101
|
Poyet C, Buser L, Roudnicky F, Detmar M, Hermanns T, Mannhard D, Höhn A, Rüschoff J, Zhong Q, Sulser T, Moch H, Wild PJ. Connexin 43 expression predicts poor progression-free survival in patients with non-muscle invasive urothelial bladder cancer. J Clin Pathol 2015; 68:819-24. [PMID: 26251520 PMCID: PMC4602233 DOI: 10.1136/jclinpath-2015-202898] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 05/25/2015] [Indexed: 12/05/2022]
Abstract
Objectives To evaluate the protein expression of connexin 43 (Cx43) in primary urothelial bladder cancer and test its association with the histopathological characteristics and clinical outcome. Methods A tissue microarray containing 348 tissue samples from 174 patients with primary urothelial carcinomas of the bladder was immunohistochemically stained for Cx43. The intensity of staining was semiquantitatively evaluated (score 0, 1+, 2+), and the association with clinicopathological features was assessed. Univariable and multivariable analyses were performed to identify predictors for progression-free survival (PFS). Results Membranous Cx43 immunoreactivity was detected in 118 (67.8%) of 174 analysable urothelial carcinomas, of which 31 (17.8%) showed even a strong (score 2+) and mainly homogeneous staining. Strong expression levels of Cx43 (score 2+) were associated with higher tumour grade, multiplicity and increased proliferation (all p<0.05). In the subgroup of patients with stage pTa and pT1 bladder tumours (n=158), strong Cx43 expression (p<0.001), solid growth pattern (p<0.001) and increased Ki-67 proliferation fraction (p<0.05) were significantly associated with shorter PFS in an univariable Cox regression analysis. In multivariable Cox regression models, Cx43 immunoreactivity and histological growth pattern remained highly significant and adverse risk factors for PFS. Conclusions The expression levels of Cx43 are frequent in non-muscle invasive bladder cancer (NMIBC), with high expression levels being associated with poor prognosis. Routine assessment of Cx43 expression may improve the identification of high-risk NMIBC.
Collapse
Affiliation(s)
- Cédric Poyet
- Department of Urology, University Hospital Zurich, Zurich, Switzerland
| | - Lorenz Buser
- Institute of Surgical Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Filip Roudnicky
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| | - Michael Detmar
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| | - Thomas Hermanns
- Department of Urology, University Hospital Zurich, Zurich, Switzerland
| | - Doris Mannhard
- Department of Urology, University Hospital Zurich, Zurich, Switzerland
| | - Andrej Höhn
- Department of Urology, University Hospital Zurich, Zurich, Switzerland
| | - Jan Rüschoff
- Institute of Surgical Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Qing Zhong
- Institute of Surgical Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Tullio Sulser
- Department of Urology, University Hospital Zurich, Zurich, Switzerland
| | - Holger Moch
- Institute of Surgical Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Peter J Wild
- Institute of Surgical Pathology, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
102
|
Tittarelli A, Guerrero I, Tempio F, Gleisner MA, Avalos I, Sabanegh S, Ortíz C, Michea L, López MN, Mendoza-Naranjo A, Salazar-Onfray F. Overexpression of connexin 43 reduces melanoma proliferative and metastatic capacity. Br J Cancer 2015; 113:259-67. [PMID: 26135897 PMCID: PMC4506378 DOI: 10.1038/bjc.2015.162] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Revised: 03/18/2015] [Accepted: 04/21/2015] [Indexed: 01/14/2023] Open
Abstract
Background: Alterations in connexin 43 (Cx43) expression and/or gap junction (GJ)-mediated intercellular communication are implicated in cancer pathogenesis. Herein, we have investigated the role of Cx43 in melanoma cell proliferation and apoptosis sensitivity in vitro, as well as metastatic capability and tumour growth in vivo. Methods: Connexin 43 expression levels, GJ coupling and proliferation rates were analysed in four different human melanoma cell lines. Furthermore, tumour growth and lung metastasis of high compared with low Cx43-expressing FMS cells were evaluated in vivo using a melanoma xenograft model. Results: Specific inhibition of Cx43 channel activity accelerated melanoma cell proliferation, whereas overexpression of Cx43 increased GJ coupling and reduced cell growth. Moreover, Cx43 overexpression in FMS cells increased basal and tumour necrosis factor-α-induced apoptosis and resulted in decreased melanoma tumour growth and lower number and size of metastatic foci in vivo. Conclusions: Our findings reveal an important role for Cx43 in intrinsically controlling melanoma growth, death and metastasis, and emphasise the potential use of compounds that selectively enhance Cx43 expression on melanoma in the future chemotherapy and/or immunotherapy protocols.
Collapse
Affiliation(s)
- A Tittarelli
- Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago 8380453, Chile
| | - I Guerrero
- Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago 8380453, Chile
| | - F Tempio
- 1] Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago 8380453, Chile [2] Millennium Institute on Immunology and Immunotherapy, Institute of Biomedical Sciences, University of Chile, Santiago 8380453, Chile
| | - M A Gleisner
- Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago 8380453, Chile
| | - I Avalos
- Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago 8380453, Chile
| | - S Sabanegh
- Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago 8380453, Chile
| | - C Ortíz
- Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago 8380453, Chile
| | - L Michea
- 1] Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago 8380453, Chile [2] Millennium Institute on Immunology and Immunotherapy, Institute of Biomedical Sciences, University of Chile, Santiago 8380453, Chile
| | - M N López
- 1] Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago 8380453, Chile [2] Millennium Institute on Immunology and Immunotherapy, Institute of Biomedical Sciences, University of Chile, Santiago 8380453, Chile [3] Research Support Office, University of Chile Clinical Hospital, Santiago 8380453, Chile
| | - A Mendoza-Naranjo
- UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6DD, UK
| | - F Salazar-Onfray
- 1] Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago 8380453, Chile [2] Millennium Institute on Immunology and Immunotherapy, Institute of Biomedical Sciences, University of Chile, Santiago 8380453, Chile
| |
Collapse
|
103
|
Nahta R, Al-Mulla F, Al-Temaimi R, Amedei A, Andrade-Vieira R, Bay S, G. Brown D, Calaf GM, Castellino RC, Cohen-Solal KA, Colacci A, Cruickshanks N, Dent P, Di Fiore R, Forte S, Goldberg GS, Hamid RA, Krishnan H, Laird DW, Lasfar A, Marignani PA, Memeo L, Mondello C, Naus CC, Ponce-Cusi R, Raju J, Roy D, Roy R, P. Ryan E, Salem HK, Scovassi AI, Singh N, Vaccari M, Vento R, Vondráček J, Wade M, Woodrick J, Bisson WH. Mechanisms of environmental chemicals that enable the cancer hallmark of evasion of growth suppression. Carcinogenesis 2015; 36 Suppl 1:S2-S18. [PMID: 26106139 PMCID: PMC4565608 DOI: 10.1093/carcin/bgv028] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2014] [Revised: 09/01/2014] [Accepted: 09/19/2014] [Indexed: 12/18/2022] Open
Abstract
As part of the Halifax Project, this review brings attention to the potential effects of environmental chemicals on important molecular and cellular regulators of the cancer hallmark of evading growth suppression. Specifically, we review the mechanisms by which cancer cells escape the growth-inhibitory signals of p53, retinoblastoma protein, transforming growth factor-beta, gap junctions and contact inhibition. We discuss the effects of selected environmental chemicals on these mechanisms of growth inhibition and cross-reference the effects of these chemicals in other classical cancer hallmarks.
Collapse
Affiliation(s)
- Rita Nahta
- *To whom correspondence should be addressed.
| | - Fahd Al-Mulla
- Department of Pathology, Kuwait University, Safat 13110, Kuwait
| | | | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Firenze, 50134 Florence, Italy
| | - Rafaela Andrade-Vieira
- Department of Biochemistry and Molecular Biology, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
| | - Sarah Bay
- Program in Genetics and Molecular Biology, Graduate Division of Biological and Biomedical Sciences, Emory University, Atlanta, GA 30322, USA
| | - Dustin G. Brown
- Department of Environmental and Radiological Health Sciences/Colorado State University/Colorado School of Public Health, Fort Collins, CO 80523-1680, USA
| | - Gloria M. Calaf
- Center for Radiological Research, Columbia University Medical Center, New York, NY 10032, USA
- Instituto de Alta Investigacion, Universidad de Tarapaca, Arica 8097877, Chile
| | - Robert C. Castellino
- Division of Hematology and Oncology, Department of Pediatrics, Children’s Healthcare of Atlanta and Emory University, Atlanta, GA 30322, USA
| | - Karine A. Cohen-Solal
- Department of Medicine/Medical Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08901-1914, USA
| | - Annamaria Colacci
- Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, Bologna 40126, Italy
| | - Nichola Cruickshanks
- Departments of Neurosurgery and Biochemistry and Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 980033, USA
| | - Paul Dent
- Departments of Neurosurgery and Biochemistry and Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 980033, USA
| | - Riccardo Di Fiore
- Department of Biological, Chemical, and Pharmaceutical Sciences and Technologies, Polyclinic Plexus, University of Palermo, 90127 Palermo, Italy
| | - Stefano Forte
- Mediterranean Institute of Oncology, 95029 Viagrande, Italy
| | - Gary S. Goldberg
- Graduate School of Biomedical Sciences and Department of Molecular Biology, School of Osteopathic Medicine, Rowan University, Stratford, NJ 08084-1501, USA
| | - Roslida A. Hamid
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, University Putra, Serdang, Selangor 43400, Malaysia
| | - Harini Krishnan
- Graduate School of Biomedical Sciences and Department of Molecular Biology, School of Osteopathic Medicine, Rowan University, Stratford, NJ 08084-1501, USA
| | - Dale W. Laird
- Department of Anatomy and Cell Biology, University of Western Ontario, London, Ontario N6A 5C1, Canada
| | - Ahmed Lasfar
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers, the State University of New Jersey, Piscataway, NJ 60503, USA
| | - Paola A. Marignani
- Department of Biochemistry and Molecular Biology, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
| | - Lorenzo Memeo
- Mediterranean Institute of Oncology, 95029 Viagrande, Italy
| | - Chiara Mondello
- Institute of Molecular Genetics, National Research Council, 27100 Pavia, Italy
| | - Christian C. Naus
- Department of Cellular & Physiological Sciences, Life Sciences Institute, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Richard Ponce-Cusi
- Instituto de Alta Investigacion, Universidad de Tarapaca, Arica 8097877, Chile
| | - Jayadev Raju
- Toxicology Research Division, Bureau of Chemical Safety Food Directorate, Health Products and Food Branch Health Canada, Ottawa, Ontario K1A0K9, Canada
| | - Debasish Roy
- Department of Natural Science, The City University of New York at Hostos Campus, Bronx, NY 10451, USA
| | - Rabindra Roy
- Molecular Oncology Program, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington DC 20057, USA
| | - Elizabeth P. Ryan
- Department of Environmental and Radiological Health Sciences/Colorado State University/Colorado School of Public Health, Fort Collins, CO 80523-1680, USA
| | - Hosni K. Salem
- Urology Dept., kasr Al-Ainy School of Medicine, Cairo University, El Manial, Cairo 12515, Egypt
| | - A. Ivana Scovassi
- Institute of Molecular Genetics, National Research Council, 27100 Pavia, Italy
| | - Neetu Singh
- Advanced Molecular Science Research Centre, King George’s Medical University, Lucknow, UP 226003, India
| | - Monica Vaccari
- Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, Bologna 40126, Italy
| | - Renza Vento
- Department of Biological, Chemical, and Pharmaceutical Sciences and Technologies, Polyclinic Plexus, University of Palermo, 90127 Palermo, Italy
- Sbarro Institute for Cancer Research and Molecular Medicine, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA
| | - Jan Vondráček
- Department of Cytokinetics, Institute of Biophysics AS CR, Brno 612 65, Czech Republic
| | - Mark Wade
- Center for Genomic Science of IIT@SEMM, Istituto Italiano di Tecnologia (IIT), Milan 16163, Italy and
| | - Jordan Woodrick
- Molecular Oncology Program, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington DC 20057, USA
| | - William H. Bisson
- Environmental and Molecular Toxicology, Environmental Health Sciences Center, Oregon State University, Corvallis, OR 97331, USA
| |
Collapse
|
104
|
Abstract
Despite considerable advancements that shattered previously held dogmas about the metastatic cascade, the evolution of therapies to treat metastatic disease has not kept up. In this Opinion article, I argue that, rather than waiting for metastases to emerge before initiating treatment, it would be more effective to target metastatic seeds before they sprout. Specifically, I advocate directing therapies towards the niches that harbour dormant disseminated tumour cells to sensitize them to cytotoxic agents. Treatment sensitization, achieved by disrupting reservoirs of leukaemic stem cells and latent HIV, argues that this approach, although unconventional, could succeed in improving patient survival by delaying or even preventing metastasis.
Collapse
Affiliation(s)
- Cyrus M. Ghajar
- Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, WA 98109 (USA)
- To whom correspondence should be addressed: Cyrus M. Ghajar, PhD, Public Health Sciences Division/ Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, P: 206.667.7080, F: 206.667.2537,
| |
Collapse
|
105
|
Pezzella F, Gatter K. Non-angiogenic tumours unveil a new chapter in cancer biology. J Pathol 2015; 235:381-3. [PMID: 25351454 DOI: 10.1002/path.4474] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Revised: 10/20/2014] [Accepted: 10/21/2014] [Indexed: 11/07/2022]
Abstract
The term 'angiogenesis' was coined in 1787 and the role of vessels in cancer has been studied ever since. In 1971 Folkman introduced the hypothesis, until now widely accepted, that tumour growth is strictly dependent on angiogenesis. However, the discovery that tumours can also grow without angiogenesis by exploiting pre-existing vessels, both in humans and more recently in mice, has demonstrated that this is not always the case. These observations highlight a new aspect of the interaction between vessels and tumours and demonstrate the existence of a previously unrecognized group of tumours that grow without angiogenesis and whose biology is, so far, largely unknown.
Collapse
Affiliation(s)
- Francesco Pezzella
- Nuffield Division of Clinical Laboratory Science, Radcliffe Department of Medicine, John Radcliffe Hospital, Oxford, UK
| | | |
Collapse
|
106
|
Karpinich NO, Caron KM. Gap junction coupling is required for tumor cell migration through lymphatic endothelium. Arterioscler Thromb Vasc Biol 2015; 35:1147-55. [PMID: 25792452 DOI: 10.1161/atvbaha.114.304752] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Accepted: 03/06/2015] [Indexed: 11/16/2022]
Abstract
OBJECTIVE The lymphatic vasculature is a well-established conduit for metastasis, but the mechanisms by which tumor cells interact with lymphatic endothelial cells (LECs) to facilitate escape remain poorly understood. Elevated levels of the lymphangiogenic peptide adrenomedullin are found in many tumors, and we previously characterized that its expression is necessary for lymphatic vessel growth within both tumors and sentinel lymph nodes and for distant metastasis. APPROACH AND RESULTS This study used a tumor cell-LEC coculture system to identify a series of adrenomedullin-induced events that facilitated transendothelial migration of the tumor cells through a lymphatic monolayer. High levels of adrenomedullin expression enhanced adhesion of tumor cells to LECs, and further analysis revealed that adrenomedullin promoted gap junction coupling between LECs as evidenced by spread of Lucifer yellow dye. Adrenomedullin also enhanced heterocellular gap junction coupling as demonstrated by Calcein dye transfer from tumor cells into LECs. This connexin-mediated gap junction intercellular communication was necessary for tumor cells to undergo transendothelial migration because pharmacological blockade of this heterocellular communication prevented the ability of tumor cells to transmigrate through the lymphatic monolayer. In addition, treatment of LECs with adrenomedullin caused nuclear translocation of β-catenin, a component of endothelial cell junctions, causing an increase in transcription of the downstream target gene C-MYC. Importantly, blockade of gap junction intercellular communication prevented β-catenin nuclear translocation. CONCLUSIONS Our findings indicate that maintenance of cell-cell communication is necessary to facilitate a cascade of events that lead to tumor cell migration through the lymphatic endothelium.
Collapse
Affiliation(s)
- Natalie O Karpinich
- From the Department of Cell Biology and Physiology (N.O.K., K.M.C.) and Department of Genetics (K.M.C.), University of North Carolina at Chapel Hill
| | - Kathleen M Caron
- From the Department of Cell Biology and Physiology (N.O.K., K.M.C.) and Department of Genetics (K.M.C.), University of North Carolina at Chapel Hill.
| |
Collapse
|
107
|
Chernet BT, Fields C, Levin M. Long-range gap junctional signaling controls oncogene-mediated tumorigenesis in Xenopus laevis embryos. Front Physiol 2015; 5:519. [PMID: 25646081 PMCID: PMC4298169 DOI: 10.3389/fphys.2014.00519] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Accepted: 12/18/2014] [Indexed: 11/21/2022] Open
Abstract
In addition to the immediate microenvironment, long-range signaling may be an important component of cancer. Molecular-genetic analyses have implicated gap junctions-key mediators of cell-cell communication-in carcinogenesis. We recently showed that the resting voltage potential of distant cell groups is a key determinant of metastatic transformation and tumor induction. Here, we show in the Xenopus laevis model that gap junctional communication (GJC) is a modulator of the long-range bioelectric signaling that regulates tumor formation. Genetic disruption of GJC taking place within tumors, within remote host tissues, or between the host and tumors significantly lowers the incidence of tumors induced by KRAS mutations. The most pronounced suppression of tumor incidence was observed upon GJC disruption taking place farther away from oncogene-expressing cells, revealing a role for GJC in distant cells in the control of tumor growth. In contrast, enhanced GJC communication through the overexpression of wild-type connexin Cx26 increased tumor incidence. Our data confirm a role for GJC in tumorigenesis, and reveal that this effect is non-local. Based on these results and on published data on movement of ions through GJs, we present a quantitative model linking the GJC coupling and bioelectrical state of cells to the ability of oncogenes to initiate tumorigenesis. When integrated with data on endogenous bioelectric signaling during left-right patterning, the model predicts differential tumor incidence outcomes depending on the spatial configurations of gap junction paths relative to tumor location and major anatomical body axes. Testing these predictions, we found that the strongest influence of GJ modulation on tumor suppression by hyperpolarization occurred along the embryonic left-right axis. Together, these data reveal new, long-range aspects of cancer control by the host's physiological parameters.
Collapse
Affiliation(s)
- Brook T. Chernet
- Department of Biology, Tufts Center for Regenerative and Developmental Biology, Tufts UniversityMedford, MA, USA
| | | | - Michael Levin
- Department of Biology, Tufts Center for Regenerative and Developmental Biology, Tufts UniversityMedford, MA, USA
| |
Collapse
|
108
|
Connexins in migration during development and cancer. Dev Biol 2014; 401:143-51. [PMID: 25553982 DOI: 10.1016/j.ydbio.2014.12.023] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Revised: 12/17/2014] [Accepted: 12/18/2014] [Indexed: 12/12/2022]
Abstract
Connexins, the gap junction proteins, through their multitude of actions are implicated in a variety of cell processes during animal development and cancer. They allow direct or paracrine/autocrine cell communication through their channel and hemi-channel functions. They enable adhesion and interact with a plethora of signalling molecules. Here, we review the common themes in developmental and pathological processes and we focus in their involvement in cell migration in four different systems: neurons, astrocytes, neural crest and cancer.
Collapse
|
109
|
Teleki I, Szasz AM, Maros ME, Gyorffy B, Kulka J, Meggyeshazi N, Kiszner G, Balla P, Samu A, Krenacs T. Correlations of differentially expressed gap junction connexins Cx26, Cx30, Cx32, Cx43 and Cx46 with breast cancer progression and prognosis. PLoS One 2014; 9:e112541. [PMID: 25383624 PMCID: PMC4226536 DOI: 10.1371/journal.pone.0112541] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2014] [Accepted: 10/06/2014] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND AND AIMS Connexins and their cell membrane channels contribute to the control of cell proliferation and compartmental functions in breast glands and their deregulation is linked to breast carcinogenesis. Our aim was to correlate connexin expression with tumor progression and prognosis in primary breast cancers. MATERIALS AND METHODS Meta-analysis of connexin isotype expression data of 1809 and 1899 breast cancers from the Affymetrix and Illumina array platforms, respectively, was performed. Expressed connexins were also monitored at the protein level in tissue microarrays of 127 patients equally representing all tumor grades, using immunofluorescence and multilayer, multichannel digital microscopy. Prognostic correlations were plotted in Kaplan-Meier curves and tested using the log-rank test and cox-regression analysis in univariate and multivariate models. RESULTS The expression of GJA1/Cx43, GJA3/Cx46 and GJB2/Cx26 and, for the first time, GJA6/Cx30 and GJB1/Cx32 was revealed both in normal human mammary glands and breast carcinomas. Within their subfamilies these connexins can form homo- and heterocellular epithelial channels. In cancer, the array datasets cross-validated each other's prognostic results. In line with the significant correlations found at mRNA level, elevated Cx43 protein levels were linked with significantly improved breast cancer outcome, offering Cx43 protein detection as an independent prognostic marker stronger than vascular invasion or necrosis. As a contrary, elevated Cx30 mRNA and protein levels were associated with a reduced disease outcome offering Cx30 protein detection as an independent prognostic marker outperforming mitotic index and necrosis. Elevated versus low Cx43 protein levels allowed the stratification of grade 2 tumors into good and poor relapse free survival subgroups, respectively. Also, elevated versus low Cx30 levels stratified grade 3 patients into poor and good overall survival subgroups, respectively. CONCLUSION Differential expression of Cx43 and Cx30 may serve as potential positive and negative prognostic markers, respectively, for a clinically relevant stratification of breast cancers.
Collapse
Affiliation(s)
- Ivett Teleki
- 1 Department of Pathology & Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | | | - Mate Elod Maros
- 1 Department of Pathology & Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Balazs Gyorffy
- MTA TTK Lendulet Cancer Biomarker Research Group, Budapest, Hungary
- 2 Department of Pediatrics, Semmelweis University, Budapest, Hungary
- MTA-SE Pediatrics and Nephrology Research Group, Budapest, Hungary
| | - Janina Kulka
- 2 Department of Pathology, Semmelweis University, Budapest, Hungary
| | - Nora Meggyeshazi
- 1 Department of Pathology & Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Gergo Kiszner
- 1 Department of Pathology & Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Peter Balla
- 1 Department of Pathology & Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Aliz Samu
- 1 Department of Pathology & Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Tibor Krenacs
- 1 Department of Pathology & Experimental Cancer Research, Semmelweis University, Budapest, Hungary
- MTA-SE Tumor Progression Research Group, Budapest, Hungary
| |
Collapse
|
110
|
Jilaveanu LB, Parisi F, Barr ML, Zito CR, Cruz-Munoz W, Kerbel RS, Rimm DL, Bosenberg MW, Halaban R, Kluger Y, Kluger HM. PLEKHA5 as a Biomarker and Potential Mediator of Melanoma Brain Metastasis. Clin Cancer Res 2014; 21:2138-47. [PMID: 25316811 DOI: 10.1158/1078-0432.ccr-14-0861] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Accepted: 09/17/2014] [Indexed: 01/13/2023]
Abstract
PURPOSE Approximately 40% of patients with metastatic melanoma develop brain metastases. Our purpose was to identify genes aberrantly expressed in melanoma that might be associated with propensity for brain homing. EXPERIMENTAL DESIGN We studied gene expression profiles in a cell line model of brain metastasis (cerebrotropic A375Br cells vs. parental A375P cells) and compared them with profiles of patients who developed early brain metastases and who did not. A tissue microarray containing 169 metastatic melanoma cases with variable time to brain metastasis was constructed to further study marker expression by quantitative immunofluorescence. An in vitro model of the blood brain barrier (BBB) was generated to evaluate potential mediators of brain metastases. RESULTS PLEKHA5 was differentially expressed in both the A375 cell line model and patient samples subjected to gene expression profiling. At the protein level, by quantitative immunofluorescence, PLEKHA5 was associated with decreased brain metastasis-free survival. PLEKHA5 overexpression was not associated with other metastatic sites. Knockdown of PLEKHA5 decreases the viability of A375Br cells, inhibits BBB transmigration and invasion in vitro. Similar results were found with YUMUL cells, cultured from a patient with overwhelming brain metastases. PLEKHA5 knockdown did not affect the viability of A375P cells. CONCLUSIONS PLEKHA5 expression in melanoma tumors was associated with early development of brain metastases. Inhibition of PLEKHA5 might decrease passage across the BBB and decrease proliferation and survival of melanoma cells both in the brain and in extracerebral sites.
Collapse
Affiliation(s)
- Lucia B Jilaveanu
- Department of Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, Connecticut
| | - Fabio Parisi
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut
| | - Meaghan L Barr
- Department of Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, Connecticut
| | - Christopher R Zito
- Department of Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, Connecticut. Department of Biology, School of Health and Natural Sciences, University of Saint Joseph, West Hartford, Connecticut
| | - William Cruz-Munoz
- Department of Medical Biophysics, Sunnybrook Research Institute, Biological Sciences Platform, University of Toronto, Toronto, Ontario, Canada
| | - Robert S Kerbel
- Department of Medical Biophysics, Sunnybrook Research Institute, Biological Sciences Platform, University of Toronto, Toronto, Ontario, Canada
| | - David L Rimm
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut
| | - Marcus W Bosenberg
- Department of Dermatology, Yale University School of Medicine, New Haven, Connecticut
| | - Ruth Halaban
- Department of Dermatology, Yale University School of Medicine, New Haven, Connecticut
| | - Yuval Kluger
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut
| | - Harriet M Kluger
- Department of Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, Connecticut.
| |
Collapse
|
111
|
Berginski ME, Creed SJ, Cochran S, Roadcap DW, Bear JE, Gomez SM. Automated analysis of invadopodia dynamics in live cells. PeerJ 2014; 2:e462. [PMID: 25071988 PMCID: PMC4103095 DOI: 10.7717/peerj.462] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 06/09/2014] [Indexed: 01/07/2023] Open
Abstract
Multiple cell types form specialized protein complexes that are used by the cell to actively degrade the surrounding extracellular matrix. These structures are called podosomes or invadopodia and collectively referred to as invadosomes. Due to their potential importance in both healthy physiology as well as in pathological conditions such as cancer, the characterization of these structures has been of increasing interest. Following early descriptions of invadopodia, assays were developed which labelled the matrix underneath metastatic cancer cells allowing for the assessment of invadopodia activity in motile cells. However, characterization of invadopodia using these methods has traditionally been done manually with time-consuming and potentially biased quantification methods, limiting the number of experiments and the quantity of data that can be analysed. We have developed a system to automate the segmentation, tracking and quantification of invadopodia in time-lapse fluorescence image sets at both the single invadopodia level and whole cell level. We rigorously tested the ability of the method to detect changes in invadopodia formation and dynamics through the use of well-characterized small molecule inhibitors, with known effects on invadopodia. Our results demonstrate the ability of this analysis method to quantify changes in invadopodia formation from live cell imaging data in a high throughput, automated manner.
Collapse
Affiliation(s)
- Matthew E Berginski
- UNC/NCSU Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill , Chapel Hill, NC , USA
| | - Sarah J Creed
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill , Chapel Hill, NC , USA
| | - Shelly Cochran
- UNC/NCSU Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill , Chapel Hill, NC , USA
| | - David W Roadcap
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill , Chapel Hill, NC , USA
| | - James E Bear
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill , Chapel Hill, NC , USA ; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill , Chapel Hill, NC , USA ; Howard Hughes Medical Institute , Chevy Chase, MD , USA
| | - Shawn M Gomez
- UNC/NCSU Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill , Chapel Hill, NC , USA ; Department of Computer Science, University of North Carolina at Chapel Hill , Chapel Hill, NC , USA ; Department of Pharmacology, University of North Carolina at Chapel Hill , Chapel Hill, NC , USA
| |
Collapse
|
112
|
Schalper KA, Carvajal-Hausdorf D, Oyarzo MP. Possible role of hemichannels in cancer. Front Physiol 2014; 5:237. [PMID: 25018732 PMCID: PMC4073485 DOI: 10.3389/fphys.2014.00237] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Accepted: 06/09/2014] [Indexed: 12/12/2022] Open
Abstract
In humans, connexins (Cxs) and pannexins (Panxs) are the building blocks of hemichannels. These proteins are frequently altered in neoplastic cells and have traditionally been considered as tumor suppressors. Alteration of Cxs and Panxs in cancer cells can be due to genetic, epigenetic and post-transcriptional/post-translational events. Activated hemichannels mediate the diffusional membrane transport of ions and small signaling molecules. In the last decade hemichannels have been shown to participate in diverse cell processes including the modulation of cell proliferation and survival. However, their possible role in tumor growth and expansion remains largely unexplored. Herein, we hypothesize about the possible role of hemichannels in carcinogenesis and tumor progression. To support this theory, we summarize the evidence regarding the involvement of hemichannels in cell proliferation and migration, as well as their possible role in the anti-tumor immune responses. In addition, we discuss the evidence linking hemichannels with cancer in diverse models and comment on the current technical limitations for their study.
Collapse
Affiliation(s)
- Kurt A Schalper
- Servicio Anatomía Patológica, Clínica Alemana de Santiago, Facultad de Medicina Clinica Alemana Universidad del Desarrollo Santiago, Chile ; Department of Pathology, Yale School of Medicine New Haven, CT, USA
| | | | - Mauricio P Oyarzo
- Servicio Anatomía Patológica, Clínica Alemana de Santiago, Facultad de Medicina Clinica Alemana Universidad del Desarrollo Santiago, Chile
| |
Collapse
|
113
|
Abstract
To comprehend the complexity of cancer, the biological characteristics acquired during the initiation and progression of tumours were classified as the 'hallmarks of cancer'. Intravital microscopy techniques have been developed to study individual cells that acquire these crucial traits, by visualizing tissues with cellular or subcellular resolution in living animals. In this Review, we highlight the latest intravital microscopy techniques that have been used in living animals (predominantly mice) to unravel fundamental and dynamic aspects of various hallmarks of cancer. In addition, we discuss the application of intravital microscopy techniques to cancer therapy, as well as limitations and future perspectives for these techniques.
Collapse
Affiliation(s)
- Saskia I J Ellenbroek
- Cancer Genomics Netherlands-Hubrecht Institute-KNAW & University Medical Centre Utrecht, Uppsalalaan 8, 3584 CT, Utrecht, The Netherlands
| | - Jacco van Rheenen
- Cancer Genomics Netherlands-Hubrecht Institute-KNAW & University Medical Centre Utrecht, Uppsalalaan 8, 3584 CT, Utrecht, The Netherlands
| |
Collapse
|
114
|
Abstract
SUMMARY Melanoma cells interact with and depend on seemingly normal cells in their tumour microenvironment to allow the acquisition of the hallmarks of solid cancer. In general, there are three types of interaction of melanoma cells with their microenvironment. First, there is bilateral communication between melanoma cells and the stroma, which includes fibroblasts, endothelial cells, immune cells, soluble molecules, and the extracellular matrix. Second, while under normal conditions keratinocytes control localisation and proliferative behaviour of melanocytes in the epidermis, once this balance is disturbed and a melanoma has developed, melanoma cells may take over the control of their epidermal tumour microenvironment. Finally, there are subcompartments within tumours with different microenvironmental milieu defined by their access to oxygen and nutrients. Therefore, different melanoma cells within a tumour face different microenvironments. Interactions between melanoma cells among each other and with the cell types in their microenvironment happen through endocrine and paracrine communication and/or through direct contact via cell-cell and cell-matrix adhesion, and gap junctional intercellular communication (GJIC). Connexins have been identified as key molecules for direct cell-cell communication and are also thought to be important for the release of signalling molecules from cells to the microenvironment. In this review we provide an update of the alterations in cell-cell communication in melanoma and the tumour microenvironment associated with melanoma development and progression.
Collapse
|
115
|
Sirnes S, Lind GE, Bruun J, Fykerud TA, Mesnil M, Lothe RA, Rivedal E, Kolberg M, Leithe E. Connexins in colorectal cancer pathogenesis. Int J Cancer 2014; 137:1-11. [PMID: 24752574 DOI: 10.1002/ijc.28911] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Accepted: 03/11/2014] [Indexed: 12/17/2022]
Abstract
The connexins constitute a family of integral membrane proteins that form channels between adjacent cells. These channels are assembled in plasma membrane domains known as gap junctions and enable cells to directly exchange ions and small molecules. Intercellular communication via gap junctions plays important roles in regulating cell growth and differentiation and in maintaining tissue homeostasis. This type of cell communication is often impaired during cancer development, and several members of the connexin protein family have been shown to act as tumor suppressors. Emerging evidence suggests that the connexin protein family has important roles in colorectal cancer development. In the normal colonic epithelial tissue, three connexin isoforms, connexin 26 (Cx26), Cx32 and Cx43, have been shown to be expressed at the protein level. Colorectal cancer development is associated with loss of connexin expression or relocalization of connexins from the plasma membrane to intracellular compartments. Downregulation of connexins in colorectal carcinomas at the transcriptional level involves cancer-specific promoter hypermethylation. Recent studies suggest that Cx43 may constrain growth of colon cancer cells by interfering with the Wnt/β-catenin pathway. There is also increasing evidence that the connexins may have potential as prognostic markers in colorectal cancer. This review discusses the role of connexins in colorectal cancer pathogenesis, as well as their potential as prognostic markers and targets in the prevention and treatment of the disease.
Collapse
Affiliation(s)
- Solveig Sirnes
- Department of Cancer Prevention, Institute for Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway; Faculty of Medicine, Centre for Cancer Biomedicine, University of Oslo, Oslo, Norway
| | | | | | | | | | | | | | | | | |
Collapse
|
116
|
Stoletov K, Bond D, Hebron K, Raha S, Zijlstra A, Lewis JD. Metastasis as a therapeutic target in prostate cancer: a conceptual framework. AMERICAN JOURNAL OF CLINICAL AND EXPERIMENTAL UROLOGY 2014; 2:45-56. [PMID: 25360439 PMCID: PMC4219290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Accepted: 03/26/2014] [Indexed: 06/04/2023]
Abstract
Metastasis is the main cause of prostate cancer-associated deaths. While significant progerss has been made in the treatment of primary tumors, efficent therapies that target the metastatic spread of prostate cancer are far from clinical reality. To efficiently treat cancer we need be able to impede its spread. Unfortunately, the majority of current therapeutics approved to treat metastatic cancer were originally selected based on their ability to inhibit primary tumor growth. This inherent flaw precluded these therapies from efficiently targeting the development of secondary metastatic lesions, a process that is distinct from that of primary tumor progression. In this review we will summarize the conceptual, cellular and molecular targets that should be considered to design effective anti-metastatic therapies.
Collapse
Affiliation(s)
- Konstantin Stoletov
- Department of Oncology, University of Alberta5-142C Katz Group Building, 114th St and 87th Ave, Edmonton, AB T6G 2E1 Canada
| | - David Bond
- Department of Oncology, University of Alberta5-142C Katz Group Building, 114th St and 87th Ave, Edmonton, AB T6G 2E1 Canada
| | - Katie Hebron
- Department of Pathology, Microbiology and Immunology, Vanderbilt University1161 21st Ave. S., C-2102C MCN, Nashville, TN 37232-2561 USA
| | - Srijan Raha
- Department of Oncology, University of Alberta5-142C Katz Group Building, 114th St and 87th Ave, Edmonton, AB T6G 2E1 Canada
| | - Andries Zijlstra
- Department of Pathology, Microbiology and Immunology, Vanderbilt University1161 21st Ave. S., C-2102C MCN, Nashville, TN 37232-2561 USA
| | - John D Lewis
- Department of Oncology, University of Alberta5-142C Katz Group Building, 114th St and 87th Ave, Edmonton, AB T6G 2E1 Canada
| |
Collapse
|
117
|
Defamie N, Chepied A, Mesnil M. Connexins, gap junctions and tissue invasion. FEBS Lett 2014; 588:1331-8. [PMID: 24457198 DOI: 10.1016/j.febslet.2014.01.012] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Revised: 01/13/2014] [Accepted: 01/14/2014] [Indexed: 12/14/2022]
Abstract
Formation of metastases negatively impacts the survival prognosis of cancer patients. Globally, if the various steps involved in their formation are relatively well identified, the molecular mechanisms responsible for the emergence of invasive cancer cells are still incompletely resolved. Elucidating what are the mechanisms that allow cancer cells to evade from the tumor is a crucial point since it is the first step of the metastatic potential of a solid tumor. In order to be invasive, cancer cells have to undergo transformations such as down-regulation of cell-cell adhesions, modification of cell-matrix adhesions and acquisition of proteolytic properties. These transformations are accompanied by the capacity to "activate" stromal cells, which may favor the motility of the invasive cells through the extracellular matrix. Since modulation of gap junctional intercellular communication is known to be involved in cancer, we were interested to consider whether these different transformations necessary for the acquisition of invasive phenotype are related with gap junctions and their structural proteins, the connexins. In this review, emerging roles of connexins and gap junctions in the process of tissue invasion are proposed.
Collapse
Affiliation(s)
- Norah Defamie
- Team IP2C, STIM laboratory, University of Poitiers, CNRS ERL 7368, 1 rue Georges Bonnet, B36, 86073 Poitiers Cedex9, France.
| | - Amandine Chepied
- Team IP2C, STIM laboratory, University of Poitiers, CNRS ERL 7368, 1 rue Georges Bonnet, B36, 86073 Poitiers Cedex9, France.
| | - Marc Mesnil
- Team IP2C, STIM laboratory, University of Poitiers, CNRS ERL 7368, 1 rue Georges Bonnet, B36, 86073 Poitiers Cedex9, France.
| |
Collapse
|
118
|
Avraham HK, Jiang S, Fu Y, Nakshatri H, Ovadia H, Avraham S. Angiopoietin-2 mediates blood-brain barrier impairment and colonization of triple-negative breast cancer cells in brain. J Pathol 2014; 232:369-81. [DOI: 10.1002/path.4304] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Revised: 11/06/2013] [Accepted: 11/11/2013] [Indexed: 12/19/2022]
Affiliation(s)
- Hava Karsenty Avraham
- Division of Experimental Medicine; Beth Israel Deaconess Medical Center and Harvard Medical School; Boston MA USA
| | - Shuxian Jiang
- Division of Experimental Medicine; Beth Israel Deaconess Medical Center and Harvard Medical School; Boston MA USA
| | - Yigong Fu
- Division of Experimental Medicine; Beth Israel Deaconess Medical Center and Harvard Medical School; Boston MA USA
| | - Harikrishna Nakshatri
- Departments of Surgery, and Biochemistry and Molecular Biology; Indiana University School of Medicine; Indianapolis IN USA
| | - Haim Ovadia
- Department of Neurology; Hadassah University Hospital; Jerusalem Israel
| | - Shalom Avraham
- Division of Experimental Medicine; Beth Israel Deaconess Medical Center and Harvard Medical School; Boston MA USA
| |
Collapse
|
119
|
Ableser MJ, Penuela S, Lee J, Shao Q, Laird DW. Connexin43 reduces melanoma growth within a keratinocyte microenvironment and during tumorigenesis in vivo. J Biol Chem 2013; 289:1592-603. [PMID: 24297173 DOI: 10.1074/jbc.m113.507228] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Connexins (Cx) have been identified as tumor suppressors or enhancers, a distinction that appears to be dependent on the type and stage of disease. However, the role of connexins in melanoma tumorigenesis and their status during cancer onset and progression remain controversial and unclear. Here, we show that the aggressive B16-BL6 mouse melanoma cell line expresses low basal levels of Cx26 and Cx43, rendering them gap junctional intercellular communication-deficient as elucidated by immunofluorescence, Western blotting, and dye transfer studies. Following ectopic expression of green fluorescent protein-tagged Cx26 and Cx43 in these connexin-deficient melanomas, punctate gap junction-like plaques were evident at sites of cell-cell apposition, and the incidence of dye transfer was significantly increased similar to connexin-rich keratinocytes. We found that the expression of Cx43, but not Cx26, significantly reduced cellular proliferation and anchorage-independent growth from control melanomas, whereas migration was unaffected. Additionally, melanomas expressing Cx43 displayed significantly reduced growth within the in situ-like microenvironment of keratinocytes, despite a lack of heterocellular gap junctional intercellular communication between the two cell types. Furthermore, when grown in vivo in the chicken chorioallantoic membrane, primary tumors derived from Cx43-expressing melanomas were significantly smaller than controls, whereas Cx26-expressing melanomas produced tumors similar to controls. Collectively, these results suggest that Cx43, and not Cx26, can act as a tumor suppressor during melanoma tumorigenesis.
Collapse
|
120
|
Alexander S, Weigelin B, Winkler F, Friedl P. Preclinical intravital microscopy of the tumour-stroma interface: invasion, metastasis, and therapy response. Curr Opin Cell Biol 2013; 25:659-71. [PMID: 23896198 DOI: 10.1016/j.ceb.2013.07.001] [Citation(s) in RCA: 111] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Revised: 07/01/2013] [Accepted: 07/02/2013] [Indexed: 01/10/2023]
Abstract
Key steps of cancer progression and therapy response depend upon interactions between cancer cells with the reactive tumour microenvironment. Intravital microscopy enables multi-modal and multi-scale monitoring of cancer progression as a dynamic step-wise process within anatomic and functional niches provided by the microenvironment. These niches deliver cell-derived and matrix-derived signals that enable cell subsets or single cancer cells to survive, migrate, grow, undergo dormancy, and escape immune surveillance. Beyond basic research, intravital microscopy has reached preclinical application to identify mechanisms of tumour-stroma interactions and outcome. We here summarise how n-dimensional 'dynamic histopathology' of tumours by intravital microscopy shapes mechanistic insight into cell-cell and cell-tissue interactions that underlie single-cell and collective cancer invasion, metastatic seeding at distant sites, immune evasion, and therapy responses.
Collapse
Affiliation(s)
- Stephanie Alexander
- David H. Koch Center for Applied Research of Genitourinary Cancers, Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | | | | | | |
Collapse
|
121
|
Gaziel-Sovran A, Osman I, Hernando E. In vivo Modeling and Molecular Characterization: A Path Toward Targeted Therapy of Melanoma Brain Metastasis. Front Oncol 2013; 3:127. [PMID: 23750336 PMCID: PMC3668495 DOI: 10.3389/fonc.2013.00127] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Accepted: 05/06/2013] [Indexed: 11/24/2022] Open
Abstract
Brain metastasis (B-Met) from melanoma remains mostly incurable and the main cause of death from the disease. Early stage clinical trials and case studies show some promise for targeted therapies in the treatment of melanoma B-Met. However, the progression-free survival for currently available therapies, although significantly improved, is still very short. The development of new potent agents to eradicate melanoma B-Met relies on the elucidation of the molecular mechanisms that allow melanoma cells to reach and colonize the brain. The discovery of such mechanisms depends heavily on pre-clinical models that enable the testing of candidate factors and therapeutic agents in vivo. In this review we summarize the effects of available targeted therapies on melanoma B-Met in the clinic. We provide an overview of existing pre-clinical models to study the disease and discuss specific molecules and mechanisms reported to modulate different aspects of melanoma B-Met and finally, by integrating both clinical and basic data, we summarize both opportunities and challenges currently presented to researchers in the field.
Collapse
Affiliation(s)
- Avital Gaziel-Sovran
- Interdisciplinary Melanoma Cooperative Group, NYU Cancer Institute, NYU Langone Medical Center , New York, NY , USA ; Department of Pathology, NYU School of Medicine , New York, NY , USA
| | | | | |
Collapse
|
122
|
Chernet B, Levin M. Endogenous Voltage Potentials and the Microenvironment: Bioelectric Signals that Reveal, Induce and Normalize Cancer. JOURNAL OF CLINICAL & EXPERIMENTAL ONCOLOGY 2013; Suppl 1:S1-002. [PMID: 25525610 PMCID: PMC4267524 DOI: 10.4172/2324-9110.s1-002] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Cancer may be a disease of geometry: a misregulation of the field of information that orchestrates individual cells' activities towards normal anatomy. Recent work identified molecular mechanisms underlying a novel system of developmental control: bioelectric gradients. Endogenous spatio-temporal differences in resting potential of non-neural cells provide instructive cues for cell regulation and complex patterning during embryogenesis and regeneration. It is now appreciated that these cues are an important layer of the dysregulation of cell: cell interactions that leads to cancer. Abnormal depolarization of resting potential (Vmem) is a convenient marker for neoplasia and activates a metastatic phenotype in genetically-normal cells in vivo. Moreover, oncogene expression depolarizes cells that form tumor-like structures, but is unable to form tumors if this depolarization is artificially prevented by misexpression of hyperpolarizing ion channels. Vmem triggers metastatic behaviors at considerable distance, mediated by transcriptional and epigenetic effects of electrically-modulated flows of serotonin and butyrate. While in vivo data on voltages in carcinogenesis comes mainly from the amphibian model, unbiased genetic screens and network profiling in rodents and human tissues reveal several ion channel proteins as bona fide oncogene and promising targets for cancer drug development. However, we propose that a focus on specific channel genes is just the tip of the iceberg. Bioelectric state is determined by post-translational gating of ion channels, not only from genetically-specified complements of ion translocators. A better model is a statistical dynamics view of spatial Vmem gradients. Cancer may not originate at the single cell level, since gap junctional coupling results in multi-cellular physiological networks with multiple stable attractors in bioelectrical state space. New medical applications await a detailed understanding of the mechanisms by which organ target morphology stored in real-time patterns of ion flows is perceived or mis-perceived by cells. Mastery of somatic voltage gradients will lead to cancer normalization or rebooting strategies, such as those that occur in regenerating and embryonic organs, resulting in transformative advances in basic biology and oncology.
Collapse
Affiliation(s)
| | - Michael Levin
- Corresponding author: Michael Levin, Department of Biology, Tufts Center for Regenerative and Developmental Biology, Tufts University, 200 Boston Ave., Suite 4600, Medford, MA 02155, USA, Tel: (617) 627-6161; Fax:(617) 627- 6121;
| |
Collapse
|