1
|
Suenaga M, Mashima T, Kawata N, Dan S, Seimiya H, Yamaguchi K. Exploratory Study Identifies Matrix Metalloproteinase-14 and -9 as Potential Biomarkers of Regorafenib Efficacy in Metastatic Colorectal Cancer. Cancers (Basel) 2024; 16:2855. [PMID: 39199626 PMCID: PMC11352555 DOI: 10.3390/cancers16162855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 08/11/2024] [Accepted: 08/14/2024] [Indexed: 09/01/2024] Open
Abstract
In identifying biomarkers for anticancer drugs, the lack of objectivity in selecting candidate factors makes interpretation difficult. We performed preclinical analysis and a translational validation study to identify candidate biomarkers for regorafenib efficacy in metastatic colorectal cancer (mCRC). Using in silico COMPARE analysis with a human cancer cell line panel, JFCR39, we selected candidate biomarkers whose expression correlates with regorafenib sensitivity. We validated predictive values in mCRC patients receiving regorafenib (discovery, n = 53) and FTD/TPI (control, n = 16). Blood samples were obtained at baseline (BL), before the second cycle (2nd), and at progressive disease (PD), and biomarker levels were measured using ELISA. Our analysis showed that high matrix metalloproteinase (MMP)-14 expression was associated with a high sensitivity to regorafenib. In the discovery cohort, high MMP-14 levels at BL and PD were correlated with tumor shrinkage and longer progression-free survival (PFS). A subsequent analysis of other related factors further indicated that the patients with decreased MMP-9 levels at the 2nd had higher disease control rates, tumor shrinkage, longer PFS, and overall survival than those with increased changes. These findings were not observed in the control cohort. Our study suggests MMP-14 and MMP-9 may serve as prognostic markers for regorafenib and provide insights into novel combination therapies with anti-MMP-9 agents or FTD/TPI.
Collapse
Affiliation(s)
- Mitsukuni Suenaga
- Gastroenterology Center, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo 135-8550, Japan; (N.K.); (K.Y.)
- Department of Clinical Oncology, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8519, Japan
| | - Tetsuo Mashima
- Division of Molecular Biotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo 135-8550, Japan; (T.M.); (H.S.)
| | - Naomi Kawata
- Gastroenterology Center, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo 135-8550, Japan; (N.K.); (K.Y.)
- Division of Molecular Biotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo 135-8550, Japan; (T.M.); (H.S.)
| | - Shingo Dan
- Division of Molecular Pharmacology, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo 135-8550, Japan;
| | - Hiroyuki Seimiya
- Division of Molecular Biotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo 135-8550, Japan; (T.M.); (H.S.)
| | - Kensei Yamaguchi
- Gastroenterology Center, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo 135-8550, Japan; (N.K.); (K.Y.)
| |
Collapse
|
2
|
Ferre-Torres J, Noguera-Monteagudo A, Lopez-Canosa A, Romero-Arias JR, Barrio R, Castaño O, Hernandez-Machado A. Modelling of chemotactic sprouting endothelial cells through an extracellular matrix. Front Bioeng Biotechnol 2023; 11:1145550. [PMID: 37362221 PMCID: PMC10285466 DOI: 10.3389/fbioe.2023.1145550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 05/26/2023] [Indexed: 06/28/2023] Open
Abstract
Sprouting angiogenesis is a core biological process critical to vascular development. Its accurate simulation, relevant to multiple facets of human health, is of broad, interdisciplinary appeal. This study presents an in-silico model replicating a microfluidic assay where endothelial cells sprout into a biomimetic extracellular matrix, specifically, a large-pore, low-concentration fibrin-based porous hydrogel, influenced by chemotactic factors. We introduce a novel approach by incorporating the extracellular matrix and chemotactic factor effects into a unified term using a single parameter, primarily focusing on modelling sprouting dynamics and morphology. This continuous model naturally describes chemotactic-induced sprouting with no need for additional rules. In addition, we extended our base model to account for matrix sensing and degradation, crucial aspects of angiogenesis. We validate our model via a hybrid in-silico experimental method, comparing the model predictions with experimental results derived from the microfluidic setup. Our results underscore the intricate relationship between the extracellular matrix structure and angiogenic sprouting, proposing a promising method for predicting the influence of the extracellular matrix on angiogenesis.
Collapse
Affiliation(s)
- Josep Ferre-Torres
- Department of Condensed Matter Physics, University of Barcelona (UB), Barcelona, Spain
| | | | - Adrian Lopez-Canosa
- Electronics and Biomedical Engineering, University of Barcelona (UB), Barcelona, Spain
- Biomaterials for Regenerative Therapies, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Spain
| | - J Roberto Romero-Arias
- Institute for Research in Applied Mathematics and Systems, National Autonomous University of Mexico , Mexico City, Mexico
| | - Rafael Barrio
- Institute of Physics, National Autonomous University of Mexico, Mexico City, Mexico
| | - Oscar Castaño
- Electronics and Biomedical Engineering, University of Barcelona (UB), Barcelona, Spain
- Biomaterials for Regenerative Therapies, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Spain
- Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona (UB), Barcelona, Spain
| | - Aurora Hernandez-Machado
- Department of Condensed Matter Physics, University of Barcelona (UB), Barcelona, Spain
- Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona (UB), Barcelona, Spain
| |
Collapse
|
3
|
Veschi V, Turdo A, Modica C, Verona F, Di Franco S, Gaggianesi M, Tirrò E, Di Bella S, Iacono ML, Pantina VD, Porcelli G, Mangiapane LR, Bianca P, Rizzo A, Sciacca E, Pillitteri I, Vella V, Belfiore A, Bongiorno MR, Pistone G, Memeo L, Colarossi L, Giuffrida D, Colarossi C, Vigneri P, Todaro M, Stassi G. Recapitulating thyroid cancer histotypes through engineering embryonic stem cells. Nat Commun 2023; 14:1351. [PMID: 36906579 PMCID: PMC10008571 DOI: 10.1038/s41467-023-36922-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 02/21/2023] [Indexed: 03/13/2023] Open
Abstract
Thyroid carcinoma (TC) is the most common malignancy of endocrine organs. The cell subpopulation in the lineage hierarchy that serves as cell of origin for the different TC histotypes is unknown. Human embryonic stem cells (hESCs) with appropriate in vitro stimulation undergo sequential differentiation into thyroid progenitor cells (TPCs-day 22), which maturate into thyrocytes (day 30). Here, we create follicular cell-derived TCs of all the different histotypes based on specific genomic alterations delivered by CRISPR-Cas9 in hESC-derived TPCs. Specifically, TPCs harboring BRAFV600E or NRASQ61R mutations generate papillary or follicular TC, respectively, whereas addition of TP53R248Q generate undifferentiated TCs. Of note, TCs arise by engineering TPCs, whereas mature thyrocytes have a very limited tumorigenic capacity. The same mutations result in teratocarcinomas when delivered in early differentiating hESCs. Tissue Inhibitor of Metalloproteinase 1 (TIMP1)/Matrix metallopeptidase 9 (MMP9)/Cluster of differentiation 44 (CD44) ternary complex, in cooperation with Kisspeptin receptor (KISS1R), is involved in TC initiation and progression. Increasing radioiodine uptake, KISS1R and TIMP1 targeting may represent a therapeutic adjuvant option for undifferentiated TCs.
Collapse
Affiliation(s)
- Veronica Veschi
- Department of Surgical Oncological and Stomatological Sciences, University of Palermo, Palermo, Italy
| | - Alice Turdo
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Chiara Modica
- Department of Surgical Oncological and Stomatological Sciences, University of Palermo, Palermo, Italy
| | - Francesco Verona
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Simone Di Franco
- Department of Surgical Oncological and Stomatological Sciences, University of Palermo, Palermo, Italy
| | - Miriam Gaggianesi
- Department of Surgical Oncological and Stomatological Sciences, University of Palermo, Palermo, Italy
| | - Elena Tirrò
- Department of Surgical Oncological and Stomatological Sciences, University of Palermo, Palermo, Italy.,Department of Clinical and Experimental Medicine, A.O.U. Policlinico-Vittorio Emanuele, Center of Experimental Oncology and Hematology, University of Catania, Catania, Italy
| | - Sebastiano Di Bella
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Melania Lo Iacono
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Vincenzo Davide Pantina
- Department of Surgical Oncological and Stomatological Sciences, University of Palermo, Palermo, Italy
| | - Gaetana Porcelli
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Laura Rosa Mangiapane
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Paola Bianca
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | | | - Elisabetta Sciacca
- Queen Mary University, Experimental Medicine & Rheumatology, London, United Kingdom
| | - Irene Pillitteri
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Veronica Vella
- Endocrinology Unit, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Hospital, Catania, Italy
| | - Antonino Belfiore
- Endocrinology Unit, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Hospital, Catania, Italy
| | - Maria Rita Bongiorno
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Giuseppe Pistone
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Lorenzo Memeo
- Department of Experimental Oncology, Mediterranean Institute of Oncology, Viagrande, Catania, Italy
| | - Lorenzo Colarossi
- Department of Experimental Oncology, Mediterranean Institute of Oncology, Viagrande, Catania, Italy
| | - Dario Giuffrida
- Department of Experimental Oncology, Mediterranean Institute of Oncology, Viagrande, Catania, Italy
| | - Cristina Colarossi
- Department of Experimental Oncology, Mediterranean Institute of Oncology, Viagrande, Catania, Italy
| | - Paolo Vigneri
- Department of Clinical and Experimental Medicine, A.O.U. Policlinico-Vittorio Emanuele, Center of Experimental Oncology and Hematology, University of Catania, Catania, Italy
| | - Matilde Todaro
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy.,A.O.U.P. "Paolo Giaccone", University of Palermo, Palermo, Italy
| | - Giorgio Stassi
- Department of Surgical Oncological and Stomatological Sciences, University of Palermo, Palermo, Italy.
| |
Collapse
|
4
|
Synthesis of a Dual-Color Fluorescent Dendrimer for Diagnosis of Cancer Metastasis in Lymph Nodes. Polymers (Basel) 2022; 14:polym14204314. [PMID: 36297891 PMCID: PMC9607438 DOI: 10.3390/polym14204314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 10/02/2022] [Accepted: 10/11/2022] [Indexed: 11/10/2022] Open
Abstract
Detection of cancer metastasis spread in lymph nodes is important in cancer diagnosis. In this study, a fluorescence imaging probe was designed for the detection of both lymph node and tumor cells using always-ON and activatable fluorescence probes with different colors. Rhodamine B (Rho), a matrix metalloproteinase-2 (MMP-2)-responsive green fluorescence probe, and a tumor-homing peptide were conjugated to a carboxy-terminal dendrimer that readily accumulates in lymph nodes. The activatable green fluorescence signal increased in the presence of MMP-2, which is secreted by tumor cells. Both the always-ON Rho signal and the activatable green fluorescence signal were observed from tumor cells, but only the weak always-ON Rho signal was from immune cells. Thus, this type of dendrimer may be useful for non-invasive imaging to diagnose cancer metastasis in lymph nodes.
Collapse
|
5
|
Kümper M, Zamek J, Steinkamp J, Pach E, Mauch C, Zigrino P. Role of MMP3 and fibroblast-MMP14 in skin homeostasis and repair. Eur J Cell Biol 2022; 101:151276. [PMID: 36162272 DOI: 10.1016/j.ejcb.2022.151276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 09/09/2022] [Accepted: 09/21/2022] [Indexed: 12/14/2022] Open
Abstract
Early lethality of mice with complete deletion of the matrix metalloproteinase MMP14 emphasized the proteases' pleiotropic functions. MMP14 deletion in adult dermal fibroblasts (MMP14Sf-/-) caused collagen type I accumulation and upregulation of MMP3 expression. To identify the compensatory role of MMP3, mice were generated with MMP3 deletion in addition to MMP14 loss in fibroblasts. These double deficient mice displayed a fibrotic phenotype in skin and tendons as detected in MMP14Sf-/- mice, but no additional obvious defects were detected. However, challenging the mice with full thickness excision wounds resulted in delayed closure of early wounds in the double deficient mice compared to wildtype and MMP14 single knockout controls. Over time wounds closed and epidermal integrity was restored. Interestingly, on day seven, post-wounding myofibroblast density was lower in the wounds of all knockout than in controls, they were higher on day 14. The delayed resolution of myofibroblasts from the granulation tissue is paralleled by reduced apoptosis of these cells, although proliferation of myofibroblasts is induced in the double deficient mice. Further analysis showed comparable TGFβ1 and TGFβR1 expression among all genotypes. In addition, in vitro, fibroblasts lacking MMP3 and MMP14 retained their ability to differentiate into myofibroblasts in response to TGFβ1 treatment and mechanical stress. However, in vivo, p-Smad2 was reduced in myofibroblasts at day 5 post-wounding, in double, but most significant in single knockout, indicating their involvement in TGFβ1 activation. Thus, although MMP3 does not compensate for the lack of fibroblast-MMP14 in tissue homeostasis, simultaneous deletion of both proteases in fibroblasts delays wound closure during skin repair. Notably, single and double deficiency of these proteases modulates myofibroblast formation and resolution in wounds.
Collapse
Affiliation(s)
- Maike Kümper
- Department of Dermatology and Venereology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany.
| | - Jan Zamek
- Department of Dermatology and Venereology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany.
| | - Joy Steinkamp
- Department of Dermatology and Venereology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany.
| | - Elke Pach
- Department of Dermatology and Venereology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany.
| | - Cornelia Mauch
- Department of Dermatology and Venereology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany.
| | - Paola Zigrino
- Department of Dermatology and Venereology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany.
| |
Collapse
|
6
|
Matsushige C, Xu X, Miyagi M, Zuo YY, Yamazaki Y. RGD-modified dextran hydrogel promotes follicle growth in three-dimensional ovarian tissue culture in mice. Theriogenology 2022; 183:120-131. [PMID: 35247849 PMCID: PMC9005264 DOI: 10.1016/j.theriogenology.2022.02.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 02/06/2022] [Accepted: 02/07/2022] [Indexed: 10/19/2022]
Abstract
In vitro follicle growth is a promising technology to preserve fertility for cancer patients. We previously developed a three-dimensional (3-D) ovarian tissue culture system supported by mouse tumor cell-derived Matrigel. When murine ovarian tissues at 14 days old were cultured in Matrigel drops, antrum formation and oocyte competence were significantly enhanced compared with those cultured without Matrigel. In this study, we tested whether nonanimal-derived dextran hydrogels can support a 3-D ovarian tissue culture. We employed chemically defined dextran hydrogels consisting of dextran polymers crosslinked with polyethylene glycol (PEG)-based cell-degradable crosslinker. To determine the optimal gel elasticity for the 3-D tissue culture, we measured Young's modulus of dextran hydrogels at four concentrations (1.75, 2.25, 2.75, and 3.25 mmol/L), and cultured ovarian tissues in these gels for 7 days. As a result, 2.25 mmol/L dextran hydrogel with Young's modulus of 224 Pa was appropriate to provide physical support as well as to promote follicle expansion in the 3-D system. To mimic the natural extracellular matrix (ECM) environment, we modified the dextran hydrogels with two bioactive factors: ECM-derived Arg-Gly-Asp (RGD) peptides as a cell-adhesive factor, and activin A. The ovarian tissues were cultured in 2.25 mmol/L dextran hydrogels under four different conditions: Activin-/RGD- (A-R-), A + R-, A-R+, and A + R+. On Day 7 of culture, follicle and oocyte sizes were significantly increased in the RGD-modified conditions compared with those without RGD. The RGD-modified hydrogels also promoted mRNA levels of steroidogenic-related genes and estradiol production in the 3-D ovarian tissue culture. In vitro maturation and developmental competence of follicular oocytes were remarkably improved in the presence of RGD. In particular, blastocyst embryos were obtained only from A-R+ or A+R+ conditions after in vitro fertilization. We also determined synergistic effects of the RGD peptides and activin A on follicle growth and oocyte development in the 3-D tissue culture. In conclusion, our results suggest that RGD-modified dextran hydrogels provide an ECM-mimetic bioactive environment to support folliculogenesis in a 3-D ovarian tissue culture system.
Collapse
|
7
|
Ding LX, Zhang J, Yang SS, Wu J, Su T, Wang WM. Heat Shock Proteins 70 Regulate Cell Motility and Invadopodia-Associated Proteins Expression in Oral Squamous Cell Carcinoma. Front Endocrinol (Lausanne) 2022; 13:890218. [PMID: 35957827 PMCID: PMC9362981 DOI: 10.3389/fendo.2022.890218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 06/20/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Many studies have shown that diabetes is often closely related to oral squamous cell carcinoma (OSCC) occurrence and metastasis. Heat shock protein 70 (Hsp70) is a molecular chaperone related to diabetes complications. This study aims to investigate the role of Hsp70 in OSCC in expression of invadopodia-associated proteins. METHODS The expressions and correlation of HSP70, Hif1α, MMP2, MMP14, and cortactin were examined using bioinformatics analysis and verified by OSCC tissue microarrays. Assay in vitro was performed to analyze cell migration capacity after treatment with or without the HSP70 inhibitor. RESULTS The expressions of invadopodia-associated proteins were enhanced in OSCC tissues compared with paracarcinoma tissues and partially correlated with HSP70. Inhibiting HSP70 significantly decreased the cell viability, proliferation, and migration of OSCC cells. CONCLUSIONS HSP70 may be involved in invadopodia-associated proteins in OSCC cells, which provides a promising method for treatment of OSCC metastasis.
Collapse
Affiliation(s)
- Le-Xi Ding
- Eye Center of Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China
- Hunan Key Laboratory of Ophthalmology, Central South University, Changsha, China
| | - Jing Zhang
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China
- Institute of Oral Precancerous Lesions, Central South University, Changsha, China
| | - Si-Si Yang
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China
- Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, China
| | - Jin Wu
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China
- Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, China
| | - Tong Su
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China
| | - Wei-Ming Wang
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Wei-Ming Wang,
| |
Collapse
|
8
|
CD147 mediates the CD44s-dependent differentiation of myofibroblasts driven by transforming growth factor-β 1. J Biol Chem 2021; 297:100987. [PMID: 34364871 PMCID: PMC8405944 DOI: 10.1016/j.jbc.2021.100987] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 06/16/2021] [Accepted: 07/15/2021] [Indexed: 11/22/2022] Open
Abstract
Progressive fibrosis leads to loss of organ function and affects many organs as a result of excessive extracellular matrix production. The ubiquitous matrix polysaccharide hyaluronan (HA) is central to this through association with its primary receptor, CD44, which exists as standard CD44 (CD44s) or multiple splice variants. Mediators such as profibrotic transforming growth factor (TGF)-β1 and proinflammatory interleukin (IL)-1β are widely associated with fibrotic progression. TGF-β1 induces myofibroblast differentiation, while IL-1β induces a proinflammatory fibroblast phenotype that promotes fibroblast binding to monocyte/macrophages. CD44 expression is essential for both responses. Potential CD44 splice variants involved, however, are unidentified. The TGF-β1-activated CD44/epidermal growth factor receptor complex induces differentiation of metastatic cells through interactions with the matrix metalloproteinase inducer, CD147. This study aimed to determine the CD44 variants involved in TGF-β1- and IL-1β-mediated responses and to investigate the potential profibrotic role of CD147. Using immunocytochemistry and quantitative PCR, standard CD44s were shown to be essential for both TGF-β1-induced fibroblast/myofibroblast differentiation and IL-1β-induced monocyte binding. Co-immunoprecipitation identified that CD147 associated with CD44s. Using CD147-siRNA and confocal microscopy, we also determined that incorporation of the myofibroblast marker, αSMA, into F-actin stress fibers was prevented in the absence of CD147 and myofibroblast-dependent collagen gel contraction was inhibited. CD147 did not associate with HA, but removal of HA prevented the association of CD44s with CD147 at points of cell–cell contact. Taken together, our data suggest that CD44s/CD147 colocalization is essential in regulating the mechanical tension required for the αSMA incorporation into F-actin stress fibers that regulates myofibroblast phenotype.
Collapse
|
9
|
Mechanical Intermittent Compression Affects the Progression Rate of Malignant Melanoma Cells in a Cycle Period-Dependent Manner. Diagnostics (Basel) 2021; 11:diagnostics11061112. [PMID: 34207144 PMCID: PMC8234529 DOI: 10.3390/diagnostics11061112] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/09/2021] [Accepted: 06/16/2021] [Indexed: 12/31/2022] Open
Abstract
Static mechanical compression is a biomechanical factor that affects the progression of melanoma cells. However, little is known about how dynamic mechanical compression affects the progression of melanoma cells. In the present study, we show that mechanical intermittent compression affects the progression rate of malignant melanoma cells in a cycle period-dependent manner. Our results suggest that intermittent compression with a cycle of 2 h on/2 h off could suppress the progression rate of melanoma cells by suppressing the elongation of F-actin filaments and mRNA expression levels related to collagen degradation. In contrast, intermittent compression with a cycle of 4 h on/4 h off could promote the progression rate of melanoma cells by promoting cell proliferation and mRNA expression levels related to collagen degradation. Mechanical intermittent compression could therefore affect the progression rate of malignant melanoma cells in a cycle period-dependent manner. Our results contribute to a deeper understanding of the physiological responses of melanoma cells to dynamic mechanical compression.
Collapse
|
10
|
Nagai K, Sato T, Kojima C. Design of a dendrimer with a matrix metalloproteinase-responsive fluorescence probe and a tumor-homing peptide for metastatic tumor cell imaging in the lymph node. Bioorg Med Chem Lett 2021; 33:127726. [PMID: 33316406 DOI: 10.1016/j.bmcl.2020.127726] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 11/18/2020] [Accepted: 11/27/2020] [Indexed: 02/01/2023]
Abstract
Fluorescence imaging is a noninvasive technique for cancer diagnosis. Dendrimers are regularly branched macromolecules with highly controllable size and structure that are a potent multifunctional nanoparticle. Anionic-terminal polyamidoamine (PAMAM) dendrimers were previously found to be accumulated in the lymph node, which is one of the main routes of tumor metastasis. In this study, we designed and synthesized a dendrimeric imaging probe for lymph node-resident tumor cell imaging. A matrix metalloproteinase-2 (MMP-2)-responsive fluorescence peptide probe and a tumor-homing peptide were conjugated to the carboxy-terminal dendrimer. The dendrimeric imaging probe treatment showed fluorescence signals inside some tumor cells (e.g., human fibrosarcoma HT-1080 and breast cancer 4T1 cells), depending on the MMP activity, but not in macrophage-like RAW264 cells.
Collapse
Affiliation(s)
- Kento Nagai
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Prefecture University, 1-2 Gakuen-cho, Naka-ku, Sakai, Osaka 599-8570, Japan
| | - Tatsumi Sato
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Prefecture University, 1-2 Gakuen-cho, Naka-ku, Sakai, Osaka 599-8570, Japan
| | - Chie Kojima
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Prefecture University, 1-2 Gakuen-cho, Naka-ku, Sakai, Osaka 599-8570, Japan.
| |
Collapse
|
11
|
Roadmap for Stroke: Challenging the Role of the Neuronal Extracellular Matrix. Int J Mol Sci 2020; 21:ijms21207554. [PMID: 33066304 PMCID: PMC7589675 DOI: 10.3390/ijms21207554] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 10/02/2020] [Accepted: 10/08/2020] [Indexed: 01/03/2023] Open
Abstract
Stroke is a major challenge in modern medicine and understanding the role of the neuronal extracellular matrix (NECM) in its pathophysiology is fundamental for promoting brain repair. Currently, stroke research is focused on the neurovascular unit (NVU). Impairment of the NVU leads to neuronal loss through post-ischemic and reperfusion injuries, as well as coagulatory and inflammatory processes. The ictal core is produced in a few minutes by the high metabolic demand of the central nervous system. Uncontrolled or prolonged inflammatory response is characterized by leukocyte infiltration of the injured site that is limited by astroglial reaction. The metabolic failure reshapes the NECM through matrix metalloproteinases (MMPs) and novel deposition of structural proteins continues within months of the acute event. These maladaptive reparative processes are responsible for the neurological clinical phenotype. In this review, we aim to provide a systems biology approach to stroke pathophysiology, relating the injury to the NVU with the pervasive metabolic failure, inflammatory response and modifications of the NECM. The available data will be used to build a protein–protein interaction (PPI) map starting with 38 proteins involved in stroke pathophysiology, taking into account the timeline of damage and the co-expression scores of their RNA patterns The application of the proposed network could lead to a more accurate design of translational experiments aiming at improving both the therapy and the rehabilitation processes.
Collapse
|
12
|
Park SS, Lee DH, Lee SM, Lee CH, Kim SY. Single-sided Deafness Leads to Changes in Vesicular Synaptic Transporters and Matrix Metalloproteinase 9 in the Primary Auditory Cortex. Neuroscience 2020; 449:189-201. [PMID: 32976983 DOI: 10.1016/j.neuroscience.2020.09.032] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 09/07/2020] [Accepted: 09/12/2020] [Indexed: 12/28/2022]
Abstract
Changes in perineuronal nets (PNNs) after hearing loss were described in previous studies. The present study aimed to examine how single-sided deafness (SSD) affects the expression of excitatory and inhibitory synaptic transporters and PNNs in the primary auditory cortex (A1). Sprague-Dawley rats (8-week-old females, n = 30) were divided into three groups: (1) the SSD 2-week group (n = 10), (2) the SSD 4-week group (n = 10), and (3) the 4-week control group (n = 10). The expression levels of vesicular glutamate transporter 1 (VGLUT1), VGLUT2, vesicular GABA transporter (VGAT), and genes related to PNNs were measured using quantitative reverse transcription-polymerase chain reaction. The A1 was immunostained for VGLUT1, glutamate acid decarboxylase (GAD) 67, neurocan, aggrecan, brevican, and Wisteria floribunda agglutinin (WFA). The expression levels of VGLUT1, VGLUT2, and VGAT were elevated in the A1 on the ipsilateral side in the SSD groups compared with those in the control groups. Aggrecan expression was elevated in the A1 on the contralateral side in the SSD 2-week group. The SSD groups had elevated expression levels of metalloproteinase (MMP) 9 on the contralateral side. The presynaptic glutamatergic and GABAergic transporters were increased in the A1 on the ipsilateral side after induction of SSD. Changes in the cortical auditory nervous system accompanied changes in the PNNs and their degradation enzymes MMP9 and MMP14.
Collapse
Affiliation(s)
- Sung-Su Park
- Department of Otorhinolaryngology, CHA University College of Medicine, Republic of Korea
| | - Da-Hye Lee
- Department of Otorhinolaryngology, CHA University College of Medicine, Republic of Korea
| | - So Min Lee
- Department of Otorhinolaryngology, CHA University College of Medicine, Republic of Korea
| | - Chang Ho Lee
- Department of Otorhinolaryngology, CHA University College of Medicine, Republic of Korea
| | - So Young Kim
- Department of Otorhinolaryngology, CHA University College of Medicine, Republic of Korea.
| |
Collapse
|
13
|
Kallikrein-Related Peptidase 14 Activates Zymogens of Membrane Type Matrix Metalloproteinases (MT-MMPs)-A CleavEx Based Analysis. Int J Mol Sci 2020; 21:ijms21124383. [PMID: 32575583 PMCID: PMC7352328 DOI: 10.3390/ijms21124383] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 06/09/2020] [Accepted: 06/17/2020] [Indexed: 01/02/2023] Open
Abstract
Kallikrein-related peptidases (KLKs) and matrix metalloproteinases (MMPs) are secretory proteinases known to proteolytically process components of the extracellular matrix, modulating the pericellular environment in physiology and in pathologies. The interconnection between these families remains elusive. To assess the cross-activation of these families, we developed a peptide, fusion protein-based exposition system (Cleavage of exposed amino acid sequences, CleavEx) aiming at investigating the potential of KLK14 to recognize and hydrolyze proMMP sequences. Initial assessment identified ten MMP activation domain sequences which were validated by Edman degradation. The analysis revealed that membrane-type MMPs (MT-MMPs) are targeted by KLK14 for activation. Correspondingly, proMMP14-17 were investigated in vitro and found to be effectively processed by KLK14. Again, the expected neo-N-termini of the activated MT-MMPs was confirmed by Edman degradation. The effectiveness of proMMP activation was analyzed by gelatin zymography, confirming the release of fully active, mature MT-MMPs upon KLK14 treatment. Lastly, MMP14 was shown to be processed on the cell surface by KLK14 using murine fibroblasts overexpressing human MMP14. Herein, we propose KLK14-mediated selective activation of cell-membrane located MT-MMPs as an additional layer of their regulation. As both, KLKs and MT-MMPs, are implicated in cancer, their cross-activation may constitute an important factor in tumor progression and metastasis.
Collapse
|
14
|
Willson JA, Damjanovski S. Spatial analysis of RECK, MT1-MMP, and TIMP-2 proteins during early Xenopus laevis development. Gene Expr Patterns 2019; 34:119066. [DOI: 10.1016/j.gep.2019.119066] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 06/19/2019] [Accepted: 07/12/2019] [Indexed: 10/26/2022]
|
15
|
Rhomboid-Like-2 Intramembrane Protease Mediates Metalloprotease-Independent Regulation of Cadherins. Int J Mol Sci 2019; 20:ijms20235958. [PMID: 31783481 PMCID: PMC6928865 DOI: 10.3390/ijms20235958] [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: 11/13/2019] [Revised: 11/25/2019] [Accepted: 11/25/2019] [Indexed: 11/17/2022] Open
Abstract
Cadherins are a major family of cell-cell adhesive receptors, which are implicated in development, tissue homeostasis, and cancer. Here, we show a novel mechanism of post-translational regulation of E-cadherin in cancer cells by an intramembrane protease of the Rhomboid family, RHBDL2, which leads to the shedding of E-cadherin extracellular domain. In addition, our data indicate that RHBDL2 mediates a similar activity on VE-cadherin, which is selectively expressed by endothelial cells. We show that RHBDL2 promotes cell migration, which is consistent with its ability to interfere with the functional role of cadherins as negative regulators of motility; moreover, the two players appear to lie in the same functional pathway. Importantly, we show that RHBDL2 expression is induced by the inflammatory chemokine TNFα. The E-cadherin extracellular domain is known to be released by metalloproteases (MMPs); however, here, we provide evidence of a novel MMP-independent, TNFα inducible, E-cadherin processing mechanism that is mediated by RHBDL2. Thus, the intramembrane protease RHBDL2 is a novel regulator of cadherins promoting cell motility.
Collapse
|
16
|
Abstract
Multiple factors involving the methionine salvage pathway (MSP) and polyamine biosynthesis have been found to be involved in cancer cell proliferation, migration, invasion and metastasis. This review summarizes the relationships of the MSP enzyme acireductone dioxygenase (ARD), the ADI1 gene encoding ARD and other gene products (ADI1GP) with carcinomas and carcinogenesis. ARD exhibits structural and functional differences depending upon the metal bound in the active site. In the penultimate step of the MSP, the Fe2+ bound form of ARD catalyzes the on-pathway oxidation of acireductone leading to methionine, whereas Ni2+ bound ARD catalyzes an off-pathway reaction producing methylthiopropionate and carbon monoxide, a biological signaling molecule and anti-apoptotic. The relationship between ADI1GP, MSP and polyamine synthesis are discussed, along with possible role(s) of metal in modulating the cellular behavior of ADI1GP and its interactions with other cellular components.
Collapse
|
17
|
Wu W, Zhou Q, Zhao W, Gong Y, Su A, Liu F, Liu Y, Li Z, Zhu J. Ginsenoside Rg3 Inhibition of Thyroid Cancer Metastasis Is Associated with Alternation of Actin Skeleton. J Med Food 2018; 21:849-857. [PMID: 30136914 DOI: 10.1089/jmf.2017.4144] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Ginsenoside Rg3, a bioactive constituent from Panax ginseng, is a worldwide well-known traditional Chinese medicine used as a tonic. It also has good antitumor activity by inhibiting tumors metastasis. Tumor metastasis is a high risk in thyroid cancer. However, the effect and molecular mechanism underlying the antimetastatic activity of Rg3 in thyroid cancer have not been reported. In our study, we found that Rg3 inhibited the growth of thyroid cancer in vitro and in vivo and significantly inhibited metastasis of thyroid cancer. Rg3 apparently inhibited the migration and invasion in four papillary thyroid cancer (PTC) cells (TPC-1, BCPAP, C643, and Ocut-2c cells) and pulmonary metastasis in lung metastasis model of C643 cells in nude mice. We further found that a possible mechanism of Rg3 inhibiting thyroid cancer cells metastasis was associated with inhibiting cells actin skeleton function. Rg3 inhibited lamellipodia formation and induced microspike formation by inhibiting Rho GTPase in thyroid cancer cells. Rg3 decreased the levels of Rac-1 and Cdc42 proteins. In addition, Rg3 decreased the expression levels of matrix metalloproteinase-2 (MMP-2) and MMP-9 proteins in four thyroid cancer cells. The results that Rg3 remarkably inhibited the expression of vascular endothelial growth factor-C (VEGF-C) protein in PTC cells and VEGF-A protein in anaplastic thyroid cancer (ATC) cells and decreased the staining of CD31 in PTC and ATC tumors hinted that Rg3 might inhibit the lymph node metastasis in PTC and angiogenesis in ATC. These studies suggested that Rg3 might be a useful agent for the treatment of metastatic thyroid cancers.
Collapse
Affiliation(s)
- Wenshuang Wu
- Department of Thyroid Surgery, West China Hospital, Sichuan University , Chengdu, China
| | - Qian Zhou
- Department of Thyroid Surgery, West China Hospital, Sichuan University , Chengdu, China
| | - Wanjun Zhao
- Department of Thyroid Surgery, West China Hospital, Sichuan University , Chengdu, China
| | - Yanping Gong
- Department of Thyroid Surgery, West China Hospital, Sichuan University , Chengdu, China
| | - Anping Su
- Department of Thyroid Surgery, West China Hospital, Sichuan University , Chengdu, China
| | - Feng Liu
- Department of Thyroid Surgery, West China Hospital, Sichuan University , Chengdu, China
| | - Yang Liu
- Department of Thyroid Surgery, West China Hospital, Sichuan University , Chengdu, China
| | - Zhihui Li
- Department of Thyroid Surgery, West China Hospital, Sichuan University , Chengdu, China
| | - Jingqiang Zhu
- Department of Thyroid Surgery, West China Hospital, Sichuan University , Chengdu, China
| |
Collapse
|
18
|
Vuong TT, Rønning SB, Ahmed TAE, Brathagen K, Høst V, Hincke MT, Suso HP, Pedersen ME. Processed eggshell membrane powder regulates cellular functions and increase MMP-activity important in early wound healing processes. PLoS One 2018; 13:e0201975. [PMID: 30080894 PMCID: PMC6078314 DOI: 10.1371/journal.pone.0201975] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 07/25/2018] [Indexed: 01/22/2023] Open
Abstract
Avian eggshell membrane (ESM) is a natural biomaterial that has been used as an alternative natural bandage to cure wounds, and is available in large quantities from egg industries. We have previously demonstrated that processed eggshell membrane powder (PEP), aiming to be used in a low cost wound healing product, possesses anti-inflammatory properties. In this study, we further investigated effects of PEP on MMP activities in vitro (a dermal fibroblast cell culture system) and in vivo (a mouse skin wound healing model). Three days incubation with PEP in cell culture led to rearrangement of the actin-cytoskeleton and vinculin in focal adhesions and increased syndecan-4 shedding. In addition, we observed increased matrix metalloproteinase type 2 (MMP-2) enzyme activation, without effects on protein levels of MMP-2 or its regulators (membrane type 1 (MT1)-MMP and tissue inhibitor of matrix metalloproteinase type 2 (TIMP-2). Longer incubation (10 days) led to increased protein levels of MMP-2 and its regulators. We also observed an increased alpha-smooth muscle actin (α-SMA) production, suggesting an effect of PEP on myofibroblast differentiation. In vivo, using the mouse skin wound healing model, PEP treatment (3 days) increased MMP activity at the wound edges, along with increased MMP-2 and MMP-9 protein levels, and increased keratinocyte cell proliferation. Altogether, our data suggest PEP stimulates MMP activity, and with a positive effect on early cellular events during wound healing.
Collapse
Affiliation(s)
| | | | - Tamer A. E. Ahmed
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Medical Biotechnology Department, Genetic Engineering and Biotechnology Research Institute, City of Scientific Research and Technology Applications (SRTA-City), Alexandria, Egypt
| | | | | | - Maxwell T. Hincke
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Department of Innovation in Medical Education, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | | | | |
Collapse
|
19
|
Zhao P, Lan F, Zhang H, Zeng G, Liu D. Down-regulation of KIF2A inhibits gastric cancer cell invasion via suppressing MT1-MMP. Clin Exp Pharmacol Physiol 2018; 45:1010-1018. [PMID: 29781531 DOI: 10.1111/1440-1681.12974] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 05/10/2018] [Accepted: 05/12/2018] [Indexed: 12/15/2022]
Abstract
Gastric cancer accounts for a sizeable proportion of global cancer mortality with high morbidity and poor prognosis. Kinesin superfamily proteins (KIFs) are microtubule-dependent motor proteins that function as oncogenes in cancer cells, it has been discovered in recent years. Kinesin family member 2a (KIF2A), a member of the KIFs, has received attention for its role in carcinogenesis and its prognostic value in several human cancers such as breast cancer, colorectal cancer, and squamous cell carcinoma. However, the role of KIF2A in human gastric cancer remains unknown. In this study we aimed to explore the expression and biological functions of KIF2A in human gastric cancer cells, as well as to reveal its potential action mechanism. First, we found that KIF2A was markedly increased in gastric cancer cells (MKN-28, MKN-45, NCI-N87 and SGC-7901) compared to normal gastric mucosa epithelial cells (GES-1). Then KIF2A was successfully silenced in MKN-45 and SGC-7901 cells to facilitate further research into its function. We discovered that KIF2A silencing can significantly inhibit the growth and invasion of MKN-45 and SGC-7901 cells in a time-independent manner, accompanying a decreased expression of Membrane type 1-matrix metalloproteinase (MT1-MMP). When MT1-MMP was reintroduced into MKN-45 and SGC-7901 cells in the KIF2A-siRNA group, only invasion inhibition effects on MKN-45 and SGC-7901 cells induced by KIF2A silencing can be reversed. In conclusion, our study reveals that down-regulation of KIF2A can inhibit gastric cancer cell invasion by suppressing MT1-MMP.
Collapse
Affiliation(s)
- Peng Zhao
- Department of Oncology, Tangdu Hospital, The Fourth Military Medical University, Shaanxi, Xi'an, China
| | - Fei Lan
- Department of Oncology, Tangdu Hospital, The Fourth Military Medical University, Shaanxi, Xi'an, China
| | - Hui Zhang
- Department of Internal Medicine, Jingyang County Hospital, Shaanxi, Xianyang, China
| | - Guangwei Zeng
- Department of Cardiovascular Medicine, Tangdu Hospital, The Fourth Military Medical University, Shaanxi, Xi'an, China
| | - Dong Liu
- Department of Oncology, Tangdu Hospital, The Fourth Military Medical University, Shaanxi, Xi'an, China
| |
Collapse
|
20
|
Shang D, Song B, Liu Y. Epirubicin suppresses proliferative and metastatic potential by downregulating transforming growth factor-β-induced expression in urothelial carcinoma. Cancer Sci 2018; 109:980-987. [PMID: 28940965 PMCID: PMC5891197 DOI: 10.1111/cas.13403] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 09/13/2017] [Accepted: 09/18/2017] [Indexed: 12/29/2022] Open
Abstract
Transforming growth factor‐β‐induced (TGFΒI) is considered to be a vital gene in several carcinomas. In this study we determined the effect of TGFBI on the proliferative and metastatic potential of human urothelial carcinoma (UC) cells as well as its mRNA and protein expression, which were detected by RT‐PCR and western blot, respectively. UC cell proliferation was analyzed by WST‐1 assay and Hoechst 33258 staining. The effect of TGFBI on UC cell metastasis was analyzed using adhesion, migration and invasion assays. We found that TGFBI increased the proliferation of UC cells. Moreover, TGFBI enhanced the adhesion, migration and invasion of UC cells by upregulating MMP‐2, MMP‐9 and calpain‐2 expression. We evaluated the effect of Epirubicin (EPI) on the regulation of TGFBI expression and found that TGFBI acts as a downstream target of EPI and is suppressed by EPI in UC cells. EPI is more effective in inhibiting the proliferation and metastasis of UC cells with high TGFBI expression. This study demonstrates that TGFBI might lead to tumorigenesis and progression of UC and those cells with high TGFBI expression may be vulnerable to relapse. EPI could prove to be a therapeutic option in patients with high TGFBI expressing UC cells.
Collapse
Affiliation(s)
- Donghao Shang
- Department of Urology, Friendship Hospital, Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Capital Medical University, Beijing, China
| | - Bo Song
- Department of Urology, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Yuting Liu
- Department of Pathology, Capital Medical University, Beijing, China
| |
Collapse
|
21
|
Metalloproteinases in atherosclerosis. Eur J Pharmacol 2017; 816:93-106. [DOI: 10.1016/j.ejphar.2017.09.007] [Citation(s) in RCA: 142] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 08/31/2017] [Accepted: 09/08/2017] [Indexed: 11/20/2022]
|
22
|
Zheng Y, Ren J, Wu Y, Meng X, Zhao Y, Wu C. Proteolytic Unlocking of Ultrastable Twin-Acylhydrazone Linkers for Lysosomal Acid-Triggered Release of Anticancer Drugs. Bioconjug Chem 2017; 28:2620-2626. [DOI: 10.1021/acs.bioconjchem.7b00471] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Yiwu Zheng
- Department of Chemistry,
College of Chemistry and Chemical Engineering, State Key Laboratory
of Physical Chemistry of Solid Surfaces, The MOE Key Laboratory of
Spectrochemical Analysis and Instrumentation, Xiamen University, Xiamen, 361005, P.R. China
| | - Jing Ren
- Department of Chemistry,
College of Chemistry and Chemical Engineering, State Key Laboratory
of Physical Chemistry of Solid Surfaces, The MOE Key Laboratory of
Spectrochemical Analysis and Instrumentation, Xiamen University, Xiamen, 361005, P.R. China
| | - Yaqi Wu
- Department of Chemistry,
College of Chemistry and Chemical Engineering, State Key Laboratory
of Physical Chemistry of Solid Surfaces, The MOE Key Laboratory of
Spectrochemical Analysis and Instrumentation, Xiamen University, Xiamen, 361005, P.R. China
| | - Xiaoting Meng
- Department of Chemistry,
College of Chemistry and Chemical Engineering, State Key Laboratory
of Physical Chemistry of Solid Surfaces, The MOE Key Laboratory of
Spectrochemical Analysis and Instrumentation, Xiamen University, Xiamen, 361005, P.R. China
| | - Yibing Zhao
- Department of Chemistry,
College of Chemistry and Chemical Engineering, State Key Laboratory
of Physical Chemistry of Solid Surfaces, The MOE Key Laboratory of
Spectrochemical Analysis and Instrumentation, Xiamen University, Xiamen, 361005, P.R. China
| | - Chuanliu Wu
- Department of Chemistry,
College of Chemistry and Chemical Engineering, State Key Laboratory
of Physical Chemistry of Solid Surfaces, The MOE Key Laboratory of
Spectrochemical Analysis and Instrumentation, Xiamen University, Xiamen, 361005, P.R. China
| |
Collapse
|
23
|
Li Q, Michaud M, Shankar R, Canosa S, Schwartz M, Madri JA. MMP-2: A modulator of neuronal precursor activity and cognitive and motor behaviors. Behav Brain Res 2017; 333:74-82. [DOI: 10.1016/j.bbr.2017.06.041] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 06/22/2017] [Accepted: 06/26/2017] [Indexed: 12/16/2022]
|
24
|
Pattni V, Vasilevskaya T, Thiel W, Heyden M. Distinct Protein Hydration Water Species Defined by Spatially Resolved Spectra of Intermolecular Vibrations. J Phys Chem B 2017. [PMID: 28636363 PMCID: PMC5607456 DOI: 10.1021/acs.jpcb.7b03966] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
![]()
In
this molecular dynamics simulation study, we analyze intermolecular
vibrations in the hydration shell of a solvated enyzme, the membrane
type 1–matrix metalloproteinase, with high spatial resolution.
Our approach allows us to characterize vibrational signatures of the
local hydrogen bond network, the translational mobility of water molecules,
as well as the molecular entropy, in specific local environments.
Our study demonstrates the heterogeneity of water properties within
the hydration shell of a complex biomolecule. We define a classification
scheme based on the vibrational density of states that allows us to
distinguish separate classes of hydration water species and facilitates
the description of hydration water properties at distinct hydration
sites. The results demonstrate that no single characteristic of the
protein surface is sufficient to determine the properties of nearby
water. The protein surface geometry, quantified here by the number
of protein atoms in the vicinity of a hydration water molecule, as
well as the chemical nature of a solvated protein functional group,
influences dynamic and thermodynamic properties of solvating water
molecules.
Collapse
Affiliation(s)
- Viren Pattni
- Max-Planck-Institut für Kohlenforschung , Kaiser-Wilhelm-Platz 1, DE-45470 Mülheim an der Ruhr, Germany
| | - Tatiana Vasilevskaya
- Max-Planck-Institut für Kohlenforschung , Kaiser-Wilhelm-Platz 1, DE-45470 Mülheim an der Ruhr, Germany
| | - Walter Thiel
- Max-Planck-Institut für Kohlenforschung , Kaiser-Wilhelm-Platz 1, DE-45470 Mülheim an der Ruhr, Germany
| | - Matthias Heyden
- Max-Planck-Institut für Kohlenforschung , Kaiser-Wilhelm-Platz 1, DE-45470 Mülheim an der Ruhr, Germany
| |
Collapse
|
25
|
Craft TR, Forrester WC. The Caenorhabditis elegans matrix non-peptidase MNP-1 is required for neuronal cell migration and interacts with the Ror receptor tyrosine kinase CAM-1. Dev Biol 2017; 424:18-27. [PMID: 28238735 DOI: 10.1016/j.ydbio.2017.02.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 02/21/2017] [Accepted: 02/22/2017] [Indexed: 10/20/2022]
Abstract
Directed cell migration is critical for metazoan development. During Caenorhabditis elegans development many neuronal, muscle and other cell types migrate. Multiple classes of proteins have been implicated in cell migration including secreted guidance cues, receptors for guidance cues and intracellular proteins that respond to cues to polarize cells and produce the forces that move them. In addition, cell surface and secreted proteases have been identified that may clear the migratory route and process guidance cues. We report here that mnp-1 is required for neuronal cell and growth cone migrations. MNP-1 is expressed by migrating cells and functions cell autonomously for cell migrations. We also find a genetic interaction between mnp-1 and cam-1, which encodes a Ror receptor tyrosine kinase required for some of the same cell migrations.
Collapse
Affiliation(s)
- Teresa R Craft
- Medical Sciences Program, Indiana University, Bloomington, IN 47405, United States
| | - Wayne C Forrester
- Medical Sciences Program, Indiana University, Bloomington, IN 47405, United States.
| |
Collapse
|
26
|
Yang J, Pei H, Luo H, Fu A, Yang H, Hu J, Zhao C, Chai L, Chen X, Shao X, Wang C, Wu W, Wan L, Ye H, Qiu Q, Peng A, Wei Y, Yang L, Chen L. Non-toxic dose of liposomal honokiol suppresses metastasis of hepatocellular carcinoma through destabilizing EGFR and inhibiting the downstream pathways. Oncotarget 2016; 8:915-932. [PMID: 27906672 PMCID: PMC5352206 DOI: 10.18632/oncotarget.13687] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 11/05/2016] [Indexed: 02/05/2023] Open
Abstract
At present, there is no specific anti-metastasis drug in HCC treatment. Drugs used for primary HCC tumors and tumor metastasis are very similar, among which cytotoxic drugs are prevalent, such as cisplatin, doxorubicin and 5-FU. The EGFR pathway plays an important role in promoting hepatocellular carcinoma (HCC) metastasis. Hence, development of non-toxic anti-metastasis drugs, such as EGFR or downstream pathways inhibitors, is of great importance. In our present study, we found non-toxic dose of liposomal honokiol (LH) could inhibit the HCC metastasis by destabilizing EGFR and inhibiting the downstream pathways. Non-toxic dose of LH significantly inhibited the motility, migration and lamellipodia formation of HepG2 cells in vitro and decreased extravasation of HepG2 cells in a novel metastasis model of transgenic zebrafish. In two lung metastasis models (HepG2 and B16F10) and a spontaneous metastasis model of HepG2 cells, LH remarkably inhibited pulmonary metastasis and regional lymph nodes metastasis without obvious toxicity. Further study showed that destabilizing EGFR and inhibiting the downstream pathways were the main mechanisms of non-toxic dose of LH on metastasis inhibition. Our results provide the preclinical rationale and the underlying mechanisms of LH to suppress HCC metastasis, implicating LH as a potential therapeutic agent to block HCC metastasis without severe side effects.
Collapse
Affiliation(s)
- Jianhong Yang
- 1 State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, P.R. China
| | - Heying Pei
- 1 State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, P.R. China
| | - Hong Luo
- 2 Department of Ultrasonic Medicine, West China Second Hospital, Sichuan University, Chengdu, China
| | - Afu Fu
- 1 State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, P.R. China
| | - Hansuo Yang
- 1 State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, P.R. China
| | - Jia Hu
- 1 State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, P.R. China
| | - Chengjian Zhao
- 1 State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, P.R. China
| | - LuLu Chai
- 1 State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, P.R. China
| | - Xiang Chen
- 1 State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, P.R. China
| | - Ximing Shao
- 1 State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, P.R. China
| | - Chunyu Wang
- 1 State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, P.R. China
| | - Wenshuang Wu
- 1 State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, P.R. China
| | - Li Wan
- 3 School of Pharmacy, Chengdu University of TCM, The Ministry of Education Key Laboratory of Standardization of Chinese Herbal Medicine, State Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu, China
| | - Haoyu Ye
- 1 State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, P.R. China
| | - Qiang Qiu
- 1 State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, P.R. China
| | - Aihua Peng
- 1 State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, P.R. China
| | - Yuquan Wei
- 1 State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, P.R. China
| | - Li Yang
- 1 State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, P.R. China
| | - Lijuan Chen
- 1 State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, P.R. China
| |
Collapse
|
27
|
Abstract
The neurovascular unit, which consists of astrocytic end-feet, neurons, pericytes, and endothelial cells, plays a key role in maintaining brain homeostasis by forming the blood-brain barrier and carefully controlling local cerebral blood flow. When the blood-brain barrier is disrupted, blood components can leak into the brain, damage the surrounding tissue and lead to cognitive impairment. This disruption in the blood-brain barrier and subsequent impairment in cognition are common after stroke and during cerebral amyloid angiopathy and Alzheimer's disease. Matrix metalloproteinases are proteases that degrade the extracellular matrix as well as tight junctions between endothelial cells and have been implicated in blood-brain barrier breakdown in neurodegenerative diseases. This review will focus on the roles of MMP2 and MMP9 in dementia, primarily post-stroke events that lead to dementia, cerebral amyloid angiopathy, and Alzheimer's disease.
Collapse
|
28
|
MT1-MMP Inhibits the Activity of Bst-2 via Their Cytoplasmic Domains Dependent Interaction. Int J Mol Sci 2016; 17:ijms17060818. [PMID: 27240342 PMCID: PMC4926352 DOI: 10.3390/ijms17060818] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2016] [Revised: 05/12/2016] [Accepted: 05/16/2016] [Indexed: 01/30/2023] Open
Abstract
Bst-2 (bone marrow stromal cell antigen 2) is a type II membrane protein, and it acts as a tetherin to inhibit virion releasing from infectious cells. Membrane type-1 matrix metalloproteinase (MT1-MMP) is a protease. It plays a pivotal role in cellular growth and migration by activating proMMP-2 into active MMP2. Our results here elaborate that MT1-MMP inhibits the tetherin activity of Bst-2 by interacting with Bst-2, and the cytoplasmic domains of both Bst-2 and MT1-MMP play critical roles within this interaction. Based on our experimental data, the assays for virion release and co-immunoprecipitation have clearly demonstrated that the activity of Bst-2 is markedly inhibited by MT1-MMP via their interaction; and both the N-terminal domain of Bst-2 and the C-terminal domain of MT1-MMP are important in the interaction. Immunostaining and Confocal Microscopy assay shows that MT1-MMP interacts with Bst-2 to form granular particles trafficking into cytoplasm from membrane and, finally, results in Bst-2 and MT1-MMP both being inhibited. In addition, mutant experiments elucidate that the N-terminal domain of Bst-2 is not only important in relating to the activity of Bst-2 itself, but is important for inhibiting the MT1-MMP/proMMP2/MMP2 pathway. These findings suggest that MT1-MMP is a novel inhibitor of Bst-2 in MT1-MMP expressed cell lines and also indicate that both the N-terminal domain of Bst-2 and the C-terminal domain of MT1-MMP are crucial in down-regulation.
Collapse
|
29
|
Tsuneki M, Madri JA. CD44 Influences Fibroblast Behaviors Via Modulation of Cell-Cell and Cell-Matrix Interactions, Affecting Survivin and Hippo Pathways. J Cell Physiol 2016; 231:731-43. [PMID: 26248063 DOI: 10.1002/jcp.25123] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 08/04/2015] [Indexed: 12/17/2022]
Abstract
CD44 has been studied in a wide variety of cell types, in a diverse array of cell behaviors and in a diverse range of signaling pathways. We now document a role for CD44 in mediating fibroblast behaviors via regulation of N-cadherin, extracellular matrix expression, Survivin and the Hippo pathway. Here, we report our findings on the roles of CD44 in modulating proliferation, apoptosis, migration and invasion of murine wild-type (WT-FB) and CD44 knockout dermal fibroblasts (CD44KO-FB). As we have documented in microvascular endothelial cells lacking CD44, we found persistent increased proliferation, reduced activation of cleaved caspase 3, increased initial attachment, but decreased strength of cell attachment in high cell density, post confluent CD44KO-FB cultures. Additionally, we found that siRNA knock-down of CD44 mimicked the behaviors of CD44KO-FB, restoring the decreases in N-cadherin, collagen type I, fibronectin, Survivin, nuclear fractions of YAP and phospho-YAP and decreased levels of cleaved caspase 3 to the levels observed in CD44KO-FB. Interestingly, plating CD44KO-FB on collagen type I or fibronectin resulted in significant decreases in secondary proliferation rates compared to plating cells on non-coated dishes, consistent with increased cell adhesion compared to their effects on WT-FB. Lastly, siRNA knockdown of CD44 in WT-FB resulted in increased fibroblast migration compared to WT-FB, albeit at reduced rates compared to CD44KO-FB. These results are consistent with CD44's pivotal role in modulating several diverse behaviors important for adhesion, proliferation, apoptosis, migration and invasion during development, growth, repair, maintenance and regression of a wide variety of mesenchymal tissues.
Collapse
Affiliation(s)
- Masayuki Tsuneki
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut.,Division of Cancer Biology, National Cancer Center Research Institute, 5-1-1, Tsukiji, Chuo-ku, Tokyo, Japan
| | - Joseph A Madri
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
30
|
Qian Y, Li L, Jiang C, Xu W, Lv Y, Zhong L, Cai K, Yang L. The effect of hyaluronan on the motility of skin dermal fibroblasts in nanofibrous scaffolds. Int J Biol Macromol 2015; 79:133-43. [DOI: 10.1016/j.ijbiomac.2015.04.059] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Revised: 04/20/2015] [Accepted: 04/23/2015] [Indexed: 10/23/2022]
|
31
|
Chiu KC, Lee CH, Liu SY, Yeh CT, Huang RY, Yuh DY, Cheng JC, Chou YT, Shieh YS. Protumoral effect of macrophage through Axl activation on mucoepidermoid carcinoma. J Oral Pathol Med 2015; 43:538-44. [PMID: 25184164 DOI: 10.1111/jop.12163] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE This study aims to test the potential involvement of Axl signaling in the protumoral effect of tumor-associated macrophages (TAMs) in mucoepidermoid carcinoma (MEC). MATERIALS AND METHODS We carried out cocultured experiments by incubation of MEC cells (UTMUC-1) and macrophages (THP-1) and examined Axl activation status. The expression of MMPs and behavior change were examined in UT-MUC-1 cells. The effect of Axl signaling on co-cultured cancer cells was further investigated by knockdown Axl expression and suppression by Axl-specific inhibitor R428. RESULTS Activation of Axl signaling and increased expression and activity of MMP-2 and MMP-9 along with increased invasion/migration ability in MEC cells were observed when co-cultured with TAMs. Upon knockdown of Axl in MEC or addition of R428 in the co-cultured system, these co-cultured effects were diminished. CONCLUSION TAMs play a protumoral role in MEC via activation of the Axl signaling pathway, up-regulating MMPs expression, and increasing invasion/migration ability.
Collapse
|
32
|
Caley MP, Martins VL, O'Toole EA. Metalloproteinases and Wound Healing. Adv Wound Care (New Rochelle) 2015; 4:225-234. [PMID: 25945285 DOI: 10.1089/wound.2014.0581] [Citation(s) in RCA: 586] [Impact Index Per Article: 58.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Accepted: 09/29/2014] [Indexed: 12/15/2022] Open
Abstract
Significance: Matrix metalloproteinases (MMPs) are present in both acute and chronic wounds. They play a pivotal role, with their inhibitors, in regulating extracellular matrix degradation and deposition that is essential for wound reepithelialization. The excess protease activity can lead to a chronic nonhealing wound. The timed expression and activation of MMPs in response to wounding are vital for successful wound healing. MMPs are grouped into eight families and display extensive homology within these families. This homology leads in part to the initial failure of MMP inhibitors in clinical trials and the development of alternative methods for modulating the MMP activity. MMP-knockout mouse models display altered wound healing responses, but these are often subtle phenotypic changes indicating the overlapping MMP substrate specificity and inter-MMP compensation. Recent Advances: Recent research has identified several new MMP modulators, including photodynamic therapy, protease-absorbing dressing, microRNA regulation, signaling molecules, and peptides. Critical Issues: Wound healing requires the controlled activity of MMPs at all stages of the wound healing process. The loss of MMP regulation is a characteristic of chronic wounds and contributes to the failure to heal. Future Directions: Further research into how MMPs are regulated should allow the development of novel treatments for wound healing.
Collapse
Affiliation(s)
- Matthew P. Caley
- Blizard Institute, Centre for Cutaneous Research, London, United Kingdom
| | - Vera L.C. Martins
- Blizard Institute, Centre for Cutaneous Research, London, United Kingdom
| | - Edel A. O'Toole
- Blizard Institute, Centre for Cutaneous Research, London, United Kingdom
| |
Collapse
|
33
|
Ma L, Zhang L, Wang B, Wei J, Liu J, Zhang L. Berberine inhibits Chlamydia pneumoniae infection-induced vascular smooth muscle cell migration through downregulating MMP3 and MMP9 via PI3K. Eur J Pharmacol 2015; 755:102-9. [PMID: 25746423 DOI: 10.1016/j.ejphar.2015.02.039] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Revised: 01/29/2015] [Accepted: 02/17/2015] [Indexed: 01/01/2023]
Abstract
The mechanisms by which Chlamydia pneumoniae infection promote vascular smooth muscle cell (VSMC) migration required in the development of atherosclerosis have not yet been fully clarified. Matrix metalloproteinases (MMPs) have important roles in VSMC migration. However, it is still unknown whether MMPs are involved in C. pneumoniae infection-induced VSMC migration. In addition, whether berberine can exert its inhibitory effects on the infection-induced VSMC migration also remains unclear. Accordingly, we investigated the effects of berberine on C. pneumoniae infection-induced VSMC migration and explored the possible mechanisms involved in this process. Herein, we found that C. pneumoniae infection could induce VSMC migration through Matrigel-coated membrane (P<0.05), which can be significantly inhibited by the broad-spectrum MMP inhibitor GM6001 (P<0.05). Our results also showed that C. pneumoniae infection upregulated both mRNA and protein expressions of MMP3 and MMP9 (P<0.05). The specific phosphoinositide 3-kinase (PI3K) inhibitor LY294002 significantly suppressed the increases in MMP3 and MMP9 protein expressions induced by C. pneumoniae infection (P<0.05). Further experiments showed that berberine significantly attenuated C. pneumoniae infection-induced VSMC migration (P<0.05). Moreover, berberine suppressed the protein expressions of MMP3 and MMP9 caused by C. pneumoniae infection in a dose-dependent manner (P<0.05). C. pneumoniae infection-induced increase in the phosphorylation level of Akt at Ser473 was inhibited by the treatment with berberine (P<0.05). Taken together, our data suggest that berberine inhibits C. pneumoniae infection-induced VSMC migration by downregulating the expressions of MMP3 and MMP9 via PI3K.
Collapse
Affiliation(s)
- Lu Ma
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Lijun Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Beibei Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Junyan Wei
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Jingya Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Lijun Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China.
| |
Collapse
|
34
|
Rajasekharan S, Rana J, Gulati S, Gupta V, Gupta S. Neuroinvasion by Chandipura virus. Acta Trop 2014; 135:122-6. [PMID: 24713200 DOI: 10.1016/j.actatropica.2014.03.028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Revised: 03/14/2014] [Accepted: 03/26/2014] [Indexed: 01/13/2023]
Abstract
Chandipura virus (CHPV) is an arthropod borne rhabdovirus associated with acute encephalitis in children below the age of 15 years in the tropical states of India. Although the entry of the virus into the nervous system is among the crucial events in the pathogenesis of CHPV, the exact mechanism allowing CHPV to invade the central nervous system (CNS) is currently poorly understood. In the present review, based on the knowledge of host interactors previously predicted for CHPV, along with the support from experimental data available for other encephalitic viruses, the authors have speculated the various plausible modes by which CHPV could surpass the blood-brain barrier and invade the CNS to cause encephalitis whilst evading the host immune surveillance. Collectively, this review provides a conservative set of potential interactions that can be employed for future experimental validation with a view to better understand the neuropathogenesis of CHPV.
Collapse
Affiliation(s)
- Sreejith Rajasekharan
- Centre for Emerging Diseases, Department of Biotechnology, Jaypee Institute of Information Technology, A-10, Sector 62, Noida, Uttar Pradesh 201 307, India
| | - Jyoti Rana
- Centre for Emerging Diseases, Department of Biotechnology, Jaypee Institute of Information Technology, A-10, Sector 62, Noida, Uttar Pradesh 201 307, India
| | - Sahil Gulati
- Centre for Emerging Diseases, Department of Biotechnology, Jaypee Institute of Information Technology, A-10, Sector 62, Noida, Uttar Pradesh 201 307, India
| | - Vandana Gupta
- Department of Microbiology, Ram Lal Anand College, University of Delhi South Campus (UDSC), Benito Juarez Marg, New Delhi 110021, India
| | - Sanjay Gupta
- Centre for Emerging Diseases, Department of Biotechnology, Jaypee Institute of Information Technology, A-10, Sector 62, Noida, Uttar Pradesh 201 307, India.
| |
Collapse
|
35
|
Tsai CL, Chen WC, Hsieh HL, Chi PL, Hsiao LD, Yang CM. TNF-α induces matrix metalloproteinase-9-dependent soluble intercellular adhesion molecule-1 release via TRAF2-mediated MAPKs and NF-κB activation in osteoblast-like MC3T3-E1 cells. J Biomed Sci 2014; 21:12. [PMID: 24502696 PMCID: PMC3926355 DOI: 10.1186/1423-0127-21-12] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2013] [Accepted: 01/28/2014] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Matrix metalloproteinase-9 (MMP-9) has been shown to be induced by cytokines including TNF-α and may contribute to bone inflammatory diseases. However, the mechanisms underlying MMP-9 expression induced by TNF-α in MC3T3-E1 cells remain unclear. RESULTS We applied gelatin zymography, Western blot, RT-PCR, real-time PCR, selective pharmacological inhibitors of transcription (actinomycin D, Act.D), translation (cycloheximide, CHI), c-Src (PP1), MEK1/2 (U0126), p38 MAPK (SB202190), JNK1/2 (SP600125), and NF-κB (Bay11-7082), respective siRNAs transfection, promoter assay, immunofluorescence staining, and ELISA to investigate the MMP-9 expression and soluble ICAM-1 (sICAM-1) release induced by TNF-α in MC3T3-E1 cells. Here we demonstrated that TNF-α-induced MMP-9 expression was attenuated by Act.D, CHI, PP1, U0126, SB202190, SP600125, and Bay11-7082, and by the transfection with siRNAs for ERK2, p38 MAPK, and JNK2. TNF-α-stimulated TNFR1, TRAF2, and c-Src complex formation was revealed by immunoprecipitation and Western blot. Furthermore, TNF-α-stimulated NF-κB phosphorylation and translocation were blocked by Bay11-7082, but not by PP1, U0126, SB202190, or SP600125. TNF-α time-dependently induced MMP-9 promoter activity which was also inhibited by PP1, U0126, SB202190, SP600125, or Bay11-7082. Up-regulation of MMP-9 was associated with the release of sICAM-1 into the cultured medium, which was attenuated by the pretreatment with MMP-2/9i, an MMP-9 inhibitor. CONCLUSIONS In this study, we demonstrated that TNF-α up-regulates MMP-9 expression via c-Src, MAPKs, and NF-κB pathways. In addition, TNF-α-induced MMP-9 expression may contribute to the production of sICAM-1 by MC3T3-E1 cells. The interplay between MMP-9 expression and sICAM-1 release may exert an important role in the regulation of bone inflammatory diseases.
Collapse
Affiliation(s)
| | | | | | | | | | - Chuen-Mao Yang
- Department of Physiology and Pharmacology and Health Ageing Research Center, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan.
| |
Collapse
|
36
|
Tsuneki M, Madri JA. CD44 regulation of endothelial cell proliferation and apoptosis via modulation of CD31 and VE-cadherin expression. J Biol Chem 2014; 289:5357-70. [PMID: 24425872 DOI: 10.1074/jbc.m113.529313] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
CD44 has been implicated in a diverse array of cell behaviors and in a diverse range of signaling pathway activations under physiological and pathophysiological conditions. We have documented a role for CD44 in mediating vascular barrier integrity via regulation of PECAM-1 (CD31) expression. We now report our findings on the roles of CD44 in modulating proliferation and apoptosis of microvascular endothelial cells via its modulation of CD31 and VE-cadherin expression and the Hippo pathway. In this report, we demonstrate persistent increased proliferation and reduced activations of both effector and initiator caspases in high cell density, postconfluent CD44 knock-out (CD44KO), and CD31KO cultures. We found that reconstitution with murine CD44 or CD31 restored the proliferative and caspase activation rates to WT levels. Moreover, we have confirmed that the CD31 ecto-domain plays a key role in specific caspase cascades as well as cell adhesion-mediated cell growth and found that CD31 deficiency results in a reduction in VE-cadherin expression. Last, we have shown that both CD44KO and CD31KO endothelial cells exhibit a reduced VE-cadherin expression correlating with increased survivin expression and YAP nuclear localization, consistent with inactivation of the Hippo pathway, resulting in increased proliferation and decreased apoptosis. These findings support the concept that CD44 mediates several of its effects on endothelia through modulation of adhesion protein expression, which, in addition to its known modulation of junctional integrity, matrix metalloproteinase levels and activation, interactions with cortical membrane proteins, and selected signaling pathways, plays a key role as a critical regulator of vascular function.
Collapse
Affiliation(s)
- Masayuki Tsuneki
- From the Department of Pathology, Yale University School of Medicine, New Haven, Connecticut 06520
| | | |
Collapse
|
37
|
Shindo K, Aishima S, Ohuchida K, Fujiwara K, Fujino M, Mizuuchi Y, Hattori M, Mizumoto K, Tanaka M, Oda Y. Podoplanin expression in cancer-associated fibroblasts enhances tumor progression of invasive ductal carcinoma of the pancreas. Mol Cancer 2013; 12:168. [PMID: 24354864 PMCID: PMC3916072 DOI: 10.1186/1476-4598-12-168] [Citation(s) in RCA: 111] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Accepted: 12/16/2013] [Indexed: 01/05/2023] Open
Abstract
Background Interactions between cancer cells and surrounding cancer-associated fibroblasts (CAFs) play an important role in cancer progression. Invasive ductal carcinoma (IDC) of the pancreas is characterized by abundant fibrous connective tissue called desmoplasia. Podoplanin (PDPN) is a lymphatic vessel marker (D2-40), and expression of PDPN by stromal CAFs has been reported to be a prognostic indicator in various types of cancer. Methods Expression of PDPN in pancreatic IDCs was assessed by immunohistochemical examination in 105 patients who underwent pancreatic resection. Primary CAFs were established from pancreatic cancer tissue obtained by surgery. Quantitative reverse transcription-polymerase chain reaction and flow cytometric analysis were performed to investigate PDPN expression in CAFs. We sorted CAFs according to PDPN expression, and analyzed the functional differences between PDPN+ CAFs and PDPN– CAFs using indirect co-culture with pancreatic cancer cell lines. We also investigated the culture conditions to regulate PDPN expression in CAFs. Results PDPN expression in stromal fibroblasts was associated with lymphatic vessel invasion (P = 0.0461), vascular invasion (P = 0.0101), tumor size ≥3 cm (P = 0.0038), histological grade (P = 0.0344), Union for International Cancer Control classification T stage (P = 0.029), and shorter survival time (P < 0.0001). Primary CAFs showed heterogeneous PDPN expression in vitro. Moreover, migration and invasion of pancreatic cancer cell lines (PANC-1 and SUIT-2) were associated with PDPN expression in CAFs (P < 0.01) and expression of CD10, matrix metalloproteinase (MMP) 2, and MMP3. In cultured CAFs, PDPN positivity changed over time under several conditions including co-culture with cancer cells, different culture media, and addition of growth factor. Conclusions PDPN-expressing CAFs enhance the progression of pancreatic IDC, and a high ratio of PDPN-expressing CAFs is an independent predictor of poor outcome. Understanding the regulation of the tumor microenvironment is an important step towards developing new therapeutic strategies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Yoshinao Oda
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Fukuoka 812-8582, Japan.
| |
Collapse
|
38
|
Langenfurth A, Rinnenthal JL, Vinnakota K, Prinz V, Carlo AS, Stadelmann C, Siffrin V, Peaschke S, Endres M, Heppner F, Glass R, Wolf SA, Kettenmann H. Membrane-type 1 metalloproteinase is upregulated in microglia/brain macrophages in neurodegenerative and neuroinflammatory diseases. J Neurosci Res 2013; 92:275-86. [DOI: 10.1002/jnr.23288] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Revised: 06/14/2013] [Accepted: 07/25/2013] [Indexed: 01/09/2023]
Affiliation(s)
- Anika Langenfurth
- Cellular Neurosciences; Max Delbrück Centre for Molecular Medicine; Berlin Germany
- Department of Neurology; Charité, Universitätsmedizin Berlin; Charité Campus Virchow Berlin Germany
| | - Jan Leo Rinnenthal
- Institute for Neuropathology; Charité, Universitätsmedizin Berlin; Charité Campus Mitte Berlin Germany
| | - Katyayni Vinnakota
- Cellular Neurosciences; Max Delbrück Centre for Molecular Medicine; Berlin Germany
| | - Vincent Prinz
- Department of Neurology and Center for Stroke Research Berlin; Charité, Universitätsmedizin Berlin; Charité Campus Mitte Berlin Germany
- Department of Neurosurgery; Charité, Universitätsmedizin Berlin; Charité Campus Virchow Berlin Germany
| | - Anne-Sophie Carlo
- Molecular Cardiovascular Research; Max Delbrück Centre for Molecular Medicine; Berlin Germany
- Max Planck Institute for Infection Biology; Berlin Germany
| | | | - Volker Siffrin
- Department of Neurology; University Medical Center Mainz; Johannes Gutenberg University Mainz; Mainz Germany
| | - Susann Peaschke
- Cellular Neurosciences; Max Delbrück Centre for Molecular Medicine; Berlin Germany
| | - Matthias Endres
- Department of Neurology; Charité, Universitätsmedizin Berlin; Charité Campus Virchow Berlin Germany
- Department of Neurology and Center for Stroke Research Berlin; Charité, Universitätsmedizin Berlin; Charité Campus Mitte Berlin Germany
| | - Frank Heppner
- Institute for Neuropathology; Charité, Universitätsmedizin Berlin; Charité Campus Mitte Berlin Germany
| | - Rainer Glass
- Neurosurgical Research; University Clinics Munich (LMU); Munich Germany
| | - Susanne A. Wolf
- Cellular Neurosciences; Max Delbrück Centre for Molecular Medicine; Berlin Germany
| | - Helmut Kettenmann
- Cellular Neurosciences; Max Delbrück Centre for Molecular Medicine; Berlin Germany
| |
Collapse
|
39
|
Identification of biomarkers for hepatocellular carcinoma using network-based bioinformatics methods. Eur J Med Res 2013; 18:35. [PMID: 24083576 PMCID: PMC4016278 DOI: 10.1186/2047-783x-18-35] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2013] [Accepted: 08/30/2013] [Indexed: 01/06/2023] Open
Abstract
Background Hepatocellular carcinoma (HCC) is one of the most common types of cancer worldwide. Despite several efforts to elucidate molecular mechanisms involved in this cancer, they are still not fully understood. Methods To acquire further insights into the molecular mechanisms of HCC, and to identify biomarkers for early diagnosis of HCC, we downloaded the gene expression profile on HCC with non-cancerous liver controls from the Gene Expression Omnibus (GEO) and analyzed these data using a combined bioinformatics approach. Results The dysregulated pathways and protein-protein interaction (PPI) network, including hub nodes that distinguished HCCs from non-cancerous liver controls, were identified. In total, 29 phenotype-related differentially expressed genes were included in the PPI network. Hierarchical clustering showed that the gene expression profile of these 29 genes was able to differentiate HCC samples from non-cancerous liver samples. Among these genes, CDC2 (Cell division control protein 2 homolog), MMP2 (matrix metalloproteinase-2) and DCN (Decorin were the hub nodes in the PPI network. Conclusions This study provides a portfolio of targets useful for future investigation. However, experimental studies should be conducted to verify our findings.
Collapse
|
40
|
Lou X, Han X, Jin C, Tian W, Yu W, Ding D, Cheng L, Huang B, Jiang H, Lin B. SOX2 targets fibronectin 1 to promote cell migration and invasion in ovarian cancer: new molecular leads for therapeutic intervention. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2013; 17:510-8. [PMID: 23895273 DOI: 10.1089/omi.2013.0058] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Abstract Ovarian cancer ranks as the second most common tumor of the female reproductive system, with a large burden on global public health. Therefore, the identification of novel molecular targets and diagnostics is an urgent need for many women affected by this disease. To this end, the human transcription factor SOX2 is involved in a wide range of pathophysiological roles, such as the maintenance of stem cell characteristics and carcinogenesis. To date, in most studies, SOX2 has been shown to promote the development of cancer, although its inhibitory roles in cancer have also been reported. However, to the best of our knowledge, the role of SOX2, specifically in ovarian cancer cells, has not been examined in detail. In this article, we report, for the first time, that SOX2 promotes migration, invasion, and clonal formation of ovarian cancer cells. We further observed that SOX2 targeted FN1, a key gene that regulates cell migration in ovarian cancer. Our findings collectively suggest that the SOX2-FN1 axis is a key pathway in mediating the migration and invasion of ovarian cancer cells. This pathway offers crucial molecular insights and promises to develop putative candidate therapeutic interventions in women with ovarian cancer.
Collapse
Affiliation(s)
- Xiaoyan Lou
- 1 Systems Biology Division and Propriumbio Research Center, Zhejiang-California International Nanosystems Institute (ZCNI), Zhejiang University , Hangzhou, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Lee H, Chang KW, Yang HY, Lin PW, Chen SU, Huang YL. MT1-MMP regulates MMP-2 expression and angiogenesis-related functions in human umbilical vein endothelial cells. Biochem Biophys Res Commun 2013; 437:232-8. [PMID: 23796708 DOI: 10.1016/j.bbrc.2013.06.046] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Accepted: 06/13/2013] [Indexed: 01/06/2023]
Abstract
Membrane type 1 (MT1)-MMP is a member of matrix metalloproteinases (MMPs) that regulates extracellular matrix remodeling. In addition, MT1-MMP also serves as a multi-functional protein. However, the functional role of MT1-MMP in human endothelial cells remains unclear. In this study we use real-time PCR and Western blotting to demonstrate for the first time that MMP-2 expression is regulated by MT1-MMP in human endothelial cells. Moreover, MMP-2 activity is also modulated by MT1-MMP. In addition we found that endothelial cells, ECM adhesion and human endothelial cell tube formation, which are known to be regulated by MMP-2, are blocked by MT1-MMP siRNA. These results suggest that MT1-MMP plays an important role in regulating angiogenesis in human endothelial cells.
Collapse
Affiliation(s)
- Hsinyu Lee
- Department of Life Science, National Taiwan University, Taipei, Taiwan, ROC
| | | | | | | | | | | |
Collapse
|
42
|
Watanabe A, Hosino D, Koshikawa N, Seiki M, Suzuki T, Ichikawa K. Critical role of transient activity of MT1-MMP for ECM degradation in invadopodia. PLoS Comput Biol 2013; 9:e1003086. [PMID: 23737743 PMCID: PMC3667784 DOI: 10.1371/journal.pcbi.1003086] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Accepted: 04/19/2013] [Indexed: 11/18/2022] Open
Abstract
Focal degradation of extracellular matrix (ECM) is the first step in the invasion of cancer cells. MT1-MMP is a potent membrane proteinase employed by aggressive cancer cells. In our previous study, we reported that MT1-MMP was preferentially located at membrane protrusions called invadopodia, where MT1-MMP underwent quick turnover. Our computer simulation and experiments showed that this quick turnover was essential for the degradation of ECM at invadopodia (Hoshino, D., et al., (2012) PLoS Comp. Biol., 8: e1002479). Here we report on characterization and analysis of the ECM-degrading activity of MT1-MMP, aiming at elucidating a possible reason for its repetitive insertion in the ECM degradation. First, in our computational model, we found a very narrow transient peak in the activity of MT1-MMP followed by steady state activity. This transient activity was due to the inhibition by TIMP-2, and the steady state activity of MT1-MMP decreased dramatically at higher TIMP-2 concentrations. Second, we evaluated the role of the narrow transient activity in the ECM degradation. When the transient activity was forcibly suppressed in computer simulations, the ECM degradation was heavily suppressed, indicating the essential role of this transient peak in the ECM degradation. Third, we compared continuous and pulsatile turnover of MT1-MMP in the ECM degradation at invadopodia. The pulsatile insertion showed basically consistent results with the continuous insertion in the ECM degradation, and the ECM degrading efficacy depended heavily on the transient activity of MT1-MMP in both models. Unexpectedly, however, low-frequency/high-concentration insertion of MT1-MMP was more effective in ECM degradation than high-frequency/low-concentration pulsatile insertion even if the time-averaged amount of inserted MT1-MMP was the same. The present analysis and characterization of ECM degradation by MT1-MMP together with our previous report indicate a dynamic nature of MT1-MMP at invadopodia and the importance of its transient peak in the degradation of the ECM.
Collapse
Affiliation(s)
- Ayako Watanabe
- Division of Mathematical Oncology, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo, Japan
| | - Daisuke Hosino
- Division of Cancer Cell Research, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo, Japan
| | - Naohiko Koshikawa
- Division of Cancer Cell Research, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo, Japan
| | - Motoharu Seiki
- Division of Cancer Cell Research, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo, Japan
- JST, CREST, Chiyoda-ku, Tokyo, Japan
| | - Takashi Suzuki
- JST, CREST, Chiyoda-ku, Tokyo, Japan
- Division of Mathematical Science, Graduate School of Engineering Science, Osaka University, Toyonaka, Osaka, Japan
| | - Kazuhisa Ichikawa
- Division of Mathematical Oncology, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo, Japan
- JST, CREST, Chiyoda-ku, Tokyo, Japan
- * E-mail:
| |
Collapse
|
43
|
Han KY, Fahd DC, Tshionyi M, Allemann N, Jain S, Chang JH, Azar DT. MT1-MMP modulates bFGF-induced VEGF-A expression in corneal fibroblasts. Protein Pept Lett 2013; 19:1334-9. [PMID: 22670674 DOI: 10.2174/092986612803521639] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2012] [Revised: 05/10/2012] [Accepted: 05/11/2012] [Indexed: 11/22/2022]
Abstract
The cornea is physiologically avascular. Following a corneal injury, wound healing often proceeds without neovascularization (NV); however, corneal NV may be induced during wound healing in certain inflammatory, infectious, degenerative, and traumatic states. Such states disrupt the physiologic balance between pro-angiogenic and antiangiogenic mediators, favoring angiogenesis. Contributors to such states are matrix metalloproteinases (MMPs), which are key factors in both extracellular matrix remodeling and angiogenesis. Similarly, vascular endothelial growth factor A (VEGF-A) and basic fibroblast growth factor (bFGF) exert pro-angiogenic effects. Here, we elaborate on the facilitative role of MMPs-specifically Membrane Type 1 MMP (MT1-MMP, MMP14)-in corneal NV. Additionally, we provide new insight into the signaling relating to MT1-MMP, Ras, and ERK in the bFGF-induced VEGF-A expression pathways within the corneal fibroblasts.
Collapse
Affiliation(s)
- Kyu-Yeon Han
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, University of Illinois at Chicago, Chicago, IL 60612, USA
| | | | | | | | | | | | | |
Collapse
|
44
|
Yang CC, Zhu LF, Xu XH, Ning TY, Ye JH, Liu LK. Membrane Type 1 Matrix Metalloproteinase induces an epithelial to mesenchymal transition and cancer stem cell-like properties in SCC9 cells. BMC Cancer 2013; 13:171. [PMID: 23548172 PMCID: PMC3637131 DOI: 10.1186/1471-2407-13-171] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2012] [Accepted: 01/30/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Tissue invasion and metastasis are acquired abilities of cancer and related to the death in oral squamous cell carcinoma (OSCC). Emerging observations indicate that the epithelial-to-mesenchymal transition (EMT) is associated with tumor progression and the generation of cells with cancer stem cells (CSCs) properties. Membrane Type 1 Matrix Metalloproteinase (MT1-MMP) is a cell surface proteinase, which is involved in degrading extracellular matrix components that can promote tumor invasion and cell migration. METHODS In the current study, we utilized SCC9 cells stably transfected with an empty vector (SCC9-N) or a vector encoding human MT1-MMP (SCC9-M) to study the role of MT1-MMP in EMT development. RESULTS Upon up-regulation of MT1-MMP, SCC9-M cells underwent EMT, in which they presented a fibroblast-like phenotype and had a decreased expression of epithelial markers (E-cadherin, cytokeratin18 and β-catenin) and an increased expression of mesenchymal markers (vimentin and fibronectin). We further demonstrated that MT1-MMP-induced morphologic changes increased the level of Twist and ZEB, and were dependent on repressing the transcription of E-cadherin. These activities resulted in low adhesive, high invasive abilities of the SCC9-M cells. Furthermore, MT1-MMP-induced transformed cells exhibited cancer stem cell (CSC)-like characteristics, such as low proliferation, self-renewal ability, resistance to chemotherapeutic drugs and apoptosis, and expression of CSCs surface markers. CONCLUSIONS In conclusion, our study indicates that overexpression of MT1-MMP induces EMT and results in the acquisition of CSC-like properties in SCC9 cells. Our growing understanding of the mechanism regulating EMT may provide new targets against invasion and metastasis in OSCC.
Collapse
Affiliation(s)
- Cong-Chong Yang
- Department of Basic Science of Stomatology, Institute of Stomatology, Nanjing Medical University, Nanjing, People's Republic of China
| | | | | | | | | | | |
Collapse
|
45
|
Vihinen P, Ala-Aho R, Kähäri VM. Diagnostic and prognostic role of matrix metalloproteases in cancer. ACTA ACUST UNITED AC 2013; 2:1025-39. [PMID: 23495924 DOI: 10.1517/17530059.2.9.1025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Matrix metalloproteases (MMPs) are key players in the progression and metastasis of cancer. MMPs cleave extracellular matrix components and in this way promote tumor growth, invasion and vascularization. MMPs also affect tumor progression by regulating availability and activity of growth factors, inflammatory cytokines and chemokines. Accordingly, several MMPs have been found to serve as prognostic indicators in solid tumors. Usually the increased levels of MMPs in patients' tumor tissue or serum/plasma are associated with poor outcome. Interestingly, recent results show that certain MMPs also serve as tumor suppressors. OBJECTIVE This review discusses the latest view on MMPs as diagnostic and prognostic indicators in cancer patients. METHODS Studies with clinical samples of 70 or more patients are included in particular. In addition, the possible roles of MMPs in future molecular diagnostics and in the evaluation of therapeutic responses are discussed. CONCLUSION MMP-9 in particular has shown prognostic value in various types of tumor, and its measurement in circulation, urine or tumor tissue might help in clinical surveillance of otherwise problematic patient cases. There is upcoming new knowledge on MMPs in therapy response evaluation, in which MMPs might be useful together with CT scans and other clinically more established prognostic factors. Certain MMPs have a dual role in terms of cancer-modulating properties and thus it is essential to evaluate their expression and function in tumor cells and host environment to select validated therapy targets but spare MMP antitargets.
Collapse
Affiliation(s)
- Pia Vihinen
- Turku University Hospital, Department of Oncology and Radiotherapy, POB 52, FIN-20521 Turku, Finland +358 2 313 0804 ; +358 2 313 2809 ;
| | | | | |
Collapse
|
46
|
Kim YH, Kwon HJ, Kim DS. Matrix metalloproteinase 9 (MMP-9)-dependent processing of βig-h3 protein regulates cell migration, invasion, and adhesion. J Biol Chem 2012; 287:38957-69. [PMID: 23019342 DOI: 10.1074/jbc.m112.357863] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Cell migration is critically involved in inflammation, cancer, and development. In this study, transforming growth factor-β-induced protein (βig-h3) was identified as a substrate of matrix metalloproteinase-9 (MMP-9) by site-directed mutagenesis. βig-h3 has two cleavage sites with the consensus sequence Pro-Xaa-Xaa-Hy-(Ser/Thr) (Hy is a hydrophobic amino acid) (PGSFT beginning at amino acid 135 and PPMGT beginning at amino acid 501). Using recombinant human βig-h3 and MMP-9, βig-h3 from βig-h3-transfected HEK293F cells, and MMP-9 from MMP-9-transfected HEK293F cells, human macrophages, and neutrophils, we found that MMP-9 proteolytically cleaves βig-h3. Cleavage leads to the loss of its adhesive property and its release from extracellular matrix proteins, collagen IV, and fibronectin. Spheroids formed by increased cell-cell interactions were observed in βig-h3-transfected HEK293F cells but not in vehicle-transfected HEK293F cells. In human glioma U87MG cells, MMP-9 constitutive overexpression resulted in endogenous βig-h3 cleavage. βig-h3 cleavage by MMP-9 led to increased cell invasion, and βig-h3 knockdown also resulted in increased cell invasion. The βig-h3 fragment cleaved by MMP-9 could bind to the surface of macrophages, and it may play a role as a peptide chemoattractant by inducing macrophage migration via focal adhesion kinase/Src-mediated signal activation. Thus, intact βig-h3 is responsible for cell migration inhibition, cell-cell contact, and cell-extracellular matrix interaction. Experimental evidence indicates that MMP-9-cleaved βig-h3 plays a role in MMP-9-mediated tumor cell and macrophage migration.
Collapse
Affiliation(s)
- Yeon Hyang Kim
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Korea.
| | | | | |
Collapse
|
47
|
KUNG CHANGI, CHEN CHENGYI, YANG CHIHCHANG, LIN CHENGYU, CHEN TIENHUA, WANG HWAISHI. Enhanced membrane-type 1 matrix metalloproteinase expression by hyaluronan oligosaccharides in breast cancer cells facilitates CD44 cleavage and tumor cell migration. Oncol Rep 2012; 28:1808-14. [DOI: 10.3892/or.2012.1993] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2012] [Accepted: 08/03/2012] [Indexed: 11/06/2022] Open
|
48
|
Myochin T, Hanaoka K, Komatsu T, Terai T, Nagano T. Design Strategy for a Near-Infrared Fluorescence Probe for Matrix Metalloproteinase Utilizing Highly Cell Permeable Boron Dipyrromethene. J Am Chem Soc 2012; 134:13730-7. [DOI: 10.1021/ja303931b] [Citation(s) in RCA: 108] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Takuya Myochin
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo
113-0033, Japan
| | - Kenjiro Hanaoka
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo
113-0033, Japan
| | - Toru Komatsu
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo
113-0033, Japan
| | - Takuya Terai
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo
113-0033, Japan
| | - Tetsuo Nagano
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo
113-0033, Japan
| |
Collapse
|
49
|
Ponte AL, Ribeiro-Fleury T, Chabot V, Gouilleux F, Langonné A, Hérault O, Charbord P, Domenech J. Granulocyte-colony-stimulating factor stimulation of bone marrow mesenchymal stromal cells promotes CD34+ cell migration via a matrix metalloproteinase-2-dependent mechanism. Stem Cells Dev 2012; 21:3162-72. [PMID: 22651889 DOI: 10.1089/scd.2012.0048] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Human hematopoietic stem/progenitor cells (HSPCs) can be mobilized into the circulation using granulocyte-colony stimulating factor (G-CSF), for graft collection in view of hematopoietic transplantation. This process has been related to bone marrow (BM) release of serine proteases and of the matrix metalloproteinase-9 (MMP-9). Yet, the role of these mediators in HSC egress from their niches remains questionable, because they are produced by nonstromal cells (mainly neutrophils and monocytes/macrophages) that are not a part of the niche. We show here that the G-CSF receptor (G-CSFR) is expressed by human BM mesenchymal stromal/stem cells (MSCs), and that G-CSF prestimulation of MSCs enhances the in vitro trans-stromal migration of CD34+ cells. Zymography analysis indicates that pro-MMP-2 (but not pro-MMP-9) is expressed in MSCs, and that G-CSF treatment increases its expression and induces its activation at the cell membrane. We further demonstrate that G-CSF-stimulated migration depends on G-CSFR expression and is mediated by a mechanism that involves MMPs. These results suggest a molecular model whereby G-CSF infusion may drive, by the direct action on MSCs, HSPC egress from BM niches via synthesis and activation of MMPs. In this model, MMP-2 instead of MMP-9 is implicated, which constitutes a major difference with mouse mobilization models.
Collapse
Affiliation(s)
- Adriana López Ponte
- UPRES-EA3855, IFR135, Faculté de Médecine, Université François Rabelais Tours, France
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Gu G, Zhao D, Yin Z, Liu P. BST-2 binding with cellular MT1-MMP blocks cell growth and migration via decreasing MMP2 activity. J Cell Biochem 2012; 113:1013-21. [PMID: 22065321 DOI: 10.1002/jcb.23433] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
MT1-MMP (membrane type 1-matrix metalloproteinase) plays important roles in cell growth and tumor invasion via mediating cleavage of MMP2/gelatinase A and a variety of substrates including type I collagen. BST-2 (bone marrow stromal cell antigen 2) is a membrane tetherin whose expression dramatically reduces the release of a broad range of enveloped viruses including HIV from infected cells. In this study, we provided evidence that both transient and IFN-α induced BST-2 could decrease the activity of MMP2 via binding to cellular MT1-MMP on its C-terminus and inhibiting its proteolytic activity; and finally block cell growth and migration. Zymography gel and Western blot experiments demonstrated that BST-2 decreased MMP2 activity, but no effect on the expression of MMP2 and MT1-MMP genes. Confocal and immunoprecipitation data showed that BST-2 co-localized and interacted with MT1-MMP. This interaction inhibited the proteolytic enzyme activity of MT1-MMP, and blocked the activation of proMMP2. Experimental results of C-terminus deletion mutant of MT1-MMP showed that activity of MMP2 was no change and also no interaction existed between the mutant and BST-2 after co-transfection with the mutant and BST-2. It meant that C-terminus of MT1-MMP played a key role in the interaction with BST-2. In addition, cell growth in 3D type I collagen gel lattice and cell migration were all inhibited by BST-2. Taken together, BST-2, as a membrane protein and a tetherin of enveloped viruses, was a novel inhibitor of MT1-MMP and could be considerable as an inhibitor of cancer cell growth and migration on clinic.
Collapse
Affiliation(s)
- Gongping Gu
- Jiangsu Province Key Laboratory for Molecular and Medicine Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, Jiangsu 210097, PR China
| | | | | | | |
Collapse
|