1
|
Ma J, Zhai Y, Ren X, Wu H, Yang M, Chai L, Chen J. Transformative insights in breast cancer: review of atomic force microscopy applications. Discov Oncol 2025; 16:256. [PMID: 40021496 PMCID: PMC11871204 DOI: 10.1007/s12672-025-02003-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 02/20/2025] [Indexed: 03/03/2025] Open
Abstract
Breast cancer remains one of the foremost global health concerns, highlighting the urgent need for innovative diagnostic and therapeutic strategies. Traditional imaging techniques, such as mammography and ultrasound, play essential roles in clinical practice; however, they often fall short in detecting early-stage tumors and providing comprehensive insights into the mechanical properties of cancer cells. In this context, Atomic Force Microscopy (AFM) has emerged as a transformative tool in breast cancer research, owing to its high-resolution imaging capabilities and nanomechanical characterization. This review explores recent advancements in AFM technology as applied to breast cancer research, emphasizing key findings that include the differentiation of various stages of tumor progression through high-resolution imaging, precise characterization of mechanical properties, and the capability for single-cell analysis. These capabilities not only enhance our understanding of tumor heterogeneity but also reveal potential biomarkers for early detection and therapeutic targets. Furthermore, the review critically examines several challenges and limitations associated with the application of AFM in breast cancer research. Issues such as complexities in sample preparation, accessibility, and the cost of AFM technology are discussed. Despite these challenges, the potential of AFM to transform our understanding of breast cancer biology is significant. Looking ahead, continued advancements in AFM technology promise to deepen our insights into breast cancer biology and guide innovative therapeutic strategies aimed at improving patient outcomes.
Collapse
Affiliation(s)
- Jiamin Ma
- Department of Breast Surgery, The Second Affiliated Hospital of Zhengzhou University, No.2 Jingba Road, Zhengzhou, 450000, Henan, China.
| | - Yuanyuan Zhai
- Department of Breast Surgery, The Second Affiliated Hospital of Zhengzhou University, No.2 Jingba Road, Zhengzhou, 450000, Henan, China
| | - Xiaoyi Ren
- Department of Breast Surgery, The Second Affiliated Hospital of Zhengzhou University, No.2 Jingba Road, Zhengzhou, 450000, Henan, China
| | - Huifang Wu
- Department of Pathology, The Second Affiliated Hospital of Zhengzhou University, No.2 Jingba Road, Zhengzhou, 450000, Henan, China
| | - Mengjie Yang
- Department of Breast Surgery, The Second Affiliated Hospital of Zhengzhou University, No.2 Jingba Road, Zhengzhou, 450000, Henan, China
| | - Lijun Chai
- Department of Breast Surgery, The Second Affiliated Hospital of Zhengzhou University, No.2 Jingba Road, Zhengzhou, 450000, Henan, China.
| | - Jianzhong Chen
- Department of Breast Surgery, The Second Affiliated Hospital of Zhengzhou University, No.2 Jingba Road, Zhengzhou, 450000, Henan, China.
| |
Collapse
|
2
|
Li M. Harnessing atomic force microscopy-based single-cell analysis to advance physical oncology. Microsc Res Tech 2024; 87:631-659. [PMID: 38053519 DOI: 10.1002/jemt.24467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 11/21/2023] [Accepted: 11/23/2023] [Indexed: 12/07/2023]
Abstract
Single-cell analysis is an emerging and promising frontier in the field of life sciences, which is expected to facilitate the exploration of fundamental laws of physiological and pathological processes. Single-cell analysis allows experimental access to cell-to-cell heterogeneity to reveal the distinctive behaviors of individual cells, offering novel opportunities to dissect the complexity of severe human diseases such as cancers. Among the single-cell analysis tools, atomic force microscopy (AFM) is a powerful and versatile one which is able to nondestructively image the fine topographies and quantitatively measure multiple mechanical properties of single living cancer cells in their native states under aqueous conditions with unprecedented spatiotemporal resolution. Over the past few decades, AFM has been widely utilized to detect the structural and mechanical behaviors of individual cancer cells during the process of tumor formation, invasion, and metastasis, yielding numerous unique insights into tumor pathogenesis from the biomechanical perspective and contributing much to the field of cancer mechanobiology. Here, the achievements of AFM-based analysis of single cancer cells to advance physical oncology are comprehensively summarized, and challenges and future perspectives are also discussed. RESEARCH HIGHLIGHTS: Achievements of AFM in characterizing the structural and mechanical behaviors of single cancer cells are summarized, and future directions are discussed. AFM is not only capable of visualizing cellular fine structures, but can also measure multiple cellular mechanical properties as well as cell-generated mechanical forces. There is still plenty of room for harnessing AFM-based single-cell analysis to advance physical oncology.
Collapse
Affiliation(s)
- Mi Li
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang, China
- Institutes for Robotics and Intelligent Manufacturing, Chinese Academy of Sciences, Shenyang, China
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
3
|
Lee EH, Chung JW, Sung E, Yoon BH, Jeon M, Park S, Chun SY, Lee JN, Kim BS, Kim HT, Kim TH, Choi SH, Yoo ES, Kwon TG, Kang HW, Kim WJ, Yun SJ, Lee S, Ha YS. Anti-Metastatic Effect of Pyruvate Dehydrogenase Kinase 4 Inhibition in Bladder Cancer via the ERK, SRC, and JNK Pathways. Int J Mol Sci 2022; 23:13240. [PMID: 36362028 PMCID: PMC9658024 DOI: 10.3390/ijms232113240] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 10/27/2022] [Accepted: 10/28/2022] [Indexed: 12/30/2023] Open
Abstract
Bladder cancer is a common global cancer with a high percentage of metastases and high mortality rate. Thus, it is necessary to identify new biomarkers that can be helpful in diagnosis. Pyruvate dehydrogenase kinase 4 (PDK4) belongs to the PDK family and plays an important role in glucose utilization in living organisms. In the present study, we evaluated the role of PDK4 in bladder cancer and its related protein changes. First, we observed elevated PDK4 expression in high-grade bladder cancers. To screen for changes in PDK4-related proteins in bladder cancer, we performed a comparative proteomic analysis using PDK4 knockdown cells. In bladder cancer cell lines, PDK4 silencing resulted in a lower rate of cell migration and invasion. In addition, a PDK4 knockdown xenograft model showed reduced bladder cancer growth in nude mice. Based on our results, PDK4 plays a critical role in the metastasis and growth of bladder cancer cells through changes in ERK, SRC, and JNK.
Collapse
Affiliation(s)
- Eun Hye Lee
- Joint Institute of Regenerative Medicine, Kyungpook National University, Daegu 41566, Korea
| | - Jae-Wook Chung
- Department of Urology, School of Medicine, Kyungpook National University, Daegu 41405, Korea
| | - Eunji Sung
- BK21 FOUR Community-Based Intelligent Novel Drug Discovery Education Unit, College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea
| | - Bo Hyun Yoon
- Joint Institute of Regenerative Medicine, Kyungpook National University, Daegu 41566, Korea
| | - Minji Jeon
- Joint Institute of Regenerative Medicine, Kyungpook National University, Daegu 41566, Korea
| | - Song Park
- Division of Biotechnology, Daegu Gyeongbuk Institute of Science and Technology, Daegu 42988, Korea
| | - So Young Chun
- BioMedical Research Institute, Kyungpook National University Hospital, Daegu 41944, Korea
| | - Jun Nyung Lee
- Department of Urology, School of Medicine, Kyungpook National University, Daegu 41405, Korea
| | - Bum Soo Kim
- Department of Urology, School of Medicine, Kyungpook National University, Daegu 41405, Korea
| | - Hyun Tae Kim
- Department of Urology, School of Medicine, Kyungpook National University, Daegu 41405, Korea
| | - Tae Hwan Kim
- Department of Urology, School of Medicine, Kyungpook National University, Daegu 41405, Korea
| | - Seock Hwan Choi
- Department of Urology, School of Medicine, Kyungpook National University, Daegu 41405, Korea
| | - Eun Sang Yoo
- Department of Urology, School of Medicine, Kyungpook National University, Daegu 41405, Korea
| | - Tae Gyun Kwon
- Department of Urology, School of Medicine, Kyungpook National University, Daegu 41405, Korea
| | - Ho Won Kang
- Department of Urology, College of Medicine, Chungbuk National University, Cheongju 28644, Korea
- Department of Urology, Chungbuk National University Hospital, Cheongju 28644, Korea
| | - Wun-Jae Kim
- Department of Urology, College of Medicine, Chungbuk National University, Cheongju 28644, Korea
- Institute of Urotech, Cheongju 28120, Korea
| | - Seok Joong Yun
- Department of Urology, College of Medicine, Chungbuk National University, Cheongju 28644, Korea
- Department of Urology, Chungbuk National University Hospital, Cheongju 28644, Korea
| | - Sangkyu Lee
- BK21 FOUR Community-Based Intelligent Novel Drug Discovery Education Unit, College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea
| | - Yun-Sok Ha
- Department of Urology, School of Medicine, Kyungpook National University, Daegu 41405, Korea
| |
Collapse
|
4
|
Proteomics-Based Identification of Dysregulated Proteins in Breast Cancer. Proteomes 2022; 10:proteomes10040035. [PMID: 36278695 PMCID: PMC9590004 DOI: 10.3390/proteomes10040035] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 10/10/2022] [Accepted: 10/18/2022] [Indexed: 11/18/2022] Open
Abstract
Immunohistochemistry (IHC) is still widely used as a morphology-based assay for in situ analysis of target proteins as specific tumor antigens. However, as a very heterogeneous collection of neoplastic diseases, breast cancer (BC) requires an accurate identification and characterization of larger panels of candidate biomarkers, beyond ER, PR, and HER2 proteins, for diagnosis and personalized treatment, without the limited availability of antibodies that are required to identify specific proteins. Top-down, middle-down, and bottom-up mass spectrometry (MS)-based proteomics approaches complement traditional histopathological tissue analysis to examine expression, modification, and interaction of hundreds to thousands of proteins simultaneously. In this review, we discuss the proteomics-based identification of dysregulated proteins in BC that are essential for the following issues: discovery and validation of new biomarkers by analysis of solid and liquid/non-invasive biopsies, cell lines, organoids and xenograft models; identification of panels of biomarkers for early detection and accurate discrimination between cancer, benign and normal tissues; identification of subtype-specific and stage-specific protein expression profiles in BC grading and measurement of disease progression; characterization of new subtypes of BC; characterization and quantitation of post-translational modifications (PTMs) and aberrant protein-protein interactions (PPI) involved in tumor development; characterization of the global remodeling of BC tissue homeostasis, diagnosis and prognostic information; and deciphering of molecular functions, biological processes and mechanisms through which the dysregulated proteins cause tumor initiation, invasion, and treatment resistance.
Collapse
|
5
|
Wang Q, Karvelsson ST, Johannsson F, Vilhjalmsson AI, Hagen L, de Miranda Fonseca D, Sharma A, Slupphaug G, Rolfsson O. UDP-glucose dehydrogenase expression is upregulated following EMT and differentially affects intracellular glycerophosphocholine and acetylaspartate levels in breast mesenchymal cell lines. Mol Oncol 2021; 16:1816-1840. [PMID: 34942055 PMCID: PMC9067156 DOI: 10.1002/1878-0261.13172] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 11/04/2021] [Accepted: 12/21/2021] [Indexed: 11/07/2022] Open
Abstract
Metabolic rewiring is one of the indispensable drivers of epithelial-mesenchymal transition (EMT) involved in breast cancer metastasis. In this study, we explored the metabolic changes during spontaneous EMT in three separately established breast EMT cell models using a proteomics approach supported by metabolomic analysis. We identified common proteomic changes, including in the expression of CDH1, CDH2, VIM, LGALS1, SERPINE1, PKP3, ATP2A2, JUP, MTCH2, RPL26L1 and PLOD2. Consistently altered metabolic enzymes included: FDFT1, SORD, TSTA3 and UDP-glucose dehydrogenase (UGDH). Of these, UGDH was most prominently altered and has previously been associated with breast cancer patient survival. siRNA-mediated knockdown of UGDH resulted in delayed cell proliferation and dampened invasive potential of mesenchymal cells, and downregulated expression of the EMT transcription factor SNAI1. Metabolomic analysis revealed that siRNA-mediated knockdown of UGDH decreased intracellular glycerophosphocholine (GPC), whereas levels of acetylaspartate (NAA) increased. Finally, our data suggested that platelet-derived growth factor receptor beta (PDGFRB) signaling was activated in mesenchymal cells. siRNA-mediated knockdown of PDGFRB downregulated UGDH expression, potentially via NFkB-p65. Our results support an unexplored relationship between UGDH and GPC, both of which have previously been independently associated with breast cancer progression.
Collapse
Affiliation(s)
- Qiong Wang
- Center for Systems Biology, Biomedical Center, Faculty of Medicine, School of Health Sciences, University of Iceland, Sturlugata 8, 101, Reykjavik, Iceland
| | - Sigurdur Trausti Karvelsson
- Center for Systems Biology, Biomedical Center, Faculty of Medicine, School of Health Sciences, University of Iceland, Sturlugata 8, 101, Reykjavik, Iceland
| | - Freyr Johannsson
- Center for Systems Biology, Biomedical Center, Faculty of Medicine, School of Health Sciences, University of Iceland, Sturlugata 8, 101, Reykjavik, Iceland
| | - Arnar Ingi Vilhjalmsson
- Center for Systems Biology, Biomedical Center, Faculty of Medicine, School of Health Sciences, University of Iceland, Sturlugata 8, 101, Reykjavik, Iceland
| | - Lars Hagen
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, NTNU, N-7491, Trondheim, Norway.,Clinic of Laboratory Medicine, St. Olavs hospital, Trondheim, Norway.,PROMEC Core Facility for Proteomics and Modomics, Norwegian University of Science and Technology, NTNU, and the Central Norway Regional Health Authority Norway, Norway
| | - Davi de Miranda Fonseca
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, NTNU, N-7491, Trondheim, Norway.,Clinic of Laboratory Medicine, St. Olavs hospital, Trondheim, Norway.,PROMEC Core Facility for Proteomics and Modomics, Norwegian University of Science and Technology, NTNU, and the Central Norway Regional Health Authority Norway, Norway
| | - Animesh Sharma
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, NTNU, N-7491, Trondheim, Norway.,Clinic of Laboratory Medicine, St. Olavs hospital, Trondheim, Norway.,PROMEC Core Facility for Proteomics and Modomics, Norwegian University of Science and Technology, NTNU, and the Central Norway Regional Health Authority Norway, Norway
| | - Geir Slupphaug
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, NTNU, N-7491, Trondheim, Norway.,Clinic of Laboratory Medicine, St. Olavs hospital, Trondheim, Norway.,PROMEC Core Facility for Proteomics and Modomics, Norwegian University of Science and Technology, NTNU, and the Central Norway Regional Health Authority Norway, Norway
| | - Ottar Rolfsson
- Center for Systems Biology, Biomedical Center, Faculty of Medicine, School of Health Sciences, University of Iceland, Sturlugata 8, 101, Reykjavik, Iceland
| |
Collapse
|
6
|
Wang Q, Karvelsson ST, Kotronoulas A, Gudjonsson T, Halldorsson S, Rolfsson O. Glutamine-fructose-6-phosphate transaminase 2 (GFPT2) is upregulated in breast epithelial-mesenchymal transition and responds to oxidative stress. Mol Cell Proteomics 2021; 21:100185. [PMID: 34923141 PMCID: PMC8803663 DOI: 10.1016/j.mcpro.2021.100185] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 10/20/2021] [Accepted: 12/13/2021] [Indexed: 12/02/2022] Open
Abstract
Breast cancer cells that have undergone partial epithelial–mesenchymal transition (EMT) are believed to be more invasive than cells that have completed EMT. To study metabolic reprogramming in different mesenchymal states, we analyzed protein expression following EMT in the breast epithelial cell model D492 with single-shot LFQ supported by a SILAC proteomics approach. The D492 EMT cell model contains three cell lines: the epithelial D492 cells, the mesenchymal D492M cells, and a partial mesenchymal, tumorigenic variant of D492 that overexpresses the oncogene HER2. The analysis classified the D492 and D492M cells as basal-like and D492HER2 as claudin-low. Comparative analysis of D492 and D492M to tumorigenic D492HER2 differentiated metabolic markers of migration from those of invasion. Glutamine-fructose-6-phosphate transaminase 2 (GFPT2) was one of the top dysregulated enzymes in D492HER2. Gene expression analysis of the cancer genome atlas showed that GFPT2 expression was a characteristic of claudin-low breast cancer. siRNA-mediated knockdown of GFPT2 influenced the EMT marker vimentin and both cell growth and invasion in vitro and was accompanied by lowered metabolic flux through the hexosamine biosynthesis pathway (HBP). Knockdown of GFPT2 decreased cystathionine and sulfide:quinone oxidoreductase (SQOR) in the transsulfuration pathway that regulates H2S production and mitochondrial homeostasis. Moreover, GFPT2 was within the regulation network of insulin and EGF, and its expression was regulated by reduced glutathione (GSH) and suppressed by the oxidative stress regulator GSK3-β. Our results demonstrate that GFPT2 controls growth and invasion in the D492 EMT model, is a marker for oxidative stress, and associated with poor prognosis in claudin-low breast cancer. GFPT2 is upregulated following EMT. GFPT2 is a marker for claudin-low breast cancer. GFPT2 affects vimentin, cell proliferation, and cell invasion. GFPT2 responds to oxidative stress. GFPT2 is regulated by insulin and EGF.
Collapse
Affiliation(s)
- Qiong Wang
- Center for Systems Biology, Biomedical Center, Faculty of Medicine, School of Health Sciences, University of Iceland, Sturlugata 8, 101 Reykjavik, Iceland
| | - Sigurdur Trausti Karvelsson
- Center for Systems Biology, Biomedical Center, Faculty of Medicine, School of Health Sciences, University of Iceland, Sturlugata 8, 101 Reykjavik, Iceland
| | - Aristotelis Kotronoulas
- Center for Systems Biology, Biomedical Center, Faculty of Medicine, School of Health Sciences, University of Iceland, Sturlugata 8, 101 Reykjavik, Iceland
| | - Thorarinn Gudjonsson
- Stem Cell Research Unit, Biomedical Center, Department of Anatomy, Faculty of Medicine, School of Health Sciences, University of Iceland, Vatnsmyrarvegi 16, 101 Reykjavík, Iceland
| | - Skarphedinn Halldorsson
- Center for Systems Biology, Biomedical Center, Faculty of Medicine, School of Health Sciences, University of Iceland, Sturlugata 8, 101 Reykjavik, Iceland
| | - Ottar Rolfsson
- Center for Systems Biology, Biomedical Center, Faculty of Medicine, School of Health Sciences, University of Iceland, Sturlugata 8, 101 Reykjavik, Iceland.
| |
Collapse
|
7
|
González-González R, Ortiz-Sarabia G, Molina-Frechero N, Salas-Pacheco JM, Salas-Pacheco SM, Lavalle-Carrasco J, López-Verdín S, Tremillo-Maldonado O, Bologna-Molina R. Epithelial-Mesenchymal Transition Associated with Head and Neck Squamous Cell Carcinomas: A Review. Cancers (Basel) 2021; 13:3027. [PMID: 34204259 PMCID: PMC8234594 DOI: 10.3390/cancers13123027] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 06/09/2021] [Accepted: 06/15/2021] [Indexed: 02/07/2023] Open
Abstract
Head and neck squamous cell carcinomas (HNSCCs) are aggressive, recurrent, and metastatic neoplasms with a high occurrence around the world and can lead to death when not treated appropriately. Several molecules and signaling pathways are involved in the malignant conversion process. Epithelial-mesenchymal transition (EMT) has been described in HNSCCs, a major type of aggressive carcinoma. EMT describes the development of epithelial cells into mesenchymal cells, which depends on several molecular interactions and signaling pathways that facilitate mesenchymal conversion. This is related to interactions with the microenvironment of the tumor, hypoxia, growth factors, matrix metalloproteinases, and the presence of viral infections. In this review, we focus on the main molecules related to EMT, their interactions with the tumor microenvironment, plasticity phenomena, epigenetic regulation, hypoxia, inflammation, their relationship with immune cells, and the inhibition of EMT in the context of HNSCCs.
Collapse
Affiliation(s)
- Rogelio González-González
- Department of Research, School of Dentistry, Universidad Juárez del Estado de Durango, Durango 34000, Mexico; (R.G.-G.); (G.O.-S.); (O.T.-M.)
| | - Gamaliel Ortiz-Sarabia
- Department of Research, School of Dentistry, Universidad Juárez del Estado de Durango, Durango 34000, Mexico; (R.G.-G.); (G.O.-S.); (O.T.-M.)
| | - Nelly Molina-Frechero
- Xochimilco Unit, Department of Health Care, Universidad Autónoma Metropolitana (UAM) Xochimilco, Mexico City 04960, Mexico; (N.M.-F.); (J.L.-C.)
| | - José Manuel Salas-Pacheco
- Scientific Research Institute, Universidad Juárez del Estado de Durango, Avenida Universidad S/N, Durango 34000, Mexico; (J.M.S.-P.); (S.M.S.-P.)
| | - Sergio Manuel Salas-Pacheco
- Scientific Research Institute, Universidad Juárez del Estado de Durango, Avenida Universidad S/N, Durango 34000, Mexico; (J.M.S.-P.); (S.M.S.-P.)
| | - Jesús Lavalle-Carrasco
- Xochimilco Unit, Department of Health Care, Universidad Autónoma Metropolitana (UAM) Xochimilco, Mexico City 04960, Mexico; (N.M.-F.); (J.L.-C.)
| | - Sandra López-Verdín
- Health Science Center, Dentistry Research Institute, Universidad de Guadalajara, Guadalajara 4430, Mexico;
| | - Omar Tremillo-Maldonado
- Department of Research, School of Dentistry, Universidad Juárez del Estado de Durango, Durango 34000, Mexico; (R.G.-G.); (G.O.-S.); (O.T.-M.)
| | - Ronell Bologna-Molina
- Department of Research, School of Dentistry, Universidad Juárez del Estado de Durango, Durango 34000, Mexico; (R.G.-G.); (G.O.-S.); (O.T.-M.)
- Molecular Pathology Area, School of Dentistry, Universidad de la República, Montevideo 11600, Uruguay
| |
Collapse
|
8
|
Vergara D, Verri T, Damato M, Trerotola M, Simeone P, Franck J, Fournier I, Salzet M, Maffia M. A Hidden Human Proteome Signature Characterizes the Epithelial Mesenchymal Transition Program. Curr Pharm Des 2020; 26:372-375. [PMID: 31995001 DOI: 10.2174/1381612826666200129091610] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 01/27/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND Molecular changes associated with the initiation of the epithelial to mesenchymal transition (EMT) program involve alterations of large proteome-based networks. The role of protein products mapping to non-coding genomic regions is still unexplored. OBJECTIVE The goal of this study was the identification of an alternative protein signature in breast cancer cellular models with a distinct expression of EMT markers. METHODS We profiled MCF-7 and MDA-MB-231 cells using liquid-chromatography mass/spectrometry (LCMS/ MS) and interrogated the OpenProt database to identify novel predicted isoforms and novel predicted proteins from alternative open reading frames (AltProts). RESULTS Our analysis revealed an AltProt and isoform protein signature capable of classifying the two breast cancer cell lines. Among the most highly expressed alternative proteins, we observed proteins potentially associated with inflammation, metabolism and EMT. CONCLUSION Here, we present an AltProts signature associated with EMT. Further studies will be needed to define their role in cancer progression.
Collapse
Affiliation(s)
- Daniele Vergara
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
| | - Tiziano Verri
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
| | - Marina Damato
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
| | - Marco Trerotola
- Department of Medical, Oral and Biotechnological Sciences, "G.d'Annunzio" University of Chieti-Pescara, Italy
| | - Pasquale Simeone
- Department of Medicine and Aging Sciences, "G.d'Annunzio" University of Chieti-Pescara, Italy; Laboratory of Cytomorphology, Center for Advanced Studies and Technology (CAST), "G.d'Annunzio" University of Chieti-Pescara, Italy
| | - Julien Franck
- University of Lille, Inserm, U-1192, Laboratoire Proteomique, Reponse Inflammatoire et Spectrometrie de Masse-PRISM, F-59000, Lille, France
| | - Isabelle Fournier
- University of Lille, Inserm, U-1192, Laboratoire Proteomique, Reponse Inflammatoire et Spectrometrie de Masse-PRISM, F-59000, Lille, France
| | - Michel Salzet
- University of Lille, Inserm, U-1192, Laboratoire Proteomique, Reponse Inflammatoire et Spectrometrie de Masse-PRISM, F-59000, Lille, France
| | - Michele Maffia
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
| |
Collapse
|
9
|
Li QH, Liu ZZ, Ge YΝ, Liu X, Xie XD, Zheng ZD, Ma YH, Liu B. Small breast epithelial mucin promotes the invasion and metastasis of breast cancer cells via promoting epithelial‑to‑mesenchymal transition. Oncol Rep 2020; 44:509-518. [PMID: 32627029 PMCID: PMC7336452 DOI: 10.3892/or.2020.7640] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 04/23/2020] [Indexed: 01/13/2023] Open
Abstract
The aim of the present study was to observe the influence of the small breast epithelial mucin (MUCL1) (also known as SBEM) gene on migration and invasion ability of breast cancer cells and to explore the potentially involved mechanism. SBEM‑interference plasmid and SBEM‑overexpressing plasmid were constructed. SBEM‑knockdown or SBEM‑overexpressing MCF‑7 and MDA‑MB‑231 breast cancer cells were established by lentivirus‑mediated stable transfection method. The scratch wound‑healing assay and Transwell chamber experiment were used to detect the influence of the SBEM gene on the migration and invasion abilities of MCF‑7 and MDA‑MB‑231 cells. Real‑time PCR (polymerase chain reaction) and western blotting were used to detect the expression of epithelial‑to‑mesenchymal transition (EMT)‑related markers and regulators. The cell morphology was observed after transfection. The SBEM‑knockdown or SBEM‑overexpressing MCF‑7 and MDA‑MB‑231 cells were established successfully. The migration and invasion abilities were decreased after SBEM was downregulated, and were increased after SBEM was overexpressed both in MCF‑7 and MDA‑MB‑231 cell lines. The mRNA and protein expressions of N‑cadherin, Twist and vimentin were elevated following SBEM overexpression, while the expression of E‑cadherin and claudin‑1 were found to be decreased following SBEM overexpression. In conclusion, SBEM has the potential to promote migration and invasion ability of breast cancer cells via promoting epithelial‑to‑mesenchymal transition.
Collapse
Affiliation(s)
- Qiu-Hua Li
- Oncology Department, The Second Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning 110034, P.R. China
| | - Zhao-Zhe Liu
- Oncology Department, General Hospital of Northern Theater Command, Shenyang, Liaoning 110016, P.R. China
| | - Ya-Νan Ge
- Oncology Department, General Hospital of Northern Theater Command, Shenyang, Liaoning 110016, P.R. China
| | - Xing Liu
- Oncology Department, General Hospital of Northern Theater Command, Shenyang, Liaoning 110016, P.R. China
| | - Xiao-Dong Xie
- Oncology Department, General Hospital of Northern Theater Command, Shenyang, Liaoning 110016, P.R. China
| | - Zhen-Dong Zheng
- Oncology Department, General Hospital of Northern Theater Command, Shenyang, Liaoning 110016, P.R. China
| | - Yue-Hai Ma
- Oncology Department, The Second Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning 110034, P.R. China
| | - Bin Liu
- Department of Medical Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning 110042, P.R. China
| |
Collapse
|
10
|
Silvestrini VC, Lanfredi GP, Masson AP, Poersch A, Ferreira GA, Thomé CH, Faça VM. A proteomics outlook towards the elucidation of epithelial-mesenchymal transition molecular events. Mol Omics 2020; 15:316-330. [PMID: 31429845 DOI: 10.1039/c9mo00095j] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The main cause of death in cancer is the spread, or metastasis, of cancer cells to distant organs with consequent tumor formation. Additionally, metastasis is a process that demands special attention, as the cellular transformations make cancer at this stage very difficult or occasionally even impossible to be cured. The main process that converts epithelial tumor cells to mesenchymal-like metastatic cells is the Epithelial to Mesenchymal Transition (EMT). This process allows stationary and polarized epithelial cells, which are connected laterally to several types of junctions as well as the basement membrane, to undergo multiple biochemical changes that enable disruption of cell-cell adherence and apical-basal polarity. Moreover, the cells undergo important reprogramming to remodel the cytoskeleton and acquire mesenchymal characteristics such as enhanced migratory capacity, invasiveness, elevated resistance to apoptosis and a large increase in the production of ECM components. As expected, the alterations of the protein complement are extensive and complex, and thus exploring this by proteomic approaches is of particular interest. Here we review the overall findings of proteome modifications during EMT, mainly focusing on molecular signatures observed in multiple proteomic studies as well as coordinated pathways, cellular processes and their clinical relevance for altered proteins. As a result, an interesting set of proteins is highlighted as potential targets to be further investigated in the context of EMT, metastasis and cancer progression.
Collapse
Affiliation(s)
- Virgínia Campos Silvestrini
- Department of Biochemistry and Immunology - FMRP - University of São Paulo, Av. Bandeirantes, 3900, 14049-900, Ribeirão Preto, SP, Brazil.
| | | | | | | | | | | | | |
Collapse
|
11
|
Expression Analysis of the Mediators of Epithelial to Mesenchymal Transition and Early Risk Assessment of Therapeutic Failure in Laryngeal Carcinoma. JOURNAL OF ONCOLOGY 2019; 2019:5649846. [PMID: 31885577 PMCID: PMC6926423 DOI: 10.1155/2019/5649846] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Revised: 07/26/2019] [Accepted: 08/10/2019] [Indexed: 02/08/2023]
Abstract
Laryngeal squamous cell carcinoma (LSCC) is an aggressive malignancy which lacks early predictors of prognosis. Here, we hypothesized that expression and prognostic characterization of the critical mediators of epithelial to mesenchymal transition (EMT) may provide key information in this regard. Linear regression and multiple correspondence analyses were performed on immunohistochemical data obtained from 20 invasive tumors. Principal component and unsupervised hierarchical clustering were used to analyze the dataset patterns associating with LSCC metastatic profile. Survival and death risk assessments were performed using Kaplan–Meier and hazard ratio tests. Data mining analysis using CHAID decision tree and logistic regression analysis was applied to define the predictive value of the risk factors of tumor aggressiveness. Our analyses showed, that in invasive LSCC tumors, cells associating with a mesenchymal profile were likely to exhibit enhanced NOS2, TGF-β, and IL-17A expression levels, concomitantly to NF-κB nuclear translocation. IHC data deciphering determined that EMT induction was also linked to the enrichment of the tumors with CD68+ populations and IL-10 signal. Strikingly, dataset cluster analysis showed that these signatures could define distinct patterns of invasive tumors, where NOS2 associated with IL-10 expression, and TGF-β and IL-17A signals associated with MMP-9 activation. Decision tree analysis identified IL-17A as a possible predictor of LSCC aggressiveness. Altogether, our results show that distinct immunological patterns would support the acquisition of EMT features in invasive LSCC and suggest that IL-17A may be useful in the early identification of patients “at-risk” of therapeutic failure.
Collapse
|
12
|
Wan Mohd Tajuddin WNB, Lajis NH, Abas F, Othman I, Naidu R. Mechanistic Understanding of Curcumin's Therapeutic Effects in Lung Cancer. Nutrients 2019; 11:E2989. [PMID: 31817718 PMCID: PMC6950067 DOI: 10.3390/nu11122989] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 11/22/2019] [Accepted: 11/30/2019] [Indexed: 12/24/2022] Open
Abstract
Lung cancer is among the most common cancers with a high mortality rate worldwide. Despite the significant advances in diagnostic and therapeutic approaches, lung cancer prognoses and survival rates remain poor due to late diagnosis, drug resistance, and adverse effects. Therefore, new intervention therapies, such as the use of natural compounds with decreased toxicities, have been considered in lung cancer therapy. Curcumin, a natural occurring polyphenol derived from turmeric (Curcuma longa) has been studied extensively in recent years for its therapeutic effects. It has been shown that curcumin demonstrates anti-cancer effects in lung cancer through various mechanisms, including inhibition of cell proliferation, invasion, and metastasis, induction of apoptosis, epigenetic alterations, and regulation of microRNA expression. Several in vitro and in vivo studies have shown that these mechanisms are modulated by multiple molecular targets such as STAT3, EGFR, FOXO3a, TGF-β, eIF2α, COX-2, Bcl-2, PI3KAkt/mTOR, ROS, Fas/FasL, Cdc42, E-cadherin, MMPs, and adiponectin. In addition, limitations, strategies to overcome curcumin bioavailability, and potential side effects as well as clinical trials were also reviewed.
Collapse
Affiliation(s)
- Wan Nur Baitty Wan Mohd Tajuddin
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, Selangor Darul Ehsan 47500, Malaysia; (W.N.B.W.M.T.); (I.O.)
| | - Nordin H. Lajis
- Laboratory of Natural Products, Faculty of Science, Universiti Putra Malaysia, UPM, Serdang 43400, Malaysia; (N.H.L.); (F.A.)
| | - Faridah Abas
- Laboratory of Natural Products, Faculty of Science, Universiti Putra Malaysia, UPM, Serdang 43400, Malaysia; (N.H.L.); (F.A.)
- Department of Food Science, Faculty of Food Science and Technology, Universiti Putra Malaysia, UPM, Serdang 43400, Malaysia
| | - Iekhsan Othman
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, Selangor Darul Ehsan 47500, Malaysia; (W.N.B.W.M.T.); (I.O.)
| | - Rakesh Naidu
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, Selangor Darul Ehsan 47500, Malaysia; (W.N.B.W.M.T.); (I.O.)
| |
Collapse
|
13
|
Vergara D, Simeone P, Damato M, Maffia M, Lanuti P, Trerotola M. The Cancer Microbiota: EMT and Inflammation as Shared Molecular Mechanisms Associated with Plasticity and Progression. JOURNAL OF ONCOLOGY 2019; 2019:1253727. [PMID: 31772577 PMCID: PMC6854237 DOI: 10.1155/2019/1253727] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 08/31/2019] [Indexed: 02/07/2023]
Abstract
With the advent of novel molecular platforms for high-throughput/next-generation sequencing, the communities of commensal and pathogenic microorganisms that inhabit the human body have been defined in depth. In the last decade, the role of microbiota-host interactions in driving human cancer plasticity and malignant progression has been well documented. Germ-free preclinical models provided an invaluable tool to demonstrate that the human microbiota can confer susceptibility to various types of cancer and can also modulate the host response to therapeutic treatments. Of interest, besides the detrimental effects of dysbiosis on cancer etiopathogenesis, specific microorganisms have been shown to exert protective activities against cancer growth. This has strong clinical implications, as restoration of the physiologic microbiota is being rapidly implemented as a novel anticancer therapeutic strategy. Here, we reviewed past and recent literature depicting the role of microbiota-host interactions in modulating key molecular mechanisms that drive human cancer plasticity and lead to malignant progression. We analyzed microbiota-host interactions occurring in the gut as well as in other anatomic sites, such as oral and nasal cavities, lungs, breast, esophagus, stomach, reproductive tract, and skin. We revealed a common ground of biological alterations and pathways modulated by a dysbiotic microbiota and potentially involved in the control of cancer progression. The molecular mechanisms most frequently affected by the pathogenic microorganisms to induce malignant progression involve epithelial-mesenchymal transition- (EMT-) dependent barrier alterations and tumor-promoting inflammation. This evidence may pave the way to better stratify high-risk cancer patients based on unique microenvironmental/microbial signatures and to develop novel, personalized, biological therapies.
Collapse
Affiliation(s)
- Daniele Vergara
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
- Laboratory of Clinical Proteomic, “Giovanni Paolo II” Hospital, ASL-Lecce, Italy
| | - Pasquale Simeone
- Department of Medicine and Aging Sciences, “G.d'Annunzio” University of Chieti-Pescara, Chieti, Italy
- Laboratory of Cytomorphology, Center for Advanced Studies and Technology (CAST), “G.d'Annunzio” University of Chieti-Pescara, Chieti, Italy
| | - Marina Damato
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
- Laboratory of Clinical Proteomic, “Giovanni Paolo II” Hospital, ASL-Lecce, Italy
| | - Michele Maffia
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
- Laboratory of Clinical Proteomic, “Giovanni Paolo II” Hospital, ASL-Lecce, Italy
| | - Paola Lanuti
- Department of Medicine and Aging Sciences, “G.d'Annunzio” University of Chieti-Pescara, Chieti, Italy
- Laboratory of Cytomorphology, Center for Advanced Studies and Technology (CAST), “G.d'Annunzio” University of Chieti-Pescara, Chieti, Italy
| | - Marco Trerotola
- Laboratory of Cancer Pathology, Center for Advanced Studies and Technology (CAST), “G.d'Annunzio” University of Chieti-Pescara, Chieti, Italy
- Department of Medical, Oral and Biotechnological Sciences, “G.d'Annunzio” University of Chieti-Pescara, Chieti, Italy
| |
Collapse
|
14
|
Najahi‐Missaoui W, Quach ND, Jenkins A, Dabke I, Somanath PR, Cummings BS. Effect of P21-activated kinase 1 (PAK-1) inhibition on cancer cell growth, migration, and invasion. Pharmacol Res Perspect 2019; 7:e00518. [PMID: 31516713 PMCID: PMC6728842 DOI: 10.1002/prp2.518] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 07/26/2019] [Indexed: 02/06/2023] Open
Abstract
P21-activated kinase-1 (PAK-1) is a serine/threonine kinase involved in multiple signaling pathways that mediate cellular functions such as cytoskeletal motility, cell proliferation, and survival. PAK-1 expression is altered in various cancers, including prostate and breast. Our recent studies showed that prostate cancer cells expressing higher levels of PAK-1 were resistant to the cytotoxic effects of the PAK-1 inhibitor, inhibitor targeting PAK-1 activation-3 (IPA-3), compared to those with lower expression. This study expanded these findings to other cancers (breast and melanoma) by testing the hypothesis that genetic and pharmacological inhibition of PAK-1 alters cell growth, migration, and invasion in prostate, breast, and skin cancer cell lines. We also tested the specificity of IPA-3 for PAK-1 and the hypothesis that gene silencing of PAK-1 altered the efficacy of sterically stabilized liposomes (SSL) containing IPA-3 (SSL-IPA-3). PAK-1 expression was identified in four different breast cancer cell lines, and in a melanoma cell line. The expression of PAK-1 correlated to the IC50 of IPA-3 as measured by MTT staining. PAK-1 inhibition using shRNA correlated with decreased cell migration and invasion in prostate cancer DU-145 and breast cancer MCF-7 cells. Decreased migration and invasion also correlated to decreased expression of E-cadherin and alterations in C-X-C Chemokine Receptor type 4 and Homing Cell Adhesion Molecule expression. PAK-1 inhibition increased the cytotoxicity of IPA-3, and the cytotoxicity of SSL-IPA-3 to levels comparable to that of free drug. These data demonstrate that both pharmacological and molecular inhibition of PAK-1 decreased growth in prostate, breast, and melanoma cancer cell lines, and increased the toxicity of IPA-3 and its liposomal formulation. These data also show the specificity of IPA-3 for PAK-1, are some of the first data suggesting that IPA-3 is a therapeutic treatment for breast cancer and melanoma, and demonstrate the efficacy of liposome-encapsulated IPA-3 in breast cancer cells.
Collapse
Affiliation(s)
- Wided Najahi‐Missaoui
- Department of Pharmaceutical and Biomedical Sciences, College of PharmacyUniversity of GeorgiaAthensGAUSA
| | - Nhat D. Quach
- Department of Pharmaceutical and Biomedical Sciences, College of PharmacyUniversity of GeorgiaAthensGAUSA
- Present address:
Department of Molecular Pharmacology, Physiology, & BiotechnologyBrown UniversityProvidenceRIUSA
| | - Amber Jenkins
- Department of Pharmaceutical and Biomedical Sciences, College of PharmacyUniversity of GeorgiaAthensGAUSA
- Present address:
Cancer Center of Middle GeorgiaDublinGAUSA
| | - Isha Dabke
- Department of Pharmaceutical and Biomedical Sciences, College of PharmacyUniversity of GeorgiaAthensGAUSA
- Present address:
Medical College of GeorgiaAugustaGAUSA
| | - Payaningal R. Somanath
- Clinical and Experimental Therapeutics, College of PharmacyUniversity of Georgia and Charlie Norwood VA Medical CenterAugustaGAUSA
- Department of Medicine, Vascular Biology Center and Cancer CenterGeorgia Regents UniversityAugustaGAUSA
| | - Brian S. Cummings
- Department of Pharmaceutical and Biomedical Sciences, College of PharmacyUniversity of GeorgiaAthensGAUSA
- Interdisciplinary Toxicology ProgramUniversity of GeorgiaAthensGAUSA
| |
Collapse
|
15
|
Trujillo-de Santiago G, Flores-Garza BG, Tavares-Negrete JA, Lara-Mayorga IM, González-Gamboa I, Zhang YS, Rojas-Martínez A, Ortiz-López R, Álvarez MM. The Tumor-on-Chip: Recent Advances in the Development of Microfluidic Systems to Recapitulate the Physiology of Solid Tumors. MATERIALS (BASEL, SWITZERLAND) 2019; 12:E2945. [PMID: 31514390 PMCID: PMC6766252 DOI: 10.3390/ma12182945] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 09/03/2019] [Accepted: 09/04/2019] [Indexed: 12/29/2022]
Abstract
The ideal in vitro recreation of the micro-tumor niche-although much needed for a better understanding of cancer etiology and development of better anticancer therapies-is highly challenging. Tumors are complex three-dimensional (3D) tissues that establish a dynamic cross-talk with the surrounding tissues through complex chemical signaling. An extensive body of experimental evidence has established that 3D culture systems more closely recapitulate the architecture and the physiology of human solid tumors when compared with traditional 2D systems. Moreover, conventional 3D culture systems fail to recreate the dynamics of the tumor niche. Tumor-on-chip systems, which are microfluidic devices that aim to recreate relevant features of the tumor physiology, have recently emerged as powerful tools in cancer research. In tumor-on-chip systems, the use of microfluidics adds another dimension of physiological mimicry by allowing a continuous feed of nutrients (and pharmaceutical compounds). Here, we discuss recently published literature related to the culture of solid tumor-like tissues in microfluidic systems (tumor-on-chip devices). Our aim is to provide the readers with an overview of the state of the art on this particular theme and to illustrate the toolbox available today for engineering tumor-like structures (and their environments) in microfluidic devices. The suitability of tumor-on-chip devices is increasing in many areas of cancer research, including the study of the physiology of solid tumors, the screening of novel anticancer pharmaceutical compounds before resourcing to animal models, and the development of personalized treatments. In the years to come, additive manufacturing (3D bioprinting and 3D printing), computational fluid dynamics, and medium- to high-throughput omics will become powerful enablers of a new wave of more sophisticated and effective tumor-on-chip devices.
Collapse
Affiliation(s)
- Grissel Trujillo-de Santiago
- Centro de Biotecnología-FEMSA, Tecnologico de Monterrey, Monterrey, Nuevo León CP 64849, Mexico.
- Departamento de Ingeniería Mecátrónica y Eléctrica, Tecnologico de Monterrey, Monterrey, Nuevo León CP 64849, Mexico.
| | | | | | - Itzel Montserrat Lara-Mayorga
- Centro de Biotecnología-FEMSA, Tecnologico de Monterrey, Monterrey, Nuevo León CP 64849, Mexico
- Departamento de Ingeniería Mecátrónica y Eléctrica, Tecnologico de Monterrey, Monterrey, Nuevo León CP 64849, Mexico
| | - Ivonne González-Gamboa
- Centro de Biotecnología-FEMSA, Tecnologico de Monterrey, Monterrey, Nuevo León CP 64849, Mexico
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Augusto Rojas-Martínez
- Centro de Investigación y Transferencia en Salud, Hospital San José, Tecnologico de Monterrey, Monterrey, Nuevo León CP 64849, Mexico
| | - Rocío Ortiz-López
- Centro de Investigación y Transferencia en Salud, Hospital San José, Tecnologico de Monterrey, Monterrey, Nuevo León CP 64849, Mexico
| | - Mario Moisés Álvarez
- Centro de Biotecnología-FEMSA, Tecnologico de Monterrey, Monterrey, Nuevo León CP 64849, Mexico.
| |
Collapse
|
16
|
Dang DK, Makena MR, Llongueras JP, Prasad H, Ko M, Bandral M, Rao R. A Ca 2+-ATPase Regulates E-cadherin Biogenesis and Epithelial-Mesenchymal Transition in Breast Cancer Cells. Mol Cancer Res 2019; 17:1735-1747. [PMID: 31076498 DOI: 10.1158/1541-7786.mcr-19-0070] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 03/25/2019] [Accepted: 05/08/2019] [Indexed: 01/01/2023]
Abstract
Progression of benign tumors to invasive, metastatic cancer is accompanied by the epithelial-to-mesenchymal transition (EMT), characterized by loss of the cell-adhesion protein E-cadherin. Although silencing mutations and transcriptional repression of the E-cadherin gene have been widely studied, not much is known about posttranslational regulation of E-cadherin in tumors. We show that E-cadherin is tightly coexpressed with the secretory pathway Ca2+-ATPase isoform 2, SPCA2 (ATP2C2), in breast tumors. Loss of SPCA2 impairs surface expression of E-cadherin and elicits mesenchymal gene expression through disruption of cell adhesion in tumorspheres and downstream Hippo-YAP signaling. Conversely, ectopic expression of SPCA2 in triple-negative breast cancer elevates baseline Ca2+ and YAP phosphorylation, enhances posttranslational expression of E-cadherin, and suppresses mesenchymal gene expression. Thus, loss of SPCA2 phenocopies loss of E-cadherin in the Hippo signaling pathway and EMT-MET transitions, consistent with a functional role for SPCA2 in E-cadherin biogenesis. Furthermore, we show that SPCA2 suppresses invasive phenotypes, including cell migration in vitro and tumor metastasis in vivo. Based on these findings, we propose that SPCA2 functions as a key regulator of EMT and may be a potential therapeutic target for treatment of metastatic cancer. IMPLICATIONS: Posttranslational control of E-cadherin and the Hippo pathway by calcium signaling regulates EMT in breast cancer cells.
Collapse
Affiliation(s)
- Donna K Dang
- Department of Physiology, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Monish Ram Makena
- Department of Physiology, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - José P Llongueras
- Department of Physiology, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Hari Prasad
- Department of Physiology, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Myungjun Ko
- Department of Physiology, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Manuj Bandral
- Department of Physiology, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Rajini Rao
- Department of Physiology, The Johns Hopkins University School of Medicine, Baltimore, Maryland.
| |
Collapse
|
17
|
Rosso M, Lapyckyj L, Besso MJ, Monge M, Reventós J, Canals F, Quevedo Cuenca JO, Matos ML, Vazquez-Levin MH. Characterization of the molecular changes associated with the overexpression of a novel epithelial cadherin splice variant mRNA in a breast cancer model using proteomics and bioinformatics approaches: identification of changes in cell metabolism and an increased expression of lactate dehydrogenase B. Cancer Metab 2019; 7:5. [PMID: 31086659 PMCID: PMC6507066 DOI: 10.1186/s40170-019-0196-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 04/04/2019] [Indexed: 12/17/2022] Open
Abstract
Background Breast cancer (BC) is the most common female cancer and the leading cause of cancer death in women worldwide. Alterations in epithelial cadherin (E-cadherin) expression and functions are associated to BC, but the underlying molecular mechanisms have not been fully elucidated. We have previously reported a novel human E-cadherin splice variant (E-cadherin variant) mRNA. Stable transfectants in MCF-7 human BC cells (MCF7Ecadvar) depicted fibroblast-like cell morphology, E-cadherin wild-type downregulation, and other molecular changes characteristic of the epithelial-to-mesenchymal transition process, reduced cell-cell adhesion, and increased cell migration and invasion. In this study, a two-dimensional differential gel electrophoresis (2D-DIGE) combined with mass spectrometry (MS) protein identification and bioinformatics analyses were done to characterize biological processes and canonical pathways affected by E-cadherin variant expression. Results By 2D-DIGE and MS analysis, 50 proteins were found differentially expressed (≥ Δ1.5) in MCF7Ecadvar compared to control cells. Validation of transcript expression was done in the ten most overexpressed and underexpressed proteins. Bioinformatics analyses revealed that 39 of the 50 proteins identified had been previously associated to BC. Moreover, metabolic processes were the most affected, and glycolysis the canonical pathway most altered. The lactate dehydrogenase B (LDHB) was the highest overexpressed protein, and transcript levels were higher in MCF7Ecadvar than in control cells. In agreement with these findings, MCF7Ecadvar conditioned media had lower glucose and higher lactate levels than control cells. MCF7Ecadvar cell treatment with 5 mM of the glycolytic inhibitor 2-deoxy-glucose led to decreased cell viability, and modulation of LDHB expression in MCF7Ecadvar cells with a specific small interfering RNA resulted in decreased cell proliferation. Finally, a positive association between expression levels of the E-cadherin variant and LDHB transcripts was demonstrated in 21 human breast tumor tissues, and breast tumor samples with higher Ki67 expression showed higher LDHB mRNA levels. Conclusions Results from this investigation contributed to further characterize molecular changes associated to the novel E-cadherin splice variant expression in BC cells. They also revealed an association between expression of the novel variant and changes related to BC progression and aggressiveness, in particular those associated to cell metabolism.
Collapse
Affiliation(s)
- Marina Rosso
- Laboratorio de Estudios de Interacción Celular en Reproducción y Cáncer, Instituto de Biología y Medicina Experimental (IBYME; CONICET-FIBYME), Vuelta de Obligado 2490, C1428ADN Buenos Aires, Argentina
| | - Lara Lapyckyj
- Laboratorio de Estudios de Interacción Celular en Reproducción y Cáncer, Instituto de Biología y Medicina Experimental (IBYME; CONICET-FIBYME), Vuelta de Obligado 2490, C1428ADN Buenos Aires, Argentina
| | - María José Besso
- Laboratorio de Estudios de Interacción Celular en Reproducción y Cáncer, Instituto de Biología y Medicina Experimental (IBYME; CONICET-FIBYME), Vuelta de Obligado 2490, C1428ADN Buenos Aires, Argentina
| | - Marta Monge
- 2Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Jaume Reventós
- 3Departament de Ciències Bàsiques, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Francesc Canals
- 2Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Jorge Oswaldo Quevedo Cuenca
- Laboratorio de Estudios de Interacción Celular en Reproducción y Cáncer, Instituto de Biología y Medicina Experimental (IBYME; CONICET-FIBYME), Vuelta de Obligado 2490, C1428ADN Buenos Aires, Argentina
| | - María Laura Matos
- Laboratorio de Estudios de Interacción Celular en Reproducción y Cáncer, Instituto de Biología y Medicina Experimental (IBYME; CONICET-FIBYME), Vuelta de Obligado 2490, C1428ADN Buenos Aires, Argentina
| | - Mónica Hebe Vazquez-Levin
- Laboratorio de Estudios de Interacción Celular en Reproducción y Cáncer, Instituto de Biología y Medicina Experimental (IBYME; CONICET-FIBYME), Vuelta de Obligado 2490, C1428ADN Buenos Aires, Argentina
| |
Collapse
|
18
|
Schuhmacher D, Sontag JM, Sontag E. Protein Phosphatase 2A: More Than a Passenger in the Regulation of Epithelial Cell-Cell Junctions. Front Cell Dev Biol 2019; 7:30. [PMID: 30895176 PMCID: PMC6414416 DOI: 10.3389/fcell.2019.00030] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 02/22/2019] [Indexed: 12/17/2022] Open
Abstract
Cell–cell adhesion plays a key role in the maintenance of the epithelial barrier and apicobasal cell polarity, which is crucial for homeostasis. Disruption of cell–cell adhesion is a hallmark of numerous pathological conditions, including invasive carcinomas. Adhesion between apposing cells is primarily regulated by three types of junctional structures: desmosomes, adherens junctions, and tight junctions. Cell junctional structures are highly regulated multiprotein complexes that also serve as signaling platforms to control epithelial cell function. The biogenesis, integrity, and stability of cell junctions is controlled by complex regulatory interactions with cytoskeletal and polarity proteins, as well as modulation of key component proteins by phosphorylation/dephosphorylation processes. Not surprisingly, many essential signaling molecules, including protein Ser/Thr phosphatase 2A (PP2A) are associated with intercellular junctions. Here, we examine how major PP2A enzymes regulate epithelial cell–cell junctions, either directly by associating with and dephosphorylating component proteins, or indirectly by affecting signaling pathways that control junctional integrity and cytoskeletal dynamics. PP2A deregulation has severe consequences on the stability and functionality of these structures, and disruption of cell–cell adhesion and cell polarity likely contribute to the link between PP2A dysfunction and human carcinomas.
Collapse
Affiliation(s)
- Diana Schuhmacher
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, Australia
| | - Jean-Marie Sontag
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, Australia.,Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Estelle Sontag
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, Australia.,Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| |
Collapse
|
19
|
Lunetti P, Di Giacomo M, Vergara D, De Domenico S, Maffia M, Zara V, Capobianco L, Ferramosca A. Metabolic reprogramming in breast cancer results in distinct mitochondrial bioenergetics between luminal and basal subtypes. FEBS J 2019; 286:688-709. [PMID: 30657636 DOI: 10.1111/febs.14756] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 12/15/2018] [Accepted: 01/14/2019] [Indexed: 12/13/2022]
Abstract
Mitochondrial dysfunction is a key feature of cancer and is frequently associated with increased aggressiveness and metastatic potential. Recent evidence has brought to light a metabolic rewiring that takes place during the epithelial-to-mesenchymal transition (EMT), a process that drives the invasive capability of malignant tumors, and highlights a mechanistic link between mitochondrial dysfunction and EMT that has been only partially investigated. In this study, we characterized mitochondrial function and bioenergetic status of cultured human breast cancer cell lines, including luminal-like and basal-like subtypes. Through a combination of biochemical and functional studies, we demonstrated that basal-like cell lines exhibit impaired, but not completely inactive, mitochondrial function, and rely on a consequent metabolic switch to glycolysis to support their ATP demand. These altered metabolic activities are linked to modifications of key electron transport chain proteins and a significant increase in levels of reactive oxygen species compared to luminal cells. Furthermore, we observed that the stable knockdown of EMT markers caused functional changes in mitochondria that result in acquisition of a hybrid glycolysis/OXPHOS phenotype in cancer cells as a means to sustain their metabolic demand.
Collapse
Affiliation(s)
- Paola Lunetti
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
| | - Mariangela Di Giacomo
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
| | - Daniele Vergara
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
| | - Stefania De Domenico
- Institute of Food Production Sciences, C.N.R. Unit of Lecce, Italy.,Biotecgen, c/o Department of Biological and Environmental Sciences and Technologies, Lecce, Italy
| | - Michele Maffia
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
| | - Vincenzo Zara
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
| | - Loredana Capobianco
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
| | - Alessandra Ferramosca
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
| |
Collapse
|
20
|
Cascione M, De Matteis V, Toma CC, Leporatti S. Morphomechanical Alterations Induced by Transforming Growth Factor-β1 in Epithelial Breast Cancer Cells. Cancers (Basel) 2018; 10:cancers10070234. [PMID: 30012949 PMCID: PMC6071091 DOI: 10.3390/cancers10070234] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 07/11/2018] [Accepted: 07/13/2018] [Indexed: 12/11/2022] Open
Abstract
The Epithelial to mesenchymal transition (EMT) is the process that drives epithelial tumor cells to acquire an invasive phenotype. The role of transforming growth factor-β1 (TGF-β1) in EMT is still debated. We used confocal laser scanning microscopy and scanning force spectroscopy to perform a morphomechanical analysis on epithelial breast cancer cells (MCF-7), comparing them before and after TGF-β1 exogenous stimulation (5 ng/mL for 48 h). After TGF-β1 treatment, loss of cell⁻cell adherence (mainly due to the reduction of E-cadherin expression of about 24%) and disaggregation of actin cortical fibers were observed in treated MCF-7. In addition, TGF-β1 induced an alteration of MCF-7 nuclei morphology as well as a decrease in the Young's modulus, owing to a rearrangement that involved the cytoskeletal networks and the nuclear region. These relevant variations in morphological features and mechanical properties, elicited by TGF-β1, suggested an increased capacity of MCF-7 to migrate, which was confirmed by a wound healing assay. By means of our biophysical approach, we highlighted the malignant progression of breast cancer cells induced by TGF-β1 exposure. We are confirming TGF-β1's role in EMT by means of morphomechanical evidence that could represent a turning point in understanding the molecular mechanisms involved in cancer progression.
Collapse
Affiliation(s)
- Mariafrancesca Cascione
- Dipartimento di Scienze Biomediche e Oncologia Umana, Università degli Studi di Bari "Aldo Moro", p.zza G. Cesare, c/o Policlinico, 70124 Bari, Italy.
| | - Valeria De Matteis
- Dipartimento di Matematica e Fisica "E. De Giorgi", Università del Salento, Via Monteroni, 73100 Lecce, Italy.
| | - Chiara C Toma
- Dipartimento di Matematica e Fisica "E. De Giorgi", Università del Salento, Via Monteroni, 73100 Lecce, Italy.
| | - Stefano Leporatti
- CNR Nanotec-Istituto di Nanotecnologia, Via Monteroni, c/o Campus Ecotekne, 73100 Lecce, Italy.
| |
Collapse
|
21
|
An SPR based immunoassay for the sensitive detection of the soluble epithelial marker E-cadherin. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2018; 14:1963-1971. [PMID: 29902526 DOI: 10.1016/j.nano.2018.05.018] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 05/04/2018] [Accepted: 05/28/2018] [Indexed: 11/20/2022]
Abstract
Protein biomarkers are important diagnostic tools for cancer and several other diseases. To be validated in a clinical context, a biomarker should satisfy some requirements including the ability to provide reliable information on a pathological state by measuring its expression levels. In parallel, the development of an approach capable of detecting biomarkers with high sensitivity and specificity would be ideally suited for clinical applications. Here, we performed an immune-based label free assay using Surface Plasmon Resonance (SPR)-based detection of the soluble form of E-cadherin, a cell-cell contact protein that is involved in the maintaining of tissue integrity. With this approach, we obtained a specific and quantitative detection of E-cadherin from a few hundred microliters of serum of breast cancer patients by obtaining a 10-fold enhancement in the detection limit over a traditional colorimetric ELISA.
Collapse
|
22
|
Vergara D, De Domenico S, Tinelli A, Stanca E, Del Mercato LL, Giudetti AM, Simeone P, Guazzelli N, Lessi M, Manzini C, Santino A, Bellina F, Maffia M. Anticancer effects of novel resveratrol analogues on human ovarian cancer cells. MOLECULAR BIOSYSTEMS 2018; 13:1131-1141. [PMID: 28429008 DOI: 10.1039/c7mb00128b] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Resveratrol, a naturally occurring phytoalexin, has long been known to play an important regulatory role in key functions in cell physiology. This multifunctional role of resveratrol is explained by its ability to interact with several targets of various cell pathways. In the recent past, synthetic chemical modifications have been made in an attempt to enhance the biological effects of resveratrol, including its anti-cancer properties. In this study, we investigated the molecular mechanisms of action of novel trans-restricted analogues of resveratrol in which the C-C double bond of the natural derivative has been replaced by diaryl-substituted imidazole analogues. In ovarian cancer models, the results of in vitro screening revealed that the resveratrol analogues exhibited enhanced anti-proliferative properties compared with resveratrol. We found that the resveratrol analogues also significantly inhibited Akt and MAPK signalling and reduced the migration of IL-6 and EGF-treated cells. Finally, in ascite-derived cancer cells, we demonstrated that the resveratrol analogues reduced the expression of epithelial mesenchymal transition (EMT) markers. Collectively, these findings indicate the enhanced anti-cancer properties of the resveratrol analogues.
Collapse
Affiliation(s)
- Daniele Vergara
- Department of Biological and Environmental Sciences and Technologies, University of Salento, via Monteroni, Lecce, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Stefania DD, Vergara D. The Many-Faced Program of Epithelial-Mesenchymal Transition: A System Biology-Based View. Front Oncol 2017; 7:274. [PMID: 29181337 PMCID: PMC5694026 DOI: 10.3389/fonc.2017.00274] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 10/31/2017] [Indexed: 12/16/2022] Open
Abstract
System biology uses a range of experimental and statistical methods to dissect complex processes that results from alterations in biological models. Given the complexity of the epithelial–mesenchymal transition (EMT) program, system biology represents a promising approach to understanding its fine molecular regulation by the interpretation of high-throughput datasets. Herein, we review recent contributions of system biology applied to the field of EMT physiology and illustrate the importance of these approaches to model biological networks that are perturbed during the transition. Together, these results allowed the definition of an EMT signature across different tumor types, the identification of dysregulated processes and new modules of regulation, making possible to reveal the EMT molecular visage underneath.
Collapse
Affiliation(s)
- De Domenico Stefania
- Biotecgen, Department of Biological and Environmental Sciences and Technologies, Lecce, Italy.,Institute of Sciences of Food Production, National Research Council, Lecce, Italy
| | - Daniele Vergara
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
| |
Collapse
|
24
|
Resveratrol Modulation of Protein Expression in parkin-Mutant Human Skin Fibroblasts: A Proteomic Approach. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:2198243. [PMID: 29138676 PMCID: PMC5613453 DOI: 10.1155/2017/2198243] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 07/19/2017] [Indexed: 01/12/2023]
Abstract
In this study, we investigated by two-dimensional gel electrophoresis (2-DE) and mass spectrometry (MS) analysis the effects of resveratrol treatment on skin primary fibroblasts from a healthy subject and from a parkin-mutant early onset Parkinson's disease patient. Parkin, an E3 ubiquitin ligase, is the most frequently mutated gene in hereditary Parkinson's disease. Functional alteration of parkin leads to impairment of the ubiquitin-proteasome system, resulting in the accumulation of misfolded or aggregated proteins accountable for the neurodegenerative process. The identification of proteins differentially expressed revealed that resveratrol treatment can act on deregulated specific biological process and molecular function such as cellular redox balance and protein homeostasis. In particular, resveratrol was highly effective at restoring the heat-shock protein network and the protein degradation systems. Moreover, resveratrol treatment led to a significant increase in GSH level, reduction of GSSG/GSH ratio, and decrease of reduced free thiol content in patient cells compared to normal fibroblasts. Thus, our findings provide an experimental evidence of the beneficial effects by which resveratrol could contribute to preserve the cellular homeostasis in parkin-mutant fibroblasts.
Collapse
|
25
|
Wang YP, Guo PT, Zhu Z, Zhang H, Xu Y, Chen YZ, Liu F, Ma SP. Pleomorphic adenoma gene like-2 induces epithelial-mesenchymal transition via Wnt/β-catenin signaling pathway in human colorectal adenocarcinoma. Oncol Rep 2017; 37:1961-1970. [PMID: 28259923 PMCID: PMC5367359 DOI: 10.3892/or.2017.5485] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2016] [Accepted: 02/15/2017] [Indexed: 12/20/2022] Open
Abstract
Epithelial-mesenchymal transition (EMT) is a critical step in the acquisition of metastatic and invasive power for tumor cells. Colorectal adenocarcinoma (CRC) is a common cancer where metastasis is directly linked to patient survival. Recent studies show that pleomorphic adenoma gene like-2 (PLAGL2) could induce tumor EMT and is an independent predictive factor associated with poor prognosis in cancer. In the present study, we confirmed the role of PLAGL2 in the prognosis of CRC patients and provide molecular evidence of PLAGL2 promoted EMT in CRC cell line SW480. We found that PLAGL2 expression was upregulated in the paraffin-embedded CRC tissues compared to borderline or benign tissues. Experimental EMT induced by PLAGL2 plasmid transfection proved PLAGL2 protein overexpression could enhance the cell scratch wound-healing and Transwell ability and significantly upregulated mesenchymal marker proteins, N-cadherin and vimentin and concurrently downregulated epithelial marker of E-cadherin. Subsequently, through western blot assay, we found that PLAGL2 could activate the Wnt-signaling component β-catenin in the nuclei. More CRC cell metastasis to the lungs was observed when the PLAGL2 overexpressing SW480 cells were injected into the tail vein of rats, compared with the cell control and PLAGL2 silence group. Our findings indicated that PLAGL2 might be a very upstream key molecule regulating EMT involved in Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Yong-Peng Wang
- Department of Colorectal Surgery, Cancer Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Peng-Tao Guo
- Department of Surgical Oncology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Zhi Zhu
- Department of Surgical Oncology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Hao Zhang
- Department of Colorectal Surgery, Cancer Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Yan Xu
- Department of Surgical Oncology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Yu-Ze Chen
- Department of Colorectal Surgery, Cancer Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Fang Liu
- Department of Colorectal Surgery, Cancer Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Si-Ping Ma
- Department of Colorectal Surgery, Cancer Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| |
Collapse
|
26
|
Jiang Z, Han B, Liu W, Peng Y. Evaluation on biological compatibility of carboxymethyl chitosan as biomaterials for antitumor drug delivery. J Biomater Appl 2017; 31:985-994. [DOI: 10.1177/0885328216688337] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Carboxymethyl-chitosan, a water-soluble derivative of chitosan, has emerged as a promising candidate for biomedical applications due to its excellent water solubility, biodegradation, biocompatibility, hydrating, antimicrobial, and nontoxicity. In this paper, the antitumor proliferation and metastasis was studied in vitro and in vivo to evaluate biocompatibility of carboxymethyl-chitosan as biomaterials for antitumor drug delivery. The results showed that carboxymethyl-chitosan could significantly reduce the clone formation and tumor migration of human cancer cells including kidney cancer cell line OS-RC-2, gastric cancer cell line SGC-7901, colon cancer cell line HT-29, and nonsmall cell lung cancer cell line NCI-H1650 in vitro. Through Lewis tumor-bearing C57BL/6 mouse model, carboxymethyl-chitosan was proved to be able to inhibit solid tumor growth and tumor metastasis to the liver and lung, meanwhile increase the level of tissue inhibitor of metalloproteinase 1 and E-cadherin, and decrease the level of mice blood serum matrix metalloproteinase 9. This study suggested that carboxymethyl-chitosan had certain antimetastasis effect and good biocompatibility and may have a potential application as a synergic antitumor reagent.
Collapse
Affiliation(s)
| | - Baoqin Han
- Ocean University of China, Qingdao, China
| | | | | |
Collapse
|
27
|
Pavan AR, Silva GDBD, Jornada DH, Chiba DE, Fernandes GFDS, Man Chin C, Dos Santos JL. Unraveling the Anticancer Effect of Curcumin and Resveratrol. Nutrients 2016; 8:nu8110628. [PMID: 27834913 PMCID: PMC5133053 DOI: 10.3390/nu8110628] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2016] [Revised: 09/24/2016] [Accepted: 09/27/2016] [Indexed: 12/16/2022] Open
Abstract
Resveratrol and curcumin are natural products with important therapeutic properties useful to treat several human diseases, including cancer. In the last years, the number of studies describing the effect of both polyphenols against cancer has increased; however, the mechanism of action in all of those cases is not completely comprehended. The unspecific effect and the ability to interfere in assays by both polyphenols make this challenge even more difficult. Herein, we analyzed the anticancer activity of resveratrol and curcumin reported in the literature in the last 11 years, in order to unravel the molecular mechanism of action of both compounds. Molecular targets and cellular pathways will be described. Furthermore, we also discussed the ability of these natural products act as chemopreventive and its use in association with other anticancer drugs.
Collapse
Affiliation(s)
- Aline Renata Pavan
- School of Pharmaceutical Sciences, UNESP-Univ Estadual Paulista, Araraquara 14800903, Brazil.
| | | | | | - Diego Eidy Chiba
- School of Pharmaceutical Sciences, UNESP-Univ Estadual Paulista, Araraquara 14800903, Brazil.
| | | | - Chung Man Chin
- School of Pharmaceutical Sciences, UNESP-Univ Estadual Paulista, Araraquara 14800903, Brazil.
| | - Jean Leandro Dos Santos
- School of Pharmaceutical Sciences, UNESP-Univ Estadual Paulista, Araraquara 14800903, Brazil.
| |
Collapse
|
28
|
Vergara D, Simeone P, De Matteis S, Carloni S, Lanuti P, Marchisio M, Miscia S, Rizzello A, Napolitano R, Agostinelli C, Maffia M. Comparative proteomic profiling of Hodgkin lymphoma cell lines. MOLECULAR BIOSYSTEMS 2016; 12:219-32. [PMID: 26588820 DOI: 10.1039/c5mb00654f] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Classical Hodgkin lymphoma (cHL) is a malignancy with complex pathogenesis. The hallmark of the disease is the presence of large mononucleated Hodgkin and bi- or multinucleated Reed/Sternberg (H/RS) cells. The origin of HRS cells in cHL is controversial as these cells show the coexpression of markers of several lineages. Using a proteomic approach, we compared the protein expression profile of cHL models of T- and B-cell derivation to find proteins differentially expressed in these cell lines. A total of 67 proteins were found differentially expressed between the two cell lines including metabolic proteins and proteins involved in the regulation of the cytoskeleton and/or cell migration, which were further validated by western blotting. Additionally, the expression of selected B- and T-cell antigens was also assessed by flow cytometry to reveal significant differences in the expression of different surface markers. Bioinformatics analysis was then applied to our dataset to find enriched pathways and networks, and to identify possible key regulators. In the present study, a proteomic approach was used to compare the protein expression profiles of two cHL cell lines. The identified proteins and/or networks, many of which not previously related to cHL, may be important to better define the pathogenesis of the disease, to identify novel diagnostic markers, and to design new therapeutic strategies.
Collapse
Affiliation(s)
- D Vergara
- Department of Biological and Environmental Sciences and Technologies, University of Salento, via Monteroni, 73100 Lecce, Italy. and Laboratory of Clinical Proteomic, "Giovanni Paolo II" Hospital, ASL-Lecce, Italy.
| | - P Simeone
- Department of Medicine and Aging Science, School of Medicine and Health Science and Unit of Cytomorphology, Research Centre on Aging (Ce.S.I), University "G. d'Annunzio" of Chieti-Pescara, 66100 Chieti, Italy
| | - S De Matteis
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy
| | - S Carloni
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy
| | - P Lanuti
- Department of Medicine and Aging Science, School of Medicine and Health Science and Unit of Cytomorphology, Research Centre on Aging (Ce.S.I), University "G. d'Annunzio" of Chieti-Pescara, 66100 Chieti, Italy
| | - M Marchisio
- Department of Medicine and Aging Science, School of Medicine and Health Science and Unit of Cytomorphology, Research Centre on Aging (Ce.S.I), University "G. d'Annunzio" of Chieti-Pescara, 66100 Chieti, Italy
| | - S Miscia
- Department of Medicine and Aging Science, School of Medicine and Health Science and Unit of Cytomorphology, Research Centre on Aging (Ce.S.I), University "G. d'Annunzio" of Chieti-Pescara, 66100 Chieti, Italy
| | - A Rizzello
- Department of Biological and Environmental Sciences and Technologies, University of Salento, via Monteroni, 73100 Lecce, Italy. and Laboratory of Clinical Proteomic, "Giovanni Paolo II" Hospital, ASL-Lecce, Italy.
| | - R Napolitano
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy
| | - C Agostinelli
- Department of Experimental, Hematopathology and Hematology Sections, Diagnostic and Specialty Medicine, S. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - M Maffia
- Department of Biological and Environmental Sciences and Technologies, University of Salento, via Monteroni, 73100 Lecce, Italy. and Laboratory of Clinical Proteomic, "Giovanni Paolo II" Hospital, ASL-Lecce, Italy.
| |
Collapse
|
29
|
Cascione M, de Matteis V, Rinaldi R, Leporatti S. Atomic force microscopy combined with optical microscopy for cells investigation. Microsc Res Tech 2016; 80:109-123. [DOI: 10.1002/jemt.22696] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 05/24/2016] [Indexed: 12/22/2022]
Affiliation(s)
- Mariafrancesca Cascione
- Dipartimento di Matematica e Fisica “Ennio De Giorgi”; Università del Salento Via Monteroni; 73100 Lecce Italy
- Euromediterranean Center for Nanomaterial Modelling and Technology (ECMT) of Consiglio Nazionale delle Ricerche; Istituto Nanoscienze; Via Arnesano 16, Lecce Italy
| | - Valeria de Matteis
- Dipartimento di Matematica e Fisica “Ennio De Giorgi”; Università del Salento Via Monteroni; 73100 Lecce Italy
| | - Rosaria Rinaldi
- Dipartimento di Matematica e Fisica “Ennio De Giorgi”; Università del Salento Via Monteroni; 73100 Lecce Italy
- Euromediterranean Center for Nanomaterial Modelling and Technology (ECMT) of Consiglio Nazionale delle Ricerche; Istituto Nanoscienze; Via Arnesano 16, Lecce Italy
| | - Stefano Leporatti
- CNR Nantotec-Istituto di Nanotecnologia, Polo di Nanotecnologia c/o Campus Ecoteckne, Via Monteroni; 73100, Lecce Italy
| |
Collapse
|
30
|
El-Gewely MR, Andreassen M, Walquist M, Ursvik A, Knutsen E, Nystad M, Coucheron DH, Myrmel KS, Hennig R, Johansen SD. Differentially Expressed MicroRNAs in Meningiomas Grades I and II Suggest Shared Biomarkers with Malignant Tumors. Cancers (Basel) 2016; 8:E31. [PMID: 26950155 PMCID: PMC4810115 DOI: 10.3390/cancers8030031] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Revised: 02/19/2016] [Accepted: 02/24/2016] [Indexed: 12/18/2022] Open
Abstract
Meningiomas represent the most common primary tumors of the central nervous system, but few microRNA (miRNA) profiling studies have been reported so far. Deep sequencing of small RNA libraries generated from two human meningioma biopsies WHO grades I (benign) and II (atypical) were compared to excess dura controls. Nineteen differentially expressed miRNAs were validated by RT-qPCR using tumor RNA from 15 patients and 5 meninges controls. Tumor suppressor miR-218 and miR-34a were upregulated relative to normal controls, however, miR-143, miR-193b, miR-451 and oncogenic miR-21 were all downregulated. From 10 selected putative mRNA targets tested by RT-qPCR only four were differentially expressed relative to normal controls. PTEN and E-cadherin (CDH1) were upregulated, but RUNX1T1 was downregulated. Proliferation biomarker p63 was upregulated with nuclear localization, but not detected in most normal arachnoid tissues. Immunoreactivity of E-cadherin was detected in the outermost layer of normal arachnoids, but was expressed throughout the tumors. Nuclear Cyclin D1 expression was positive in all studied meningiomas, while its expression in arachnoid was limited to a few trabecular cells. Meningiomas of grades I and II appear to share biomarkers with malignant tumors, but with some additional tumor suppressor biomarkers expression. Validation in more patients is of importance.
Collapse
Affiliation(s)
- Mohamed Raafat El-Gewely
- Department of Medical Biology, Faculty of Health Sciences, UiT-The Arctic University of Norway, NO-9037 Tromsø, Norway.
| | - Morten Andreassen
- Department of Medical Biology, Faculty of Health Sciences, UiT-The Arctic University of Norway, NO-9037 Tromsø, Norway.
| | - Mari Walquist
- Department of Medical Biology, Faculty of Health Sciences, UiT-The Arctic University of Norway, NO-9037 Tromsø, Norway.
| | - Anita Ursvik
- Department of Medical Biology, Faculty of Health Sciences, UiT-The Arctic University of Norway, NO-9037 Tromsø, Norway.
| | - Erik Knutsen
- Department of Medical Biology, Faculty of Health Sciences, UiT-The Arctic University of Norway, NO-9037 Tromsø, Norway.
| | - Mona Nystad
- Department of Clinical Medicine, Women's Health and Perinatology Research Group, Faculty of Health Sciences, UiT-The Arctic University of Norway, NO-9037 Tromsø, Norway.
- Department of Obstetrics and Gynecology, University Hospital of North Norway, NO-9038 Tromsø, Norway.
- Department of Medical Genetics, Division of Child and Adolescent Health, University Hospital of North Norway, NO-9038 Tromsø, Norway.
| | - Dag H Coucheron
- Department of Medical Biology, Faculty of Health Sciences, UiT-The Arctic University of Norway, NO-9037 Tromsø, Norway.
| | | | - Rune Hennig
- Department of Neurosurgery, University Hospital of North Norway, NO-9038 Tromsø, Norway.
- Department of Clinical Medicine, Division of Neurosurgery, Faculty of Health Sciences, UiT-The Arctic University of Norway, NO-9037 Tromsø, Norway.
| | - Steinar D Johansen
- Department of Medical Biology, Faculty of Health Sciences, UiT-The Arctic University of Norway, NO-9037 Tromsø, Norway.
- Marine Genomics Group, Faculty of Biosciences and Aquaculture, Nord University NO-8049 Bodø, Norway.
| |
Collapse
|
31
|
Vergara D, Simeone P, Franck J, Trerotola M, Giudetti A, Capobianco L, Tinelli A, Bellomo C, Fournier I, Gaballo A, Alberti S, Salzet M, Maffia M. Translating epithelial mesenchymal transition markers into the clinic: Novel insights from proteomics. EUPA OPEN PROTEOMICS 2016; 10:31-41. [PMID: 29900098 PMCID: PMC5988589 DOI: 10.1016/j.euprot.2016.01.003] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 11/30/2015] [Accepted: 01/05/2016] [Indexed: 12/15/2022]
Abstract
The growing understanding of the molecular mechanisms underlying epithelial-to-mesenchymal transition (EMT) may represent a potential source of clinical markers. Despite EMT drivers have not yet emerged as candidate markers in the clinical setting, their association with established clinical markers may improve their specificity and sensitivity. Mass spectrometry-based platforms allow analyzing multiple samples for the expression of EMT candidate markers, and may help to diagnose diseases or monitor treatment efficiently. This review highlights proteomic approaches applied to elucidate the differences between epithelial and mesenchymal tumors and describes how these can be used for target discovery and validation.
Collapse
Affiliation(s)
- Daniele Vergara
- Department of Biological and Environmental Sciences and Technologies, University of Salento, via Monteroni, 73100 Lecce, Italy.,Laboratory of Clinical Proteomic, "Giovanni Paolo II" Hospital, ASL-Lecce, Italy
| | - Pasquale Simeone
- Research Centre on Aging (Ce.S.I), Unit of Cytomorphology, "University G. d'Annunzio" Foundation, 66100 Chieti, Italy.,Department of Medicine and Aging Science, School of Medicine and Health Science, University "G. d'Annunzio" of Chieti-Pescara, 66100 Chieti, Italy
| | - Julien Franck
- U1192 INSERM, Laboratoire PRISM: Protéomique, Réponse Inflammatoire, Spectrométrie de Masse, Université Lille 1, Villeneuve D'Ascq, France
| | - Marco Trerotola
- Unit of Cancer Pathology, CeSI, Foundation University 'G. d'Annunzio', Chieti, Italy.,Department of Medical, Oral and Biotechnological Sciences, University "G. D'Annunzio" of Chieti-Pescara, 66100 Chieti, Italy
| | - Anna Giudetti
- Department of Biological and Environmental Sciences and Technologies, University of Salento, via Monteroni, 73100 Lecce, Italy
| | - Loredana Capobianco
- Department of Biological and Environmental Sciences and Technologies, University of Salento, via Monteroni, 73100 Lecce, Italy
| | - Andrea Tinelli
- Department of Gynecology and Obstetrics, Division of Experimental Endoscopic Surgery, Imaging, Minimally Invasive Therapy and Technology, "Vito Fazzi" Hospital, ASL-Lecce, Italy.,International Translational Medicine and Biomodelling Research Group, Department of Applied Mathematics, Moscow Institute of Physics and Technology (MIPT), State University, Moscow, Russia
| | - Claudia Bellomo
- Department of Medical Biochemistry and Microbiology and Ludwig Institute for Cancer Research, Science for Life Laboratory, Biomedical Center, Uppsala University, Box 582, SE 75 123 Uppsala, Sweden
| | - Isabelle Fournier
- U1192 INSERM, Laboratoire PRISM: Protéomique, Réponse Inflammatoire, Spectrométrie de Masse, Université Lille 1, Villeneuve D'Ascq, France
| | - Antonio Gaballo
- CNR NANOTEC-Institute of Nanotechnology, Polo di Nanotecnologia c/o Campus Ecotekne, via Monteroni, 73100 Lecce, Italy
| | - Saverio Alberti
- Unit of Cancer Pathology, CeSI, Foundation University 'G. d'Annunzio', Chieti, Italy
| | - Michel Salzet
- U1192 INSERM, Laboratoire PRISM: Protéomique, Réponse Inflammatoire, Spectrométrie de Masse, Université Lille 1, Villeneuve D'Ascq, France
| | - Michele Maffia
- Department of Biological and Environmental Sciences and Technologies, University of Salento, via Monteroni, 73100 Lecce, Italy.,Laboratory of Clinical Proteomic, "Giovanni Paolo II" Hospital, ASL-Lecce, Italy
| |
Collapse
|
32
|
Bottoni P, Isgrò MA, Scatena R. The epithelial-mesenchymal transition in cancer: a potential critical topic for translational proteomic research. Expert Rev Proteomics 2015; 13:115-33. [PMID: 26567562 DOI: 10.1586/14789450.2016.1112742] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The epithelial-mesenchymal transition (EMT) is a morphogenetic process that results in a loss of epithelial characteristics and the acquisition of a mesenchymal phenotype. First described in embryogenesis, the EMT has been recently implicated in carcinogenesis and tumor progression. In addition, recent evidence has shown that stem-like cancer cells present the hallmarks of the EMT. Some of the molecular mechanisms related to the interrelationships between cancer pathophysiology and the EMT are well-defined. Nevertheless, the precise molecular mechanism by which epithelial cancer cells acquire the mesenchymal phenotype remains largely unknown. This review focuses on various proteomic strategies with the goal of better understanding the physiological and pathological mechanisms of the EMT process.
Collapse
Affiliation(s)
- Patrizia Bottoni
- a Institute of Biochemistry and Clinical Biochemistry , School of Medicine - Catholic University , Rome , Italy
| | - Maria Antonietta Isgrò
- b Department of Diagnostic and Molecular Medicine , Catholic University of the Sacred Heart , Rome , Italy
| | - Roberto Scatena
- a Institute of Biochemistry and Clinical Biochemistry , School of Medicine - Catholic University , Rome , Italy
| |
Collapse
|
33
|
The Relationship Between E-Cadherin and its Transcriptional Repressors in Spontaneously Arising Canine Invasive Micropapillary Mammary Carcinoma. J Comp Pathol 2015; 153:256-65. [PMID: 26385325 DOI: 10.1016/j.jcpa.2015.08.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 08/05/2015] [Accepted: 08/17/2015] [Indexed: 01/08/2023]
Abstract
E-cadherin downregulation is related to metastatic behaviour and a poor prognosis in cancer. It might be induced by transcriptional repression mediated by the transcription factors SNAIL, ZEB1, ZEB2 and TWIST. Here, we investigated E-cadherin expression and its relationship to those transcriptional repressors (i.e. SNAIL, ZEB1, ZEB2 and TWIST) in the progression from carcinoma 'in situ' to invasion to lymph node metastasis in spontaneously arising canine invasive micropapillary carcinoma (IMPC). E-cadherin expression decreased from carcinoma in situ to invasive progression and was likely to increase with lymph node metastasis. Expression of SNAIL decreased from carcinoma in situ to invasive areas and from invasive areas to lymph nodes. Metastatic lymph nodes had higher expression of ZEB1 than carcinoma in situ and invasive areas. ZEB2 expression was observed in 52%, 38% and 33% of carcinoma in situ areas, invasive areas and lymph node metastases, respectively. TWIST expression was observed in 52%, 38% and 33% of carcinoma in situ areas, invasive areas and lymph node metastases, respectively. In invasive areas, E-cadherin downregulation correlated significantly with SNAIL and TWIST upregulation. Additionally, in infiltrating components of IMPCs, E-cadherin(-)SNAIL(+) neoplastic epithelial cells were observed by immunofluorescence. Taken together, canine mammary IMPCs had a loss of E-cadherin from carcinoma in situ to invasive areas, which appears to be induced by the transcription factor SNAIL. In lymph node metastasis, ZEB1 appears to not exert E-cadherin transcriptional repression activity.
Collapse
|
34
|
Expression of SIP1 is strongly correlated with LDHA and shows a significantly poor outcome in gastric cancer. Tumour Biol 2015; 36:7521-30. [DOI: 10.1007/s13277-015-3470-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Accepted: 04/15/2015] [Indexed: 10/23/2022] Open
|