51
|
Panda M, Biswal BK. Cell signaling and cancer: a mechanistic insight into drug resistance. Mol Biol Rep 2019; 46:5645-5659. [PMID: 31280421 DOI: 10.1007/s11033-019-04958-6] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 06/27/2019] [Indexed: 12/22/2022]
Abstract
Drug resistance is a major setback for advanced therapeutics in multiple cancers. The increasing prevalence of this resistance is a growing concern and bitter headache for the researchers since a decade. Hence, it is essential to revalidate the existing strategies available for cancer treatment and to look after a novel therapeutic approach for target based killing of cancer cells at the genetic level. This review outlines the different mechanisms enabling resistance including drug efflux, drug target alternation, alternative splicing, the release of the extracellular vesicle, tumor heterogeneity, epithelial-mesenchymal transition, tumor microenvironment, the secondary mutation in the receptor, epigenetic alternation, heterodimerization of receptors, amplification of target and amplification of components rather than the target. Furthermore, existing evidence and the role of various signaling pathways like EGFR, Ras, PI3K/Akt, Wnt, Notch, TGF-β, Integrin-ECM signaling in drug resistance are explained. Lastly, the prevention of this resistance by a contemporary therapeutic strategy, i.e., a combination of specific signaling pathway inhibitors and the cocktail of a cancer drug is summarized showing the new treatment strategies.
Collapse
Affiliation(s)
- Munmun Panda
- Cancer Drug Resistance Laboratory, Department of Life Science, National Institute of Technology, Sundargarh, Rourkela, Odisha, 769008, India
| | - Bijesh K Biswal
- Cancer Drug Resistance Laboratory, Department of Life Science, National Institute of Technology, Sundargarh, Rourkela, Odisha, 769008, India.
| |
Collapse
|
52
|
Beta1 integrin blockade overcomes doxorubicin resistance in human T-cell acute lymphoblastic leukemia. Cell Death Dis 2019; 10:357. [PMID: 31043590 PMCID: PMC6494825 DOI: 10.1038/s41419-019-1593-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 04/09/2019] [Accepted: 04/12/2019] [Indexed: 02/06/2023]
Abstract
Growing evidence indicates that cell adhesion to extracellular matrix (ECM) plays an important role in cancer chemoresistance. Leukemic T cells express several adhesion receptors of the β1 integrin subfamily with which they interact with ECM. However, the role of β1 integrins in chemoresistance of T-cell acute lymphoblastic leukemia (T-ALL) is still ill defined. In this study, we demonstrate that interactions of human T-ALL cell lines and primary blasts with three-dimensional matrices including Matrigel and collagen type I gel promote their resistance to doxorubicin via β1 integrin. The blockade of β1 integrin with a specific neutralizing antibody sensitized xenografted CEM leukemic cells to doxorubicin, diminished the leukemic burden in the bone marrow and resulted in the extension of animal survival. Mechanistically, Matrigel/β1 integrin interaction enhanced T-ALL chemoresistance by promoting doxorubicin efflux through the activation of the ABCC1 drug transporter. Finally, our findings showed that Matrigel/β1 interaction enhanced doxorubicin efflux and chemoresistance by activating the FAK-related proline-rich tyrosine kinase 2 (PYK2) as both PYK2 inhibitor and siRNA diminished the effect of Matrigel. Collectively, these results support the role of β1 integrin in T-ALL chemoresistance and suggest that the β1 integrin pathway can constitute a therapeutic target to avoid chemoresistance and relapsed-disease in human T-ALL.
Collapse
|
53
|
Zhao Y, Wetter NM, Wang X. Imaging Integrin Tension and Cellular Force at Submicron Resolution with an Integrative Tension Sensor. J Vis Exp 2019. [PMID: 31081814 DOI: 10.3791/59476] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Molecular tension transmitted by integrin-ligand bonds is the fundamental mechanical signal in the integrin pathway that plays significant roles in many cell functions and behaviors. To calibrate and image integrin tension with high force sensitivity and spatial resolution, we developed an integrative tension sensor (ITS), a DNA-based fluorescent tension sensor. The ITS is activated to fluoresce if sustaining a molecular tension, thus converting force to fluorescent signal at the molecular level. The tension threshold for ITS activation is tunable in the range of 10-60 pN that well covers the dynamic range of integrin tension in cells. On a substrate grafted with an ITS, the integrin tension of adherent cells is visualized by fluorescence and imaged at submicron resolution. The ITS is also compatible with cell structural imaging in both live cells and fixed cells. The ITS has been successfully applied to the study of platelet contraction and cell migration. This paper details the procedure for the synthesis and application of the ITS in the study of integrin-transmitted cellular force.
Collapse
Affiliation(s)
- Yuanchang Zhao
- Department of Physics and Astronomy, Iowa State University
| | | | - Xuefeng Wang
- Department of Physics and Astronomy, Molecular, Cellular, and Developmental Biology Interdepartmental Program, Iowa State University;
| |
Collapse
|
54
|
Identification of pathways associated with chemosensitivity through network embedding. PLoS Comput Biol 2019; 15:e1006864. [PMID: 30893303 PMCID: PMC6443184 DOI: 10.1371/journal.pcbi.1006864] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 04/01/2019] [Accepted: 02/09/2019] [Indexed: 12/27/2022] Open
Abstract
Basal gene expression levels have been shown to be predictive of cellular response to cytotoxic treatments. However, such analyses do not fully reveal complex genotype- phenotype relationships, which are partly encoded in highly interconnected molecular networks. Biological pathways provide a complementary way of understanding drug response variation among individuals. In this study, we integrate chemosensitivity data from a large-scale pharmacogenomics study with basal gene expression data from the CCLE project and prior knowledge of molecular networks to identify specific pathways mediating chemical response. We first develop a computational method called PACER, which ranks pathways for enrichment in a given set of genes using a novel network embedding method. It examines a molecular network that encodes known gene-gene as well as gene-pathway relationships, and determines a vector representation of each gene and pathway in the same low-dimensional vector space. The relevance of a pathway to the given gene set is then captured by the similarity between the pathway vector and gene vectors. To apply this approach to chemosensitivity data, we identify genes whose basal expression levels in a panel of cell lines are correlated with cytotoxic response to a compound, and then rank pathways for relevance to these response-correlated genes using PACER. Extensive evaluation of this approach on benchmarks constructed from databases of compound target genes and large collections of drug response signatures demonstrates its advantages in identifying compound-pathway associations compared to existing statistical methods of pathway enrichment analysis. The associations identified by PACER can serve as testable hypotheses on chemosensitivity pathways and help further study the mechanisms of action of specific cytotoxic drugs. More broadly, PACER represents a novel technique of identifying enriched properties of any gene set of interest while also taking into account networks of known gene-gene relationships and interactions. Gene expression levels have been used to study the cellular response to drug treatments. However, analysis of gene expression without considering gene interactions cannot fully reveal complex genotype-phenotype relationships. Biological pathways reveal the interactions among genes, thus providing a complementary way of understanding the drug response variation among individuals. In this paper, we aim to identify pathways that mediate the chemical response of each drug. We used the recently generated CTRP pharmacogenomics data and CCLE basal expression data to identify these pathways. We showed that using the prior knowledge encoded in molecular networks substantially improves pathway identification. In particular, we integrate genes and pathways into a large heterogeneous network in which links are protein-protein interactions and gene-pathway affiliations. We then project this heterogeneous network onto a low-dimensional space, which enables more precise similarity measurements between pathways and drug-response-correlated genes. Extensive experiments on two benchmarks show that our method substantially improved the pathway identification performance by using the molecular networks. More importantly, our method represents a novel technique of identifying enriched properties of any gene set of interest while also taking into account networks of known gene-gene relationships and interactions.
Collapse
|
55
|
Biophysical properties of cells for cancer diagnosis. J Biomech 2019; 86:1-7. [PMID: 30803699 DOI: 10.1016/j.jbiomech.2019.02.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 11/23/2018] [Accepted: 02/09/2019] [Indexed: 02/06/2023]
Abstract
Biophysical properties associated with the microenvironment of a tumor has been recognized as an important modulator for cell behaviour and function. Particularly, tissue rigidity is important during tumor carcinogenesis as it affects the tumor's ability to metastasis. Multiple downstream pathways are affected with a difference in rigidity of the extracellular matrix. The insight into tumor mechanosignalling represents a promising field that may lead to novel approaches for cancer diagnostics. Measurement of rigidity of the extracellular matrix or the tissue is a potential diagnostics approach for cancer detection. Altered extracellular matrix states persist for a long period of time and have lower heterogeneity compared to protein or genetic markers, therefore are more reliable as biomarkers. On the other hand, measurement of different kinase associated proteins or transcripts provide an early insight into potential transition of cells towards metastasis. Co-localization of transcriptional factors like YAP/TAZ provide an insight to determine if the cells are undergoing metastatic changes. This review explains the unique biophysical properties of the tumor microenvironment that present the potential targets for the diagnosis of cancer.
Collapse
|
56
|
Imaninezhad M, Hill L, Kolar G, Vogt K, Zustiak SP. Templated Macroporous Polyethylene Glycol Hydrogels for Spheroid and Aggregate Cell Culture. Bioconjug Chem 2019; 30:34-46. [PMID: 30562006 DOI: 10.1021/acs.bioconjchem.8b00596] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Macroporous cell-laden hydrogels have recently gained recognition for a wide range of biomedical and bioengineering applications. There are various approaches to create porosity in hydrogels, including lyophilization or foam formation. However, many do not allow a precise control over pore size or are not compatible with in situ cell encapsulation. Here, we developed novel templated macroporous hydrogels by encapsulating uniform degradable hydrogel microspheres produced via microfluidics into a hydrogel slab. The microspheres degraded completely leaving macropores behind. Microsphere degradation was dependent on the incubation medium, microsphere size, microsphere confinement in the hydrogel as well as cell encapsulation. Uniquely, the degradable microspheres were biocompatible and when laden with cells, the cells were deposited in the macropores upon microsphere degradation and formed multicellular aggregates. The hydrogel-encapsulated cell aggregates were used in a small drug screen to demonstrate the relevance of cell-matrix interactions for multicellular spheroid drug responsiveness. Hydrogel-grown spheroid cultures are increasingly important in applications such as in vitro tumor, hepatocellular, and neurosphere cultures and drug screening; hence, the templated cell aggregate-laden hydrogels described here would find utility in various applications.
Collapse
Affiliation(s)
- Mozhdeh Imaninezhad
- Department of Biomedical Engineering , Saint Louis University , Saint Louis , Missouri 63103 , United States
| | - Lindsay Hill
- Department of Biomedical Engineering , Saint Louis University , Saint Louis , Missouri 63103 , United States
| | - Grant Kolar
- Department of Pathology , Saint Louis University , Saint Louis , Missouri 63104 , United States
| | - Kyle Vogt
- Department of Biomedical Engineering , Saint Louis University , Saint Louis , Missouri 63103 , United States
| | - Silviya Petrova Zustiak
- Department of Biomedical Engineering , Saint Louis University , Saint Louis , Missouri 63103 , United States
| |
Collapse
|
57
|
Zhu Q, Sun H, Yang D, Tighe S, Liu Y, Zhu Y, Hu M. Cellular Substrates for Cell-Based Tissue Engineering of Human Corneal Endothelial Cells. Int J Med Sci 2019; 16:1072-1077. [PMID: 31523168 PMCID: PMC6743271 DOI: 10.7150/ijms.34440] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 05/21/2019] [Indexed: 12/15/2022] Open
Abstract
Corneal endothelial tissue engineering aims to find solutions for blindness due to endothelial dysfunction. A suitable combination of endothelial cells, substrates and environmental cues should be deployed for engineering functional endothelial tissues. This manuscript reviews up-to-date topics of corneal endothelial tissue engineering with special emphasis on biomaterial substrates and their properties, efficacy, and mechanisms of supporting functional endothelial cells in vitro.
Collapse
Affiliation(s)
- Qin Zhu
- Department of Ophthalmology, The Second People's Hospital of Yunnan Province (Fourth Affiliated Hospital of Kunming Medical University); Yunnan Eye Institute; Key Laboratory of Yunnan Province for the Prevention and Treatment of ophthalmology (2017DG008); Provincial Innovation Team for Cataract and Ocular Fundus Disease (2017HC010); Expert Workstation of Yao Ke (2017IC064), Kunming 650021, China
| | - Hong Sun
- Department of Ophthalmology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Dongmei Yang
- Department of Ophthalmology, The Second People's Hospital of Yunnan Province (Fourth Affiliated Hospital of Kunming Medical University); Yunnan Eye Institute; Key Laboratory of Yunnan Province for the Prevention and Treatment of ophthalmology (2017DG008); Provincial Innovation Team for Cataract and Ocular Fundus Disease (2017HC010); Expert Workstation of Yao Ke (2017IC064), Kunming 650021, China
| | - Sean Tighe
- Tissue Tech, Inc., Ocular Surface Center, and Ocular Surface Research & Education Foundation, Miami, FL, 33173 USA
| | - Yongsong Liu
- Department of Ophthalmology, Yan' An Hospital of Kunming City, Kunming, 650051, China
| | - Yingting Zhu
- Tissue Tech, Inc., Ocular Surface Center, and Ocular Surface Research & Education Foundation, Miami, FL, 33173 USA
| | - Min Hu
- Department of Ophthalmology, The Second People's Hospital of Yunnan Province (Fourth Affiliated Hospital of Kunming Medical University); Yunnan Eye Institute; Key Laboratory of Yunnan Province for the Prevention and Treatment of ophthalmology (2017DG008); Provincial Innovation Team for Cataract and Ocular Fundus Disease (2017HC010); Expert Workstation of Yao Ke (2017IC064), Kunming 650021, China
| |
Collapse
|
58
|
Abderrazak A, El Azreq MA, Naci D, Fortin PR, Aoudjit F. Alpha2beta1 Integrin (VLA-2) Protects Activated Human Effector T Cells From Methotrexate-Induced Apoptosis. Front Immunol 2018; 9:2269. [PMID: 30374344 PMCID: PMC6197073 DOI: 10.3389/fimmu.2018.02269] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 09/12/2018] [Indexed: 01/18/2023] Open
Abstract
β1 integrins are critical for T cell migration, survival and costimulation. The integrin α2β1, which is a receptor for collagen, also named VLA-2, is a major costimulatory pathway of effector T cells and has been implicated in arthritis pathogenesis. Herein, we have examined its ability to promote methotrexate (MTX) resistance by enhancing effector T cells survival. Our results show that attachment of anti-CD3-activated human polarized Th17 cells to collagen but not to fibronectin or laminin led to a significant reduction of MTX-induced apoptosis. The anti-CD3+collagen-rescued cells still produce significant amounts of IL-17 and IFNγ upon their reactivation indicating that their inflammatory nature is preserved. Mechanistically, we found that the prosurvival role of anti-CD3+collagen involves activation of the MTX transporter ABCC1 (ATP Binding Cassette subfamily C Member 1). Finally, the protective effect of collagen/α2β1 integrin on MTX-induced apoptosis also occurs in memory CD4+ T cells isolated from rheumatoid arthritis (RA) patients suggesting its clinical relevance. Together these results show that α2β1 integrin promotes MTX resistance of effector T cells, and suggest that it could contribute to the development of MTX resistance that is seen in RA.
Collapse
Affiliation(s)
- Amna Abderrazak
- Axe de Recherche sur les Maladies Infectieuses et Immunitaires, Centre de Recherche du CHU De Québec-Université Laval, Québec, QC, Canada
| | - Mohammed-Amine El Azreq
- Axe de Recherche sur les Maladies Infectieuses et Immunitaires, Centre de Recherche du CHU De Québec-Université Laval, Québec, QC, Canada
| | - Dalila Naci
- Axe de Recherche sur les Maladies Infectieuses et Immunitaires, Centre de Recherche du CHU De Québec-Université Laval, Québec, QC, Canada
| | - Paul R Fortin
- Axe de Recherche sur les Maladies Infectieuses et Immunitaires, Centre de Recherche du CHU De Québec-Université Laval, Québec, QC, Canada.,Division de Rhumatologie, Département de Médecine, Faculté de Médecine, Université Laval, Québec, QC, Canada
| | - Fawzi Aoudjit
- Axe de Recherche sur les Maladies Infectieuses et Immunitaires, Centre de Recherche du CHU De Québec-Université Laval, Québec, QC, Canada.,Département de Microbiologie-Infectiologie et D'immunologie, Faculté de Médecine, Université Laval, Québec, QC, Canada
| |
Collapse
|
59
|
Fu A, Peh YM, Ngan W, Wei N, Luo KQ. Rapid identification of antimicrometastases drugs using integrated model systems with two dimensional monolayer, three dimensional spheroids, and zebrafish xenotransplantation tumors. Biotechnol Bioeng 2018; 115:2828-2843. [DOI: 10.1002/bit.26816] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 07/10/2018] [Accepted: 08/09/2018] [Indexed: 01/28/2023]
Affiliation(s)
- Afu Fu
- School of Chemical and Biomedical Engineering, Nanyang Technological UniversitySingapore Singapore
| | - Yu Ming Peh
- School of Chemical and Biomedical Engineering, Nanyang Technological UniversitySingapore Singapore
| | - Weida Ngan
- School of Chemical and Biomedical Engineering, Nanyang Technological UniversitySingapore Singapore
| | - Na Wei
- School of Chemical and Biomedical Engineering, Nanyang Technological UniversitySingapore Singapore
| | - Kathy Qian Luo
- Faculty of Health Sciences, University of Macau, TaipaMacau China
| |
Collapse
|
60
|
Hou S, Tiriac H, Sridharan BP, Scampavia L, Madoux F, Seldin J, Souza GR, Watson D, Tuveson D, Spicer TP. Advanced Development of Primary Pancreatic Organoid Tumor Models for High-Throughput Phenotypic Drug Screening. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2018; 23:574-584. [PMID: 29673279 PMCID: PMC6013403 DOI: 10.1177/2472555218766842] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 02/26/2018] [Accepted: 03/05/2018] [Indexed: 12/30/2022]
Abstract
Traditional high-throughput drug screening in oncology routinely relies on two-dimensional (2D) cell models, which inadequately recapitulate the physiologic context of cancer. Three-dimensional (3D) cell models are thought to better mimic the complexity of in vivo tumors. Numerous methods to culture 3D organoids have been described, but most are nonhomogeneous and expensive, and hence impractical for high-throughput screening (HTS) purposes. Here we describe an HTS-compatible method that enables the consistent production of organoids in standard flat-bottom 384- and 1536-well plates by combining the use of a cell-repellent surface with a bioprinting technology incorporating magnetic force. We validated this homogeneous process by evaluating the effects of well-characterized anticancer agents against four patient-derived pancreatic cancer KRAS mutant-associated primary cells, including cancer-associated fibroblasts. This technology was tested for its compatibility with HTS automation by completing a cytotoxicity pilot screen of ~3300 approved drugs. To highlight the benefits of the 3D format, we performed this pilot screen in parallel in both the 2D and 3D assays. These data indicate that this technique can be readily applied to support large-scale drug screening relying on clinically relevant, ex vivo 3D tumor models directly harvested from patients, an important milestone toward personalized medicine.
Collapse
Affiliation(s)
- Shurong Hou
- The Scripps Research Institute Molecular
Screening Center, Department of Molecular Medicine, Scripps Florida, Jupiter, FL,
USA
- These authors contributed equally to
this work
| | - Hervé Tiriac
- Cancer Center, Cold Spring Harbor
Laboratory, Cold Spring Harbor, NY, USA
- These authors contributed equally to
this work
| | - Banu Priya Sridharan
- The Scripps Research Institute Molecular
Screening Center, Department of Molecular Medicine, Scripps Florida, Jupiter, FL,
USA
| | - Louis Scampavia
- The Scripps Research Institute Molecular
Screening Center, Department of Molecular Medicine, Scripps Florida, Jupiter, FL,
USA
| | - Franck Madoux
- The Scripps Research Institute Molecular
Screening Center, Department of Molecular Medicine, Scripps Florida, Jupiter, FL,
USA
- Amgen, Inc., Thousand Oaks, CA,
USA
| | - Jan Seldin
- Greiner Bio-One North America, Inc.,
Monroe, NC, USA
| | - Glauco R. Souza
- Nano3D Biosciences, Inc. and University
of Texas Health Science Center at Houston, Houston, TX, USA
| | | | - David Tuveson
- Cancer Center, Cold Spring Harbor
Laboratory, Cold Spring Harbor, NY, USA
- Co-communicated by D.T. and T.P.S
| | - Timothy P. Spicer
- The Scripps Research Institute Molecular
Screening Center, Department of Molecular Medicine, Scripps Florida, Jupiter, FL,
USA
- Co-communicated by D.T. and T.P.S
| |
Collapse
|
61
|
Lunasin is a novel therapeutic agent for targeting melanoma cancer stem cells. Oncotarget 2018; 7:84128-84141. [PMID: 27566591 PMCID: PMC5356649 DOI: 10.18632/oncotarget.11554] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 08/13/2016] [Indexed: 12/31/2022] Open
Abstract
Recent studies provide compelling evidence that melanoma is initiated and maintained by a small population of malignant cells called cancer-initiating cells (CICs) that exhibit stem-cell-like properties. Observations that CICs have a distinct biology when compared to that of the bulk tumor cells and, importantly, are resistant to chemotherapies and radiation, suggest that CICs are involved in invasion, metastasis, and ultimately relapse. Lunasin, a bioactive peptide present in soybean, has both chemopreventive activity and chemotherapeutic activity against multiple cancer types. In this study, we tested the potential of Lunasin to specifically target CICs in melanoma tumor cell populations. In vitro studies using human melanoma cell lines showed that Lunasin treatment decreased the size of a subpopulation of melanoma cells expressing the surrogate CIC marker, Aldehyde Dehydrogenase, concomitant with a reduction in the ability to form colonies in soft agar assays, and reduced tumor growth in mouse xenografts. Similarly, Lunasin inhibited colony formation by isolated melanoma CICs in soft agar and reduced oncosphere formation in vitro and substantially inhibited tumor growth in mouse xenografts. Mechanistic studies revealed that Lunasin treatment of isolated melanoma CICs induced expression of the melanocyte-associated differentiation markers Tyrosinase and Microphthalmia-associated Transcription Factor concomitant with reduced expression of the stemness factor NANOG. These findings document for the first time that Lunasin has significant therapeutic activity against melanoma by specifically targeting melanoma CICs, and inducing a more differentiated, non-CIC phenotype. Thus, Lunasin may represent a novel therapeutic option for both chemoresistant and advanced metastatic melanoma management.
Collapse
|
62
|
Addressing intra-tumoral heterogeneity and therapy resistance. Oncotarget 2018; 7:72322-72342. [PMID: 27608848 PMCID: PMC5342165 DOI: 10.18632/oncotarget.11875] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 08/24/2016] [Indexed: 12/12/2022] Open
Abstract
In the last several years, our appreciation of intra-tumoral heterogeneity has greatly increased due to accumulating evidence for the co-existence of genetically and epigenetically divergent cancer cells residing in different microenvironments within a tumor. Herein, we review recent literature discussing intra-tumoral heterogeneity in the context of therapy resistance mechanisms at the genetic, epigenetic and microenvironmental levels. We illustrate the influence of tumor microenvironment on therapy resistance and epigenetic states of cancer cells by highlighting the role of cancer stem cells in therapy resistance. We also summarize different strategies that have been employed to address various resistance mechanisms at genetic, epigenetic, and microenvironmental levels in preclinical and clinical studies. We propose that future personalized cancer therapy design needs to incorporate dynamic and comprehensive analyses of tumor heterogeneity landscape and multi-dimensional mechanisms of therapy resistance.
Collapse
|
63
|
Chai AWY, Cheung AKL, Dai W, Ko JMY, Ip JCY, Chan KW, Kwong DLW, Ng WT, Lee AWM, Ngan RKC, Yau CC, Tung SY, Lee VHF, Lam AKY, Pillai S, Law S, Lung ML. Metastasis-suppressing NID2, an epigenetically-silenced gene, in the pathogenesis of nasopharyngeal carcinoma and esophageal squamous cell carcinoma. Oncotarget 2018; 7:78859-78871. [PMID: 27793011 PMCID: PMC5346683 DOI: 10.18632/oncotarget.12889] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 10/19/2016] [Indexed: 12/13/2022] Open
Abstract
Nidogen-2 (NID2) is a key component of the basement membrane that stabilizes the extracellular matrix (ECM) network. The aim of the study is to analyze the functional roles of NID2 in the pathogenesis of nasopharyngeal carcinoma (NPC) and esophageal squamous cell carcinoma (ESCC). We performed genome-wide methylation profiling of NPC and ESCC and validated our findings using the methylation-sensitive high-resolution melting (MS-HRM) assay. Results showed that promoter methylation of NID2 was significantly higher in NPC and ESCC samples than in their adjacent non-cancer counterparts. Consistently, down-regulation of NID2 was observed in the clinical samples and cell lines of both NPC and ESCC. Re-expression of NID2 suppresses clonogenic survival and migration abilities of transduced NPC and ESCC cells. We showed that NID2 significantly inhibits liver metastasis. Mechanistic studies of signaling pathways also confirm that NID2 suppresses the EGFR/Akt and integrin/FAK/PLCγ metastasis-related pathways. This study provides novel insights into the crucial tumor metastasis suppression roles of NID2 in cancers.
Collapse
Affiliation(s)
- Annie Wai Yeeng Chai
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong (SAR), People's Republic of China
| | - Arthur Kwok Leung Cheung
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong (SAR), People's Republic of China
| | - Wei Dai
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong (SAR), People's Republic of China
| | - Josephine Mun Yee Ko
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong (SAR), People's Republic of China
| | - Joseph Chok Yan Ip
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong (SAR), People's Republic of China
| | - Kwok Wah Chan
- Center for Cancer Research, The University of Hong Kong, Hong Kong (SAR), People's Republic of China.,Department of Pathology, The University of Hong Kong, Hong Kong (SAR), People's Republic of China
| | - Dora Lai-Wan Kwong
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong (SAR), People's Republic of China.,Center for Nasopharyngeal Carcinoma Research, The University of Hong Kong, Hong Kong (SAR), People's Republic of China
| | - Wai Tong Ng
- Center for Nasopharyngeal Carcinoma Research, The University of Hong Kong, Hong Kong (SAR), People's Republic of China.,Department of Clinical Oncology, Pamela Youde Nethersole Eastern Hospital, Hong Kong (SAR), People's Republic of China
| | - Anne Wing Mui Lee
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong (SAR), People's Republic of China.,Center for Nasopharyngeal Carcinoma Research, The University of Hong Kong, Hong Kong (SAR), People's Republic of China
| | - Roger Kai Cheong Ngan
- Center for Nasopharyngeal Carcinoma Research, The University of Hong Kong, Hong Kong (SAR), People's Republic of China.,Department of Clinical Oncology, Queen Elizabeth Hospital, Hong Kong (SAR), People's Republic of China
| | - Chun Chung Yau
- Center for Nasopharyngeal Carcinoma Research, The University of Hong Kong, Hong Kong (SAR), People's Republic of China.,Department of Oncology, Princess Margaret Hospital, Hong Kong (SAR), People's Republic of China
| | - Stewart Yuk Tung
- Center for Nasopharyngeal Carcinoma Research, The University of Hong Kong, Hong Kong (SAR), People's Republic of China.,Department of Clinical Oncology, Tuen Mun Hospital, Hong Kong (SAR), People's Republic of China
| | - Victor Ho Fun Lee
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong (SAR), People's Republic of China.,Center for Nasopharyngeal Carcinoma Research, The University of Hong Kong, Hong Kong (SAR), People's Republic of China
| | - Alfred King-Yin Lam
- Department of Cancer Molecular Pathology, Griffith Medical School and Menzies Health Institute Queensland, Griffith University, Gold Coast, Australia
| | - Suja Pillai
- Department of Cancer Molecular Pathology, Griffith Medical School and Menzies Health Institute Queensland, Griffith University, Gold Coast, Australia
| | - Simon Law
- Center for Cancer Research, The University of Hong Kong, Hong Kong (SAR), People's Republic of China.,Department of Surgery, The University of Hong Kong, Hong Kong (SAR), People's Republic of China
| | - Maria Li Lung
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong (SAR), People's Republic of China.,Center for Cancer Research, The University of Hong Kong, Hong Kong (SAR), People's Republic of China.,Center for Nasopharyngeal Carcinoma Research, The University of Hong Kong, Hong Kong (SAR), People's Republic of China
| |
Collapse
|
64
|
MicroRNA-31 functions as a tumor suppressor and increases sensitivity to mitomycin-C in urothelial bladder cancer by targeting integrin α5. Oncotarget 2018; 7:27445-57. [PMID: 27050274 PMCID: PMC5053662 DOI: 10.18632/oncotarget.8479] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2016] [Accepted: 03/16/2016] [Indexed: 11/25/2022] Open
Abstract
Urothelial bladder cancer (UBC) is a common genitourinary malignancy. MiR-31, a well-identified miRNA, exhibits diverse properties in different cancers. However, the specific functions and mechanisms of miR-31 in UBC have not been investigated. In this study, tumor samples, especially invasive UBC, showed significantly reduced level of miR-31, as compared with normal urothelium. Prognostic analysis using the EORTC model showed that down-regulation of miR-31 correlated with higher risks of recurrence and progression in noninvasive UBC cases. Remarkably, overexpression of miR-31 mimics in UBC cell lines inhibited cell proliferation, migration and invasion. Integrin α5 (ITGA5), an integrin family member, was subsequently identified as a direct target of miR-31 in UBC cells. When treated with mitomycin-C (MMC), miR-31-expressing UBC cells displayed lower survival and higher apoptotic rates, and deactivated Akt and ERK. These effects arising from miR-31 overexpression were abrogated by ITGA5 restoration. Furthermore, miR-31 markedly inhibited tumor growth and increased the effectiveness of MMC in UBC xenografts. In summary, our data suggest that miR-31 is a prognostic predictor and can serve as a potential therapeutic target of UBC.
Collapse
|
65
|
Lovitt CJ, Shelper TB, Avery VM. Doxorubicin resistance in breast cancer cells is mediated by extracellular matrix proteins. BMC Cancer 2018; 18:41. [PMID: 29304770 PMCID: PMC5756400 DOI: 10.1186/s12885-017-3953-6] [Citation(s) in RCA: 220] [Impact Index Per Article: 31.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 12/21/2017] [Indexed: 11/24/2022] Open
Abstract
Background Cancer cell resistance to therapeutics can result from acquired or de novo-mediated factors. Here, we have utilised advanced breast cancer cell culture models to elucidate de novo doxorubicin resistance mechanisms. Methods The response of breast cancer cell lines (MCF-7 and MDA-MB-231) to doxorubicin was examined in an in vitro three-dimensional (3D) cell culture model. Cells were cultured with Matrigel™ enabling cellular arrangements into a 3D architecture in conjunction with cell-to-extracellular matrix (ECM) contact. Results Breast cancer cells cultured in a 3D ECM-based model demonstrated altered sensitivity to doxorubicin, when compared to those grown in corresponding two-dimensional (2D) monolayer culture conditions. Investigations into the factors triggering the observed doxorubicin resistance revealed that cell-to-ECM interactions played a pivotal role. This finding correlated with the up-regulation of pro-survival proteins in 3D ECM-containing cell culture conditions following exposure to doxorubicin. Inhibition of integrin signalling in combination with doxorubicin significantly reduced breast cancer cell viability. Furthermore, breast cancer cells grown in a 3D ECM-based model demonstrated a significantly reduced proliferation rate in comparison to cells cultured in 2D conditions. Conclusion Collectively, these novel findings reveal resistance mechanisms which may contribute to reduced doxorubicin sensitivity.
Collapse
Affiliation(s)
- Carrie J Lovitt
- Discovery Biology, Griffith Institute for Drug Discovery, Griffith University, Building N27, Brisbane Innovation Park, Nathan, QLD, 4111, Australia
| | - Todd B Shelper
- Discovery Biology, Griffith Institute for Drug Discovery, Griffith University, Building N27, Brisbane Innovation Park, Nathan, QLD, 4111, Australia
| | - Vicky M Avery
- Discovery Biology, Griffith Institute for Drug Discovery, Griffith University, Building N27, Brisbane Innovation Park, Nathan, QLD, 4111, Australia.
| |
Collapse
|
66
|
Vert A, Castro J, Ribó M, Vilanova M, Benito A. Transcriptional profiling of NCI/ADR-RES cells unveils a complex network of signaling pathways and molecular mechanisms of drug resistance. Onco Targets Ther 2018; 11:221-237. [PMID: 29379303 PMCID: PMC5757493 DOI: 10.2147/ott.s154378] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Background Ovarian cancer has the highest mortality rate among all the gynecological cancers. This is mostly due to the resistance of ovarian cancer to current chemotherapy regimens. Therefore, it is of crucial importance to identify the molecular mechanisms associated with chemoresistance. Methods NCI/ADR-RES is a multidrug-resistant cell line that is a model for the study of drug resistance in ovarian cancer. We carried out a microarray-derived transcriptional profiling analysis of NCI/ADR-RES to identify differentially expressed genes relative to its parental OVCAR-8. Results Gene-expression profiling has allowed the identification of genes and pathways that may be important for the development of drug resistance in ovarian cancer. The NCI/ADR-RES cell line has differential expression of genes involved in drug extrusion, inactivation, and efficacy, as well as genes involved in the architectural and functional reorganization of the extracellular matrix. These genes are controlled through different signaling pathways, including MAPK–Akt, Wnt, and Notch. Conclusion Our findings highlight the importance of using orthogonal therapies that target completely independent pathways to overcome mechanisms of resistance to both classical chemotherapeutic agents and molecularly targeted drugs.
Collapse
Affiliation(s)
- Anna Vert
- Protein Engineering Laboratory, Department of Biology, Faculty of Sciences, Universitat de Girona.,Biomedical Research Institute of Girona (IDIBGi), Girona, Spain
| | - Jessica Castro
- Protein Engineering Laboratory, Department of Biology, Faculty of Sciences, Universitat de Girona.,Biomedical Research Institute of Girona (IDIBGi), Girona, Spain
| | - Marc Ribó
- Protein Engineering Laboratory, Department of Biology, Faculty of Sciences, Universitat de Girona.,Biomedical Research Institute of Girona (IDIBGi), Girona, Spain
| | - Maria Vilanova
- Protein Engineering Laboratory, Department of Biology, Faculty of Sciences, Universitat de Girona.,Biomedical Research Institute of Girona (IDIBGi), Girona, Spain
| | - Antoni Benito
- Protein Engineering Laboratory, Department of Biology, Faculty of Sciences, Universitat de Girona.,Biomedical Research Institute of Girona (IDIBGi), Girona, Spain
| |
Collapse
|
67
|
Skhinas JN, Cox TR. The interplay between extracellular matrix remodelling and kinase signalling in cancer progression and metastasis. Cell Adh Migr 2017; 12:529-537. [PMID: 29168660 DOI: 10.1080/19336918.2017.1405208] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The extracellular matrix (ECM) is a master regulator of cellular phenotype and behaviour. It plays a crucial role in both normal tissue homeostasis and complex diseases such as cancer. The interplay between the intrinsic factors of cancer cells themselves, including their genotype and signalling networks; and the extrinsic factors of the tumour stroma, such as the ECM and ECM remodelling; together determine the fate and behaviour of cancer cells. As a consequence, tumour progression, metastatic spread and response to therapy are ultimately controlled by ECM-driven fine-tuning of intracellular kinase signalling. The ability to target and uncouple this interaction presents an emerging and promising potential in the treatment of cancer.
Collapse
Affiliation(s)
- Joanna N Skhinas
- a The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Cancer Division, St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney , Sydney, NSW , Australia
| | - Thomas R Cox
- a The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Cancer Division, St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney , Sydney, NSW , Australia
| |
Collapse
|
68
|
Petpiroon N, Sritularak B, Chanvorachote P. Phoyunnanin E inhibits migration of non-small cell lung cancer cells via suppression of epithelial-to-mesenchymal transition and integrin αv and integrin β3. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2017; 17:553. [PMID: 29284478 PMCID: PMC5747023 DOI: 10.1186/s12906-017-2059-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 12/12/2017] [Indexed: 01/17/2023]
Abstract
BACKGROUND The conversion of the epithelial phenotype of cancer cells into cells with a mesenchymal phenotype-so-called epithelial-mesenchymal transition (EMT)-has been shown to enhance the capacity of the cells to disseminate throughout the body. EMT is therefore becoming a potential target for anti-cancer drug discovery. Here, we showed that phoyunnanin E, a compound isolated from Dendrobium venustum, possesses anti-migration activity and addressed its mechanism of action. METHODS The cytotoxic and proliferative effects of phoyunnanin E on human non-small cell lung cancer-derived H460, H292, and A549 cells and human keratinocyte HaCaT cells were investigated by MTT assay. The effect of phoyunnanin E on EMT was evaluated by determining the colony formation and EMT markers. The migration and invasion of H460, H292, A549 and HaCaT cells was evaluated by wound healing assay and transwell invasion assay, respectively. EMT markers, integrins and migration-associated proteins were examined by western blot analysis. RESULTS Phoyunnanin E at the concentrations of 5 and 10 μM, which are non-toxic to H460, H292, A549 and HaCaT cells showed good potential to inhibit the migratory activity of three types of human lung cancer cells. The anti-migration effect of phoyunnanin E was shown to relate to the suppressed EMT phenotypes, including growth in anchorage-independent condition, cell motility, and EMT-specific protein markers (N-cadherin, vimentin, slug, and snail). In addition to EMT suppression, we found that phoyunnanin E treatment with 5 and 10 μM could decrease the cellular level of integrin αv and integrin β3, these integrins are frequently up-regulated in highly metastatic tumor cells. We further characterized the regulatory proteins in cell migration and found that the cells treated with phoyunnanin E exhibited a significantly lower level of phosphorylated focal adhesion kinase (p-FAK) and phosphorylated ATP-dependent tyrosine kinase (p-AKT), and their downstream effectors (including Ras-related C3 botulinum (Rac-GTP); Cell division cycle 42 (Cdc42); and Ras homolog gene family, member A (Rho-GTP)) in comparison to those of the non-treated control. CONCLUSIONS We have determined for the first time that phoyunnanin E could inhibit the motility of lung cancer cells via the suppression of EMT and metastasis-related integrins. This new information could support further development of this compound for anti-metastasis approaches.
Collapse
|
69
|
Bruney L, Liu Y, Grisoli A, Ravosa MJ, Stack MS. Integrin-linked kinase activity modulates the pro-metastatic behavior of ovarian cancer cells. Oncotarget 2017; 7:21968-81. [PMID: 26959113 PMCID: PMC5008337 DOI: 10.18632/oncotarget.7880] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 02/20/2016] [Indexed: 11/25/2022] Open
Abstract
Epithelial ovarian cancer (EOC) is the most fatal gynecologic cancer in the U.S., resulting in >14,000 deaths/year. Most women are diagnosed at late stage with widely disseminated intra-peritoneal metastatic disease, resulting in a 5-year survival rate of <30%. EOCs spread via direct extension and exfoliation into the peritoneal cavity, adhesion to peritoneal mesothelial cells, mesothelial cell retraction to expose sub-mseothelial matrix and anchoring in the type I collagen-rich matrix to generate secondary lesions. As a molecular-level understanding of EOC metastasis may identify novel therapeutic targets, the current study evaluated the expression and activity of integrin-linked kinase (ILK), a Ser/Thr protein kinase activated upon integrin-mediated adhesion. Results show that ILK is co-expressed in EOC with the pro-metastatic enzyme membrane type 1 matrix metalloproteinase (MT1-MMP) and catalyzed phosphorylation of the cytoplasmic tail of the proteinase. Downregulation of ILK expression or activity reduced adhesion to and invasion of collagen gels and organotypic meso-mimetic cultures. As an initial early event in EOC metastasis is integrin-mediated adhesion, these results suggest that further evaluation of ILK inhibitors as anti-metastatic agents in EOC is warranted.
Collapse
Affiliation(s)
- Lana Bruney
- Department of Medical Physiology & Pharmacology, University of Missouri School of Medicine, Columbia, MO, USA.,Harper Cancer Research Institute, University of Notre Dame, South Bend, IN, USA
| | - Yueying Liu
- Harper Cancer Research Institute, University of Notre Dame, South Bend, IN, USA.,Departments of Chemistry & Biochemistry and University of Notre Dame, Notre Dame, IN, USA
| | - Anne Grisoli
- Harper Cancer Research Institute, University of Notre Dame, South Bend, IN, USA
| | - Matthew J Ravosa
- Biological Sciences, University of Notre Dame, Notre Dame, IN, USA
| | - M Sharon Stack
- Department of Medical Physiology & Pharmacology, University of Missouri School of Medicine, Columbia, MO, USA.,Harper Cancer Research Institute, University of Notre Dame, South Bend, IN, USA.,Departments of Chemistry & Biochemistry and University of Notre Dame, Notre Dame, IN, USA
| |
Collapse
|
70
|
Augsburger D, Nelson PJ, Kalinski T, Udelnow A, Knösel T, Hofstetter M, Qin JW, Wang Y, Gupta AS, Bonifatius S, Li M, Bruns CJ, Zhao Y. Current diagnostics and treatment of fibrosarcoma -perspectives for future therapeutic targets and strategies. Oncotarget 2017; 8:104638-104653. [PMID: 29262667 PMCID: PMC5732833 DOI: 10.18632/oncotarget.20136] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 07/29/2017] [Indexed: 12/18/2022] Open
Abstract
Adult-type fibrosarcoma is a rare and highly aggressive subtype of soft tissue sarcomas. Due to the existence of other spindle-cell shaped sarcomas, its diagnosis is always one of exclusion. The likelihood of misdiagnoses between similar tumour entities is high, and often leads to inappropriate tumour treatment. We summarize here the main features of fibrosarcoma. When fibrosarcoma is appropriately diagnosed, the patient`s overall prognosis is generally quite poor. Fibrosarcoma is characterized by its low sensitivity towards radio- and chemotherapy as well as by its high rate of tumour recurrences. Thus it is important to identify new methods to improve treatment of this tumour entity. We discuss some promising new directions in fibrosarcoma research, specifically focusing on more effective targeting of the tumour microenvironment. Communication between tumour cells and their surrounding stromal tissue play a crucial role in cancer progression, invasion, metastasis and chemosensitivity. The therapeutic potential of targeting the tumour microenvironment is addressed.
Collapse
Affiliation(s)
- Daniela Augsburger
- Department of General, Visceral und Vascular Surgery, Otto-von-Guericke University, Magdeburg, Germany
| | - Peter J. Nelson
- Clinical Biochemistry Group, Medizinische Klinik und Poliklinik IV, University of Munich, Munich, Germany
| | - Thomas Kalinski
- Department of Pathology, Otto-von-Guericke University, Magdeburg, Germany
| | - Andrej Udelnow
- Department of General, Visceral und Vascular Surgery, Otto-von-Guericke University, Magdeburg, Germany
| | - Thomas Knösel
- Institute of Pathology, Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Monika Hofstetter
- Clinical Biochemistry Group, Medizinische Klinik und Poliklinik IV, University of Munich, Munich, Germany
| | - Ji Wei Qin
- Department of General, Visceral und Vascular Surgery, Otto-von-Guericke University, Magdeburg, Germany
| | - Yan Wang
- Department of General, Visceral und Vascular Surgery, Otto-von-Guericke University, Magdeburg, Germany
| | - Arvid Sen Gupta
- Department of General, Visceral und Vascular Surgery, Otto-von-Guericke University, Magdeburg, Germany
| | - Susanne Bonifatius
- Department of General, Visceral und Vascular Surgery, Otto-von-Guericke University, Magdeburg, Germany
| | - Minglun Li
- Department of Radiation Oncology, Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Christiane J. Bruns
- Department of General, Visceral und Vascular Surgery, Otto-von-Guericke University, Magdeburg, Germany
- Department of General, Visceral and Cancer Surgery, University Hospital of Cologne, Cologne, Germany
- Present address: Department of General, Visceral and Cancer Surgery, University Hospital of Cologne, Cologne, Germany
| | - Yue Zhao
- Department of General, Visceral und Vascular Surgery, Otto-von-Guericke University, Magdeburg, Germany
- Department of General, Visceral and Cancer Surgery, University Hospital of Cologne, Cologne, Germany
- Present address: Department of General, Visceral and Cancer Surgery, University Hospital of Cologne, Cologne, Germany
| |
Collapse
|
71
|
Schwenk R, Stehning T, Bischoff I, Ullrich A, Kazmaier U, Fürst R. The pretubulysin-induced exposure of collagen is caused by endothelial cell retraction that results in an increased adhesion and decreased transmigration of tumor cells. Oncotarget 2017; 8:77622-77633. [PMID: 29100413 PMCID: PMC5652804 DOI: 10.18632/oncotarget.20746] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Accepted: 08/04/2017] [Indexed: 12/11/2022] Open
Abstract
Microtubule-targeting agents (MTAs) are the most widely used chemotherapeutic drugs. Pretubulysin (PT), a biosynthetic precursor of the myxobacterial tubulysins, was recently identified as a novel MTA. Besides its strong anti-tumoral activities, PT attenuates tumor angiogenesis, exerts anti-vascular actions on tumor vessels and decreases cancer metastasis formation in vivo. The aim of the present study was to analyze the impact of PT on the interaction of endothelial and tumor cells in vitro to gain insights into the mechanism underlying its anti-metastatic effect. The influence of PT on tumor cell adhesion and transmigration onto/through the endothelium as well as its influence on cell adhesion molecules and the chemokine system CXCL12/CXCR4 was investigated. Treatment of human endothelial cells with PT increased the adhesion of breast cancer cells to the endothelial monolayer, whereas their transmigration through the endothelium was strongly reduced. Interestingly, the PT-induced upregulation of ICAM-1, VCAM-1 and CXCL12 were dispensable for the PT-evoked tumor cell adhesion. Tumor cells preferred to adhere to collagen exposed within PT-triggered endothelial gaps via β1-integrins on the tumor cell surface. Taken together, our study provides, at least in part, an explanation for the anti-metastatic potential of PT.
Collapse
Affiliation(s)
- Rebecca Schwenk
- Institute of Pharmaceutical Biology, Goethe University, Frankfurt, Germany
| | - Tanja Stehning
- Institute of Pharmaceutical Biology, Goethe University, Frankfurt, Germany
| | - Iris Bischoff
- Institute of Pharmaceutical Biology, Goethe University, Frankfurt, Germany
| | - Angelika Ullrich
- Institute of Organic Chemistry, Saarland University, Saarbrücken, Germany
| | - Uli Kazmaier
- Institute of Organic Chemistry, Saarland University, Saarbrücken, Germany
| | - Robert Fürst
- Institute of Pharmaceutical Biology, Goethe University, Frankfurt, Germany
| |
Collapse
|
72
|
Abstract
OPINION STATEMENT The survival rate for patients with advanced stages of squamous cell carcinoma of the head and neck (SCCHN) remains poor despite multimodal treatment options. Cetuximab, an anti-EGFR inhibitor, is the only FDA-approved targeted agent for this disease. Recent findings have implicated modifications of the microenvironment and, consequently, phenotypical modifications of the cancer cell, in treatment resistance mechanisms. For many years, cancer research has focused mainly on targetable sites on or inside the cancer cell. Nowadays, in preclinical and clinical studies, a greater emphasis is being placed on drugs that target the tumor microenvironment. Potential targets relate to tumor vascularization, immunology, extracellular matrix components, or cancer-associated fibroblasts. The combination of these new agents with standard treatment options is of particular interest to overcome resistance mechanisms and/or to increase treatment efficacy. Whereas antiangiogenic agents show poor clinical activity, immunotherapy seems to be a more promising tool with an objective response rate (ORR) of 20 % in patients with recurrent and/or metastatic squamous cell carcinoma (R/M SCC). Other targets, located inside the extracellular matrix or on cancer associated fibroblasts, are under preclinical investigation. These new agents all need to be tested in clinical trials alone, or in combination with standard treatment modalities, based on preclinical data. To increase our knowledge of the complex network between the cancer cell and its environment, preclinical studies should consider co-culture models, and clinical studies should incorporate a translational research objective.
Collapse
|
73
|
Wang X, Xu T, Gao F, He H, Zhu Y, Shen Z. Targeting of CCN2 suppresses tumor progression and improves chemo-sensitivity in urothelial bladder cancer. Oncotarget 2017; 8:66316-66327. [PMID: 29029514 PMCID: PMC5630414 DOI: 10.18632/oncotarget.19987] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2017] [Accepted: 07/19/2017] [Indexed: 11/25/2022] Open
Abstract
Urothelial bladder cancer (UBC) is the most common urinary neoplasm in China. CCN family protein 2 (CCN2), a cysteine-rich matricellular protein, is abnormally expressed in several cancer types and involved in tumor progression or chemo-resistance. However, detailed expression patterns and effects of CCN2 in UBC still remain unknown. We found that down-regulation of CCN2 suppressed proliferation, migration and invasion of UBC cells in vitro and targeting of CCN2 decelerated xenograft growth in vivo. When treated with mitomycin C (MMC), CCN2-scilencing UBC cells showed lower survival and higher apoptotic rates and these effects were probably mediated via inactivation of Akt and Erk pathways. We also demonstrated the clinical significance of CCN2 expression, which was higher in UBC tissues and associated with advanced tumor stage and high pathologic grade. Taken together, our data suggest that CCN2 is an oncogene in UBC and might serve as a matricellular target for improving chemotherapeutic efficacy.
Collapse
Affiliation(s)
- Xiaojing Wang
- Department of Urology, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Tianyuan Xu
- Department of Urology, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Fengbin Gao
- Department of Urology, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Hongchao He
- Department of Urology, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yu Zhu
- Department of Urology, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Zhoujun Shen
- Department of Urology, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China.,Department of Urology, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
74
|
Deshmukh SK, Srivastava SK, Zubair H, Bhardwaj A, Tyagi N, Al-Ghadhban A, Singh AP, Dyess DL, Carter JE, Singh S. Resistin potentiates chemoresistance and stemness of breast cancer cells: Implications for racially disparate therapeutic outcomes. Cancer Lett 2017; 396:21-29. [PMID: 28302531 PMCID: PMC5437742 DOI: 10.1016/j.canlet.2017.03.010] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 03/04/2017] [Accepted: 03/07/2017] [Indexed: 12/27/2022]
Abstract
Breast cancer (BC) continues to be the most frequently diagnosed cancer in American women, which disproportionately affects women of African-American (AA) descent. Previously, we reported greater serum levels of resistin in AA BC patients relative to Caucasian-American (CA) patients, and established its role in growth and aggressiveness of breast tumor cells. Here we have investigated the role of resistin in BC-chemoresistance. MDA-MB-231 and MDA-MB-468 BC cells of CA and AA origin, respectively, were incubated with resistin prior to doxorubicin treatment. Our data suggest that resistin conferred chemoresistance to both BC cell lines; however, the effect on AA cells was more profound. Furthermore, the resistin-induced doxorubicin-resistance was shown to occur due to suppression of apoptosis. Resistin treatment also affected the stemness of BC cells, as suggested by reduced cell surface expression of CD24, induced expression of CD44 and ALDH1, and increased capability of cells to form mammospheres. Mechanistic studies revealed that resistin-induced chemoresistance, apoptosis and stemness of BC cells were mediated through STAT3 activation. Taken together, our findings provide novel insight into the role of resistin in BC biology, and strengthen its role in racially disparate clinical outcomes.
Collapse
Affiliation(s)
- Sachin K Deshmukh
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, USA
| | - Sanjeev K Srivastava
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, USA
| | - Haseeb Zubair
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, USA
| | - Arun Bhardwaj
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, USA
| | - Nikhil Tyagi
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, USA
| | - Ahmed Al-Ghadhban
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, USA
| | - Ajay P Singh
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, USA; Department of Biochemistry and Molecular Biology, College of Medicine, University of South Alabama, Mobile, AL, USA
| | - Donna L Dyess
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, USA
| | - James E Carter
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, AL, USA
| | - Seema Singh
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, USA; Department of Biochemistry and Molecular Biology, College of Medicine, University of South Alabama, Mobile, AL, USA.
| |
Collapse
|
75
|
Tripathi SC, Fahrmann JF, Celiktas M, Aguilar M, Marini KD, Jolly MK, Katayama H, Wang H, Murage EN, Dennison JB, Watkins DN, Levine H, Ostrin EJ, Taguchi A, Hanash SM. MCAM Mediates Chemoresistance in Small-Cell Lung Cancer via the PI3K/AKT/SOX2 Signaling Pathway. Cancer Res 2017. [PMID: 28646020 DOI: 10.1158/0008-5472.can-16-2874] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Despite favorable responses to initial therapy, small-cell lung cancer (SCLC) relapse occurs within a year and exhibits resistance to multiple drugs. Because of limited accessibility of patient tissues for research purposes, SCLC patient-derived xenografts (PDX) have provided the best opportunity to address this limitation. Here, we sought to identify novel mechanisms involved in SCLC chemoresistance. Through in-depth proteomic profiling, we identified MCAM as a markedly upregulated surface receptor in chemoresistant SCLC cell lines and in chemoresistant PDX compared with matched treatment-naïve tumors. MCAM depletion in chemoresistant cells reduced cell proliferation and reduced the IC50 inhibitory concentration of chemotherapeutic drugs in vitro This MCAM-mediated sensitization to chemotherapy occurred via SOX2-dependent upregulation of mitochondrial 37S ribosomal protein 1/ATP-binding cassette subfamily C member 1 (MRP1/ABCC1) and the PI3/AKT pathway. Metabolomic profiling revealed that MCAM modulated lactate production in chemoresistant cells that exhibit a distinct metabolic phenotype characterized by low oxidative phosphorylation. Our results suggest that MCAM may serve as a novel therapeutic target to overcome chemoresistance in SCLC. Cancer Res; 77(16); 4414-25. ©2017 AACR.
Collapse
Affiliation(s)
- Satyendra C Tripathi
- Department of Clinical Cancer Prevention, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Johannes F Fahrmann
- Department of Clinical Cancer Prevention, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Muge Celiktas
- Department of Clinical Cancer Prevention, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mitzi Aguilar
- Department of Clinical Cancer Prevention, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Kieren D Marini
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Mohit K Jolly
- Center for Theoretical Biological Physics, Rice University, Houston, Texas
| | - Hiroyuki Katayama
- Department of Clinical Cancer Prevention, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hong Wang
- Department of Clinical Cancer Prevention, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Eunice N Murage
- Department of Clinical Cancer Prevention, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jennifer B Dennison
- Department of Clinical Cancer Prevention, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - D Neil Watkins
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Herbert Levine
- Center for Theoretical Biological Physics, Rice University, Houston, Texas
| | - Edwin J Ostrin
- Department of Pulmonary Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ayumu Taguchi
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Samir M Hanash
- Department of Clinical Cancer Prevention, University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
76
|
Zhang K, Myllymäki SM, Gao P, Devarajan R, Kytölä V, Nykter M, Wei GH, Manninen A. Oncogenic K-Ras upregulates ITGA6 expression via FOSL1 to induce anoikis resistance and synergizes with αV-Class integrins to promote EMT. Oncogene 2017; 36:5681-5694. [PMID: 28604746 PMCID: PMC5658677 DOI: 10.1038/onc.2017.177] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Revised: 04/11/2017] [Accepted: 05/04/2017] [Indexed: 12/17/2022]
Abstract
In many cancer types, integrin-mediated signaling regulates proliferation, survival and invasion of tumorigenic cells. However, it is still unclear how integrins crosstalk with oncogenes to regulate tumorigenesis and metastasis. Here we show that oncogenic K-RasV12 upregulates α6-integrin expression in Madin–Darby canine kidney (MDCK) cells via activation of the mitogen-activated protein kinase/extracellular signal-regulated kinase (ERK)/Fos-related antigen 1-signaling cascade. Activated α6-integrins promoted metastatic capacity and anoikis resistance, and led to perturbed growth of MDCK cysts. Transcriptomic analysis of K-RasV12-transformed MDCK cells also revealed robust downregulation of αV-class integrins. Re-expression of αV-integrin in K-RasV12-transformed MDCK cells synergistically upregulated the expression of Zinc finger E-box-binding homeobox 1 and Twist-related protein 1 and triggered epithelial-mesenchymal transition leading to induced cell motility and invasion. These results delineate the signaling cascades connecting oncogenic K-RasV12 with α6- and αV-integrin functions to modulate cancer cell survival and tumorigenesis, and reveal new possible strategies to target highly oncogenic K-RasV12 mutants.
Collapse
Affiliation(s)
- K Zhang
- Biocenter Oulu, Centre of Excellence in Cell-Extracellular Matrix Research, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - S-M Myllymäki
- Biocenter Oulu, Centre of Excellence in Cell-Extracellular Matrix Research, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - P Gao
- Biocenter Oulu, Centre of Excellence in Cell-Extracellular Matrix Research, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - R Devarajan
- Biocenter Oulu, Centre of Excellence in Cell-Extracellular Matrix Research, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - V Kytölä
- Prostate Cancer Research Center, Institute of Biomedical Technology and BioMediTech, University of Tampere and Tampere University Hospital, Tampere, Finland
| | - M Nykter
- Prostate Cancer Research Center, Institute of Biomedical Technology and BioMediTech, University of Tampere and Tampere University Hospital, Tampere, Finland
| | - G-H Wei
- Biocenter Oulu, Centre of Excellence in Cell-Extracellular Matrix Research, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - A Manninen
- Biocenter Oulu, Centre of Excellence in Cell-Extracellular Matrix Research, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| |
Collapse
|
77
|
Hamurcu Z, Kahraman N, Ashour A, Ozpolat B. FOXM1 transcriptionally regulates expression of integrin β1 in triple-negative breast cancer. Breast Cancer Res Treat 2017; 163:485-493. [PMID: 28361350 DOI: 10.1007/s10549-017-4207-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 03/15/2017] [Indexed: 01/10/2023]
Abstract
PURPOSE Triple-negative breast cancer (TNBC) is an aggressive type of breast cancer and associated with early metastasis, drug resistance, and poor patient survival. Fork head box M1 (FOXM1) is considered as an emerging molecular target due to its oncogenic role and high overexpression profile in 85% in TNBC. However, molecular mechanisms by which FOXM1 transcription factor mediate its oncogenic effects are not fully understood. Integrin β1 is often upregulated in invasive breast cancers and associated with poor clinical outcome and shorter overall patient survival in TNBC. However, the mechanisms regulating integrin β1 (ITGB1) gene expression have not been well elucidated. METHODS Normal breast epithelium (MCF10A) and TNBC cells (i.e., MDA-MB-231, BT-20 MDA-MB436) were used for the study. Small interfering RNA (siRNA)-based knockdown was used to inhibit Integrin β1 gene (mRNA) and protein expressions, which are detected by RT-PCR and Western blot, respectively. Chromatin immunoprecipitation (ChiP) and gene reporter (Luciferase) assays were used to demonstrate that FOXM1 transcription factor binds to the promoter of Integrin β1 gene and drives its expression. RESULTS We demonstrated that FOXM1 directly binds to the promoter of integrin β1 gene and transcriptionally regulates its expression and activity of focal adhesion kinase (FAK) in TNBC cells. CONCLUSION Our study suggests that FOXM1 transcription factor regulates Integrin β1 gene expression and that FOXM1/ Integrin-β1/FAK axis may play an important role in the progression of TNBC.
Collapse
Affiliation(s)
- Zuhal Hamurcu
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 422, Houston, TX, 77030, USA.,Faculty of Medicine, Department of Medical Biology, Erciyes University, Kayseri, Turkey.,Betül-Ziya Eren Genome and Stem Cell Center, Erciyes University, Kayseri, Turkey
| | - Nermin Kahraman
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 422, Houston, TX, 77030, USA
| | - Ahmed Ashour
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 422, Houston, TX, 77030, USA
| | - Bulent Ozpolat
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 422, Houston, TX, 77030, USA. .,Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
78
|
Grabowski B, Schmidt MA, Rüter C. Immunomodulatory Yersinia outer proteins (Yops)-useful tools for bacteria and humans alike. Virulence 2017; 8:1124-1147. [PMID: 28296562 DOI: 10.1080/21505594.2017.1303588] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Human-pathogenic Yersinia produce plasmid-encoded Yersinia outer proteins (Yops), which are necessary to down-regulate anti-bacterial responses that constrict bacterial survival in the host. These Yops are effectively translocated directly from the bacterial into the target cell cytosol by the type III secretion system (T3SS). Cell-penetrating peptides (CPPs) in contrast are characterized by their ability to autonomously cross cell membranes and to transport cargo - independent of additional translocation systems. The recent discovery of bacterial cell-penetrating effector proteins (CPEs) - with the prototype being the T3SS effector protein YopM - established a new class of autonomously translocating immunomodulatory proteins. CPEs represent a vast source of potential self-delivering, anti-inflammatory therapeutics. In this review, we give an update on the characteristic features of the plasmid-encoded Yops and, based on recent findings, propose the further development of these proteins for potential therapeutic applications as natural or artificial cell-penetrating forms of Yops might be of value as bacteria-derived biologics.
Collapse
Affiliation(s)
- Benjamin Grabowski
- a Institute of Infectiology - Centre for Molecular Biology of Inflammation (ZMBE), University of Münster , Münster , Germany
| | - M Alexander Schmidt
- a Institute of Infectiology - Centre for Molecular Biology of Inflammation (ZMBE), University of Münster , Münster , Germany
| | - Christian Rüter
- a Institute of Infectiology - Centre for Molecular Biology of Inflammation (ZMBE), University of Münster , Münster , Germany
| |
Collapse
|
79
|
Abstract
During cancer progression, the extracellular matrix (ECM) undergoes dramatic changes, which promote cancer cell migration and invasion. In the remodeled tumor ECM, fibronectin (FN) level is upregulated to assist tumor growth, progression, and invasion. FN serves as a central organizer of ECM molecules and mediates the crosstalk between the tumor microenvironment and cancer cells. Its upregulation is correlated with angiogenesis, cancer progression, metastasis, and drug resistance. A number of FN-targeting ligands have been developed for cancer imaging and therapy. Thus far, FN-targeting imaging agents have been tested for nuclear imaging, MRI, and fluorescence imaging, for tumor detection and localization. FN-targeting therapeutics, including nuclear medicine, chemotherapy drugs, cytokines, and photothermal moieties, were also developed in cancer therapy. Because of the prevalence of FN overexpression in cancer, FN targeting imaging agents and therapeutics have the promise of broad applications in the diagnosis, treatment, and image-guided interventions of many types of cancers. This review will summarize current understanding on the role of FN in cancer, discuss the design and development of FN-targeting agents, and highlight the applications of these FN-targeting agents in cancer imaging and therapy.
Collapse
Affiliation(s)
- Zheng Han
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Zheng-Rong Lu
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| |
Collapse
|
80
|
Sison EAR, Kurre P, Kim YM. Understanding the bone marrow microenvironment in hematologic malignancies: A focus on chemokine, integrin, and extracellular vesicle signaling. Pediatr Hematol Oncol 2017; 34:365-378. [PMID: 29211600 PMCID: PMC6516746 DOI: 10.1080/08880018.2017.1395938] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Signaling between leukemia cells and nonhematopoietic cells in the bone marrow microenvironment contributes to leukemia cell growth and survival. This complicated extrinsic mechanism of chemotherapy resistance relies on a number of pathways and factors, some of which have yet to be determined. Research on cell-cell crosstalk the bone marrow microenvironment in acute leukemia was presented at the 2016 annual Therapeutic Advances in Childhood Leukemia (TACL) investigator meeting. This review summarizes the mini-symposium proceedings and focuses on chemokine signaling via the cell surface receptor CXCR4, adhesion molecule signaling via integrin α4, and crosstalk between leukemia cells and the bone marrow microenvironment that is mediated through extracellular vesicles.
Collapse
Affiliation(s)
| | - Peter Kurre
- Doernbecher Children’s Hospital, Oregon Health and Science University, Portland, Oregon
| | - Yong-Mi Kim
- Children’s Hospital of Los Angeles, Keck School of Medicine of the University of Southern California, Los Angeles, California
| |
Collapse
|
81
|
The fibronectin III-1 domain activates a PI3-Kinase/Akt signaling pathway leading to αvβ5 integrin activation and TRAIL resistance in human lung cancer cells. BMC Cancer 2016; 16:574. [PMID: 27484721 PMCID: PMC4970220 DOI: 10.1186/s12885-016-2621-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 07/26/2016] [Indexed: 12/20/2022] Open
Abstract
Background Fibronectin is a mechanically sensitive protein which is organized in the extracellular matrix as a network of interacting fibrils. The lung tumor stroma is enriched for fibronectin which is thought to contribute to metastasis and drug resistance. Fibronectin is an elastic, multi-modular protein made up of individually folded domains, some of which can stretch in response to increased mechanical tension. Very little is known about the relationship of fibronectin’s unfolded domains to lung cancer resistance to chemotherapy. In the present study, we evaluated the impact of unfolding the first Type III domain of fibronectin (FnIII-1c) on TNF-related apoptosis inducing ligand (TRAIL) resistance. Methods NCI-H460 non-small cell lung cancer cells were treated with FnIII-1c then assessed for TRAIL-induced apoptosis. Subsequent analysis of FnIII-1c-mediated signaling pathways was also completed. Human non-small cell lung cancer tissue sections were assessed for the expression of vitronectin by immunohistochemistry. Results FnIII-1c inhibited TRAIL-induced activation of caspase 8 and subsequent apoptosis in NCI-H460 lung cancer cells. FnIII-1c treatment was associated with the activation of the phosphatidylinositol-3-kinase/alpha serine/threonine kinase (PI3K/Akt) pathway and the αvβ5 integrin receptor for vitronectin, both of which were required for TRAIL resistance. Immunohistochemical staining of sections from non-small cell lung cancers showed that vitronectin was localized around blood vessels and in the tumor-stroma interface. Conclusions Unfolding of Type III domains within the fibronectin matrix may promote TRAIL resistance through the activation of a PI3K/Akt/αvβ5 signaling axis and point to a novel mechanism by which changes in secondary structure of fibronectin contribute to cancer cell resistance to apoptosis.
Collapse
|
82
|
Myers JS, von Lersner AK, Sang QXA. Proteomic Upregulation of Fatty Acid Synthase and Fatty Acid Binding Protein 5 and Identification of Cancer- and Race-Specific Pathway Associations in Human Prostate Cancer Tissues. J Cancer 2016; 7:1452-64. [PMID: 27471561 PMCID: PMC4964129 DOI: 10.7150/jca.15860] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2016] [Accepted: 05/23/2016] [Indexed: 12/25/2022] Open
Abstract
Protein profiling studies of prostate cancer have been widely used to characterize molecular differences between diseased and non-diseased tissues. When combined with pathway analysis, profiling approaches are able to identify molecular mechanisms of prostate cancer, group patients by cancer subtype, and predict prognosis. This strategy can also be implemented to study prostate cancer in very specific populations, such as African Americans who have higher rates of prostate cancer incidence and mortality than other racial groups in the United States. In this study, age-, stage-, and Gleason score-matched prostate tumor specimen from African American and Caucasian American men, along with non-malignant adjacent prostate tissue from these same patients, were compared. Protein expression changes and altered pathway associations were identified in prostate cancer generally and in African American prostate cancer specifically. In comparing tumor to non-malignant samples, 45 proteins were significantly cancer-associated and 3 proteins were significantly downregulated in tumor samples. Notably, fatty acid synthase (FASN) and epidermal fatty acid-binding protein (FABP5) were upregulated in human prostate cancer tissues, consistent with their known functions in prostate cancer progression. Aldehyde dehydrogenase family 1 member A3 (ALDH1A3) was also upregulated in tumor samples. The Metastasis Associated Protein 3 (MTA3) pathway was significantly enriched in tumor samples compared to non-malignant samples. While the current experiment was unable to detect statistically significant differences in protein expression between African American and Caucasian American samples, differences in overrepresentation and pathway enrichment were found. Structural components (Cytoskeletal Proteins and Extracellular Matrix Protein protein classes, and Biological Adhesion Gene Ontology (GO) annotation) were overrepresented in African American but not Caucasian American tumors. Additionally, 5 pathways were enriched in African American prostate tumors: the Small Cell Lung Cancer, Platelet-Amyloid Precursor Protein, Agrin, Neuroactive Ligand-Receptor Interaction, and Intrinsic pathways. The protein components of these pathways were either basement membrane proteins or coagulation proteins.
Collapse
Affiliation(s)
- Jennifer S Myers
- 1. Department of Chemistry & Biochemistry, Florida State University, Tallahassee, FL, USA
| | - Ariana K von Lersner
- 1. Department of Chemistry & Biochemistry, Florida State University, Tallahassee, FL, USA
| | - Qing-Xiang Amy Sang
- 1. Department of Chemistry & Biochemistry, Florida State University, Tallahassee, FL, USA.; 2. Institute of Molecular Biophysics, Florida State University, Tallahassee, FL, USA
| |
Collapse
|
83
|
Liu CC, Leclair P, Monajemi M, Sly LM, Reid GS, Lim CJ. α-Integrin expression and function modulates presentation of cell surface calreticulin. Cell Death Dis 2016; 7:e2268. [PMID: 27310876 PMCID: PMC5143402 DOI: 10.1038/cddis.2016.176] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 05/10/2016] [Accepted: 05/26/2016] [Indexed: 12/22/2022]
Abstract
Calreticulin presentation on the cell surface is an important hallmark of immunogenic cell death (ICD), serving as the prophagocytic signal for macrophages. Cell adhesion is a physiologically relevant stimulus previously shown to increase calreticulin interaction with α-integrins via the juxtamembrane, cytosolic GFFKR motif. This study assessed whether integrin function can regulate surface calreticulin levels in ICD. We generated calreticulin-null T-lymphoblasts and confirmed the loss of surface calreticulin expression on cells treated with doxorubicin, an ICD inducer. Reconstituted expression with full-length calreticulin targeted to the endoplasmic reticulum (ER) successfully rescued doxorubicin-induced surface calreticulin. Reconstitution with a truncation mutant calreticulin targeted to the cytosol led to constitutively high surface calreticulin that was not further elevated by doxorubicin, suggesting calreticulin released from the stressed ER transits the cytosol before its translocation to the cell surface. When stimulated to engage integrin substrates, doxorubicin-treated wild-type T-lymphoblasts exhibited decreased surface calreticulin compared with cells under non-adherent conditions. The inhibitory effect on surface calreticulin was recapitulated for cells in suspension treated with a β1-integrin-activating antibody, 9EG7. Similarly, cells expressing a truncated α-integrin cytosolic tail, bearing only the juxtamembrane GFFKR calreticulin-binding motif, exhibited low surface calreticulin with doxorubicin treatment under non-adherent conditions. Using partial permeabilization techniques to distinguish between cytosolic and ER staining, we found that ICD inducers promoted the accumulation of cytosolic calreticulin with negligible change in total calreticulin, suggesting that integrin-mediated inhibition of surface calreticulin was due to reduced cytosolic to surface translocation. T-lymphoblasts co-treated with an ICD inducer and 9EG7 exhibited reduced phagocytosis by macrophages when compared with treatment with only ICD inducer. This study reveals a previously uncharacterized function of integrins as negative regulators of ICD by suppressing presentation of cell surface calreticulin.
Collapse
Affiliation(s)
- C-C Liu
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada V5Z 4H4.,Department of Medicine, University of British Columbia, Vancouver, BC, Canada V5Z 4H4
| | - P Leclair
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada V5Z 4H4
| | - M Monajemi
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada V5Z 4H4
| | - L M Sly
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada V5Z 4H4
| | - G S Reid
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada V5Z 4H4.,Michael Cuccione Childhood Cancer Research Program, BC Children's Hospital, Vancouver, BC, Canada V5Z 4H4
| | - C J Lim
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada V5Z 4H4.,Michael Cuccione Childhood Cancer Research Program, BC Children's Hospital, Vancouver, BC, Canada V5Z 4H4
| |
Collapse
|
84
|
Huang Y, Yang X, Xu T, Kong Q, Zhang Y, Shen Y, Wei Y, Wang G, Chang KJ. Overcoming resistance to TRAIL-induced apoptosis in solid tumor cells by simultaneously targeting death receptors, c-FLIP and IAPs. Int J Oncol 2016; 49:153-63. [PMID: 27210546 PMCID: PMC4902065 DOI: 10.3892/ijo.2016.3525] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 04/15/2016] [Indexed: 12/12/2022] Open
Abstract
The discovery of the TRAIL protein and its death receptors DR4/5 changed the horizon of cancer research because TRAIL specifically kills cancer cells. However, the validity of TRAIL-based cancer therapies has yet to be established, as most cancer cells are TRAIL-resistant. In this report, we demonstrate that TRAIL-resistance of many cancer cell lines can be overcome after siRNA- or rocaglamide-mediated downregulation of c-FLIP expression and simultaneous inhibition of IAPs activity using AT406, a pan-antagonist of IAPs. Combined triple actions of the TRAIL, the IAPs inhibitor, AT406, and the c-FLIP expression inhibitor, rocaglamide (ART), markedly improve TRAIL-induced apoptotic effects in most solid cancer cell lines through the activation of an extrinsic apoptosis pathway. Furthermore, this ART combination does not harm normal cells. Among the 18 TRAIL-resistant cancer cell lines used, 15 cell lines become sensitive or highly sensitive to ART, and two out of three glioma cell lines exhibit high resistance to ART treatment due to very low levels of procaspase-8. This study provides a rationale for the development of TRAIL-induced apoptosis-based cancer therapies.
Collapse
Affiliation(s)
- Ying Huang
- Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming, Yunnan 650500, P.R. China
| | - Xiang Yang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650500, P.R. China
| | - Tianrui Xu
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650500, P.R. China
| | - Qinghong Kong
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650500, P.R. China
| | - Yaping Zhang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650500, P.R. China
| | - Yuehai Shen
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650500, P.R. China
| | - Yunlin Wei
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650500, P.R. China
| | - Guanlin Wang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650500, P.R. China
| | - Kwen-Jen Chang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650500, P.R. China
| |
Collapse
|
85
|
Mohseni M, Samadi N, Ghanbari P, Yousefi B, Tabasinezhad M, Sharifi S, Nazemiyeh H. Co-treatment by docetaxel and vinblastine breaks down P-glycoprotein mediated chemo-resistance. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2016; 19:300-9. [PMID: 27114800 PMCID: PMC4834120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
OBJECTIVES Chemoresistance remains the main causes of treatment failure and mortality in cancer patients. There is an urgent need to investigate novel approaches to improve current therapeutic modalities and increase cancer patients' survival. Induction of drug efflux due to overexpression of P-glycoproteins is considered as an important leading cause of multidrug resistance. In this study, we investigated the role of combination treatments of docetaxel and vinblastine in overcoming P-glycoprotein mediated inhibition of apoptosis and induction of cell proliferation in human non-small cell lung carcinoma cells. MATERIALS AND METHODS Cell proliferation and apoptosis were assessed using MTT assay and DAPI staining, respectively. P-glycoprotein expression was evaluated in gene and protein levels by Real-time RT-PCR and Western blot analysis, respectively. RESULTS Combination treatment of the cells with docetaxel and vinblastine decreased the IC50 values for docetaxel from (30±3.1) to (15±2.6) nM and for vinblastine from (30±5.9) to (5±5.6) nM (P≤0.05). P-glycoprotein mRNA expression level showed a significant up-regulation in the cells incubated with each drug alone (P≤0.001). Incubation of the cells with combined concentrations of both agents neutralized P-glycoprotein overexpression (P≤0.05). Adding verapamil, a P-glycoprotein inhibitor caused a further increase in the percentage of apoptotic cells when the cells were treated with both agents. CONCLUSION Our results suggest that combination therapy along with P-glycoprotein inhibition can be considered as a novel approach to improve the efficacy of chemotherapeutics in cancer patients with high P-glycoprotein expression.
Collapse
Affiliation(s)
- Mahsa Mohseni
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran,Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nasser Samadi
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran,Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran,Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran,Corresponding author: Nasser Samadi, Department of Medical Biotechnology, Faculty of Advanced Medical Sciences and Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran. Tel: +98-413-3355789; Fax: +98-413-3355789;
| | - Parisa Ghanbari
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran,Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Bahman Yousefi
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran,Proteomics laboratory, Department of Clinical Biochemistry, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Tabasinezhad
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran,Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Simin Sharifi
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran,Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Nazemiyeh
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran,Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
86
|
Discovery and characterization of a high-affinity and high-specificity peptide ligand LXY30 for in vivo targeting of α3 integrin-expressing human tumors. EJNMMI Res 2016; 6:18. [PMID: 26922417 PMCID: PMC4769701 DOI: 10.1186/s13550-016-0165-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 01/19/2016] [Indexed: 12/20/2022] Open
Abstract
Background α3β1 integrin is overexpressed in several types of human cancer and is associated with poor prognosis, metastasis, and resistance to cancer treatment. We previously identified a cyclic peptide ligand LXY1 that specifically binds to the α3β1 integrin on human glioblastoma U-87MG cells. Here, we optimized LXY1 through one-bead one-compound combinatorial library screening and site-specific modifications to improve its in vivo binding property. Methods Three bead libraries were synthesized and whole-cell binding assays were performed. The binding capacity of individual peptide ligands against different tumor cells was determined by flow cytometry and confirmed by optical imaging. A complex joining biotinylated ligand with streptavidin-Cy5.5 was used for in vivo target imaging in both subcutaneous and orthotopic U-87MG xenograft mouse models. Results LXY30, a cyclic peptide with the sequence cdG-Phe(3,5-diF)-G-Hyp-NcR, emerged as the most potent and selective ligand for the α3 subunit of α3β1 integrin with improved in vitro and in vivo tumor-targeting effects compared to LXY1 in U-87MG cells. LXY30 is considerably stable in plasma as demonstrated in an in vitro stability study in 90 % human plasma. LXY30 also binds to several other known α3β1 integrin-expressing glioblastoma, lung, and breast cancer cell lines with various affinities. Conclusions Our data support further investigating the role of LXY30 as a human tumor-targeting peptide ligand for systemic and intracranial delivery of imaging agents and cancer therapeutics. Electronic supplementary material The online version of this article (doi:10.1186/s13550-016-0165-z) contains supplementary material, which is available to authorized users.
Collapse
|
87
|
Yuan Y, Wu L, Shen S, Wu S, Burdick MM. Effect of alpha 2,6 sialylation on integrin-mediated adhesion of breast cancer cells to fibronectin and collagen IV. Life Sci 2016; 149:138-45. [PMID: 26903292 DOI: 10.1016/j.lfs.2016.02.071] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 02/17/2016] [Accepted: 02/18/2016] [Indexed: 02/06/2023]
Abstract
AIMS To determine the role of sialylation on α5β1 and α2β1 integrins in the regulation of adhesion between breast cancer cells and extracellular matrix (ECM). MAIN METHODS Static cell adhesion assays were performed to quantify avidity of breast cancer cells to ECM. The effects of sialidases on α2,6 sialylation was assessed by flow cytometry using biotin conjugated Sambucus nigra lectin. Lectin affinity assays were used to determine expression of α2,6 sialylated integrins. Cell migration and invasion were investigated by wound healing and transwell invasion assays. KEY FINDINGS α2, α5 and β1 integrins had considerable α2,6 sialylation on MDA-MB-231 cells, whereas signals from MCF-7 cells were undetectable. Cleavage of α2,6 sialylation increased adhesion of MDA-MB-231 cells to ECM, while adhesion of MCF-7 cells was unaffected, consistent with the latter's lack of endogenous α2,6 sialylated surface integrins. Neither surface expression of α2β1 and α5β1 integrins, nor activated β1 integrin, changed in MDA-MB-231 cells after sialidase treatment. However, sialidase treatment did not have significant impact on migration or invasion of MDA-MB-231 cells. SIGNIFICANCE Cell adhesion is an important early step of cancer metastasis, yet the roles of sialylation in regulating integrin-mediated breast cancer cell adhesion in comparison to migration and invasion are not well-understood. Our data suggest desialylation of α2,6-sialylated integrins increases adhesion, but not migration or invasion, of MDA-MB-231 cells to ECM without altering integrin expression. It should be considered that α2,6 sialylation may play different roles in regulating cell adhesion of different cancer cells when developing potential therapeutics targeting α2,6 sialylation.
Collapse
Affiliation(s)
- Ye Yuan
- Edison Biotechnology Institute, Ohio University, Athens, OH, United States; Department of Chemistry and Biochemistry, Ohio University, Athens, OH, United States
| | - Larry Wu
- Department of Chemical and Biomolecular Engineering, Ohio University, Athens, OH, United States
| | - Siqi Shen
- College of Life Sciences, Capital Normal University, Beijing, China
| | - Shiyong Wu
- Edison Biotechnology Institute, Ohio University, Athens, OH, United States; Department of Chemistry and Biochemistry, Ohio University, Athens, OH, United States.
| | - Monica M Burdick
- Department of Chemical and Biomolecular Engineering, Ohio University, Athens, OH, United States.
| |
Collapse
|
88
|
Blandin AF, Renner G, Lehmann M, Lelong-Rebel I, Martin S, Dontenwill M. β1 Integrins as Therapeutic Targets to Disrupt Hallmarks of Cancer. Front Pharmacol 2015; 6:279. [PMID: 26635609 PMCID: PMC4656837 DOI: 10.3389/fphar.2015.00279] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 11/05/2015] [Indexed: 01/11/2023] Open
Abstract
Integrins belong to a large family of αβ heterodimeric transmembrane proteins first recognized as adhesion molecules that bind to dedicated elements of the extracellular matrix and also to other surrounding cells. As important sensors of the cell microenvironment, they regulate numerous signaling pathways in response to structural variations of the extracellular matrix. Biochemical and biomechanical cues provided by this matrix and transmitted to cells via integrins are critically modified in tumoral settings. Integrins repertoire are subjected to expression level modifications, in tumor cells, and in surrounding cancer-associated cells, implicated in tumor initiation and progression as well. As critical players in numerous cancer hallmarks, defined by Hanahan and Weinberg (2011), integrins represent pertinent therapeutic targets. We will briefly summarize here our current knowledge about integrin implications in those different hallmarks focusing primarily on β1 integrins.
Collapse
Affiliation(s)
- Anne-Florence Blandin
- Department "Tumoral Signaling and Therapeutic Targets," Faculty of Pharmacy, UMR7213 Centre National de la Recherche Scientifique, University of Strasbourg Illkirch, France
| | - Guillaume Renner
- Department "Tumoral Signaling and Therapeutic Targets," Faculty of Pharmacy, UMR7213 Centre National de la Recherche Scientifique, University of Strasbourg Illkirch, France
| | - Maxime Lehmann
- Department "Tumoral Signaling and Therapeutic Targets," Faculty of Pharmacy, UMR7213 Centre National de la Recherche Scientifique, University of Strasbourg Illkirch, France
| | - Isabelle Lelong-Rebel
- Department "Tumoral Signaling and Therapeutic Targets," Faculty of Pharmacy, UMR7213 Centre National de la Recherche Scientifique, University of Strasbourg Illkirch, France
| | - Sophie Martin
- Department "Tumoral Signaling and Therapeutic Targets," Faculty of Pharmacy, UMR7213 Centre National de la Recherche Scientifique, University of Strasbourg Illkirch, France
| | - Monique Dontenwill
- Department "Tumoral Signaling and Therapeutic Targets," Faculty of Pharmacy, UMR7213 Centre National de la Recherche Scientifique, University of Strasbourg Illkirch, France
| |
Collapse
|
89
|
Wang M, Topalovski M, Toombs JE, Wright CM, Moore ZR, Boothman DA, Yanagisawa H, Wang H, Witkiewicz A, Castrillon DH, Brekken RA. Fibulin-5 Blocks Microenvironmental ROS in Pancreatic Cancer. Cancer Res 2015; 75:5058-69. [PMID: 26577699 DOI: 10.1158/0008-5472.can-15-0744] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 08/15/2015] [Indexed: 12/16/2022]
Abstract
Elevated oxidative stress is an aberration seen in many solid tumors, and exploiting this biochemical difference has the potential to enhance the efficacy of anticancer agents. Homeostasis of reactive oxygen species (ROS) is important for normal cell function, but excessive production of ROS can result in cellular toxicity, and therefore ROS levels must be balanced finely. Here, we highlight the relationship between the extracellular matrix and ROS production by reporting a novel function of the matricellular protein Fibulin-5 (Fbln5). We used genetically engineered mouse models of pancreatic ductal adenocarcinoma (PDAC) and found that mutation of the integrin-binding domain of Fbln5 led to decreased tumor growth, increased survival, and enhanced chemoresponse to standard PDAC therapies. Through mechanistic investigations, we found that improved survival was due to increased levels of oxidative stress in Fbln5-mutant tumors. Furthermore, loss of the Fbln5-integrin interaction augmented fibronectin signaling, driving integrin-induced ROS production in a 5-lipooxygenase-dependent manner. These data indicate that Fbln5 promotes PDAC progression by functioning as a molecular rheostat that modulates cell-ECM interactions to reduce ROS production, and thus tip the balance in favor of tumor cell survival and treatment-refractory disease.
Collapse
Affiliation(s)
- Miao Wang
- Hamon Center for Therapeutic Oncology Research, UT Southwestern Medical Center, Dallas, Texas
| | - Mary Topalovski
- Hamon Center for Therapeutic Oncology Research, UT Southwestern Medical Center, Dallas, Texas
| | - Jason E Toombs
- Hamon Center for Therapeutic Oncology Research, UT Southwestern Medical Center, Dallas, Texas
| | - Christopher M Wright
- Hamon Center for Therapeutic Oncology Research, UT Southwestern Medical Center, Dallas, Texas
| | - Zachary R Moore
- Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, Texas
| | - David A Boothman
- Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, Texas
| | - Hiromi Yanagisawa
- Department of Molecular Biology, UT Southwestern Medical Center, Dallas, Texas
| | - Huamin Wang
- Department of Pathology, UT MD Anderson Cancer Center, Houston, Texas
| | | | | | - Rolf A Brekken
- Hamon Center for Therapeutic Oncology Research, UT Southwestern Medical Center, Dallas, Texas. Department of Surgery, UT Southwestern Medical Center, Dallas, Texas. Department of Pharmacology, UT Southwestern Medical Center, Dallas, Texas.
| |
Collapse
|
90
|
Zustiak SP, Dadhwal S, Medina C, Steczina S, Chehreghanianzabi Y, Ashraf A, Asuri P. Three-dimensional matrix stiffness and adhesive ligands affect cancer cell response to toxins. Biotechnol Bioeng 2015; 113:443-52. [DOI: 10.1002/bit.25709] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 06/15/2015] [Accepted: 07/08/2015] [Indexed: 02/06/2023]
Affiliation(s)
| | - Smritee Dadhwal
- Department of Bioengineering; Santa Clara University; Santa Clara California
| | - Carlos Medina
- Department of Bioengineering; Santa Clara University; Santa Clara California
| | - Sonette Steczina
- Department of Bioengineering; Santa Clara University; Santa Clara California
| | | | - Anisa Ashraf
- Department of Biomedical Engineering; Saint Louis University; St. Louis Missouri
| | - Prashanth Asuri
- Department of Bioengineering; Santa Clara University; Santa Clara California
| |
Collapse
|
91
|
Wang W, Chen P, Tang M, Li J, Pei Y, Cai S, Zhou X, Chen S. Tumstatin 185-191 increases the sensitivity of non-small cell lung carcinoma cells to cisplatin by blocking proliferation, promoting apoptosis and inhibiting Akt activation. Am J Transl Res 2015; 7:1332-1344. [PMID: 26396665 PMCID: PMC4568790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2015] [Accepted: 07/31/2015] [Indexed: 06/05/2023]
Abstract
PURPOSE This study aimed to investigate the synergistic anti-tumor effects of tumstatin 185-191 and cisplatin in non-small cell lung carcinoma cells (NSCLC) (A549 cells and cisplatin resistant A549/DDP cells), and the potential role of Akt signaling pathway was also explored. METHODS A549 or A549/DDP cells were treated with Tum185-191 or Tum185-191 plus cisplatin. Cell viability was assessed by modified MTT assay. 50% inhibiting concentration (IC50) and reversing drug-resistance index (RI) of chemotherapeutics were determined by MTT assay. Cell apoptosis was measured by Hoechst 33258 staining and flow cytometry. The activation of Akt signaling pathway was evaluated by immunocytochemistry and Western blot assay. RESULTS Tum185-191 inhibited the proliferation of A549 cells and A549/DDP cells. In the presence of Tum185-191 (20 and 40 μM), IC50 of cisplatin reduced significantly in A549 cells and A549/DDP cells. Combined use of tumstatin 185-191 and cisplatin exerted synergistic effects in promoting apoptosis. A549 and A549/DDP cells had a high expression of p-Akt, and Tum185-191, but not cisplatin, significantly inhibited p-Akt expression. Combined use of cisplatin and Tum185-191 failed to further inhibit p-Akt expression. After Tum185-191 treatment, the increased p-Akt expression was observed at 15 min, peaked at 30-60 min, but disappeared at 120 min. CONCLUSION Tum185-191 increases the apoptosis, inhibit the proliferation, enhance the sensitivity of A549 cells to cisplatin and also partly reverse the resistance of A549-DDP cells to cisplatin, which is at least partially mediated by inactivating Akt pathway. These findings provide evidence for the chemotherapy of NSCLC with Tum185-191 and cisplatin.
Collapse
Affiliation(s)
- Wei Wang
- Department of Thoracic Medicine, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South UniversityChangsha 410013, China
| | - Ping Chen
- Department of Respiratory Medicine, The Second Xiangya Hospital, Central South UniversityChangsha 410011, China
| | - Min Tang
- Department of Thoracic Medicine, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South UniversityChangsha 410013, China
| | - Junli Li
- Department of Respiratory Medicine, The Second Xiangya Hospital, Central South UniversityChangsha 410011, China
| | - Yanfang Pei
- Department of Respiratory Medicine, The Second Xiangya Hospital, Central South UniversityChangsha 410011, China
| | - Shan Cai
- Department of Respiratory Medicine, The Second Xiangya Hospital, Central South UniversityChangsha 410011, China
| | - Xiao Zhou
- Department of Oncology Plastic Surgery, Department of Head and Neck Surgery, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South UniversityChangsha 410013, China
| | - Senlin Chen
- Department of Pathology, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South UniversityChangsha 410013, China
| |
Collapse
|
92
|
Alfarouk KO, Stock CM, Taylor S, Walsh M, Muddathir AK, Verduzco D, Bashir AHH, Mohammed OY, Elhassan GO, Harguindey S, Reshkin SJ, Ibrahim ME, Rauch C. Resistance to cancer chemotherapy: failure in drug response from ADME to P-gp. Cancer Cell Int 2015; 15:71. [PMID: 26180516 PMCID: PMC4502609 DOI: 10.1186/s12935-015-0221-1] [Citation(s) in RCA: 420] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 06/30/2015] [Indexed: 12/15/2022] Open
Abstract
Cancer chemotherapy resistance (MDR) is the innate and/or acquired ability of cancer cells to evade the effects of chemotherapeutics and is one of the most pressing major dilemmas in cancer therapy. Chemotherapy resistance can arise due to several host or tumor-related factors. However, most current research is focused on tumor-specific factors and specifically genes that handle expression of pumps that efflux accumulated drugs inside malignantly transformed types of cells. In this work, we suggest a wider and alternative perspective that sets the stage for a future platform in modifying drug resistance with respect to the treatment of cancer.
Collapse
Affiliation(s)
- Khalid O Alfarouk
- Institute of Endemic Diseases, University of Khartoum, Khartoum, Sudan
| | | | - Sophie Taylor
- School of Veterinary Medicine and Science, University of Nottingham, Nottingham, UK
| | - Megan Walsh
- School of Veterinary Medicine and Science, University of Nottingham, Nottingham, UK
| | | | | | - Adil H H Bashir
- Institute of Endemic Diseases, University of Khartoum, Khartoum, Sudan
| | | | - Gamal O Elhassan
- Uneizah Pharmacy College, Qassim University, AL-Qassim, Kingdom of Saudi Arabia ; Faculty of Pharmacy, Omdurman Islamic University, Khartoum, Sudan
| | | | - Stephan J Reshkin
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy
| | | | - Cyril Rauch
- School of Veterinary Medicine and Science, University of Nottingham, Nottingham, UK
| |
Collapse
|
93
|
Matsuzaki S, Darcha C. Co-operation between the AKT and ERK signaling pathways may support growth of deep endometriosis in a fibrotic microenvironment in vitro. Hum Reprod 2015; 30:1606-16. [PMID: 25976656 DOI: 10.1093/humrep/dev108] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 04/22/2015] [Indexed: 12/13/2022] Open
Abstract
STUDY QUESTION How can deep endometriotic stromal cells proliferate and persist in a fibrotic environment? SUMMARY ANSWER The serine/threonine kinase AKT and extracellular regulated kinase (ERK) signaling pathways may co-operate to support growth of deep endometriotic lesions by enhancing endometriotic stromal cell proliferation and survival in a fibrotic microenvironment in vitro. WHAT IS KNOWN ALREADY Endometriosis, particularly deep infiltrating endometriosis, is characterized histologically by dense fibrous tissue that is primarily composed of type I collagen. This tissue may cause pelvic pain and infertility, which are major clinical issues associated with endometriosis. Proliferation of normal fibroblasts is tightly regulated, and fibrillar, polymerized type I collagen inhibits normal fibroblast proliferation. However, no studies to date have investigated how deep endometriotic stromal cells can proliferate and persist in a fibrotic environment. STUDY DESIGN, SIZE, DURATION Endometrial and/or endometriotic tissues from 104 patients (61 with and 43 without endometriosis) of reproductive age with normal menstrual cycles were analyzed. A total of 25 nude mice received a single injection of endometrial fragments from a total of five samples. PARTICIPANTS/MATERIALS, SETTING, METHODS We evaluated cell proliferation, caspase 3/7 activity, and the AKT and ERK signaling pathways in endometrial and endometriotic stromal cells on three-dimensional (3D) polymerized collagen matrices in vitro. In addition, to determine whether aberrant activation of the AKT and ERK pathways is involved during progression of fibrosis in endometriosis in vivo, we evaluated the expression of phosphorylated AKT and ERK1/2 in endometriotic implants in a nude mouse model of endometriosis. Finally, we evaluated the effects of MK2206 (an AKT inhibitor) and U0126 (a MEK inhibitor) on cell proliferation, caspase 3/7 activity, and phosphorylation of AKT and ERK1/2 of endometriotic stromal cells on 3D polymerized collagen matrices. MAIN RESULTS AND THE ROLE OF CHANCE Proliferation of endometriotic stromal cells was significantly less inhibited than that of endometrial stromal cells (P < 0.05) on 3D polymerized collagen. Levels of phosphorylated AKT, phosphorylated p70S6K and phosphorylated ERK1/2 were significantly higher in endometriotic stromal cells than in endometrial stromal cells at 24 h (P < 0.05) and at 72 h (P < 0.05) on 3D polymerized collagen. Phosphorylated AKT expression was significantly increased on Days 21 and 28 compared with those on Days 3 and 7 (all P < 0.05) in endometriotic implants during progression of fibrosis in a nude mouse model of endometriosis. Inhibition of AKT or ERK1/2 with MK2206 or U0126, respectively, did not significantly increase caspase 3/7 activity in endometriotic stromal cells on either two-dimensional or 3D collagen matrices. Western blot analysis showed that MK2206 alone decreased levels of phosphorylated AKT; however, it increased levels of phosphorylated ERK in endometriotic cells compared with vehicle-treated cells (both P < 0.05). In addition, U0126 treatment decreased levels of phosphorylated ERK; however, it resulted in increased levels of phosphorylated AKT in endometriotic stromal cells compared with vehicle-treated cells (both P < 0.05). LIMITATIONS, REASONS FOR CAUTION Endometriosis involves a number of processes, such as invasion, metastasis, angiogenesis, and apoptosis resistance, and a variety of signaling pathways may be involved in promoting development and progression of the disease. In addition, further animal experiments are required to determine whether the AKT and ERK signaling pathways co-operate to support growth of endometriotic lesions in a fibrotic microenvironment in vivo. WIDER IMPLICATIONS OF THE FINDINGS Co-targeting the AKT and ERK pathways may be an effective therapeutic strategy for endometriosis treatment. STUDY FUNDING/COMPETING INTERESTS This study was supported in part by Karl Storz Endoscopy & GmbH (Tuttlingen, Germany). No competing interests are declared.
Collapse
Affiliation(s)
- Sachiko Matsuzaki
- CHU Clermont-Ferrand, CHU Estaing, Chirurgie Gynécologique, Clermont-Ferrand, France Clermont Université, Université d'Auvergne, ISIT UMR6284, Clermont-Ferrand, France CNRS, ISIT UMR6284, Clermont-Ferrand, France
| | - Claude Darcha
- CHU Clermont-Ferrand, Service d'Anatomie et Cytologie Pathologiques, Clermont-Ferrand, France
| |
Collapse
|
94
|
Deep G, Kumar R, Jain AK, Agarwal C, Agarwal R. Silibinin inhibits fibronectin induced motility, invasiveness and survival in human prostate carcinoma PC3 cells via targeting integrin signaling. Mutat Res 2015; 768:35-46. [PMID: 25285031 DOI: 10.1016/j.mrfmmm.2014.05.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Prostate cancer (PCA) is the 2nd leading cause of cancer-related deaths among men in the United States. Preventing or inhibiting metastasis-related events through non-toxic agents could be a useful approach for lowering high mortality among PCA patients. We have earlier reported that natural flavonoid silibinin possesses strong anti-metastatic efficacy against PCA however, mechanism/s of its action still remains largely unknown. One of the major events during metastasis is the replacement of cell-cell interaction with integrins-based cell-matrix interaction that controls motility, invasiveness and survival of cancer cells. Accordingly, here we examined silibinin effect on advanced human PCA PC3 cells' interaction with extracellular matrix component fibronectin. Silibinin (50-200 μM) treatment significantly decreased the fibronectin (5 μg/ml)-induced motile morphology via targeting actin cytoskeleton organization in PC3 cells. Silibinin also decreased the fibronectin-induced cell proliferation and motility but significantly increased cell death in PC3 cells. Silibinin also inhibited the PC3 cells invasiveness in Transwell invasion assays with fibronectin or cancer associated fibroblasts (CAFs) serving as chemoattractant. Importantly, PC3-luc cells cultured on fibronectin showed rapid dissemination and localized in lungs following tail vein injection in athymic male nude mice; however, in silibinin-treated PC3-luc cells, dissemination and lung localization was largely compromised. Molecular analyses revealed that silibinin treatment modulated the fibronectin-induced expression of integrins (α5, αV, β1 and β3), actin-remodeling (FAK, Src, GTPases, ARP2 and cortactin), apoptosis (cPARP and cleaved caspase 3), EMT (E-cadherin and β-catenin), and cell survival (survivin and Akt) related signaling molecules in PC3 cells. Furthermore, PC3-xenograft tissue analyses confirmed the inhibitory effect of silibinin on fibronectin and integrins expression. Together, these results showed that silibinin targets PCA cells' interaction with fibronectin and inhibits their motility, invasiveness and survival; thus further supporting silibinin use in PCA intervention including its metastatic progression.
Collapse
|
95
|
Hu Y, Chen HY, Yu CY, Xu J, Wang JL, Qian J, Zhang X, Fang JY. A long non-coding RNA signature to improve prognosis prediction of colorectal cancer. Oncotarget 2015; 5:2230-42. [PMID: 24809982 PMCID: PMC4039159 DOI: 10.18632/oncotarget.1895] [Citation(s) in RCA: 136] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Increasing evidence suggests long non-coding RNAs (lncRNAs) are frequently aberrantly expressed in cancers, however, few related lncRNA signatures have been established for prediction of cancer prognosis. We aimed to develop a lncRNA signature to improve prognosis prediction of colorectal cancer (CRC). Using a lncRNA-mining approach, we performed lncRNA expression profiling in large CRC cohorts from Gene Expression Ominus (GEO), including GSE39582 test series(N=436), internal validation series (N=117); and two independent validation series GSE14333 (N=197) and GSE17536(N=145). We established a set of six lncRNAs that were significantly correlated with the disease free survival (DFS) in the test series. Based on this six-lncRNA signature, the test series patients could be classified into high-risk and low-risk subgroups with significantly different DFS (HR=2.670; P<0.0001). The prognostic value of this six-lncRNA signature was confirmed in the internal validation series and another two independent CRC sets. Gene set enrichment analysis (GSEA) analysis suggested that risk score positively correlated with several cancer metastasis related pathways. Functional experiments demonstrated three dysregulated lncRNAs, AK123657, BX648207 and BX649059 were required for efficient invasion and proliferation suppression in CRC cell lines. Our results might provide an efficient classification tool for clinical prognosis evaluation of CRC.
Collapse
Affiliation(s)
- Ye Hu
- Division of Gastroenterology and Hepatology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institution of Digestive Disease; Key Laboratory of Gastroenterology and Hepatology, Ministry of Health; State Key Laboratory of Oncogene and Related Genes., Shanghai, China
| | | | | | | | | | | | | | | |
Collapse
|
96
|
Valcz G, Sipos F, Tulassay Z, Molnar B, Yagi Y. Importance of carcinoma-associated fibroblast-derived proteins in clinical oncology. J Clin Pathol 2014; 67:1026-1031. [PMID: 25135950 DOI: 10.1136/jclinpath-2014-202561] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Carcinoma-associated fibroblast (CAF) as prominent cell type of the tumour microenvironment has complex interaction with both the cancer cells and other non-neoplastic surrounding cells. The CAF-derived regulators and extracellular matrix proteins can support cancer progression by providing a protective microenvironment for the cancer cells via reduction of chemotherapy sensitivity. On the other hand, these proteins may act as powerful prognostic markers as well as potential targets of anticancer therapy. In this review, we summarise the clinical importance of the major CAF-derived signals influencing tumour behaviour and determining the outcome of chemotherapy.
Collapse
Affiliation(s)
- Gabor Valcz
- Department of Pathology, Harvard Medical School, Boston, Massachusetts, USA
| | - Ferenc Sipos
- 2nd Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| | - Zsolt Tulassay
- Molecular Medicine Research Unit, Hungarian Academy of Sciences, Budapest, Hungary
| | - Bela Molnar
- Molecular Medicine Research Unit, Hungarian Academy of Sciences, Budapest, Hungary
| | - Yukako Yagi
- Department of Pathology, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
97
|
Somatic mutations favorable to patient survival are predominant in ovarian carcinomas. PLoS One 2014; 9:e112561. [PMID: 25390899 PMCID: PMC4229214 DOI: 10.1371/journal.pone.0112561] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Accepted: 10/09/2014] [Indexed: 11/19/2022] Open
Abstract
Somatic mutation accumulation is a major cause of abnormal cell growth. However, some mutations in cancer cells may be deleterious to the survival and proliferation of the cancer cells, thus offering a protective effect to the patients. We investigated this hypothesis via a unique analysis of the clinical and somatic mutation datasets of ovarian carcinomas published by the Cancer Genome Atlas. We defined and screened 562 macro mutation signatures (MMSs) for their associations with the overall survival of 320 ovarian cancer patients. Each MMS measures the number of mutations present on the member genes (except for TP53) covered by a specific Gene Ontology (GO) term in each tumor. We found that somatic mutations favorable to the patient survival are predominant in ovarian carcinomas compared to those indicating poor clinical outcomes. Specially, we identified 19 (3) predictive MMSs that are, usually by a nonlinear dose-dependent effect, associated with good (poor) patient survival. The false discovery rate for the 19 "positive" predictors is at the level of 0.15. The GO terms corresponding to these MMSs include "lysosomal membrane" and "response to hypoxia", each of which is relevant to the progression and therapy of cancer. Using these MMSs as features, we established a classification tree model which can effectively partition the training samples into three prognosis groups regarding the survival time. We validated this model on an independent dataset of the same disease (Log-rank p-value < 2.3 × 10(-4)) and a dataset of breast cancer (Log-rank p-value < 9.3 × 10(-3)). We compared the GO terms corresponding to these MMSs and those enriched with expression-based predictive genes. The analysis showed that the GO term pairs with large similarity are mainly pertinent to the proteins located on the cell organelles responsible for material transport and waste disposal, suggesting the crucial role of these proteins in cancer mortality.
Collapse
|
98
|
Ghanbari P, Mohseni M, Tabasinezhad M, Yousefi B, Saei AA, Sharifi S, Rashidi MR, Samadi N. Inhibition of survivin restores the sensitivity of breast cancer cells to docetaxel and vinblastine. Appl Biochem Biotechnol 2014; 174:667-81. [PMID: 25086926 DOI: 10.1007/s12010-014-1125-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Accepted: 07/23/2014] [Indexed: 12/19/2022]
Abstract
Combination therapy is considered a viable strategy to overcome the resistance to chemotherapeutics. Survivin as a member of the inhibitor of apoptosis protein (IAP) family, which is involved in resistance to various drugs. We investigated the role of combination therapy in downregulating survivin and increasing drug's efficacy in MDA-MB-231 cells. MTT assay and DAPI staining were applied to study the anti-proliferative activity and apoptosis response of the agents. Real-time RT-PCR and Western blot analysis were applied to study survivin mRNA and protein. Our findings showed that combined treatment of cells with docetaxel and vinblastine reduces survivin expression and consequently decreases the IC50 value of docetaxel from 70 to 5 nM (p < 0.05). Furthermore, combination therapy with deguelin, a survivin inhibitor, exerted a considerable enhancement in synergistic efficacy of docetaxel and vinblastine (p < 0.05). Survivin downregulation may thus be considered a potential strategy in increasing the efficacy of chemotherapeutics in cancer patients.
Collapse
Affiliation(s)
- Parisa Ghanbari
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran,
| | | | | | | | | | | | | | | |
Collapse
|
99
|
Dvorkin-Gheva A, Hassell JA. Identification of a novel luminal molecular subtype of breast cancer. PLoS One 2014; 9:e103514. [PMID: 25076125 PMCID: PMC4116208 DOI: 10.1371/journal.pone.0103514] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Accepted: 07/03/2014] [Indexed: 12/31/2022] Open
Abstract
The molecular classification of human breast tumors has afforded insights into subtype specific biological processes, patient prognosis and response to therapies. However, using current methods roughly one quarter of breast tumors cannot be classified into one or another molecular subtype. To explore the possibility that the unclassifiable samples might comprise one or more novel subtypes we employed a collection of publically available breast tumor datasets with accompanying clinical information to assemble 1,593 transcript profiles: 25% of these samples could not be assigned to one of the current molecular subtypes of breast cancer. All of the unclassifiable samples could be grouped into a new molecular subtype, which we termed “luminal-like”. We also identified the luminal-like subtype in an independent collection of tumor samples (NKI295). We found that patients harboring tumors of the luminal-like subtype have a better prognosis than those with basal-like breast cancer, a similar prognosis to those with ERBB2+, luminal B or claudin-low tumors, but a worse prognosis than patients with luminal A or normal-like breast tumors. Our findings suggest the occurrence of another molecular subtype of breast cancer that accounts for the vast majority of previously unclassifiable breast tumors.
Collapse
Affiliation(s)
- Anna Dvorkin-Gheva
- Centre for Functional Genomics, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - John A. Hassell
- Centre for Functional Genomics, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- * E-mail:
| |
Collapse
|
100
|
Martinez-Serra J, Gutierrez A, Muñoz-Capó S, Navarro-Palou M, Ros T, Amat JC, Lopez B, Marcus TF, Fueyo L, Suquia AG, Gines J, Rubio F, Ramos R, Besalduch J. xCELLigence system for real-time label-free monitoring of growth and viability of cell lines from hematological malignancies. Onco Targets Ther 2014; 7:985-94. [PMID: 24959085 PMCID: PMC4061162 DOI: 10.2147/ott.s62887] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The xCELLigence system is a new technological approach that allows the real-time cell analysis of adherent tumor cells. To date, xCELLigence has not been able to monitor the growth or cytotoxicity of nonadherent cells derived from hematological malignancies. The basis of its technology relies on the use of culture plates with gold microelectrodes located in their base. We have adapted the methodology described by others to xCELLigence, based on the pre-coating of the cell culture surface with specific substrates, some of which are known to facilitate cell adhesion in the extracellular matrix. Pre-coating of the culture plates with fibronectin, compared to laminin, collagen, or gelatin, significantly induced the adhesion of most of the leukemia/lymphoma cells assayed (Jurkat, L1236, KMH2, and K562). With a fibronectin substrate, nonadherent cells deposited in a monolayer configuration, and consequently, the cell growth and viability were robustly monitored. We further demonstrate the feasibility of xCELLigence for the real-time monitoring of the cytotoxic properties of several antineoplastic agents. In order to validate this technology, the data obtained through real-time cell analysis was compared with that obtained from using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide method. This provides an excellent label-free tool for the screening of drug efficacy in nonadherent cells and discriminates optimal time points for further molecular analysis of cellular events associated with treatments, reducing both time and costs.
Collapse
Affiliation(s)
- Jordi Martinez-Serra
- Department of Hematology, University Hospital Son Espases, Palma de Mallorca, Balearic Islands, Spain
| | - Antonio Gutierrez
- Department of Hematology, University Hospital Son Espases, Palma de Mallorca, Balearic Islands, Spain
| | - Saúl Muñoz-Capó
- Department of Hematology, University Hospital Son Espases, Palma de Mallorca, Balearic Islands, Spain
| | - María Navarro-Palou
- Department of Hematology, University Hospital Son Espases, Palma de Mallorca, Balearic Islands, Spain
| | - Teresa Ros
- Department of Hematology, University Hospital Son Espases, Palma de Mallorca, Balearic Islands, Spain
| | - Juan Carlos Amat
- Department of Hematology, University Hospital Son Espases, Palma de Mallorca, Balearic Islands, Spain
| | - Bernardo Lopez
- Department of Hematology, University Hospital Son Espases, Palma de Mallorca, Balearic Islands, Spain
| | - Toni F Marcus
- Department of Hematology, University Hospital Son Espases, Palma de Mallorca, Balearic Islands, Spain
| | - Laura Fueyo
- Department of Clinical Analysis, University Hospital Son Espases, Palma de Mallorca, Balearic Islands, Spain
| | - Angela G Suquia
- Department of Clinical Analysis, University Hospital Son Espases, Palma de Mallorca, Balearic Islands, Spain
| | - Jordi Gines
- Department of Pharmacy, University Hospital Son Espases, Palma de Mallorca, Balearic Islands, Spain
| | - Francisco Rubio
- Department of Hematology, University Hospital Son Espases, Palma de Mallorca, Balearic Islands, Spain
| | - Rafael Ramos
- Department of Pathology, University Hospital Son Espases, Palma de Mallorca, Balearic Islands, Spain
| | - Joan Besalduch
- Department of Hematology, University Hospital Son Espases, Palma de Mallorca, Balearic Islands, Spain
| |
Collapse
|