis. Intraoperative peritumoral application of GPI-anchored TIMP-1 as an adjuvant to surgery may help maintain tumor control by targeting microscopic residual fibrosarcoma cells and increasing their sensitivity to chemotherapy
Collapse
|
11
|
Zhao X, Pang L, Qian Y, Wang Q, Li Y, Wu M, Ouyang Z, Gao Z, Qiu L. An animal model of buccal mucosa cancer and cervical lymph node metastasis induced by U14 squamous cell carcinoma cells. Exp Ther Med 2013; 5:1083-1088. [PMID: 23599733 PMCID: PMC3628591 DOI: 10.3892/etm.2013.938] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2012] [Accepted: 01/18/2013] [Indexed: 11/06/2022] Open
Abstract
The buccal mucosa is the site with the highest risk of contracting a malignancy in habitual betel quid chewers who expose the buccal mucosa to high doses of carcinogens. Of all oral cancers, those of the buccal mucosa are associated with the poorest prognoses. Therefore, it would be helpful to have an animal model to evaluate new treatment modalities for buccal mucosa cancer. In the present study, we evaluated whether the imprinting control region (ICR) mouse animal model could be employed as a cancer model for buccal mucosa cancer. Sixty male ICR mice were randomly divided into two groups, a normal group (n=10) and a cancer-induced group (n=50). Each mouse in the cancer group was inoculated with 0.05 ml U14 cancer cell suspension (1×107/ml) on the buccal mucosa. Histological staining and gene expression assays revealed that neck lymph node metastasis animal models were established. After 20 days, the cheek tumor formation rate of the ICR mice reached 100%. Furthermore, the neck lymph node metastasis rate was 53%. We identified that U14 cells produce strong metastasis in ICR mice. Metastasis of the tumor to the lymph node began with carcinoma metastasis encroaching on the marginal sinus. Then it infiltrated to the cortex and medulla and the infiltration continued until the normal lymph node structure was completely damaged. This animal model may be employed in medical research on buccal mucosa cancer and cervical lymph node metastasis. In conclusion, our findings indicate that U14 cell-induced mouse buccal mucosa cancer may be a potential cancer model for human buccal mucosa squamous cell carcinoma.
Collapse
Affiliation(s)
- Xin Zhao
- Department of Biological and Chemical Engineering, Chongqing University of Education, Chongqing 400067
| | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Osawa Y, Suetsugu A, Matsushima-Nishiwaki R, Yasuda I, Saibara T, Moriwaki H, Seishima M, Kozawa O. Liver acid sphingomyelinase inhibits growth of metastatic colon cancer. J Clin Invest 2013; 123:834-43. [PMID: 23298833 DOI: 10.1172/jci65188] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2012] [Accepted: 11/13/2012] [Indexed: 12/16/2022] Open
Abstract
Acid sphingomyelinase (ASM) regulates the homeostasis of sphingolipids, including ceramides and sphingosine-1-phosphate (S1P). These sphingolipids regulate carcinogenesis and proliferation, survival, and apoptosis of cancer cells. However, the role of ASM in host defense against liver metastasis remains unclear. In this study, the involvement of ASM in liver metastasis of colon cancer was examined using Asm-/- and Asm+/+ mice that were inoculated with SL4 colon cancer cells to produce metastatic liver tumors. Asm-/- mice demonstrated enhanced tumor growth and reduced macrophage accumulation in the tumor, accompanied by decreased numbers of hepatic myofibroblasts (hMFs), which express tissue inhibitor of metalloproteinase 1 (TIMP1), around the tumor margin. Tumor growth was increased by macrophage depletion or by Timp1 deficiency, but was decreased by hepatocyte-specific ASM overexpression, which was associated with increased S1P production. S1P stimulated macrophage migration and TIMP1 expression in hMFs in vitro. These findings indicate that ASM in the liver inhibits tumor growth through cytotoxic macrophage accumulation and TIMP1 production by hMFs in response to S1P. Targeting ASM may represent a new therapeutic strategy for treating liver metastasis of colon cancer.
Collapse
Affiliation(s)
- Yosuke Osawa
- Department of Pharmacology, Gifu University Graduate School of Medicine, 1-1 Yanagido Gifu 501-1194, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Zhao X, Kim SY, Park KY. Bamboo Salt HasIn VitroAnticancer Activity in HCT-116 Cells and Exerts Anti-Metastatic EffectsIn Vivo. J Med Food 2013; 16:9-19. [DOI: 10.1089/jmf.2012.2316] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Xin Zhao
- Department of Food Science and Nutrition, Pusan National University, Busan, South Korea
- Department of Biological and Chemical Engineering, Chongqing University of Education, Chongqing, China
| | - So-Young Kim
- Functional Food and Nutrition Division, Department of Agrofood Resources, National Academy of Agricultural Sciences (NAAS), Rural Development Administration, Suwon, South Korea
| | - Kun-Young Park
- Department of Food Science and Nutrition, Pusan National University, Busan, South Korea
| |
Collapse
|
14
|
Zhao X, Wang Q, Qian Y, Pang L. Cassia tora L. (Jue-ming-zi) has anticancer activity in TCA8113 cells in vitro and exerts anti-metastatic effects in vivo. Oncol Lett 2012; 5:1036-1042. [PMID: 23426077 PMCID: PMC3576362 DOI: 10.3892/ol.2012.1097] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2012] [Accepted: 12/21/2012] [Indexed: 11/06/2022] Open
Abstract
Cassia tora L. (Jue-ming-zi) is a traditional Chinese medicine widely used in East Asia. The in vitro anticancer effects of Jue-ming-zi were evaluated in TCA8113 human tongue carcinoma cells using a 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyltetrazolium bromide (MTT) assay. At a concentration of 1.0 mg/ml, Cassia tora L. inhibited the growth of TCA8113 cells by 72%; this inhibiton was greater than that by 0.5 and 0.25 mg/ml Cassia tora L. (43 and 16%, respectively). To elucidate the inhibitory mechanisms underlying the anticancer effect of Cassia tora L. in cancer cells, the expression of genes associated with apoptosis, inflammation and metastasis were measured using RT-PCR and western blot analysis. Cassia tora L. significantly induced apoptosis in cancer cells (P<0.05) by upregulating Bax, caspase-3 and caspase-9, and by downregulating Bcl-2. The expression of genes associated with inflammation, including NF-κB, iNOS and COX-2, was significantly downregulated (P<0.05) by Cassia tora L., demonstrating its anti-inflammatory properties. Cassia tora L. also exerted a significant anti-metastatic effect on cancer cells as demonstrated by decreased mRNA expression of matrix metalloprotease (MMP) genes and increased expression of tissue inhibitors of metalloproteinases (TIMPs), and as confirmed by the inhibition of induced tumor metastasis induced in 26-M3.1 colon cells in BALB/c mice. Our results demonstrated that Cassia tora L. exhibited the most potent in vitro anticancer effects, induced apoptosis, had anti-inflammatory activities and exerted in vivo anti-metastatic effects. Additionally, the anticancer, anti-inflammatory and anti-metastatic effects of the higher Cassia tora L. concentrations were stronger compared with those of the lower Cassia tora L. concentrations tested.
Collapse
Affiliation(s)
- Xin Zhao
- Department of Biological and Chemical Engineering, Chongqing University of Education, Chongqing 400067
| | | | | | | |
Collapse
|
15
|
Bandapalli OR, Paul E, Schirmacher P, Brand K. Opposite effects of tissue inhibitor of metalloproteinases-1 (TIMP-1) over-expression and knockdown on colorectal liver metastases. BMC Res Notes 2012; 5:14. [PMID: 22230683 PMCID: PMC3284431 DOI: 10.1186/1756-0500-5-14] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2011] [Accepted: 01/09/2012] [Indexed: 11/10/2022] Open
Abstract
Background Tissue inhibitors of metalloproteinases (TIMPs) and the corresponding metalloproteinases are integral parts of the protease network and have been shown to be involved in cancer development and metastasis. Paradoxically, for TIMP-1, tumor promoting as well as tumor inhibitory effects have been observed. Methods To address this paradox, we utilized the BALB/c/CT26 mouse model that reliably leads to liver metastasis after splenic tumor cell injection and variegated the type of target cells for therapeutic intervention and the modalities of gene transfer. Since we have observed before that over-expression of TIMP-1 in liver host cells leads to efficient tumor growth inhibition in this model, we now examined whether targeting the tumor cells themselves will have a similar effect. Results In concordance with the earlier results, TIMP-1 over-expression in tumor cells led to a dramatic reduction of tumor growth as well. To evaluate any influence of treatment modality, we further examined whether TIMP-1 knockdown in the same animal model would have the opposite effect on tumor growth than TIMP-1 over-expression. Indeed, TIMP-1 knockdown led to a marked increase in tumor burden. Conclusion These data indicate that in the BALB/c/CT26 model, the modification of TIMP-1 has concordant effects irrespective of the type of target cell or the technique of modulation of TIMP-1 activity, and that TIMP-1 is unequivocally tumor inhibitory in this model.
Collapse
Affiliation(s)
- Obul R Bandapalli
- Institute of Pathology, University Hospital Heidelberg, University of Heidelberg, Im Neuenheimer Feld 220/1, 69120 Heidelberg, Germany.
| | | | | | | |
Collapse
|
16
|
Tumor cell-derived Timp-1 is necessary for maintaining metastasis-promoting Met-signaling via inhibition of Adam-10. Clin Exp Metastasis 2011; 28:793-802. [PMID: 21789719 DOI: 10.1007/s10585-011-9410-z] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2011] [Accepted: 07/11/2011] [Indexed: 01/03/2023]
Abstract
In many different tumor entities, increased expression of tissue inhibitor of metalloproteinases-1 (Timp-1) is associated with poor prognosis. We previously reported in mouse models that elevated systemic levels of Timp-1 induce a gene expression signature in the liver microenvironment increasing the susceptibility of this organ to tumor cells. This host effect was dependent on increased activity of the hepatocyte growth factor (Hgf)/hepatocyte growth factor receptor (Met) signaling pathway. In a recent study we showed that Met signaling is regulated by Timp-1 as it inhibits the Met sheddase A disintegrin and metalloproteinase-10 (Adam-10). The aim of the present study was to elucidate whether the metastatic potential of tumor cells benefits from autocrine Timp-1 as well and involves Adam-10 and Met signaling. In a syngeneic murine model of experimental liver metastasis Timp-1 expression and Met signaling were localized within metastatic colonies and expressed by tumor cells. Knock down of tumor cell Timp-1 suppressed Met signaling in metastases and inhibited metastasis formation and tumor cell-scattering in the liver. In vitro, knock down of tumor cell Timp-1 prevented Hgf-induced Met phosphorylation. Consequently, knock down of Met sheddase Adam-10 triggered auto-phosphorylation and responsiveness to Hgf. Accordingly, Adam-10 knock down increased Met phosphorylation in metastatic foci and induced tumor cell scattering into the surrounding liver parenchyma. In conclusion, these findings show that tumor cell-derived Timp-1 acts as a positive regulator of the metastatic potential and support the concept that proteases and their natural inhibitors, as members of the protease web, are major players of signaling during normal homeostasis and disease.
Collapse
|
17
|
von Steinburg SP, Krüger A, Fischer T, Mario Schneider KT, Schmitt M. Placental expression of proteases and their inhibitors in patients with HELLP syndrome. Biol Chem 2009; 390:1199-204. [PMID: 19663680 DOI: 10.1515/bc.2009.123] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
In preeclampsia and hemolysis, elevated liver enzymes and low platelet (HELLP) syndrome, impaired trophoblast invasion and excessive fibrin deposition in the placental intervillous space is associated with fetal compromise. However, little information is available whether modulation of placental protease expression--potentially causing impaired trophoblast invasion--is associated with the HELLP syndrome. Total RNA and protein were extracted from placental tissue from 11 females with HELLP syndrome and 8 controls matched for gestational age. mRNA expression of matrix metalloprotease (MMP) -2 and -9, tissue inhibitors of metalloprotease (TIMP) -1, -2, and -3, and urokinase-type plasminogen activator receptor (uPAR) was determined by Northern blotting. Protein expression of MMP-2 and -9, and TIMP-1 and -2 was detected by Western blotting and that of uPA, uPAR, and plasminogen activator inhibitor (PAI) -1 by ELISA. In patients with HELLP syndrome, mRNA expression of MMP-2 and TIMP-2 was decreased, whereas TIMP-1 and -3 levels were unchanged. MMP-9 and uPAR mRNA was undetectable in both groups. Protein expression of all investigated proteolytic factors remained unchanged. Our findings at the mRNA level suggest a decrease in matrix remodeling in placentae from patients with HELLP syndrome compared with control pregnancies, although this is not supported at the protein level.
Collapse
Affiliation(s)
- Stephanie Pildner von Steinburg
- Abteilung Perinatalmedizin, Frauenklinik und Poliklinik der Technischen Universität München, Klinikum rechts der Isar, D-81675 München, Germany.
| | | | | | | | | |
Collapse
|
18
|
Haviernik P, Diaz MT, Haviernikova E, Tse W, Stetler-Stevenson WG, Bunting KD. Hematopoiesis in mice is extremely resilient to wide variation in TIMP/MMP balance. Blood Cells Mol Dis 2008; 41:179-87. [PMID: 18487063 DOI: 10.1016/j.bcmd.2008.03.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2007] [Revised: 02/28/2008] [Accepted: 03/24/2008] [Indexed: 11/25/2022]
Abstract
Tissue inhibitors of matrix metalloproteinases (TIMPs) are natural inhibitors of matrix metalloproteinases (MMPs) and are associated with normal and pathologic extracellular matrix turnover. Because the microenvironment is critical for normal hematopoietic stem/progenitor cell function, we aimed to determine whether alterations in the TIMP/MMP balance impact upon normal hematopoiesis in mice. We have used both overexpression and knockout mouse models to determine whether early hematopoiesis is susceptible to potentially pathologic changes in TIMP/MMP level. These studies used TIMP-1(-/-) mice and retroviral vectors co-expressing human TIMP-1 or TIMP-2 linked with the green fluorescent protein (GFP) transduced into bone marrow (BM) cells and transplanted into lethally-irradiated recipient mice. Loss of TIMP-1 in knockout mice or retroviral overexpression of TIMP-1 or TIMP-2 did not alter hematopoietic stem/progenitor function during steady-state hematopoiesis. Surprisingly, even when applying hematopoietic stress through mobilization, chemotaxis, or myelosuppression, murine hematopoiesis was not adversely affected by TIMP-1 or TIMP-2 level. We conclude that TIMP/MMP balance alone does not exert significant influence on blood cell development and homeostasis. An important corollary of these studies is that specific modulation using MMP inhibitors for cancer or immunologic therapy is unlikely to have adverse hematopoietic side effects.
Collapse
Affiliation(s)
- Peter Haviernik
- Department of Medicine, Division of Hematology-Oncology, Case Western Reserve University, Cleveland, OH 44106, USA
| | | | | | | | | | | |
Collapse
|
19
|
Kopitz C, Gerg M, Bandapalli OR, Ister D, Pennington CJ, Hauser S, Flechsig C, Krell HW, Antolovic D, Brew K, Nagase H, Stangl M, von Weyhern CWH, Brücher BLDM, Brand K, Coussens LM, Edwards DR, Krüger A. Tissue inhibitor of metalloproteinases-1 promotes liver metastasis by induction of hepatocyte growth factor signaling. Cancer Res 2007; 67:8615-23. [PMID: 17875701 DOI: 10.1158/0008-5472.can-07-0232] [Citation(s) in RCA: 117] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Balanced expression of proteases and their inhibitors is one prerequisite of tissue homeostasis. Metastatic spread of tumor cells through the organism depends on proteolytic activity and is the death determinant for cancer patients. Paradoxically, increased expression of tissue inhibitor of metalloproteinases-1 (TIMP-1), a natural inhibitor of several endometalloproteinases, including matrix metalloproteinases and a disintegrin and metalloproteinase-10 (ADAM-10), in cancer patients is negatively correlated with their survival, although TIMP-1 itself inhibits invasion of some tumor cells. Here, we show that elevated stromal expression of TIMP-1 promotes liver metastasis in two independent tumor models by inducing the hepatocyte growth factor (HGF) signaling pathway and expression of several metastasis-associated genes, including HGF and HGF-activating proteases, in the liver. We also found in an in vitro assay that suppression of ADAM-10 is in principle able to prevent shedding of cMet, which may be one explanation for the increase of cell-associated HGF receptor cMet in livers with elevated TIMP-1. Similar TIMP-1-associated changes in gene expression were detected in livers of patients with metastatic colorectal cancer. The newly identified role of TIMP-1 to create a prometastatic niche may also explain the TIMP-1 paradoxon.
Collapse
Affiliation(s)
- Charlotte Kopitz
- Institut für Experimentelle Onkologie und Therapieforschung, Universität München, Munich, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Tse JC, Kalluri R. Mechanisms of metastasis: epithelial-to-mesenchymal transition and contribution of tumor microenvironment. J Cell Biochem 2007; 101:816-29. [PMID: 17243120 DOI: 10.1002/jcb.21215] [Citation(s) in RCA: 259] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Every year about 500,000 people in the United States die as a result of cancer. Among them, 90% exhibit systemic disease with metastasis. Considering this high rate of incidence and mortality, it is critical to understand the mechanisms behind metastasis and identify new targets for therapy. In recent years, two broad mechanisms for metastasis have received significant attention: epithelial-to-mesenchymal transition (EMT) and tumor microenvironment interactions. EMT is believed to be a major mechanism by which cancer cells become migratory and invasive. Various cancer cells--both in vivo and in vitro--demonstrate features of epithelial-to-mesenchymal-like transition. In addition, many steps of metastasis are influenced by host contributions from the tumor microenvironment, which help determine the course and severity of metastasis. Here we evaluate the diverse mechanisms of EMT and tumor microenvironment interactions in the progression of cancer, and construct a rational argument for targeting these pathways to control metastasis.
Collapse
Affiliation(s)
- Joyce C Tse
- Division of Matrix Biology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | | |
Collapse
|
21
|
Bogaczewicz J, Jasielski P, Mosiewicz A, Trojanowski T, Suchozebrska-Jesionek D, Stryjecka-Zimmer M. [The role of matrix metalloproteinases and tissue inhibitors of metalloproteinases in invasion of tumours of neuroepithelial tissue]. Neurol Neurochir Pol 2007; 45:291-338. [PMID: 17103354 DOI: 10.1080/10408360801973244] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Tumour invasion requires degradation of extracellular matrix components and migration of cells through degraded structures into surrounding tissues. Matrix metalloproteinases (MMP) constitute a family of zinc and calcium-dependent endopeptidases that play a key role in the breakdown of extracellular matrix, and in processing of cytokines, growth factors, chemokines and cell surface receptors. Their activity is regulated at the levels of transcription, activation and inhibition by tissue inhibitors of metalloproteinases (TIMP). Changes in expression of MMP and TIMP are implicated in tumour invasion, because they may contribute to both migration of tumour cells and angiogenesis. Alterations of MMP expression observed in brain tumours arouse interest in the development and evaluation of synthetic matrix metalloproteinase inhibitors as antitumour agents.
Collapse
Affiliation(s)
- Jarosław Bogaczewicz
- Katedra i Klinika Neurochirurgii i Neurochirurgii Dzieciêcej, Akademia Medyczna im. prof. Feliksa Skubiszewskiego, ul. Jaczewskiego 8, 20-954 Lublin.
| | | | | | | | | | | |
Collapse
|
22
|
Cruz-Muñoz W, Kim I, Khokha R. TIMP-3 deficiency in the host, but not in the tumor, enhances tumor growth and angiogenesis. Oncogene 2006; 25:650-5. [PMID: 16186800 DOI: 10.1038/sj.onc.1209104] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Tumor cells, stromal cell compartment and the extracellular matrix (ECM) together generate a multifaceted tumor microenvironment. Matrix metalloproteinases and their tissue inhibitors (TIMPs) provide a means for tumor-stromal interaction during tumorigenesis. Among TIMPs, TIMP-3 is uniquely localized to the ECM and is frequently silenced in human cancers. Here, we asked whether the absence of TIMP-3 in the tumor cell or the host affects the process of tumorigenesis. Timp-3(-/-) ES-cell clones were generated and used to develop teratomas in nude mice. Timp-3(-/-) teratomas showed similar tumor take, growth, and angiogenesis compared to timp-3(+/+) teratomas. To study the effect of TIMP-3 ablation in the host stroma, we measured the growth kinetics of subcutaneous B16F10 melanomas in timp-3(-/-) and wild-type littermates. Tumors grew significantly faster in timp-3(-/-) than in wild-type mice and their CD31 content was significantly higher indicating increased angiogenesis. Augmented angiogenesis in timp-3(-/-) mice was directly tested using Matrigel plug and Gelfoam assays. In response to FGF-2, timp-3(-/-) endothelial cells invaded more efficiently, leading to enhanced formation of functional blood vessels. Thus, TIMP-3 deficiency in the host, but not in the tumor per se, leads to enhanced tumor growth and angiogenesis. TIMP-3 located within the tumor microenvironment inhibits tumorigenesis.
Collapse
Affiliation(s)
- W Cruz-Muñoz
- Department of Laboratory Medicine and Pathobiology, Toronto, Ontario, Canada
| | | | | |
Collapse
|
23
|
Rust R, Visser L, van der Leij J, Harms G, Blokzijl T, Deloulme JC, van der Vlies P, Kamps W, Kok K, Lim M, Poppema S, van den Berg A. High expression of calcium-binding proteins, S100A10, S100A11 and CALM2 in anaplastic large cell lymphoma. Br J Haematol 2005; 131:596-608. [PMID: 16351635 DOI: 10.1111/j.1365-2141.2005.05816.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Anaplastic large cell lymphomas (ALCL) are characterised by the presence of CD30-positive large cells, which usually are of T-cell type. Based on the presence or absence of translocations involving the anaplastic lymphoma kinase (ALK) locus, ALCL cases can be divided into two groups. To gain more insight in the biology of ALCL, we applied serial analysis of gene expression (SAGE) on the Karpas299 cell line and identified 25 up- and 19 downregulated genes. Comparison of the differentially expressed genes with DNA copy number changes in Karpas299 revealed that two overexpressed genes, S100A10 and S100A11, were located in an amplicon suggesting that the increased mRNA levels were caused by DNA amplification. Quantitative reverse transcription polymerase chain reaction on 5 ALCL cell lines and 12 ALCL tissues confirmed the SAGE data for 13 out of 14 up- and one out of four downregulated genes. Immunohistochemical staining confirmed the presence of S100A10, a calcium-binding protein, in three out of five ALK+ and all 7 ALK- ALCL cases. S100A11 staining was confirmed in all ALK+ and six of seven ALK- ALCL cases. Three of the upregulated genes represented calcium-binding proteins, which suggest that altered intracellular signaling might be associated with the oncogenesis of ALCL.
Collapse
Affiliation(s)
- Renata Rust
- Department of Pathology and Laboratory Medicine, University Medical Centre Groningen, Groningen, the Netherlands
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Bianchini G, Aschi M, Cavicchio G, Crucianelli M, Preziuso S, Gallina C, Nastari A, Gavuzzo E, Mazza F. Design, modelling, synthesis and biological evaluation of peptidomimetic phosphinates as inhibitors of matrix metalloproteinases MMP-2 and MMP-8. Bioorg Med Chem 2005; 13:4740-9. [PMID: 15935680 DOI: 10.1016/j.bmc.2005.04.079] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2005] [Revised: 04/21/2005] [Accepted: 04/28/2005] [Indexed: 10/25/2022]
Abstract
Three novel peptidomimetic phosphinate inhibitors have been synthesized and evaluated as inhibitors of matrix metalloproteinases MMP-2 and MMP-8. Their IC50 values are in the micromolar range, and one of them showed to be the most effective inhibitor of MMP-2. The differences in binding affinities for MMP-2 and MMP-8 of the three phosphinates have been rationalized by means of modelling studies and molecular dynamics simulations.
Collapse
Affiliation(s)
- Gianluca Bianchini
- Dipartimento di Chimica, Ingegneria Chimica e Materiali, Università dell'Aquila, via Vetoio, I-67010-Coppito, AQ, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Ruokolainen H, Pääkkö P, Turpeenniemi-Hujanen T. Tissue Inhibitor of Matrix Metalloproteinase-1 Is Prognostic in Head and Neck Squamous Cell Carcinoma: Comparison of the Circulating and Tissue Immunoreactive Protein. Clin Cancer Res 2005; 11:3257-64. [PMID: 15867221 DOI: 10.1158/1078-0432.ccr-04-2277] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Tissue inhibitors of metalloproteinases (TIMP) are capable of inhibiting the matrix metalloproteinases, but they also possess other biological functions. Little is known about the role of TIMP-1 in the progression and spreading of cancer cells among patients with head and neck squamous cell carcinoma (HNSCC). In this study, the pretreatment serum levels of TIMP-1 or the overexpression of TIMP-1 immunoreactive protein in the primary tumor was correlated to the clinical course in patients with HNSCC. EXPERIMENTAL DESIGN The TIMP-1 immunoreactive protein was studied in 74 cases representing HNSCC. The tissue immunoreactive protein was evaluated from paraffin-embedded tumor sections in 68 cases using immunohistologic staining with a specific antibody, and in 68 cases the pretreatment serum levels of TIMP-1 were quantitatively measured by ELISA assay. The results were compared with the clinicopathologic factors of the disease and the patients' outcome. RESULTS A positive correlation was found between the size of the primary tumor (T) and the circulating TIMP-1 level (P = 0.021) or the positive immunoreaction of TIMP-1 in tumor (P = 0.039). The 5-year cause-specific survival was significantly lower in patients presenting with a high serum TIMP-1 level than in those with a low level of TIMP-1 (38% versus 64%, P = 0.034). They also had an unfavorable 5-year relapse-free survival rate (37% versus 56%, respectively). Similarly, the expression of TIMP-1 in tumor was prognostic for shortened survival, the 5-year cumulative relapse-free survival being 42% in patients with a TIMP-1-positive tumor versus 75% in cases with a negative tumor (P = 0.035). Tissue TIMP-1 positivity also seemed associated to the cause-specific survival (P = 0.075) and to be connected with later lymph node or hematogenic relapses. CONCLUSIONS This study shows for the first time that both circulating and tissue TIMP-1 immunoreactive protein predicts the clinical course and dissemination in HNSCC, suggesting that TIMP-1 might be related to both tumor growth and metastasis in HNSCC.
Collapse
Affiliation(s)
- Henni Ruokolainen
- Department of Oncology, University of Oulu and Oulu University Hospital, Oulu, Finland
| | | | | |
Collapse
|
26
|
Sternlicht MD, Bergers G. Matrix metalloproteinases as emerging targets in anticancer therapy: status and prospects. ACTA ACUST UNITED AC 2005. [DOI: 10.1517/14728222.4.5.609] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
27
|
Rust R, Blokzijl T, Harms G, Lim M, Visser L, Kamps WA, Poppema S, van den Berg A. TIMP-1 expression in anaplastic large cell lymphoma is usually restricted to macrophages and only seldom observed in tumour cells. J Pathol 2005; 206:445-50. [PMID: 15920698 DOI: 10.1002/path.1796] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Anaplastic large cell lymphomas (ALCLs) can be subdivided into two subgroups on the basis of their expression of the ALK protein. ALK protein expression leads to activation of signal transducer and activator of transcription (STAT) 3, which is more commonly expressed in ALK-positive than in ALK-negative tumours. Activated STAT3 leads to the induction of several genes such as Mcl-1, Bcl-2 and Bcl-X(L), and tissue inhibitor of metalloproteinase (TIMP)-1. In this study, we analysed TIMP-1 expression in five ALCL cell lines and 11 tumours by quantitative RT-PCR and immunohistochemistry. We identified high-level TIMP-1 expression by RT-PCR in three ALK-positive ALCL-derived cell lines and in all ALK-positive ALCLs, whereas ALK-negative ALCLs generally demonstrated a lower level of TIMP-1 expression. Concordant with these results, we observed TIMP-1 immunostaining in all ALK-positive ALCLs and in only two of six ALK-negative ALCLs. No relationship was observed between the levels of ALK and TIMP-1 expression in the ALK-positive tumours. STAT3 expression levels were similar in all ALCL samples. Double staining with either CD30 or CD68 demonstrated that TIMP-1 expression was restricted to macrophages in the majority of TIMP-1-positive tumours. Expression of the TIMP-1 substrate MMP-2 was more prominent in ALK-negative tumours, while MMP-9 levels were low in all cases. Expression levels of IL-6 and TGF-beta1, which are cytokines known to induce TIMP-1, were higher in ALK-negative ALCLs and moderate in ALK-positive tumours. No clear relationship was observed between IL-10 expression and ALK positivity. Overall, no correlation was seen in ALCLs between the expression of TIMP-1 and that of cytokines that induce TIMP-1. Lack of TIMP-1 expression in the tumour cells of ALK-positive ALCLs argues against a direct role for ALK-induced activation of STAT3 in the regulation of TIMP-1 expression in ALCL.
Collapse
Affiliation(s)
- Renata Rust
- Department of Pathology and Laboratory Medicine, University Medical Center Groningen and University of Groningen, Groningen, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Haviernik P, Lahoda C, Bradley HL, Hawley TS, Ramezani A, Hawley RG, Stetler-Stevenson M, Stetler-Stevenson WG, Bunting KD. Tissue inhibitor of matrix metalloproteinase-1 overexpression in M1 myeloblasts impairs IL-6-induced differentiation. Oncogene 2004; 23:9212-9. [PMID: 15516987 DOI: 10.1038/sj.onc.1208096] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The balance between matrix metalloproteinase (MMP) and tissue inhibitor of matrix metalloproteinase (TIMP) is important for extracellular matrix interactions of hematopoietic cells. MMP-independent growth modulating activity for TIMP-1 on B lymphocytes and erythroid progenitors has also been described, but a role for TIMP-1 in myelomonocytic differentiation has not been previously reported. In this study, we demonstrate that TIMP-1 overexpression impairs differentiation of the myeloblastic M1 cell line following interleukin (IL)-6 stimulation. We generated retroviral vectors coexpressing human TIMP-1 and the green fluorescent protein (GFP) and stably transduced murine M1 myeloid cells. TIMP-1 expressing cells showed a large reduction in IL-6-induced macrophage differentiation in vitro that was reversible with a specific monoclonal antibody. The differentiation delay in M1/TIMP-1 cells was also specifically reversible by pharmacologic phosphatidylinositol-3 kinase (PI3-K) inhibition. Additionally, overexpression of a TIMP-1/GFP fusion protein also impaired M1 differentiation and this protein was localized to the cell surface, consistent with an autocrine receptor-mediated mechanism. Surprisingly, TIMP-1 transduced cells had a selective advantage for growth in IL-6, indicating that functional effects on growth and differentiation of M1 cells were primarily through an autocrine mechanism. Intrinsic TIMP-1 expression in myeloid leukemia cells might thus impact upon survival or differentiation.
Collapse
Affiliation(s)
- Peter Haviernik
- Hematopoiesis Department, American Red Cross, Jerome H Holland Laboratory for the Biomedical Sciences, Rockville, MD 20855, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Elezkurtaj S, Kopitz C, Baker AH, Perez-Cantó A, Arlt MJE, Khokha R, Gansbacher B, Anton M, Brand K, Krüger A. Adenovirus-mediated overexpression of tissue inhibitor of metalloproteinases-1 in the liver: efficient protection against T-cell lymphoma and colon carcinoma metastasis. J Gene Med 2004; 6:1228-37. [PMID: 15390257 DOI: 10.1002/jgm.637] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Matrix metalloproteinases (MMPs) are critical for metastasis of tumor cells. Tissue inhibitor of metalloproteinases-1 (TIMP-1), a natural MMP inhibitor, was shown to reduce metastasis in different models. Here, we investigated whether increased TIMP-1 levels in the liver achieved by adenoviral gene transfer will effectively inhibit liver metastasis of two independent tumor cell lines. METHOD TIMP-1 was transferred with adenoviral vectors into the livers of DBA/2 and Balb/c mice, which were subsequently challenged by hematogenous experimental metastases of the T-cell lymphoma cell line L-CI.5s or the colorectal carcinoma cell line CT-26, respectively. RESULTS MMP-9 expression in the liver was induced upon metastasis in both tumor types. Adenoviral gene transfer led to high transduction efficacy as indicated by lacZ expression in 60% of hepatocytes. TIMP-1, a key inhibitor of MMP-9, was expressed at 10(5)-fold higher levels by adenoviral gene transfer as compared with levels achieved in TIMP-1 transgenic mice, previously shown to be inefficient to reduce T-cell lymphoma metastasis. High local and systemic (serum) levels of TIMP-1 led to substantial (94%) reduction of T-cell lymphoma and colorectal carcinoma (73%) experimental liver metastasis. CONCLUSIONS Adenoviral gene transfer led to systemic and local TIMP-1 levels sufficient to inhibit metastasis of a highly aggressive T-cell lymphoma, pointing at the requirement of threshold levels for effective anti-metastatic efficacy. This approach was also efficient in a colon carcinoma solid tumor model. We propose that viral gene transfer of TIMP-1 can provide a suitable defense strategy to prevent metastatic spread to the liver.
Collapse
MESH Headings
- Adenoviridae/genetics
- Animals
- Cell Line, Tumor
- Colonic Neoplasms/pathology
- Gene Transfer Techniques
- Genetic Therapy
- Genetic Vectors
- Liver/metabolism
- Liver Neoplasms, Experimental/blood supply
- Liver Neoplasms, Experimental/prevention & control
- Liver Neoplasms, Experimental/secondary
- Lymphoma, T-Cell/pathology
- Matrix Metalloproteinase 9/biosynthesis
- Matrix Metalloproteinase 9/genetics
- Mice
- Mice, Inbred BALB C
- Mice, Inbred DBA
- Mice, Knockout
- Neovascularization, Pathologic/enzymology
- Neovascularization, Pathologic/pathology
- Tissue Inhibitor of Metalloproteinase-1/biosynthesis
- Tissue Inhibitor of Metalloproteinase-1/genetics
Collapse
Affiliation(s)
- Sefer Elezkurtaj
- Institute for Biology, Humboldt University Berlin, at the Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Lai R, Rassidakis GZ, Medeiros LJ, Ramdas L, Goy AH, Cutler C, Fujio Y, Kunisada K, Amin HM, Gilles F. Signal transducer and activator of transcription-3 activation contributes to high tissue inhibitor of metalloproteinase-1 expression in anaplastic lymphoma kinase-positive anaplastic large cell lymphoma. THE AMERICAN JOURNAL OF PATHOLOGY 2004; 164:2251-8. [PMID: 15161657 PMCID: PMC1615762 DOI: 10.1016/s0002-9440(10)63781-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The tissue inhibitor of metalloproteinase-1 (TIMP1) is expressed in a subset of malignant lymphomas and can inhibit tumor spread and promote cell survival. Recent data suggest that TIMP1 expression may be regulated by signal transducer and activator of transcription (STAT)-3. Thus, we tested the hypothesis that TIMP1 expression is related to STAT3 activation in lymphomas, with a focus on anaplastic large cell lymphomas (ALCLs), which are known to express high levels of phosphorylated/active STAT3 (pSTAT3). Specific inhibition of STAT3 with a dominant-negative construct led to concentration-dependent down-regulation of TIMP1 expression in two anaplastic lymphoma kinase (ALK)(+) ALCL cell lines, Karpas 299 and SU-DHL-1. Using cDNA microarrays, ALK(+) ALCL cell lines consistently expressed the highest TIMP1 level among 29 lymphoma cell lines of various subtypes. The association between TIMP1 expression and high level of STAT3 activation was validated by Western blots and immunostaining using antibodies specific for pSTAT3 and TIMP1. We further evaluated the relationship between TIMP1 expression and STAT3 activation in 43 ALCL tumors (19 ALK(+) and 24 ALK(-)) using immunohistochemistry and a tissue microarray. The TIMP1(+) group had a mean of 64% pSTAT3(+) cells as compared to 23% pSTAT3(+) cells in the TIMP1(-) group (P = 0.002). As expected, TIMP1 positivity was higher in the ALK(+) group (15 of 19, 79%) compared with the ALK(-) group (5 of 24, 21%; P = 0.0002) because NPM-ALK restricted to ALK(+) tumors was previously shown to activate STAT3. In conclusion, STAT3 directly contributes to the high level of TIMP1 expression in ALK(+) ALCL, and TIMP1 expression correlates with high level of STAT3 activation in ALCL. TIMP1, as a downstream target of STAT3, may mediate the anti-apoptotic effects of STAT3.
Collapse
Affiliation(s)
- Raymond Lai
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Freije JMP, Balbín M, Pendás AM, Sánchez LM, Puente XS, López-Otín C. Matrix metalloproteinases and tumor progression. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2004; 532:91-107. [PMID: 12908552 DOI: 10.1007/978-1-4615-0081-0_9] [Citation(s) in RCA: 114] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The matrix metalloproteinases (MMPs) are a family of more than 20 distinct enzymes that are frequently overexpressed in human tumors. Functional studies have shown that MMPs play an important role in the proteolytic destruction of extracellular matrix and basement membranes, thereby facilitating tumor invasion and metastasis. In addition, these enzymes may also be important in other steps of tumor evolution including neoplastic cell proliferation and angiogenesis stimulation. On the basis of the relevance of MMPs in tumor progression, a number of different strategies aimed to block the unwanted activity of these enzymes in cancer have been developed. Unfortunately, most clinical trials with the first series of MMP inhibitors have failed to show clear benefit in patients with advanced cancer. Explanations for this lack of success include the failure to recognize the role of these enzymes in early stages of the disease as well as inadequacy of either the employed inhibitors or the proteases to be targeted. The introduction of novel concepts such as tumor degradome, and global approaches to protease analysis, may facilitate the identification of the relevant MMPs that must be targeted in each individual cancer patient. On the other hand, the finding that MMPs are enzymes whose effects on biologically active substrates can have profound consequences on cell behaviour, suggests that selective inhibition of a limited set of MMPs at early stages of tumor evolution might be much more effective than using wide-spectrum inhibitors active against most family members, and administered to patients at late stages of the disease. Further studies directed to elucidate these questions will be necessary to clarify whether any of the multiple strategies of MMP inhibition may be part of future therapeutic approaches to control tumor progression.
Collapse
Affiliation(s)
- José M P Freije
- Departamento de Bioquímica, Instituto Universitario de Oncología, Universidad de Oviedo, 33006-Oviedo, Spain
| | | | | | | | | | | |
Collapse
|
32
|
Tarantul VZ. Transgenic Mice as an In Vivo Model of Lymphomagenesis. INTERNATIONAL REVIEW OF CYTOLOGY 2004; 236:123-80. [PMID: 15261738 DOI: 10.1016/s0074-7696(04)36004-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
This review covers multiple data obtained on genetically modified mice that help to elucidate various intricate molecular mechanisms of lymphomagenesis in humans. We are in a "golden age" of mouse genetics. The mouse is by far the most accessible mammalian system physiologically similar to humans. Transgenic mouse models have illuminated how different genes contribute to human lymphomagenesis. Multiple experiments with transgenic mice have not only confirmed the data obtained for human lymphomas but also gave additional evidence for the role of some genes and cooperative participation of their products in the development of human lymphomas. Genes and gene networks detected on transgenic mice can successfully serve as molecular targets for tumor therapy. This review demonstrates the extraordinary possibilities of transgenic technology, which is presently one of the readily available, efficient, and accurate tools to solve the problem of cancer.
Collapse
Affiliation(s)
- V Z Tarantul
- Institute of Molecular Genetics, Russian Academy of Sciences, Moscow 123182, Russia
| |
Collapse
|
33
|
Kuittinen O, Apaja-Sarkkinen M, Turpeenniemi-Hujanen T. Gelatinases (MMP-2 and MMP-9), TIMP-1 expression and the extent of neovascularization in aggressive non-Hodgkin's lymphomas. Eur J Haematol 2003; 71:91-9. [PMID: 12890147 DOI: 10.1034/j.1600-0609.2003.00101.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
OBJECTIVES The present study was carried out to clarify the role of matrix metalloproteinase-2 and -9 (MMP-2 and MMP-9), tissue inhibitor of metalloproteinase-1 (TIMP-1) and the extent of neovascularization in the clinicopathologic behavior of non-Hodgkin's lymphomas. METHODS Paraffin-embedded histologic sections from 57 patients with aggressive non-Hodgkin's lymphomas were stained with MMP-2, MMP-9, TIMP-1, and factor VIII antibodies to correlate the expression of these markers to the clinical disease characteristics. RESULTS Strong MMP-9 staining was found to be an adverse prognostic factor among patients with aggressive B-cell lymphomas, the probabilities for 5-yr disease-free survival being 73%, 63%, 50%, and 0% in patients with grades 0, 1, 2, and 3 staining, respectively. Among the patients with strong (grades 2 and 3) MMP-9 staining, however, positivity for TIMP-1 indicated a trend toward a more favorable prognosis. TIMP-1 expression also correlated with the immunoblastic and anaplastic lymphoma subtypes. The expression of the proteins for MMP-2 and factor VIII had no independent prognostic role. None of the study parameters correlated with disease stage, the occurrence of extranodal infiltrates, the occurrence of bulky tumor, or the IPI scores. CONCLUSIONS Positivity for MMP-9 immunoreactive protein is an independent sign of an unfavorable prognosis in non-Hodgkin's lymphomas. This is not mediated through influences in tumor dissemination or neovascularization indicating it to carry other important biological functions.
Collapse
Affiliation(s)
- Outi Kuittinen
- Department of Oncology and Radiotherapy, Oulu University Hospital, Oulu, Finland
| | | | | |
Collapse
|
34
|
Ikenaka Y, Yoshiji H, Kuriyama S, Yoshii J, Noguchi R, Tsujinoue H, Yanase K, Namisaki T, Imazu H, Masaki T, Fukui H. Tissue inhibitor of metalloproteinases-1 (TIMP-1) inhibits tumor growth and angiogenesis in the TIMP-1 transgenic mouse model. Int J Cancer 2003; 105:340-346. [PMID: 12704667 DOI: 10.1002/ijc.11094] [Citation(s) in RCA: 85] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The tissue inhibitor of matrix metalloproteinases-1 (TIMP-1) has been recognized as a multifunctional protein. The role of TIMPs in cancer remains the subject of conflicting reports with an antitumor activity or a tumor growth stimulation activity by several mechanisms. The aim of our study is to investigate the effect of ectopic TIMP-1 overexpression on the primary transplanted tumor growth. We employed transgenic mice overexpressing the human TIMP-1 (hTIMP-1) in the liver under control of the albumin promoter/enhancer (TIMP-Tg-mice) and producing high serum levels of TIMP-1. We used the transplantable Ehrlich tumor cells in the current study. The allograft study revealed that the tumor growth in the TIMP-Tg-mice was more significantly inhibited than control (Cont) mice by associated suppression of neovascularization in the tumor. The in vitro studies showed that the recombinant TIMP-1 (rTIMP-1) did not affect the proliferation of the endothelial cells (ECs) and tumor cells, suggesting that the tumor suppressive effect of TIMP-1 was not due to cytotoxicity. TIMP-1 significantly inhibited EC tubular formation in vitro. Furthermore, TIMP-1 treatment did not affect the levels of matrix metalloproteinase (MMP)-2 and MMP-9 mRNA in the Ehrlich tumor cells in vitro, although these expressions in the tumor were markedly suppressed in the TIMP-Tg-mice, compared to the Cont-mice at the end of the experiment. These results suggested that the ectopically overexpressed TIMP-1 inhibited the tumor growth by angiogenesis suppression.
Collapse
Affiliation(s)
- Yasuhide Ikenaka
- Third Department of Internal Medicine, Nara Medical University, Nara, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Banke IJ, Arlt MJE, Pennington C, Kopitz C, Steinmetzer T, Schweinitz A, Gansbacher B, Quigley JP, Edwards DR, Stürzebecher J, Krüger A. Increase of Anti-Metastatic Efficacy by Selectivity- But Not Affinity-Optimization of Synthetic Serine Protease Inhibitors. Biol Chem 2003; 384:1515-25. [PMID: 14669995 DOI: 10.1515/bc.2003.168] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Although tumors frequently show elevated protease activities, the concept of anti-proteolytic cancer therapy has lost momentum after failure of clinical trials with broad-spectrum matrix metalloproteinase inhibitors. Thus we need to adapt our design strategies for protease inhibitors. Here, we employed a series of seven structurally fine-modulated and pharmacokinetically closely related synthetic 4-amidinobenzylamine-based inhibitors with distinct selectivity for prototypical serine proteases in a murine T cell lymphoma liver metastasis model. This in vivo screening revealed efficacy of urokinase inhibitors but no correlation between urokinase selectivity or affinity and anti-metastatic effect. In contrast, factor Xa-selective inhibitors were more potent, demonstrating factor Xa or a factor Xa-like serine protease likely to be more determinant in this model. Factor Xa selectivity, but not affinity, significantly improved anti-metastatic efficacy. For example, factor Xa inhibitors CJ-504 and CJ-510 exert similar affinity for factor Xa (K(i)=14 nM versus 8.8 nM) but CJ-504 was 70-fold more selective for factor Xa. This correlated with higher anti-metastatic efficacy (58.8% with CJ-504; 28.2% with CJ-510). Our results show that among the protease inhibitors employed that have affinities in the nanomolar range, the strategy of selectivity-optimization is superior to further improvement of affinity to significantly enhance anti-metastatic efficacy. This appreciation may be important for the future rational design of new anti-proteolytic agents for cancer therapy.
Collapse
Affiliation(s)
- Ingo J Banke
- Institut für Experimentelle Onkologie und Therapieforschung, Technische Universität München, Ismaninger Str. 22, D-81675 München, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Abstract
For more than two decades, the view that tumour-associated matrix metalloproteinases (MMPs) were required for peritumour tissue degradation and metastasis dominated the drive to develop MMP inhibitors as anticancer therapeutics. Until recently, clinical trials with MMP inhibitors have yielded disappointing results, highlighting the need for better insight into the mechanisms by which this growing family of multifunctional enzymes contribute to tumour growth. It is now recognized that MMP activity is tightly regulated at several levels, providing new avenues for blocking these enzymes. What are the different approaches that can be used to target MMPs, and which of these might lead to new therapeutic strategies for cancer?
Collapse
Affiliation(s)
- Christopher Mark Overall
- Department of Oral Biological and Medical Sciences, C.I.H.R. Group in Matrix Dynamics, University of British Columbia, V6T 1Z3, Canada
| | | |
Collapse
|
37
|
Thorns C, Gaiser T, Lange K, Merz H, Feller AC. cDNA arrays: gene expression profiles of Hodgkin's disease and anaplastic large cell lymphoma cell lines. Pathol Int 2002; 52:578-85. [PMID: 12406187 DOI: 10.1046/j.1320-5463.2002.01400.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
cDNA arrays are a powerful tool for the identification of differentially expressed genes in malignant tumors. We used this technique to study the gene expression profiles of anaplastic large cell lymphoma (ALCL) and Hodgkin's disease (HD). Gene expression of 11 lymphoma cell lines was analyzed covering 1176 cDNA sequences. Comparing these data to the expression profiles of B- and T-lymphocytes, we identified 27 genes that were deregulated in all cell lines or in a particular entity. For the establishment of gene expression profiles the 27 genes were assigned to four groups composed of genes deregulated in (i) all lymphoma cell lines, (ii) ALCL and HD, (iii) only HD, and (iv) ALCL exclusively. Our results indicate that ALCL and HD share the differential expression of at least five genes. In addition, both entities are characterized by the differentially deregulated expression of four genes in HD and seven genes in ALCL. Because the expression profiling was performed on cell lines, further studies are needed to clarify the biological significance of the differentially expressed genes.
Collapse
Affiliation(s)
- Christoph Thorns
- Department of Pathology, German Consultation and Reference Center for Lymphomas, Medical University of Luebeck, Luebeck, Germany.
| | | | | | | | | |
Collapse
|
38
|
Lin H, Chen X, Wang J, Yu Z. Inhibition of apoptosis in rat mesangial cells by tissue inhibitor of metalloproteinase-1. Kidney Int 2002; 62:60-9. [PMID: 12081564 DOI: 10.1046/j.1523-1755.2002.00403.x] [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: 11/20/2022]
Abstract
BACKGROUND Tissue inhibitor of metalloproteinase-1 (TIMP-1) is an important inhibitor of extracellular matrix degradation. Recently, it was reported that TIMP-1 also could inhibit apoptosis in B type lymphocyte. This study was designed to examine the effects of TIMP-1 on mesangial cell apoptosis. METHODS The full-length cDNA of TIMP-1 was cloned and used to construct two recombinant vectors, TIMP-1S and TIMP-1AS, encoding sense TIMP-1 and antisense TIMP-1, respectively. The vectors were transfected into rat mesangial cells (RMC) and their expressions detected by Northern and Western blotting. Apoptosis was induced by serum deprivation, and was monitored for DNA fragmentation by TUNEL assay and DNA laddering. In addition, the expression of endogenous TIMP-1, matrix metalloprotein-2 (MMP-2), and MMP-9, as well as apoptosis-related genes Bcl-2 and Bax were investigated. RESULTS TIMP-1AS transfection induced a suppression of TIMP-1 expression accompanied by an earlier onset of apoptosis, and TIMP-1S transfection induced TIMP-1 over-expression accompanied by a much later onset of apoptosis. A neutralizing antibody of TIMP-1 restored the sensitivity of TIMP-1S-transfected RMC to serum deprivation, but a synthetic matrix metalloproteinase inhibitor BB-94 did not influence the sensitivity of TIMP-1S-transfected RMC to serum deprivation. Finally, TIMP-1 over-expression inhibited the expression of Bax but with no effect on the expression of Bcl-2. CONCLUSION TIMP-1 inhibits the serum deprivation-induced apoptosis in RMC, in which Bax might be involved.
Collapse
Affiliation(s)
- Hongli Lin
- Department of Nephrology, Kidney Center & Key Lab of Chinese PLA, General Hospital of Chinese PLA, 28 Fu-xing Road, Beijing 100853, People's Republic of China
| | | | | | | |
Collapse
|
39
|
Trocmé C, Gaudin P, Berthier S, Morel F. Regulation of TIMP-1 phenotypic expression in Epstein--Barr virus-immortalized B lymphocytes. BIOCHIMICA ET BIOPHYSICA ACTA 2002; 1590:167-76. [PMID: 12063180 DOI: 10.1016/s0167-4889(02)00210-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Normal B lymphocytes as well as malignant B cells extravasate from blood circulation during physiological and pathological processes and require matrix metalloproteinases (MMPs) to facilitate trafficking through the subendothelial basal lamina and the extracellular matrix. We have previously shown that Epstein-Barr virus (EBV)-immortalized B lymphocytes constitutively synthesized low levels of MMP-9 and huge amounts of its preferential inhibitor, tissue inhibitor of matrix metalloproteinase-1 (TIMP-1). In the present study, TIMP-1 phenotypic expression was extensively investigated in response to various mediators including interleukins, chemokines, growth factors and tumor promotor, and was compared to MMP-9 synthesis. Results showed a roughly constitutive TIMP-1 expression opposed to an inducible MMP-9 synthesis. Nevertheless, further analysis of TIMP-1 synthesis showed the existence of regulation mechanisms: modulation of intracellular Ca(2+) concentration as well as cation ionophore monensin were demonstrated to influence TIMP-1 production and secretion. The precise pathways implicated in these regulation mechanisms are currently under survey.
Collapse
Affiliation(s)
- Candice Trocmé
- GREPI, Laboratoire d'Enzymologie, CHU Albert Michallon, BP 217, 38043 Grenoble Cedex 9, France.
| | | | | | | |
Collapse
|
40
|
Abstract
Matrix metalloproteinases (MMPs) have long been associated with cancer-cell invasion and metastasis. This provided the rationale for clinical trials of MMP inhibitors, unfortunately with disappointing results. We now know, however, that the MMPs have functions other than promotion of invasion, have substrates other than components of the extracellular matrix, and that they function before invasion in the development of cancer. With this knowledge in hand, can we rethink the use of MMP inhibitors in the clinic?
Collapse
Affiliation(s)
- Mikala Egeblad
- Department of Anatomy, University of California at San Francisco, 94143-0452, USA.
| | | |
Collapse
|
41
|
Cianfrocca M, Cooley TP, Lee JY, Rudek MA, Scadden DT, Ratner L, Pluda JM, Figg WD, Krown SE, Dezube BJ. Matrix metalloproteinase inhibitor COL-3 in the treatment of AIDS-related Kaposi's sarcoma: a phase I AIDS malignancy consortium study. J Clin Oncol 2002; 20:153-9. [PMID: 11773164 DOI: 10.1200/jco.2002.20.1.153] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE Matrix metalloproteinases (MMPs) are involved in tumor invasion and metastasis and are overexpressed in Kaposi's sarcoma (KS) cells. The primary aim was to define the safety and toxicity of the MMP inhibitor COL-3 in patients with AIDS-related KS. Secondary aims were to evaluate tumor response, pharmacokinetics, and changes in blood levels of MMP-2, MMP-9, vascular endothelial growth factor (VEGF), and basic fibroblast growth factor (bFGF). PATIENTS AND METHODS COL-3 was administered orally once daily, and doses were escalated in cohorts of three to six subjects. Patients with symptomatic visceral KS or severe tumor-associated edema were excluded. Antiretroviral therapy was permitted but not required. Study end points were grade 3 or 4 toxicity or progressive KS. Serial blood specimens were obtained for pharmacokinetics and levels of MMP-2, MMP-9, VEGF, and bFGF. RESULTS Eighteen patients received COL-3 in dosing cohorts of 25, 50, and 70 mg/m(2)/d. Prior KS therapy was reported by 17 patients (94%). COL-3-related grade 3 or 4 adverse events were reported by six patients and included photosensitivity, rash, and headache. There was one complete response and seven partial responses, for an overall response rate of 44%, with a median response duration of 25+ weeks. The median COL-3 half-life was 39.3 hours (range, 4.1 to 251.1 hours). There was a significant difference between responders and nonresponders with respect to the change in MMP-2 serum levels from baseline to minimum value on treatment (P =.037). CONCLUSION COL-3 administered orally once daily to patients with AIDS-related KS is reasonably well tolerated. The most common adverse event was dose-related photosensitivity. Antitumor activity was noted. Further evaluation of COL-3 for the treatment of KS is warranted.
Collapse
Affiliation(s)
- Mary Cianfrocca
- Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Boston, MA 02215, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Oelmann E, Herbst H, Zühlsdorf M, Albrecht O, Nolte A, Schmitmann C, Manzke O, Diehl V, Stein H, Berdel WE. Tissue inhibitor of metalloproteinases 1 is an autocrine and paracrine survival factor, with additional immune-regulatory functions, expressed by Hodgkin/Reed-Sternberg cells. Blood 2002; 99:258-67. [PMID: 11756180 DOI: 10.1182/blood.v99.1.258] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Tissue inhibitor of metalloproteinases (TIMP)-1 and TIMP-2 are proteins with proteinase-inhibiting and cytokine properties. TIMP-1 is active primarily in B cells and B-cell lymphomas, whereas TIMP-2 expression is restricted to T cells. The expression of TIMP-1 and TIMP-2 in lymph nodes from patients with Hodgkin disease (HD) and in Hodgkin-derived cell lines was investigated. In situ hybridization showed TIMP-1 RNA expression in 3% to 80% of Hodgkin/Reed-Sternberg (H/R-S) cells from 14 of 15 patients, with results in one patient being at the lowest detection limit; no expression of TIMP-2 in H/R-S cells; and only weak expression of TIMP-2 in reactive lymphoid tissue. Production of TIMP-1 protein by H/R-S cells was accordingly found on immunohistochemical analysis of lymph nodes from patients with HD. There was only low expression of matrix metalloproteinase (MMP)-2, which is mainly inhibited by TIMP-2; no expression of MMP-1 and MMP-3 in reactive lymphoid tissue; and no expression of these MMPs in H/R-S cells. Thus, TIMP-1 expression in lymph nodes was not correlated with metalloproteinase expression. Five of 7 Hodgkin-derived cell lines expressed TIMP-1 at the protein level. Only one of these cell lines expressed TIMP-2, at the lowest detection limit. TIMP-1 levels in plasma from patients with HD were within the same range as those in plasma from healthy controls. Recombinant human TIMP-1 inhibited induced cell death in Hodgkin-derived cell lines in vitro. TIMP-1 and TIMP-2 inhibited T-cell cytotoxicity against autologous cells presenting tumor-associated antigens and in allogeneic mixed lymphocyte cultures. Thus, TIMP-1, aside from its role in proteinase equilibrium, is an autocrine and paracrine survival factor for H/R-S cells and an immunosuppressive protein expressed in Hodgkin lymphomas.
Collapse
Affiliation(s)
- Elisabeth Oelmann
- Department of Medicine (Hematology/Oncology), Westfaelische Wilhelms Universitaet Muenster, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Ho AT, Voura EB, Soloway PD, Watson KL, Khokha R. MMP inhibitors augment fibroblast adhesion through stabilization of focal adhesion contacts and up-regulation of cadherin function. J Biol Chem 2001; 276:40215-24. [PMID: 11500488 DOI: 10.1074/jbc.m101647200] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Increased pericellular proteolysis due to an imbalance between MMPs (matrix metalloproteinases) and TIMPs (tissue inhibitors of metalloproteinases) promotes early stages of tumorigenesis. We have reported that TIMP-1 down-regulation confers tumorigenicity on immortal Swiss 3T3 fibroblasts. In pursuit of the mechanism involved in this transformation, we asked whether MMP inhibitors modulate contact inhibition and cell adhesion, because the dysregulation of these events is essential for cellular transformation. Using both genetic and biochemical means, we demonstrate that MMP inhibitors regulate fibroblast cell adhesion. TIMP-1 down-regulated cells formed dense, multilayered colonies, suggesting a loss of contact inhibition. Recombinant TIMP-1 and synthetic MMP inhibitors (MMPi) restored normal cell contact and density of these cells in a dose-dependent manner. Consequently, the effect of MMPi on both cell-extracellular matrix (ECM) and cell-cell adhesion were investigated. Upon MMPi treatment, p125(FAK) was redistributed, together with vinculin, to points of cell-ECM contact. Furthermore, phosphorylation of p125(FAK) was restored to levels similar to that of wild type. In parallel, MMPi treatment increased cadherin levels and stabilized cadherin-mediated cell-cell contacts. Moreover, enhanced cadherin function was evident as increased calcium-dependent cell-cell aggregation and co-localization of cadherin and beta-catenin at the cell membrane. We also obtained independent evidence of altered cadherin function using timp-1(-/-) mouse embryonic fibroblasts. Our data provide provocative evidence that increased pericellular proteolysis impacts cell adhesion systems to offset normal contact inhibition, with subsequent effects on cell transformation and tumorigenesis.
Collapse
Affiliation(s)
- A T Ho
- Department of Medical Biophysics, Ontario Cancer Institute, University Health Network, University of Toronto, 610 University Ave., Toronto, Ontario M5G 2M9, Canada
| | | | | | | | | |
Collapse
|
44
|
Leco KJ, Waterhouse P, Sanchez OH, Gowing KL, Poole AR, Wakeham A, Mak TW, Khokha R. Spontaneous air space enlargement in the lungs of mice lacking tissue inhibitor of metalloproteinases-3 (TIMP-3). J Clin Invest 2001. [DOI: 10.1172/jci200112067] [Citation(s) in RCA: 187] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
45
|
Guedez L, McMarlin AJ, Kingma DW, Bennett TA, Stetler-Stevenson M, Stetler-Stevenson WG. Tissue inhibitor of metalloproteinase-1 alters the tumorigenicity of Burkitt's lymphoma via divergent effects on tumor growth and angiogenesis. THE AMERICAN JOURNAL OF PATHOLOGY 2001; 158:1207-15. [PMID: 11290537 PMCID: PMC1891911 DOI: 10.1016/s0002-9440(10)64070-9] [Citation(s) in RCA: 89] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Epstein-Barr virus (EBV)-positive Burkitt's lymphoma cells and EBV-infected B cells elicit humoral factors that inhibit tumor-induced angiogenesis, resulting in tumor necrosis and regression. Of the chemokine factors identified in association with this growth behavior, none have induced complete tumor regression. We have previously identified tissue inhibitors of metalloproteinase (TIMP)-1 in various B cell lymphoma cell lines. Here we show that induction of TIMP-1 expression in an EBV-negative Burkitt's lymphoma cell line results in a biphasic, in vivo tumor growth pattern in the nude mouse that is essentially identical to EBV-positive Burkitt's lymphoma cell lines. The initial effect of TIMP-1 is to enhance tumor growth, consistent with the reported anti-apoptotic effect of TIMP-1 on B cell growth. Tumor necrosis and regression then follow the initial period of rapid, increased tumor growth. Only microscopic foci of residual, proliferating tumor cells are observed on biopsy of the tumor site. This latter effect is mediated by TIMP-1 inhibition of an angiogenic response within the developing tumor mass, as demonstrated by immunostaining and microvessel counts. These findings suggest that TIMP-1 is an important mediator of the in vivo growth properties of EBV-positive Burkitt's lymphoma.
Collapse
Affiliation(s)
- L Guedez
- National Institutes of Health, National Cancer Institute, Extracellular Matrix Section, Laboratory of Pathology, Division of Clinical Sciences, Bethesda, MD 20892, USA
| | | | | | | | | | | |
Collapse
|
46
|
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]
|
47
|
Abstract
The matrix metalloproteinases (MMPs) constitute a multigene family of over 25 secreted and cell surface enzymes that process or degrade numerous pericellular substrates. Their targets include other proteinases, proteinase inhibitors, clotting factors, chemotactic molecules, latent growth factors, growth factor-binding proteins, cell surface receptors, cell-cell adhesion molecules, and virtually all structural extracellular matrix proteins. Thus MMPs are able to regulate many biologic processes and are closely regulated themselves. We review recent advances that help to explain how MMPs work, how they are controlled, and how they influence biologic behavior. These advances shed light on how the structure and function of the MMPs are related and on how their transcription, secretion, activation, inhibition, localization, and clearance are controlled. MMPs participate in numerous normal and abnormal processes, and there are new insights into the key substrates and mechanisms responsible for regulating some of these processes in vivo. Our knowledge in the field of MMP biology is rapidly expanding, yet we still do not fully understand how these enzymes regulate most processes of development, homeostasis, and disease.
Collapse
Affiliation(s)
- Mark D. Sternlicht
- Department of Anatomy, University of California, San Francisco, California 94143-0452
| | - Zena Werb
- Department of Anatomy, University of California, San Francisco, California 94143-0452
| |
Collapse
|
48
|
Kossakowska AE, Urbanski SJ, Janowska-Wieczorek A. Matrix metalloproteinases and their tissue inhibitors - expression, role and regulation in human malignant non-Hodgkin's lymphomas. Leuk Lymphoma 2000; 39:485-93. [PMID: 11342332 DOI: 10.3109/10428190009113379] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Human malignant non-Hodgkin's lymphomas (NHL) represent a heterogeneous group of neoplasms, which vary in their clinical behavior and pathophysiology. Matrix metalloproteinases (MMPs) and their tissue inhibitors (TIMPs) have been shown to play a role in the pathophysiology and clinical aggressiveness of human NHL. In this setting, MMP-9 and TIMP-1 appear to be the most important members of the MMP and TIMP families, and overexpression of both correlates with a poor clinical outcome of patients with NHL. MMP-9 and TIMP-1, however, act through different mechanisms and are produced by different cell types. Expression of both is upregulated by interleukin-6 (IL-6), a cytokine that is known as one of the factors involved in the pathophysiology of human NHL. In this review we summarize the complex regulation of MMP and TIMP expression in human NHL and propose a mechanism by which MMP-9, TIMP-1 and IL-6 may influence the biology of these tumors.
Collapse
Affiliation(s)
- A E Kossakowska
- Department of Pathology, University of Calgary and Calgary Laboratory Services, Calgary, Alberta, Canada
| | | | | |
Collapse
|
49
|
Sternlicht MD, Bissell MJ, Werb Z. The matrix metalloproteinase stromelysin-1 acts as a natural mammary tumor promoter. Oncogene 2000; 19:1102-13. [PMID: 10713697 PMCID: PMC2933206 DOI: 10.1038/sj.onc.1203347] [Citation(s) in RCA: 196] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Extracellular matrix-degrading matrix metalloproteinases (MMPs) are invariably upregulated in epithelial cancers and are key agonists in angiogenesis, invasion and metastasis. Yet most MMPs are secreted not by the cancer cells themselves, but by stromal cells within and around the tumor mass. Because the stromal environment can influence tumor formation, and because MMPs can alter this environment, MMPs may also contribute to the initial stages of cancer development. Several recent studies in MMP-overexpressing and MMP-deficient mice support this possibility, but have required carcinogens or pre-existing oncogenic mutations to initiate tumorigenesis. Here we review the spontaneous development of premalignant and malignant lesions in the mammary glands of transgenic mice that express an autoactivating form of MMP-3/stromelysin-1 under the control of the whey acidic protein gene promoter. These changes were absent in nontransgenic littermates and were quenched by co-expression of a human tissue inhibitor of metalloproteinases-1 (TIMP-1) transgene. Thus by altering the cellular microenvironment, stromelysin-1 can act as a natural tumor promoter and enhance cancer susceptibility.
Collapse
Affiliation(s)
- M D Sternlicht
- Department of Anatomy, University of California, 513 Parnassus Avenue, HSW-1301, San Francisco, California, CA 94143-0452, USA
| | | | | |
Collapse
|
50
|
Abstract
Our recent finding that resistance to lymphoma cell metastasis in intercellular adhesion molecule-1-(ICAM-1)–deficient mice was manifested after homing suggested that the mechanism could involve the capacity of ICAM-1 to induce, via leukocyte function-associated antigen-1 (LFA-1) signaling, the expression of new genes necessary for migration and survival of lymphoma cells after homing. This hypothesis would imply that lymphoma cells, on repeated metastatic cycles, would acquire such a highly aggressive phenotype that they no longer require contact with ICAM-1 at later stages of metastasis. We addressed this question by generating highly aggressive lymphoma variants to determine if increased tumorigenicity would allow lymphoma cells to grow into tumors in ICAM-1–deficient mice. We found that on repeated in vivo passages, a selective pressure favored the lymphoma cells that constitutively express high levels of matrix metalloproteainse-9 (MMP-9), a gene associated with a poor clinical outcome in non-Hodgkins's lymphoma. We further found that although the parent lymphoma cells could not grow tumors in ICAM-1–deficient mice, the aggressive lymphoma variants could. This indicates that, at late stages of the disease, tumor cells with a high metastatic efficiency, encoded by the repertoire of selected genes, no longer require some of the signals normally delivered by cell adhesion molecules. In light of these findings, the possibility of inhibiting dissemination of lymphoma cells at the late stage of the disease by acting against cell adhesion molecules must be reconsidered. (Blood. 2000;95:314-319)
Collapse
|