51
|
Fujita S, Sumi M, Tatsukawa E, Nagano K, Katase N. Expressions of extracellular matrix-remodeling factors in lymph nodes from oral cancer patients. Oral Dis 2020; 26:1424-1431. [PMID: 32419185 DOI: 10.1111/odi.13419] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 04/02/2020] [Accepted: 04/26/2020] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Most malignant tumors require remodeling extracellular matrices (ECMs) for invasive growth and metastasis. Cancer cells and stromal cells remodel ECM. We investigated the relationship between regional lymph node (LN) metastasis and expression of ECM-remodeling factors in oral squamous cell carcinoma (OSCC). METHODS Using primary OSCC and cervical LNs obtained surgically, we performed immunohistochemical evaluation of the ECM-remodeling factors, lysyl oxidase (LOX), MT1-MMP, S100A8, and TIMP-1 in primary tumor and marginal sinus histiocytosis (MSH) in LNs, and determined the statistical significance of the positive rates between metastatic and metastasis-free groups. RESULTS Marginal sinus histiocytosis was more frequently formed in the metastatic group compared to the metastasis-free group. Lymphatic metastasis correlated with the immunopositivity rates of tumor cells expressing LOX, MT1-MMP, and TIMP-1, and of stromal cells expressing TIMP-1. The case rates of MSH containing macrophages positive for LOX and MT1-MMP in the metastasis group were significantly higher than in the metastasis-free group. ECM-remodeling-associated macrophages accumulate in marginal sinus in conjunction with lymphatic metastasis. CONCLUSION Expression of LOX, MT1-MMP, and TIMP-1 in the parenchyma, and stromal expression of TIMP-1 in primary tumor may predict lymphatic metastasis. LOX and MT1-MMP have a possibility to participate in formation of pre-metastatic niche in LNs.
Collapse
Affiliation(s)
- Shuichi Fujita
- Department of Oral Pathology, Medical and Dental Sciences, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Misa Sumi
- Department of Radiology and Cancer Biology, Medical and Dental Sciences, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Eri Tatsukawa
- Department of Oral Pathology, Medical and Dental Sciences, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Kenichi Nagano
- Department of Oral Pathology, Medical and Dental Sciences, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Naoki Katase
- Department of Oral Pathology, Medical and Dental Sciences, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| |
Collapse
|
52
|
Raeeszadeh-Sarmazdeh M, Do LD, Hritz BG. Metalloproteinases and Their Inhibitors: Potential for the Development of New Therapeutics. Cells 2020; 9:E1313. [PMID: 32466129 PMCID: PMC7290391 DOI: 10.3390/cells9051313] [Citation(s) in RCA: 168] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 05/18/2020] [Accepted: 05/19/2020] [Indexed: 02/06/2023] Open
Abstract
The metalloproteinase (MP) family of zinc-dependent proteases, including matrix metalloproteinases (MMPs), a disintegrin and metalloproteases (ADAMs), and a disintegrin and metalloproteinase with thrombospondin motifs (ADAMTSs) plays a crucial role in the extracellular matrix (ECM) remodeling and degradation activities. A wide range of substrates of the MP family includes ECM components, chemokines, cell receptors, and growth factors. Metalloproteinases activities are tightly regulated by proteolytic activation and inhibition via their natural inhibitors, tissue inhibitors of metalloproteinases (TIMPs), and the imbalance of the activation and inhibition is responsible in progression or inhibition of several diseases, e.g., cancer, neurological disorders, and cardiovascular diseases. We provide an overview of the structure, function, and the multifaceted role of MMPs, ADAMs, and TIMPs in several diseases via their cellular functions such as proteolysis of other cell signaling factors, degradation and remodeling of the ECM, and other essential protease-independent interactions in the ECM. The significance of MP inhibitors targeting specific MMP or ADAMs with high selectivity is also discussed. Recent advances and techniques used in developing novel MP inhibitors and MP responsive drug delivery tools are also reviewed.
Collapse
Affiliation(s)
- Maryam Raeeszadeh-Sarmazdeh
- Chemical and Materials Engineering Department, University of Nevada, Reno, NV 89557, USA; (L.D.D.); (B.G.H.)
| | | | | |
Collapse
|
53
|
Abreu M, Cabezas-Sainz P, Alonso-Alconada L, Ferreirós A, Mondelo-Macía P, Lago-Lestón RM, Abalo A, Díaz E, Palacios-Zambrano S, Rojo-Sebastian A, López-López R, Sánchez L, Moreno-Bueno G, Muinelo-Romay L. Circulating Tumor Cells Characterization Revealed TIMP1 as a Potential Therapeutic Target in Ovarian Cancer. Cells 2020; 9:cells9051218. [PMID: 32423054 PMCID: PMC7291036 DOI: 10.3390/cells9051218] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Accepted: 05/11/2020] [Indexed: 12/19/2022] Open
Abstract
Background: Recent studies showed a relevant role of hematogenous spread in ovarian cancer and the interest of circulating tumor cells (CTCs) monitoring as a prognosis marker. The aim of the present study was the characterization of CTCs from ovarian cancer patients, paying special attention to cell plasticity characteristics to better understand the biology of these cells. Methods: CTCs isolation was carried out in 38 patients with advanced high-grade serous ovarian cancer using in parallel CellSearch and an alternative EpCAM-based immunoisolation followed by RT-qPCR analysis to characterize these cells. Results: Epithelial CTCs were found in 21% of patients, being their presence higher in patients with extraperitoneal metastasis. Importantly, this population was characterized by the expression of epithelial markers as MUC1 and CK19, but also by genes associated with mesenchymal and more malignant features as TIMP1, CXCR4 and the stem markers CD24 and CD44. In addition, we evidenced the relevance of TIMP1 expression to promote tumor proliferation, suggesting its interest as a therapeutic target. Conclusions: Overall, we evidenced the utility of the molecular characterization of EpCAM+ CTCs from advanced ovarian cancer patients to identify biomarkers with potential applicability for disseminated disease detection and as therapeutic targets such as TIMP1.
Collapse
Affiliation(s)
- Manuel Abreu
- Liquid Biopsy Analysis Unit, Translational Medical Oncology (Oncomet), Health Research Institute of Santiago (IDIS), 15706 Santiago de Compostela, Spain; (M.A.); (P.M.-M.); (R.M.L.-L.); (A.A.)
| | - Pablo Cabezas-Sainz
- Department of Genetics, University of Santiago de Compostela, Campus de Lugo, 27002 Lugo, Spain; (P.C.-S.); (L.S.)
| | | | - Alba Ferreirós
- Nasasbiotech, S.L., Canton Grande 3, 15003 A Coruña, Spain; (L.A.-A.); (A.F.)
| | - Patricia Mondelo-Macía
- Liquid Biopsy Analysis Unit, Translational Medical Oncology (Oncomet), Health Research Institute of Santiago (IDIS), 15706 Santiago de Compostela, Spain; (M.A.); (P.M.-M.); (R.M.L.-L.); (A.A.)
| | - Ramón Manuel Lago-Lestón
- Liquid Biopsy Analysis Unit, Translational Medical Oncology (Oncomet), Health Research Institute of Santiago (IDIS), 15706 Santiago de Compostela, Spain; (M.A.); (P.M.-M.); (R.M.L.-L.); (A.A.)
| | - Alicia Abalo
- Liquid Biopsy Analysis Unit, Translational Medical Oncology (Oncomet), Health Research Institute of Santiago (IDIS), 15706 Santiago de Compostela, Spain; (M.A.); (P.M.-M.); (R.M.L.-L.); (A.A.)
| | - Eva Díaz
- Fundación MD Anderson Internacional, C/Gómez Hemans 2, 28033 Madrid, Spain; (E.D.); (A.R.-S.)
| | | | | | - Rafael López-López
- Translational Medical Oncology (Oncomet), Health Research Institute of Santiago de Compostela (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela (SERGAS), 15706 Santiago de Compostela, Spain;
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Monforte de Lemos 3-5, 28029 Madrid, Spain
| | - Laura Sánchez
- Department of Genetics, University of Santiago de Compostela, Campus de Lugo, 27002 Lugo, Spain; (P.C.-S.); (L.S.)
| | - Gema Moreno-Bueno
- Fundación MD Anderson Internacional, C/Gómez Hemans 2, 28033 Madrid, Spain; (E.D.); (A.R.-S.)
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Monforte de Lemos 3-5, 28029 Madrid, Spain
- Departamento de Bioquímica, Universidad Autónoma de Madrid (UAM), Instituto de Investigaciones Biomédicas ‘Alberto Sols’ (CSIC-UAM), IdiPaz, Arzobispo Morcillo 4, 28029 Madrid, Spain
- Correspondence: (G.M.-B.); (L.M.-R.); Tel.: +34-91-787-86-00 (G.M.-B.); +34-981955073 (L.M.-R.)
| | - Laura Muinelo-Romay
- Liquid Biopsy Analysis Unit, Translational Medical Oncology (Oncomet), Health Research Institute of Santiago (IDIS), 15706 Santiago de Compostela, Spain; (M.A.); (P.M.-M.); (R.M.L.-L.); (A.A.)
- Departamento de Bioquímica, Universidad Autónoma de Madrid (UAM), Instituto de Investigaciones Biomédicas ‘Alberto Sols’ (CSIC-UAM), IdiPaz, Arzobispo Morcillo 4, 28029 Madrid, Spain
- Correspondence: (G.M.-B.); (L.M.-R.); Tel.: +34-91-787-86-00 (G.M.-B.); +34-981955073 (L.M.-R.)
| |
Collapse
|
54
|
Huang B, Huang M, Li Q. Cancer-Associated Fibroblasts Promote Angiogenesis of Hepatocellular Carcinoma by VEGF-Mediated EZH2/VASH1 Pathway. Technol Cancer Res Treat 2020; 18:1533033819879905. [PMID: 31757187 PMCID: PMC6876164 DOI: 10.1177/1533033819879905] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background: Hepatocellular carcinoma is a highly vascularized tumor, so it is critical to study its angiogenesis. Cancer-associated fibroblasts and enhancer of zeste homolog 2 play an important role in tumor angiogenesis and became significant hallmarks of cancer. But the relationship between enhancer of zeste homolog-2 and cancer-associated fibroblasts in response to angiogenesis and its precise mechanism remains unclear. Methods: The separation of cancer-associated fibroblasts was identified by immunofluorescence. 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide analysis was used to reveal the proliferation of human umbilical vein endothelial cells. Vascular endothelial growth factor level was quantified by enzyme-linked immunosorbent assay. The wound healing, transwell, and vascular tube formation assays were used to identify the capability of migration, invasion, and tube formation of human umbilical vein endothelial cells in vitro. The detection of tumor angiogenesis was also performed in vivo. Finally, the level of enhancer of zeste homolog-2 and vasohibin 1 were determined by quantitative real-time polymerase chain reaction and Western blotting. Results: In comparison to control and condition medium noncancerous fibroblasts groups, the condition medium cancer-associated fibroblasts could significantly promote the proliferation, migration, invasion, and angiogenesis of human umbilical vein endothelial cells. We found that cancer-associated fibroblasts promoted angiogenesis of human umbilical vein endothelial cells via vascular endothelial growth factor secretion in vitro and in vivo. The upregulation of enhancer of zeste homolog 2 by vascular endothelial growth factor inhibited the expression of vasohibin 1, thus promoting the proliferation and angiogenesis of human umbilical vein endothelial cells. Taken together, the cancer-associated fibroblasts of hepatocellular carcinoma regulate the enhancer of zeste homolog-2/vasohibin 1 pathway via vascular endothelial growth factor secretion, thereby promoting the proliferation and angiogenesis of human umbilical vein endothelial cells. Conclusion: Our study identified the relationship between cancer-associated fibroblasts and enhancer of zeste homolog-2 and confirmed the pivotal role of cancer-associated fibroblasts in angiogenesis of hepatocellular carcinoma. Cancer-associated fibroblasts promote angiogenesis of hepatocellular carcinoma by vascular endothelial growth factor–mediated enhancer of zeste homolog-2/vasohibin 1 pathway and may be a potentially useful therapeutic target for hepatocellular carcinoma.
Collapse
Affiliation(s)
- Bin Huang
- Department of Intervention, Hunan Provincial Cancer Hospital, Changsha, People's Republic of China
| | - Manping Huang
- Department of Intervention, Hunan Provincial Cancer Hospital, Changsha, People's Republic of China
| | - Qin Li
- Department of Gynecology, Affiliated Hospital of Hunan Institute of Traditional Chinese Medicine, Changsha, People's Republic of China
| |
Collapse
|
55
|
Hashem S, Nisar S, Sageena G, Macha MA, Yadav SK, Krishnankutty R, Uddin S, Haris M, Bhat AA. Therapeutic Effects of Curcumol in Several Diseases; An Overview. Nutr Cancer 2020; 73:181-195. [PMID: 32285707 DOI: 10.1080/01635581.2020.1749676] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Sheema Hashem
- Translational Medicine, Research Branch, Sidra Medical and Research Center, Doha, Qatar
| | - Sabah Nisar
- Translational Medicine, Research Branch, Sidra Medical and Research Center, Doha, Qatar
| | | | - Muzafar A. Macha
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Department of Biotechnology, Central University of Kashmir, Ganderbal, India
| | - Santosh K. Yadav
- Translational Medicine, Research Branch, Sidra Medical and Research Center, Doha, Qatar
| | - Roopesh Krishnankutty
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Mohammad Haris
- Translational Medicine, Research Branch, Sidra Medical and Research Center, Doha, Qatar
| | - Ajaz A. Bhat
- Translational Medicine, Research Branch, Sidra Medical and Research Center, Doha, Qatar
| |
Collapse
|
56
|
Matrix Effect in the Isolation of Breast Cancer-Derived Nanovesicles by Immunomagnetic Separation and Electrochemical Immunosensing-A Comparative Study. SENSORS 2020; 20:s20040965. [PMID: 32054015 PMCID: PMC7071381 DOI: 10.3390/s20040965] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Revised: 02/05/2020] [Accepted: 02/06/2020] [Indexed: 01/16/2023]
Abstract
Exosomes are cell-derived nanovesicles released into biological fluids, which are involved in cell-to-cell communication. The analysis of the content and the surface of the exosomes allow conclusions about the cells they are originating from and the underlying condition, pathology or disease. Therefore, the exosomes are currently considered good candidates as biomarkers to improve the current methods for clinical diagnosis, including cancer. However, due to their low concentration, conventional procedures for exosome detection including biosensing usually require relatively large sample volumes and involve preliminary purification and preconcentration steps by ultracentrifugation. In this paper, the immunomagnetic separation is presented as an alternative method for the specific isolation of exosomes in serum. To achieve that, a rational study of the surface proteins in exosomes, which can be recognized by magnetic particles, is presented. The characterization was performed in exosomes obtained from cell culture supernatants of MCF7, MDA-MB-231 and SKBR3 breast cancer cell lines, including TEM and nanoparticle tracking analysis (NTA). For the specific characterization by flow cytometry and confocal microscopy, different commercial antibodies against selected receptors were used, including the general tetraspanins CD9, CD63 and CD81, and cancer-related receptors (CD24, CD44, CD54, CD326 and CD340). The effect of the serum matrix on the immunomagnetic separation was then carefully evaluated by spiking the exosomes in depleted human serum. Based on this study, the exosomes were preconcentrated by immunomagnetic separation on antiCD81-modified magnetic particles in order to achieve further magnetic actuation on the surface of the electrode for the electrochemical readout. The performance of this approach is discussed and compared with classical characterization methods.
Collapse
|
57
|
Knight BE, Kozlowski N, Havelin J, King T, Crocker SJ, Young EE, Baumbauer KM. TIMP-1 Attenuates the Development of Inflammatory Pain Through MMP-Dependent and Receptor-Mediated Cell Signaling Mechanisms. Front Mol Neurosci 2019. [PMID: 31616247 DOI: 10.3389/fnmol.2019.00220/bibtex] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023] Open
Abstract
Unresolved inflammation is a significant predictor for developing chronic pain, and targeting the mechanisms underlying inflammation offers opportunities for therapeutic intervention. During inflammation, matrix metalloproteinase (MMP) activity contributes to tissue remodeling and inflammatory signaling, and is regulated by tissue inhibitors of metalloproteinases (TIMPs). TIMP-1 and -2 have known roles in pain, but only in the context of MMP inhibition. However, TIMP-1 also has receptor-mediated cell signaling functions that are not well understood. Here, we examined how TIMP-1-dependent cell signaling impacts inflammatory hypersensitivity and ongoing pain. We found that hindpaw injection of complete Freund's adjuvant (CFA) increased cutaneous TIMP-1 expression that peaked prior to development of mechanical hypersensitivity, suggesting that TIMP-1 inhibits the development of inflammatory hypersensitivity. To examine this possibility, we injected TIMP-1 knockout (T1KO) mice with CFA and found that T1KO mice exhibited rapid onset thermal and mechanical hypersensitivity at the site of inflammation that was absent or attenuated in WT controls. We also found that T1KO mice exhibited hypersensitivity in adjacent tissues innervated by different sets of afferents, as well as skin contralateral to the site of inflammation. Replacement of recombinant murine (rm)TIMP-1 alleviated hypersensitivity when administered at the site and time of inflammation. Administration of either the MMP inhibiting N-terminal or the cell signaling C-terminal domains recapitulated the antinociceptive effect of full-length rmTIMP-1, suggesting that rmTIMP-1inhibits hypersensitivity through MMP inhibition and receptor-mediated cell signaling. We also found that hypersensitivity was not due to genotype-specific differences in MMP-9 activity or expression, nor to differences in cytokine expression. Administration of rmTIMP-1 prevented mechanical hypersensitivity and ongoing pain in WT mice, collectively suggesting a novel role for TIMP-1 in the attenuation of inflammatory pain.
Collapse
Affiliation(s)
- Brittany E Knight
- Department of Neuroscience, UConn Health, Farmington, CT, United States
| | - Nathan Kozlowski
- School of Nursing, University of Connecticut, Storrs, CT, United States
| | - Joshua Havelin
- Center for Excellence in the Neurosciences, University of New England, Biddeford, ME, United States.,Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, United States
| | - Tamara King
- Center for Excellence in the Neurosciences, University of New England, Biddeford, ME, United States.,Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, United States.,College of Osteopathic Medicine, University of New England, Biddeford, ME, United States
| | - Stephen J Crocker
- Department of Neuroscience, UConn Health, Farmington, CT, United States.,Institute for Systems Genomics, UConn Health, Farmington, CT, United States
| | - Erin E Young
- School of Nursing, University of Connecticut, Storrs, CT, United States.,Institute for Systems Genomics, UConn Health, Farmington, CT, United States.,The Center for Advancement in Managing Pain, School of Nursing, University of Connecticut, Storrs, CT, United States.,Genetics and Genome Sciences, UConn Health, Farmington, CT, United States
| | - Kyle M Baumbauer
- School of Nursing, University of Connecticut, Storrs, CT, United States.,School of Nursing, University of Connecticut, Storrs, CT, United States.,School of Nursing, University of Connecticut, Storrs, CT, United States.,School of Nursing, University of Connecticut, Storrs, CT, United States.,School of Nursing, University of Connecticut, Storrs, CT, United States
| |
Collapse
|
58
|
Ahn CB, Lee JH, Han DG, Kang HW, Lee SH, Lee JI, Son KH, Lee JW. Simulated microgravity with floating environment promotes migration of non-small cell lung cancers. Sci Rep 2019; 9:14553. [PMID: 31601869 PMCID: PMC6787256 DOI: 10.1038/s41598-019-50736-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 09/06/2019] [Indexed: 11/09/2022] Open
Abstract
A migration of cancer is one of the most important factors affecting cancer therapy. Particularly, a cancer migration study in a microgravity environment has gained attention as a tool for developing cancer therapy. In this study, we evaluated the proliferation and migration of two types (adenocarcinoma A549, squamous cell carcinoma H1703) of non-small cell lung cancers (NSCLC) in a floating environment with microgravity. When we measured proliferation of two NSCLCs in the microgravity (MG) and ground-gravity (CONT), although initial cell adhesion in MG was low, a normalized proliferation rate of A549 in MG was higher than that in CONT. Wound healing results of A549 and H1703 showed rapid recovery in MG; particularly, the migration rate of A549 was faster than that of H1703 both the normal and low proliferating conditions. Gene expression results showed that the microgravity accelerated the migration of NSCLC. Both A549 and H1703 in MG highly expressed the migration-related genes MMP-2, MMP-9, TIMP-1, and TIMP-2 compared to CONT at 24 h. Furthermore, analysis of MMP-2 protein synthesis revealed weaker metastatic performance of H1703 than that of A549. Therefore, the simulated microgravity based cancer culture environment will be a potential for migration and metastasis studies of lung cancers.
Collapse
Affiliation(s)
- Chi Bum Ahn
- Department of Molecular Medicine, College of Medicine, Gachon University, Incheon, Republic of Korea
| | - Ji-Hyun Lee
- Department of Molecular Medicine, College of Medicine, Gachon University, Incheon, Republic of Korea
| | - Dae Geun Han
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, Republic of Korea
| | - Hyun-Wook Kang
- Department of Biomedical Engineering, School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Sung-Ho Lee
- Department of Thoracic and Cardiovascular Surgery, Korea University Medical College, Korea University, Seoul, Republic of Korea
| | - Jae-Ik Lee
- Department of Thoracic and Cardiovascular Surgery, Gachon University Gil Medical Center, School of Medicine, Gachon University, Incheon, Republic of Korea
| | - Kuk Hui Son
- Department of Thoracic and Cardiovascular Surgery, Gachon University Gil Medical Center, School of Medicine, Gachon University, Incheon, Republic of Korea.
| | - Jin Woo Lee
- Department of Molecular Medicine, College of Medicine, Gachon University, Incheon, Republic of Korea. .,Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, Republic of Korea.
| |
Collapse
|
59
|
Knight BE, Kozlowski N, Havelin J, King T, Crocker SJ, Young EE, Baumbauer KM. TIMP-1 Attenuates the Development of Inflammatory Pain Through MMP-Dependent and Receptor-Mediated Cell Signaling Mechanisms. Front Mol Neurosci 2019; 12:220. [PMID: 31616247 PMCID: PMC6764257 DOI: 10.3389/fnmol.2019.00220] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Accepted: 08/30/2019] [Indexed: 12/12/2022] Open
Abstract
Unresolved inflammation is a significant predictor for developing chronic pain, and targeting the mechanisms underlying inflammation offers opportunities for therapeutic intervention. During inflammation, matrix metalloproteinase (MMP) activity contributes to tissue remodeling and inflammatory signaling, and is regulated by tissue inhibitors of metalloproteinases (TIMPs). TIMP-1 and -2 have known roles in pain, but only in the context of MMP inhibition. However, TIMP-1 also has receptor-mediated cell signaling functions that are not well understood. Here, we examined how TIMP-1-dependent cell signaling impacts inflammatory hypersensitivity and ongoing pain. We found that hindpaw injection of complete Freund’s adjuvant (CFA) increased cutaneous TIMP-1 expression that peaked prior to development of mechanical hypersensitivity, suggesting that TIMP-1 inhibits the development of inflammatory hypersensitivity. To examine this possibility, we injected TIMP-1 knockout (T1KO) mice with CFA and found that T1KO mice exhibited rapid onset thermal and mechanical hypersensitivity at the site of inflammation that was absent or attenuated in WT controls. We also found that T1KO mice exhibited hypersensitivity in adjacent tissues innervated by different sets of afferents, as well as skin contralateral to the site of inflammation. Replacement of recombinant murine (rm)TIMP-1 alleviated hypersensitivity when administered at the site and time of inflammation. Administration of either the MMP inhibiting N-terminal or the cell signaling C-terminal domains recapitulated the antinociceptive effect of full-length rmTIMP-1, suggesting that rmTIMP-1inhibits hypersensitivity through MMP inhibition and receptor-mediated cell signaling. We also found that hypersensitivity was not due to genotype-specific differences in MMP-9 activity or expression, nor to differences in cytokine expression. Administration of rmTIMP-1 prevented mechanical hypersensitivity and ongoing pain in WT mice, collectively suggesting a novel role for TIMP-1 in the attenuation of inflammatory pain.
Collapse
Affiliation(s)
- Brittany E Knight
- Department of Neuroscience, UConn Health, Farmington, CT, United States
| | - Nathan Kozlowski
- School of Nursing, University of Connecticut, Storrs, CT, United States
| | - Joshua Havelin
- Center for Excellence in the Neurosciences, University of New England, Biddeford, ME, United States.,Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, United States
| | - Tamara King
- Center for Excellence in the Neurosciences, University of New England, Biddeford, ME, United States.,Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, United States.,College of Osteopathic Medicine, University of New England, Biddeford, ME, United States
| | - Stephen J Crocker
- Department of Neuroscience, UConn Health, Farmington, CT, United States.,Institute for Systems Genomics, UConn Health, Farmington, CT, United States
| | - Erin E Young
- School of Nursing, University of Connecticut, Storrs, CT, United States.,Institute for Systems Genomics, UConn Health, Farmington, CT, United States.,The Center for Advancement in Managing Pain, School of Nursing, University of Connecticut, Storrs, CT, United States.,Genetics and Genome Sciences, UConn Health, Farmington, CT, United States
| | - Kyle M Baumbauer
- School of Nursing, University of Connecticut, Storrs, CT, United States.,School of Nursing, University of Connecticut, Storrs, CT, United States.,School of Nursing, University of Connecticut, Storrs, CT, United States.,School of Nursing, University of Connecticut, Storrs, CT, United States.,School of Nursing, University of Connecticut, Storrs, CT, United States
| |
Collapse
|
60
|
Elechalawar CK, Bhattacharya D, Ahmed MT, Gora H, Sridharan K, Chaturbedy P, Sinha SH, Chandra Sekhar Jaggarapu MM, Narayan KP, Chakravarty S, Eswaramoorthy M, Kundu TK, Banerjee R. Dual targeting of folate receptor-expressing glioma tumor-associated macrophages and epithelial cells in the brain using a carbon nanosphere-cationic folate nanoconjugate. NANOSCALE ADVANCES 2019; 1:3555-3567. [PMID: 36133563 PMCID: PMC9417975 DOI: 10.1039/c9na00056a] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 07/22/2019] [Indexed: 05/21/2023]
Abstract
Glioblastoma multiforme (GBM), the highly invasive form of glioma, exhibits the highest mortality in patients with brain malignancies. Increasing glioma patients' survivability is challenging, as targeting only tumor-associated malignant cells would not reduce the overall aggressiveness of the tumor mass. This is due to the inadequacy in countering pro-proliferative, invasive and metastatic factors released by tumor-mass associated macrophages (TAMs). Hence, strategically, dual targeting both tumor cells and TAMs is necessary for effective glioma treatment and increased survivability. Conventional FR-targeting systems can easily target cancer cells that overtly express folate receptors (FRs). However, FRs are expressed only moderately in both glioma cells and in TAMs. Hence, it is more challenging to coordinate dual targeting of glioma cells and TAMs with lower levels of FR expression. A recently developed carbon nanosphere (CSP) with effective blood-brain barrier (BBB) penetrability was modified with a new folic acid-cationic lipid conjugate (F8) as a targeting ligand. The uniqueness of the cationic lipid-folate conjugate is that it stably associates with the negatively charged CSP surface at about >22 mol% surface concentration, a concentration at least 5-fold higher than what is achieved for conventional FR-targeting delivery systems. This enabled dual uptake of the CSP on TAMs and tumor cells via FRs. A doxorubicin-associated FR-targeting formulation (CFD), in an orthotopic glioma model and in a glioma subcutaneous model, induced the maximum anticancer effect with enhanced average mice survivability twice that of untreated mice and without any systemic liver toxicity. Additionally, we observed a significant decrease of TAM-released pro-aggressive factors, TGF-β, STAT3, invasion and migration related sICAM-1, and other cytokines indicating anti-TAM activity of the CFD. Taken together, we principally devised, to the best of our knowledge, the first FR-targeting nano-delivery system for targeting brain-associated TAMs and tumor cells as an efficient glioma therapeutic.
Collapse
Affiliation(s)
- Chandra Kumar Elechalawar
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology Hyderabad 500 007 India
- Academy of Scientific & Innovative Research (AcSIR) Taramani Chennai 600113 India
| | - Dwaipayan Bhattacharya
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology Hyderabad 500 007 India
- Department of Biological Sciences, BITS Pilani Hyderabad Campus, Jawahar Nagar, Shameerpet Mandal Hyderabad 500078 India
| | - Mohammed Tanveer Ahmed
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology Hyderabad 500 007 India
- Academy of Scientific & Innovative Research (AcSIR) Taramani Chennai 600113 India
| | - Halley Gora
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology Hyderabad 500 007 India
| | - Kathyayani Sridharan
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology Hyderabad 500 007 India
- Academy of Scientific & Innovative Research (AcSIR) Taramani Chennai 600113 India
| | - Piyush Chaturbedy
- Chemistry and Physics of Materials Unit, Jawaharlal Nehru Centre for Advanced Scientific Research Jakkur P.O Bangalore 560 064 India
| | - Sarmistha Halder Sinha
- Transcription and Disease Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research Jakkur P.O Bangalore 560 064 India
| | - Madhan Mohan Chandra Sekhar Jaggarapu
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology Hyderabad 500 007 India
- Academy of Scientific & Innovative Research (AcSIR) Taramani Chennai 600113 India
| | - Kumar Pranav Narayan
- Department of Biological Sciences, BITS Pilani Hyderabad Campus, Jawahar Nagar, Shameerpet Mandal Hyderabad 500078 India
| | - Sumana Chakravarty
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology Hyderabad 500 007 India
| | - Muthusamy Eswaramoorthy
- Chemistry and Physics of Materials Unit, Jawaharlal Nehru Centre for Advanced Scientific Research Jakkur P.O Bangalore 560 064 India
| | - Tapas Kumar Kundu
- Transcription and Disease Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research Jakkur P.O Bangalore 560 064 India
| | - Rajkumar Banerjee
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology Hyderabad 500 007 India
- Academy of Scientific & Innovative Research (AcSIR) Taramani Chennai 600113 India
| |
Collapse
|
61
|
Roy R, Morad G, Jedinak A, Moses MA. Metalloproteinases and their roles in human cancer. Anat Rec (Hoboken) 2019; 303:1557-1572. [PMID: 31168956 DOI: 10.1002/ar.24188] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 12/27/2018] [Accepted: 03/04/2019] [Indexed: 02/06/2023]
Abstract
It is now widely appreciated that members of the matrix metalloproteinase (MMP) family of enzymes play a key role in cancer development and progression along with many of the hallmarks associated with them. The activity of these enzymes has been directly implicated in extracellular matrix remodeling, the processing of growth factors and receptors, the modulation of cell migration, proliferation, and invasion, the epithelial to mesenchymal transition, the regulation of immune responses, and the control of angiogenesis. Certain MMP family members have been validated as biomarkers of a variety of human cancers including those of the breast, brain, pancreas, prostate, ovary, and others. The related metalloproteinases, the A disintegrin and metalloproteinases (ADAMs), share a number of these functions as well. Here, we explore these essential metalloproteinases and some of their disease-associated activities in detail as well as some of their complementary translational potential. Anat Rec, 2019. © 2019 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Roopali Roy
- The Vascular Biology Program, Boston Children's Hospital and the Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Golnaz Morad
- The Vascular Biology Program, Boston Children's Hospital and the Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Andrej Jedinak
- The Vascular Biology Program, Boston Children's Hospital and the Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Marsha A Moses
- The Vascular Biology Program, Boston Children's Hospital and the Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
62
|
Zhang Y, Xiong X, Huai Y, Dey A, Hossen NM, Roy RV, Elechalawar CK, Rao G, Bhattacharya R, Mukherjee P. Gold Nanoparticles Disrupt Tumor Microenvironment - Endothelial Cell Cross Talk To Inhibit Angiogenic Phenotypes in Vitro. Bioconjug Chem 2019; 30:1724-1733. [PMID: 31067032 PMCID: PMC6939887 DOI: 10.1021/acs.bioconjchem.9b00262] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
It is currently recognized that perpetual cross talk among key players in tumor microenvironment such as cancer cells (CCs), cancer associated fibroblasts (CAFs), and endothelial cells (ECs) plays a critical role in tumor progression, metastasis, and therapy resistance. Disruption of the cross talk may be useful to improve the outcome of therapeutics for which limited options are available. In the current study we investigate the use of gold nanoparticles (AuNPs) as a therapeutic tool to disrupt the multicellular cross talk within the TME cells with an emphasis on inhibiting angiogenesis. We demonstrate here that AuNPs disrupt signal transduction from TME cells (CCs, CAFs, and ECs) to ECs and inhibit angiogenic phenotypes in vitro. We show that conditioned media (CM) from ovarian CCs, CAFs, or ECs themselves induce tube formation and migration of ECs in vitro. Migration of ECs is also induced when ECs are cocultured with CCs, CAFs, or ECs. In contrast, CM from the cells treated with AuNPs or cocultured cells pretreated with AuNPs demonstrate diminished effects on ECs tube formation and migration. Mechanistically, AuNPs deplete ∼95% VEGF165 from VEGF single-protein solution and remove up to ∼45% of VEGF165 from CM, which is reflected on reduced activation of VEGF-Receptor 2 (VEGFR2) as compared to control CM. These results demonstrate that AuNPs inhibit angiogenesis via blockade of VEGF-VEGFR2 signaling from TME cells to endothelial cells.
Collapse
Affiliation(s)
- Yushan Zhang
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| | - Xunhao Xiong
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| | - Yanyan Huai
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| | - Anindya Dey
- Department of Obstetrics and Gynecology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| | - Nazir Md Hossen
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| | - Ram Vinod Roy
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| | - Chandra Kumar Elechalawar
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| | - Geeta Rao
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| | - Resham Bhattacharya
- Department of Obstetrics and Gynecology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
- Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| | - Priyabrata Mukherjee
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
- Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| |
Collapse
|
63
|
Houthuijzen JM, Jonkers J. Cancer-associated fibroblasts as key regulators of the breast cancer tumor microenvironment. Cancer Metastasis Rev 2019; 37:577-597. [PMID: 30465162 DOI: 10.1007/s10555-018-9768-3] [Citation(s) in RCA: 132] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Tumor cells exist in close proximity with non-malignant cells. Extensive and multilayered crosstalk between tumor cells and stromal cells tailors the tumor microenvironment (TME) to support survival, growth, and metastasis. Fibroblasts are one of the largest populations of non-malignant host cells that can be found within the TME of breast, pancreatic, and prostate tumors. Substantial scientific evidence has shown that these cancer-associated fibroblasts (CAFs) are not only associated with tumors by proximity but are also actively recruited to developing tumors where they can influence other cells of the TME as well as influencing tumor cell survival and metastasis. This review discusses the impact of CAFs on breast cancer biology and highlights their heterogeneity, origin and their role in tumor progression, ECM remodeling, therapy resistance, metastasis, and the challenges ahead of targeting CAFs to improve therapy response.
Collapse
Affiliation(s)
- J M Houthuijzen
- Department of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands.
| | - J Jonkers
- Department of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| |
Collapse
|
64
|
Alvim RG, Hughes C, Somma A, Nagar KK, Wong NC, La Rosa S, Monette S, Kim K, Coleman JA. The potential risk of tumor progression after use of dehydrated human amnion/chorion membrane allograft in a positive margin resection model. Ther Adv Urol 2019; 11:1756287219837771. [PMID: 30956688 PMCID: PMC6444417 DOI: 10.1177/1756287219837771] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 02/10/2019] [Indexed: 11/16/2022] Open
Abstract
Objective: The objective of this study was to examine the impact of dehydrated human amnion/chorion membrane (dHACM) allografts on prostate and bladder cancer growth in the setting of residual disease and positive surgical margins. Materials and methods: A commercially available version of dHACM was used. Cytokines were identified and quantified, followed by comparative analysis of cell growth in two different human cell lines: prostate cancer (LNCaP) and bladder cancer (UM-UC-3), in vitro and in vivo. Tumor growth between the two groups, membrane versus no membrane implant, was compared and immunohistochemistry studies were conducted to quantify CD-31, Ki-67, and vimentin. A Student’s unpaired t-test was used to determine statistical significance. Results: The UM-UC-3 and LNCaP cells grew quicker in medium plus 10% serum and dHACM extract than in the other media (p = 0.03). A total of 28 distinct cytokines were found in the extract, 11 of which had relatively high concentrations and are associated with prostate and bladder cancer tumor progression. In vivo LNCaP model, after 10 weeks, the median tumor volume in the membrane group was almost threefold larger than the partial resection alone (p = 0.01). Two weeks after resection, in the UM-UC-3 model, the membrane group reached fourfold larger than the partial resection without membrane group (p < 0.01). In both groups, the expression of CD-31 and Ki-67 markers were similar and showed no statistical significance (p > 0.05). It was only in the LNCaP tumors that vimentin expression was significantly higher in the group without membrane compared with the membrane group (p = 0.008). Conclusion: The use of dHACM after partial tumor resection is related to faster tumor relapse and growth in prostate and urothelial cancer in vivo models, showing a potential risk of rapid local recurrence in patients at high risk of positive margins.
Collapse
Affiliation(s)
- Ricardo G Alvim
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Christopher Hughes
- Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Alexander Somma
- Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Karan K Nagar
- Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nathan C Wong
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Stephen La Rosa
- Department of Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sebastien Monette
- Laboratory of Comparative Pathology and the Genetically Modified Animal Phenotyping Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kwanghee Kim
- Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jonathan A Coleman
- Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| |
Collapse
|
65
|
Dourado MR, Korvala J, Åström P, De Oliveira CE, Cervigne NK, Mofatto LS, Campanella Bastos D, Pereira Messetti AC, Graner E, Paes Leme AF, Coletta RD, Salo T. Extracellular vesicles derived from cancer-associated fibroblasts induce the migration and invasion of oral squamous cell carcinoma. J Extracell Vesicles 2019; 8:1578525. [PMID: 30788085 PMCID: PMC6374932 DOI: 10.1080/20013078.2019.1578525] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 12/11/2018] [Accepted: 01/18/2019] [Indexed: 12/15/2022] Open
Abstract
As one of the most abundant constituents of the tumour microenvironment (TME), cancer-associated fibroblasts (CAF) display critical roles during tumour progression and metastasis. Multiple classes of molecules including growth factors, cytokines, proteases and extracellular matrix proteins, are produced by CAF to act as mediators of the stroma-tumour interactions. One of the main channels for this communication is associated with extracellular vesicles (EV), which are secreted particles loaded with protein and genetic information. In this study, we evaluated the effects of EV derived from CAF primary human cell lines (n = 5) on proliferation, survival, migration, and invasion of oral squamous cell carcinoma (OSCC) cells. As controls, EV from human primary-established normal oral fibroblasts (NOF, n = 5) were used. Our in vitro assays showed that CAF-EV significantly induces migration and invasion of OSCC cells and promote a disseminated pattern of HSC-3 cell invasion in the 3D organotypic assay. Furthermore, gene expression analysis of EV-treated cancer cells revealed changes in the pathways associated with tumour metabolism and up-regulation of tumour invasion genes. Our findings suggest a significant role of CAF-EV in promoting the migration and invasion of OSCC cells, which are related to the activation of cancer-related pathways.
Collapse
Affiliation(s)
- Mauricio Rocha Dourado
- Department of Oral Diagnosis, Piracicaba Dental School, University of Campinas, Piracicaba, Brazil.,Cancer and Translational Medicine Research Unit, Faculty of Medicine and Medical Research Center Oulu, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Johanna Korvala
- Cancer and Translational Medicine Research Unit, Biocenter Oulu and Faculty of Medicine, University of Oulu, Oulu, Finland
| | - Pirjo Åström
- Cancer and Translational Medicine Research Unit, Faculty of Medicine and Medical Research Center Oulu, Oulu University Hospital, University of Oulu, Oulu, Finland
| | | | - Nilva K Cervigne
- Department of Morphology and Basic Pathology, Faculty of Medicine of Jundiai, Jundiai, Brazil
| | - Luciana Souto Mofatto
- Genomics and Expression Laboratory, Department of Genetics, Evolution and Bioagents, Institute of Biology, University of Campinas, Piracicaba, Brazil
| | - Debora Campanella Bastos
- Department of Oral Diagnosis, Piracicaba Dental School, University of Campinas, Piracicaba, Brazil
| | | | - Edgard Graner
- Department of Oral Diagnosis, Piracicaba Dental School, University of Campinas, Piracicaba, Brazil
| | | | - Ricardo D Coletta
- Department of Oral Diagnosis, Piracicaba Dental School, University of Campinas, Piracicaba, Brazil
| | - Tuula Salo
- Cancer and Translational Medicine Research Unit, Faculty of Medicine and Medical Research Center Oulu, Oulu University Hospital, University of Oulu, Oulu, Finland.,Institute of Oral and Maxillofacial Disease, University of Helsinki, and HUSLAB, Department of Pathology, Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
66
|
Grünwald B, Schoeps B, Krüger A. Recognizing the Molecular Multifunctionality and Interactome of TIMP-1. Trends Cell Biol 2019; 29:6-19. [DOI: 10.1016/j.tcb.2018.08.006] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 08/24/2018] [Accepted: 08/28/2018] [Indexed: 01/31/2023]
|
67
|
Dynamic matrisome: ECM remodeling factors licensing cancer progression and metastasis. Biochim Biophys Acta Rev Cancer 2018; 1870:207-228. [DOI: 10.1016/j.bbcan.2018.09.002] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 09/07/2018] [Accepted: 09/30/2018] [Indexed: 01/04/2023]
|
68
|
Spinnen J, Ringe J, Sittinger M. CCL25 chemokine-guided stem cell attraction: an assessment of possible benefits and risks. Regen Med 2018; 13:833-844. [PMID: 30284497 DOI: 10.2217/rme-2018-0016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Due to its chemoattraction potential on mesenchymal stromal cells of the CCL25/CCR9 axis, local application of CCL25 to severely damaged tissues may be a promising approach for regenerative therapies. Analysis of the given data revealed that CCL25/CCR9 signaling has a crucial role in regulation of an adult immune homeostasis. CCR9 expression variations resulted in dysfunctional immune response in colitis, rheumatoid arthritis and endometriosis. Regarding oncology, different neoplastic tissues exploit CCL25-dependent CCR9 signaling for either local proliferation or migration processes. The CCR9 pathway likely can trigger crosstalk between the Akt and NOTCH pathway and thus participate in the regulation of the neoplastic behavior. In conclusion, the designated application-tissue requires precise molecular analysis of possible CCR9 expression due to its proto-oncogenic characteristics.
Collapse
Affiliation(s)
- Jacob Spinnen
- Tissue Engineering Laboratory, Berlin-Brandenburg Center for Regenerative Therapies, Department of Rheumatology & Clinical Immunology, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Jochen Ringe
- Tissue Engineering Laboratory, Berlin-Brandenburg Center for Regenerative Therapies, Department of Rheumatology & Clinical Immunology, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Michael Sittinger
- Tissue Engineering Laboratory, Berlin-Brandenburg Center for Regenerative Therapies, Department of Rheumatology & Clinical Immunology, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| |
Collapse
|
69
|
Investigating Pathogenic and Hepatocarcinogenic Mechanisms from Normal Liver to HCC by Constructing Genetic and Epigenetic Networks via Big Genetic and Epigenetic Data Mining and Genome-Wide NGS Data Identification. DISEASE MARKERS 2018; 2018:8635329. [PMID: 30344796 PMCID: PMC6174771 DOI: 10.1155/2018/8635329] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 08/02/2018] [Accepted: 08/13/2018] [Indexed: 12/14/2022]
Abstract
The prevalence of hepatocellular carcinoma (HCC) is still high worldwide because liver diseases could develop into HCC. Recent reports indicate nonalcoholic fatty liver disease and nonalcoholic steatohepatitis (NAFLD&NASH) and primary biliary cirrhosis and primary sclerosing cholangitis (PBC&PSC) are significant of HCC. Therefore, understanding the cellular mechanisms of the pathogenesis and hepatocarcinogenesis from normal liver cells to HCC through NAFLD&NASH or PBC&PSC is a priority to prevent the progression of liver damage and reduce the risk of further complications. By the genetic and epigenetic data mining and the system identification through next-generation sequencing data and its corresponding DNA methylation profiles of liver cells in normal, NAFLD&NASH, PBC&PSC, and HCC patients, we identified the genome-wide real genetic and epigenetic networks (GENs) of normal, NAFLD&NASH, PBC&PSC, and HCC patients. In order to get valuable insight into these identified genome-wide GENs, we then applied a principal network projection method to extract the corresponding core GENs for normal liver cells, NAFLD&NASH, PBC&PSC, and HCC. By comparing the signal transduction pathways involved in the identified core GENs, we found that the hepatocarcinogenesis through NAFLD&NASH was induced through DNA methylation of HIST2H2BE, HSPB1, RPL30, and ALDOB and the regulation of miR-21 and miR-122, and the hepatocarcinogenesis through PBC&PSC was induced through DNA methylation of RPL23A, HIST2H2BE, TIMP1, IGF2, RPL30, and ALDOB and the regulation of miR-29a, miR-21, and miR-122. The genetic and epigenetic changes in the pathogenesis and hepatocarcinogenesis potentially serve as potential diagnostic biomarkers and/or therapeutic targets.
Collapse
|
70
|
Alkasalias T, Moyano-Galceran L, Arsenian-Henriksson M, Lehti K. Fibroblasts in the Tumor Microenvironment: Shield or Spear? Int J Mol Sci 2018; 19:ijms19051532. [PMID: 29883428 PMCID: PMC5983719 DOI: 10.3390/ijms19051532] [Citation(s) in RCA: 166] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Revised: 05/15/2018] [Accepted: 05/16/2018] [Indexed: 12/11/2022] Open
Abstract
Tumorigenesis is a complex process involving dynamic interactions between malignant cells and their surrounding stroma, including both the cellular and acellular components. Within the stroma, fibroblasts represent not only a predominant cell type, but also a major source of the acellular tissue microenvironment comprising the extracellular matrix (ECM) and soluble factors. Normal fibroblasts can exert diverse suppressive functions against cancer initiating and metastatic cells via direct cell-cell contact, paracrine signaling by soluble factors, and ECM integrity. The loss of such suppressive functions is an inherent step in tumor progression. A tumor cell-induced switch of normal fibroblasts into cancer-associated fibroblasts (CAFs), in turn, triggers a range of pro-tumorigenic signals accompanied by distraction of the normal tissue architecture, thus creating an optimal niche for cancer cells to grow extensively. To further support tumor progression and metastasis, CAFs secrete factors such as ECM remodeling enzymes that further modify the tumor microenvironment in combination with the altered adhesive forces and cell-cell interactions. These paradoxical tumor suppressive and promoting actions of fibroblasts are the focus of this review, highlighting the heterogenic molecular properties of both normal and cancer-associated fibroblasts, as well as their main mechanisms of action, including the emerging impact on immunomodulation and different therapy responses.
Collapse
Affiliation(s)
- Twana Alkasalias
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Biomedicum, Solnavägen 9, SE-17177 Stockholm, Sweden.
- Department of Biology, College of Science, Salahaddin University, Irbil 44002, Kurdistan-Iraq.
| | - Lidia Moyano-Galceran
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Biomedicum, Solnavägen 9, SE-17177 Stockholm, Sweden.
| | - Marie Arsenian-Henriksson
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Biomedicum, Solnavägen 9, SE-17177 Stockholm, Sweden.
| | - Kaisa Lehti
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Biomedicum, Solnavägen 9, SE-17177 Stockholm, Sweden.
- Research Programs Unit, Genome-Scale Biology and Medicum, University of Helsinki, and Helsinki University Hospital, P.O. Box 63, FI-00014 Helsinki, Finland.
| |
Collapse
|
71
|
Nguyen EV, Centenera MM, Moldovan M, Das R, Irani S, Vincent AD, Chan H, Horvath LG, Lynn DJ, Daly RJ, Butler LM. Identification of Novel Response and Predictive Biomarkers to Hsp90 Inhibitors Through Proteomic Profiling of Patient-derived Prostate Tumor Explants. Mol Cell Proteomics 2018; 17:1470-1486. [PMID: 29632047 DOI: 10.1074/mcp.ra118.000633] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 03/26/2018] [Indexed: 12/16/2022] Open
Abstract
Inhibition of the heat shock protein 90 (Hsp90) chaperone is a promising therapeutic strategy to target expression of the androgen receptor (AR) and other oncogenic drivers in prostate cancer cells. However, identification of clinically-relevant responses and predictive biomarkers is essential to maximize efficacy and treatment personalization. Here, we combined mass spectrometry (MS)-based proteomic analyses with a unique patient-derived explant (PDE) model that retains the complex microenvironment of primary prostate tumors. Independent discovery and validation cohorts of PDEs (n = 16 and 30, respectively) were cultured in the absence or presence of Hsp90 inhibitors AUY922 or 17-AAG. PDEs were analyzed by LC-MS/MS with a hyper-reaction monitoring data independent acquisition (HRM-DIA) workflow, and differentially expressed proteins identified using repeated measure analysis of variance (ANOVA; raw p value <0.01). Using gene set enrichment, we found striking conservation of the most significantly AUY922-altered gene pathways between the discovery and validation cohorts, indicating that our experimental and analysis workflows were robust. Eight proteins were selectively altered across both cohorts by the most potent inhibitor, AUY922, including TIMP1, SERPINA3 and CYP51A (adjusted p < 0.01). The AUY922-mediated decrease in secretory TIMP1 was validated by ELISA of the PDE culture medium. We next exploited the heterogeneous response of PDEs to 17-AAG in order to detect predictive biomarkers of response and identified PCBP3 as a marker with increased expression in PDEs that had no response or increased in proliferation. Also, 17-AAG treatment led to increased expression of DNAJA1 in PDEs that exhibited a cytostatic response, revealing potential drug resistance mechanisms. This selective regulation of DNAJA1 was validated by Western blot analysis. Our study establishes "proof-of-principle" that proteomic profiling of drug-treated PDEs represents an effective and clinically-relevant strategy for identification of biomarkers that associate with certain tumor-specific responses.
Collapse
Affiliation(s)
- Elizabeth V Nguyen
- From the ‡Cancer Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia.,§Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria 3800, Australia
| | - Margaret M Centenera
- ¶Adelaide Medical School and Freemasons Foundation Centre for Men's Health, University of Adelaide, Adelaide, South Australia 5005, Australia.,‖South Australian Health and Medical Research Institute, Adelaide, South Australia 5000, Australia
| | - Max Moldovan
- ‖South Australian Health and Medical Research Institute, Adelaide, South Australia 5000, Australia
| | - Rajdeep Das
- ¶Adelaide Medical School and Freemasons Foundation Centre for Men's Health, University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Swati Irani
- ¶Adelaide Medical School and Freemasons Foundation Centre for Men's Health, University of Adelaide, Adelaide, South Australia 5005, Australia.,‖South Australian Health and Medical Research Institute, Adelaide, South Australia 5000, Australia
| | - Andrew D Vincent
- ¶Adelaide Medical School and Freemasons Foundation Centre for Men's Health, University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Howard Chan
- From the ‡Cancer Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia.,§Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria 3800, Australia
| | - Lisa G Horvath
- **Cancer Division, The Kinghorn Cancer Centre/Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia.,‡‡Royal Prince Alfred Hospital, Camperdown, New South Wales 2050, Australia.,§§Department of Medical Oncology, Chris O'Brien Lifehouse, Camperdown, New South Wales 2050, Australia
| | - David J Lynn
- ‖South Australian Health and Medical Research Institute, Adelaide, South Australia 5000, Australia.,¶¶School of Medicine, Flinders University, Bedford Park, SA 5042, Australia
| | - Roger J Daly
- From the ‡Cancer Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia; .,§Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria 3800, Australia
| | - Lisa M Butler
- ¶Adelaide Medical School and Freemasons Foundation Centre for Men's Health, University of Adelaide, Adelaide, South Australia 5005, Australia.,‖South Australian Health and Medical Research Institute, Adelaide, South Australia 5000, Australia
| |
Collapse
|
72
|
Xu Y, Wang K, Yu Q. FRMD6 inhibits human glioblastoma growth and progression by negatively regulating activity of receptor tyrosine kinases. Oncotarget 2018; 7:70080-70091. [PMID: 27661120 PMCID: PMC5342536 DOI: 10.18632/oncotarget.12148] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 09/02/2016] [Indexed: 02/06/2023] Open
Abstract
FRMD6 is an Ezrin/Radixin/Moesin (ERM) family protein and a human homologue of Drosophila expanded (ex). Ex functions in parallel of Drosophila merlin at upstream of the Hippo signaling pathway that controls proliferation, apoptosis, tissue regeneration, and tumorigenesis. Even though the core kinase cascade (MST1/2-Lats1/2-YAP/TAZ) of the Hippo pathway has been well established, its upstream regulators are not well understood. Merlin promotes activation of the Hippo pathway. However, the effect of FRMD6 on the Hippo pathway is controversial. Little is known about how FRMD6 functions and the potential role of FRMD in gliomagenesis and glioblastoma (GBM) progression. We demonstrate for the first time that FRMD6 is down-regulated in human GBM cells and tissues and that increased FRMD6 expression inhibits whereas FRMD6 knockdown promotes GBM cell proliferation/invasion in vitro and GBM growth/progression in vivo. Furthermore, we demonstrate that unlike increased expression of merlin, which enhances the stress induced activation of the Hippo pathway, increased FRMD6 expression displays little effect on the pathway. In contrast, we show that FRMD6 inhibits activation of a couple of receptor tyrosine kinases (RTKs) including c-Met and PDGFR and their downstream Erk and AKT kinases. Moreover, we show that expression of constitutively active c-Met, the TPR-Met fusion protein, largely reverses the anti-GBM effect of FRMD6 in vivo, suggesting that FRMD6 functions at least partially through inhibiting activity of RTKs especially c-Met. These results establish a novel function of FRMD6 in inhibiting human GBM growth and progression and uncover a novel mechanism by which FRMD6 exerts its anti-GBM activity.
Collapse
Affiliation(s)
- Yin Xu
- Department of Oncological Sciences Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kaiqiang Wang
- Department of Oncological Sciences Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Qin Yu
- Department of Oncological Sciences Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
73
|
Ghoshal-Gupta S, Kutiyanawalla A, Lee BR, Ojha J, Nurani A, Mondal AK, Kolhe R, Rojiani AM, Rojiani MV. TIMP-1 downregulation modulates miR-125a-5p expression and triggers the apoptotic pathway. Oncotarget 2018; 9:8941-8956. [PMID: 29507665 PMCID: PMC5823642 DOI: 10.18632/oncotarget.23832] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 12/26/2017] [Indexed: 12/11/2022] Open
Abstract
Matrix metalloproteinases and their natural inhibitors (TIMPs) are important elements in a wide range of oncology settings. Elevated levels of tissue inhibitor of metalloproteinase-1 (TIMP-1) have often been associated with increased tumorigenesis. This has been demonstrated in a number of clinical and experimental models which include breast, gastric, colorectal and non-small cell lung carcinoma (NSCLC). Our earlier studies have identified increased angiogenic activity and aggressive tumor kinetics in TIMP-1 overexpressing H2009 lung adenocarcinoma cells. TIMP-1 overexpression has also been implicated in antiapoptotic responses, inducing a significant upregulation of Bcl-2. These TIMP-1 functions have been shown to be MMP-independent and provide insight into its pleiotropic activities. The current study examines microRNA (miRNA) interactions with this molecule. We have sought to define the relationship between TIMP-1 and miRNA by knocking down TIMP-1 in high TIMP-1 expressing lung adenocarcinoma cell lines. TIMP-1 knockdown resulted in increased expression of miR-125a-5p with a concomitant increase in apoptosis and attenuation of the tumorigenic features of these cells. We have identified TIMP-1 as a bona fide target of miR-125a-5p, and their interaction resulted in an increase in p53 expression. We further corroborated our in vitro data with patient samples, which exhibited an inverse correlation between TIMP-1 and miR-125a-5p expression. Our study lends support to the notion that elevated TIMP-1 levels, which are frequently associated with poor prognosis, cause aberrant modulation of miRNAs.
Collapse
Affiliation(s)
- Sampa Ghoshal-Gupta
- Department of Pathology, Medical College of Georgia-Augusta University, Augusta, GA, United States of America
| | - Ammar Kutiyanawalla
- Department of Pathology, Medical College of Georgia-Augusta University, Augusta, GA, United States of America
| | - Byung Rho Lee
- Department of Pathology, Medical College of Georgia-Augusta University, Augusta, GA, United States of America
| | - Juhi Ojha
- Department of Pathology, Medical College of Georgia-Augusta University, Augusta, GA, United States of America
| | - Aliya Nurani
- Department of Pathology, Medical College of Georgia-Augusta University, Augusta, GA, United States of America
| | - Ashis K Mondal
- Department of Pathology, Medical College of Georgia-Augusta University, Augusta, GA, United States of America
| | - Ravindra Kolhe
- Department of Pathology, Medical College of Georgia-Augusta University, Augusta, GA, United States of America
| | - Amyn M Rojiani
- Department of Pathology, Medical College of Georgia-Augusta University, Augusta, GA, United States of America
| | - Mumtaz V Rojiani
- Department of Pathology, Medical College of Georgia-Augusta University, Augusta, GA, United States of America.,Department of Medicine, Medical College of Georgia-Augusta University, Augusta, GA, United States of America
| |
Collapse
|
74
|
Kuppusamy P, Govindan N, Yusoff MM, Ichwan SJ. Proteins are potent biomarkers to detect colon cancer progression. Saudi J Biol Sci 2017; 24:1212-1221. [PMID: 28855814 PMCID: PMC5562385 DOI: 10.1016/j.sjbs.2014.09.017] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Revised: 09/21/2014] [Accepted: 09/24/2014] [Indexed: 01/11/2023] Open
Abstract
Colon cancer is the most common type of cancer and major cause of death worldwide. The detection of colon cancer is difficult in early stages. However, the secretory proteins have been used as ideal biomarker for the detection of colon cancer progress in cancer patients. Serum/tissue protein expression could help general practitioners to identify colon cancer at earlier stages. By this way, we use the biomarkers to evaluate the anticancer drugs and their response to therapy in cancer models. Recently, the biomarker discovery is important in cancer biology and disease management. Also, many measurable specific molecular components have been studied in colon cancer therapeutics. The biomolecules are mainly DNA, RNA, metabolites, enzymes, mRNA, aptamers and proteins. Thus, in this review we demonstrate the important protein biomarker in colon cancer development and molecular identification of protein biomarker discovery.
Collapse
Affiliation(s)
- Palaniselvam Kuppusamy
- Mammalian Cell Technology Laboratory, Faculty of Industrial Sciences and Technology, Universiti Malaysia Pahang, Lebuhraya Tun Razak 26300, Gambang, Kuantan, Pahang, Malaysia
| | - Natanamurugaraj Govindan
- Mammalian Cell Technology Laboratory, Faculty of Industrial Sciences and Technology, Universiti Malaysia Pahang, Lebuhraya Tun Razak 26300, Gambang, Kuantan, Pahang, Malaysia
| | - Mashitah M. Yusoff
- Mammalian Cell Technology Laboratory, Faculty of Industrial Sciences and Technology, Universiti Malaysia Pahang, Lebuhraya Tun Razak 26300, Gambang, Kuantan, Pahang, Malaysia
| | - Solachuddin J.A. Ichwan
- Kulliyyah of Dentistry, International Islamic University Malaysia, Bandar Indera Mahkota 125200, Kuantan, Pahang, Malaysia
| |
Collapse
|
75
|
Bui TP, Hoang AN, Le PL, Pham BT, Nguyen LTT, Do HM, Ta TV, Trinh TH. Matrix metalloproteinases in Vietnamese patients with colorectal cancer. Oncol Lett 2017; 13:2097-2104. [PMID: 28454367 PMCID: PMC5403361 DOI: 10.3892/ol.2017.5680] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 11/30/2016] [Indexed: 01/12/2023] Open
Abstract
Matrix metalloproteinases (MMPs), a family of endopeptidases also known as gelatinases, have been reported to affect the acquisition of the cell proliferative, cell invasive and metastatic phenotype of several types of cancer. In particular, the gelatinases MMP-2 and −9 have been revealed to facilitate tumor growth and invasion in patients with colorectal cancer (CRC). However, it is not known whether the gelatinase activity of MMP-2 and −9 is also elevated in Vietnamese patients with CRC. The activity of MMP-2 and −9 in the tissue samples of 103 patients with CRC was evaluated by gelatin zymography and quantified using ImageJ. The association between the level of activity of MMP-2 and −9 and various clinicopathological factors was analyzed, and Chisio BioPAX Editor software was used to visualize the biological pathways regulating the activity of the MMPs. The present study noticed significantly increased activity of active MMP-2 and MMP-9 in tumor tissues (P<0.01), and significantly decreased levels of pro-form MMP-2 and MMP-9 in tumor tissues (P<0.01), compared with that in adjacent tissues in patients with CRC. A correlation between the normalized different activity of MMP-2 and −9 and various clinicopathological features was observed. Furthermore, bioinformatics analysis indicated that the alteration in the activity of MMP-2 and MMP-9 may have been controlled by biological pathways involving the tissue inhibitors of metalloprotease-2 and −1. These findings indicate that the activity of the gelatinases MMP-2 and −9 affects the tumor progression and metastasis of patients with CRC, providing a potential novel approach for determining the prognosis of CRC.
Collapse
Affiliation(s)
- Thao Phuong Bui
- Department of Biology and Key Laboratory of Enzyme and Protein Technology, College of Science, Vietnam National University, Hanoi 10000, Vietnam
| | - Anh Ngoc Hoang
- Department of Biology and Key Laboratory of Enzyme and Protein Technology, College of Science, Vietnam National University, Hanoi 10000, Vietnam
| | - Phuong Lan Le
- Department of Biology and Key Laboratory of Enzyme and Protein Technology, College of Science, Vietnam National University, Hanoi 10000, Vietnam
| | - Bich Thi Pham
- Department of Biology and Key Laboratory of Enzyme and Protein Technology, College of Science, Vietnam National University, Hanoi 10000, Vietnam
| | - Linh Thi Tu Nguyen
- Department of Biology and Key Laboratory of Enzyme and Protein Technology, College of Science, Vietnam National University, Hanoi 10000, Vietnam
| | - Ha Minh Do
- Department of Biology and Key Laboratory of Enzyme and Protein Technology, College of Science, Vietnam National University, Hanoi 10000, Vietnam
| | - To Van Ta
- Department of Anatomical Pathology-Cytopathology, Vietnam National Cancer Hospital, Hanoi 10000, Vietnam
| | - Thai Hong Trinh
- Department of Biology and Key Laboratory of Enzyme and Protein Technology, College of Science, Vietnam National University, Hanoi 10000, Vietnam
| |
Collapse
|
76
|
Abstract
A compelling long-term goal of cancer biology is to understand the crucial players during tumorigenesis in order to develop new interventions. Here, we review how the four non-redundant tissue inhibitors of metalloproteinases (TIMPs) regulate the pericellular proteolysis of a vast range of matrix and cell surface proteins, generating simultaneous effects on tumour architecture and cell signalling. Experimental studies demonstrate the contribution of TIMPs to the majority of cancer hallmarks, and human cancers invariably show TIMP deregulation in the tumour or stroma. Of the four TIMPs, TIMP1 overexpression or TIMP3 silencing is consistently associated with cancer progression or poor patient prognosis. Future efforts will align mouse model systems with changes in TIMPs in patients, will delineate protease-independent TIMP function, will pinpoint therapeutic targets within the TIMP-metalloproteinase-substrate network and will use TIMPs in liquid biopsy samples as biomarkers for cancer prognosis.
Collapse
Affiliation(s)
- Hartland W Jackson
- Department of Medical Biophysics, University of Toronto, Princess Margaret Cancer Centre, TMDT 301-13, 101 College Street, Toronto, Ontario, M5G IL7 Canada
- Bodenmiller Laboratory, University of Zürich, Institute for Molecular Life Sciences, Winterthurstrasse 190, 8057 Zürich, Switzerland
| | - Virginie Defamie
- Department of Medical Biophysics, University of Toronto, Princess Margaret Cancer Centre, TMDT 301-13, 101 College Street, Toronto, Ontario, M5G IL7 Canada
| | - Paul Waterhouse
- Department of Medical Biophysics, University of Toronto, Princess Margaret Cancer Centre, TMDT 301-13, 101 College Street, Toronto, Ontario, M5G IL7 Canada
| | - Rama Khokha
- Department of Medical Biophysics, University of Toronto, Princess Margaret Cancer Centre, TMDT 301-13, 101 College Street, Toronto, Ontario, M5G IL7 Canada
| |
Collapse
|
77
|
Boufraqech M, Zhang L, Nilubol N, Sadowski SM, Kotian S, Quezado M, Kebebew E. Lysyl Oxidase (LOX) Transcriptionally Regulates SNAI2 Expression and TIMP4 Secretion in Human Cancers. Clin Cancer Res 2016; 22:4491-504. [PMID: 27029493 PMCID: PMC8201910 DOI: 10.1158/1078-0432.ccr-15-2461] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2015] [Accepted: 03/24/2016] [Indexed: 01/04/2023]
Abstract
PURPOSE Epithelial-to-mesenchymal transition (EMT) is important in cancer progression and metastasis. We and others have previously reported that lysyl oxidase (LOX) is overexpressed in aggressive cancers, is associated with increased mortality, and regulates EMT. However, the mechanism by which LOX mediates EMT is unknown. In this study, we investigated the effect of LOX on mediators of EMT. EXPERIMENTAL DESIGN We used chromatin immunoprecipitation and promoter luciferase assays to determine the target gene of LOX. To determine the effects of SNAI2 in vivo, we used our metastatic anaplastic thyroid cancer (ATC) mouse model. To investigate the effects of LOX and SNAI2 on MMPs and TIMPs, protein arrays were used. Primary tumors from patients with metastatic, breast and colon cancer, and tissue array for thyroid cancer were assessed for SNAI2 and TIMP4 expression by immunohistochemistry. RESULTS We found that LOX knockdown decreases SNAI2 expression in cancer cell lines. Furthermore, knockdown of LOX reduced SNAI2 expression in a metastatic mouse model of thyroid cancer. We also demonstrated that LOX binds and transactivates the SNAI2 promoter. We found a direct correlation in thyroid and breast cancer samples between LOX and SNAI2 expression. To understand how LOX/SNAI2 axis mediates these effects, we performed a comprehensive analysis of MMPs/TIMPs. LOX and SNAI2 depletion reduced TIMP4 secretion. Analysis of SNAI2 and TIMP4 expression showed overexpression of both proteins in aggressive thyroid, colon, and breast tumors. CONCLUSIONS Our findings provide new evidence that LOX regulates SNAI2 expression and that SNAI2-mediated TIMP4 secretion plays a role in cancer progression. Clin Cancer Res; 22(17); 4491-504. ©2016 AACR.
Collapse
Affiliation(s)
- Myriem Boufraqech
- Endocrine Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Lisa Zhang
- Endocrine Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Naris Nilubol
- Endocrine Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Samira M Sadowski
- Endocrine Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Shweta Kotian
- Endocrine Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Martha Quezado
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Electron Kebebew
- Endocrine Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland.
| |
Collapse
|
78
|
Mo A, Jackson S, Varma K, Carpino A, Giardina C, Devers TJ, Rosenberg DW. Distinct Transcriptional Changes and Epithelial-Stromal Interactions Are Altered in Early-Stage Colon Cancer Development. Mol Cancer Res 2016; 14:795-804. [PMID: 27353028 DOI: 10.1158/1541-7786.mcr-16-0156] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 06/11/2016] [Indexed: 12/11/2022]
Abstract
UNLABELLED Although the progression of mutated colonic cells is dependent upon interactions between the initiated epithelium and surrounding stroma, the nature of these interactions is poorly understood. Here, the development of an ultrasensitive laser capture microdissection (LCM)/RNA-seq approach for studying the epithelial and stromal compartments of aberrant crypt foci (ACF) is described. ACF are the earliest identifiable preneoplastic lesion found within the human colon and are detected using high-definition endoscopy with contrast dye spray. The current analysis focused on the epithelium of ACF with somatic mutations to either KRAS, BRAF, or APC, and expression patterns compared with normal mucosa from each patient. By comparing gene expression patterns among groups, an increase in a number of proinflammatory NF-κB target genes was identified that was specific to ACF epithelium, including TIMP1, RELA, and RELB Distinct transcriptional changes associated with each somatic mutation were observed and a subset of ACF display BRAF(V600E)-mediated senescence-associated transcriptome characterized by increased expression of CDKN2A Finally, LCM-captured ACF-associated stroma was found to be transcriptionally distinct from normal-appearing stroma, with an upregulation of genes related to immune cell infiltration and fibroblast activation. Immunofluorescence confirmed increased CD3(+) T cells within the stromal microenvironment of ACF and an abundance of activated fibroblasts. Collectively, these results provide new insight into the cellular interplay that occurs at the earliest stages of colonic neoplasia, highlighting the important role of NF-κB, activated stromal fibroblasts, and lymphocyte infiltration. IMPLICATIONS Fibroblasts and immune cells in the stromal microenvironment play an important role during the earliest stages of colon carcinogenesis. Mol Cancer Res; 14(9); 795-804. ©2016 AACR.
Collapse
Affiliation(s)
- Allen Mo
- Center for Molecular Medicine, School of Medicine, UConn Health, Farmington, Connecticut. Colon Cancer Prevention Program, Neag Comprehensive Cancer Center, School of Medicine, UConn Health, Farmington, Connecticut
| | | | - Kamini Varma
- Thermo Fisher Scientific, South San Francisco, California
| | - Alan Carpino
- Thermo Fisher Scientific, South San Francisco, California
| | - Charles Giardina
- Department of Molecular & Cell Biology, University of Connecticut, Storrs, Connecticut
| | - Thomas J Devers
- Division of Gastroenterology, School of Medicine, UConn Health, Farmington, Connecticut
| | - Daniel W Rosenberg
- Center for Molecular Medicine, School of Medicine, UConn Health, Farmington, Connecticut. Colon Cancer Prevention Program, Neag Comprehensive Cancer Center, School of Medicine, UConn Health, Farmington, Connecticut.
| |
Collapse
|
79
|
Song T, Dou C, Jia Y, Tu K, Zheng X. TIMP-1 activated carcinoma-associated fibroblasts inhibit tumor apoptosis by activating SDF1/CXCR4 signaling in hepatocellular carcinoma. Oncotarget 2016; 6:12061-79. [PMID: 25909286 PMCID: PMC4494923 DOI: 10.18632/oncotarget.3616] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 03/14/2015] [Indexed: 12/29/2022] Open
Abstract
Tissue inhibitor of metalloproteinase 1 (TIMP-1) is an endogenous inhibitor for MMPs that regulates the remodeling and turnover of the ECM during normal development and pathological conditions. Intriguingly, recent studies have shown that TIMP-1 plays a dual role in cancer progression. In this study, we found that TIMP-1 expression in HCC tissues is associated with advanced TNM stage, intrahepatic metastasis, portal vein invasion, and vasculature invasion. Notably, TIMP-1 expression in HCC tissue is significantly related to worse overall survival for patients with HCC after liver resection. Ectopic TIMP1 expression promoted the growth of HCC xenografts in nude mice. Both co-culture with Huh7 cells with a high level of TIMP-1 and TIMP1 treatment resulted in up-regulation of hallmarks of carcinoma-associated fibroblasts (CAFs) and accelerated cell proliferation, migration and invasion in immortalized liver fibroblasts (LFs) isolated from human normal liver tissue. By co-culture with CAFs, SDF-1/CXCR4/PI3K/AKT signaling was activated and apoptosis was markedly repressed with an increased Bcl-2/BAX ratio in Huh7 cells. Taken together, our observations suggest that TIMP-1 induces the trans-differentiation of LFs into CAFs, suppresses apoptosis via SDF-1/CXCR4/PI3K/AKT signaling and then promotes HCC progression. This protein may be a potential prognostic biomarker and therapeutic target for HCC.
Collapse
Affiliation(s)
- Tao Song
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Changwei Dou
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Yuli Jia
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Kangsheng Tu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Xin Zheng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| |
Collapse
|
80
|
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: 2.4] [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
|
81
|
Chan R, Sethi P, Jyoti A, McGarry R, Upreti M. Investigating the Radioresistant Properties of Lung Cancer Stem Cells in the Context of the Tumor Microenvironment. Radiat Res 2016; 185:169-81. [PMID: 26836231 DOI: 10.1667/rr14285.1] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Lung cancer is the most common cause of cancer-related deaths worldwide and non-small cell lung cancer (NSCLC) accounts for ~85% of all lung cancer. While recent research has shown that cancer stem cells (CSC) exhibit radioresistant and chemoresistant properties, current cancer therapy targets the bulk of the tumor burden without accounting for the CSC and the contribution of the tumor microenvironment. CSC interaction with the stroma enhances NSCLC survival, thus limiting the efficacy of treatment. The aim of this study was to elucidate the role of CSC and the microenvironment in conferring radio- or chemoresistance in an in vitro tumor model for NSCLC. The novel in vitro three-dimensional (3D) NSCLC model of color-coded tumor tissue analogs (TTA) that we have developed is comprised of human lung adenocarcinoma cells, fibroblasts, endothelial cells and NSCLC cancer stem cells maintained in low oxygen conditions (5% O2) to recapitulate the physiologic conditions in tumors. Using this model, we demonstrate that a single 5 Gy radiation dose does not inhibit growth of TTA containing CSC and results in elevated expression of cytokines (TGF-α, RANTES, ENA-78) and factors (vimentin, MMP and TIMP), indicative of an invasive and aggressive phenotype. However, combined treatment of single dose or fractionated doses with cisplatin was found to either attenuate or decrease the proliferative effect that radiation exposure alone had on TTA containing CSC maintained in hypoxic conditions. In summary, we utilized a 3D NSCLC model, which had characteristics of the tumor microenvironment and tumor cell heterogeneity, to elucidate the multifactorial nature of radioresistance in tumors.
Collapse
Affiliation(s)
- Ryan Chan
- a Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky, 40536-0596; and b Department of Radiation Medicine, University of Kentucky Albert B. Chandler Hospital, Lexington, Kentucky 40536-0293
| | - Pallavi Sethi
- a Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky, 40536-0596; and b Department of Radiation Medicine, University of Kentucky Albert B. Chandler Hospital, Lexington, Kentucky 40536-0293
| | - Amar Jyoti
- a Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky, 40536-0596; and b Department of Radiation Medicine, University of Kentucky Albert B. Chandler Hospital, Lexington, Kentucky 40536-0293
| | - Ronald McGarry
- a Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky, 40536-0596; and b Department of Radiation Medicine, University of Kentucky Albert B. Chandler Hospital, Lexington, Kentucky 40536-0293
| | - Meenakshi Upreti
- a Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky, 40536-0596; and b Department of Radiation Medicine, University of Kentucky Albert B. Chandler Hospital, Lexington, Kentucky 40536-0293
| |
Collapse
|
82
|
Huynh PT, Beswick EJ, Coronado YA, Johnson P, O'Connell MR, Watts T, Singh P, Qiu S, Morris K, Powell DW, Pinchuk IV. CD90(+) stromal cells are the major source of IL-6, which supports cancer stem-like cells and inflammation in colorectal cancer. Int J Cancer 2015; 138:1971-81. [PMID: 26595254 DOI: 10.1002/ijc.29939] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Revised: 10/01/2015] [Accepted: 10/29/2015] [Indexed: 12/11/2022]
Abstract
IL-6 is a pleiotropic cytokine increased in CRC and known to directly promote tumor growth. Colonic myofibroblasts/fibroblasts (CMFs or stromal cells) are CD90(+) innate immune cells representing up to 30% of normal colonic mucosal lamina propria cells. They are expanded in CRC tumor stroma, where they also known as a cancer associated fibroblasts (CAFs). Cells of mesenchymal origin, such as normal myofibroblasts/fibroblasts, are known to secrete IL-6; however, their contribution to the increase in IL-6 in CRC and to tumor-promoting inflammation is not well defined. Using in situ, ex vivo and coculture analyses we have demonstrated that the number of IL-6 producing CMFs is increased in CRC (C-CMFs) and they represent the major source of IL-6 in T2-T3 CRC tumors. Activity/expression of stem cell markers-aldehyde dehydrogenase and LGR5- was significantly up-regulated in colon cancer cells (SW480, Caco-2 or HT29) cultured in the presence of conditioned medium from tumor isolated C-CMFs in an IL-6 dependent manner. C-CMF and its derived condition medium, but not normal CMF isolated from syngeneic normal colons, induced differentiation of tumor promoting inflammatory T helper 17 cells (Th17) cell responses in an IL-6 dependent manner. Our study suggests that CD90(+) fibroblasts/myofibroblasts may be the major source of IL-6 in T2-T3 CRC tumors, which supports the stemness of tumor cells and induces an immune adaptive inflammatory response (a.k.a. Th17) favoring tumor growth. Taken together our data supports the notion that IL-6 producing CAFs (a.k.a. C-CMFs) may provide a useful target for treating or preventing CRCs.
Collapse
Affiliation(s)
- Phuong T Huynh
- Departments of Internal Medicine, Institute of Translational Science at University of Texas Medical Branch, Galveston, TX
| | - Ellen J Beswick
- Department of Molecular Genetics, University of New Mexico, Albuquerque, NM
| | - Yun A Coronado
- Departments of Internal Medicine, Institute of Translational Science at University of Texas Medical Branch, Galveston, TX
| | - Paul Johnson
- Departments of Surgery, Institute of Translational Science at University of Texas Medical Branch, Galveston, TX
| | - Malaney R O'Connell
- Departments of Neuoroscience and Cell Biology, Institute of Translational Science at University of Texas Medical Branch, Galveston, TX
| | - Tammara Watts
- Departments of Otolaryngology, Institute of Translational Science at University of Texas Medical Branch, Galveston, TX
| | - Pomila Singh
- Departments of Neuoroscience and Cell Biology, Institute of Translational Science at University of Texas Medical Branch, Galveston, TX
| | - Suimin Qiu
- Departments of Pathology, Institute of Translational Science at University of Texas Medical Branch, Galveston, TX
| | - Katherine Morris
- Department of Surgery, University of New Mexico, Albuquerque, NM
| | - Don W Powell
- Departments of Internal Medicine, Institute of Translational Science at University of Texas Medical Branch, Galveston, TX.,Departments of Neuoroscience and Cell Biology, Institute of Translational Science at University of Texas Medical Branch, Galveston, TX.,Institute of Translational Science at University of Texas Medical Branch, Galveston, TX
| | - Irina V Pinchuk
- Departments of Internal Medicine, Institute of Translational Science at University of Texas Medical Branch, Galveston, TX.,Institute of Translational Science at University of Texas Medical Branch, Galveston, TX.,Departments of Microbiology and Immunology, Institute of Translational Science at University of Texas Medical Branch, Galveston, TX
| |
Collapse
|
83
|
LNA aptamer based multi-modal, Fe3O4-saturated lactoferrin (Fe3O4-bLf) nanocarriers for triple positive (EpCAM, CD133, CD44) colon tumor targeting and NIR, MRI and CT imaging. Biomaterials 2015; 71:84-99. [PMID: 26318819 DOI: 10.1016/j.biomaterials.2015.07.055] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2015] [Revised: 07/29/2015] [Accepted: 07/31/2015] [Indexed: 12/28/2022]
Abstract
This is the first ever attempt to combine anti-cancer therapeutic effects of emerging anticancer biodrug bovine lactoferrin (bLf), and multimodal imaging efficacy of Fe3O4 nanoparticles (NPs) together, as a saturated Fe3O4-bLf. For cancer stem cell specific uptake of nanocapsules/nanocarriers (NCs), Fe3O4-bLf was encapsulated in alginate enclosed chitosan coated calcium phosphate (AEC-CP) NCs targeted (Tar) with locked nucleic acid (LNA) modified aptamers against epithelial cell adhesion molecule (EpCAM) and nucleolin markers. The nanoformulation was fed orally to mice injected with triple positive (EpCAM, CD133, CD44) sorted colon cancer stem cells in the xenograft cancer stem cell mice model. The complete regression of tumor was observed in 70% of mice fed on non-targeted (NT) NCs, with 30% mice showing tumor recurrence after 30 days, while only 10% mice fed with Tar NCs showed tumor recurrence indicating a significantly higher survival rate. From tumor tissue analyses of 35 apoptotic markers, 55 angiogenesis markers, 40 cytokines, 15 stem cell markers and gene expression studies of important signaling molecules, it was revealed that the anti-cancer mechanism of Fe3O4-bLf was intervened through TRAIL, Fas, Fas-associated protein with death domain (FADD) mediated phosphorylation of p53, to induce activation of second mitochondria-derived activator of caspases (SMAC)/DIABLO (inhibiting survivin) and mitochondrial depolarization leading to release of cytochrome C. Induction of apoptosis was observed by inhibition of the Akt pathway and activation of cytokines released from monocytes/macrophages and dendritic cells (interleukin (IL) 27, keratinocyte chemoattractant (KC)). On the other hand, the recurrence of tumor in AEC-CP-Fe3O4-bLf NCs fed mice mainly occurred due to activation of alternative pathways such as mitogen-activated protein kinases (MAPK)/extracellular signal-regulated kinases (ERK) and Wnt signaling leading to an increase in expression of survivin, survivin splice variant (survivin 2B) and other anti-apoptotic proteins Bad, Bcl-2 and XIAP. Apart from the promising anti-cancer efficacy and the exceptional tumor targeting ability observed by multimodal imaging using near-infrared (NIR) imaging, magnetic resonance imaging (MRI) and computerized tomographic (CT) techniques, these NCs also maintained the immunomodulatory benefits of bLf as they were able to increase the RBC, hemoglobin, iron calcium and zinc levels in mice.
Collapse
|
84
|
Gupta P, Sharma PK, Mir H, Singh R, Singh N, Kloecker GH, Lillard JW, Singh S. CCR9/CCL25 expression in non-small cell lung cancer correlates with aggressive disease and mediates key steps of metastasis. Oncotarget 2015; 5:10170-9. [PMID: 25296976 PMCID: PMC4259413 DOI: 10.18632/oncotarget.2526] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Poor clinical outcome of lung cancer (LuCa) is primarily due to lack of knowledge about specific molecules involved in its progression and metastasis. In this study, we for the first time show the clinical and biological significance of CC chemokine receptor-9 (CCR9) in non-small cell lung cancer (NSCLC). Expression of CCR9 and CCL25, the only natural ligand of CCR9, was significantly higher (p < 0.0001) in NSCLC tissues and serum respectively, compared to their respective controls. Interestingly, expression of both CCR9 and CCL25 was significantly higher in adenocarcinomas (ACs) compared to squamous cell carcinomas (SCCs) (p = 0.04, and p < 0.0001). Similar to tissues, AC and SCC cell lines were positive for CCR9 expression. Despite of marginal difference in CCR9 expression, AC cells showed higher migratory and invasive potential in response to CCL25, compared to SCC cells. This differential biological response of AC cells was primarily due to differential expression of matrix metalloproteinases and tissue inhibitor of metalloproteinases under the influence of CCL25. Our results suggest CCR9 as a potential target for developing new treatment modality for NSCLC. Additionally, differential serum CCL25 level in ACs and SCCs, two NSCLC subtypes, suggest its potential as a non-invasive diagnostic/prognostic biomarker.
Collapse
Affiliation(s)
| | - Praveen K Sharma
- School of Natural Sciences, Center of Life Sciences, Central University of Jharkhand, Ranchi, India
| | - Hina Mir
- Morehouse School of Medicine, Atlanta, GA, USA
| | | | | | - Goetz H Kloecker
- James Graham Brown Cancer Center, University of Louisville, School of Medicine, Louisville, KY, USA
| | | | | |
Collapse
|
85
|
Rapamycin increases CCN2 expression of lung fibroblasts via phosphoinositide 3-kinase. J Transl Med 2015; 95:846-59. [PMID: 26192087 DOI: 10.1038/labinvest.2015.68] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2014] [Revised: 03/27/2015] [Accepted: 04/14/2015] [Indexed: 02/03/2023] Open
Abstract
Excessive production of connective tissue growth factor (CTGF, CCN2) and increased motor ability of the activated fibroblast phenotype contribute to the pathogenesis of idiopathic pulmonary fibrosis (IPF). However, molecules and signal pathways regulating CCN2 expression and migration of lung fibroblasts are still elusive. We hypothesize that rapamycin, via binding and blocking mammalian target of rapamycin (mTOR) complex (mTORC), affects CCN2 expression and migration of lung fibroblasts in vitro. Primary normal and fibrotic human lung fibroblasts were isolated from lung tissues of three patients with primary spontaneous pneumothorax and three with IPF. Cells were incubated with regular medium, or medium containing rapamycin, human recombinant transforming growth factor (TGF)-β1, or both. CCN2 and tissue inhibitor of metalloproteinase (TIMP)-1 expression in cells or supernatant was detected. Wound healing and migration assay was used to measure the migratory potential. TGF-β type I receptor (TβRI)/Smad inhibitor, SB431542 and phosphoinositide 3-kinase (PI3K) inhibitor, LY294002 were used to determine rapamycin's mechanism of action. We demonstrated that rapamycin amplified basal or TGF-β1-induced CCN2 mRNA and protein expression in normal or fibrotic fibroblasts by Smad-independent but PI3K-dependent pathway. Additionally, rapamycin also enhanced TIMP-1 expression as indicated by ELISA. However, wound healing and migrating assay showed rapamycin did not affect the mobility of fibroblasts. Collectively, this study implies a significant fibrogenic induction activity of rapamycin by activating AKT and inducing CCN2 expression in vitro and provides the possible mechanisms for the in vivo findings which previously showed no antifibrotic effect of rapamycin on lung fibrosis.
Collapse
|
86
|
Gong Y, Chippada-Venkata UD, Galsky MD, Huang J, Oh WK. Elevated circulating tissue inhibitor of metalloproteinase 1 (TIMP-1) levels are associated with neuroendocrine differentiation in castration resistant prostate cancer. Prostate 2015; 75:616-27. [PMID: 25560638 DOI: 10.1002/pros.22945] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 11/17/2014] [Indexed: 01/30/2023]
Abstract
BACKGROUND Tissue inhibitor of metalloproteinase-1 (TIMP-1) is a 28.5 kDa secreted glycoprotein that inhibits matrix metalloproteinase (MMP) activity. Our group has previously shown that elevated plasma TIMP-1 levels predict poor survival in metastatic castration-resistant prostate cancer (CRPC) patients; however, the underlying source and impact of elevated circulating TIMP-1 protein is unknown. METHODS In this study, we used qRT-PCR, ELISA and immunohistochemistry to evaluate TIMP-1 expression in androgen-sensitive and resistant prostate cancer (PC) cell lines, tumor tissues and patient sera, and to correlate TIMP-1 levels to expression of chromogranin A (CGA), an established marker of neuroendocrine differentiation (NED). We also explored the relationship between TIMP-1 overexpression and induction of NED by overexpressing TIMP-1 in androgen-sensitive LNCaP cells, as well as by inducing NED of LNCaP cells with IL-6. RESULTS Patients with CRPC have significantly higher serum TIMP-1 levels compared to patients with hormone-sensitive disease. Although circulating TIMP-1 levels were increased, peripheral blood cells were not the source of elevation. Instead, elevated TIMP-1 expression was associated with higher expression of CGA in both blood and metastatic tumor tissue. We further show that androgen receptor (AR) and PSA non-expressing prostate cancer cell lines known to display NED phenotypes such as PC-3, PC-3M, and DU145 cells, expressed high levels of TIMP-1, in contrast to AR (+) and PSA (+) adenocarcinoma cell lines such as LNCaP, VCaP, and LAPC-4, which had barely detectable levels of TIMP-1. In addition, ectopic overexpression of TIMP-1 in LNCaP cells did not induce NED. However, TIMP-1 mRNA expression was elevated >10-fold during IL-6-induced NED of LNCaP cells, suggesting that TIMP-1 overexpression accompanies, but is not the driving force for NED. Finally, we show that conditioned media from androgen-resistant PC-3, PC-3M, and DU145 cells induced TIMP-1 mRNA expression in primary prostate stromal fibroblasts in an ERK and NF-κB dependent manner. CONCLUSIONS We provide in vitro and clinical evidence to support the association between NED and elevated circulating TIMP-1 expression in CRPC. Our observation supports further evaluation of TIMP-1 as a tissue and serum biomarker for NED in CRPC.
Collapse
Affiliation(s)
- Yixuan Gong
- Division of Hematology and Medical Oncology, The Tisch Cancer Institute, Mount Sinai School of Medicine, New York, New York
| | | | | | | | | |
Collapse
|
87
|
Brunckhorst MK, Xu Y, Lu R, Yu Q. Angiopoietins promote ovarian cancer progression by establishing a procancer microenvironment. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 184:2285-96. [PMID: 25043619 DOI: 10.1016/j.ajpath.2014.05.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Revised: 04/16/2014] [Accepted: 05/06/2014] [Indexed: 02/03/2023]
Abstract
Despite decades of research, the survival rate of ovarian cancer patients is largely unchanged. Current chemotherapeutic drugs are effective only transiently because patients with advanced disease eventually develop resistance. Thus, there is a pressing need for identifying novel therapeutic targets in ovarian cancer. Mounting evidence suggests that angiopoietins (Angpts) may play an essential role in cancer progression; however, the expression profiles and biological effects of Angpts on ovarian cancer remain largely unknown. Here, we show that, compared with their normal counterparts, expressions of Angpt1, Angpt2, and Angpt4 are increased in ovarian cancer cells and tissues and that human ovarian cancer cells also express the Angpt receptor Tie-2-receptor tyrosine kinase. We show that increased expression of Angpt1, Angpt2, or Angpt4 promotes intraperitoneal growth of ovarian cancers and shortens survival of the experimental mice. We further show, for the first time, that Angpts promote accumulation of cancer-associated fibroblasts and tumor angiogenesis in the ovarian cancer microenvironment, as well as enhance ovarian cancer cell proliferation and invasion in vivo. In addition, we establish a novel function of Angpts in promoting proliferation and invasion and inducing Tie-2 and extracellular signal-regulated kinase 1/2 activation in ovarian cancer-associated fibroblasts. Taken together, these data suggest that the Angpt-Tie-2 functional axis is an important player in ovarian cancer progression and an attractive target for ovarian cancer therapy.
Collapse
Affiliation(s)
- Melissa K Brunckhorst
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Yin Xu
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Rong Lu
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Qin Yu
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York.
| |
Collapse
|
88
|
Tommelein J, Verset L, Boterberg T, Demetter P, Bracke M, De Wever O. Cancer-associated fibroblasts connect metastasis-promoting communication in colorectal cancer. Front Oncol 2015; 5:63. [PMID: 25853091 PMCID: PMC4369728 DOI: 10.3389/fonc.2015.00063] [Citation(s) in RCA: 146] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Accepted: 03/02/2015] [Indexed: 12/17/2022] Open
Abstract
Colorectal cancer (CRC) progression and eventually metastasis is directed in many aspects by a circuitous ecosystem consisting of an extracellular matrix scaffold populated by cancer-associated fibroblasts (CAFs), endothelial cells, and diverse immune cells. CAFs are recruited from local tissue-resident fibroblasts or pericryptal fibroblasts and distant fibroblast precursors. CAFs are highly abundant in CRC. In this review, we apply the metastasis-promoting communication of colorectal CAFs to 10 cancer hallmarks described by Hanahan and Weinberg. CAFs influence innate and adaptive tumor immune responses. Using datasets from previously published work, we re-explore the potential messages implicated in this process. Fibroblasts present in metastasis (metastasis-associated fibroblasts) from CRC may have other characteristics and functional roles than CAFs in the primary tumor. Since CAFs connect metastasis-promoting communication, CAF markers are potential prognostic biomarkers. CAFs and their products are possible targets for novel therapeutic strategies.
Collapse
Affiliation(s)
- Joke Tommelein
- Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University , Ghent , Belgium
| | - Laurine Verset
- Department of Pathology, Erasme University Hospital, Université Libre de Bruxelles , Brussels , Belgium
| | - Tom Boterberg
- Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University , Ghent , Belgium
| | - Pieter Demetter
- Department of Pathology, Erasme University Hospital, Université Libre de Bruxelles , Brussels , Belgium
| | - Marc Bracke
- Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University , Ghent , Belgium
| | - Olivier De Wever
- Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University , Ghent , Belgium
| |
Collapse
|
89
|
Takawale A, Sakamuri SS, Kassiri Z. Extracellular Matrix Communication and Turnover in Cardiac Physiology and Pathology. Compr Physiol 2015; 5:687-719. [DOI: 10.1002/cphy.c140045] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
90
|
Down-regulation of TIMP-1 inhibits cell migration, invasion, and metastatic colonization in lung adenocarcinoma. Tumour Biol 2015; 36:3957-67. [PMID: 25578494 DOI: 10.1007/s13277-015-3039-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Accepted: 01/02/2015] [Indexed: 10/24/2022] Open
Abstract
Tissue inhibitor metalloproteinase-1 (TIMP-1) is clinically associated with a poor prognosis for various cancers, but the roles of TIMP-1 in lung cancer metastasis are controversial. Our previous secretomic study revealed that TIMP-1 is highly abundant in high invasiveness cells of lung adenocarcinoma. In the current study, TIMP-1 abundances in primary lung adenocarcinoma tissues, as revealed by immunohistochemistry, are significantly higher in patients with lymph invasion and distant metastasis than in those without. Receiver operating characteristic curve analyses suggest 73.7 and 86.2 % accuracy to separate patients with lymph node and distant metastasis and those without, respectively. Moreover, we demonstrate that the expression level of TIMP-1 positively associates with cell mobility, invasiveness, and metastatic colonization. Most notably, the novel mechanism in which TIMP-1 facilitates metastatic colonization through the mediation of pericellular polyFN1 assembly was revealed. In summary, this study presents novel functions of TIMP-1 in promoting cancer metastasis and suggests TIMP-1 is a potential tissue biomarker for lymph invasion and distant metastasis of lung adenocarcinoma.
Collapse
|
91
|
Vinther L, Lademann U, Andersen EV, Højrup P, Thaysen-Andersen M, Krogh BO, Viuff B, Brünner N, Stenvang J, Moreira JMA. Purification and characterization of bioactive his6-tagged recombinant human tissue inhibitor of metalloproteinases-1 (TIMP-1) protein expressed at high yields in mammalian cells. Protein Expr Purif 2014; 101:157-64. [PMID: 24998777 DOI: 10.1016/j.pep.2014.06.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Revised: 06/18/2014] [Accepted: 06/23/2014] [Indexed: 12/15/2022]
Abstract
Tissue inhibitor of metalloproteinases-1 (TIMP-1) is an endogenous inhibitor of matrix metalloproteinases (MMPs) with reported tumor promoting, as well as inhibitory, effects. These paradoxical properties are presumably mediated by different biological functions, MMP-dependent as well as -independent, and probably related to TIMP-1 levels of protein expression, post-translational modifications, and cellular localization. TIMP-1 is an N-glycosylated protein that folds into two functional domains, a C- and an N-terminal domain, with six disulfide bonds. Furthermore, TIMP-1 is processed in the N-terminal sequence. These three biochemical properties make TIMP-1 difficult to produce in conventional bacterial, insect, or yeast expression systems. We describe here a HEK293 cell-based strategy for production and purification of secreted and N-glycosylated recombinant his6-tagged human TIMP-1 (his6-rTIMP-1), which resulted in large amounts of highly purified and bioactive protein. Matrix-assisted laser desorption ionization mass spectrometry confirmed the N- and C-termini of his6-rTIMP-1, and N-glycosylation profiling showed a match to the N-glycosylation of human plasma TIMP-1. The his6-rTIMP-1 was bioactive as shown by its proper inhibitory effect on MMP-2 activity, and its stimulatory effect on cell growth when added to the growth medium of four different breast cancer cell lines. This study provides an easy set-up for large scale production and purification of bioactive, tagged recombinant human TIMP-1, which structurally and functionally is similar to endogenous human TIMP-1, while using an expression system that is adaptable to most biochemical and biomedical laboratories including those that do not perform protein purifications routinely.
Collapse
Affiliation(s)
- Lena Vinther
- Section for Molecular Disease Biology and Sino-Danish Breast Cancer Research Centre, Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Ulrik Lademann
- Section for Molecular Disease Biology and Sino-Danish Breast Cancer Research Centre, Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Elisabeth Veyhe Andersen
- Section for Molecular Disease Biology and Sino-Danish Breast Cancer Research Centre, Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Peter Højrup
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
| | - Morten Thaysen-Andersen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
| | | | - Birgitte Viuff
- Section for Molecular Disease Biology and Sino-Danish Breast Cancer Research Centre, Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Nils Brünner
- Section for Molecular Disease Biology and Sino-Danish Breast Cancer Research Centre, Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Jan Stenvang
- Section for Molecular Disease Biology and Sino-Danish Breast Cancer Research Centre, Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - José M A Moreira
- Section for Molecular Disease Biology and Sino-Danish Breast Cancer Research Centre, Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark.
| |
Collapse
|
92
|
Gong Y, Chippada-Venkata UD, Oh WK. Roles of matrix metalloproteinases and their natural inhibitors in prostate cancer progression. Cancers (Basel) 2014; 6:1298-327. [PMID: 24978435 PMCID: PMC4190542 DOI: 10.3390/cancers6031298] [Citation(s) in RCA: 118] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Revised: 05/31/2014] [Accepted: 06/09/2014] [Indexed: 01/16/2023] Open
Abstract
Matrix metalloproteinases (MMPs), a group of zinc-dependent endopeptidases involved in the degradation of the extracellular matrix, play an important role in tissue remodeling associated with various physiological processes such as morphogenesis, angiogenesis, and tissue repair, as well as pathological processes including cirrhosis, arthritis and cancer. The MMPs are well established as mediators of tumor invasion and metastasis by breaking down connective tissue barriers. Although there has been a vast amount of literature on the role of MMPs in invasion, metastasis and angiogenesis of various cancers, the role of these endopeptidases in prostate cancer progression has not been systematically reviewed. This overview summarizes findings on the tissue and blood expression of MMPs, their function, regulation and prognostic implication in human prostate cancer, with a focus on MMP-2, -7, -9, MT1-MMP and tissue inhibitor of metalloproteinase 1 (TIMP-1). This review also summarizes the efficacy and failure of early-generation matrix metalloproteinase inhibitors (MMPIs) in the treatment of metastatic prostate cancer and highlights the lessons and challenges for next generation MMPIs.
Collapse
Affiliation(s)
- Yixuan Gong
- Division of Hematology and Medical Oncology, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Uma D Chippada-Venkata
- Division of Hematology and Medical Oncology, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - William K Oh
- Division of Hematology and Medical Oncology, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
93
|
Spinale FG, Villarreal F. Targeting matrix metalloproteinases in heart disease: lessons from endogenous inhibitors. Biochem Pharmacol 2014; 90:7-15. [PMID: 24780447 DOI: 10.1016/j.bcp.2014.04.011] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Revised: 04/16/2014] [Accepted: 04/17/2014] [Indexed: 01/13/2023]
Abstract
Basic pharmacological/transgenic studies have clearly demonstrated a cause-effect relationship between the induction and activation of matrix metalloproteinases (MMPs) and adverse changes in the structure and function of the left ventricle (LV). Thus, regulation of MMP induction and/or activation would appear to be a potential therapeutic target in the context of cardiovascular disease, such as following myocardial infarction (MI). However, pharmacological approaches to inhibit MMPs have yet to be realized for clinical applications. The endogenous inhibitors of the MMPs (TIMPs) constitute a set of 4 small molecules with unique functionality and specificity. Thus, improved understanding on the function and roles of individual TIMPs may provide important insight into the design and targets for pharmacological applications in LV remodeling processes, such as MI. Therefore, the purpose of this review will be to briefly examine biological functions and relevance of the individual TIMPs in terms of adverse LV remodeling post-MI. Second is to examine the past outcomes and issues surrounding clinical trials targeting MMPs in the post MI context and how new insights into TIMP biology may provide new pharmacological targets. This review will put forward the case that initial pharmacological attempts at MMP inhibition were over-simplistic and that future strategies must recognize the diversity of this matrix proteolytic system and that lessons from TIMP biology may lead to future therapeutic strategies.
Collapse
Affiliation(s)
- Francis G Spinale
- Cardiovascular Translational Research Center, University of South Carolina School of Medicine, Columbia, SC, USA; Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, USA; WJB Dorn Veteran Affairs Medical Center, Columbia, SC, USA.
| | - Francisco Villarreal
- Division of Cardiology, University of California-San Diego School of Medicine, La Jolla, CA, USA
| |
Collapse
|