1
|
Granados-Aparici S, Vieco-Martí I, López-Carrasco A, Navarro S, Noguera R. Real-time morphometric analysis of targeted therapy for neuroblastoma cells in monolayer and 3D hydrogels using digital holographic microscopy. iScience 2024; 27:111231. [PMID: 39569369 PMCID: PMC11576390 DOI: 10.1016/j.isci.2024.111231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 08/26/2024] [Accepted: 10/21/2024] [Indexed: 11/22/2024] Open
Abstract
High-risk neuroblastoma (HR-NB) patient treatment is currently insufficient and challenging due to its high clinical, morphological, and genetic heterogeneity as well as the scarcity of available samples for research. We used a gelatin- and silk fibroin-based hydrogel system with cross-linked vitronectin (VN) as an artificial biomimetic three-dimensional (3D) environment to mirror aggressive neuroblastoma (NB) tumors and tested long-term cell response to Cilengitide (CLG). Based on our previous studies and others using the integrin inhibitor CLG as a potential mechanotherapy drug, we show that CLG caused cell detachment in monolayer cultures of MYCN-amplified SK-N-BE (2) and ALK-mutated SH-SY5Y human neuroblastoma cell lines. Cell detachment and aggregation were maintained in hydrogel-free monolayer cells whereas cells embedded in hydrogels presented different responses to treatment, suggesting differential anoikis resistance between the two cell lines. This underscores the advantages of testing therapeutic approaches using real-time imaging of tumor cells in 3D biomimetic models and its contribution to precision medicine.
Collapse
Affiliation(s)
- Sofia Granados-Aparici
- Pathology Department, Medical School, University of Valencia-INCLIVA, Valencia, Spain
- Centro de Investigación Biomédica en Red de Cáncer, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Isaac Vieco-Martí
- Pathology Department, Medical School, University of Valencia-INCLIVA, Valencia, Spain
- Centro de Investigación Biomédica en Red de Cáncer, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Amparo López-Carrasco
- Pathology Department, Medical School, University of Valencia-INCLIVA, Valencia, Spain
- Centro de Investigación Biomédica en Red de Cáncer, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Samuel Navarro
- Pathology Department, Medical School, University of Valencia-INCLIVA, Valencia, Spain
- Centro de Investigación Biomédica en Red de Cáncer, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Rosa Noguera
- Pathology Department, Medical School, University of Valencia-INCLIVA, Valencia, Spain
- Centro de Investigación Biomédica en Red de Cáncer, Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
2
|
Jokela TA, Dane MA, Smith RL, Devlin KL, Shalabi S, Lopez JC, Miyano M, Stampfer MR, Korkola JE, Gray JW, Heiser LM, LaBarge MA. Functional delineation of the luminal epithelial microenvironment in breast using cell-based screening in combinatorial microenvironments. Cell Signal 2024; 113:110958. [PMID: 37935340 PMCID: PMC10696611 DOI: 10.1016/j.cellsig.2023.110958] [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: 09/22/2023] [Revised: 10/24/2023] [Accepted: 10/30/2023] [Indexed: 11/09/2023]
Abstract
Microenvironment signals are potent determinants of cell fate and arbiters of tissue homeostasis, however understanding how different microenvironment factors coordinately regulate cellular phenotype has been experimentally challenging. Here we used a high-throughput microenvironment microarray comprised of 2640 unique pairwise signals to identify factors that support proliferation and maintenance of primary human mammary luminal epithelial cells. Multiple microenvironment factors that modulated luminal cell number were identified, including: HGF, NRG1, BMP2, CXCL1, TGFB1, FGF2, PDGFB, RANKL, WNT3A, SPP1, HA, VTN, and OMD. All of these factors were previously shown to modulate luminal cell numbers in painstaking mouse genetics experiments, or were shown to have a role in breast cancer, demonstrating the relevance and power of our high-dimensional approach to dissect key microenvironmental signals. RNA-sequencing of primary epithelial and stromal cell lineages identified the cell types that express these signals and the cognate receptors in vivo. Cell-based functional studies confirmed which effects from microenvironment factors were reproducible and robust to individual variation. Hepatocyte growth factor (HGF) was the factor most robust to individual variation and drove expansion of luminal cells via cKit+ progenitor cells, which expressed abundant MET receptor. Luminal cells from women who are genetically high risk for breast cancer had significantly more MET receptor and may explain the characteristic expansion of the luminal lineage in those women. In ensemble, our approach provides proof of principle that microenvironment signals that control specific cellular states can be dissected with high-dimensional cell-based approaches.
Collapse
Affiliation(s)
- Tiina A Jokela
- Department of Population Sciences, Center for Cancer and Aging, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Mark A Dane
- Department of Biomedical Engineering, Oregon Health Sciences University, Portland, OR, USA
| | - Rebecca L Smith
- Department of Biomedical Engineering, Oregon Health Sciences University, Portland, OR, USA
| | - Kaylyn L Devlin
- Department of Biomedical Engineering, Oregon Health Sciences University, Portland, OR, USA
| | - Sundus Shalabi
- Department of Population Sciences, Center for Cancer and Aging, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA; Faculty of Medicine, Arab American University of Palestine, Jenin, Palestine
| | - Jennifer C Lopez
- Department of Population Sciences, Center for Cancer and Aging, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Masaru Miyano
- Department of Population Sciences, Center for Cancer and Aging, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Martha R Stampfer
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - James E Korkola
- Department of Biomedical Engineering, Oregon Health Sciences University, Portland, OR, USA
| | - Joe W Gray
- Department of Biomedical Engineering, Oregon Health Sciences University, Portland, OR, USA
| | - Laura M Heiser
- Department of Biomedical Engineering, Oregon Health Sciences University, Portland, OR, USA.
| | - Mark A LaBarge
- Department of Population Sciences, Center for Cancer and Aging, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA; Center for Cancer Biomarkers Research (CCBIO), University of Bergen, Bergen, Norway; Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA.
| |
Collapse
|
3
|
Burgos-Panadero R, El Moukhtari SH, Noguera I, Rodríguez-Nogales C, Martín-Vañó S, Vicente-Munuera P, Cañete A, Navarro S, Blanco-Prieto MJ, Noguera R. Unraveling the extracellular matrix-tumor cell interactions to aid better targeted therapies for neuroblastoma. Int J Pharm 2021; 608:121058. [PMID: 34461172 DOI: 10.1016/j.ijpharm.2021.121058] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 08/24/2021] [Accepted: 08/25/2021] [Indexed: 12/17/2022]
Abstract
Treatment in children with high-risk neuroblastoma remains largely unsuccessful due to the development of metastases and drug resistance. The biological complexity of these tumors and their microenvironment represent one of the many challenges to face. Matrix glycoproteins such as vitronectin act as bridge elements between extracellular matrix and tumor cells and can promote tumor cell spreading. In this study, we established through a clinical cohort and preclinical models that the interaction of vitronectin and its ligands, such as αv integrins, are related to the stiffness of the extracellular matrix in high-risk neuroblastoma. These marked alterations found in the matrix led us to specifically target tumor cells within these altered matrices by employing nanomedicine and combination therapy. Loading the conventional cytotoxic drug etoposide into nanoparticles significantly increased its efficacy in neuroblastoma cells. We noted high synergy between etoposide and cilengitide, a high-affinity cyclic pentapeptide αv integrin antagonist. The results of this study highlight the need to characterize cell-extracellular matrix interactions, to improve patient care in high-risk neuroblastoma.
Collapse
Affiliation(s)
- Rebeca Burgos-Panadero
- Department of Pathology, Medical School, University of Valencia - INCLIVA Biomedical Health Research Institute, 46010 Valencia, Spain; Low Prevalence Tumors, Centro de investigación biomédica en red de cáncer (CIBERONC), Instituto de Salud Carlos III, 28029 Madrid, Spain.
| | - Souhaila H El Moukhtari
- Department of Pharmaceutical Technology and Chemistry, School of Pharmacy and Nutrition, University of Navarra, 31008 Pamplona, Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain.
| | - Inmaculada Noguera
- Central Support Service for Experimental Research (SCSIE), University of Valencia, Burjassot, Valencia, Spain.
| | - Carlos Rodríguez-Nogales
- Department of Pharmaceutical Technology and Chemistry, School of Pharmacy and Nutrition, University of Navarra, 31008 Pamplona, Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain.
| | - Susana Martín-Vañó
- Department of Pathology, Medical School, University of Valencia - INCLIVA Biomedical Health Research Institute, 46010 Valencia, Spain; Low Prevalence Tumors, Centro de investigación biomédica en red de cáncer (CIBERONC), Instituto de Salud Carlos III, 28029 Madrid, Spain.
| | - Pablo Vicente-Munuera
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla and Departamento de Biología Celular, Universidad de Sevilla, Seville 41013, Spain.
| | - Adela Cañete
- Pediatric Oncology, La Fe Hospital, Av. Fernando Abril Martorell 106, 46026 Valencia, Spain.
| | - Samuel Navarro
- Department of Pathology, Medical School, University of Valencia - INCLIVA Biomedical Health Research Institute, 46010 Valencia, Spain; Low Prevalence Tumors, Centro de investigación biomédica en red de cáncer (CIBERONC), Instituto de Salud Carlos III, 28029 Madrid, Spain.
| | - María J Blanco-Prieto
- Department of Pharmaceutical Technology and Chemistry, School of Pharmacy and Nutrition, University of Navarra, 31008 Pamplona, Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain.
| | - Rosa Noguera
- Department of Pathology, Medical School, University of Valencia - INCLIVA Biomedical Health Research Institute, 46010 Valencia, Spain; Low Prevalence Tumors, Centro de investigación biomédica en red de cáncer (CIBERONC), Instituto de Salud Carlos III, 28029 Madrid, Spain.
| |
Collapse
|
4
|
Nohawica M, Errachid A, Wyganowska-Swiatkowska M. Adipose-PAS interactions in the context of its localised bio-engineering potential (Review). Biomed Rep 2021; 15:70. [PMID: 34276988 PMCID: PMC8278035 DOI: 10.3892/br.2021.1446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 05/05/2021] [Indexed: 11/24/2022] Open
Abstract
Adipocytes are a known source of stem cells. They are easy to harvest, and are a suitable candidate for autogenous grafts. Adipose derived stem cells (ADSCs) have multiple target tissues which they can differentiate into, including bone and cartilage. In adipose tissue, ADSCs are able to differentiate, as well as providing energy and a supply of cytokines/hormones to manage the hypoxic and lipid/hormone saturated adipose environment. The plasminogen activation system (PAS) controls the majority of proteolytic activities in both adipose and wound healing environments, allowing for rapid cellular migration and tissue remodelling. While the primary activation pathway for PAS occurs through the urokinase plasminogen activator (uPA), which is highly expressed by endothelial cells, its function is not limited to enabling revascularisation. Proteolytic activity is dependent on protease activation, localisation, recycling mechanisms and substrate availability. uPA and uPA activated plasminogen allows pluripotent cells to arrive to new local environments and fulfil the niche demands. However, overstimulation, the acquisition of a migratory phenotype and constant protein turnover can be unconducive to the formation of structured hard and soft tissues. To maintain a suitable healing pattern, the proteolytic activity stimulated by uPA is modulated by plasminogen activator inhibitor 1. Depending on the physiological settings, different parts of the remodelling mechanism are activated with varying results. Utilising the differences within each microenvironment to recreate a desired niche is a valid therapeutic bio-engineering approach. By controlling the rate of protein turnover combined with a receptive stem cell lineage, such as ADSC, a novel avenue on the therapeutic opportunities may be identified, which can overcome limitations, such as scarcity of stem cells, low angiogenic potential or poor host tissue adaptation.
Collapse
Affiliation(s)
- Michal Nohawica
- Chair and Department of Dental Surgery and Periodontology, Poznan University of Medicinal Sciences, Poznan, Greater Poland 60-812, Poland
| | - Abdelmounaim Errachid
- Chair and Department of Dental Surgery and Periodontology, Poznan University of Medicinal Sciences, Poznan, Greater Poland 60-812, Poland
- Earth and Life Institute, University Catholique of Louvain, Louvain-la-Neuve B-1348, Belgium
| | - Marzena Wyganowska-Swiatkowska
- Chair and Department of Dental Surgery and Periodontology, Poznan University of Medicinal Sciences, Poznan, Greater Poland 60-812, Poland
| |
Collapse
|
5
|
Riccardi C, Napolitano E, Musumeci D, Montesarchio D. Dimeric and Multimeric DNA Aptamers for Highly Effective Protein Recognition. Molecules 2020; 25:E5227. [PMID: 33182593 PMCID: PMC7698228 DOI: 10.3390/molecules25225227] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/06/2020] [Accepted: 11/08/2020] [Indexed: 12/14/2022] Open
Abstract
Multivalent interactions frequently occur in biological systems and typically provide higher binding affinity and selectivity in target recognition than when only monovalent interactions are operative. Thus, taking inspiration by nature, bivalent or multivalent nucleic acid aptamers recognizing a specific biological target have been extensively studied in the last decades. Indeed, oligonucleotide-based aptamers are suitable building blocks for the development of highly efficient multivalent systems since they can be easily modified and assembled exploiting proper connecting linkers of different nature. Thus, substantial research efforts have been put in the construction of dimeric/multimeric versions of effective aptamers with various degrees of success in target binding affinity or therapeutic activity enhancement. The present review summarizes recent advances in the design and development of dimeric and multimeric DNA-based aptamers, including those forming G-quadruplex (G4) structures, recognizing different key proteins in relevant pathological processes. Most of the designed constructs have shown improved performance in terms of binding affinity or therapeutic activity as anti-inflammatory, antiviral, anticoagulant, and anticancer agents and their number is certainly bound to grow in the next future.
Collapse
Affiliation(s)
- Claudia Riccardi
- Department of Chemical Sciences, University of Naples Federico II, via Cintia 21, I-80126 Naples, Italy; (E.N.); (D.M.); (D.M.)
- Department of Advanced Medical and Surgical Sciences, 2nd Division of Neurology, Center for Rare Diseases and InterUniversity Center for Research in Neurosciences, University of Campania Luigi Vanvitelli, via Sergio Pansini, 5, I-80131 Naples, Italy
| | - Ettore Napolitano
- Department of Chemical Sciences, University of Naples Federico II, via Cintia 21, I-80126 Naples, Italy; (E.N.); (D.M.); (D.M.)
| | - Domenica Musumeci
- Department of Chemical Sciences, University of Naples Federico II, via Cintia 21, I-80126 Naples, Italy; (E.N.); (D.M.); (D.M.)
- Institute of Biostructures and Bioimages, CNR, via Mezzocannone 16, I-80134 Naples, Italy
| | - Daniela Montesarchio
- Department of Chemical Sciences, University of Naples Federico II, via Cintia 21, I-80126 Naples, Italy; (E.N.); (D.M.); (D.M.)
| |
Collapse
|
6
|
Ghimire H, Garlapati C, Janssen EAM, Krishnamurti U, Qin G, Aneja R, Perera AGU. Protein Conformational Changes in Breast Cancer Sera Using Infrared Spectroscopic Analysis. Cancers (Basel) 2020; 12:E1708. [PMID: 32605072 PMCID: PMC7407230 DOI: 10.3390/cancers12071708] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 06/19/2020] [Accepted: 06/25/2020] [Indexed: 01/08/2023] Open
Abstract
Protein structural alterations, including misfolding and aggregation, are a hallmark of several diseases, including cancer. However, the possible clinical application of protein conformational analysis using infrared spectroscopy to detect cancer-associated structural changes in proteins has not been established yet. The present study investigates the applicability of Fourier transform infrared spectroscopy in distinguishing the sera of healthy individuals and breast cancer patients. The cancer-associated alterations in the protein structure were analyzed by fitting the amide I (1600-1700 cm-1) band of experimental curves, as well as by comparing the ratio of the absorbance values at the amide II and amide III bands, assigning those as the infrared spectral signatures. The snapshot of the breast cancer-associated alteration in circulating DNA and RNA was also evaluated by extending the spectral fitting protocol to the complex region of carbohydrates and nucleic acids, 1140-1000 cm-1. The sensitivity and specificity of these signatures, representing the ratio of the α-helix and β-pleated sheet in proteins, were both 90%. Likewise, the ratio of amides II and amide III (I1556/I1295) had a sensitivity and specificity of 100% and 80%, respectively. Thus, infrared spectroscopy can serve as a powerful tool to understand the protein structural alterations besides distinguishing breast cancer and healthy serum samples.
Collapse
Affiliation(s)
- Hemendra Ghimire
- Department of Physics and Astronomy, Georgia State University, Atlanta, GA 30303, USA;
| | | | - Emiel A. M. Janssen
- Department of Pathology, Stavanger University Hospital, Stavanger NO-4068, Norway;
| | - Uma Krishnamurti
- Department of Pathology, Emory University School of Medicine, Atlanta, GA 30322, USA;
| | - Gengsheng Qin
- Department of Mathematics and Statistics, Georgia State University, Atlanta, GA 30303, USA;
| | - Ritu Aneja
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA; (C.G.); (R.A.)
- Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA 30303, USA
| | - A. G. Unil Perera
- Department of Physics and Astronomy, Georgia State University, Atlanta, GA 30303, USA;
- Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA 30303, USA
| |
Collapse
|
7
|
Yadav VK, Lee TY, Hsu JBK, Huang HD, Yang WCV, Chang TH. Computational analysis for identification of the extracellular matrix molecules involved in endometrial cancer progression. PLoS One 2020; 15:e0231594. [PMID: 32315343 PMCID: PMC7173926 DOI: 10.1371/journal.pone.0231594] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 03/26/2020] [Indexed: 12/16/2022] Open
Abstract
Recurrence and poorly differentiated (grade 3 and above) and atypical cell type endometrial cancer (EC) have poor prognosis outcome. The mechanisms and characteristics of recurrence and distal metastasis of EC remain unclear. The extracellular matrix (ECM) of the reproductive tract in women undergoes extensive structural remodelling changes every month. Altered ECMs surrounding cells were believed to play crucial roles in a cancer progression. To decipher the associations between ECM and EC development, we generated a PAN-ECM Data list of 1516 genes including ECM molecules (ECMs), synthetic and degradation enzymes for ECMs, ECM receptors, and soluble molecules that regulate ECM and used RNA-Seq data from The Cancer Genome Atlas (TCGA) for the studies. The alterations of PAN-ECM genes by comparing the RNA-Seq expressions profiles of EC samples which have been grouped as tumorigenesis and metastasis group based on their pathological grading were identified. Differential analyses including functional enrichment, co-expression network, and molecular network analysis were carried out to identify the specific PAN-ECM genes that may involve in the progression of EC. Eight hundred and thirty-one and 241 PAN-ECM genes were significantly involved in tumorigenesis (p-value <1.571e-15) and metastasis (p-value <2.2e-16), respectively, whereas 140 genes were in the intersection of tumorigenesis and metastasis. Interestingly, 92 of the 140 intersecting PAN-ECM genes showed contrasting fold changes between the tumorigenesis and metastasis datasets. Enrichment analysis for the contrast PAN-ECM genes indicated pathways such as GP6 signaling, ILK signaling, and interleukin (IL)-8 signaling pathways were activated in metastasis but inhibited in tumorigenesis. The significantly activated ECM and ECM associated genes in GP6 signaling, ILK signaling, and interleukin (IL)-8 signaling pathways may play crucial roles in metastasis of EC. Our study provides a better understanding of the etiology and the progression of EC.
Collapse
Affiliation(s)
- Vijesh Kumar Yadav
- The Program for Translational Medicine, Graduate Institute of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Tzong-Yi Lee
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen, Longgang District, Shenzhen, Guangdong Province, China
- School of Life and Health Science, The Chinese University of Hong Kong, Shenzhen, Longgang District, Shenzhen, Guangdong Province, China
| | - Justin Bo-Kai Hsu
- Department of Medical Research, Taipei Medical University Hospital, Taipei, Taiwan
| | - Hsien-Da Huang
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen, Longgang District, Shenzhen, Guangdong Province, China
- School of Life and Health Science, The Chinese University of Hong Kong, Shenzhen, Longgang District, Shenzhen, Guangdong Province, China
| | - Wei-Chung Vivian Yang
- The Program for Translational Medicine, Graduate Institute of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- The PhD Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- * E-mail: (W-CVY); (T-HC)
| | - Tzu-Hao Chang
- Graduate Institute of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- Clinical Big Data Research Center, Taipei Medical University Hospital, Taipei, Taiwan
- * E-mail: (W-CVY); (T-HC)
| |
Collapse
|
8
|
Jandrey EHF, Moura RP, Andrade LNS, Machado CL, Campesato LF, Leite KRM, Inoue LT, Asprino PF, da Silva APM, de Barros ACSD, Carvalho A, de Lima VC, Carraro DM, Brentani HP, da Cunha IW, Soares FA, Parmigiani RB, Chammas R, Camargo AA, Costa ÉT. NDRG4 promoter hypermethylation is a mechanistic biomarker associated with metastatic progression in breast cancer patients. NPJ Breast Cancer 2019; 5:11. [PMID: 30963110 PMCID: PMC6450950 DOI: 10.1038/s41523-019-0106-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Accepted: 03/11/2019] [Indexed: 01/27/2023] Open
Abstract
The risk of developing metastatic disease in breast cancer patients is traditionally predictable based on the number of positive axillary lymph nodes, complemented with additional clinicopathological factors. However, since lymph node-negative patients have a 20-30% probability of developing metastatic disease, lymph node information alone is insufficient to accurately assess individual risk. Molecular approaches, such as multigene expression panels, analyze a set of cancer-related genes that more accurately predict the early risk of metastasis and the treatment response. Here, we present N-Myc downstream-regulated gene 4 (NDRG4) epigenetic silencing as a mechanistic biomarker of metastasis in ductal invasive breast tumors. While aberrant NDRG4 DNA hypermethylation is significantly associated with the development of metastatic disease, downregulation of NDRG4 transcription and protein expression is functionally associated with enhanced lymph node adhesion and cell mobility. Here, we show that epigenetic silencing of NDRG4 modulates integrin signaling by assembling β1-integrins into large punctate clusters at the leading edge of tumor cells to promote an "adhesive switch," decreasing cell adhesion to fibronectin and increasing cell adhesion and migration towards vitronectin, an important component of human lymph nodes. Taken together, our functional and clinical observations suggest that NDRG4 is a potential mechanistic biomarker in breast cancer that is functionally associated with metastatic disease.
Collapse
Affiliation(s)
| | | | - Luciana N. S. Andrade
- Laboratório de Oncologia Experimental, Centro de Investigação Translacional em Oncologia, Instituto do Câncer do Estado de São Paulo, São Paulo, SP Brazil
| | - Camila L. Machado
- Laboratório de Oncologia Experimental, Centro de Investigação Translacional em Oncologia, Instituto do Câncer do Estado de São Paulo, São Paulo, SP Brazil
| | | | | | - Lilian T. Inoue
- Centro de Oncologia Molecular, Hospital Sírio-Libanês, São Paulo, SP Brazil
| | - Paula F. Asprino
- Centro de Oncologia Molecular, Hospital Sírio-Libanês, São Paulo, SP Brazil
| | | | | | | | - Vladmir C. de Lima
- Centro Internacional de Pesquisa, A.C. Camargo Cancer Center, Fundação Antônio Prudente, São Paulo, SP Brazil
| | - Dirce M. Carraro
- Centro Internacional de Pesquisa, A.C. Camargo Cancer Center, Fundação Antônio Prudente, São Paulo, SP Brazil
| | - Helena P. Brentani
- LIM23-Instituto de Psiquiatria, Faculdade de Medicina da Universidade de São Paulo (USP), São Paulo, Brazil
| | | | | | | | - Roger Chammas
- Laboratório de Oncologia Experimental, Centro de Investigação Translacional em Oncologia, Instituto do Câncer do Estado de São Paulo, São Paulo, SP Brazil
| | - Anamaria A. Camargo
- Centro de Oncologia Molecular, Hospital Sírio-Libanês, São Paulo, SP Brazil
- Ludwig Institute for Cancer Research (LICR), São Paulo, Brazil
| | - Érico T. Costa
- Centro de Oncologia Molecular, Hospital Sírio-Libanês, São Paulo, SP Brazil
- Ludwig Institute for Cancer Research (LICR), São Paulo, Brazil
| |
Collapse
|
9
|
Combinatorial Electrophoresis and Mass Spectrometry-Based Proteomics in Breast Milk for Breast Cancer Biomarker Discovery. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1140:451-467. [PMID: 31347064 DOI: 10.1007/978-3-030-15950-4_26] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Innovations in approaches for early detection and individual risk assessment of different cancers, including breast cancer (BC), are needed to reduce cancer morbidity and associated mortality. The assessment of potential cancer biomarkers in accessible bodily fluids provides a novel approach to identify the risk and/or onset of cancer. Biomarkers are biomolecules, such as proteins, that are indicative of an abnormality or a disease. Human milk is vastly underutilized biospecimen that offers the opportunity to investigate potential protein BC-biomarkers in young, reproductively active women. As a first step, we have examined the entire protein pattern in human milk samples from breastfeeding mothers with cancer, who were diagnosed either before or after milk donation, and from women without cancer, using mass spectrometry (MS)-based proteomics.
Collapse
|
10
|
Aslebagh R, Channaveerappa D, Arcaro KF, Darie CC. Comparative two-dimensional polyacrylamide gel electrophoresis (2D-PAGE) of human milk to identify dysregulated proteins in breast cancer. Electrophoresis 2018; 39:1723-1734. [PMID: 29756217 DOI: 10.1002/elps.201800025] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 03/11/2018] [Accepted: 04/16/2018] [Indexed: 12/20/2022]
Abstract
Breast cancer (BC) remains a major cause of mortality, and early detection is considered important for reducing BC-associated deaths. Early detection of BC is challenging in young women, due to the limitations of mammography on the dense breast tissue of young women. We recently reported results of a pilot proteomics study, using one-dimensional polyacrylamide gel electrophoresis (1D-PAGE) and mass spectrometry (MS) to investigate differences in milk proteins from women with and without BC. Here, we applied two-dimensional polyacrylamide gel electrophoresis (2D-PAGE) and MS to compare the protein pattern in milk from the breasts of a single woman who was diagnosed with BC in one breast 24 months after donating her milk. Statistically different gel spots were picked for protein digestion followed by nanoliquid chromatography tandem MS (nanoLC-MS/MS) analysis. The upregulated proteins in BC versus control are alpha-amylase, gelsolin isoform a precursor, alpha-2-glycoprotein 1 zinc isoform CRA_b partial, apoptosis-inducing factor 2 and vitronectin. Several proteins were downregulated in the milk of the breast later diagnosed with cancer as compared to the milk from the healthy breast, including different isoforms of albumin, cholesterol esterase, different isoforms of lactoferrin, different proteins from the casein family and different isoforms of lysozyme. Results warrant further studies to determine the usefulness of these milk proteins for assessing risk and detecting occult disease. MS data is available via ProteomeXchange with identifier PXD009860.
Collapse
Affiliation(s)
- Roshanak Aslebagh
- Biochemistry & Proteomics Group, Department of Chemistry & Biomolecular Science, Clarkson University, Potsdam, NY, USA
| | - Devika Channaveerappa
- Biochemistry & Proteomics Group, Department of Chemistry & Biomolecular Science, Clarkson University, Potsdam, NY, USA
| | - Kathleen F Arcaro
- Department of Veterinary & Animal Sciences, University of Massachusetts Amherst, Amherst, MA, USA
| | - Costel C Darie
- Biochemistry & Proteomics Group, Department of Chemistry & Biomolecular Science, Clarkson University, Potsdam, NY, USA
| |
Collapse
|
11
|
Chang PY, Liao YP, Wang HC, Chen YC, Huang RL, Wang YC, Yuan CC, Lai HC. An epigenetic signature of adhesion molecules predicts poor prognosis of ovarian cancer patients. Oncotarget 2017; 8:53432-53449. [PMID: 28881822 PMCID: PMC5581121 DOI: 10.18632/oncotarget.18515] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 05/10/2017] [Indexed: 12/31/2022] Open
Abstract
DNA methylation is a promising biomarker for cancer. The epigenetic effects of cell adhesion molecules may affect the therapeutic outcome and the present study examined their effects on survival in ovarian cancer. We integrated methylomics and genomics datasets in The Cancer Genome Atlas (n = 391) and identified 106 highly methylated adhesion-related genes in ovarian cancer tissues. Univariate analysis revealed the methylation status of eight genes related to progression-free survival. In multivariate Cox regression analysis, four highly methylated genes (CD97, CTNNA1, DLC1, HAPLN2) and three genes (LAMA4, LPP, MFAP4) with low methylation were significantly associated with poor progression-free survival. Low methylation of VTN was an independent poor prognostic factor for overall survival after adjustment for age and stage. Patients who carried any two of CTNNA1, DLC1 or MFAP4 were significantly associated with poor progression-free survival (hazard ratio: 1.59; 95% confidence interval: 1.23, 2.05). This prognostic methylation signature was validated in a methylomics dataset generated in our lab (n = 37, hazard ratio: 16.64; 95% confidence interval: 2.68, 103.14) and in another from the Australian Ovarian Cancer Study (n = 91, hazard ratio: 2.43; 95% confidence interval: 1.11, 5.36). Epigenetics of cell adhesion molecules is related to ovarian cancer prognosis. A more comprehensive methylomics of cell adhesion molecules is needed and may advance personalized treatment with adhesion molecule-related drugs.
Collapse
Affiliation(s)
- Ping-Ying Chang
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Republic of China.,Division of Hematology & Oncology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Republic of China
| | - Yu-Ping Liao
- Department of Obstetrics and Gynecology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Republic of China
| | - Hui-Chen Wang
- Department of Obstetrics and Gynecology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Republic of China
| | - Yu-Chih Chen
- Division of Research and Analysis, Food and Drug Administration, Ministry of Health and Welfare, Taipei, Republic of China
| | - Rui-Lan Huang
- Department of Obstetrics and Gynecology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Republic of China
| | - Yu-Chi Wang
- Department of Obstetrics and Gynecology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Republic of China
| | - Chiou-Chung Yuan
- Department of Obstetrics and Gynecology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Republic of China
| | - Hung-Cheng Lai
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Republic of China.,Department of Obstetrics and Gynecology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Republic of China.,Department of Obstetrics and Gynecology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Republic of China.,Translational Epigenetic Center, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Republic of China.,Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, P. R. China.,Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, P. R. China
| |
Collapse
|
12
|
Stuart CH, Riley KR, Boyacioglu O, Herpai DM, Debinski W, Qasem S, Marini FC, Colyer CL, Gmeiner WH. Selection of a Novel Aptamer Against Vitronectin Using Capillary Electrophoresis and Next Generation Sequencing. MOLECULAR THERAPY. NUCLEIC ACIDS 2016; 5:e386. [PMID: 27845768 PMCID: PMC5155323 DOI: 10.1038/mtna.2016.91] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 09/02/2016] [Indexed: 01/04/2023]
Abstract
Breast cancer (BC) results in ~40,000 deaths each year in the United States and even among survivors treatment of the disease may have devastating consequences, including increased risk for heart disease and cognitive impairment resulting from the toxic effects of chemotherapy. Aptamer-mediated drug delivery can contribute to improved treatment outcomes through the selective delivery of chemotherapy to BC cells, provided suitable cancer-specific antigens can be identified. We report here the use of capillary electrophoresis in conjunction with next generation sequencing to develop the first vitronectin (VN) binding aptamer (VBA-01; Kd 405 nmol/l, the first aptamer to vitronectin (VN; Kd = 405 nmol/l) , a protein that plays an important role in wound healing and that is present at elevated levels in BC tissue and in the blood of BC patients relative to the corresponding nonmalignant tissues. We used VBA-01 to develop DVBA-01, a dimeric aptamer complex, and conjugated doxorubicin (Dox) to DVBA-01 (7:1 ratio) using pH-sensitive, covalent linkages. Dox conjugation enhanced the thermal stability of the complex (60.2 versus 46.5°C) and did not decrease affinity for the VN target. The resulting DVBA-01-Dox complex displayed increased cytotoxicity to MDA-MB-231 BC cells that were cultured on plasticware coated with VN (1.8 × 10-6mol/l) relative to uncoated plates (2.4 × 10-6 mol/l), or plates coated with the related protein fibronectin (2.1 × 10-6 mol/l). The VBA-01 aptamer was evaluated for binding to human BC tissue using immunohistochemistry and displayed tissue specific binding and apparent association with BC cells. In contrast, a monoclonal antibody that preferentially binds to multimeric VN primarily stained extracellular matrix and vessel walls of BC tissue. Our results indicate a strong potential for using VN-targeting aptamers to improve drug delivery to treat BC.
Collapse
Affiliation(s)
- Christopher H Stuart
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
- Department of Molecular Medicine and Translation Science, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
- Current address: RayBiotech, Norcross, Georgia, USA
| | - Kathryn R Riley
- Department of Chemistry, Wake Forest University, Winston-Salem, North Carolina, USA
- Current address: Department of Chemistry and Biochemistry, Swarthmore College,Swarthmore, Pennsylvania, USA
| | - Olcay Boyacioglu
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
- Current address: Department of Food Engineering, Faculty of Engineering, Adnan Menderes University, Aydin, Turkey
| | - Denise M Herpai
- Brain Tumor Center of Excellence, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Waldemar Debinski
- Brain Tumor Center of Excellence, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Shadi Qasem
- Department of Pathology, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Frank C Marini
- Wake Forest Institute of Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Christa L. Colyer
- Department of Chemistry, Wake Forest University, Winston-Salem, North Carolina, USA
| | - William H Gmeiner
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
- Department of Molecular Medicine and Translation Science, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| |
Collapse
|
13
|
De Lorenzi V, Sarra Ferraris GM, Madsen JB, Lupia M, Andreasen PA, Sidenius N. Urokinase links plasminogen activation and cell adhesion by cleavage of the RGD motif in vitronectin. EMBO Rep 2016; 17:982-98. [PMID: 27189837 DOI: 10.15252/embr.201541681] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 04/19/2016] [Indexed: 11/09/2022] Open
Abstract
Components of the plasminogen activation system including urokinase (uPA), its inhibitor (PAI-1) and its cell surface receptor (uPAR) have been implicated in a wide variety of biological processes related to tissue homoeostasis. Firstly, the binding of uPA to uPAR favours extracellular proteolysis by enhancing cell surface plasminogen activation. Secondly, it promotes cell adhesion and signalling through binding of the provisional matrix protein vitronectin. We now report that uPA and plasmin induces a potent negative feedback on cell adhesion through specific cleavage of the RGD motif in vitronectin. Cleavage of vitronectin by uPA displays a remarkable receptor dependence and requires concomitant binding of both uPA and vitronectin to uPAR Moreover, we show that PAI-1 counteracts the negative feedback and behaves as a proteolysis-triggered stabilizer of uPAR-mediated cell adhesion to vitronectin. These findings identify a novel and highly specific function for the plasminogen activation system in the regulation of cell adhesion to vitronectin. The cleavage of vitronectin by uPA and plasmin results in the release of N-terminal vitronectin fragments that can be detected in vivo, underscoring the potential physiological relevance of the process.
Collapse
Affiliation(s)
- Valentina De Lorenzi
- Unit of Cell Matrix Signalling, IFOM The FIRC Institute of Molecular Oncology, Milan, Italy
| | | | - Jeppe B Madsen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Michela Lupia
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| | - Peter A Andreasen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Nicolai Sidenius
- Unit of Cell Matrix Signalling, IFOM The FIRC Institute of Molecular Oncology, Milan, Italy
| |
Collapse
|
14
|
Kashyap AS, Shooter GK, Shokoohmand A, McGovern J, Sivaramakrishnan M, Croll TI, Cane G, Leavesley DI, Söderberg O, Upton Z, Hollier BG. Antagonists of IGF:Vitronectin Interactions Inhibit IGF-I-Induced Breast Cancer Cell Functions. Mol Cancer Ther 2016; 15:1602-13. [PMID: 27196774 DOI: 10.1158/1535-7163.mct-15-0907] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 04/28/2016] [Indexed: 11/16/2022]
Abstract
We provide proof-of-concept evidence for a new class of therapeutics that target growth factor:extracellular matrix (GF:ECM) interactions for the management of breast cancer. Insulin-like growth factor-I (IGF-I) forms multiprotein complexes with IGF-binding proteins (IGFBP) and the ECM protein vitronectin (VN), and stimulates the survival, migration and invasion of breast cancer cells. For the first time we provide physical evidence for IGFBP-3:VN interactions in breast cancer patient tissues; these interactions were predominantly localized to tumor cell clusters and in stroma surrounding tumor cells. We show that disruption of IGF-I:IGFBP:VN complexes with L(27)-IGF-II inhibits IGF-I:IGFBP:VN-stimulated breast cancer cell migration and proliferation in two- and three-dimensional assay systems. Peptide arrays screened to identify regions critical for the IGFBP-3/-5:VN and IGF-II:VN interactions demonstrated IGFBP-3/-5 and IGF-II binds VN through the hemopexin-2 domain, and VN binds IGFBP-3 at residues not involved in the binding of IGF-I to IGFBP-3. IGFBP-interacting VN peptides identified from these peptide arrays disrupted the IGF-I:IGFBP:VN complex, impeded the growth of primary tumor-like spheroids and, more importantly, inhibited the invasion of metastatic breast cancer cells in 3D assay systems. These studies provide first-in-field evidence for the utility of small peptides in antagonizing GF:ECM-mediated biologic functions and present data demonstrating the potential of these peptide antagonists as novel therapeutics. Mol Cancer Ther; 15(7); 1602-13. ©2016 AACR.
Collapse
Affiliation(s)
- Abhishek S Kashyap
- Queensland University of Technology, Tissue Repair and Regeneration Program, Institute of Health and Biomedical Innovation, Brisbane, Queensland, Australia. Cancer Immunology, Department of Biomedicine, University Hospital Basel, Basel, Switzerland.
| | - Gary K Shooter
- Queensland University of Technology, Tissue Repair and Regeneration Program, Institute of Health and Biomedical Innovation, Brisbane, Queensland, Australia
| | - Ali Shokoohmand
- Queensland University of Technology, Tissue Repair and Regeneration Program, Institute of Health and Biomedical Innovation, Brisbane, Queensland, Australia
| | - Jacqui McGovern
- Queensland University of Technology, Tissue Repair and Regeneration Program, Institute of Health and Biomedical Innovation, Brisbane, Queensland, Australia
| | | | - Tristan I Croll
- Queensland University of Technology, Tissue Repair and Regeneration Program, Institute of Health and Biomedical Innovation, Brisbane, Queensland, Australia
| | - Gaëlle Cane
- Department of Immunology, Genetics and Pathology Science for Life Laboratory, BMC, Uppsala University, Uppsala, Sweden
| | - David I Leavesley
- Queensland University of Technology, Tissue Repair and Regeneration Program, Institute of Health and Biomedical Innovation, Brisbane, Queensland, Australia
| | - Ola Söderberg
- Department of Immunology, Genetics and Pathology Science for Life Laboratory, BMC, Uppsala University, Uppsala, Sweden
| | - Zee Upton
- Queensland University of Technology, Tissue Repair and Regeneration Program, Institute of Health and Biomedical Innovation, Brisbane, Queensland, Australia
| | - Brett G Hollier
- Queensland University of Technology, Tissue Repair and Regeneration Program, Institute of Health and Biomedical Innovation, Brisbane, Queensland, Australia. Australian Prostate Cancer Research Centre - Queensland, Institute of Health and Biomedical Innovation, Translational Research Institute, Brisbane, Queensland, Australia.
| |
Collapse
|
15
|
Vitronectin: a promising breast cancer serum biomarker for early diagnosis of breast cancer in patients. Tumour Biol 2016; 37:8909-16. [PMID: 26753956 DOI: 10.1007/s13277-015-4750-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 12/29/2015] [Indexed: 10/22/2022] Open
Abstract
Breast cancer is the most common cancer in women worldwide, identification of new biomarkers for early diagnosis and detection will improve the clinical outcome of breast cancer patients. In the present study, we determined serum levels of vitronectin (VN) in 93 breast cancer patients, 30 benign breast lesions, 9 precancerous lesions, and 30 healthy individuals by enzyme-linked immunosorbent assays. Serum VN level was significantly higher in patients with stage 0-I primary breast cancer than in healthy individuals, patients with benign breast lesion or precancerous lesions, as well as those with breast cancer of higher stages. Serum VN level was significantly and negatively correlated with tumor size, lymph node status, and clinical stage (p < 0.05 in all cases). In addition, VN displayed higher area under curve (AUC) value (0.73, 95 % confidence interval (CI) [0.62-0.84]) than carcinoembryonic antigen (CEA) (0.64, 95 % CI [0.52-0.77]) and cancer antigen 15-3 (CA 15-3) (0.69, 95 % CI [0.58-0.81]) when used to distinguish stage 0-I cancer and normal control. Importantly, the combined use of three biomarkers yielded an improvement in receiver operating characteristic curve with an AUC of 0.83, 95 % CI [0.74-0.92]. Taken together, our current study showed for the first time that serum VN is a promising biomarker for early diagnosis of breast cancer when combined with CEA and CA15-3.
Collapse
|
16
|
Beretov J, Wasinger VC, Millar EKA, Schwartz P, Graham PH, Li Y. Proteomic Analysis of Urine to Identify Breast Cancer Biomarker Candidates Using a Label-Free LC-MS/MS Approach. PLoS One 2015; 10:e0141876. [PMID: 26544852 PMCID: PMC4636393 DOI: 10.1371/journal.pone.0141876] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2014] [Accepted: 10/14/2015] [Indexed: 01/11/2023] Open
Abstract
INTRODUCTION Breast cancer is a complex heterogeneous disease and is a leading cause of death in women. Early diagnosis and monitoring progression of breast cancer are important for improving prognosis. The aim of this study was to identify protein biomarkers in urine for early screening detection and monitoring invasive breast cancer progression. METHOD We performed a comparative proteomic analysis using ion count relative quantification label free LC-MS/MS analysis of urine from breast cancer patients (n = 20) and healthy control women (n = 20). RESULTS Unbiased label free LC-MS/MS-based proteomics was used to provide a profile of abundant proteins in the biological system of breast cancer patients. Data analysis revealed 59 urinary proteins that were significantly different in breast cancer patients compared to the normal control subjects (p<0.05, fold change >3). Thirty-six urinary proteins were exclusively found in specific breast cancer stages, with 24 increasing and 12 decreasing in their abundance. Amongst the 59 significant urinary proteins identified, a list of 13 novel up-regulated proteins were revealed that may be used to detect breast cancer. These include stage specific markers associated with pre-invasive breast cancer in the ductal carcinoma in-situ (DCIS) samples (Leucine LRC36, MAST4 and Uncharacterized protein CI131), early invasive breast cancer (DYH8, HBA, PEPA, uncharacterized protein C4orf14 (CD014), filaggrin and MMRN2) and metastatic breast cancer (AGRIN, NEGR1, FIBA and Keratin KIC10). Preliminary validation of 3 potential markers (ECM1, MAST4 and filaggrin) identified was performed in breast cancer cell lines by Western blotting. One potential marker MAST4 was further validated in human breast cancer tissues as well as individual human breast cancer urine samples with immunohistochemistry and Western blotting, respectively. CONCLUSIONS Our results indicate that urine is a useful non-invasive source of biomarkers and the profile patterns (biomarkers) identified, have potential for clinical use in the detection of BC. Validation with a larger independent cohort of patients is required in the following study.
Collapse
Affiliation(s)
- Julia Beretov
- Cancer Care Centre, St George Hospital, Kogarah, Australia
- St George and Sutherland Clinical School, Faculty of Medicine, University of New South Wales (UNSW), Kensington, Australia
- SEALS, Anatomical Pathology, St George Hospital, Kogarah, Australia
| | - Valerie C. Wasinger
- Bioanalytical Mass Spectrometry Facility, Mark Wainwright Analytical Centre, UNSW, Kensington, Australia
- School of Medical Sciences, UNSW, Kensington, Australia
| | - Ewan K. A. Millar
- SEALS, Anatomical Pathology, St George Hospital, Kogarah, Australia
- School of Medical Sciences, UNSW, Kensington, Australia
- Cancer Research Program, Kinghorn Cancer Centre and Garvan Institute of Medical Research, Darlinghurst, Australia
- School of Medicine and Health Sciences, University of Western Sydney, Campbelltown, Australia
| | - Peter Schwartz
- Breast Surgery, St George Private Hospital, Kogarah, Australia
| | - Peter H. Graham
- Cancer Care Centre, St George Hospital, Kogarah, Australia
- St George and Sutherland Clinical School, Faculty of Medicine, University of New South Wales (UNSW), Kensington, Australia
| | - Yong Li
- Cancer Care Centre, St George Hospital, Kogarah, Australia
- St George and Sutherland Clinical School, Faculty of Medicine, University of New South Wales (UNSW), Kensington, Australia
- * E-mail:
| |
Collapse
|
17
|
Leavesley DI, Kashyap AS, Croll T, Sivaramakrishnan M, Shokoohmand A, Hollier BG, Upton Z. Vitronectin--master controller or micromanager? IUBMB Life 2013; 65:807-18. [PMID: 24030926 DOI: 10.1002/iub.1203] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Accepted: 07/18/2013] [Indexed: 11/07/2022]
Abstract
The concept that the mammalian glycoprotein vitronectin acts as a biological 'glue' and key controller of mammalian tissue repair and remodelling activity is emerging from nearly 50 years of experimental in vitro and in vivo data. Unexpectedly, the vitronectin-knockout (VN-KO) mouse was found to be viable and to have largely normal phenotype. However, diligent observation revealed that the VN-KO animal exhibits delayed coagulation and poor wound healing. This is interpreted to indicate that VN occupies a role in the earliest events of thrombogenesis and tissue repair. VN is the foundation upon which the thrombus grows in an organised structure. In addition to sealing the wound, the thrombus also serves to protect the underlying tissue from oxidation, is a reservoir of mitogens and tissue repair mediators, and provides a provisional scaffold for the repairing tissue. In the absence of VN (e.g., VN-KO animal), this cascade is disrupted before it begins. A wide variety of biologically active species associate with VN. Although initial studies were focused on mitogens, other classes of bioactives (e.g., glycosaminoglycans and metalloproteinases) are now also known to specifically interact with VN. Although some interactions are transient, others are long-lived and often result in multi-protein complexes. Multi-protein complexes provide several advantages: prolonging molecular interactions, sustaining local concentrations, facilitating co-stimulation of cell surface receptors and thereby enhancing cellular/biological responses. We contend that these, or equivalent, multi-protein complexes facilitate VN polyfunctionality in vivo. It is also likely that many of the species demonstrated to associate with VN in vitro, also associate with VN in vivo in similar multi-protein complexes. Thus, the predominant biological function of VN is that of a master controller of the extracellular environment; informing, and possibly instructing cells 'where' to behave, 'when' to behave and 'how' to behave (i.e., appropriately for the current circumstance).
Collapse
Affiliation(s)
- David I Leavesley
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, QLD, Australia
| | | | | | | | | | | | | |
Collapse
|
18
|
Pola C, Formenti SC, Schneider RJ. Vitronectin–αvβ3 Integrin Engagement Directs Hypoxia-Resistant mTOR Activity and Sustained Protein Synthesis Linked to Invasion by Breast Cancer Cells. Cancer Res 2013; 73:4571-8. [DOI: 10.1158/0008-5472.can-13-0218] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
19
|
Direct evidence of the importance of vitronectin and its interaction with the urokinase receptor in tumor growth. Blood 2013; 121:2316-23. [PMID: 23327926 DOI: 10.1182/blood-2012-08-451187] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Extensive evidence implicates the urokinase plasminogen activator receptor (uPAR) in tumor growth, invasion, and metastasis. Recent studies have substantiated the importance of the interaction between uPAR and the extracellular matrix protein vitronectin (VN) for the signaling activity of the receptor in vitro, however, the possible relevance of this interaction for the activity of uPAR in tumor growth and metastasis has not been assessed. We generated a panel of HEK293 cell lines expressing mouse uPAR (muPAR(WT)), an uPAR mutant specifically deficient in VN binding (muPAR(W32A)), and a truncation variant (muPAR(ΔD1)) deficient in both VN and uPA binding. In vitro cells expressing muPAR(WT) display increased cell adhesion, spreading, migration, and proliferation associated with increased p130Cas and MAPK signaling. Disruption of VN binding or ablation of both VN and uPA binding specifically abrogates these activities of uPAR. When xenografted into SCID (severe combined immunodeficiency) mice, the expression of muPAR(WT), but not muPAR(W32A) or muPAR(ΔD1), accelerates tumor development, demonstrating that VN binding is responsible for the tumor-promoting activity of uPAR in vivo. In an orthotopic xenograft model using MDA-MB-231 cells in RAG1(-/-)/VN(-/-) mice, we document that host deficiency in VN strongly impairs tumor formation. These 2 lines of in vivo experimentation independently demonstrate an important role for VN in tumor growth even if the uPAR dependence of the effect in the MDA-MB-231 model remains to be ascertained.
Collapse
|
20
|
Curran CS, Keely PJ. Breast tumor and stromal cell responses to TGF-β and hypoxia in matrix deposition. Matrix Biol 2012; 32:95-105. [PMID: 23262216 DOI: 10.1016/j.matbio.2012.11.016] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2012] [Revised: 11/06/2012] [Accepted: 11/06/2012] [Indexed: 02/07/2023]
Abstract
The components that comprise the extracellular matrix (ECM) are integral to normal tissue homeostasis as well as the development and progression of breast tumors. The secretion, construction, and remodeling of the ECM are each regulated by a complex interplay between tumor cells, fibroblasts and macrophages. Transforming growth factor-β (TGF-β) is an essential molecule in regulating the cellular production of ECM molecules and the adhesive interactions of cells with the ECM. Additionally, hypoxic cell signals, initiated by oxygen deprivation, additional metabolic factors or receptor activation, are associated with ECM formation and the progression of breast cancer. Both TGF-β and hypoxic cell signals are implicated in the functional and morphological changes of cancer-associated-fibroblasts and tumor-associated-macrophages. Moreover, the enhanced recruitment of tumor and stromal cells in response to hypoxia-induced chemokines leads to increased ECM deposition and remodeling, increased blood vessel formation, and enhanced tumor migration. Thus, elucidation of the collaborative networks between tumor and stromal cells in response to the combined signals of TGF-β and hypoxia may yield insight into treatment parameters that target both tumor and stromal cells.
Collapse
Affiliation(s)
- Colleen S Curran
- Laboratory of Cell and Molecular Biology, Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53706, United States.
| | | |
Collapse
|
21
|
Georgoulis A, Havaki S, Drosos Y, Goutas N, Vlachodimitropoulos D, Aleporou-Marinou V, Kittas C, Marinos E, Kouloukoussa M. RGD binding to integrin alphavbeta3 affects cell motility and adhesion in primary human breast cancer cultures. Ultrastruct Pathol 2012. [PMID: 23181508 DOI: 10.3109/01913123.2012.681834] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Integrins mediate cell adhesion to the extracellular matrix. Integrin alphavbeta3 recognizes the RGD motif as a ligand-binding site and has been associated with high malignant potential in breast cancer cells, signaling the onset of widespread metastasis. In recent years, several antagonists of integrin alphavbeta3, including RGD peptides, have been used as potential anti-cancer agents. In the present work, the effect of the linear RGD hexapeptide GRGDSP was studied, for the first time, on breast tumor explants, as well as on well-spread human breast cancer cells from primary cultures, using the explant technique, to clarify the role of this peptide in the suppression of breast cancer cell migration. The results showed that incubation of breast tumor explants with RGD peptide at the beginning of culture development inhibited completely the migration of cancer cells out of the tissue fragment as revealed by electron microscopy. RGD incubation of well-spread breast cancer cells from primary culture resulted in rounding and shrinkage of the cells accompanied by altered distribution of integrin alphavbeta3 and concomitant F-actin cytoskeletal disorganization, as revealed by immunofluorescence. Electron immunocytochemistry showed aggregation of integrin alphavbeta3 at the cell periphery and its detection in noncoated vesicles. However, Western immunoblotting showed no change in beta3 subunit expression, despite the altered distribution of the integrin alphavbeta3. In light of the above, it appears that the RGD peptide plays an important role in the modulation of cell motility and in the perturbation of cell attachment affecting the malignant potential of breast cancer cells in primary cultures.
Collapse
Affiliation(s)
- Anastasios Georgoulis
- Laboratory of Histology and Embryology, Medical School, University of Athens, Athens, Greece
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
SHEN JING, MA XIAOYAN, YI FEI, RASUL AZHAR, CUI MANHUA, MA TONGHUI. Increased expression levels of vitronectin in the maternal-fetal interface of placenta in early-onset severe preeclampsia. Mol Med Rep 2012; 7:53-8. [DOI: 10.3892/mmr.2012.1141] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2012] [Accepted: 09/25/2012] [Indexed: 11/05/2022] Open
|
23
|
Cartier-Michaud A, Malo M, Charrière-Bertrand C, Gadea G, Anguille C, Supiramaniam A, Lesne A, Delaplace F, Hutzler G, Roux P, Lawrence DA, Barlovatz-Meimon G. Matrix-bound PAI-1 supports cell blebbing via RhoA/ROCK1 signaling. PLoS One 2012; 7:e32204. [PMID: 22363817 PMCID: PMC3283740 DOI: 10.1371/journal.pone.0032204] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2011] [Accepted: 01/24/2012] [Indexed: 11/19/2022] Open
Abstract
The microenvironment of a tumor can influence both the morphology and the behavior of cancer cells which, in turn, can rapidly adapt to environmental changes. Increasing evidence points to the involvement of amoeboid cell migration and thus of cell blebbing in the metastatic process; however, the cues that promote amoeboid cell behavior in physiological and pathological conditions have not yet been clearly identified. Plasminogen Activator Inhibitor type-1 (PAI-1) is found in high amount in the microenvironment of aggressive tumors and is considered as an independent marker of bad prognosis. Here we show by immunoblotting, activity assay and immunofluorescence that, in SW620 human colorectal cancer cells, matrix-associated PAI-1 plays a role in the cell behavior needed for amoeboid migration by maintaining cell blebbing, localizing PDK1 and ROCK1 at the cell membrane and maintaining the RhoA/ROCK1/MLC-P pathway activation. The results obtained by modeling PAI-1 deposition around tumors indicate that matrix-bound PAI-1 is heterogeneously distributed at the tumor periphery and that, at certain spots, the elevated concentrations of matrix-bound PAI-1 needed for cancer cells to undergo the mesenchymal-amoeboid transition can be observed. Matrix-bound PAI-1, as a matricellular protein, could thus represent one of the physiopathological requirements to support metastatic formation.
Collapse
Affiliation(s)
| | - Michel Malo
- IBISC EA 4526, Evry Val d'Essonne University, Evry, France
| | - Cécile Charrière-Bertrand
- IBISC EA 4526, Evry Val d'Essonne University, Evry, France
- University Paris-Est Créteil, Créteil, France
| | - Gilles Gadea
- CRBM UMR 5237 CNRS, Montpellier University, Montpellier, France
| | | | | | - Annick Lesne
- Institut des Hautes Etudes Scientifiques, Bures-sur-Yvette, France
- LPTMC UMR 7600 CNRS, Paris, France
| | | | | | - Pierre Roux
- CRBM UMR 5237 CNRS, Montpellier University, Montpellier, France
| | - Daniel A. Lawrence
- Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor, Michigan, United States of America
| | - Georgia Barlovatz-Meimon
- IBISC EA 4526, Evry Val d'Essonne University, Evry, France
- University Paris-Est Créteil, Créteil, France
- * E-mail:
| |
Collapse
|
24
|
PAI-1: An Integrator of Cell Signaling and Migration. Int J Cell Biol 2011; 2011:562481. [PMID: 21837240 PMCID: PMC3151495 DOI: 10.1155/2011/562481] [Citation(s) in RCA: 136] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2011] [Revised: 05/09/2011] [Accepted: 05/17/2011] [Indexed: 12/23/2022] Open
Abstract
Cellular migration, over simple surfaces or through complex stromal barriers, requires coordination between detachment/re-adhesion cycles, involving structural components of the extracellular matrix and their surface-binding elements (integrins), and the precise regulation of the pericellular proteolytic microenvironment. It is now apparent that several proteases and protease inhibitors, most notably urokinase plasminogen activator (uPA) and plasminogen activator inhibitor type-1 (PAI-1), also interact with several cell surface receptors transducing intracellular signals that significantly affect both motile and proliferative programs. These events appear distinct from the original function of uPA/PAI-1 as modulators of the plasmin-based proteolytic cascade. The multifaceted interactions of PAI-1 with specific matrix components (i.e., vitronectin), the low-density lipoprotein receptor-related protein-1 (LRP1), and the uPA/uPA receptor complex have dramatic consequences on the migratory phenotype and may underlie the pathophysiologic sequalae of PAI-1 deficiency and overexpression. This paper focuses on the increasingly intricate role of PAI-1 as a major mechanistic determinant of the cellular migratory phenotype.
Collapse
|
25
|
Kadowaki M, Sangai T, Nagashima T, Sakakibara M, Yoshitomi H, Takano S, Sogawa K, Umemura H, Fushimi K, Nakatani Y, Nomura F, Miyazaki M. Identification of vitronectin as a novel serum marker for early breast cancer detection using a new proteomic approach. J Cancer Res Clin Oncol 2011; 137:1105-15. [PMID: 21253761 DOI: 10.1007/s00432-010-0974-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2010] [Accepted: 12/28/2010] [Indexed: 01/22/2023]
Abstract
PURPOSE Breast cancer is the most frequent malignancy in women. However, no useful serum markers with high sensitivity and specificity for the detection of early breast cancer have been identified. The search for biological markers of early breast cancer is of continual interest in experimental and clinical breast cancer research. We recently described a simple and highly reproducible three-step proteome analysis for identifying potential disease-marker candidates among the low-abundance serum proteins. METHODS Serum samples from breast ductal carcinoma in situ (DCIS) patients and normal controls were subjected to a three-step serum proteome analysis. The steps were the following: first, immunodepletion of most abundant proteins; second, fractionation using reverse-phase high-performance liquid chromatography; and third, separation using two-dimensional electrophoresis (2-DE). Differences revealed by protein staining were further confirmed by Western blotting, immunohistochemical staining, and enzyme-linked immunosorbent assays (ELISA). RESULTS Twenty-two upregulated and 26 downregulated spots were detected on the 2-DE gels, and a total of 33 proteins were identified by liquid chromatography and tandem mass spectrometry. Western blotting confirmed that the level of vitronectin was significantly increased in DCIS patients compared with that of normal controls. Immunohistochemical staining of vitronectin in breast cancer tissue revealed high expression in small vessel walls surrounding cancer cells and the extracellular matrix of stroma. Moreover, vitronectin serum concentrations, as measured by ELISA, were significantly increased in patients with DCIS or more advanced breast cancer compared with those of normal controls. CONCLUSIONS Vitronectin could serve as a promising serum marker for the detection of primary breast cancer.
Collapse
MESH Headings
- Aged
- Biomarkers, Tumor/blood
- Blotting, Western
- Breast Neoplasms/blood
- Breast Neoplasms/diagnosis
- Carcinoma, Ductal, Breast/blood
- Carcinoma, Ductal, Breast/diagnosis
- Carcinoma, Intraductal, Noninfiltrating/blood
- Carcinoma, Intraductal, Noninfiltrating/diagnosis
- Early Detection of Cancer
- Electrophoresis, Gel, Two-Dimensional
- Enzyme-Linked Immunosorbent Assay
- Female
- Humans
- Middle Aged
- Proteome/analysis
- Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
- Vitronectin/blood
Collapse
Affiliation(s)
- Masami Kadowaki
- Department of General Surgery, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8670, Japan.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Blouse GE, Dupont DM, Schar CR, Jensen JK, Minor KH, Anagli JY, Gårdsvoll H, Ploug M, Peterson CB, Andreasen PA. Interactions of plasminogen activator inhibitor-1 with vitronectin involve an extensive binding surface and induce mutual conformational rearrangements. Biochemistry 2010; 48:1723-35. [PMID: 19193026 DOI: 10.1021/bi8017015] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
In order to explore early events during the association of plasminogen activator inhibitor-1 (PAI-1) with its cofactor vitronectin, we have applied a robust strategy that combines protein engineering, fluorescence spectroscopy, and rapid reaction kinetics. Fluorescence stopped-flow experiments designed to monitor the rapid association of PAI-1 with vitronectin indicate a fast, concentration-dependent, biphasic binding of PAI-1 to native vitronectin but only a monophasic association with the somatomedin B (SMB) domain, suggesting that multiple phases of the binding interaction occur only when full-length vitronectin is present. Nonetheless, in all cases, the initial fast interaction is followed by slower fluorescence changes attributed to a conformational change in PAI-1. Complementary experiments using an engineered, fluorescently silent PAI-1 with non-natural amino acids showed that concomitant structural changes occur as well in native vitronectin. Furthermore, we have measured the effect of vitronectin on the rate of insertion of the reactive center loop into beta-sheet A of PAI-1 during reaction with target proteases. With a variety of PAI-1 variants, we observe that both full-length vitronectin and the SMB domain have protease-specific effects on the rate of loop insertion but that the two exhibit clearly different effects. These results support a model for PAI-1 binding to vitronectin in which the interaction surface extends beyond the region of PAI-1 occupied by the SMB domain. In support of this model are recent results that define a PAI-1-binding site on vitronectin that lies outside the somatomedin B domain (Schar, C. R., Blouse, G. E., Minor, K. H., and Peterson, C. B. (2008) J. Biol. Chem. 283, 10297-10309) and the complementary site on PAI-1 (Schar, C. R., Jensen, J. K., Christensen, A., Blouse, G. E., Andreasen, P. A., and Peterson, C. B. (2008) J. Biol. Chem. 283, 28487-28496).
Collapse
Affiliation(s)
- Grant E Blouse
- Laboratory of Cellular Protein Science, Department of Molecular Biology, University of Aarhus, Gustav Wieds Vej 10C, DK-8000 Arhus C, Denmark
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Francischetti IMB, Kotsyfakis M, Andersen JF, Lukszo J. Cyr61/CCN1 displays high-affinity binding to the somatomedin B(1-44) domain of vitronectin. PLoS One 2010; 5:e9356. [PMID: 20195466 PMCID: PMC2829074 DOI: 10.1371/journal.pone.0009356] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2009] [Accepted: 01/22/2010] [Indexed: 01/20/2023] Open
Abstract
Background Cyr61 is a member of the CCN (Cyr61, connective tissue growth, NOV) family of extracellular-associated (matricellular) proteins that present four distinct functional modules, namely insulin-like growth factor binding protein (IGFBP), von Willebrand factor type C (vWF), thrombospondin type 1 (TSP), and C-terminal growth factor cysteine knot (CT) domain. While heparin sulphate proteoglycans reportedly mediate the interaction of Cyr61 with the matrix and cell surface, the role of other extracellular associated proteins has not been revealed. Methods and Findings In this report, surface plasmon resonance (SPR) experiments and solid-phase binding assays demonstrate that recombinant Cyr61 interacts with immobilized monomeric or multimeric vitronectin (VTNC) with KD in the nanomolar range. Notably, the binding site for Cyr61 was identified as the somatomedin B domain (SMTB 1–44) of VTNC, which mediates its interaction with PAI-1, uPAR, and integrin αvβ3. Accordingly, PAI-1 outcompetes Cyr61 for binding to immobilized SMTB 1–44, and Cyr61 attenuates uPAR-mediated U937 adhesion to VTNC. In contrast, isothermal titration calorimetry shows that Cyr61 does not display high-affinity binding for SMTB 1-44 in solution. Nevertheless, competitive ELISA revealed that multimeric VTNC, heat-modified monomeric VTNC, or SMTB 1–44 at high concentrations attenuate Cyr61 binding to immobilized VTNC, while monomeric VTNC was ineffective. Therefore, immobilization of VTNC exposes cryptic epitopes that recognize Cyr61 with high affinity, as reported for a number of antibodies, β-endorphin, and other molecules. Conclusions The finding that Cyr61 interacts with the SMTB 1–44 domain suggests that VTNC represent a point of anchorage for CCN family members to the matrix. Results are discussed in the context of the role of CCN and VTNC in matrix biology and angiogenesis.
Collapse
Affiliation(s)
- Ivo M B Francischetti
- Section of Vector Biology, Laboratory of Malaria and Vector Research, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America.
| | | | | | | |
Collapse
|
28
|
Hollier BG, Kricker JA, Van Lonkhuyzen DR, Leavesley DI, Upton Z. Substrate-bound insulin-like growth factor (IGF)-I-IGF binding protein-vitronectin-stimulated breast cell migration is enhanced by coactivation of the phosphatidylinositide 3-Kinase/AKT pathway by alphav-integrins and the IGF-I receptor. Endocrinology 2008; 149:1075-90. [PMID: 18079201 DOI: 10.1210/en.2007-0740] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
IGF-I can bind to the extracellular matrix protein vitronectin (VN) through the involvement of IGF-binding proteins-2, -3, -4, and -5. Because IGF-I and VN have established roles in tumor cell dissemination, we were keen to investigate the functional consequences of the interaction of IGF-I, IGF binding proteins (IGFBPs), and VN in tumor cell biology. Hence, functional responses of MCF-7 breast carcinoma cells and normal nontumorgenic MCF-10A mammary epithelial cells were investigated to allow side-by-side comparisons of these complexes in both cancerous and normal breast cells. We demonstrate that substrate-bound IGF-I-IGFBP-VN complexes stimulate synergistic increases in cellular migration in both cell types. Studies using IGF-I analogs determined this stimulation to be dependent on both heterotrimeric IGF-I-IGFBP-VN complex formation and the involvement of the IGF-I receptor (IGF-IR). Furthermore, the enhanced cellular migration was abolished on incubation of MCF-7 and MCF-10A cells with function blocking antibodies directed at VN-binding integrins and the IGF-IR. Analysis of the signal transduction pathways underlying the enhanced cell migration revealed that the complexes stimulate a transient activation of the ERK/MAPK signaling pathway while simultaneously producing a sustained activation of the phosphatidylinositide 3-kinase/AKT pathway. Experiments using pharmacological inhibitors of these pathways determined a requirement for phosphatidylinositide 3-kinase/AKT activation in the observed response. Overexpression of wild type and activated AKT further increases substrate-bound IGF-I-IGFBP-VN-stimulated migration. This study provides the first mechanistic insights into the action of IGF-I-IGFBP-VN complexes and adds further evidence to support the involvement of VN-binding integrins and their cooperativity with the IGF-IR in the promotion of tumor cell migration.
Collapse
Affiliation(s)
- Brett G Hollier
- Tissue Repair and Regeneration ProgramInstitute of Health and Biomedical Innovation, Queensland University of Technology, 60 Musk Avenue, Kelvin Grove, Queensland 4059, Australia.
| | | | | | | | | |
Collapse
|
29
|
Ghosh MC, Grass L, Soosaipillai A, Sotiropoulou G, Diamandis EP. Human kallikrein 6 degrades extracellular matrix proteins and may enhance the metastatic potential of tumour cells. Tumour Biol 2005; 25:193-9. [PMID: 15557757 DOI: 10.1159/000081102] [Citation(s) in RCA: 92] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2004] [Accepted: 07/07/2004] [Indexed: 11/19/2022] Open
Abstract
Human kallikrein 6 (hK6), a trypsin-like serine protease, is a newly identified member of the kallikrein gene family. Its involvement in inflammatory CNS lesions and in demyelination has been reported. Recent work has suggested that expression of this enzyme is significantly elevated in patients with ovarian cancer. We have identified many tumour cell lines that secrete hK6, but its physiological role is unknown. Here, we try to unveil the role of this kallikrein in the metastasis and invasion of tumour cells. We demonstrate that purified human recombinant hK6 can cleave gelatin in zymography and can efficiently degrade high-molecular-weight extracellular matrix proteins such as fibronectin, laminin, vitronectin and collagen. In Boyden chamber assays, we found that tumour cells treated with a neutralizing hK6 antibody migrate less than control cells. We conclude that hK6 might play a role in the invasion and metastasis of tumour cells and may be a candidate therapeutic target.
Collapse
Affiliation(s)
- Manik C Ghosh
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Canada
| | | | | | | | | |
Collapse
|
30
|
Väisänen T, Väisänen MR, Autio-Harmainen H, Pihlajaniemi T. Type XIII collagen expression is induced during malignant transformation in various epithelial and mesenchymal tumours. J Pathol 2005; 207:324-35. [PMID: 16110459 DOI: 10.1002/path.1836] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Little information is available on the expression of transmembrane type XIII collagen in human diseases. The present study has investigated the expression of this collagen in cancer, in particular during malignant transformation. By combining the tissue microarray technique with in situ hybridization, a consistent pattern of clearly increased type XIII collagen mRNA expression was found in the stromal compartment of epithelial tumours and throughout mesenchymal tumours. Slightly elevated mRNA expression was observed in dysplastic samples and in malignant epithelial cells. It is also demonstrated that factors secreted into the culture medium by tumour cells, in particular the growth factor TGF-beta, contribute to the induction of type XIII collagen expression, and trigger concomitantly a profound phenotypic and morphological transition of cultured primary fibroblasts. Reciprocally, type XIII collagen may alter the growth milieu of malignant cells as the soluble type XIII collagen ectodomain influenced the adherence and spreading of cells cultured on vitronectin-rich matrix. It is proposed that malignant transformation stimulates the expression of type XIII collagen, particularly in the tumour stroma and to a lesser extent in the epithelium, and that this high type XIII collagen expression may contribute to tumour progression and behaviour by modulating cell-matrix interactions.
Collapse
Affiliation(s)
- Timo Väisänen
- Collagen Research Unit, Biocenter Oulu and Department of Medical Biochemistry and Molecular Biology, University of Oulu, PO Box 5000, 90014 University of Oulu, Finland
| | | | | | | |
Collapse
|
31
|
Väisänen MR, Väisänen T, Pihlajaniemi T. The shed ectodomain of type XIII collagen affects cell behaviour in a matrix-dependent manner. Biochem J 2004; 380:685-93. [PMID: 15005656 PMCID: PMC1224210 DOI: 10.1042/bj20031974] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2003] [Revised: 02/26/2004] [Accepted: 03/09/2004] [Indexed: 11/17/2022]
Abstract
Transmembrane type XIII collagen resides in adhesive structures of cells and tissues, and has therefore been implicated in cell adhesion and in adhesion-dependent cell functions. This collagen also exists as a soluble protein in the pericellular matrix, as the ectodomain is released from the plasma membrane by proteolytic cleavage. Analysis with various protease inhibitors led to confirmation of the furin family of proprotein convertases as the protease group responsible for the shedding of the ectodomain, cleaving at a site conforming to the consensus sequence for the proprotein convertases at the stem of the ectodomain. Both the trans -Golgi network and the plasma membrane were used as cleavage locations. Mammalian cells employed various intracellular mechanisms to modulate shedding of the ectodomain, all resulting in a similar cleavage event. Cell detachment from the underlying substratum was also found to augment the excision. The released ectodomain rendered the pericellular surroundings less supportive of cell adhesion, migration and proliferation, as seen specifically on a vitronectin substratum. Type XIII collagen ectodomain shedding thus resulted in the formation of a soluble, biologically active molecule, which eventually modulated cell behaviour in a reciprocal and substratum-specific manner. The dual existence of membrane-bound and soluble variants widens our biological understanding of type XIII collagen.
Collapse
Affiliation(s)
- Marja-Riitta Väisänen
- Collagen Research Unit, Biocenter Oulu and Department of Medical Biochemistry and Molecular Biology, University of Oulu, P.O. Box 5000, 90014 Oulu, Finland
| | | | | |
Collapse
|
32
|
Wilkins-Port CE, Sanderson RD, Tominna-Sebald E, McKeown-Longo PJ. Vitronectin's basic domain is a syndecan ligand which functions in trans to regulate vitronectin turnover. ACTA ACUST UNITED AC 2004; 10:85-103. [PMID: 14681059 DOI: 10.1080/cac.10.2.85.103] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
During the process of tissue remodeling, vitronectin (Vn) is deposited in the extracellular matrix where it plays a key role in the regulation of pericellular proteolysis and cell motility. In previous studies we have shown that extracellular levels of vitronectin are controlled by receptor-mediated endocytosis and that this process is dependent upon vitronectin binding to sulfated proteoglycans. We have now identified vitronectin's 12 amino acid "basic domain" which is contained within the larger 40 amino acid heparin binding domain, as a syndecan binding site. Recombinant vitronectins representing wild type vitronectin (rVn) and vitronectin with the basic domain deleted (rVnDelta347-358) were prepared in a baculoviral expression system. The rVn as well as a glutathione S-transferase (GST) fusion protein, consisting of vitronectin's 40 amino acid heparin binding domain (GST-VnHBD), exhibited dose dependent binding to HT-1080 cell surfaces, which was attenuated following deletion of the basic domain. In addition, GST-VnHBD supported both HT-1080 and dermal fibroblast cell adhesion, which was also dependent upon the basic domain. Similarly, ARH-77 cells transfected with syndecans -1, -2, or -4, but not Glypican-1, adhered to GST-VnHBD coated wells, while adhesion of these same cells was lost following deletion of the basic domain. HT-1080 cells were unable to degrade rVnDelta347-358. Degradation of rVnDelta347-358 was completely recovered in the presence of GST-VnHBD but not in the presence of GST-VnHBDDelta347-358. These results indicate that turnover of soluble vitronectin requires ligation of vitronectin's basic domain and that this binding event can work in trans to regulate vitronectin degradation.
Collapse
Affiliation(s)
- Cynthia E Wilkins-Port
- Center for Cell Biology and Cancer Research, Neil Hellman Medical Research Building, Albany Medical College, 47 New Scotland Avenue, Albany, NY 12208, USA
| | | | | | | |
Collapse
|
33
|
Mayasundari A, Whittemore NA, Serpersu EH, Peterson CB. The Solution Structure of the N-terminal Domain of Human Vitronectin. J Biol Chem 2004; 279:29359-66. [PMID: 15123712 DOI: 10.1074/jbc.m401279200] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The three-dimensional structure of an N-terminal fragment comprising the first 51 amino acids from human plasma vitronectin, the somatomedin B (SMB) domain, has been determined by two-dimensional NMR approaches. An average structure was calculated, representing the overall fold from a set of 20 minimized structures. The core residues (18-41) overlay with a root mean square deviation of 2.29 +/- 0.62 A. The N- and C-terminal segments exhibit higher root mean square deviations, reflecting more flexibility in solution and/or fewer long-range NOEs for these regions. Residues 26-30 form a unique single-turn alpha-helix, the locus where plasminogen activator inhibitor type-1 (PAI-1) is bound. This structure of this helix is highly homologous with that of a recombinant SMB domain solved in a co-crystal with PAI-1 (Zhou, A., Huntington, J. A., Pannu, N. S., Carrell, R. W., and Read, R. J. (2003) Nat. Struct. Biol. 10, 541-544), although the remainder of the structure differs. Significantly, the pattern of disulfide cross-links observed in this material isolated from human plasma is altogether different from the disulfides proposed for recombinant forms. The NMR structure reveals the relative orientation of binding sites for cell surface receptors, including an integrin-binding site at residues 45-47, which was disordered and did not diffract in the co-crystal, and a site for the urokinase receptor, which overlaps with the PAI-1-binding site.
Collapse
Affiliation(s)
- Anand Mayasundari
- Department of Biochemistry and Cellular and Molecular Biology and the Center of Excellence in Structural Biology, University of Tennessee, Knoxville, Tennessee 37996, USA
| | | | | | | |
Collapse
|