1
|
Wang Q, Serda M, Li Q, Sun T. Recent Advancements on Self-Immolative System Based on Dynamic Covalent Bonds for Delivering Heterogeneous Payloads. Adv Healthc Mater 2023; 12:e2300138. [PMID: 36943096 DOI: 10.1002/adhm.202300138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 03/10/2023] [Indexed: 03/23/2023]
Abstract
The precisely spatial-temporal delivery of heterogeneous payloads from a single system with the same pulse is in great demand in realizing versatile and synergistic functions. Very few molecular architectures can satisfy the strict requirements of dual-release translated from single triggers, while the self-immolative systems based on dynamic covalent bonds represent the "state-of-art" of ultimate solution strategy. Embedding heterogeneous payloads symmetrically onto the self-immolative backbone with dynamic covalent bonds as the trigger, can respond to the quasi-bio-orthogonal hallmarks which are higher at the disease's microenvironment to simultaneously yield the heterogeneous payloads (drug A/drug B or drug/reporter). In this review, the modular design principles are concentrated to illustrate the rules in tailoring useful structures, then the rational applications are enumerated on the aspects of drug codelivery and visualized drug-delivery. This review, hopefully, can give the general readers a comprehensive understanding of the self-immolative systems based on dynamic covalent bonds for delivering heterogeneous payloads.
Collapse
Affiliation(s)
- Qingbing Wang
- Department of Interventional Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin Er Road, Shanghai, 200025, P. R. China
- Key Laboratory of Smart Drug Delivery Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai, 201203, P. R. China
| | - Maciej Serda
- Institute of Chemistry, University of Silesia in Katowice, Katowice, 40-006, Poland
| | - Quan Li
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, 10 Boyanghu Road, Tianjin, 301617, P. R. China
- College of Chemistry and Chemical Engineering, Hubei University, 368 Youyidadao Avenue, Wuhan, 430062, P. R. China
| | - Tao Sun
- Key Laboratory of Smart Drug Delivery Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai, 201203, P. R. China
| |
Collapse
|
2
|
Kim KM, Jung J. Upregulation of G Protein-Coupled Estrogen Receptor by Chrysin-Nanoparticles Inhibits Tumor Proliferation and Metastasis in Triple Negative Breast Cancer Xenograft Model. Front Endocrinol (Lausanne) 2020; 11:560605. [PMID: 33042020 PMCID: PMC7522162 DOI: 10.3389/fendo.2020.560605] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Accepted: 08/17/2020] [Indexed: 12/18/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is associated with a high mortality rate among women globally. TNBC shows a high rate of recurrence and distant metastasis. Particularly, the chemotherapy is limited because hormone therapy of breast cancer is ineffective. Thus, an effective chemotherapeutic agent is needed for tumor suppression. Chrysin-nanoparticles (chrysin-NPs) were investigated for their inhibitory effect on a MDA-MB-231-derived xenograft model. To gain insight into the underlying mechanisms, we conducted human matrix metalloproteinase (MMP) array, western blot, and immunohistochemistry analysis. Furthermore, in vivo imaging was used to monitor the chemotherapeutic efficacy of chrysin-NPs in a metastasis mouse model. Chrysin-NPs significantly inhibited the proliferation of MDA-MB-231 cells via the PI3K/JNK pathway and induced cell death through the p53-apoptosis pathway, leading to delayed MDA-MB-231-derived tumor growth. Interestingly, chrysin-NPs significantly induced G protein-coupled estrogen receptor (GPER) expression, which suppresses MMPs and NF-κB expression. Chrysin-NPs acted as effective metastasis inhibitors. Our results suggest that chrysin-NPs may be used as an effective adjuvant formulation to inhibit TNBC progression.
Collapse
|
3
|
Influence of Immune Myeloid Cells on the Extracellular Matrix During Cancer Metastasis. CANCER MICROENVIRONMENT 2016; 9:45-61. [PMID: 26956475 PMCID: PMC4842183 DOI: 10.1007/s12307-016-0181-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 02/12/2016] [Indexed: 01/04/2023]
Abstract
The extracellular matrix (ECM) is one of the most important components within the tumor microenvironment that supports cancer development and metastasis. Under normal physiological conditions, the ECM is a tightly regulated network providing structural and biochemical support. However, the ECM becomes highly disorganized during neoplastic progression and consequently, stimulates cancer cell transformation, growth and spread. Cancer development and progression is also known to greatly benefit from the support of immune myeloid cells, which have multiple pro-tumorigenic functions including promoting tumor growth, migration and invasion, stimulating angiogenesis and suppressing anti-tumor responses. An increasing number of studies have shown that myeloid cells alter the ECM to support metastatic cancer progression and in turn, the ECM can influence the function of infiltrating myeloid cells. However, the exact nature of this relationship, such as the mechanisms employed and their molecular targets remains unclear. This review discusses evidence for the reciprocal dependence of myeloid cells and the tumor ECM for efficient tumor development and explores potential mechanisms involved in these interactions. A better understanding of this relationship has exciting implications for the development of new therapeutic treatments for metastatic cancer.
Collapse
|
4
|
Kurniawan NA, Chaudhuri PK, Lim CT. Mechanobiology of cell migration in the context of dynamic two-way cell-matrix interactions. J Biomech 2015; 49:1355-1368. [PMID: 26747513 DOI: 10.1016/j.jbiomech.2015.12.023] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Revised: 11/30/2015] [Accepted: 12/14/2015] [Indexed: 12/31/2022]
Abstract
Migration of cells is integral in various physiological processes in all facets of life. These range from embryonic development, morphogenesis, and wound healing, to disease pathology such as cancer metastasis. While cell migratory behavior has been traditionally studied using simple assays on culture dishes, in recent years it has been increasingly realized that the physical, mechanical, and chemical aspects of the matrix are key determinants of the migration mechanism. In this paper, we will describe the mechanobiological changes that accompany the dynamic cell-matrix interactions during cell migration. Furthermore, we will review what is to date known about how these changes feed back to the dynamics and biomechanical properties of the cell and the matrix. Elucidating the role of these intimate cell-matrix interactions will provide not only a better multi-scale understanding of cell motility in its physiological context, but also a more holistic perspective for designing approaches to regulate cell behavior.
Collapse
Affiliation(s)
- Nicholas A Kurniawan
- Department of Biomedical Engineering, Eindhoven University of Technology, P.O. Box 513, 5600MB Eindhoven, The Netherlands; Department of Systems Biophysics, FOM Institute AMOLF, Amsterdam, The Netherlands.
| | | | - Chwee Teck Lim
- Mechanobiology Institute, National University of Singapore, Singapore; Department of Biomedical Engineering, National University of Singapore, Singapore.
| |
Collapse
|
5
|
O'Sullivan S, Medina C, Ledwidge M, Radomski MW, Gilmer JF. Nitric oxide-matrix metaloproteinase-9 interactions: biological and pharmacological significance--NO and MMP-9 interactions. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2014; 1843:603-17. [PMID: 24333402 DOI: 10.1016/j.bbamcr.2013.12.006] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Revised: 12/02/2013] [Accepted: 12/05/2013] [Indexed: 12/24/2022]
Abstract
Nitric oxide (NO) and matrix metalloproteinase 9 (MMP-9) levels are found to increase in inflammation states and in cancer, and their levels may be reciprocally modulated. Understanding interactions between NO and MMP-9 is of biological and pharmacological relevance and may prove crucial in designing new therapeutics. The reciprocal interaction between NO and MMP-9 have been studied for nearly twenty years but to our knowledge, are yet to be the subject of a review. This review provides a summary of published data regarding the complex and sometimes contradictory effects of NO on MMP-9. We also analyse molecular mechanisms modulating and mediating NO-MMP-9 interactions. Finally, a potential therapeutic relevance of these interactions is presented.
Collapse
|
6
|
Study of the binding interaction between fluorinated matrix metalloproteinase inhibitors and Human Serum Albumin. Eur J Med Chem 2014; 79:13-23. [DOI: 10.1016/j.ejmech.2014.03.064] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Revised: 02/18/2014] [Accepted: 03/21/2014] [Indexed: 11/18/2022]
|
7
|
Li H, Yu SS, Miteva M, Nelson CE, Werfel T, Giorgio TD, Duvall CL. Matrix Metalloproteinase Responsive, Proximity-activated Polymeric Nanoparticles for siRNA Delivery. ADVANCED FUNCTIONAL MATERIALS 2013; 23:3040-3052. [PMID: 25214828 PMCID: PMC4159188 DOI: 10.1002/adfm.201202215] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Small interfering RNA (siRNA) has significant potential to evolve into a new class of pharmaceutical inhibitors, but technologies that enable robust, tissue-specific intracellular delivery must be developed before effective clinical translation can be achieved. A pH-responsive, smart polymeric nanoparticle (SPN) with matrix metalloproteinase (MMP)-7-dependent proximity-activated targeting (PAT) is described here. The PAT-SPN was designed to trigger cellular uptake and cytosolic delivery of siRNA once activated by MMP-7, an enzyme whose overexpression is a hallmark of cancer initiation and progression. The PAT-SPN is composed of a corona-forming PEG block, an MMP-7-cleavable peptide, a cationic siRNA-condensing block, and a pH-responsive, endosomolytic terpolymer block that drives self-assembly and forms the PAT-SPN core. With this novel design, the PEG corona shields cellular interactions until it is cleaved in MMP-7-rich environments, shifting SPNζ-potential from +5.8 to +14.4 mV and triggering a 2.5 fold increase in carrier internalization. The PAT-SPN exhibited pH-dependent membrane disruptive behavior that enabled siRNA escape from endo-lysosomal pathways. Efficient intracellular siRNA delivery and knockdown of the model enzyme luciferase in R221A-Luc mammary tumor cellssignificantly depended on MMP-7 pre-activation. These combined data indicate that the PAT-SPN provides a promising new platform for tissue-specific, proximity-activated siRNA delivery to MMP-rich pathological environments.
Collapse
Affiliation(s)
- Hongmei Li
- Department of Biomedical Engineering, Vanderbilt University, VU Station B, Box 351631, Nashville, TN, USA; Vanderbilt Institute of Nanoscale Science and Engineering, Vanderbilt University, Nashville, TN, USA
| | - Shann S. Yu
- Department of Biomedical Engineering, Vanderbilt University, VU Station B, Box 351631, Nashville, TN, USA
| | - Martina Miteva
- Department of Biomedical Engineering, Vanderbilt University, VU Station B, Box 351631, Nashville, TN, USA
| | - Christopher E. Nelson
- Department of Biomedical Engineering, Vanderbilt University, VU Station B, Box 351631, Nashville, TN, USA
| | - Thomas Werfel
- Vanderbilt Institute of Nanoscale Science and Engineering, Vanderbilt University, Nashville, TN, USA; Department of Engineering and Physics, Murray State University, Murray, KY, USA
| | - Todd D. Giorgio
- Department of Biomedical Engineering, Vanderbilt University, VU Station B, Box 351631, Nashville, TN, USA; Department of Cancer Biology, Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Craig L. Duvall
- Department of Biomedical Engineering, Vanderbilt University, VU Station B, Box 351631, Nashville, TN, USA; Vanderbilt Institute of Nanoscale Science and Engineering, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
8
|
Casalini F, Fugazza L, Esposito G, Cabella C, Brioschi C, Cordaro A, D’Angeli L, Bartoli A, Filannino AM, Gringeri CV, Longo DL, Muzio V, Nuti E, Orlandini E, Figlia G, Quattrini A, Tei L, Digilio G, Rossello A, Maiocchi A. Synthesis and Preliminary Evaluation in Tumor Bearing Mice of New 18F-Labeled Arylsulfone Matrix Metalloproteinase Inhibitors as Tracers for Positron Emission Tomography. J Med Chem 2013; 56:2676-89. [DOI: 10.1021/jm4001743] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Francesca Casalini
- Department of Science and Technological
Innovation, Università del Piemonte Orientale “Amedeo Avogadro”, Viale T. Michel 11,
I-15121 Alessandria, Italy
| | - Lorenza Fugazza
- Research and Development, Advanced Accelerator Applications, Via Ribes 5, I-10010
Colleretto Giacosa (TO), Italy
| | - Giovanna Esposito
- Molecular Imaging Centre, University of Torino, Via Nizza 52, I-10126 Torino,
Italy
| | - Claudia Cabella
- Centro Ricerche Bracco, Bracco Imaging S.p.A., Via Ribes 5, I-10010 Colleretto
Giacosa (TO), Italy
| | - Chiara Brioschi
- Centro Ricerche Bracco, Bracco Imaging S.p.A., Via Ribes 5, I-10010 Colleretto
Giacosa (TO), Italy
| | - Alessia Cordaro
- Centro Ricerche Bracco, Bracco Imaging S.p.A., Via Ribes 5, I-10010 Colleretto
Giacosa (TO), Italy
| | - Luca D’Angeli
- Molecular Imaging Centre, University of Torino, Via Nizza 52, I-10126 Torino,
Italy
| | - Antonietta Bartoli
- Molecular Imaging Centre, University of Torino, Via Nizza 52, I-10126 Torino,
Italy
| | - Azzurra M. Filannino
- Research and Development, Advanced Accelerator Applications, Via Ribes 5, I-10010
Colleretto Giacosa (TO), Italy
| | - Concetta V. Gringeri
- Department of Science and Technological
Innovation, Università del Piemonte Orientale “Amedeo Avogadro”, Viale T. Michel 11,
I-15121 Alessandria, Italy
| | - Dario L. Longo
- Molecular Imaging Centre, University of Torino, Via Nizza 52, I-10126 Torino,
Italy
| | - Valeria Muzio
- Research and Development, Advanced Accelerator Applications, Via Ribes 5, I-10010
Colleretto Giacosa (TO), Italy
| | - Elisa Nuti
- Department
of Pharmacy, University of Pisa, Via Bonanno
6, I-56126 Pisa, Italy
| | | | - Gianluca Figlia
- Institute of Experimental Neurology,
Division of Neuroscience, San Raffaele Scientific Institute, Via Olgettina 58, 20132 Milan, Italy
| | - Angelo Quattrini
- Institute of Experimental Neurology,
Division of Neuroscience, San Raffaele Scientific Institute, Via Olgettina 58, 20132 Milan, Italy
| | - Lorenzo Tei
- Department of Science and Technological
Innovation, Università del Piemonte Orientale “Amedeo Avogadro”, Viale T. Michel 11,
I-15121 Alessandria, Italy
| | - Giuseppe Digilio
- Department of Science and Technological
Innovation, Università del Piemonte Orientale “Amedeo Avogadro”, Viale T. Michel 11,
I-15121 Alessandria, Italy
- Molecular Imaging Centre, University of Torino, Via Nizza 52, I-10126 Torino,
Italy
| | - Armando Rossello
- Department
of Pharmacy, University of Pisa, Via Bonanno
6, I-56126 Pisa, Italy
| | - Alessandro Maiocchi
- Centro Ricerche Bracco, Bracco Imaging S.p.A., Via Ribes 5, I-10010 Colleretto
Giacosa (TO), Italy
| |
Collapse
|
9
|
Zhang CY, Zhang L, Yu HX, Bao JD, Sun Z, Lu RR. Curcumin inhibits invasion and metastasis in K1 papillary thyroid cancer cells. Food Chem 2013; 139:1021-8. [PMID: 23561205 DOI: 10.1016/j.foodchem.2013.02.016] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2012] [Revised: 12/12/2012] [Accepted: 02/05/2013] [Indexed: 12/11/2022]
Abstract
Curcumin, the active constituent of dietary spice turmeric, possesses a strong potential for cancer prevention and treatment. However, there is no study to address the effects of curcumin on invasion and metastasis of thyroid cancers. Thyroid cancer is the most common malignancy of endocrine organs, and its incidence rates have steadily increased over recent decades. Although most indolent tumours can be effectively managed, metastatic tumours at distant secondary sites behave aggressively and currently there is no effective form of treatment. Here, for the first time it has been reported that curcumin inhibit multiple metastasis steps of K1 papillary thyroid cancer cells. Curcumin dose-dependently suppressed viability of K1 cells as well as its cell attachment, spreading, migration and invasion abilities. Moreover, curcumin could also down-regulate the expression and activity of matrix metalloproteinase-9 (MMP-9). The findings showed that curcumin might be an effective tumouristatic agent for the treatment of aggressive papillary thyroid carcinomas.
Collapse
Affiliation(s)
- Chi-Yu Zhang
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Institute of Nuclear Medicine, 20 Qian Rong Road, Wuxi, Jiangsu 214063, China
| | | | | | | | | | | |
Collapse
|
10
|
Gringeri CV, Menchise V, Rizzitelli S, Cittadino E, Catanzaro V, Dati G, Chaabane L, Digilio G, Aime S. Novel Gd(III)-based probes for MR molecular imaging of matrix metalloproteinases. CONTRAST MEDIA & MOLECULAR IMAGING 2012; 7:175-84. [PMID: 22434630 DOI: 10.1002/cmmi.478] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Two novel Gd-based contrast agents (CAs) for the molecular imaging of matrix metalloproteinases (MMPs) were synthetized and characterized in vitro and in vivo. These probes were based on the PLG*LWAR peptide sequence, known to be hydrolyzed between Gly and Leu by a broad panel of MMPs. A Gd-DOTA chelate was conjugated to the N-terminal position through an amide bond, either directly to proline (compd Gd-K11) or through a hydrophilic spacer (compd Gd-K11N). Both CA were made strongly amphiphilic by conjugating an alkyl chain at the C-terminus of the peptide sequence. Gd-K11 and Gd-K11N have a good affinity for β-cyclodextrins (K(D) 310 and 670 µ m respectively) and for serum albumin (K(D) 350 and 90 µ m respectively), and can be efficiently cleaved in vitro at the expected site by MMP-2 and MMP-12. Upon MMP-dependent cleavage, the CAs lose the C-terminal tetrapeptide and the alkyl chain, thus undergoing to an amphiphilic-to-hydrophilic transformation that is expected to alter tissue pharmacokinetics. To prove this, Gd-K11 was systemically administered to mice bearing a subcutaneous B16.F10 melanoma, either pre-treated or not with the broad spectrum MMP inhibitor GM6001 (Ilomastat). The washout of the Gd-contrast enhancement in MR images was significantly faster for untreated subjects (displaying MMP activity) with respect to treated ones (MMP activity inhibited). The washout kinetics of Gd-contrast enhancement from the tumor microenvironment could be then interpreted in terms of the local activity of MMPs.
Collapse
Affiliation(s)
- Concetta V Gringeri
- Department of Environmental and Life Sciences, Università del Piemonte Orientale A. Avogadro, Viale T. Michel 11, 15121, Alessandria, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
11
|
The rs522616 polymorphism in the matrix metalloproteinase-3 (MMP-3) gene is associated with sporadic brain arteriovenous malformation in a Chinese population. J Clin Neurosci 2010; 17:1568-72. [DOI: 10.1016/j.jocn.2010.04.023] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2010] [Revised: 03/29/2010] [Accepted: 04/04/2010] [Indexed: 01/25/2023]
|
12
|
Li H, Yoneda M, Takeyama M, Sugita I, Tsunekawa H, Yamada H, Watanabe D, Mukai T, Yamamura M, Iwaki M, Zako M. Effect of Infliximab on Tumor Necrosis Factor-Alpha-Induced Alterations in Retinal Microvascular Endothelial Cells and Retinal Pigment Epithelial Cells. J Ocul Pharmacol Ther 2010; 26:549-56. [DOI: 10.1089/jop.2010.0079] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- Huili Li
- Department of Ophthalmology, Aichi Medical University, Aichi, Japan
| | - Masahiko Yoneda
- Department of Biochemistry and Molecular biology, Aichi Prefectural College of Nursing and Health, Aichi, Japan
| | | | - Iichiro Sugita
- Department of Ophthalmology, Aichi Medical University, Aichi, Japan
| | - Hinako Tsunekawa
- Department of Ophthalmology, Aichi Medical University, Aichi, Japan
| | - Hiroshi Yamada
- Department of Ophthalmology, Aichi Medical University, Aichi, Japan
| | - Daisuke Watanabe
- Department of Dermatology, Aichi Medical University, Aichi, Japan
| | - Tomoyuki Mukai
- Department of Rheumatology, Aichi Medical University, Aichi, Japan
| | | | - Masayoshi Iwaki
- Department of Ophthalmology, Aichi Medical University, Aichi, Japan
| | - Masahiro Zako
- Department of Ophthalmology, Aichi Medical University, Aichi, Japan
| |
Collapse
|
13
|
Nalla AK, Gorantla B, Gondi CS, Lakka SS, Rao JS. Targeting MMP-9, uPAR, and cathepsin B inhibits invasion, migration and activates apoptosis in prostate cancer cells. Cancer Gene Ther 2010; 17:599-613. [PMID: 20448670 DOI: 10.1038/cgt.2010.16] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Prostate cancer is one of the most commonly diagnosed cancers and the second leading cause of cancer deaths in Americans. The high mortality rate is mainly attributed to the invasiveness and metastasis of advanced prostate cancer. Targeting the molecules involved in metastasis could be an effective mode of treatment for prostate cancer. In this study, the therapeutic potential of siRNA-mediated targeting of matrix metalloproteinase-9 (MMP-9), urokinase plasminogen activator receptor (uPAR), and cathepsin B (CB) in prostate cancer was carried out using single and bi-cistronic siRNA-expressing constructs. Downregulation of MMP-9, uPAR, and CB inhibited matrigel invasion, in vitro angiogenesis and wound-healing migration ability of PC3 and DU145 prostate cancer cell lines. In addition, the siRNA treatments induced apoptosis in the tumor cells as determined by TUNEL and DNA laddering assays. An attempt to elucidate the apoptotic pathway showed the involvement of FAS-mediated activation of caspases-8 and -7. Further, mice with orthotopic prostate tumors treated with siRNA-expressing vectors showed significant inhibition in tumor growth and migration. In conclusion, we report that the siRNA-mediated knockdown of MMP-9, uPAR, and CB inhibits invasiveness and migration of prostate cancer cells and leads to apoptosis both in vitro and in vivo.
Collapse
Affiliation(s)
- A K Nalla
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine, Peoria, IL 61605, USA
| | | | | | | | | |
Collapse
|
14
|
Singh S, Singh R, Singh UP, Rai SN, Novakovic KR, Chung LWK, Didier PJ, Grizzle WE, Lillard JW. Clinical and biological significance of CXCR5 expressed by prostate cancer specimens and cell lines. Int J Cancer 2009; 125:2288-95. [PMID: 19610059 DOI: 10.1002/ijc.24574] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Chemokines and chemokine receptors have been shown to be involved in metastatic process of prostate cancer (PCa). In this study, we show primary PCa tissues and cell lines (LNCaP and PC3) express CXCR5, a specific chemokine receptor for CXCL13. Expression of CXCR5 was significantly higher (p < 0.001) in PCa cases than compared to normal match (NM) tissues. CXCR5 intensity correlated (R(2) = 0.97) with Gleason score. While prostate tumor tissues with Gleason scores >or= 7, displayed predominantly nuclear CXCR5 expression patterns, PCa specimens with Gleason scores <or= 6 showed predominantly membrane and cytoplasmic expression patterns that were comparable to benign prostatic hyperplasia (BPH). Similar to tissue expression, PCa cell lines expressed significantly more CXCR5 than normal prostatic epithelial cells (PrECs), and CXCR5 expression was distributed among intracellular and extracellular compartments. Functional in vitro assays showed higher migratory and invasive potentials toward CXCL13, an effect that was mediated by CXCR5. In both PCa cell lines, CXCL13 treatment increased the expression of collagenase-1 or matrix metalloproteinase-1 (MMP-1), collagenase-3 (MMP-13), stromelysin-1 (MMP-3), stromelysin-2 (MMP-10) and stromelysin-3 (MMP-11). These data demonstrate the clinical and biological relevance of the CXCL13-CXCR5 pathway and its role in PCa cell invasion and migration.
Collapse
Affiliation(s)
- Shailesh Singh
- Department of Microbiology and Immunology, James Graham Brown Cancer Center, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Li Y, Jia JH, Kang S, Zhang XJ, Zhao J, Wang N, Zhou RM, Sun DL, Duan YN, Wang DJ. The functional polymorphisms on promoter region of matrix metalloproteinase-12, -13 genes may alter the risk of epithelial ovarian carcinoma in Chinese. Int J Gynecol Cancer 2009; 19:129-33. [PMID: 19258954 DOI: 10.1111/igc.0b013e31819a1d8e] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
BACKGROUNDS AND AIMS Growing evidences indicate that single nucleotide polymorphisms (SNPs) of matrix metalloproteinases (MMPs) gene promoter may alter MMPs protein expression levels to influence malignant tumors developing and progressing. Our study was to assess the effects of the SNPs in the promoter region of MMP-12 and MMP-13 on the risk of epithelial ovarian carcinoma (EOC) developing and progressing. METHODS MMP-12 A-82G and MMP-13 A-77G SNPs were genotyped by polymerase chain reaction-restriction fragment length polymorphism in 256 EOC patients and 329 controls. RESULTS The A/G genotype frequency of MMP-12 was significantly higher in patients than in controls (7.0% vs 3.3%, P = 0.04); similarly, the frequency of MMP-12 82G allele was higher in patients too (P = 0.04). Compared with A/A genotype, A/G genotype significantly increased the risk of EOC (odds ratio, 2.19; 95% confidence interval, 1.01-4.72). Age-stratified analysis showed that individuals with A/G genotype had a higher risk in the final diagnosis aged younger than 50 years. We observed no overall association between MMP-13-77A/G polymorphism and EOC. However, an elevated positive association was observed for A/A versus G/G + A/G genotypes in mucinous ovarian cancer. Combining the analyzed 2 SNPs, the haplotype distributions in patients were not significantly different from that in controls. CONCLUSION These results suggested that the G allele of the MMP-12 82A/G polymorphism might be a risk factor for the development and progression of EOC and that the A/A genotype of MMP-13-77A/G polymorphism was associated with special pathological subtype and clinical stage in EOC at least in Chinese women.
Collapse
Affiliation(s)
- Yan Li
- Department of Obstetrics and Gynaecology, Hebei Medical University, Fourth Hospital, Shijiazhuang, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Abstract
Early detection of both primary tumors and metastatic disease remains a major challenge in the diagnosis and staging of cancer. The recognition of the role of MMPs in both the growth and metastasis of tumors has guided the development not only of therapeutic strategies utilizing synthetic, small-molecule MMP inhibitors (MMPIs), but has also catalyzed methods to detect and image tumors in vivo by means of tumor-associated proteolytic activity. These imaging approaches target MMPs involved in cancer progression via contrast agents linked to MMPIs or to MMP selective and specific substrates with sensitivity enhanced by amplification during enzymatic processing. This review draws attention to a variety of strategies utilized to image MMP activity in vivo.
Collapse
|
17
|
Grzywa R, Oleksyszyn J. First synthesis of α-aminoalkyl-(N-substituted)thiocarbamoyl-phosphinates: Inhibitors of aminopeptidase N (APN/CD13) with the new zinc-binding group. Bioorg Med Chem Lett 2008; 18:3734-6. [DOI: 10.1016/j.bmcl.2008.05.050] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2008] [Revised: 05/14/2008] [Accepted: 05/14/2008] [Indexed: 11/25/2022]
|
18
|
Burk J, Burggraf D, Vosko M, Dichgans M, Hamann GF. Protection of cerebral microvasculature after moderate hypothermia following experimental focal cerebral ischemia in mice. Brain Res 2008; 1226:248-55. [PMID: 18586014 DOI: 10.1016/j.brainres.2008.06.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2008] [Revised: 06/08/2008] [Accepted: 06/10/2008] [Indexed: 11/29/2022]
Abstract
Clinical studies have shown that the treatment of ischemic stroke with hypothermia is promising. In this animal study, we investigated the fate of the microvasculature following focal cerebral ischemia in mice with and without hypothermia. Focal cerebral ischemia was induced by occlusion of the middle cerebral artery (MCAO) (3 h) with an intraluminal filament technique. Eight mice received normothermia (36.5 degrees C, NT) and eight received hypothermia (32-34 degrees C, HT) treatment during 24 h of reperfusion. Another six mice represented the sham group. Analysis of the hypothermic group in comparison to the normothermic group revealed a significantly reduced infarct volume (NT: 63.56+/-4.62 mm3 SEM, HT: 38.09+/-4.83 mm3 SEM; P<0.01) and showed considerably ameliorated neurological deficits (Garcia-score) after 24 h (P<0.01). In addition, the degradation of the microvascular basal lamina antigen collagen type IV after normothermia was strongly reduced (P<0.05) compared to sham. Hypothermia diminished this effect so that collagen type IV was not significantly reduced compared to sham. Moreover the hemoglobin extravasation was strongly reduced under hypothermic treatment compared to the normothermic group (P<0.01). In the hypothermia group the urokinase plasminogen-activator (uPA) activity (P=0.01) was significantly decreased compared to the normothermia group. Also MMP-9 was significantly reduced (P<0.05) during hypothermic treatment. In conclusion, for the first time we show in mice that hypothermia preserves the microvascular wall structures after ischemia. We have demonstrated that hypothermia protects the basal lamina, reduces the infarct volume and hemorrhage, and reduces proteolytic enzymes. These protective effects in an additional animal model of ischemia and reperfusion strongly recommend hypothermia as a potential beneficial treatment for stroke.
Collapse
Affiliation(s)
- Jan Burk
- Department of Neurology, Ludwig-Maximilians University, Klinikum Grosshadern, Marchioninistr. 15, 81377 Munich, Germany
| | | | | | | | | |
Collapse
|
19
|
Hayashi R, Jin X, Cook GR. Synthesis and evaluation of novel heterocyclic MMP inhibitors. Bioorg Med Chem Lett 2007; 17:6864-70. [PMID: 18029173 PMCID: PMC2574968 DOI: 10.1016/j.bmcl.2007.10.020] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2007] [Revised: 10/02/2007] [Accepted: 10/05/2007] [Indexed: 11/23/2022]
Abstract
A variety of novel heterocyclic compounds were synthesized and evaluated for MMP inhibition. Broad spectrum inhibition of MMPs 1, 2, 9, and 12 was found with pyridinone-based compounds while N-heterocyclic triazoles and tetrazoles were largely ineffective. A highly selective tetrazole inhibitor for MMP-2 was discovered.
Collapse
Affiliation(s)
- Ryuji Hayashi
- Center for Protease Research, Department of Chemistry and Molecular Biology, North Dakota State University, Fargo, ND 58105, USA
| | - Xiaomin Jin
- Center for Protease Research, Department of Chemistry and Molecular Biology, North Dakota State University, Fargo, ND 58105, USA
| | - Gregory R. Cook
- Center for Protease Research, Department of Chemistry and Molecular Biology, North Dakota State University, Fargo, ND 58105, USA
| |
Collapse
|
20
|
Kunigal S, Lakka SS, Gondi CS, Estes N, Rao JS. RNAi-mediated downregulation of urokinase plasminogen activator receptor and matrix metalloprotease-9 in human breast cancer cells results in decreased tumor invasion, angiogenesis and growth. Int J Cancer 2007; 121:2307-16. [PMID: 17657740 PMCID: PMC2396459 DOI: 10.1002/ijc.22962] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The serine protease urokinase-type plasminogen activator (uPA) plays a significant role in tumor cell invasion and metastasis when bound to its specific receptor, uPAR (also known as CD87). In addition to the uPA-uPAR system, matrix metalloproteinases (MMPs) are involved in tumor cell invasion and metastasis. In this study, we achieved specific inhibition of uPAR and MMP-9 using RNAi technology. We introduced small interfering RNA to downregulate the expression of uPAR and MMP-9 (pUM) in breast cancer cell lines (MDA MB 231 and ZR 75 1). In vitro angiogenesis studies indicated a decrease in the angiogenic potential of the treated cells; in particular, a remarkable decrease was observed in the cells treated with bicistronic construct (pUM) in comparision to the controls. Additionally, bicistronic construct inhibited the formation of capillary-like structures in in vivo models of angiogenesis. Similarly, the invasive potential and migration decreased dramatically when treated with the bicistronic construct as shown by matrigel invasion and migration assays. These results suggest a synergistic effect from the simultaneous downregulation of uPAR and MMP-9. We also assessed the levels of phosphorylated forms of MAPK, ERK and AKT signaling pathway molecules and found reduction in the levels of these molecules in cells treated with the bicistronic construct as compared to the control cells. Furthermore, targeting both uPAR and MMP-9 totally regressed orthotopic breast tumors in nude mice. In conclusion, our results provide evidence that the simultaneous downregulation of uPAR and MMP-9 using RNAi technology may provide an effective tool for breast cancer therapy.
Collapse
Affiliation(s)
- Sateesh Kunigal
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, One Illini Drive, Peoria, IL, 61605
| | - Sajani S. Lakka
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, One Illini Drive, Peoria, IL, 61605
| | - Christopher S. Gondi
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, One Illini Drive, Peoria, IL, 61605
| | - Norman Estes
- Department of Surgery, University of Illinois College of Medicine at Peoria, One Illini Drive, Peoria, IL, 61605
| | - Jasti S. Rao
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, One Illini Drive, Peoria, IL, 61605
- Department of Neurosurgery, University of Illinois College of Medicine at Peoria, One Illini Drive, Peoria, IL, 61605
- *Correspondence: J.S. Rao, Ph.D., Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, One Illini Drive, Peoria, IL 61605, USA: e-mail: ., FAX (309) 671-3442
| |
Collapse
|
21
|
Abdallah MA, Abdullah HI, Kang S, Taylor DD, Nakajima ST, Gercel-Taylor C. Effects of the components of hormone therapy on matrix metalloproteinases in breast-cancer cells: an in vitro study. Fertil Steril 2007; 87:978-81. [PMID: 17207794 DOI: 10.1016/j.fertnstert.2006.08.091] [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] [Received: 12/28/2005] [Revised: 08/02/2006] [Accepted: 08/02/2006] [Indexed: 10/23/2022]
Abstract
A combination of E and progestogens significantly increased matrix metalloproteinase (MMP)-2 in both T47D cells (E(2)-medroxyprogesteroneacetate [MPA] and E(2)-P) and MCF-7 cells (E(2)-MPA, E(2)-P, and equilin-MPA). All combinations resulted in higher MMP-9 levels in MCF-7 cells, but higher MMP-9 levels resulted only with equilin-norethinderone in T47D cells.
Collapse
|
22
|
Heist RS, Marshall AL, Liu G, Zhou W, Su L, Neuberg D, Lynch TJ, Wain J, Christiani DC. Matrix Metalloproteinase Polymorphisms and Survival in Stage I Non–Small Cell Lung Cancer. Clin Cancer Res 2006; 12:5448-53. [PMID: 17000679 DOI: 10.1158/1078-0432.ccr-06-0262] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE The matrix metalloproteinases (MMP) are a family of enzymes that can degrade extracellular matrix and facilitate invasion through the basement membrane. Several polymorphisms in MMP-1, MMP-2, MMP-3, and MMP-12 have been described, some of which lead to differential transcription. We hypothesized that polymorphisms in these MMP genes may be associated with survival outcomes in early-stage non-small cell lung cancer (NSCLC). EXPERIMENTAL DESIGN We evaluated the relationship between MMP-1, MMP-2, MMP-3, and MMP-12 polymorphisms and both recurrence-free survival (RFS) and overall survival (OS) among 382 patients with stage I NSCLC. Analyses of genotype associations with survival outcomes were done using Cox proportional hazards models and Kaplan-Meier methods and the log-rank test. RESULTS Patients carrying the variant G allele of the MMP-12 1082A/G polymorphism had significantly worse outcomes [crude hazard ratio (HR) for OS 1.74; 95% confidence interval (95% CI), 1.18-2.58, P=0.006; crude HR for RFS, 1.53; 95% CI, 1.05-2.23, P=0.03]. After adjusting for age, sex, stage, pack-years of smoking, and histologic subtype, the MMP-12 1082A/G polymorphism remained significantly associated with survival outcomes [adjusted HR (AHR) for OS, 1.94; 95% CI, 1.28-2.97, P=0.002; AHR for RFS, 1.61; 95% CI, 1.07-2.41, P=0.02]. None of the other MMP polymorphisms was significantly associated with survival. CONCLUSIONS Our results show that patients with stage I NSCLC carrying the variant G allele of the MMP-12 1082A/G polymorphism have worse survival.
Collapse
Affiliation(s)
- Rebecca Suk Heist
- Massachusetts General Hospital and Harvard School of Public Health, Boston, Massachusetts 02115, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Leyden J, Murray D, Moss A, Arumuguma M, Doyle E, McEntee G, O'Keane C, Doran P, MacMathuna P. Net1 and Myeov: computationally identified mediators of gastric cancer. Br J Cancer 2006; 94:1204-12. [PMID: 16552434 PMCID: PMC2361249 DOI: 10.1038/sj.bjc.6603054] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Gastric adenocarcinoma (GA) is a significant cause of mortality worldwide. The molecular mechanisms of GA remain poorly characterised. Our aim was to characterise the functional activity of the computationally identified genes, NET 1 and MYEOV in GA. Digital Differential Display was used to identify genes altered expression in GA-derived EST libraries. mRNA levels of a subset of genes were quantitated by qPCR in a panel of cell lines and tumour tissue. The effect of pro- and anti-inflammatory stimuli on gene expression was investigated. Cell proliferation and invasion were measured using in an in-vitro GA model following inhibition of expression using siRNA. In all, 23 genes not previously reported in association with GA were identified. Two genes, Net1 and Myeov, were selected for further analysis and increased expression was detected in GA tissue compared to paired normal tissue using quantitative PCR. siRNA-mediated downregulation of Net1 and Myeov resulted in decreased proliferation and invasion of gastric cancer cells in vitro. These functional studies highlight a putative role for NET1 and Myeov in the development and progression of gastric cancer. These genes may provide important targets for intervention in GA, evidenced by their role in promoting invasion and proliferation, key phenotypic hallmarks of cancer cells.
Collapse
Affiliation(s)
- J Leyden
- Gastrointestinal Unit, Mater Misericordiae University Hospital, University College Dublin, Ireland
| | - D Murray
- Genome Resource Unit, Department of Medicine and Therapeutics, Mater Misericordiae University Hospital, University College Dublin, Dublin 7, Ireland
| | - A Moss
- Gastrointestinal Unit, Mater Misericordiae University Hospital, University College Dublin, Ireland
| | - M Arumuguma
- Department of Surgery, Mater Misericordiae University Hospital, University College Dublin, Ireland
| | - E Doyle
- Department of Pathology, Mater Misericordiae University Hospital, University College Dublin, Ireland
| | - G McEntee
- Department of Surgery, Mater Misericordiae University Hospital, University College Dublin, Ireland
| | - C O'Keane
- Department of Pathology, Mater Misericordiae University Hospital, University College Dublin, Ireland
| | - P Doran
- Genome Resource Unit, Department of Medicine and Therapeutics, Mater Misericordiae University Hospital, University College Dublin, Dublin 7, Ireland
- Genome Resource Unit, Department of Medicine and Therapeutics, Mater Misericordiae University Hospital, University College Dublin, Dublin 7, Ireland. E-mail:
| | - P MacMathuna
- Gastrointestinal Unit, Mater Misericordiae University Hospital, University College Dublin, Ireland
| |
Collapse
|
24
|
Lee CZ, Yao JS, Huang Y, Zhai W, Liu W, Guglielmo BJ, Lin E, Yang GY, Young WL. Dose-response effect of tetracyclines on cerebral matrix metalloproteinase-9 after vascular endothelial growth factor hyperstimulation. J Cereb Blood Flow Metab 2006; 26:1157-64. [PMID: 16395286 DOI: 10.1038/sj.jcbfm.9600268] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Brain arteriovenous malformations (BAVMs) are a potentially life-threatening disorder. Matrix metalloproteinase (MMP)-9 activity was greatly increased in BAVM tissue specimens. Doxycycline was shown to decrease cerebral MMP-9 activities and angiogenesis induced by vascular endothelial growth factor (VEGF). In the present study, we determined the dose-response effects of doxycycline and minocycline on cerebral MMP-9 using our mouse model with VEGF focal hyperstimulation delivered with adenoviral vector (AdVEGF) in the brain. Mice were treated with doxycycline or minocycline, respectively, at 1, 5, 10, 30, 50, or 100 mg/kg/day through drinking water for 1 week. Our results have shown that MMP-9 messenger ribonucleic acid (mRNA) expression was inhibited by doxycycline starting at 10 mg/kg/day (P<0.02). Minocycline showed more potent inhibition on MMP-9 mRNA expression, starting at 1 (P<0.005) and further at more than 30 (P<0.001) mg/kg/day. At the enzymatic activity level, doxycycline started to suppress MMP-9 activity at 5 mg/kg/day (P<0.001), while minocycline had an effect at a lower dose, 1 mg/kg/day (P<0.02). The inhibition of cerebral MMP-9 mRNA and activity were highly correlated with drug levels in the brain tissue. We also assessed the potential relevant signaling pathway in vitro to elucidate the mechanisms underlying the MMP-9 inhibition by tetracyclines. In vitro, minocycline, but not doxycycline, inhibits MMP-9, at least in part, via the extracellular signaling-related kinase 1/2 (ERK1/2)-mediated pathway. This study provided the evidence that the tetracyclines inhibit stimulated cerebral MMP-9 at multiple levels and are effective at very low doses, offering great potential for therapeutic use.
Collapse
MESH Headings
- Animals
- Anti-Bacterial Agents/metabolism
- Anti-Bacterial Agents/pharmacology
- Blotting, Western
- Brain/metabolism
- Brain Chemistry/drug effects
- Cell Movement/drug effects
- Cells, Cultured
- Dose-Response Relationship, Drug
- Doxycycline/metabolism
- Doxycycline/pharmacology
- Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors
- Gelatin/metabolism
- Intracranial Arteriovenous Malformations/enzymology
- Male
- Matrix Metalloproteinase 9/metabolism
- Mice
- Minocycline/metabolism
- Minocycline/pharmacology
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- RNA, Messenger/biosynthesis
- RNA, Messenger/genetics
- Reverse Transcriptase Polymerase Chain Reaction
- Stimulation, Chemical
- Tetracyclines/metabolism
- Tetracyclines/pharmacology
- Vascular Endothelial Growth Factor A/pharmacology
Collapse
Affiliation(s)
- Chanhung Z Lee
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, 94110, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Kaliski A, Maggiorella L, Cengel KA, Mathe D, Rouffiac V, Opolon P, Lassau N, Bourhis J, Deutsch E. Angiogenesis and tumor growth inhibition by a matrix metalloproteinase inhibitor targeting radiation-induced invasion. Mol Cancer Ther 2006; 4:1717-28. [PMID: 16275993 DOI: 10.1158/1535-7163.mct-05-0179] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In this study, we have evaluated the interactions between ionizing radiation and a matrix metalloproteinase (MMP) inhibitor. Using Matrigel invasion assays, we show that ionizing radiation induced a dose-dependent increase in the invasive phenotype of cultured B16 melanoma cells and that conditioned medium from these irradiated B16 cells promoted endothelial cell [human microvascular endothelial cells (HMEC)] invasiveness. To determine whether the radiation-induced changes in invasive phenotype could be due to changes in MMP activation, we have tested the ability of the MMP inhibitor Metastat to modulate the ionizing radiation-induced invasive phenotype using both an in vitro melanoma model and a mouse s.c. tumor model. In these studies, Metastat inhibited the ionizing radiation-induced invasive phenotype in cultured B16 cells and similarly inhibited the increase in HMEC invasion induced by conditioned medium from irradiated B16 cells. Conversely, ionizing radiation increased B16 MMP-2 activity and the conditioned medium from irradiated B16 induced HMEC MMP-2 activity. To further investigate the interaction between ionizing radiation and MMP activation, we then studied the effects of ionizing radiation on downstream effectors of the MMP system. We found that ionizing radiation induced vascular endothelial growth factor (VEGF) secretion by B16 melanoma cells and that this secretion was inhibited by Metastat. Similarly, conditioned medium from irradiated B16 was also able to increase VEGF secretion in HMECs. Moreover, ionizing radiation-induced melanoma cell invasiveness was partially inhibited by an anti-VEGF monoclonal antibody. In vivo, ionizing radiation plus concomitant Metastat yielded the greatest growth inhibition of melanoma s.c. tumors and this effect correlated with inhibition of angiogenesis as measured by both Doppler ultrasonography and platelet/endothelial cell adhesion molecule-1 staining. Finally, ionizing radiation modulated MMP-2, VEGF, and VEGF receptor expression in these tumor samples using immunohistochemistry. Taken together, these results suggest that there is an ionizing radiation-induced tumor survival pathway and a possible paracrine ionizing radiation-induced stimulatory pathway emanating from tumor cells toward the endothelial bed that is impeded when Metastat is given simultaneously. This model could provide in vivo evidence of the antitumor efficacy of combining a MMP inhibitor with ionizing radiation to target radiation-induced invasion and angiogenesis.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/chemistry
- Antineoplastic Agents/pharmacology
- Cell Line, Tumor
- Cell Proliferation
- Cell Survival
- Cells, Cultured
- Collagen/chemistry
- Collagen/pharmacology
- Culture Media, Conditioned/pharmacology
- Dose-Response Relationship, Radiation
- Drug Combinations
- Endothelium, Vascular/pathology
- Enzyme Inhibitors/pharmacology
- Enzyme-Linked Immunosorbent Assay
- Humans
- Immunohistochemistry
- Laminin/chemistry
- Laminin/pharmacology
- Matrix Metalloproteinase 2/metabolism
- Matrix Metalloproteinase 9/metabolism
- Matrix Metalloproteinase Inhibitors
- Melanoma, Experimental/drug therapy
- Mice
- Neoplasm Invasiveness
- Neoplasm Transplantation
- Neoplasms, Radiation-Induced/pathology
- Neovascularization, Pathologic
- Phenotype
- Platelet Endothelial Cell Adhesion Molecule-1/biosynthesis
- Proteoglycans/chemistry
- Proteoglycans/pharmacology
- Radiation, Ionizing
- Time Factors
- Ultrasonography
- Ultrasonography, Doppler
- Vascular Endothelial Growth Factor A/immunology
- Vascular Endothelial Growth Factor A/metabolism
Collapse
Affiliation(s)
- Alexandre Kaliski
- Unité Propre de Recherche de l'Enseignement Supérieur-Equipe d'Accueil 27-10, Villejuif, France
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Fu L, Ishizuya-Oka A, Buchholz DR, Amano T, Matsuda H, Shi YB. A Causative Role of Stromelysin-3 in Extracellular Matrix Remodeling and Epithelial Apoptosis during Intestinal Metamorphosis in Xenopus laevis. J Biol Chem 2005; 280:27856-65. [PMID: 15929979 DOI: 10.1074/jbc.m413275200] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The matrix metalloproteinases are a family of proteases capable of degrading various components of the extracellular matrix. Expression studies have implicated the involvement of the matrix metalloproteinase stromelysin-3 (ST3) in tissue remodeling and pathogenesis. However, the in vivo role of ST3 has been difficult to study because of a lack of good animal models. Here we used intestinal remodeling during thyroid hormone-dependent metamorphosis of Xenopus laevis as a model to investigate in vivo the role of ST3 during postembryonic organ development in vertebrates. We generated transgenic tadpoles expressing ST3 under control of a heat shock-inducible promoter. We showed for the first time in vivo that wild type ST3 but not a catalytically inactive mutant was sufficient to induce larval epithelial cell death and fibroblast activation, events that normally occur only in the presence of thyroid hormone. We further demonstrated that these changes in cell fate are associated with altered gene expression in the intestine and remodeling of the intestinal basal lamina. These results thus suggest that ST3 regulates cell fate and tissue morphogenesis through direct or indirect ECM remodeling.
Collapse
Affiliation(s)
- Liezhen Fu
- Laboratory of Gene Regulation and Development, NICHD, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | | | |
Collapse
|
27
|
Abstract
Tissues maintain homeostasis by monitoring and responding to varied physical interactions between cells and their microenvironment. In situations where acute tissue damage occurs, such as wounding, pathogenic assault, or toxic exposure, regulatory circuits that monitor tissue homeostasis are rapidly engaged to initiate tissue repair by regulating cell polarity, proliferation and death, matrix metabolism, inflammation, and vascular and lymphatic function. The critical feature of regulating these acute responses is the innate ability to discriminate between homeostatic versus damaged tissue states and engage or disengage regulatory machinery as appropriate; thus, a major distinction between acute versus chronic disease is the altered ability to appropriately activate and?or inactivate reparative regulatory programs. Since cancer is a chronic disease characterized by altered cell polarity, enhanced cell survival, inflammation, increased matrix metabolism, and enhanced vascular and lymphatic function, considerable attention is now focused on understanding the cellular and molecular mechanisms regulating these responsive pathways. Since chemoattractant cytokines are important mediators of leukocyte recruitment following acute tissue stress, and demonstrate altered characteristics of expression and activation in chronically inflamed tissue, they have been implicated as key regulators of inflammation and angiogenesis during cancer development. This chapter focuses on the clinical and experimental data implicating proinflammatory cytokines and chemokines as important potentiators of carcinogenesis.
Collapse
Affiliation(s)
- Stephen C Robinson
- Cancer Research Institute, University of California, San Francisco, 94143, USA
| | | |
Collapse
|
28
|
Hashimoto T, Matsumoto MM, Li JF, Lawton MT, Young WL. Suppression of MMP-9 by doxycycline in brain arteriovenous malformations. BMC Neurol 2005; 5:1. [PMID: 15667660 PMCID: PMC547916 DOI: 10.1186/1471-2377-5-1] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2004] [Accepted: 01/24/2005] [Indexed: 11/16/2022] Open
Abstract
Background The primary aim of this study is to demonstrate the feasibility of utilizing doxycycline to suppress matrix metalloproteinase-9 (MMP-9) in brain arteriovenous malformations (AVMs). Methods Ex-vivo treatment of AVM tissues: Intact AVM tissues were treated with doxycycline for 48 hours. Active and total MMP-9 in the medium were measured. Pilot trial: AVM patients received either doxycycline (100 mg) or placebo twice a day for one week prior to AVM resection. Active and total MMP-9 in BVM tissues were measured. Results Ex-vivo treatment of AVM tissues: Doxycycline at 10 and 100 μg/ml significantly decreased MMP-9 levels in AVM tissues ex-vivo (total: control vs 10 vs 100 μg/ml = 100 ± 6 vs 60 ± 16 vs 61 ± 9%; active: 100 ± 8 vs 48 ± 16 vs 59 ± 10%). Pilot trial: 10 patients received doxycycline, and 4 patients received placebo. There was a trend for both MMP-9 levels to be lower in the doxycycline group than in the placebo group (total: 2.18 ± 1.94 vs 3.26 ± 3.58, P = .50; active: 0.48 ± 0.48 vs 0.95 ± 1.01 ng/100 μg protein, P = .25). Conclusions A clinically relevant concentration of doxycycline decreased MMP-9 in ex-vivo AVM tissues. Furthermore, there was a trend that oral doxycycline for as short as one week resulted in a decrease in MMP-9 in AVM tissues. Further studies are warranted to justify a clinical trial to test effects of doxycycline on MMP-9 expression in AVM tissues.
Collapse
Affiliation(s)
- Tomoki Hashimoto
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, San Francisco, California, USA
- Center for Cerebrovascular Research, University of California, San Francisco, San Francisco, San Francisco, California, USA
| | - Melissa M Matsumoto
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, San Francisco, California, USA
- Center for Cerebrovascular Research, University of California, San Francisco, San Francisco, San Francisco, California, USA
| | - Jenny F Li
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, San Francisco, California, USA
- Center for Cerebrovascular Research, University of California, San Francisco, San Francisco, San Francisco, California, USA
| | - Michael T Lawton
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, San Francisco, California, USA
- Center for Cerebrovascular Research, University of California, San Francisco, San Francisco, San Francisco, California, USA
| | - William L Young
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, San Francisco, California, USA
- Department of Neurology, University of California, San Francisco, San Francisco, San Francisco, California, USA
- Center for Cerebrovascular Research, University of California, San Francisco, San Francisco, San Francisco, California, USA
| | | |
Collapse
|
29
|
Cook GR, Manivannan E, Underdahl T, Lukacova V, Zhang Y, Balaz S. Synthesis and evaluation of novel oxazoline MMP inhibitors. Bioorg Med Chem Lett 2004; 14:4935-9. [PMID: 15341955 DOI: 10.1016/j.bmcl.2004.07.023] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2004] [Revised: 07/09/2004] [Accepted: 07/12/2004] [Indexed: 10/26/2022]
Abstract
MMP inhibitors with novel oxazoline zinc binding groups have been synthesized and evaluated. Selectivity for the inhibition of MMP-9 over MMP-1, MMP-2, and MMP-12 has been achieved in several cases.
Collapse
Affiliation(s)
- Gregory R Cook
- Center for Protease Research, Department of Chemistry, Biochemistry and Molecular Biology, North Dakota State University, Fargo, ND 58105, USA.
| | | | | | | | | | | |
Collapse
|
30
|
Lee CZ, Xu B, Hashimoto T, McCulloch CE, Yang GY, Young WL. Doxycycline Suppresses Cerebral Matrix Metalloproteinase-9 and Angiogenesis Induced by Focal Hyperstimulation of Vascular Endothelial Growth Factor in a Mouse Model. Stroke 2004; 35:1715-9. [PMID: 15166398 DOI: 10.1161/01.str.0000129334.05181.b6] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background and Purpose—
A number of central nervous system (CNS) disorders are associated with abnormalities in or activation of angiogenesis, including vascular malformations. To test the hypothesis that the nonspecific matrix metalloproteinase (MMP) inhibitor, doxycycline, suppresses vascular endothelial growth factor (VEGF)-induced cerebral angiogenesis through inhibition of MMPs, we used a mouse model with enhanced cerebral angiogenesis induced by focal hyperstimulation of VEGF from adenovirus DNA (AdVEGF) transduction.
Methods—
The time course study of MMP activity was performed at 7 and 14 days after AdVEGF transduction. MMP activity and expression were examined by zymography and immunohistochemistry, respectively. As an index of cerebral angiogenesis, microvessel counting was performed in the brains of 3 groups of mice (n=6): (1) control; (2) AdVEGF only; and (3) AdVEGF plus doxycycline (30 mg/kg per day).
Results—
Brain MMP-9 activities increased 4-fold (883±137 versus 179±179; 1-sided
P
<0.001) at 7 days after AdVEGF transduction. VEGF transduction increased vessel counts by 19% (255±27 versus 215±15, 1-sided
P
<0.01). Doxycycline treatment decreased MMP-9 activity (89±72 versus 883±137; 1-sided
P
<0.001) and cerebral microvessel number (231±17 versus 255±27; 1-sided
P
<0.05).
Conclusions—
Doxycycline is effective in decreasing stimulated cerebral MMP-9 activity and parenchymal angiogenesis. The decrease in MMP-9 activity is associated with decreased microvessel counts. Brain pathophysiological processes that involve abnormally enhanced angiogenesis may be amenable to manipulation by MMP inhibitors, including tetracycline derivatives.
Collapse
Affiliation(s)
- Chanhung Z Lee
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, USA
| | | | | | | | | | | |
Collapse
|
31
|
Kamiguti AS, Lee ES, Till KJ, Harris RJ, Glenn MA, Lin K, Chen HJ, Zuzel M, Cawley JC. The role of matrix metalloproteinase 9 in the pathogenesis of chronic lymphocytic leukaemia. Br J Haematol 2004; 125:128-40. [PMID: 15059134 DOI: 10.1111/j.1365-2141.2004.04877.x] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Matrix metalloproteinases (MMPs) are important for the pathogenesis and progression of different tumours. MMPs-2 and -9 are the principal MMPs produced by lymphocytes; these enzymes can degrade a number of matrix proteins but are the two main MMPs that digest type IV collagen, the major component of basement membranes. Therefore, these enzymes are potentially important for tissue invasion and remodelling by malignant lymphocytes. This study showed that chronic lymphocytic leukaemia (CLL) cells produce and secrete variable amounts of pro-MMP-9, but no MMP-2 or tissue inhibitor of metalloproteinase 1 (TIMP-1). The pro-enzyme was found in monomeric and dimeric forms and also complexed with lipocalin. Moreover, a small fraction of secreted monomer became associated with the cell surface and activated upon cell adhesion to insolubilized type IV collagen. High levels of intracellular MMP-9 were associated with advanced (stage C) disease and with poor patient survival. Immunohistochemical studies demonstrated that MMP-9 was associated with areas of tissue invasion and remodelling. The relatively specific MMP-9 inhibitors, Ro31-9790 (3 micromol/l) and TIMP-1, reduced CLL-cell migration through type IV collagen and through endothelial monolayers suggesting that the enzyme may also be important in malignant cell entry and egress to and from involved tissue. Our data raise the possibility that MMP-9 modulation may have therapeutic potential in advanced CLL.
Collapse
Affiliation(s)
- Aura S Kamiguti
- Department of Haematology, Royal Liverpool Hospital, University of Liverpool, Duncan Building 3rd Floor, Daulby Street, Liverpool L69 3GA, UK.
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Bonfil RD, Osenkowski P, Fridman R, Cher ML. Matrix metalloproteinaes and bone metastasis. Cancer Treat Res 2004; 118:173-95. [PMID: 15043193 DOI: 10.1007/978-1-4419-9129-4_8] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Affiliation(s)
- R Daniel Bonfil
- Department of Urology, Wayne State University School of Medicine, Barbara Ann Karmanos Cancer Institute, Detroit, MI 48202, USA
| | | | | | | |
Collapse
|
33
|
Abstract
The early detection of both primary tumors and metastatic disease continue to be significant challenges in the diagnosis and staging of cancer. The growing recognition of the role of proteinases and proteolytic cascades in both the growth and metastasis of tumors has led to the development not only of therapeutic strategies using proteinase inhibitors, but also of methods to detect and image tumors in vivo via tumor-associated proteolytic activities. These imaging strategies derive from the enhanced sensitivity afforded by amplification that can be obtained by enzymatic processing to increase the efficacy of imaging "contrast agents" coupled with the inherent substrate specificity and selectivity of proteinases. This review describes key proteinases important in cancer progression, the strategies that have been devised to detect and image proteolytic activity in vivo, and the potential for this kind of functional imaging to serve as a marker for targeted therapy. The intent is to draw attention to the developing methods of molecular imaging to facilitate not only cancer diagnosis, but also for devising strategies for individualized targeted therapy and non-invasive monitoring of therapeutic efficacy.
Collapse
Affiliation(s)
- J Oliver McIntyre
- Department of Cancer Biology, Vanderbilt University, Nashville, Tennessee 37232-6840, USA.
| | | |
Collapse
|
34
|
Abstract
Solid tumors co-opt the body's endogenous extracellular proteolytic machinery for their invasion and metastasis. This is supported by a large number of independent observations ranging from histochemical and prognostic studies of cancer patient material to animal experiments. There are several extracellular proteolytic systems that are relevant in the context of cancer, but the plasminogen activation (PA) system and the matrix metalloproteases (MMPs) remain the most thoroughly investigated. Localization studies by immunohistochemistry and in situ mRNA hybridization in tumors of common human cancers have repeatedly identified members of the PA and MMP systems in stromal cells. The cancer cells, of epithelial origin, contribute PA and MMP components in some cases, but their contribution fades in comparison with the overwhelming expression of proteolytic components by fibroblasts, macrophages, endothelial cells, and other stromal cells. Ideal animal models of human cancers should recapitulate this fundamental proteolytic aspect of tumor biology. However, in the transplantable tumor models where PA or MMP components have been studied at the cellular level in vivo, this is most often not the case. Transgenic cancer models may provide a closer parallel to the human situation, in that PA and MMP components are synthesized by the tumor stroma. The pivotal role of stromal cells has been confirmed experimentally in mouse models in which the expression pattern of proteolytic components is strongly reminiscent of human tumors. In these models it is possible to reconstitute the wild-type tumor characteristics of proteolytically deficient tumor-bearing mice by transplantation with wild-type fibroblasts or hemapoietic cells. These studies collectively show that cancer-associated proteolysis is a collaborative effort of malignant cancer cells and various stromal cells--a collaboration in which stromal cells contribute the majority of the active proteolytic components that are necessary for the invasive behavior of the tumors. This cellular division of labor positions the stromal cells as prime targets for future research and possibly therapy. Vascular endothelial cells are already the focus of intense therapeutically relevant research, but tumor-associated fibroblasts, macrophages, neutrophils, lymphendothelial cells, etc. provide additional largely unexplored territory in the ongoing search for efficient countermeasures against invasive cancer.
Collapse
Affiliation(s)
- Kasper Almholt
- The Finsen Laboratory, Rigshospitalet, Strandboulevarden 49, 2100 Copenhagen, Denmark
| | | |
Collapse
|
35
|
Bergers G, Hanahan D. Combining antiangiogenic agents with metronomic chemotherapy enhances efficacy against late-stage pancreatic islet carcinomas in mice. COLD SPRING HARBOR SYMPOSIA ON QUANTITATIVE BIOLOGY 2003; 67:293-300. [PMID: 12858552 DOI: 10.1101/sqb.2002.67.293] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Affiliation(s)
- G Bergers
- Department of Neurological Surgery, Cancer Centers, University of California, San Francisco, California 94143, USA
| | | |
Collapse
|
36
|
Fingleton B. Matrix metalloproteinase inhibitors for cancer therapy:the current situation and future prospects. Expert Opin Ther Targets 2003; 7:385-97. [PMID: 12783574 DOI: 10.1517/14728222.7.3.385] [Citation(s) in RCA: 98] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Inhibition of matrix metalloproteinases (MMPs), a family of proteolytic enzymes linked to many aspects of cancer progression, has been explored as a therapeutic goal for almost two decades. Thus far, all tested MMP inhibitors (MMPIs) have failed to reach primary end points in Phase III clinical trials, although secondary analyses suggest benefits in particular patient groups. The clinical development of these agents has been hampered by problems related to determination of effective dosages and side effects that necessitate dose lowering or drug holidays. Imaging technologies offer hope as a means to measure enzyme activity and hence effective enzyme inhibition in vivo. Meanwhile, recent results from genetic studies of both mice and man have given some clues to possible causes of musculoskeletal side effects. Future progress in the therapeutic use of MMPIs is dependent on the ability to selectively target cancer-associated MMPs at the correct stage in tumour progression and the development of surrogate markers of in vivo efficacy.
Collapse
Affiliation(s)
- Barbara Fingleton
- Vanderbilt University Medical Center, Department of Cancer Biology, 736 PRB 23rd and Pierce Avenues, Nashville, TN 37232-6840, USA.
| |
Collapse
|
37
|
Hashimoto T, Wen G, Lawton MT, Boudreau NJ, Bollen AW, Yang GY, Barbaro NM, Higashida RT, Dowd CF, Halbach VV, Young WL. Abnormal expression of matrix metalloproteinases and tissue inhibitors of metalloproteinases in brain arteriovenous malformations. Stroke 2003; 34:925-31. [PMID: 12649522 DOI: 10.1161/01.str.0000061888.71524.df] [Citation(s) in RCA: 120] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
BACKGROUND AND PURPOSE Excessive degradation of the vascular matrix by matrix metalloproteinases (MMPs) can lead to structural instability of vessels. In this study we examined the expression of MMPs and tissue inhibitors of metalloproteinases (TIMPs) in brain arteriovenous malformations (BAVMs). METHODS We performed gelatin zymography for MMPs and Western blot for MMP-9, MMP-2, TIMP-1, TIMP-2, TIMP-3, and TIMP-4. MMP-9 expression was localized by immunohistochemistry. RESULTS We analyzed 37 BAVM specimens and 9 control brain specimens from epilepsy surgery. Thirty-two BAVM patients had embolization treatment before resection. Eighteen BAVM patients had a history of hemorrhage from BAVMs. Neither MMP-2 nor TIMP-2 was detected in BAVMs or control brain specimens. Compared with control brain samples, BAVM samples had higher levels of total MMP-9, active MMP-9, pro-MMP-9, TIMP-1, and TIMP-3. TIMP-4 levels were higher in the control brain than in BAVM specimens. MMP-9 was localized to the endothelial cell/peri-endothelial cell layer and infiltrating neutrophils of BAVMs. BAVMs with venous stenosis >or=50% had higher expression of MMP-9 than BAVMs with venous stenosis <50%. There was no apparent association between total MMP-9, pro-MMP-9, or active MMP-9 levels and (1) feeding artery pressure, (2) pattern of draining vein (exclusively deep venous drainage versus any superficial drainage), and (3) BAVM size. CONCLUSIONS We found increased levels of MMP-9 and TIMPs in BAVMs. Abnormal balance of MMP-9 and TIMPs may contribute to vascular instability of BAVMs.
Collapse
Affiliation(s)
- Tomoki Hashimoto
- Department of Anesthesia and Perioperative Care, University of California, San Francisco 94110, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Nelson FC, Delos Santos E, Levin JI, Chen JM, Skotnicki JS, DiJoseph JF, Sharr MA, Sung A, Killar LM, Cowling R, Jin G, Roth CE, Albright JD. Benzodiazepine inhibitors of the MMPs and TACE. Bioorg Med Chem Lett 2003; 12:2867-70. [PMID: 12270165 DOI: 10.1016/s0960-894x(02)00633-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
A series of benzodiazepine inhibitors of the MMPs and TACE has been developed. These compounds display an interesting selectivity profile and should be useful tools for exploring the biological relevance of such selectivity.
Collapse
Affiliation(s)
- Frances C Nelson
- Wyeth Research, 401N. Middletown Road, Pearl River, NY 10965, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Spurbeck WW, Ng CYC, Vanin EF, Davidoff AM. Retroviral vector-producer cell-mediated in vivo gene transfer of TIMP-3 restricts angiogenesis and neuroblastoma growth in mice. Cancer Gene Ther 2003; 10:161-7. [PMID: 12637936 DOI: 10.1038/sj.cgt.7700577] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Destruction and remodeling of the extracellular matrix occurs during the formation of new blood vessels that are required for tumor growth. We sought to determine whether gene-therapy mediated in vivo delivery of tissue inhibitor of matrix metalloproteinase-3 (TIMP-3), using retroviral vector-producer cells, could suppress angiogenesis and subsequent tumor growth in a murine neuroblastoma model. Tumor volume 28 days after coinjection of tumor cells with producer cells generating TIMP-3-encoding retroviral vectors was 21% that of controls, as was the mean tumor vascular index, a measure of blood vessel maturity. When tumors were allowed to reach a mean volume of 0.05 cm(3) before treatment, their size 2 weeks later was 47% relative to controls; larger tumors were not significantly affected. When producer cells were injected at surgical sites following excision of subcutaneous tumors, local recurrence 14 days later was only 22% in TIMP-3 producer cell treated mice as compared to 71% in controls. Unsuccessful transduction of melanoma cells in situ, another tumor of neural crest origin, resulted in unimpaired tumor growth, despite the fact that these tumors are susceptible to TIMP-3 overexpression, demonstrating the importance of tumor cell transduction in this approach. Thus, retroviral vector-producer cell-mediated in vivo gene transfer of TIMP-3 to tumor cells can significantly restrict tumor-induced angiogenesis and tumor growth. This approach may be an effective adjuvant in the treatment of neuroblastoma and other solid tumors refractory to traditional therapy, although it appears to be most effective in smaller tumors or in the setting of minimal residual disease, and the tumor cells must be susceptible to retroviral vector-mediated transduction.
Collapse
Affiliation(s)
- William W Spurbeck
- Department of Surgery, St Jude Children's Research Hospital, and University of Tennessee College of Medicine, Memphis, 38105, USA
| | | | | | | |
Collapse
|
40
|
Erickson AC, Barcellos-Hoff MH. The not-so innocent bystander: the microenvironment as a therapeutic target in cancer. Expert Opin Ther Targets 2003; 7:71-88. [PMID: 12556204 DOI: 10.1517/14728222.7.1.71] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The microenvironment in which cancer arises is often regarded as a bystander to the clonal expansion and acquisition of malignant characteristics of the tumour. However, a major function of the microenvironment is to suppress cancer, and its disruption is required for the establishment of cancer. In addition, tumour cells can further distort the microenvironment to promote growth, recruit non-malignant cells that provide physiological resources, and facilitate invasion. In this review, the authors discuss the contribution of the microenvironment, i.e., the stroma and its resident vasculature, inflammatory cells, growth factors and the extracellular matrix (ECM), in the development of cancer, and focus on two components as potential therapeutic targets in breast cancer. First, the ECM, which imparts crucial signalling via integrins and other receptors, is a first-line barrier to invasion, modulates aggressive behaviour and may be manipulated to provide novel impediments to tumour growth. Second, the authors discuss the involvement of TGF-beta1 as an example of one of many growth factors that can regulate ECM composition and degradation and that play complex roles in cancer. Compared to the variable routes taken by cells to become cancers, the response of tissues to cancer is relatively consistent. Therefore, controlling and eliminating cancer may be more readily achieved indirectly via the tissue microenvironment.
Collapse
Affiliation(s)
- Anna C Erickson
- Life Sciences Division, Building 74-174, 1 Cyclotron Road, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | | |
Collapse
|
41
|
Hengge UR, Ruzicka T, Tyring SK, Stuschke M, Roggendorf M, Schwartz RA, Seeber S. Update on Kaposi's sarcoma and other HHV8 associated diseases. Part 1: epidemiology, environmental predispositions, clinical manifestations, and therapy. THE LANCET. INFECTIOUS DISEASES 2002; 2:281-92. [PMID: 12062994 DOI: 10.1016/s1473-3099(02)00263-3] [Citation(s) in RCA: 195] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Kaposi's sarcoma (KS) is a mesenchymal tumour involving blood and lymphatic vessels. Only recently has the pathogenesis of this extraordinary neoplasm been elucidated. Viral oncogenesis and cytokine-induced growth together with some state of immunocompromise represent important conditions for this tumour to develop. In 1994, a novel virus was discovered and termed human herpesvirus 8 (HHV8), also known as Kaposi's sarcoma-associated herpes virus, which can be found in all types of KS, whether related to HIV or not. In the era of highly active antiretroviral therapy (HAART), the incidence of AIDS-KS has considerably declined, probably due to enhanced immune reconstitution and anti-HHV8-specific immune responses. If HAART is able to prevent spreading of KS, local therapy of KS may become an essential component of patient management. Part 1 of the review covers the epidemiology, environmental predispositions, clinical manifestations, and therapy of KS. Newer treatments such as pegylated liposomal anthracyclines and experimental strategies are discussed. We also present rationales and graduated treatment algorithms for local and systemic therapy in patients with KS to appropriately meet the challenges of this extraordinary neoplasm. Part 2, to be published next month, will summarise recent insights in the pathogenesis of KS and will discuss other HHV8-related diseases such as Castleman's disease and primary effusion lymphoma.
Collapse
Affiliation(s)
- Ulrich R Hengge
- Department of Dermatology, Venereology and Allergology, University of Essen, Germany.
| | | | | | | | | | | | | |
Collapse
|
42
|
Coussens LM, Fingleton B, Matrisian LM. Matrix metalloproteinase inhibitors and cancer: trials and tribulations. Science 2002; 295:2387-92. [PMID: 11923519 DOI: 10.1126/science.1067100] [Citation(s) in RCA: 2073] [Impact Index Per Article: 90.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
For at least 30 years, matrix metalloproteinases (MMPs) have been heralded as promising targets for cancer therapy on the basis of their massive up-regulation in malignant tissues and their unique ability to degrade all components of the extracellular matrix. Preclinical studies testing the efficacy of MMP suppression in tumor models were so compelling that synthetic metalloproteinase inhibitors (MPIs) were rapidly developed and routed into human clinical trials. The results of these trials have been disappointing. Here we review the studies that brought MPIs into clinical testing and discuss the design and outcome of the trials in light of new information about the cellular source, substrates, and mode of action of MMPs at different stages of tumor progression. The important lessons learned from the MPI experience may be of great value for future studies of MPIs and for cancer drug development in general.
Collapse
Affiliation(s)
- Lisa M Coussens
- Department of Pathology and Cancer Research Institute, University of California, 2340 Sutter Street, San Francisco, CA 94143, USA
| | | | | |
Collapse
|
43
|
Brinckerhoff CE, Matrisian LM. Matrix metalloproteinases: a tail of a frog that became a prince. Nat Rev Mol Cell Biol 2002; 3:207-14. [PMID: 11994741 DOI: 10.1038/nrm763] [Citation(s) in RCA: 831] [Impact Index Per Article: 36.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
It is 40 years since the first member of what came to be known as the matrix metalloproteinase (MMP) family was described. Structural, molecular and biochemical approaches have subsequently contributed to piecing together the puzzle of how MMPs work, and how they contribute to various disease processes.
Collapse
|
44
|
Chang C, Werb Z. The many faces of metalloproteases: cell growth, invasion, angiogenesis and metastasis. Trends Cell Biol 2002. [PMID: 11684441 DOI: 10.1016/s0962-8924(01)82222-4] [Citation(s) in RCA: 219] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Metalloproteases are important in many aspects of biology, ranging from cell proliferation, differentiation and remodeling of the extracellular matrix (ECM) to vascularization and cell migration. These events occur several times during organogenesis in both normal development and during tumor progression. Mechanisms of metalloprotease action underlying these events include the proteolytic cleavage of growth factors so that they can become available to cells not in direct physical contact, degradation of the ECM so that founder cells can move across tissues into nearby stroma, and regulated receptor cleavage to terminate migratory signaling. Most of these processes require a delicate balance between the functions of matrix metalloproteases (MMPs) or metalloprotease-disintegrins (ADAMs) and natural tissue inhibitors of metalloproteases (TIMPs). In this review, we discuss recent progress in identifying an essential role for metalloproteases in axon outgrowth, as an example of a focal invasive event. We also discuss the evolving concept of how MMPs might regulate stem cell fate during tumor development.
Collapse
Affiliation(s)
- C Chang
- Department of Anatomy, Box 0452, University of California San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143-0452, USA
| | | |
Collapse
|
45
|
Ruangpanit N, Price JT, Holmbeck K, Birkedal-Hansen H, Guenzler V, Huang X, Chan D, Bateman JF, Thompson EW. MT1-MMP-dependent and -independent regulation of gelatinase A activation in long-term, ascorbate-treated fibroblast cultures: regulation by fibrillar collagen. Exp Cell Res 2002; 272:109-18. [PMID: 11777335 DOI: 10.1006/excr.2001.5403] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Human skin fibroblasts were cultured long-term in the presence of ascorbic acid to allow formation of a three-dimensional collagen matrix, and the effects of this on activation of secreted matrix metalloproteinase-2 (MMP-2) were examined. Accumulation of collagen over time correlated with increased levels of both mature MMP-2 and cell-associated membrane type 1-MMP (MT1-MMP), and subsequently increased mRNA levels for MT1-MMP, providing temporal resolution of the "nontranscriptional" and "transcriptional" effects of collagen on MT-1MMP functionality. MMP-2 activation by these cultures was blocked by inhibitors of prolyl-4-hydroxylase, or when fibroblasts derived from the collagen alpha1(I) gene-deficient Mov-13 mouse were used. MMP-2 activation by the Mov-13 fibroblasts was rescued by transfection of a full-length alpha1(I) collagen cDNA, and to our surprise, also by transfection with an alpha1(I) collagen cDNA carrying a mutation at the C-proteinase cleavage, which almost abrogated fibrillogenesis. Although studies with ascorbate-cultured MT1-MMP-/- fibroblasts showed that MT1-MMP played a significant role in the collagen-induced MMP-2 activation, a residual MT1-MMP-independent activation of MMP-2 was seen which resembled the level of MMP-2 activation persisting when wild-type fibroblasts were cultured in the presence of both ascorbic acid and MMP inhibitors. We were also unable to block this residual activation with inhibitors specific for serinyl, aspartyl, or cysteinyl enzymes.
Collapse
Affiliation(s)
- Neeracha Ruangpanit
- VBCRC Breast Cancer Invasion and Metastasis Unit, St. Vincent's Institute of Medical Research, Melbourne, Australia
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Fingleton B, Vargo-Gogola T, Crawford HC, Matrisian LM. Matrilysin [MMP-7] expression selects for cells with reduced sensitivity to apoptosis. Neoplasia 2001; 3:459-68. [PMID: 11774028 PMCID: PMC1506562 DOI: 10.1038/sj.neo.7900190] [Citation(s) in RCA: 131] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2001] [Accepted: 08/01/2001] [Indexed: 11/09/2022] Open
Abstract
The matrix metalloproteinase matrilysin (MMP-7) has been demonstrated to contribute to tumor development. We have shown previously that members of the TNF family of apoptosis-inducing proteins are substrates for this enzyme, resulting in increased death pathway signaling. The goal of the current study was to reconcile the proapoptotic and tumor-promoting functions of matrilysin. In the human HBL100 and murine NMuMG cell lines that represent early stages of tumor progression and that express both Fas ligand and its receptor, exposure to matrilysin results in cell death that can be blocked by FasL neutralizing antibodies. Constitutive expression of matrilysin in these cell lines selects for cells with reduced sensitivity to Fas-mediated apoptosis as demonstrated both with a receptor-activating antibody and with in vitro activated splenocytes. Matrilysin-expressing cells are also significantly less sensitive to chemical inducers of apoptosis. We propose that the expression of matrilysin that has been reported at early stages in various tumor types can act to select cells with a significantly decreased chance of removal due to immune surveillance. As a result, these cells are more likely to acquire additional genetic modifications and develop further as tumors.
Collapse
Affiliation(s)
- B Fingleton
- Department of Cancer Biology, Vanderbilt University School of Medicine, PRB 23rd and Pierce, Nashville, TN 37232-6840, USA.
| | | | | | | |
Collapse
|
47
|
Barcellos-Hoff MH, Brooks AL. Extracellular signaling through the microenvironment: a hypothesis relating carcinogenesis, bystander effects, and genomic instability. Radiat Res 2001; 156:618-27. [PMID: 11604083 DOI: 10.1667/0033-7587(2001)156[0618:esttma]2.0.co;2] [Citation(s) in RCA: 204] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Cell growth, differentiation and death are directed in large part by extracellular signaling through the interactions of cells with other cells and with the extracellular matrix; these interactions are in turn modulated by cytokines and growth factors, i.e. the microenvironment. Here we discuss the idea that extracellular signaling integrates multicellular damage responses that are important deterrents to the development of cancer through mechanisms that eliminate abnormal cells and inhibit neoplastic behavior. As an example, we discuss the action of transforming growth factor beta (TGFB1) as an extracellular sensor of damage. We propose that radiation-induced bystander effects and genomic instability are, respectively, positive and negative manifestations of this homeostatic process. Bystander effects exhibited predominantly after a low-dose or a nonhomogeneous radiation exposure are extracellular signaling pathways that modulate cellular repair and death programs. Persistent disruption of extracellular signaling after exposure to relatively high doses of ionizing radiation may lead to the accumulation of aberrant cells that are genomically unstable. Understanding radiation effects in terms of coordinated multicellular responses that affect decisions regarding the fate of a cell may necessitate re-evaluation of radiation dose and risk concepts and provide avenues for intervention.
Collapse
Affiliation(s)
- M H Barcellos-Hoff
- Life Sciences Division, Lawrence Berkeley National Laboratory, University of California, Berkeley, California 94720, USA.
| | | |
Collapse
|
48
|
Abstract
Metalloproteases are important in many aspects of biology, ranging from cell proliferation, differentiation and remodeling of the extracellular matrix (ECM) to vascularization and cell migration. These events occur several times during organogenesis in both normal development and during tumor progression. Mechanisms of metalloprotease action underlying these events include the proteolytic cleavage of growth factors so that they can become available to cells not in direct physical contact, degradation of the ECM so that founder cells can move across tissues into nearby stroma, and regulated receptor cleavage to terminate migratory signaling. Most of these processes require a delicate balance between the functions of matrix metalloproteases (MMPs) or metalloprotease-disintegrins (ADAMs) and natural tissue inhibitors of metalloproteases (TIMPs). In this review, we discuss recent progress in identifying an essential role for metalloproteases in axon outgrowth, as an example of a focal invasive event. We also discuss the evolving concept of how MMPs might regulate stem cell fate during tumor development.
Collapse
|
49
|
Abstract
The matrix metalloproteinases (MMPs) have been viewed as bulldozers, destroying the extracellular matrix to permit normal remodeling and contribute to pathological tissue destruction and tumor cell invasion. More recently, the identification of specific matrix and non-matrix substrates for MMPs and the elucidation of the biological consequence of cleavage indicates that perhaps MMPs should be viewed more as pruning shears, playing sophisticated roles in modulating normal cellular behavior, cell-cell communication and tumor progression.
Collapse
Affiliation(s)
- L J McCawley
- Department of Cancer Biology, Vanderbilt University Medical Center, 771 PRB, 23rd and Pierce, Nashville, Tennessee 37232-6840, USA
| | | |
Collapse
|
50
|
Abstract
Acquired immune deficiency syndrome-associated Kaposi sarcoma is a progressive and occasionally fatal condition. The strong angiogenic component of this disease makes it particularly suitable for treatment with the emerging class of drugs that act as antiangiogenic agents. Matrix metalloproteinases have been shown to play prominent roles in the angiogenic process, and small molecule inhibitors of these enzymes are currently being tested as antiangiogenic agents in various malignancies. Given that matrix metalloproteinases contribute to multiple steps of the angiogenic process, inhibitors of these enzymes, either alone or in combination with other agents, may represent a particularly effective therapeutic approach for Kaposi sarcoma.
Collapse
Affiliation(s)
- B Fingleton
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | | |
Collapse
|