151
|
Cancer-associated fibroblasts modulate growth factor signaling and extracellular matrix remodeling to regulate tumor metastasis. Biochem Soc Trans 2017; 45:229-236. [PMID: 28202677 DOI: 10.1042/bst20160387] [Citation(s) in RCA: 379] [Impact Index Per Article: 47.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 11/13/2016] [Accepted: 12/08/2016] [Indexed: 02/07/2023]
Abstract
Cancer-associated fibroblasts (CAFs) are major components of the surrounding stroma of carcinomas that emerge in the tumor microenvironment as a result of signals derived from the cancer cells. Biochemical cross-talk between cancer cells and CAFs as well as mechanical remodeling of the stromal extracellular matrix (ECM) by CAFs are important contributors to tumor cell migration and invasion, which are critical for cancer progression from a primary tumor to metastatic disease. In this review, we discuss key paracrine signaling pathways between CAFs and cancer cells that promote cancer cell migration and invasion. In addition, we discuss physical changes that CAFs exert on the stromal ECM to facilitate migration and invasion of cancer cells.
Collapse
|
152
|
Tumor-stroma ratio(TSR) as a potential novel predictor of prognosis in digestive system cancers: A meta-analysis. Clin Chim Acta 2017; 472:64-68. [PMID: 28729135 DOI: 10.1016/j.cca.2017.07.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 07/03/2017] [Accepted: 07/03/2017] [Indexed: 12/15/2022]
Abstract
MAIN PROBLEM The tumor-stroma ratio (TSR) has been reported as a prognosis predictor in multiple cancers. The aim of this meta-analysis was to investigate the potential value of TSR as a prognostic predictor of cancer in the digestive system. METHODS We searched PubMed, Embase, Elsevier and Web of Science. All studies exploring the association of TSR with overall survival (OS) or disease-free survival (DFS), and lymph node metastasis (LNM) were identified. RESULTS In total, eight studies were eligible for analysis, and they included 1959 patients. Meta-analysis showed that the low TSR in the tumor could predict poor overall survival (OS) in multiple cancers (pooled Hazard Ratio [HR]: 2.15, 95%CI: 1.80-2.57, P<0.00001, fixed effects). For disease-free survival (DFS), low TSR was also a significant predictor (pooled Hazard Ratio [HR]: 2.31, 95%CI: 1.88-2.83, P<0.00001, fixed effects). In addition, low TSR was correlated with tumor stage. DISCUSSION The tumor-stroma ratio (TSR) may potentially serve as a poor prognostic predictor for the metastasis and prognosis of cancer.
Collapse
|
153
|
Bahrami A, Hassanian SM, Khazaei M, Hasanzadeh M, Shahidsales S, Maftouh M, Ferns GA, Avan A. The Therapeutic Potential of Targeting Tumor Microenvironment in Breast Cancer: Rational Strategies and Recent Progress. J Cell Biochem 2017; 119:111-122. [DOI: 10.1002/jcb.26183] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 06/01/2017] [Indexed: 01/05/2023]
Affiliation(s)
- Afsane Bahrami
- Department of Modern Sciences and Technologies, Faculty of MedicineMashhad University of Medical SciencesMashhadIran
- Metabolic Syndrome Research CenterMashhad University of Medical SciencesMashhadIran
- Student Research Committee, Faculty of MedicineMashhad University of Medical SciencesMashhadIran
| | - Seyed Mahdi Hassanian
- Metabolic Syndrome Research CenterMashhad University of Medical SciencesMashhadIran
- Department of Medical Biochemistry, School of MedicineMashhad University of Medical SciencesMashhadIran
| | - Majid Khazaei
- Department of Physiology, Faculty of MedicineMashhad University of Medical SciencesMashhadIran
| | - Malihe Hasanzadeh
- Department of Gynecology Oncology, Woman Health Research CenterMashhad University of Medical SciencesMashhadIran
| | | | - Mina Maftouh
- Metabolic Syndrome Research CenterMashhad University of Medical SciencesMashhadIran
| | - Gordon A. Ferns
- Brighton & Sussex Medical SchoolDivision of Medical EducationFalmer, BrightonSussex BN1 9PH, UK
| | - Amir Avan
- Metabolic Syndrome Research CenterMashhad University of Medical SciencesMashhadIran
- Cancer Research CenterMashhad University of Medical SciencesMashhadIran
| |
Collapse
|
154
|
Targeting Tumor Microenvironment: Effects of Chinese Herbal Formulae on Macrophage-Mediated Lung Cancer in Mice. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2017. [PMID: 28630636 PMCID: PMC5467330 DOI: 10.1155/2017/7187168] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Our previous studies have shown that Qing-Re-Huo-Xue (QRHX) formulae had significant anti-inflammatory effects in chronic airway diseases such as asthma and chronic obstructive lung disease. Here, we examined the effects of QRHX on lung cancer cell invasion and the potential associated mechanism(s), mainly polarization of macrophages in the tumor microenvironment. In vivo, QRHX both inhibited tumor growth and decreased the number of tumor-associated macrophages (TAMs) in mice with lung cancer. Further study indicated that QRHX inhibited cancer-related inflammation in tumor by decreasing infiltration of TAMs and IL-6 and TNF-α production and meanwhile decreased arginase 1 (Arg-1) expression and increased inducible NO synthase (iNOS) expression. QRHX could markedly inhibit CD31 and VEGF protein expression. Additionally, CXCL12/CXCR4 expression and JAK2/STAT3 phosphorylation were reduced in QRHX treatment group. Thus, we draw that QRHX played a more important role in inhibiting tumor growth by regulating TAMs in mice, which was found to be associated with the inhibition of inflammation and the CXCL12/CXCR4/JAK2/STAT3 signaling pathway.
Collapse
|
155
|
Adenylate kinase hCINAP determines self-renewal of colorectal cancer stem cells by facilitating LDHA phosphorylation. Nat Commun 2017; 8:15308. [PMID: 28516914 PMCID: PMC5454382 DOI: 10.1038/ncomms15308] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 03/17/2017] [Indexed: 12/18/2022] Open
Abstract
Targeting the specific metabolic phenotypes of colorectal cancer stem cells (CRCSCs) is an innovative therapeutic strategy for colorectal cancer (CRC) patients with poor prognosis and relapse. However, the context-dependent metabolic traits of CRCSCs remain poorly elucidated. Here we report that adenylate kinase hCINAP is overexpressed in CRC tissues. Depletion of hCINAP inhibits invasion, self-renewal, tumorigenesis and chemoresistance of CRCSCs with a loss of mesenchymal signature. Mechanistically, hCINAP binds to the C-terminal domain of LDHA, the key regulator of glycolysis, and depends on its adenylate kinase activity to promote LDHA phosphorylation at tyrosine 10, resulting in the hyperactive Warburg effect and the lower cellular ROS level and conferring metabolic advantage to CRCSC invasion. Moreover, hCINAP expression is positively correlated with the level of Y10-phosphorylated LDHA in CRC patients. This study identifies hCINAP as a potent modulator of metabolic reprogramming in CRCSCs and a promising drug target for CRC invasion and metastasis.
Collapse
|
156
|
Simultaneous Detection of EGFR and VEGF in Colorectal Cancer using Fluorescence-Raman Endoscopy. Sci Rep 2017; 7:1035. [PMID: 28432289 PMCID: PMC5430917 DOI: 10.1038/s41598-017-01020-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 03/24/2017] [Indexed: 12/14/2022] Open
Abstract
Fluorescence endomicroscopy provides quick access to molecular targets, while Raman spectroscopy allows the detection of multiple molecular targets. Using a simultaneous fluorescence-Raman endoscopic system (FRES), we herein demonstrate its potential in cancer diagnosis in an orthotopically induced colorectal cancer (CRC) xenograft model. In the model, epidermal growth factor receptor (EGFR) and vascular endothelial growth factor (VEGF) were targeted with antibody-conjugated fluorescence and surface-enhanced Raman scattering (F-SERS) dots. FRES demonstrated fast signal detection and multiplex targeting ability using fluorescence and Raman signals to detect the F-SERS dots. In addition, FRES showed a multiplex targeting ability even on a subcentimeter-sized CRC after spraying with a dose of 50 µg F-SERS dots. In conclusion, molecular characteristics of tumor cells (EGFR in cancer cell membranes) and tumor microenvironments (VEGF in the extracellular matrix) could be simultaneously investigated when performing a colonoscopy.
Collapse
|
157
|
Secondini C, Coquoz O, Spagnuolo L, Spinetti T, Peyvandi S, Ciarloni L, Botta F, Bourquin C, Rüegg C. Arginase inhibition suppresses lung metastasis in the 4T1 breast cancer model independently of the immunomodulatory and anti-metastatic effects of VEGFR-2 blockade. Oncoimmunology 2017; 6:e1316437. [PMID: 28680747 DOI: 10.1080/2162402x.2017.1316437] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 03/31/2017] [Accepted: 03/31/2017] [Indexed: 01/18/2023] Open
Abstract
Tumor angiogenesis promotes tumor growth and metastasis. Anti-angiogenic therapy in combination with chemotherapy is used for the treatment of metastatic cancers, including breast cancer but therapeutic benefits are limited. Mobilization and accumulation of myeloid-derived suppressor cells (MDSC) during tumor progression and therapy have been implicated in metastasis formation and resistance to anti-angiogenic treatments. Here, we used the 4T1 orthotopic syngenic mouse model of mammary adenocarcinoma to investigate the effect of VEGF/VEGFR-2 axis inhibition on lung metastasis, MDSC and regulatory T cells (Tregs). We show that treatment with the anti-VEGFR-2 blocking antibody DC101 inhibits primary tumor growth, angiogenesis and lung metastasis. DC101 treatment had no effect on MDSC mobilization, but partially attenuated the inhibitory effect of mMDSC on T cell proliferation and decreased the frequency of Tregs in primary tumors and lung metastases. Strikingly, DC101 treatment induced the expression of the immune-suppressive molecule arginase I in mMDSC. Treatment with the arginase inhibitor Nω-hydroxy-nor-Arginine (Nor-NOHA) reduced the inhibitory effect of MDSC on T cell proliferation and inhibited number and size of lung metastasis but had little or no additional effects in combination with DC101. In conclusion, DC101 treatment suppresses 4T1 tumor growth and metastasis, partially reverses the inhibitory effect of mMDSC on T cell proliferation, decreases Tregs in tumors and increases arginase I expression in mMDSC. Arginase inhibition suppresses lung metastasis independently of DC101 effects. These observations contribute to the further characterization of the immunomodulatory effect of anti-VEGF/VEGFR2 therapy and provide a rationale to pursue arginase inhibition as potential anti-metastatic therapy.
Collapse
Affiliation(s)
- Chiara Secondini
- Department of Medicine, Faculty of Science, University of Fribourg, Fribourg, Switzerland
| | - Oriana Coquoz
- Department of Medicine, Faculty of Science, University of Fribourg, Fribourg, Switzerland
| | - Lorenzo Spagnuolo
- Department of Medicine, Faculty of Science, University of Fribourg, Fribourg, Switzerland.,School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, Lausanne, Switzerland
| | - Thibaud Spinetti
- Department of Medicine, Faculty of Science, University of Fribourg, Fribourg, Switzerland
| | - Sanam Peyvandi
- Department of Medicine, Faculty of Science, University of Fribourg, Fribourg, Switzerland
| | - Laura Ciarloni
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, Lausanne, Switzerland
| | - Francesca Botta
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, Lausanne, Switzerland
| | - Carole Bourquin
- Department of Medicine, Faculty of Science, University of Fribourg, Fribourg, Switzerland.,School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, Lausanne, Switzerland
| | - Curzio Rüegg
- Department of Medicine, Faculty of Science, University of Fribourg, Fribourg, Switzerland.,Division of Experimental Oncology, University Hospital and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
158
|
Abstract
Now clear evidences are available to support the hypothesis that inflammation accelerates the conditions including events and molecules that reach to various types of cancers. Inflammation is a normal response to infection containing the innate and adaptive immune systems. However, when allowed to continue, unresolved, perturbation of cellular microenvironment takes place; therefore, it leads to adaptations in genes that are linked to cancer. In addition, a lot of data are accessible confirming the concept that tumour microenvironment is orchestrated by various inflammatory cells and goes to neoplastic process and finally invasion, migration and metastasis. However, infiltrations of leucocytes lead to angiogenesis, propagation and invasion. An inflammatory microenvironment that perhaps fostering impact of angiogenesis include cytokines, chemokines, enzymes and growth factors that play key role for expansion and invasion of cancer cells. This insight highlights the pathogenesis of inflammation-associated cancers and also touches and fosters the role of acetamides for the treatment and chemoprevention of carcinomas that are allied with inflammation.
Collapse
Affiliation(s)
- Priyanka Rani
- a Department of Chemistry , School of Sciences, IFTM University Moradabad , Uttar Pradesh , India
| | - Dilipkumar Pal
- b Department of Pharmaceutical Sciences , Guru Ghasidas Vishwavidyalaya (A Central University) , Koni, Bilaspur , CG , India
| | - Rahul Rama Hegde
- c Department of Pharmaceutics , School of Pharmaceutical Sciences, IFTM University Moradabad , Uttar Pradesh , India
| | - Syed Riaz Hashim
- d Department of Chemistry , School of Pharmaceutical Sciences, IFTM University Moradabad , Uttar Pradesh , India
| |
Collapse
|
159
|
Kanda Y, Osaki M, Onuma K, Sonoda A, Kobayashi M, Hamada J, Nicolson GL, Ochiya T, Okada F. Amigo2-upregulation in Tumour Cells Facilitates Their Attachment to Liver Endothelial Cells Resulting in Liver Metastases. Sci Rep 2017; 7:43567. [PMID: 28272394 PMCID: PMC5341090 DOI: 10.1038/srep43567] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 01/25/2017] [Indexed: 02/06/2023] Open
Abstract
Since liver metastasis is the main cause of death in cancer patients, we attempted to identify the driver gene involved. QRsP-11 fibrosarcoma cells were injected into the spleens of syngeneic mice to isolate tumour sub-populations that colonize the liver. Cells from liver metastatic nodules were established and subsequently injected intrasplenically for selection. After 12 cycles, the cell subline LV12 was obtained. Intravenous injection of LV12 cells produced more liver metastases than QRsP-11 cells, whereas the incidence of lung metastases was similar to that of QRsP-11 cells. LV12 cells adhered to liver-derived but not to lung-derived endothelial cells. DNA chip analysis showed that amphoterin-induced gene and open reading frame 2 (Amigo2) was overexpressed in LV12 cells. siRNA-mediated knockdown of Amigo2 expression in LV12 cells attenuated liver endothelial cell adhesion. Ex vivo imaging showed that suppression of Amigo2 in luciferase-expressing LV12 cells reduced attachment/metastasis to liver to the same level as that observed with QRsP-11 cells. Forced expression of Amigo2 in QRsP-11 cells increased liver endothelial cell adhesion and liver metastasis. Additionally, Amigo2 expression in human cancers was higher in liver metastatic lesions than in primary lesions. Thus, Amigo2 regulated tumour cell adhesion to liver endothelial cells and formation of liver metastases.
Collapse
Affiliation(s)
- Yusuke Kanda
- Division of Pathological Biochemistry, Tottori University Faculty of Medicine, Yonago, Japan
| | - Mitsuhiko Osaki
- Division of Pathological Biochemistry, Tottori University Faculty of Medicine, Yonago, Japan.,Chromosome Engineering Research Center, Tottori University, Yonago, Japan
| | - Kunishige Onuma
- Division of Pathological Biochemistry, Tottori University Faculty of Medicine, Yonago, Japan
| | - Ayana Sonoda
- Division of Pathological Biochemistry, Tottori University Faculty of Medicine, Yonago, Japan
| | - Masanobu Kobayashi
- Health Sciences University of Hokkaido, School of Nursing and Social Services, Ishikari-Tobetsu, Japan
| | - Junichi Hamada
- Health Sciences University of Hokkaido, School of Nursing and Social Services, Ishikari-Tobetsu, Japan
| | - Garth L Nicolson
- Department of Molecular Pathology, The Institute for Molecular Medicine, South Laguna Beach, CA, USA
| | - Takahiro Ochiya
- Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, Tsukiji, Chuo-ku, Tokyo, Japan
| | - Futoshi Okada
- Division of Pathological Biochemistry, Tottori University Faculty of Medicine, Yonago, Japan.,Chromosome Engineering Research Center, Tottori University, Yonago, Japan
| |
Collapse
|
160
|
Bergamo A, Pelillo C, Chambery A, Sava G. Influence of components of tumour microenvironment on the response of HCT-116 colorectal cancer to the ruthenium-based drug NAMI-A. J Inorg Biochem 2017; 168:90-97. [DOI: 10.1016/j.jinorgbio.2016.11.031] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 11/22/2016] [Accepted: 11/30/2016] [Indexed: 12/30/2022]
|
161
|
Vinnakota K, Zhang Y, Selvanesan BC, Topi G, Salim T, Sand-Dejmek J, Jönsson G, Sjölander A. M2-like macrophages induce colon cancer cell invasion via matrix metalloproteinases. J Cell Physiol 2017; 232:3468-3480. [PMID: 28098359 DOI: 10.1002/jcp.25808] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 01/13/2017] [Accepted: 01/17/2017] [Indexed: 12/19/2022]
Abstract
The inflammatory milieu plays an important role in colon cancer development and progression. Previously, we have shown that tumor-associated macrophages (TAMs), an important component of the tumor microenvironment, are enriched in tumors compared with normal tissue and confer a poorer prognosis. In the present study, we found that matrix metallopeptidase-9 (MMP-9), which degrades extracellular matrix proteins, was increased in biopsies from colon cancer patients and in mouse xenografts with SW480 cell-derived tumors. SW480 colon cancer cells exposed to M2-like macrophage-conditioned medium (M2-medium) exhibited increased MMP-9 mRNA, protein expression and gelatinase activity. A similar effect was obtained by the addition of tumor necrosis factor-α (TNFα) and leukotriene D4 (LTD4 ). MMP-9 expression and activity were reduced by a TNFα blocking antibody adalimumab and a cysteinyl leukotriene receptor 1 (CysLTR1, the receptor for LTD4 ) antagonist montelukast. M2-medium also induced changes in the epithelial-mesenchymal transition (EMT) markers E-cadherin, β-catenin, vimentin, and snail in SW480 cells. We also found that both M2-medium and TNFα and LTD4 induced stabilization/nuclear translocation of β-catenin. Furthermore, we also observed an elongated phenotype that may indicate increased invasiveness, as confirmed in a collagen I invasion assay. M2-medium increased the invasive ability, and a similar effect was also obtained by the addition of TNFα and LTD4 . The specific MMP inhibitor I or adalimumab and montelukast reduced the number of invasive cells. In conclusion, our findings show that M2-medium enriched in TNFα and LTD4 promote colon cancer cell invasion via MMP-9 expression and activation and the induction of EMT.
Collapse
Affiliation(s)
- Katyayni Vinnakota
- Division of Cell and Experimental Pathology, Department of Translational Medicine, Clinical Research Centre, Lund University, Skåne University Hospital, Malmö, Sweden
| | - Yuan Zhang
- Division of Cell and Experimental Pathology, Department of Translational Medicine, Clinical Research Centre, Lund University, Skåne University Hospital, Malmö, Sweden
| | - Benson Chellakkan Selvanesan
- Division of Cell and Experimental Pathology, Department of Translational Medicine, Clinical Research Centre, Lund University, Skåne University Hospital, Malmö, Sweden
| | - Geriolda Topi
- Division of Cell and Experimental Pathology, Department of Translational Medicine, Clinical Research Centre, Lund University, Skåne University Hospital, Malmö, Sweden
| | - Tavga Salim
- Division of Cell and Experimental Pathology, Department of Translational Medicine, Clinical Research Centre, Lund University, Skåne University Hospital, Malmö, Sweden
| | - Janna Sand-Dejmek
- Division of Cell and Experimental Pathology, Department of Translational Medicine, Clinical Research Centre, Lund University, Skåne University Hospital, Malmö, Sweden
| | - Gunilla Jönsson
- Division of Cell and Experimental Pathology, Department of Translational Medicine, Clinical Research Centre, Lund University, Skåne University Hospital, Malmö, Sweden
| | - Anita Sjölander
- Division of Cell and Experimental Pathology, Department of Translational Medicine, Clinical Research Centre, Lund University, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
162
|
Al-Ghorbani M, Thirusangu P, Gurupadaswamy HD, Vigneshwaran V, Mohammed YHE, Prabhakar BT, Khanum SA. Synthesis of novel morpholine conjugated benzophenone analogues and evaluation of antagonistic role against neoplastic development. Bioorg Chem 2017; 71:55-66. [PMID: 28139247 DOI: 10.1016/j.bioorg.2017.01.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 11/10/2016] [Accepted: 01/15/2017] [Indexed: 10/20/2022]
Abstract
A series of novel 4-benzyl-morpholine-2-carboxylic acid N'-[2-(4-benzoyl-phenoxy)-acetyl]-hydrazide derivatives 8a-j has been synthesized from (4-hydroxy-aryl)-aryl methanones through a multi-step reaction sequence and then evaluated for anti-proliferative activity in vitro against various types of neoplastic cells of mouse and human such as DLA, EAC, MCF-7 and A549 cells. From the cytotoxic studies and structural activity relationship of compounds 8a-j, it is clear that methyl group on the B ring of benzophenone is essential for antiproliferative activity and bromo at ortho position (compound 8b) and methyl at para position (compound 8f) on A ring of benzophenone are significant for extensive anti-mitogenic activity. Investigation on clonogenesis and Fluorescence-activated cell sorting suggests that compounds 8b and 8f have the potency to exhibit the prolonged activity with cell cycle arrest on G2/M phase against cancer progression. Further, the compounds 8b and 8f inhibit murine ascites lymphoma through caspase activated DNase mediated apoptosis.
Collapse
Affiliation(s)
- Mohammed Al-Ghorbani
- Department of Chemistry, Yuvaraja's College, University of Mysore, Mysore 570005, Karnataka, India
| | - Prabhu Thirusangu
- Molecular Onco-medicine Laboratory, Postgraduate Department of Studies and Research in Biotechnology, Sahyadri Science College (A), Kuvempu University, Shimoga 577203, Karnataka, India
| | - H D Gurupadaswamy
- Department of Chemistry, Yuvaraja's College, University of Mysore, Mysore 570005, Karnataka, India
| | - V Vigneshwaran
- Molecular Onco-medicine Laboratory, Postgraduate Department of Studies and Research in Biotechnology, Sahyadri Science College (A), Kuvempu University, Shimoga 577203, Karnataka, India
| | - Yasser H E Mohammed
- Department of Chemistry, Yuvaraja's College, University of Mysore, Mysore 570005, Karnataka, India
| | - B T Prabhakar
- Molecular Onco-medicine Laboratory, Postgraduate Department of Studies and Research in Biotechnology, Sahyadri Science College (A), Kuvempu University, Shimoga 577203, Karnataka, India
| | - Shaukath Ara Khanum
- Department of Chemistry, Yuvaraja's College, University of Mysore, Mysore 570005, Karnataka, India.
| |
Collapse
|
163
|
Hughes C, Baker MJ. Can mid-infrared biomedical spectroscopy of cells, fluids and tissue aid improvements in cancer survival? A patient paradigm. Analyst 2017; 141:467-75. [PMID: 26501136 DOI: 10.1039/c5an01858g] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
This review will take a fresh approach from the patient perspective; offering insight into the applications of mid-infrared biomedical spectroscopy in a scenario whereby the patient presents with non-specific symptoms and via an extensive diagnostic process multiple lesions are discovered but no clear sign of the primary tumour; a condition known as cancer of unknown primary (CUP). With very limited options to diagnose the cancer origin, treatment options are likely to be ineffective and prognosis is consequentially very poor. CUP has not yet been targeted by infrared biospectroscopy, however, this timely, concise dissemination will focus on a series of research highlights and breakthroughs from the field for the management of a variety of cancer-related diseases - many examples of which have occurred within this year alone. The case for integration of mid-infrared (MIR) technology into clinical practice will be demonstrated largely via diagnostic, but also therapeutic and prognostic avenues by means of including cytological, bio-fluid and tissue analysis. The review is structured around CUP but is relevant for all cancer diagnoses. Infrared spectroscopy is fast developing a reputation as a valid and powerful tool for the detection and diagnosis of cancer using a variety of sample formats. The technology will produce data and tools that are designed to complement routine clinical practice; enhancing the ability of the clinician to make a reliable and non-subjective decision and enabling decreased levels of mortality and morbidity and gains in patient quality of life.
Collapse
Affiliation(s)
- Caryn Hughes
- School of Chemical Engineering & Analytical Sciences, Faculty of Engineering & Physical Science, University of Manchester, Brunswick Street, Manchester, M13 9PL, UK. and WestCHEM, Department of Pure and Applied Chemistry, University of Strathclyde, Technology and Innovation Centre, 99 George Street, Glasgow, G1 1RD, UK.
| | - Matthew J Baker
- WestCHEM, Department of Pure and Applied Chemistry, University of Strathclyde, Technology and Innovation Centre, 99 George Street, Glasgow, G1 1RD, UK.
| |
Collapse
|
164
|
Majidinia M, Yousefi B. Breast tumor stroma: A driving force in the development of resistance to therapies. Chem Biol Drug Des 2017; 89:309-318. [PMID: 28042683 DOI: 10.1111/cbdd.12893] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2016] [Revised: 09/15/2016] [Accepted: 10/11/2016] [Indexed: 12/12/2022]
Abstract
Breast cancer is the most common cancer and the second leading cause of cancer-related death in women worldwide. In spite of huge advancements in early detection and ever-increasing knowledge of breast cancer biology, approximately 30% of patients with early-stage breast cancer experience disease recurrence. Most patients are chemosensitive and cancer free immediately after the treatment. About 50% to 70% of breast cancer patients, however, will relapse within 1 year. Such a relapse is usually concomitant with adenocarcinoma cells acquiring a chemoresistant phenotype. Both de novo and acquired chemoresistance are poorly understood and present a major burden in the treatment of breast cancer. Although, previously, chemoresistance was largely linked to genetic alterations within the cancer cells, recent investigations are indicating that chemoresistance can also be associated with the tumor microenvironment. Nowadays, it is widely believed that tumor microenvironment is a key player in tumor progression and response to treatment. In this study, we will review the interactions of breast tumor cells with their microenvironment, present the latest research on the resistance mediated by the stromal component in breast cancer, and discuss the potential therapeutic strategies that can be exploited to treat breast cancers by targeting tumor microenvironment.
Collapse
Affiliation(s)
- Maryam Majidinia
- Department of Clinical Biochemistry, Faculty of Medicine, Urmia University Medical Sciences, Urmia, Iran.,Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Bahman Yousefi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
165
|
Biswal BM, Yusoff Z. Application of Nanotechnology in Cancer Treatment. ENGINEERING APPLICATIONS OF NANOTECHNOLOGY 2017. [DOI: 10.1007/978-3-319-29761-3_11] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
166
|
Huang YT, Lan Q, Ponsonnet L, Blanquet M, Christofori G, Zaric J, Rüegg C. The matricellular protein CYR61 interferes with normal pancreatic islets architecture and promotes pancreatic neuroendocrine tumor progression. Oncotarget 2016; 7:1663-74. [PMID: 26625209 PMCID: PMC4811488 DOI: 10.18632/oncotarget.6411] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 11/15/2015] [Indexed: 12/22/2022] Open
Abstract
The significance of matricellular proteins during development and cancer progression is widely recognized. However, how these proteins actively contribute to physiological development and pathological cancer progression is only partially elucidated. In this study, we investigated the role of the matricellular protein Cysteine-rich 61 (CYR61) in pancreatic islet development and carcinogenesis. Transgenic expression of CYR61 in β cells (Rip1CYR mice) caused irregular islets morphology and distorted sorting of α cells, but did not alter islets size, number or vascularization. To investigate the function of CYR61 during carcinogenesis, we crossed Rip1CYR mice with Rip1Tag2 mice, a well-established model of β cell carcinogenesis. Beta tumors in Rip1Tag2CYR mice were larger, more invasive and more vascularized compared to tumors in Rip1Tag2 mice. The effect of CYR61 on angiogenesis was fully abrogated by treating mice with the anti-VEGFR2 mAb DC101. Results from in vitro assays demonstrated that CYR61 modulated integrin α6β1-dependent invasion and adhesion without altering its expression. Taken together, these results show that CYR61 expression in pancreatic β cells interferes with normal islet architecture, promotes islet tumor growth, invasion and VEGF/VERGFR-2-dependent tumor angiogenesis. Taken together, these observations demonstrate that CYR61 acts as a tumor-promoting gene in pancreatic neuroendocrine tumors.
Collapse
Affiliation(s)
- Yu-Ting Huang
- Department of Medicine, Faculty of Science, University of Fribourg, Fribourg, Switzerland.,National Center for Competence in Research (NCCR), Molecular Oncology, Swiss Institute for Experimental Cancer Research (ISREC)-Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Qiang Lan
- Department of Medicine, Faculty of Science, University of Fribourg, Fribourg, Switzerland.,National Center for Competence in Research (NCCR), Molecular Oncology, Swiss Institute for Experimental Cancer Research (ISREC)-Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Lionel Ponsonnet
- Department of Medicine, Faculty of Science, University of Fribourg, Fribourg, Switzerland.,National Center for Competence in Research (NCCR), Molecular Oncology, Swiss Institute for Experimental Cancer Research (ISREC)-Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Marisa Blanquet
- Department of Medicine, Faculty of Science, University of Fribourg, Fribourg, Switzerland
| | - Gerhard Christofori
- National Center for Competence in Research (NCCR), Molecular Oncology, Swiss Institute for Experimental Cancer Research (ISREC)-Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.,Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Jelena Zaric
- Department of Medicine, Faculty of Science, University of Fribourg, Fribourg, Switzerland.,National Center for Competence in Research (NCCR), Molecular Oncology, Swiss Institute for Experimental Cancer Research (ISREC)-Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Curzio Rüegg
- Department of Medicine, Faculty of Science, University of Fribourg, Fribourg, Switzerland.,National Center for Competence in Research (NCCR), Molecular Oncology, Swiss Institute for Experimental Cancer Research (ISREC)-Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| |
Collapse
|
167
|
Chen B, Dai W, Mei D, Liu T, Li S, He B, He B, Yuan L, Zhang H, Wang X, Zhang Q. Comprehensively priming the tumor microenvironment by cancer-associated fibroblast-targeted liposomes for combined therapy with cancer cell-targeted chemotherapeutic drug delivery system. J Control Release 2016; 241:68-80. [DOI: 10.1016/j.jconrel.2016.09.014] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Revised: 08/16/2016] [Accepted: 09/14/2016] [Indexed: 12/18/2022]
|
168
|
Xu B, Rodenhizer D, Lakhani S, Zhang X, Soleas JP, Ailles L, McGuigan AP. Patterning cellular compartments within TRACER cultures using sacrificial gelatin printing. Biofabrication 2016; 8:035018. [PMID: 27631341 DOI: 10.1088/1758-5090/8/3/035018] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
In the past decade, it has been well recognised that the tumour microenvironment contains microenvironmental components such as hypoxia that significantly influence tumour cell behaviours such, invasiveness and therapy resistance, all of which provide new targets for studying cancer biology and developing anticancer therapeutics. In response, a large number of two-dimensional and three-dimensional (3D) in vitro tumour models have been developed to recapitulate different aspects of the tumour microenvironment and enable the study of related biological questions. While more complex models enable new biological insight, such models often involve time-consuming and complex fabrication or analysis processes, which limit their adoption by the broader cancer biology community. To address this, we recently reported the development of a new platform that enables easy assembly and analysis of 3D tumour cultures, the tissue roll for analysis of cellular environment response (TRACER). The TRACER platform enables recapitulation of many spatial aspects of the tumour microenvironment to ask a variety of questions, however its original design contains only one cell type. In contrast tumours in vivo often contain a neoplastic and stromal compartment. To expand the types of questions the TRACER system is useful for asking, here we present a strategy to pattern distinct cell type domains into TRACER layers using a custom-built gelatin-dispensing pen. The pen allows deposition of a temporary gelatin barrier into the TRACER scaffold to define domain boundaries between cell populations. The gelatin can be melted away after cell seeding to allow interaction of cell populations from adjacent domains. Our device offers a simple strategy to generate complex multi-cell type tumour cultures for analysis of fundamental biology and drug development applications.
Collapse
Affiliation(s)
- Bin Xu
- University of Toronto, Department of Chemical Engineering and Applied Chemistry, 200 College St., Toronto, ON M5S 3E5, Canada
| | | | | | | | | | | | | |
Collapse
|
169
|
Mavrofrydi O, Mavroeidi P, Papazafiri P. Comparative assessment of HIF-1α and Akt responses in human lung and skin cells exposed to benzo[α]pyrene: Effect of conditioned medium from pre-exposed primary fibroblasts. ENVIRONMENTAL TOXICOLOGY 2016; 31:1103-1112. [PMID: 25728052 DOI: 10.1002/tox.22119] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Revised: 01/08/2015] [Accepted: 01/10/2015] [Indexed: 06/04/2023]
Abstract
Exposure to atmospheric pollutants has been accused for many adverse health effects. Benzo[α]pyrene (Β[α]Ρ) in particular, the most extensively studied member of pollutants, is implicated in both cancer initiation and promotion. In the present study, we compared the effects of noncytotoxic doses of Β[α]Ρ, between human skin and lung epithelial cells A431 and A549, respectively, focusing on Akt kinase and HIF-1α, as it is well known that these proteins are upregulated in various human cancers promoting survival, angiogenesis and metastasis of tumor cells. Also, taking into consideration that fibroblasts are involved in cancer progression, we tested the possible modulation of epithelial cell response by paracrine factors secreted by Β[α]Ρ-treated fibroblasts. Low doses of Β[α]Ρ were found to enhance epithelial cell proliferation and upregulate both Akt kinase and HIF-1α, with A549 cells exhibiting a more sustained profile of upregulation. It is to notice that, the response of HIF-1α was remarkably early, acting as a sensitive marker in response to airborne pollutants. Also, HIF-1α was induced by Β[α]Ρ in both lung and skin fibroblasts indicating that this effect may be conserved throughout different cell types and tissues. Interestingly however, the response of both proteins was differentially modified upon treatment with conditioned medium from Β[α]Ρ-exposed fibroblasts. This is particularly evident in A459 cells and confirms the critical role of intercellular and paracrine factors in the modulation of the final response to an extracellular signal. © 2015 Wiley Periodicals, Inc. Environ Toxicol 31: 1103-1112, 2016.
Collapse
Affiliation(s)
- Olga Mavrofrydi
- Division of Animal and Human Physiology, Department of Biology, University of Athens, 15784 Panepistimiopolis, Ilissia, Athens, Greece
| | - Panagiota Mavroeidi
- Division of Animal and Human Physiology, Department of Biology, University of Athens, 15784 Panepistimiopolis, Ilissia, Athens, Greece
| | - Panagiota Papazafiri
- Division of Animal and Human Physiology, Department of Biology, University of Athens, 15784 Panepistimiopolis, Ilissia, Athens, Greece
| |
Collapse
|
170
|
Inoue T, Adachi K, Kawana K, Taguchi A, Nagamatsu T, Fujimoto A, Tomio K, Yamashita A, Eguchi S, Nishida H, Nakamura H, Sato M, Yoshida M, Arimoto T, Wada-Hiraike O, Oda K, Osuga Y, Fujii T. Cancer-associated fibroblast suppresses killing activity of natural killer cells through downregulation of poliovirus receptor (PVR/CD155), a ligand of activating NK receptor. Int J Oncol 2016. [PMID: 27499237 DOI: 10.3892/ijo.2016.3631.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Cancer-associated fibroblasts (CAFs) play an important role in cancer expansion and progression in tumor microenvironment (TME), via both direct and indirect interactions. Natural killer (NK) cells play a crucial role in anticancer immunity. We investigated the inhibitory effects of CAFs on NK cell activity. CAFs were isolated from endometrial cancer tissue, while normal endometrial fibroblasts (NEFs) were obtained from normal endometrium with no pathological abnormality. NK cells were obtained from allogenic healthy volunteers. CAFs or NEFs were co-cultured at an NK/fibroblast ratio of 1:1 with or without inserted membrane. For NK cell activity, K562 cells were cultured as target cells. NK cell-killing activity was determined by calculating the ratio of PI-positive K562 cells in the presence of NK cells co-cultured with fibroblasts versus NK cells alone. To examine whether NK cell activity was suppressed by IDO pathway, we inhibited IDO activity using the IDO inhibitor 1-MT. We demonstrated that CAFs derived from endometrial cancer induced greater suppression of the killing activity of allogenic NK cells compared with normal endometrial fibroblasts (NEFs). The suppression of NK cell activity by CAFs was inhibited when a membrane was inserted between the CAFs and NK cells, but not by 1-MT, an inhibitor of IDO. We focused on receptor-ligand interactions between CAFs and NK cell and found that cell-surface poliovirus receptor (PVR/CD155), a ligand of activating NK receptor DNAM-1, was downregulated in the CAFs compared with NEFs. To confirm whether PVR downregulation results in the decrease of NK cell-killing activity, PVR expression in NEFs was knocked down using siRNA against PVR (PVRsi). NK cell activity was suppressed by co-culture with PVR-knockdown NEFs, to a similar extent than CAF-induced suppression. CAFs showed increased suppression of NK cell-killing activity compared with NEFs, due to decreased PVR cell surface expression, a ligand of an NK activating receptor. This study demonstrated a novel mechanism of suppression of NK cell activity by CAFs in the TME.
Collapse
Affiliation(s)
- Tomoko Inoue
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Katsuyuki Adachi
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Kei Kawana
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Ayumi Taguchi
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Takeshi Nagamatsu
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Asaha Fujimoto
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Kensuke Tomio
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Aki Yamashita
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Satoko Eguchi
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Haruka Nishida
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Hiroe Nakamura
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Masakazu Sato
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Mitsuyo Yoshida
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Takahide Arimoto
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Osamu Wada-Hiraike
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Katsutoshi Oda
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Yutaka Osuga
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Tomoyuki Fujii
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-8655, Japan
| |
Collapse
|
171
|
Onec B, Okutan H, Albayrak M, Saribacak Can E, Aslan V, Unver Koluman B, Soyer Kosemehmetoglu O, Albayrak A, Kos DM. The Predictive Role of the Neutrophil/Lymphocyte Ratio in Survival with Multiple Myeloma: A Single Center Experience. J Clin Lab Anal 2016; 31. [PMID: 27500465 DOI: 10.1002/jcla.22032] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2016] [Accepted: 07/01/2016] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Recent studies have shown a positive correlation between tumor-related immune response markers and the poor outcome in solid tumors. In this study, we aimed to investigate the neutrophil/lymphocyte ratio (NLR) in multiple myeloma. To the best of our knowledge, this would be the second report concerning this topic. METHODS We retrospectively reviewed the data for 52 multiple myeloma patients. The patients were grouped using the baseline NLR as NLR ≤ 1.72 and NLR > 1.72 using receiver operating characteristic analysis to determine a cut off. We compared the two groups in terms of both the known prognostic factors of the myeloma and the overall survival (OS). RESULTS Our study showed that NLR is associated with C-reactive protein and β2 microglobulin (P = 0.02 and P = 0.001, respectively). The patients with NLR > 1.72 had significantly worse stages, performance status, and kidney functions. The whole group's OS was estimated as 35.1 months while the patients with lower NLR had better OS when compared with those with NLR > 1.72 (42.75 and 26.14 months, respectively, P: 0.04). CONCLUSION Neutrophil/lymphocyte ratio, which is associated with stage, performance status, and kidney functions, can be used in daily practice as a predictor for survival in multiple myeloma. Simply adding NLR to the routine charts may enrich our data for larger studies.
Collapse
Affiliation(s)
- Birgul Onec
- Department of Hematology, Dışkapı Training and Research Hospital, Ankara, Turkey
| | - Harika Okutan
- Department of Hematology, Dışkapı Training and Research Hospital, Ankara, Turkey
| | - Murat Albayrak
- Department of Hematology, Dışkapı Training and Research Hospital, Ankara, Turkey
| | - Esra Saribacak Can
- Department of Hematology, Dışkapı Training and Research Hospital, Ankara, Turkey
| | - Vedat Aslan
- Department of Hematology, Dışkapı Training and Research Hospital, Ankara, Turkey
| | - Basak Unver Koluman
- Department of Hematology, Dışkapı Training and Research Hospital, Ankara, Turkey
| | | | - Aynur Albayrak
- Department of Pathology, Dışkapı Training and Research Hospital, Ankara, Turkey
| | - Durdu Mehmet Kos
- Department of Internal Medicine, Faculty of Medicine, Düzce University, Düzce, Turkey
| |
Collapse
|
172
|
Mao XY, Li QQ, Gao YF, Zhou HH, Liu ZQ, Jin WL. Gap junction as an intercellular glue: Emerging roles in cancer EMT and metastasis. Cancer Lett 2016; 381:133-7. [PMID: 27490999 DOI: 10.1016/j.canlet.2016.07.037] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 07/27/2016] [Accepted: 07/27/2016] [Indexed: 12/31/2022]
Abstract
Metastasis is a common phenomenon in the progression and dissemination of cancer. It is estimated that metastasis accounts for 90% cancer-related mortality. Although the formation of tumor metastasis is relatively well understood, the underlying molecular mechanisms responsible for the emergence of aggressive cancer phenotype are still elusive. Figuring out the mechanisms by which cancer cells evade from the tumor is beneficial for obtaining novel and effectively therapeutic approaches. Primary tumors are composed of various subpopulations of cells with heterogeneous metastatic characteristics and the occurrence of metastatic dissemination is mainly dependent upon the interactions between tumor and the surrounding microenvironment. Tumor microenvironment (TME) such as extracellular matrix, macrophages, fibroblasts, stem cells and endothelial cells can orchestrate events critical to tumor evolution toward metastasis. GJ serves as an important communication between tumor cells and stromal cells. Increased GJs coupling blocks metastatic potential in some cancer animal models such as breast cancer and melanoma. Besides, epithelial-to-mesenchymal transition (EMT) is also a crucial step in the metastatic process and there are signs that GJs contribute to cell adhesion and migration (the pathological feature of EMT) in breast cancer. Therefore, we propose that GJ serves as an intercellular glue to suppress EMT and cancer metastasis.
Collapse
Affiliation(s)
- Xiao-Yuan Mao
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China; Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha 410078, China.
| | - Qiu-Qi Li
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China; Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha 410078, China
| | - Yuan-Feng Gao
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China; Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha 410078, China
| | - Hong-Hao Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China; Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha 410078, China
| | - Zhao-Qian Liu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China; Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha 410078, China.
| | - Wei-Lin Jin
- Institute of Nano Biomedicine and Engineering, Department of Instrument Science and Engineering, Key Lab. for Thin Film and Microfabrication Technology of Ministry of Education, School of Electronic Information and Electronic Engineering, Shanghai Jiao Tong University, Shanghai 200240, China; National Centers for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China.
| |
Collapse
|
173
|
Inoue T, Adachi K, Kawana K, Taguchi A, Nagamatsu T, Fujimoto A, Tomio K, Yamashita A, Eguchi S, Nishida H, Nakamura H, Sato M, Yoshida M, Arimoto T, Wada-Hiraike O, Oda K, Osuga Y, Fujii T. Cancer-associated fibroblast suppresses killing activity of natural killer cells through downregulation of poliovirus receptor (PVR/CD155), a ligand of activating NK receptor. Int J Oncol 2016; 49:1297-304. [PMID: 27499237 PMCID: PMC5021244 DOI: 10.3892/ijo.2016.3631] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 07/14/2016] [Indexed: 12/11/2022] Open
Abstract
Cancer-associated fibroblasts (CAFs) play an important role in cancer expansion and progression in tumor microenvironment (TME), via both direct and indirect interactions. Natural killer (NK) cells play a crucial role in anticancer immunity. We investigated the inhibitory effects of CAFs on NK cell activity. CAFs were isolated from endometrial cancer tissue, while normal endometrial fibroblasts (NEFs) were obtained from normal endometrium with no pathological abnormality. NK cells were obtained from allogenic healthy volunteers. CAFs or NEFs were co-cultured at an NK/fibroblast ratio of 1:1 with or without inserted membrane. For NK cell activity, K562 cells were cultured as target cells. NK cell-killing activity was determined by calculating the ratio of PI-positive K562 cells in the presence of NK cells co-cultured with fibroblasts versus NK cells alone. To examine whether NK cell activity was suppressed by IDO pathway, we inhibited IDO activity using the IDO inhibitor 1-MT. We demonstrated that CAFs derived from endometrial cancer induced greater suppression of the killing activity of allogenic NK cells compared with normal endometrial fibroblasts (NEFs). The suppression of NK cell activity by CAFs was inhibited when a membrane was inserted between the CAFs and NK cells, but not by 1-MT, an inhibitor of IDO. We focused on receptor-ligand interactions between CAFs and NK cell and found that cell-surface poliovirus receptor (PVR/CD155), a ligand of activating NK receptor DNAM-1, was downregulated in the CAFs compared with NEFs. To confirm whether PVR downregulation results in the decrease of NK cell-killing activity, PVR expression in NEFs was knocked down using siRNA against PVR (PVRsi). NK cell activity was suppressed by co-culture with PVR-knockdown NEFs, to a similar extent than CAF-induced suppression. CAFs showed increased suppression of NK cell-killing activity compared with NEFs, due to decreased PVR cell surface expression, a ligand of an NK activating receptor. This study demonstrated a novel mechanism of suppression of NK cell activity by CAFs in the TME.
Collapse
Affiliation(s)
- Tomoko Inoue
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Katsuyuki Adachi
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Kei Kawana
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Ayumi Taguchi
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Takeshi Nagamatsu
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Asaha Fujimoto
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Kensuke Tomio
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Aki Yamashita
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Satoko Eguchi
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Haruka Nishida
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Hiroe Nakamura
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Masakazu Sato
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Mitsuyo Yoshida
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Takahide Arimoto
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Osamu Wada-Hiraike
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Katsutoshi Oda
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Yutaka Osuga
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Tomoyuki Fujii
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-8655, Japan
| |
Collapse
|
174
|
Yang L, Xia L, Wang Y, Hong S, Chen H, Liang S, Peng P, Chen Y. Low Prognostic Nutritional Index (PNI) Predicts Unfavorable Distant Metastasis-Free Survival in Nasopharyngeal Carcinoma: A Propensity Score-Matched Analysis. PLoS One 2016; 11:e0158853. [PMID: 27399281 PMCID: PMC4939954 DOI: 10.1371/journal.pone.0158853] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 06/22/2016] [Indexed: 02/06/2023] Open
Abstract
Background Poor nutritional status is associated with progression and advanced disease in patients with cancer. The prognostic nutritional index (PNI) may represent a simple method of assessing host immunonutritional status. This study was designed to investigate the prognostic value of the PNI for distant metastasis-free survival (DMFS) in patients with nasopharyngeal carcinoma (NPC). Methods A training cohort of 1,168 patients with non-metastatic NPC from two institutions was retrospectively analyzed. The optimal PNI cutoff value for DMFS was identified using the online tool “Cutoff Finder”. DMFS was analyzed using stratified and adjusted analysis. Propensity score-matched analysis was performed to balance baseline characteristics between the high and low PNI groups. Subsequently, the prognostic value of the PNI for DMFS was validated in an external validation cohort of 756 patients with NPC. The area under the receiver operating characteristics curve (AUC) was calculated to compare the discriminatory ability of different prognostic scores. Results The optimal PNI cutoff value was determined to be 51. Low PNI was significantly associated with poorer DMFS than high PNI in univariate analysis (P<0.001) as well as multivariate analysis (P<0.001) before propensity score matching. In subgroup analyses, PNI could also stratify different risks of distant metastases. Propensity score-matched analyses confirmed the prognostic value of PNI, excluding other interpretations and selection bias. In the external validation cohort, patients with high PNI also had significantly lower risk of distant metastases than those with low PNI (Hazards Ratios, 0.487; P<0.001). The PNI consistently showed a higher AUC value at 1-year (0.780), 3-year (0.793) and 5-year (0.812) in comparison with other prognostic scores. Conclusion PNI, an inexpensive and easily assessable inflammatory index, could aid clinicians in developing individualized treatment and follow-up strategies for patients with non-metastatic NPC.
Collapse
Affiliation(s)
- Lin Yang
- Sun Yat-sen University cancer center, Guangzhou, China.,State Key Laboratory of Oncology in Southern China, Guangzhou, China.,Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Liangping Xia
- Sun Yat-sen University cancer center, Guangzhou, China.,State Key Laboratory of Oncology in Southern China, Guangzhou, China.,Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yan Wang
- Sun Yat-sen University cancer center, Guangzhou, China.,State Key Laboratory of Oncology in Southern China, Guangzhou, China.,Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Shaodong Hong
- Sun Yat-sen University cancer center, Guangzhou, China.,State Key Laboratory of Oncology in Southern China, Guangzhou, China.,Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Haiyang Chen
- The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | | | - Peijian Peng
- The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Yong Chen
- Sun Yat-sen University cancer center, Guangzhou, China.,State Key Laboratory of Oncology in Southern China, Guangzhou, China.,Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| |
Collapse
|
175
|
Michiels C, Tellier C, Feron O. Cycling hypoxia: A key feature of the tumor microenvironment. Biochim Biophys Acta Rev Cancer 2016; 1866:76-86. [PMID: 27343712 DOI: 10.1016/j.bbcan.2016.06.004] [Citation(s) in RCA: 128] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2016] [Revised: 06/18/2016] [Accepted: 06/21/2016] [Indexed: 01/06/2023]
Abstract
A compelling body of evidence indicates that most human solid tumors contain hypoxic areas. Hypoxia is the consequence not only of the chaotic proliferation of cancer cells that places them at distance from the nearest capillary but also of the abnormal structure of the new vasculature network resulting in transient blood flow. Hence two types of hypoxia are observed in tumors: chronic and cycling (intermittent) hypoxia. Most of the current work aims at understanding the role of chronic hypoxia in tumor growth, response to treatment and metastasis. Only recently, cycling hypoxia, with spatial and temporal fluctuations in oxygen levels, has emerged as another key feature of the tumor environment that triggers different responses in comparison to chronic hypoxia. Either type of hypoxia is associated with distinct effects not only in cancer cells but also in stromal cells. In particular, cycling hypoxia has been demonstrated to favor, to a higher extent than chronic hypoxia, angiogenesis, resistance to anti-cancer treatments, intratumoral inflammation and tumor metastasis. These review details these effects as well as the signaling pathway it triggers to switch on specific transcriptomic programs. Understanding the signaling pathways through which cycling hypoxia induces these processes that support the development of an aggressive cancer could convey to the emergence of promising new cancer treatments.
Collapse
Affiliation(s)
- Carine Michiels
- URBC-NARILIS, University of Namur, 61 rue de Bruxelles, 5000 Namur, Belgium.
| | - Céline Tellier
- URBC-NARILIS, University of Namur, 61 rue de Bruxelles, 5000 Namur, Belgium
| | - Olivier Feron
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain, 53 Avenue Mounier, B1.53.09, B-1200 Brussels, Belgium.
| |
Collapse
|
176
|
Nguyen T, Mège RM. N-Cadherin and Fibroblast Growth Factor Receptors crosstalk in the control of developmental and cancer cell migrations. Eur J Cell Biol 2016; 95:415-426. [PMID: 27320194 DOI: 10.1016/j.ejcb.2016.05.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 05/13/2016] [Accepted: 05/24/2016] [Indexed: 12/12/2022] Open
Abstract
Cell migrations are diverse. They constitutemajor morphogenetic driving forces during embryogenesis, but they contribute also to the loss of tissue homeostasis and cancer growth. Capabilities of cells to migrate as single cells or as collectives are controlled by internal and external signalling, leading to the reorganisation of their cytoskeleton as well as by the rebalancing of cell-matrix and cell-cell adhesions. Among the genes altered in numerous cancers, cadherins and growth factor receptors are of particular interest for cell migration regulation. In particular, cadherins such as N-cadherin and a class of growth factor receptors, namely FGFRs cooperate to regulate embryonic and cancer cell behaviours. In this review, we discuss on reciprocal crosstalk between N-cadherin and FGFRs during cell migration. Finally, we aim at clarifying the synergy between N-cadherin and FGFR signalling that ensure cellular reorganization during cell movements, mainly during cancer cell migration and metastasis but also during developmental processes.
Collapse
Affiliation(s)
- Thao Nguyen
- Institut Jacques Monod, CNRS, Université Paris Diderot, Paris, France
| | - René Marc Mège
- Institut Jacques Monod, CNRS, Université Paris Diderot, Paris, France.
| |
Collapse
|
177
|
Fibroblast surface-associated FGF-2 promotes contact-dependent colorectal cancer cell migration and invasion through FGFR-SRC signaling and integrin αvβ5-mediated adhesion. Oncotarget 2016; 6:14300-17. [PMID: 25973543 PMCID: PMC4546468 DOI: 10.18632/oncotarget.3883] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2014] [Accepted: 04/21/2015] [Indexed: 12/15/2022] Open
Abstract
Carcinoma-associated fibroblasts were reported to promote colorectal cancer (CRC) invasion by secreting motility factors and extracellular matrix processing enzymes. Less is known whether fibroblasts may induce CRC cancer cell motility by contact-dependent mechanisms. To address this question we characterized the interaction between fibroblasts and SW620 and HT29 colorectal cancer cells in 2D and 3D co-culture models in vitro. Here we show that fibroblasts induce contact-dependent cancer cell elongation, motility and invasiveness independently of deposited matrix or secreted factors. These effects depend on fibroblast cell surface-associated fibroblast growth factor (FGF) -2. Inhibition of FGF-2 or FGF receptors (FGFRs) signaling abolishes these effects. FGFRs activate SRC in cancer cells and inhibition or silencing of SRC in cancer cells, but not in fibroblasts, prevents fibroblasts-mediated effects. Using an RGD-based integrin antagonist and function-blocking antibodies we demonstrate that cancer cell adhesion to fibroblasts requires integrin αvβ5. Taken together, these results demonstrate that fibroblasts induce cell-contact-dependent colorectal cancer cell migration and invasion under 2D and 3D conditions in vitro through fibroblast cell surface-associated FGF-2, FGF receptor-mediated SRC activation and αvβ5 integrin-dependent cancer cell adhesion to fibroblasts. The FGF-2-FGFRs-SRC-αvβ5 integrin loop might be explored as candidate therapeutic target to block colorectal cancer invasion.
Collapse
|
178
|
Tanner Y, Grose RP. Dysregulated FGF signalling in neoplastic disorders. Semin Cell Dev Biol 2016; 53:126-35. [DOI: 10.1016/j.semcdb.2015.10.012] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 10/08/2015] [Indexed: 12/31/2022]
|
179
|
Ciarloni L, Ehrensberger SH, Imaizumi N, Monnier-Benoit S, Nichita C, Myung SJ, Kim JS, Song SY, Kim TI, van der Weg B, Meier R, Borovicka J, Beglinger C, Vallet C, Maerten P, Rüegg C, Dorta G. Development and Clinical Validation of a Blood Test Based on 29-Gene Expression for Early Detection of Colorectal Cancer. Clin Cancer Res 2016; 22:4604-11. [PMID: 27126992 DOI: 10.1158/1078-0432.ccr-15-2057] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 04/09/2016] [Indexed: 11/16/2022]
Abstract
PURPOSE A blood test for early detection of colorectal cancer is a valuable tool for testing asymptomatic individuals and reducing colorectal cancer-related mortality. The objective of this study was to develop and validate a novel blood test able to differentiate patients with colorectal cancer and adenomatous polyps (AP) from individuals with a negative colonoscopy. EXPERIMENTAL DESIGN A case-control, multicenter clinical study was designed to collect blood samples from patients referred for colonoscopy or surgery. Predictive algorithms were developed on 75 controls, 61 large AP (LAP) ≥1 cm, and 45 colorectal cancer cases and independently validated on 74 controls, 42 LAP, and 52 colorectal cancer cases (23 stages I-II) as well as on 245 cases including other colorectal findings and diseases other than colorectal cancer. The test is based on a 29-gene panel expressed in peripheral blood mononuclear cells alone or in combination with established plasma tumor markers. RESULTS The 29-gene algorithm detected colorectal cancer and LAP with a sensitivity of 79.5% and 55.4%, respectively, with 90.0% specificity. Combination with the protein tumor markers carcinoembryonic antigen (CEA) and CYFRA21-2 resulted in a specificity increase (92.2%) with a sensitivity for colorectal cancer and LAP detection of 78.1% and 52.3%, respectively. CONCLUSIONS We report the validation of a novel blood test, Colox®, for the detection of colorectal cancer and LAP based on a 29-gene panel and the CEA and CYFRA21-1 plasma biomarkers. The performance and convenience of this routine blood test provide physicians a useful tool to test average-risk individuals unwilling to undergo upfront colonoscopy. Clin Cancer Res; 22(18); 4604-11. ©2016 AACR.
Collapse
Affiliation(s)
| | | | | | | | - Cristina Nichita
- Department of Gastroenterology and Hepatology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Seung-Jae Myung
- Asan Medical Centre, Department of Gastroenterology, University of Ulsan College of Medicine, Seoul, South Korea
| | - Joo Sung Kim
- Department of Internal Medicine, College of Medicine, Seoul National University, Seoul, South Korea
| | - Si Young Song
- Severance Hospital, Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Tae Il Kim
- Severance Hospital, Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | | | - Rémy Meier
- Kantonsspital Liestal, Gastroenterology, Hepatology and Nutrition Department, University Hospital, Liestal, Switzerland
| | - Jan Borovicka
- Kantonsspital St. Gallen, Department of Gastroenterology and Hepatology, St. Gallen, Switzerland
| | | | - Cédric Vallet
- Ensemble Hospitalier de la Côte, Surgery Services, Morges, Switzerland
| | | | - Curzio Rüegg
- Novigenix SA, Epalinges, Switzerland. Department of Medicine, Faculty of Science, University of Fribourg, Fribourg, Switzerland.
| | - Gian Dorta
- Department of Gastroenterology and Hepatology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland.
| |
Collapse
|
180
|
Mei L, Liu Y, Zhang H, Zhang Z, Gao H, He Q. Antitumor and Antimetastasis Activities of Heparin-based Micelle Served As Both Carrier and Drug. ACS APPLIED MATERIALS & INTERFACES 2016; 8:9577-9589. [PMID: 27058058 DOI: 10.1021/acsami.5b12347] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Effective treatments for tumors are not easy to achieve due to the existence of metastases, which are responsible for most tumor death. Hence, a new drug delivery system is a pressing need, which should be biocompatible, stimuli-responsive, and multifunctional, including antitumor, antimetastasis, and antiangiogenesis effects. However, it is challenging to achieve all of these properties in one drug delivery system. Here, we developed a system of drug DOX and heparin into one self-assemble nanoparticle via pH-sensitive hydrazone bond and hydrophobic groups, deoxycholate. In the process, heparin itself was not only as the hydrophilic segments of the carrier, but also processed multiple biological functions such as antiangiogenesis and antimetastasis effect. The micelle nanoparticle HD-DOX processed good stability and acidic pH-triggered drug release property. After systemic administration, heparin-based micelle nanoparticle showed longer half-time and enhanced accumulation of DOX in tumors through the enhanced permeability and retention effect, leading to more efficient antitumor effects. In addition, heparin could hinder platelet-induced tumor cells epithelial-mesenchymal transition (EMT) and partially affect cell actin cytoskeletal arrangement, resulting in the disorganization of the actin cytoskeleton. Therefore, HD-DOX exhibited significant inhibitory effect on the metastasis in melanoma animal model in C57BL/6 mouse. Meanwhile, benefited from the antiangiogenesis effect of heparin, tube formations in endothelial cells were effectively inhibited and tumor vascular density was decreased by HD-DOX. Taken together, our study developed a self-assembly nanoplatform that both the drug and carrier had therapeutic effects with ideal antitumor efficacy.
Collapse
Affiliation(s)
- Ling Mei
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University . No. 17, Block 3, Southern Renmin Road, Chengdu 610041, China
| | - Yayuan Liu
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University . No. 17, Block 3, Southern Renmin Road, Chengdu 610041, China
| | - HuaJin Zhang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University . No. 17, Block 3, Southern Renmin Road, Chengdu 610041, China
| | - Zhirong Zhang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University . No. 17, Block 3, Southern Renmin Road, Chengdu 610041, China
| | - Huile Gao
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University . No. 17, Block 3, Southern Renmin Road, Chengdu 610041, China
| | - Qin He
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University . No. 17, Block 3, Southern Renmin Road, Chengdu 610041, China
| |
Collapse
|
181
|
Tran TB, Baek C, Min J. Electric Cell-Substrate Impedance Sensing (ECIS) with Microelectrode Arrays for Investigation of Cancer Cell-Fibroblasts Interaction. PLoS One 2016; 11:e0153813. [PMID: 27088611 PMCID: PMC4835071 DOI: 10.1371/journal.pone.0153813] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 04/04/2016] [Indexed: 12/30/2022] Open
Abstract
The tumor microenvironment, including stromal cells, surrounding blood vessels and extracellular matrix components, has been defined as a crucial factor that influences the proliferation, drug-resistance, invasion and metastasis of malignant epithelial cells. Among other factors, the communications and interaction between cancer cells and stromal cells have been reported to play pivotal roles in cancer promotion and progression. To investigate these relationships, an on-chip co-culture model was developed to study the cellular interaction between A549—human lung carcinoma cells and MRC-5—human lung epithelial cells in both normal proliferation and treatment conditions. In brief, a co-culture device consisting of 2 individual fluidic chambers in parallel, which were separated by a 100 μm fence was utilized for cell patterning. Microelectrodes arrays were installed within each chamber including electrodes at various distances away from the confrontation line for the electrochemical impedimetric sensing assessment of cell-to-cell influence. After the fence was removed and cell-to-cell contact occurred, by evaluating the impedance signal responses representing cell condition and behavior, both direct and indirect cell-to-cell interactions through conditioned media were investigated. The impact of specific distances that lead to different influences of fibroblast cells on cancer cells in the co-culture environment was also defined.
Collapse
Affiliation(s)
- Trong Binh Tran
- School of Integrative Engineering, Chung-Ang University, Heukseok-dong, Dongjak-gu, Seoul, Republic of Korea
| | - Changyoon Baek
- School of Integrative Engineering, Chung-Ang University, Heukseok-dong, Dongjak-gu, Seoul, Republic of Korea
| | - Junhong Min
- School of Integrative Engineering, Chung-Ang University, Heukseok-dong, Dongjak-gu, Seoul, Republic of Korea
- * E-mail:
| |
Collapse
|
182
|
Kirstein JM, Hague MN, McGowan PM, Tuck AB, Chambers AF. Primary melanoma tumor inhibits metastasis through alterations in systemic hemostasis. J Mol Med (Berl) 2016; 94:899-910. [PMID: 27048169 DOI: 10.1007/s00109-016-1415-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 03/21/2016] [Accepted: 03/24/2016] [Indexed: 02/07/2023]
Abstract
Progression from a primary tumor to distant metastases requires extensive interactions between tumor cells and their microenvironment. The primary tumor is not only the source of metastatic cells but also can also modulate host responses to these cells, leading to an enhancement or inhibition of metastasis. Tumor-mediated stimulation of bone marrow can result in pre-metastatic niche formation and increased metastasis. However, a primary tumor can also inhibit metastasis through concomitant tumor resistance-inhibition of metastatic growth by existing tumor mass. Here, we report that the presence of a B16F10 primary tumor significantly restricted numbers and sizes of experimental lung metastases through reduction of circulating platelets and reduced formation of metastatic tumor cell-associated thrombi. Tumor-bearing mice displayed splenomegaly, correlated with primary tumor size and platelet count. Reduction in platelet numbers in tumor-bearing animals was responsible for metastatic inhibition, as restoration of platelet numbers using isolated platelets re-established both tumor cell-associated thrombus formation and experimental metastasis. Consumption of platelets due to a B16F10 primary tumor is a form of concomitant tumor resistance and demonstrates the systemic impact of a growing tumor. Understanding the interplay between primary tumors and metastases is essential, as clarification of concomitant tumor resistance mechanisms may allow inhibition of metastatic growth following tumor resection. Key messages Mice with a primary B16F10 tumor had reduced metastasis vs. mice without a primary tumor. Tumor-bearing mice had splenomegaly and fewer platelets and tumor-associated thrombi. Restoring platelets restored tumor-associated thrombi and increased metastasis. This work shows the impact that a primary tumor can have on systemic metastasis. Understanding these interactions may lead to improved ways to inhibit metastasis.
Collapse
Affiliation(s)
- Jennifer M Kirstein
- London Regional Cancer Program, London Health Sciences Centre, 790 Commissioners Road East, London, ON, N6A 4L6, Canada.,Department of Medical Biophysics, University of Western Ontario, London, ON, Canada
| | - M Nicole Hague
- London Regional Cancer Program, London Health Sciences Centre, 790 Commissioners Road East, London, ON, N6A 4L6, Canada
| | - Patricia M McGowan
- London Regional Cancer Program, London Health Sciences Centre, 790 Commissioners Road East, London, ON, N6A 4L6, Canada
| | - Alan B Tuck
- London Regional Cancer Program, London Health Sciences Centre, 790 Commissioners Road East, London, ON, N6A 4L6, Canada.,Department of Pathology and Laboratory Medicine, University of Western Ontario, London, ON, Canada.,Department of Oncology, University of Western Ontario, London, ON, Canada
| | - Ann F Chambers
- London Regional Cancer Program, London Health Sciences Centre, 790 Commissioners Road East, London, ON, N6A 4L6, Canada. .,Department of Medical Biophysics, University of Western Ontario, London, ON, Canada. .,Department of Pathology and Laboratory Medicine, University of Western Ontario, London, ON, Canada. .,Department of Oncology, University of Western Ontario, London, ON, Canada.
| |
Collapse
|
183
|
Xie Y, Zhong Y, Gao T, Zhang X, Li LI, Ruan H, Li D. Human lymphatic endothelial cells contribute to epithelial ovarian carcinoma metastasis by promoting lymphangiogenesis and tumour cell invasion. Exp Ther Med 2016; 11:1587-1594. [PMID: 27168777 PMCID: PMC4840642 DOI: 10.3892/etm.2016.3134] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 01/11/2016] [Indexed: 12/25/2022] Open
Abstract
The microenvironment of a tumour is an important factor in ovarian cancer metastasis. The present study aimed to simulate the in vivo microenvironment of an ovarian carcinoma using a co-culture system consisting of human lymphatic endothelial cells (HLECs) and human ovarian carcinoma cells with directional high lymphatic metastasis (SKOV3-PM4s) in order to investigate the role of both cell types in ovarian carcinoma metastasis. The SKOV3-PM4s cultured in the HLEC-conditioned medium exhibited increased numbers of pseudopodia and mitotic figures, proliferated at a faster rate and exhibited enhanced invasion and migratory abilities. Furthermore, the HLECs cultured in SKOV3-PM4-conditioned medium exhibited significant morphological alterations and vacuolisation of the cytoplasm, as well as increased invasion, migratory and tube forming abilities. In addition, spontaneous fusion of the SKOV3-PM4s and HLECs was observed in the co-culture system using laser confocal microscopy. The gelatin zymography assay demonstrated that matrix metalloproteinase-2, which was downregulated in the SKOV3-PM4s, was upregulated in the co-culture system. The results of the present study suggested that the invasion ability of the SKOV3-PM4s was increased in the in vitro co-culture system of SKOV3-PM4 and HLECs. Therefore, alterations in the cell microenvironment may represent a novel strategy for ovarian cancer therapy.
Collapse
Affiliation(s)
- Yihong Xie
- Research Department, Affiliated Tumour Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Yanping Zhong
- Medical Scientific Research Centre, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Ting Gao
- Research Department, Affiliated Tumour Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Xinying Zhang
- Research Department, Affiliated Tumour Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - L I Li
- Research Department, Affiliated Tumour Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Heyun Ruan
- Research Department, Affiliated Tumour Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Danrong Li
- Research Department, Affiliated Tumour Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| |
Collapse
|
184
|
Mukherjee S, Manna A, Bhattacharjee P, Mazumdar M, Saha S, Chakraborty S, Guha D, Adhikary A, Jana D, Gorain M, Mukherjee SA, Kundu GC, Sarkar DK, Das T. Non-migratory tumorigenic intrinsic cancer stem cells ensure breast cancer metastasis by generation of CXCR4+ migrating cancer stem cells. Oncogene 2016; 35:4937-48. [DOI: 10.1038/onc.2016.26] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Revised: 11/02/2015] [Accepted: 11/27/2015] [Indexed: 12/18/2022]
|
185
|
Zhong J, Chen Y, Wang LJ. Emerging molecular basis of hematogenous metastasis in gastric cancer. World J Gastroenterol 2016; 22:2434-2440. [PMID: 26937132 PMCID: PMC4768190 DOI: 10.3748/wjg.v22.i8.2434] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Revised: 10/29/2015] [Accepted: 11/19/2015] [Indexed: 02/06/2023] Open
Abstract
Lymphatic metastasis is commonly observed in gastric cancer (GC), but hematogenous metastasis is more likely responsible for the cancer-related mortality. Since Stephen Paget first introduced the “seed and soil hypothesis” a century ago, growing evidence recognizes that numerous essential secreted factors and signaling pathway effectors participate in the pre-metastatic niche formation and distant organ metastasis. The cross-talk between GC cells and surrounding microenvironment may consist of a series of interrelated steps, including epithelial mesenchymal transition, intravasation into blood vessels, circulating tumor cell translocation, and secondary organ metastasis. Secreted factors including vascular endothelial growth factor (VEGF), matrix metalloproteinases and cancer-derived extracellular vesicles, especially exosomes, are essential in formation of premetastatic niche. Circulating tumor cells and microRNAs represent as ‘‘metastatic intermediates’’ between primary tumors and sites of dissemination. Many biomarkers have been identified as novel metastatic markers and prognostic effectors. In addition, molecular therapy has been designed to target biomarkers such as growth factors (human epidermal growth factor receptor 2, VEGF) and chemokines, although they have not clearly proven to be effective in inhibiting GC metastasis in clinical trials. In this review, we will systematically discuss the emerging molecules and their microenvironment in hematogenous metastasis of GC, which may help us to find new therapeutic strategies in the future.
Collapse
|
186
|
Proinflammatory and Anti-Inflammatory Cytokines Mediated by NF-κB Factor as Prognostic Markers in Mammary Tumors. Mediators Inflamm 2016; 2016:9512743. [PMID: 26989335 PMCID: PMC4771900 DOI: 10.1155/2016/9512743] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Revised: 01/14/2016] [Accepted: 01/19/2016] [Indexed: 01/08/2023] Open
Abstract
Inflammation results in the production of cytokines, such as interleukin- (IL-) 4 and IL-10 with immunosuppressive properties or IL-6 and TNF-α with procarcinogenic activity. Furthermore, NF-κB is the major link between inflammation and tumorigenesis. This study verified the interaction between active inflammatory cytokines in the tumor microenvironment and serum of female dogs with mammary tumors and their correlation with the clinicopathological characteristics and overall survival. Measurement of gene expression was performed by qPCR and protein levels by ELISA/Luminex. High gene and protein expression levels of NF-κB, IL-6, and TNF-α were found in association with characteristics that reflect worse prognosis and a negative correlation between TNF-α protein expression and survival time was observed (p < 0.05). In contrast, high gene and protein expression levels of IL-4 and IL-10 were associated with characteristics of better prognosis and an increased level of IL-4 and a longer survival time of animals were obtained (p < 0.05). In addition, there was a positive correlation between TNF-α and IL-6 expression in association with NF-κB. The results show a significant correlation of these cytokines with tumor development, associated with NF-κB expression and cytokines promodulation, showing that these biological factors could be used as predictive and prognostic markers in breast cancer.
Collapse
|
187
|
Huang FJ, Zhou XY, Ye L, Fei XC, Wang S, Wang W, Ning G. Follicular thyroid carcinoma but not adenoma recruits tumor-associated macrophages by releasing CCL15. BMC Cancer 2016; 16:98. [PMID: 26875556 PMCID: PMC4753660 DOI: 10.1186/s12885-016-2114-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 02/03/2016] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The differential diagnosis of follicular thyroid carcinoma (FTC) and follicular adenoma (FA) before surgery is a clinical challenge. Many efforts have been made but most focusing on tumor cells, while the roles of tumor associated macrophages (TAMs) remained unclear in FTC. Here we analyzed the differences between TAMs in FTC and those in FA. METHODS We first analyzed the density of TAMs by CD68 immunostaining in 59 histologically confirmed FTCs and 47 FAs. Cytokines produced by FTC and FA were profiled using antibody array, and validated by quantitative PCR. Chemotaxis of monocyte THP-1 was induced by condition medium of FTC cell lines (FTC133 and WRO82-1) with and without anti-CCL15 neutralizing antibody. Finally, we analyzed CCL15 protein level in FTC and FA by immunohistochemistry. RESULTS The average density of CD68(+) cells was 9.5 ± 5.4/field in FTC, significantly higher than that in FA (4.9 ± 3.4/field, p < 0.001). Subsequently profiling showed that CCL15 was the most abundant chemokine in FTC compared with FA. CCL15 mRNA in FTC was 51.4-folds of that in FA. CM of FTC cell lines induced THP-1 cell chemotaxis by 33 ~ 77%, and anti-CCL15 neutralizing antibody reduced THP-1 cell migration in a dose-dependent manner. Moreover, we observed positive CCL15 immunostaining in 67.8% of FTCs compared with 23.4% of FAs. CONCLUSION Our study suggested FTC might induce TAMs infiltration by producing CCL15. Measurement of TAMs and CCL15 in follicular thyroid lesions may be applied clinically to differentiate FTC from FA pre-operation.
Collapse
Affiliation(s)
- Feng-Jiao Huang
- Shanghai Key Laboratoryfor Endocrine Tumors, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases and Shanghai E-institute for Endocrinology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, 197 Ruijin 2nd Road, Shanghai, 200025, P.R. China.
| | - Xiao-Yi Zhou
- Shanghai Key Laboratoryfor Endocrine Tumors, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases and Shanghai E-institute for Endocrinology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, 197 Ruijin 2nd Road, Shanghai, 200025, P.R. China.
| | - Lei Ye
- Shanghai Key Laboratoryfor Endocrine Tumors, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases and Shanghai E-institute for Endocrinology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, 197 Ruijin 2nd Road, Shanghai, 200025, P.R. China.
| | - Xiao-Chun Fei
- Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University, School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P.R. China.
| | - Shu Wang
- Shanghai Key Laboratoryfor Endocrine Tumors, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases and Shanghai E-institute for Endocrinology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, 197 Ruijin 2nd Road, Shanghai, 200025, P.R. China. .,Laboratory for Endocrine & Metabolic Diseases of Institute of Health Science, Shanghai Jiao Tong University School of Medicine and Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 227 South Chongqing Road, Shanghai, 200025, P.R. China.
| | - Weiqing Wang
- Shanghai Key Laboratoryfor Endocrine Tumors, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases and Shanghai E-institute for Endocrinology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, 197 Ruijin 2nd Road, Shanghai, 200025, P.R. China.
| | - Guang Ning
- Shanghai Key Laboratoryfor Endocrine Tumors, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases and Shanghai E-institute for Endocrinology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, 197 Ruijin 2nd Road, Shanghai, 200025, P.R. China. .,Laboratory for Endocrine & Metabolic Diseases of Institute of Health Science, Shanghai Jiao Tong University School of Medicine and Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 227 South Chongqing Road, Shanghai, 200025, P.R. China.
| |
Collapse
|
188
|
Tissot T, Ujvari B, Solary E, Lassus P, Roche B, Thomas F. Do cell-autonomous and non-cell-autonomous effects drive the structure of tumor ecosystems? Biochim Biophys Acta Rev Cancer 2016; 1865:147-54. [PMID: 26845682 DOI: 10.1016/j.bbcan.2016.01.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2015] [Revised: 01/28/2016] [Accepted: 01/30/2016] [Indexed: 12/21/2022]
Abstract
By definition, a driver mutation confers a growth advantage to the cancer cell in which it occurs, while a passenger mutation does not: the former is usually considered as the engine of cancer progression, while the latter is not. Actually, the effects of a given mutation depend on the genetic background of the cell in which it appears, thus can differ in the subclones that form a tumor. In addition to cell-autonomous effects generated by the mutations, non-cell-autonomous effects shape the phenotype of a cancer cell. Here, we review the evidence that a network of biological interactions between subclones drives cancer cell adaptation and amplifies intra-tumor heterogeneity. Integrating the role of mutations in tumor ecosystems generates innovative strategies targeting the tumor ecosystem's weaknesses to improve cancer treatment.
Collapse
Affiliation(s)
- Tazzio Tissot
- CREEC/MIVEGEC, UMR IRD/CNRS/UM 5290, 911 Avenue Agropolis, BP 64501, 34394 Montpellier Cedex 5, France.
| | - Beata Ujvari
- Centre for Integrative Ecology, School of Life and Environmental Sciences, Deakin University, Waurn Ponds, Australia
| | - Eric Solary
- INSERM U1170, Gustave Roussy, 94805 Villejuif, France; University Paris-Saclay, Faculty of Medicine, 94270 Le Kremlin-Bicêtre, France
| | - Patrice Lassus
- CNRS, UMR 5535, Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, Montpellier, France
| | - Benjamin Roche
- CREEC/MIVEGEC, UMR IRD/CNRS/UM 5290, 911 Avenue Agropolis, BP 64501, 34394 Montpellier Cedex 5, France; Unité mixte internationale de Modélisation Mathématique et Informatique des Systèmes Complexes (UMI IRD/UPMC UMMISCO), 32 Avenue Henri Varagnat, 93143 Bondy Cedex, France
| | - Frédéric Thomas
- CREEC/MIVEGEC, UMR IRD/CNRS/UM 5290, 911 Avenue Agropolis, BP 64501, 34394 Montpellier Cedex 5, France
| |
Collapse
|
189
|
Lei ZG, Ren XH, Wang SS, Liang XH, Tang YL. Immunocompromised and immunocompetent mouse models for head and neck squamous cell carcinoma. Onco Targets Ther 2016; 9:545-55. [PMID: 26869799 PMCID: PMC4734789 DOI: 10.2147/ott.s95633] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Mouse models can closely mimic human oral squamous epithelial carcinogenesis, greatly expand the in vivo research possibilities, and play a critical role in the development of diagnosis, monitoring, and treatment of head and neck squamous cell carcinoma. With the development of the recent research on the contribution of immunity/inflammation to cancer initiation and progression, mouse models have been divided into two categories, namely, immunocompromised and immunocompetent mouse models. And thus, this paper will review these two kinds of models applied in head and neck squamous cell carcinoma to provide a platform to understand the complicated histological, molecular, and genetic changes of oral squamous epithelial tumorigenesis.
Collapse
Affiliation(s)
- Zhen-Ge Lei
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital Affiliated to Nanchang University, Nanchang, Jiangxi, People's Republic of China
| | - Xiao-Hua Ren
- Department of Stomatology, Sichuan Medical Science Academy and Sichuan Provincial People's Hospital, Chengdu, Sichuan, People's Republic of China
| | - Sha-Sha Wang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Xin-Hua Liang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China; Department of Oral and Maxillofacial Surgery, West China College of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Ya-Ling Tang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China; Department of Oral Pathology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
| |
Collapse
|
190
|
Richichi B, Comito G, Renaudet O, Fiore M, Marra A, Stecca B, Pasquato L, Chiarugi P, Nativi C. Role of a Preorganized Scaffold Presenting Four Residues of a GM-3 Lactone Mimetic on Melanoma Progression. ACS Med Chem Lett 2016; 7:28-33. [PMID: 26819661 DOI: 10.1021/acsmedchemlett.5b00283] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 11/10/2015] [Indexed: 12/14/2022] Open
Abstract
Two tetravalent architectures, the glycocalix 7 and the RAFT 9, presenting four residues of a GM-3 ganglioside lactone mimetic, target the host compartment of melanoma and significantly abrogate the effect induced by cancer-associated fibroblasts (CAFs) contact + hypoxia in the motility and invasiveness of tumor cells. The data reported support the involvement of glycosphingolipids (GSLs) in hypoxia and show an interesting role played by compound 9 in targeting melanoma cells thereby interfering with melanoma progression. The unprecedented findings reported for the glycocluster 9 may contribute to the understanding of the critical and complex interactions between tumor cells and their local environment paving the way for new therapeutic agents.
Collapse
Affiliation(s)
- Barbara Richichi
- Department
of Chemistry “Ugo Schiff″, University of Florence, via della Lastruccia, 13 50019 Sesto F.no (FI), Italy
| | - Giuseppina Comito
- Department
of Experimental and Clinical Biomedical Sciences, Biochemistry, Human
Health Medical School, University of Florence, viale Morgagni 50, 50134 Firenze, Italy
| | - Olivier Renaudet
- Université Grenoble-Alpes, and CNRS, DCM, 38000 Grenoble, France
- Institut Universitaire de France, 103 Boulevard Saint-Michel, 75005 Paris, France
| | - Michele Fiore
- Université Grenoble-Alpes, and CNRS, DCM, 38000 Grenoble, France
| | - A. Marra
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, Ecole Nationale Supérieure de Chimie de Montpellier, 8 Rue de l’Ecole Normale, 34296 Montpellier cedex 5, France
| | - B. Stecca
- Core Research
Laboratory, Istituto Toscano Tumori, viale Pieraccini, 6, 50139 Firenze, Italy
| | - L. Pasquato
- Department
of Chemical and Pharmaceutical Sciences, University of Trieste, via L. Giorgieri 1, 34127 Trieste, Italy
| | - P. Chiarugi
- Department
of Experimental and Clinical Biomedical Sciences, Biochemistry, Human
Health Medical School, University of Florence, viale Morgagni 50, 50134 Firenze, Italy
| | - C. Nativi
- Department
of Chemistry “Ugo Schiff″, University of Florence, via della Lastruccia, 13 50019 Sesto F.no (FI), Italy
- FiorGen, Polo Scientifico e Tecnologico, via Sacconi, 6 50019 Sesto F.no (FI), Italy
| |
Collapse
|
191
|
Identification of two benzopyrroloxazines acting as selective GPER antagonists in breast cancer cells and cancer-associated fibroblasts. Future Med Chem 2016; 7:437-48. [PMID: 25875871 DOI: 10.4155/fmc.15.3] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND G-protein coupled estrogen receptor (GPER) is involved in numerous intracellular physiological and pathological events including cancer cell migration and proliferation. Its characterization is yet incomplete due to the limited number of specific ligands. RESULTS Two novel selective GPER antagonists, based on a benzo[b]pyrrolo[1,2-d][1,4]oxazin-4-one structure, have been designed and synthesized. Their binding to the receptor was confirmed by a competition assay, while the antagonist effects were ascertained by their capability to prevent the ligand-stimulated action of GPER. The transcription mediated by the classical estrogen receptor was not influenced, demonstrating selectivity for GPER. CONCLUSION These novel compounds may be considered useful leads toward the dissection of the GPER signaling and the development of new pharmacological treatments in breast cancer.
Collapse
|
192
|
Li J, Lan T, Zhang C, Zeng C, Hou J, Yang Z, Zhang M, Liu J, Liu B. Reciprocal activation between IL-6/STAT3 and NOX4/Akt signalings promotes proliferation and survival of non-small cell lung cancer cells. Oncotarget 2015; 6:1031-48. [PMID: 25504436 PMCID: PMC4359215 DOI: 10.18632/oncotarget.2671] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Accepted: 11/02/2014] [Indexed: 12/14/2022] Open
Abstract
Inflammatory cytokines and oxidative stress are two critical mediators in inflammation-associated cancer. Interleukin-6 (IL-6) is one of the most critical tumor-promoting cytokines in non-small cell lung cancer (NSCLC). In our recent study, we confirmed that NADPH oxidase 4 (NOX4), an important source of reactive oxygen species (ROS) production in NSCLC cells, promotes malignant progression of NSCLC. However, whether the crosstalk of NOX4 and IL-6 signalings exists in NSCLC remains undentified. In this study, we show that NOX4 expression is positively correlated with IL-6 expression in NSCLC tissues. Exogenous IL-6 treatment significantly enhances NOX4/ROS/Akt signaling in NSCLC cells. NOX4 also enhances IL-6 production and activates IL-6/STAT3 signaling in NSCLC cells. Specifically, NOX4 is confirmed to functionally interplay with IL-6 to promote NSCLC cell proliferation and survival. The in vivo results were similar to those obtained in vitro. These data indicate a novel NOX4-dependent link among IL-6 in the NSCLC microenvironment, oxidative stress in NSCLC cells and autocrined IL-6 in NSCLC cells. NOX4/Akt and IL-6/STAT3 signalings can reciprocally and positively regulate each other, leading to enhanced NSCLC cell proliferation and survival. Therefore, NOX4 may serve as a promising target against NSCLC alone with IL-6 signaling.
Collapse
Affiliation(s)
- Juan Li
- Clinical Pharmacy Department, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Tian Lan
- Vascular Biology Research Institute, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Cuixiang Zhang
- Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 110300, China.,Beijing Key Laboratory of Pharmacology of Chinese Materia Medica, Beijing 110300, China
| | - Cheng Zeng
- Clinical Pharmacy Department, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Jincai Hou
- Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 110300, China.,Beijing Key Laboratory of Pharmacology of Chinese Materia Medica, Beijing 110300, China
| | - Zhicheng Yang
- Department of Pharmacology, School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Min Zhang
- Department of Health Statistics, School of Public Health, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Jianxun Liu
- Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 110300, China.,Beijing Key Laboratory of Pharmacology of Chinese Materia Medica, Beijing 110300, China
| | - Bing Liu
- Clinical Pharmacy Department, Guangdong Pharmaceutical University, Guangzhou 510006, China
| |
Collapse
|
193
|
CD11b deficiency suppresses intestinal tumor growth by reducing myeloid cell recruitment. Sci Rep 2015; 5:15948. [PMID: 26526388 PMCID: PMC4630647 DOI: 10.1038/srep15948] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 10/01/2015] [Indexed: 02/07/2023] Open
Abstract
Mac-1 (CD11b) is expressed on bone marrow-derived immune cells. CD11b binds to ligands to regulate leukocyte adhesion and migration across the endothelium or epithelium. Here, we employed CD11b knockout mice and an ApcMin/+ spontaneous intestinal adenoma mouse model to clarify the function of CD11b in intestinal tumorigenesis. We showed that CD11b deficiency may contribute to the inhibition of myeloid cell trafficking to the tumor microenvironment and inactivated Wnt/β-catenin pathway to suppress tumor growth. This effect was partly mediated by inhibiting the myeloid cell-mediated decrease in TNF-α secretion, which inhibits the recruitment of myeloid-derived suppressor cells to the tumor microenvironment and subsequently induces IFN-γ and CXCL9 production. This work provides evidence for the mechanism by which CD11b may function as an important oncogene and highlights the potential of CD11b as a therapeutic target in CRC.
Collapse
|
194
|
Yan X, Li W, Pan L, Fu E, Xie Y, Chen M, Mu D. Lewis Lung Cancer Cells Promote SIGNR1(CD209b)-Mediated Macrophages Polarization Induced by IL-4 to Facilitate Immune Evasion. J Cell Biochem 2015; 117:1158-66. [PMID: 26447454 DOI: 10.1002/jcb.25399] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 10/06/2015] [Indexed: 01/16/2023]
Abstract
Tumor-associated macrophages are a prominent component of lung cancer and contribute to tumor progression by facilitating the immune evasion of cancer cells. DC-SIGN (CD209) assists in the immune evasion of a broad spectrum of pathogens and neoplasms by inhibiting the maturation of DCs and subsequent cytokines production. However, the expression of DC-SIGN in macrophages and its role in mediating immune evasion in lung cancer and the underlying mechanism remain unclear. Our study aimed to identify the immunosuppressive role of SIGNR1 in murine macrophage differentiation and lung cancer progression. We found that SIGNR1-positive RAW264.7 macrophages were enriched in mixed cultures with Lewis lung cancer cells (LLC) (ratio of RAW 264.7 to LLC being 1:1) after stimulation with IL-4. Moreover, LLC-educated macrophages exhibited significantly higher levels of IL-10 but lower IL-12 in response to IL-4 treatment as determined by RT-PCR and ELISA. However, inhibition of SIGNR1 markedly hampered the production of IL-10, indicating that SIGNR1 was indispensable for IL-4+LLC induced macrophage polarization towards the M2 subtype. Furthermore, polarized M2 cells immersed in a tumor microenvironment promoted the migration of LLCs, as measured by transwell assays, but migration was suppressed after blockade of SIGNR1 using CD209b antibody. In addition, IL-4+LLC-educated macrophages reduced the proliferation of the activated T cells and reduced IFN-γ-mediated Th1 response in T cells, while SIGNR1 inhibition rescued Th1 cell functions. In conclusion, murine SIGNR1 expressed in LLC-educated macrophages appears to mediate IL-4-induced RAW264.7 macrophage polarization and thus facilitate lung cancer evasion.
Collapse
Affiliation(s)
- Xiaolong Yan
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, Shaanxi Province, China
| | - Wenhai Li
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, Shaanxi Province, China
| | - Lei Pan
- Department of Respiration Medicine, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, Shaanxi Province, China
| | - Enqing Fu
- Department of Respiration Medicine, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, Shaanxi Province, China
| | - Yonghong Xie
- Department of Respiration Medicine, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, Shaanxi Province, China
| | - Min Chen
- Department of Respiration Medicine, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, Shaanxi Province, China
| | - Deguang Mu
- Department of Respiration Medicine, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, Shaanxi Province, China
| |
Collapse
|
195
|
Xia Q, Bai QR, Dong M, Sun X, Zhang H, Cui J, Xi H, Hu XL, Shen Q, Chen L. Interaction Between Gastric Carcinoma Cells and Neural Cells Promotes Perineural Invasion by a Pathway Involving VCAM1. Dig Dis Sci 2015; 60:3283-92. [PMID: 26108418 DOI: 10.1007/s10620-015-3758-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 06/10/2015] [Indexed: 12/12/2022]
Abstract
BACKGROUND Perineural invasion (PNI) is one of the important routes for local spread of gastric carcinoma associated with poor prognosis. However, the exact cellular characteristics and molecular mechanisms of PNI are still unclear. AIM To identify the interaction between gastric carcinoma cells and neural cells, and whether vascular cell adhesion molecule-1 (VCAM1) is involved in this process. METHODS We adopted in vitro cell coculture assays to investigate the cellular and molecular interaction between gastric cancer cells and neural cells. RESULTS We find upregulation of VCAM1 in clinical gastric cancer tissue samples. In in vitro tumor-neural cell coculture system, gastric cancer cells with high level of VCAM1 promote proliferation of neural progenitor cells and induce the process outgrowth and branching of neural cells. Reciprocally, neural cells enhance neurotropic migration and mobility of tumor cells. Repressing VCAM1 function through VCAM1 blocking antibody can attenuate these effects. CONCLUSIONS Our study indicates that VCAM1 is significantly involved in tumor invasion via mediating nerve-tumor interaction, which is a mutually beneficial process. It is possible that interaction between neural cells and tumor cells might contribute to PNI of gastric carcinoma. Inhibiting the activity of VCAM1 could be a potential strategy targeting PNI in gastric carcinoma therapy.
Collapse
Affiliation(s)
- Qijun Xia
- Department of General Surgery, General Hospital of Chinese People's Liberation Army (PLA), Medical College of PLA, Beijing, 100853, People's Republic of China.
| | - Qing-Ran Bai
- Center for Stem Cell Biology and Regenerative Medicine, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, People's Republic of China.
- School of Life Science, Tsinghua University, Beijing, 100084, People's Republic of China.
| | - Maosheng Dong
- Department of General Surgery, General Hospital of the PLA Second Artillery Force, Beijing, 100088, People's Republic of China.
| | - Xicai Sun
- Center for Stem Cell Biology and Regenerative Medicine, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, People's Republic of China.
| | - Haihong Zhang
- Department of General Surgery, General Hospital of Chinese People's Liberation Army (PLA), Medical College of PLA, Beijing, 100853, People's Republic of China.
| | - Jianxin Cui
- Department of General Surgery, General Hospital of Chinese People's Liberation Army (PLA), Medical College of PLA, Beijing, 100853, People's Republic of China.
| | - Hongqin Xi
- Department of General Surgery, General Hospital of Chinese People's Liberation Army (PLA), Medical College of PLA, Beijing, 100853, People's Republic of China.
| | - Xiao-Ling Hu
- Center for Stem Cell Biology and Regenerative Medicine, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, People's Republic of China.
| | - Qin Shen
- Center for Stem Cell Biology and Regenerative Medicine, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, People's Republic of China.
| | - Lin Chen
- Department of General Surgery, General Hospital of Chinese People's Liberation Army (PLA), Medical College of PLA, Beijing, 100853, People's Republic of China.
| |
Collapse
|
196
|
Ruksha TG, Aksenenko MB, Shvetsova YI. [Molecular and pathomorphological prognostic markers for melanoma: Current approaches and prospects]. Arkh Patol 2015; 77:71-77. [PMID: 26485783 DOI: 10.17116/patol201577471-77] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The incidence of melanoma demonstrates a persistent increasing tendency, which justifies the need to study and identify new prognostic markers for the development and course of this disease. The given paper shows current approaches to melanoma staging, including those to applying pathomorphological prognostic criteria, and discusses prospects for using the results of genomic and epigenomic studies of the carcinoma in clinical practice.
Collapse
Affiliation(s)
- T G Ruksha
- Professor V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, Ministry of Health of the Russia, Krasnoyarsk
| | - M B Aksenenko
- Professor V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, Ministry of Health of the Russia, Krasnoyarsk
| | - Yu I Shvetsova
- Professor V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, Ministry of Health of the Russia, Krasnoyarsk
| |
Collapse
|
197
|
Tellier C, Desmet D, Petit L, Finet L, Graux C, Raes M, Feron O, Michiels C. Cycling hypoxia induces a specific amplified inflammatory phenotype in endothelial cells and enhances tumor-promoting inflammation in vivo. Neoplasia 2015; 17:66-78. [PMID: 25622900 PMCID: PMC4309725 DOI: 10.1016/j.neo.2014.11.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Revised: 11/04/2014] [Accepted: 11/06/2014] [Indexed: 12/13/2022] Open
Abstract
Abnormal architecture of the tumor blood network, as well as heterogeneous erythrocyte flow, leads to temporal fluctuations in tissue oxygen tension exposing tumor and stromal cells to cycling hypoxia. Inflammation is another feature of tumor microenvironment and is considered as a new enabling characteristic of tumor progression. As cycling hypoxia is known to participate in tumor aggressiveness, the purpose of this study was to evaluate its role in tumor-promoting inflammation. Firstly, we assessed the impact of cycling hypoxia in vitro on endothelial inflammatory response induced by tumor necrosis factor α. Results showed that endothelial cells exposed to cycling hypoxia displayed an amplified proinflammatory phenotype, characterized by an increased expression of inflammatory cytokines, namely, interleukin (IL)-6 and IL-8; by an increased expression of adhesion molecules, in particular intercellular adhesion molecule-1 (ICAM-1); and consequently by an increase in THP-1 monocyte adhesion. This exacerbation of endothelial inflammatory phenotype occurs through nuclear factor-κB overactivation. Secondly, the role of cycling hypoxia was studied on overall tumor inflammation in vivo in tumor-bearing mice. Results showed that cycling hypoxia led to an enhanced inflammation in tumors as prostaglandin-endoperoxide synthase 2 (PTGS2), IL-6, CXCL1 (C-X-C motif ligand 1), and macrophage inflammatory protein 2 (murine IL-8 functional homologs) mRNA expression was increased and as a higher leukocyte infiltration was evidenced. Furthermore, cycling hypoxia-specific inflammatory phenotype, characterized by a simultaneous (baculoviral inhibitor of apoptosis repeat-containing 5)(low)/PTGS2(high)/ICAM-1(high)/IL-6(high)/IL-8(high) expression, is associated with a poor prognosis in human colon cancer. This new phenotype could thus be used in clinic to more precisely define prognosis for colon cancer patients. In conclusion, our findings evidenced for the first time the involvement of cycling hypoxia in tumor-promoting inflammation amplification.
Collapse
Affiliation(s)
- Céline Tellier
- Unit of Biochemistry and Cellular Biology (URBC), Namur Research Institute for LIfe Sciences (NARILIS), University of Namur (UNamur), 61 Rue de Bruxelles, B-5000 Namur, Belgium
| | - Déborah Desmet
- Unit of Biochemistry and Cellular Biology (URBC), Namur Research Institute for LIfe Sciences (NARILIS), University of Namur (UNamur), 61 Rue de Bruxelles, B-5000 Namur, Belgium
| | - Laurenne Petit
- Unit of Pharmacology and Therapeutics (FATH 5349), University of Louvain Medical School (UCL), 52 Avenue Mounier, B-1200 Bruxelles, Belgium
| | - Laure Finet
- Biobank, CHU Dinant-Mont-Godinne, UCL, 1 Avenue Docteur G.Thérasse, B-5530 Yvoir, Belgium
| | - Carlos Graux
- Biobank, CHU Dinant-Mont-Godinne, UCL, 1 Avenue Docteur G.Thérasse, B-5530 Yvoir, Belgium
| | - Martine Raes
- Unit of Biochemistry and Cellular Biology (URBC), Namur Research Institute for LIfe Sciences (NARILIS), University of Namur (UNamur), 61 Rue de Bruxelles, B-5000 Namur, Belgium
| | - Olivier Feron
- Unit of Pharmacology and Therapeutics (FATH 5349), University of Louvain Medical School (UCL), 52 Avenue Mounier, B-1200 Bruxelles, Belgium
| | - Carine Michiels
- Unit of Biochemistry and Cellular Biology (URBC), Namur Research Institute for LIfe Sciences (NARILIS), University of Namur (UNamur), 61 Rue de Bruxelles, B-5000 Namur, Belgium.
| |
Collapse
|
198
|
Li M, Luan F, Zhao Y, Hao H, Zhou Y, Han W, Fu X. Epithelial-mesenchymal transition: An emerging target in tissue fibrosis. Exp Biol Med (Maywood) 2015; 241:1-13. [PMID: 26361988 DOI: 10.1177/1535370215597194] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 06/19/2015] [Indexed: 12/18/2022] Open
Abstract
Epithelial-mesenchymal transition (EMT) is involved in a variety of tissue fibroses. Fibroblasts/myofibroblasts derived from epithelial cells contribute to the excessive accumulation of fibrous connective tissue in damaged tissue, which can lead to permanent scarring or organ malfunction. Therefore, EMT-related fibrosis cannot be neglected. This review highlights the findings that demonstrate the EMT to be a direct contributor to the fibroblast/myofibroblast population in the development of tissue fibrosis and helps to elucidate EMT-related anti-fibrotic strategies, which may enable the development of therapeutic interventions to suppress EMT and potentially reverse organ fibrosis.
Collapse
Affiliation(s)
- Meirong Li
- Wound Healing and Cell Biology Laboratory, Institute of Basic Medical Science, Chinese PLA General Hospital, Beijing 100853, P. R. China Trauma Treatment Center, Central Laboratory, Chinese PLA General Hospital Hainan Branch, Sanya 572014, P. R. China
| | - Fuxin Luan
- Trauma Treatment Center, Central Laboratory, Chinese PLA General Hospital Hainan Branch, Sanya 572014, P. R. China
| | - Yali Zhao
- Wound Healing and Cell Biology Laboratory, Institute of Basic Medical Science, Chinese PLA General Hospital, Beijing 100853, P. R. China Trauma Treatment Center, Central Laboratory, Chinese PLA General Hospital Hainan Branch, Sanya 572014, P. R. China
| | - Haojie Hao
- Wound Healing and Cell Biology Laboratory, Institute of Basic Medical Science, Chinese PLA General Hospital, Beijing 100853, P. R. China
| | - Yong Zhou
- Wound Healing and Cell Biology Laboratory, Institute of Basic Medical Science, Chinese PLA General Hospital, Beijing 100853, P. R. China
| | - Weidong Han
- Wound Healing and Cell Biology Laboratory, Institute of Basic Medical Science, Chinese PLA General Hospital, Beijing 100853, P. R. China
| | - Xiaobing Fu
- Wound Healing and Cell Biology Laboratory, Institute of Basic Medical Science, Chinese PLA General Hospital, Beijing 100853, P. R. China
| |
Collapse
|
199
|
Kakkad S, Glunde K, Penet MF, Bhujwalla ZM. Structural and functional roles of collagen 1 fibers in breast cancer metastasis: collagen 1 fiber density increases in lymph node-positive breast cancers. BREAST CANCER MANAGEMENT 2015. [DOI: 10.2217/bmt.15.8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Samata Kakkad
- JHU ICMIC Program, Division of Cancer Imaging Research, The Russell H Morgan Department of Radiology & Radiological Science, Johns Hopkins University School of Medicine, 208C Traylor Building, 720 Rutland Avenue, Baltimore, MD 21205, USA
| | - Kristine Glunde
- JHU ICMIC Program, Division of Cancer Imaging Research, The Russell H Morgan Department of Radiology & Radiological Science, Johns Hopkins University School of Medicine, 208C Traylor Building, 720 Rutland Avenue, Baltimore, MD 21205, USA
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Marie-France Penet
- JHU ICMIC Program, Division of Cancer Imaging Research, The Russell H Morgan Department of Radiology & Radiological Science, Johns Hopkins University School of Medicine, 208C Traylor Building, 720 Rutland Avenue, Baltimore, MD 21205, USA
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Zaver M Bhujwalla
- JHU ICMIC Program, Division of Cancer Imaging Research, The Russell H Morgan Department of Radiology & Radiological Science, Johns Hopkins University School of Medicine, 208C Traylor Building, 720 Rutland Avenue, Baltimore, MD 21205, USA
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
200
|
Anti-angiogenesis therapy and gap junction inhibition reduce MDA-MB-231 breast cancer cell invasion and metastasis in vitro and in vivo. Sci Rep 2015. [PMID: 26218768 PMCID: PMC4517444 DOI: 10.1038/srep12598] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Cancer cells secrete VEGF, which plays a key role in their growth, invasion, extravasation and metastasis. Direct cancer cell-endothelial cell interaction, mediated by gap junctions, is of critical importance in the extravasation process. In this study, we evaluated avastin (Av), an anti-VEGF antibody; and oleamide (OL), a gap junction inhibitor, using MDA-MB-231 human breast cancer cells in vitro and a xenograft murine model in vivo. Results showed that Av/OL significantly decreased proliferation, induced cell cycle arrest and decreased migration and invasion of MDA-MB-231 cells in vitro. In addition, Av/OL significantly decreased homo and hetero-cellular communication interaction between MDA-MDA and MDA-endothelial cells, respectively. The expression levels of several factors including VEGF, HIF1α, CXCR4, Cx26, Cx43, and MMP9 were attenuated upon Av/OL treatment in vitro. On the other hand, avastin, but not oleamide, reduced tumor size of NSG mice injected subdermally (s.d.) with MDA-MB-231 cells, which was also associated with increased survival. Furthermore, Av but also OL, separately, significantly increased the survival rate, and reduced pulmonary and hepatic metastatic foci, of intravenously (i.v.) injected mice. Finally, OL reduced MMP9 protein expression levels, better than Av and in comparisons to control, in the lungs of MDA-MB-231 i.v. injected NSG mice. In conclusion, while avastin has anti-angiogenic, anti-tumor and anti-metastatic activities, oleamide has anti-metastatic activity, presumably at the extravasation level, providing further evidence for the role of gap junction intercellular communication (GJIC) in cancer cell extravasation.
Collapse
|