1
|
Hamada M, Varkoly KS, Riyadh O, Beladi R, Munuswamy-Ramanujam G, Rawls A, Wilson-Rawls J, Chen H, McFadden G, Lucas AR. Urokinase-Type Plasminogen Activator Receptor (uPAR) in Inflammation and Disease: A Unique Inflammatory Pathway Activator. Biomedicines 2024; 12:1167. [PMID: 38927374 PMCID: PMC11201033 DOI: 10.3390/biomedicines12061167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 04/24/2024] [Accepted: 05/10/2024] [Indexed: 06/28/2024] Open
Abstract
The urokinase-type plasminogen activator receptor (uPAR) is a unique protease binding receptor, now recognized as a key regulator of inflammation. Initially, uPA/uPAR was considered thrombolytic (clot-dissolving); however, recent studies have demonstrated its predominant immunomodulatory functions in inflammation and cancer. The uPA/uPAR complex has a multifaceted central role in both normal physiological and also pathological responses. uPAR is expressed as a glycophosphatidylinositol (GPI)-linked receptor interacting with vitronectin, integrins, G protein-coupled receptors, and growth factor receptors within a large lipid raft. Through protein-to-protein interactions, cell surface uPAR modulates intracellular signaling, altering cellular adhesion and migration. The uPA/uPAR also modifies extracellular activity, activating plasminogen to form plasmin, which breaks down fibrin, dissolving clots and activating matrix metalloproteinases that lyse connective tissue, allowing immune and cancer cell invasion and releasing growth factors. uPAR is now recognized as a biomarker for inflammatory diseases and cancer; uPAR and soluble uPAR fragments (suPAR) are increased in viral sepsis (COVID-19), inflammatory bowel disease, and metastasis. Here, we provide a comprehensive overview of the structure, function, and current studies examining uPAR and suPAR as diagnostic markers and therapeutic targets. Understanding uPAR is central to developing diagnostic markers and the ongoing development of antibody, small-molecule, nanogel, and virus-derived immune-modulating treatments that target uPAR.
Collapse
Affiliation(s)
- Mostafa Hamada
- College of Medicine, Kansas City University, 1750 Independence Ave, Kansas City, MO 64106, USA; (M.H.); (O.R.)
| | - Kyle Steven Varkoly
- Department of Internal Medicine, McLaren Macomb Hospital, Michigan State University College of Human Medicine, 1000 Harrington St., Mt Clemens, MI 48043, USA
| | - Omer Riyadh
- College of Medicine, Kansas City University, 1750 Independence Ave, Kansas City, MO 64106, USA; (M.H.); (O.R.)
| | - Roxana Beladi
- Department of Neurosurgery, Ascension Providence Hospital, Michigan State University College of Human Medicine, 16001 W Nine Mile Rd, Southfield, MI 48075, USA;
| | - Ganesh Munuswamy-Ramanujam
- Molecular Biology and Immunobiology Division, Interdisciplinary Institute of Indian System of Medicine, SRM Institute of Science and Technology, Kattankulathur 603203, India;
| | - Alan Rawls
- School of Life Sciences, Arizona State University, 427 E Tyler Mall, Tempe, AZ 85281, USA; (A.R.); (J.W.-R.)
| | - Jeanne Wilson-Rawls
- School of Life Sciences, Arizona State University, 427 E Tyler Mall, Tempe, AZ 85281, USA; (A.R.); (J.W.-R.)
| | - Hao Chen
- Department of Tumor Center, Lanzhou University Second Hospital, Lanzhou 730030, China;
| | - Grant McFadden
- Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, 727 E Tyler St., Tempe, AZ 85287, USA;
| | - Alexandra R. Lucas
- Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, 727 E Tyler St., Tempe, AZ 85287, USA;
| |
Collapse
|
2
|
Cecchi M, Anceschi C, Silvano A, Coniglio ML, Chinnici A, Magnelli L, Lapucci A, Laurenzana A, Parenti A. Unveiling the Role of Tryptophan 2,3-Dioxygenase in the Angiogenic Process. Pharmaceuticals (Basel) 2024; 17:558. [PMID: 38794128 PMCID: PMC11124529 DOI: 10.3390/ph17050558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 04/23/2024] [Accepted: 04/24/2024] [Indexed: 05/26/2024] Open
Abstract
BACKGROUND Indoleamine 2,3-dioxygenase (IDO1) and tryptophan-2,3-dioxygenase (TDO) are the two principals enzymes involved in the catabolization of tryptophan (Trp) into kynurenine (Kyn). Despite their well-established role in the immune escape, their involvement in angiogenesis remains uncertain. We aimed to characterize TDO and IDO1 in human umbilical venular endothelial cells (HUVECs) and human endothelial colony-forming cells (ECFCs). METHODS qRT-PCR and immunofluorescence were used for TDO and IDO1 expression while their activity was measured using ELISA assays. Cell proliferation was examined via MTT tests and in in vitro angiogenesis by capillary morphogenesis. RESULTS HUVECs and ECFCs expressed TDO and IDO1. Treatment with the selective TDO inhibitor 680C91 significantly impaired HUVEC proliferation and 3D-tube formation in response to VEGF-A, while IDO1 inhibition showed no effect. VEGF-induced mTor phosphorylation and Kyn production were hindered by 680C91. ECFC morphogenesis was also inhibited by 680C91. Co-culturing HUVECs with A375 induced TDO up-regulation in both cell types, whose inhibition reduced MMP9 activity and prevented c-Myc and E2f1 upregulation. CONCLUSIONS HUVECs and ECFCs express the key enzymes of the kynurenine pathway. Significantly, TDO emerges as a pivotal player in in vitro proliferation and capillary morphogenesis, suggesting a potential pathophysiological role in angiogenesis beyond its well-known immunomodulatory effects.
Collapse
Affiliation(s)
- Marta Cecchi
- Department of Neuroscience, Psychology, Drug Research and Child Health, (NEUROFARBA) Pharmacology and Toxicology Section, University of Florence, 50139 Florence, Italy; (M.C.); (A.C.)
| | - Cecilia Anceschi
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50121 Florence, Italy; (C.A.); (L.M.); (A.L.)
| | - Angela Silvano
- Department of Health Sciences, Division of Obstetrics and Gynecology, Careggi Hospital, University of Florence, 50134 Florence, Italy;
| | - Maria Luisa Coniglio
- Centre of Excellence, Division of Pediatric Oncology/Hematology, Meyer Children’s Hospital IRCCS, 50139 Florence, Italy;
| | - Aurora Chinnici
- Department of Neuroscience, Psychology, Drug Research and Child Health, (NEUROFARBA) Pharmacology and Toxicology Section, University of Florence, 50139 Florence, Italy; (M.C.); (A.C.)
- Centre of Excellence, Division of Pediatric Oncology/Hematology, Meyer Children’s Hospital IRCCS, 50139 Florence, Italy;
| | - Lucia Magnelli
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50121 Florence, Italy; (C.A.); (L.M.); (A.L.)
| | - Andrea Lapucci
- Department of Health Sciences, Clinical Pharmacology and Oncology Section, University of Florence, V. le G. Pieraccini, 6, 50139 Florence, Italy
| | - Anna Laurenzana
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50121 Florence, Italy; (C.A.); (L.M.); (A.L.)
| | - Astrid Parenti
- Department of Health Sciences, Clinical Pharmacology and Oncology Section, University of Florence, V. le G. Pieraccini, 6, 50139 Florence, Italy
| |
Collapse
|
3
|
Penkov D, Beloglazova I, Parfyonova Y. Endothelial-specific Enhancer as a Cis Element of PLAUR Regulation by TNF-alpha, IL-1beta, and VEGF. Curr Pharm Des 2024; 30:1630-1640. [PMID: 38715331 DOI: 10.2174/0113816128296376240424072322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Accepted: 04/03/2024] [Indexed: 01/23/2025]
Abstract
The expression of human PLAUR gene, which encodes the urokinase plasminogen activator receptor (uPAR), is cell- and process-specific and elevated in inflammation, cancer and senescence. Its tight regulation is achieved by regulatory elements in the gene locus, such as the promoter and several enhancers. The promoter activity is not specific to a particular cell type and has been described earlier. The proximal enhancer is endothelial-specific and responsible for the PLAUR expression pattern in endothelial cells. In this study we described the enhancer activity and its cis-regulatory elements based on the published data. We showed a possible connection of the enhancer activity with known cellular phenotypes.
Collapse
Affiliation(s)
- Dmitry Penkov
- Laboratory of Angiogenesis, Institute of Experimental Cardiology Named after Academician V.N. Smirnov, Federal State Budgetary Institution National Medical Research Center of Cardiology Named after Academician E.I. Chazov, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Irina Beloglazova
- Laboratory of Angiogenesis, Institute of Experimental Cardiology Named after Academician V.N. Smirnov, Federal State Budgetary Institution National Medical Research Center of Cardiology Named after Academician E.I. Chazov, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Yelena Parfyonova
- Laboratory of Angiogenesis, Institute of Experimental Cardiology Named after Academician V.N. Smirnov, Federal State Budgetary Institution National Medical Research Center of Cardiology Named after Academician E.I. Chazov, Ministry of Health of the Russian Federation, Moscow, Russia
| |
Collapse
|
4
|
Schwarz N, Yadegari H. Potentials of Endothelial Colony-Forming Cells: Applications in Hemostasis and Thrombosis Disorders, from Unveiling Disease Pathophysiology to Cell Therapy. Hamostaseologie 2023; 43:325-337. [PMID: 37857295 DOI: 10.1055/a-2101-5936] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2023] Open
Abstract
Endothelial colony-forming cells (ECFCs) are endothelial progenitor cells circulating in a limited number in peripheral blood. They can give rise to mature endothelial cells (ECs) and, with intrinsically high proliferative potency, contribute to forming new blood vessels and restoring the damaged endothelium in vivo. ECFCs can be isolated from peripheral blood or umbilical cord and cultured to generate large amounts of autologous ECs in vitro. Upon differentiation in culture, ECFCs are excellent surrogates for mature ECs showing the same phenotypic, genotypic, and functional features. In the last two decades, the ECFCs from various vascular disease patients have been widely used to study the diseases' pathophysiology ex vivo and develop cell-based therapeutic approaches, including vascular regenerative therapy, tissue engineering, and gene therapy. In the current review, we will provide an updated overview of past studies, which have used ECFCs to elucidate the molecular mechanisms underlying the pathogenesis of hemostatic disorders in basic research. Additionally, we summarize preceding studies demonstrating the utility of ECFCs as cellular tools for diagnostic or therapeutic clinical applications in thrombosis and hemostasis.
Collapse
Affiliation(s)
- Nadine Schwarz
- Institute of Experimental Hematology and Transfusion Medicine, University Hospital Bonn, Bonn, Germany
| | - Hamideh Yadegari
- Institute of Experimental Hematology and Transfusion Medicine, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
5
|
Chillà A, Anceschi C, Frediani E, Scavone F, Del Rosso T, Pelagio G, Tufaro A, De Palma G, Del Rosso M, Fibbi G, Chiarugi P, Laurenzana A, Margheri F. Inhibition of MMPs supports amoeboid angiogenesis hampering VEGF-targeted therapies via MLC and ERK 1/2 signaling. J Transl Med 2023; 21:102. [PMID: 36759828 PMCID: PMC9912547 DOI: 10.1186/s12967-023-03954-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 02/01/2023] [Indexed: 02/11/2023] Open
Abstract
BACKGROUND In the past decades studies on anti-tumoral drugs inhibiting matrix metalloproteinase (MMPs) were disappointing. Recently, we demonstrated that mature endothelial cells (ECs) and endothelial colony forming cells (ECFCs) can switch between invasion modes to cope with challenging environments, performing the "amoeboid angiogenesis" in the absence of proteases activity. METHODS We first set out to investigate by ELISA if the inhibitors of the main protease family involved in angiogenesis were differently expressed during breast cancer progression. We used Marimastat, a broad-spectrum MMP inhibitor, as a means of inducing amoeboid characteristics and studied VEGF role in amoeboid angiogenesis. Thus, we performed invasion and capillary morphogenesis assay, morphological, cell signaling and in vivo mouse studies. RESULTS Our data showed that TIMP1, TIMP2, alpha2-antiplasmin, PAI-1 and cystatin increase in breast cancer serum of patients with primary cancer and lymph node positive compared to healthy women. In vitro results revealed that the most high-powered protease inhibitors able to induce amoeboid invasion of ECFCs were TIMP1, 2 and 3. Surprisingly, Marimastat promotes ECFC invasion and tubular formation in vitro and in vivo, inducing amoeboid characteristics. We observed that the combination of Marimastat plus VEGF doesn't boost neither cell invasion nor vessel formation capacity. Moreover, inhibition of VEGF activity with Bevacizumab in the presence of Marimastat confirmed that amoeboid angiogenesis is independent from the stimulus of the main vascular growth factor, VEGF. CONCLUSIONS We underline the importance to consider the amoeboid mechanism of endothelial and cancer cell invasion, probably responsible for the failure of synthetic metalloproteinase inhibitors as cancer therapy and tumor resistance to VEGF-targeted therapies, to set-up new drugs to be used in cancer therapy.
Collapse
Affiliation(s)
- Anastasia Chillà
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale G.B. Morgagni, 50, 50134, Florence, Italy.
| | - Cecilia Anceschi
- grid.8404.80000 0004 1757 2304Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Viale G.B. Morgagni, 50, 50134 Florence, Italy
| | - Elena Frediani
- grid.8404.80000 0004 1757 2304Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Viale G.B. Morgagni, 50, 50134 Florence, Italy
| | - Francesca Scavone
- grid.8404.80000 0004 1757 2304Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Viale G.B. Morgagni, 50, 50134 Florence, Italy
| | - Tommaso Del Rosso
- grid.4839.60000 0001 2323 852XDepartment of Physics, Pontifical Catholic University of Rio de Janeiro, Rio de Janeiro, RJ 22451-900 Brazil
| | - Giuseppe Pelagio
- IRCCS Istituto Tumori Giovanni Paolo II Bari, Viale Orazio Flacco 65, 70124 Bari, Italy
| | - Antonio Tufaro
- IRCCS Istituto Tumori Giovanni Paolo II Bari, Viale Orazio Flacco 65, 70124 Bari, Italy
| | - Giuseppe De Palma
- IRCCS Istituto Tumori Giovanni Paolo II Bari, Viale Orazio Flacco 65, 70124 Bari, Italy
| | - Mario Del Rosso
- grid.8404.80000 0004 1757 2304Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Viale G.B. Morgagni, 50, 50134 Florence, Italy
| | - Gabriella Fibbi
- grid.8404.80000 0004 1757 2304Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Viale G.B. Morgagni, 50, 50134 Florence, Italy
| | - Paola Chiarugi
- grid.8404.80000 0004 1757 2304Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Viale G.B. Morgagni, 50, 50134 Florence, Italy
| | - Anna Laurenzana
- grid.8404.80000 0004 1757 2304Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Viale G.B. Morgagni, 50, 50134 Florence, Italy
| | - Francesca Margheri
- grid.8404.80000 0004 1757 2304Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Viale G.B. Morgagni, 50, 50134 Florence, Italy
| |
Collapse
|
6
|
Lv T, Zhao Y, Jiang X, Yuan H, Wang H, Cui X, Xu J, Zhao J, Wang J. uPAR: An Essential Factor for Tumor Development. J Cancer 2021; 12:7026-7040. [PMID: 34729105 PMCID: PMC8558663 DOI: 10.7150/jca.62281] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 10/02/2021] [Indexed: 02/06/2023] Open
Abstract
Tumorigenesis is closely related to the loss of control of many genes. Urokinase-type plasminogen activator receptor (uPAR), a glycolipid-anchored protein on the cell surface, is controlled by many factors in tumorigenesis and is expressed in many tumor tissues. In this review, we summarize the regulatory effects of the uPAR signaling pathway on processes and factors related to tumor progression, such as tumor cell proliferation, adhesion, metastasis, glycolysis, tumor microenvironment and angiogenesis. Overall, the evidence accumulated to date suggests that uPAR induction by tumor progression may be one of the most important factors affecting therapeutic efficacy. An improved understanding of the interactions between uPAR and its coreceptors in cancer will provide critical biomolecular information that may help to better predict the disease course and response to therapy.
Collapse
Affiliation(s)
- Tao Lv
- College of Biological Resource and Food Engineering, Qujing Normal University, Qujing, Yunnan, China 655011.,Key Laboratory of Yunnan Province Universities of the Diversity and Ecological Adaptive Evolution for Animals and Plants on YunGui Plateau, Qujing Normal University, Qujing, China 655011
| | - Ying Zhao
- College of Biological Resource and Food Engineering, Qujing Normal University, Qujing, Yunnan, China 655011
| | - Xinni Jiang
- School of Biological Sciences and Technology, Chengdu Medical College, Chengdu, Sichuan, China 610500
| | - Hemei Yuan
- College of Biological Resource and Food Engineering, Qujing Normal University, Qujing, Yunnan, China 655011
| | - Haibo Wang
- College of Biological Resource and Food Engineering, Qujing Normal University, Qujing, Yunnan, China 655011.,Key Laboratory of Yunnan Province Universities of the Diversity and Ecological Adaptive Evolution for Animals and Plants on YunGui Plateau, Qujing Normal University, Qujing, China 655011
| | - Xuelin Cui
- College of Biological Resource and Food Engineering, Qujing Normal University, Qujing, Yunnan, China 655011
| | - Jiashun Xu
- College of Biological Resource and Food Engineering, Qujing Normal University, Qujing, Yunnan, China 655011
| | - Jingye Zhao
- College of Biological Resource and Food Engineering, Qujing Normal University, Qujing, Yunnan, China 655011
| | - Jianlin Wang
- College of Chemistry and Environmental Science, Qujing Normal University, Qujing, Yunnan, China 655011
| |
Collapse
|
7
|
Lai X, Guo Y, Chen M, Wei Y, Yi W, Shi Y, Xiong L. Caveolin1: its roles in normal and cancer stem cells. J Cancer Res Clin Oncol 2021; 147:3459-3475. [PMID: 34498146 DOI: 10.1007/s00432-021-03793-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 09/03/2021] [Indexed: 12/09/2022]
Abstract
PURPOSE Stem cells are characterized by the capability of self-renewal and multi-differentiation. Normal stem cells, which are important for tissue repair and tissue regeneration, can be divided into embryonic stem cells (ESCs) and somatic stem cells (SSCs) depending on their origin. As a subpopulation of cells within cancer, cancer stem cells (CSCs) are at the root of therapeutic resistance. Tumor-initiating cells (TICs) are necessary for tumor initiation. Caveolin1 (Cav1), a membrane protein located at the caveolae, participates in cell lipid transport, cell migration, cell proliferation, and cell signal transduction. The purpose of this review was to explore the relationship between Cav1 and stem cells. RESULTS In ESCs, Cav1 is beneficial for self-renewal, proliferation, and migration. In SSCs, Cav1 exhibits positive or/and negative effects on stem cell self-renewal, differentiation, proliferation, migration, and angiogenic capacity. Cav1 deficiency impairs normal stem cell-based tissue repair. In CSCs, Cav1 inhibits or/and promotes CSC self-renewal, differentiation, invasion, migration, tumorigenicity ability, and CSC formation. And suppressing Cav1 promotes chemo-sensitivity in CSCs and TICs. CONCLUSION Cav1 shows dual roles in stem cell biology. Targeting the Cav1-stem cell axis would be a new way for tissue repair and cancer drug resistance.
Collapse
Affiliation(s)
- Xingning Lai
- Department of Pathophysiology, Medical College, Nanchang University, 461 Bayi Road, Nanchang, China.,Second Clinical Medical College, Nanchang University, Nanchang, 330006, China
| | - Yiling Guo
- Department of Pathophysiology, Medical College, Nanchang University, 461 Bayi Road, Nanchang, China.,Second Clinical Medical College, Nanchang University, Nanchang, 330006, China
| | - Miaomiao Chen
- Department of Pathophysiology, Medical College, Nanchang University, 461 Bayi Road, Nanchang, China.,First Clinical Medical College, Nanchang University, Nanchang, 330006, China
| | - Yuxuan Wei
- Department of Pathophysiology, Medical College, Nanchang University, 461 Bayi Road, Nanchang, China.,Queen Mary School, Jiangxi Medical College of Nanchang University, Nanchang, 330006, China
| | - Wanting Yi
- Department of Pathophysiology, Medical College, Nanchang University, 461 Bayi Road, Nanchang, China.,First Clinical Medical College, Nanchang University, Nanchang, 330006, China
| | - Yubo Shi
- Department of Pathophysiology, Medical College, Nanchang University, 461 Bayi Road, Nanchang, China.,Queen Mary School, Jiangxi Medical College of Nanchang University, Nanchang, 330006, China
| | - Lixia Xiong
- Department of Pathophysiology, Medical College, Nanchang University, 461 Bayi Road, Nanchang, China. .,Jiangxi Province Key Laboratory of Tumor Pathogenesis and Molecular Pathology, Nanchang, 330006, China.
| |
Collapse
|
8
|
The Urokinase Receptor: A Multifunctional Receptor in Cancer Cell Biology. Therapeutic Implications. Int J Mol Sci 2021; 22:ijms22084111. [PMID: 33923400 PMCID: PMC8073738 DOI: 10.3390/ijms22084111] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 04/12/2021] [Accepted: 04/13/2021] [Indexed: 12/19/2022] Open
Abstract
Proteolysis is a key event in several biological processes; proteolysis must be tightly controlled because its improper activation leads to dramatic consequences. Deregulation of proteolytic activity characterizes many pathological conditions, including cancer. The plasminogen activation (PA) system plays a key role in cancer; it includes the serine-protease urokinase-type plasminogen activator (uPA). uPA binds to a specific cellular receptor (uPAR), which concentrates proteolytic activity at the cell surface, thus supporting cell migration. However, a large body of evidence clearly showed uPAR involvement in the biology of cancer cell independently of the proteolytic activity of its ligand. In this review we will first describe this multifunctional molecule and then we will discuss how uPAR can sustain most of cancer hallmarks, which represent the biological capabilities acquired during the multistep cancer development. Finally, we will illustrate the main data available in the literature on uPAR as a cancer biomarker and a molecular target in anti-cancer therapy.
Collapse
|
9
|
Abdelgawad ME, Desterke C, Uzan G, Naserian S. Single-cell transcriptomic profiling and characterization of endothelial progenitor cells: new approach for finding novel markers. Stem Cell Res Ther 2021; 12:145. [PMID: 33627177 PMCID: PMC7905656 DOI: 10.1186/s13287-021-02185-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 01/24/2021] [Indexed: 12/14/2022] Open
Abstract
Background Endothelial progenitor cells (EPCs) are promising candidates for the cellular therapy of peripheral arterial and cardiovascular diseases. However, hitherto there is no specific marker(s) defining precisely EPCs. Herein, we are proposing a new in silico approach for finding novel EPC markers. Methods We assembled five groups of chosen EPC-related genes/factors using PubMed literature and Gene Ontology databases. This shortened database of EPC factors was fed into publically published transcriptome matrix to compare their expression between endothelial colony-forming cells (ECFCs), HUVECs, and two adult endothelial cell types (ECs) from the skin and adipose tissue. Further, the database was used for functional enrichment on Mouse Phenotype database and protein-protein interaction network analyses. Moreover, we built a digital matrix of healthy donors’ PBMCs (33 thousand single-cell transcriptomes) and analyzed the expression of these EPC factors. Results Transcriptome analyses showed that BMP2, 4, and ephrinB2 were exclusively highly expressed in EPCs; the expression of neuropilin-1 and VEGF-C were significantly higher in EPCs and HUVECs compared with other ECs; Notch 1 was highly expressed in EPCs and skin-ECs; MIR21 was highly expressed in skin-ECs; PECAM-1 was significantly higher in EPCs and adipose ECs. Moreover, functional enrichment of EPC-related genes on Mouse Phenotype and STRING protein database has revealed significant relations between chosen EPC factors and endothelial and vascular functions, development, and morphogenesis, where ephrinB2, BMP2, and BMP4 were highly expressed in EPCs and were connected to abnormal vascular functions. Single-cell RNA-sequencing analyses have revealed that among the EPC-regulated markers in transcriptome analyses, (i) ICAM1 and Endoglin were weekly expressed in the monocyte compartment of the peripheral blood; (ii) CD163 and CD36 were highly expressed in the CD14+ monocyte compartment whereas CSF1R was highly expressed in the CD16+ monocyte compartment, (iii) L-selectin and IL6R were globally expressed in the lymphoid/myeloid compartments, and (iv) interestingly, PLAUR/UPAR and NOTCH2 were highly expressed in both CD14+ and CD16+ monocytic compartments. Conclusions The current study has identified novel EPC markers that could be used for better characterization of EPC subpopulation in adult peripheral blood and subsequent usage of EPCs for various cell therapy and regenerative medicine applications.
Collapse
Affiliation(s)
- Mohamed Essameldin Abdelgawad
- Biochemistry & Molecular Biotechnology Division, Chemistry Department, Faculty of Science; Innovative Cellular Microenvironment Optimization Platform (ICMOP), Helwan University, Cairo, Egypt. .,Inserm UMR-S-MD 1197, Hôpital Paul Brousse - Bâtiment Lavoisier, 12-14 avenue Paul Vaillant Couturier, 94800, Villejuif, France. .,Paris-Saclay University, Villejuif, France.
| | - Christophe Desterke
- Paris-Saclay University, Villejuif, France.,Inserm UMR-S-MD A9, Hôpital Paul Brousse, Villejuif, France
| | - Georges Uzan
- Inserm UMR-S-MD 1197, Hôpital Paul Brousse - Bâtiment Lavoisier, 12-14 avenue Paul Vaillant Couturier, 94800, Villejuif, France.,Paris-Saclay University, Villejuif, France
| | - Sina Naserian
- Inserm UMR-S-MD 1197, Hôpital Paul Brousse - Bâtiment Lavoisier, 12-14 avenue Paul Vaillant Couturier, 94800, Villejuif, France. .,Paris-Saclay University, Villejuif, France. .,CellMedEx, Saint Maur des Fossés, France.
| |
Collapse
|
10
|
Armanetti P, Chillà A, Margheri F, Biagioni A, Menichetti L, Margheri G, Ratto F, Centi S, Bianchini F, Severi M, Traversi R, Bani D, Lulli M, Del Rosso T, Mocali A, Rovida E, Del Rosso M, Fibbi G, Laurenzana A. Enhanced Antitumoral Activity and Photoacoustic Imaging Properties of AuNP-Enriched Endothelial Colony Forming Cells on Melanoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2001175. [PMID: 33643785 PMCID: PMC7887578 DOI: 10.1002/advs.202001175] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 09/18/2020] [Indexed: 05/03/2023]
Abstract
Near infrared (NIR)-resonant gold nanoparticles (AuNPs) hold great promise in cancer diagnostics and treatment. However, translating the theranostic potential of AuNPs into clinical applications still remains a challenge due to the difficulty to improve the efficiency and specificity of tumor delivery in vivo as well as the clearance from liver and spleen to avoid off target toxicity. In this study, endothelial colony forming cells (ECFCs) are exploited as vehicles to deliver AuNPs to tumors. It is first demonstrated that ECFCs display a great capability to intake AuNPs without losing viability, and exert antitumor activity per se. Using a human melanoma xenograft mouse model, it is next demonstrated that AuNP-loaded ECFCs retain their capacity to migrate to tumor sites in vivo 1 day after injection and stay in the tumor mass for more than 1 week. In addition, it is demonstrated that ECFC-loaded AuNPs are efficiently cleared by the liver over time and do not elicit any sign of damage to healthy tissue.
Collapse
Affiliation(s)
- Paolo Armanetti
- Institute of Clinical Physiology (IFC)National Research CouncilPisa56124Italy
| | - Anastasia Chillà
- Department of Experimental and Clinical Biomedical SciencesUniversity of FlorenceFlorence50134Italy
| | - Francesca Margheri
- Department of Experimental and Clinical Biomedical SciencesUniversity of FlorenceFlorence50134Italy
| | - Alessio Biagioni
- Department of Experimental and Clinical Biomedical SciencesUniversity of FlorenceFlorence50134Italy
| | - Luca Menichetti
- Institute of Clinical Physiology (IFC)National Research CouncilPisa56124Italy
| | - Giancarlo Margheri
- Institute for Complex SystemsNational Research CouncilSesto Fiorentino50019Italy
| | - Fulvio Ratto
- Institute of Applied Physics “N. Carrara”National Research CouncilSesto Fiorentino50019Italy
| | - Sonia Centi
- Institute of Applied Physics “N. Carrara”National Research CouncilSesto Fiorentino50019Italy
| | - Francesca Bianchini
- Department of Experimental and Clinical Biomedical SciencesUniversity of FlorenceFlorence50134Italy
| | - Mirko Severi
- Department of Chemistry “Ugo Schiff”University of FlorenceSesto Fiorentino50019Italy
| | - Rita Traversi
- Department of Chemistry “Ugo Schiff”University of FlorenceSesto Fiorentino50019Italy
| | - Daniele Bani
- Department of Clinical and Experimental MedicineUniversity of FlorenceFlorence50134Italy
| | - Matteo Lulli
- Department of Experimental and Clinical Biomedical SciencesUniversity of FlorenceFlorence50134Italy
| | - Tommaso Del Rosso
- Department of PhysicsPontifícia Universidade Católica do Rio de JaneiroRio de Janeiro22451‐900Brazil
| | - Alessandra Mocali
- Department of Experimental and Clinical Biomedical SciencesUniversity of FlorenceFlorence50134Italy
| | - Elisabetta Rovida
- Department of Experimental and Clinical Biomedical SciencesUniversity of FlorenceFlorence50134Italy
| | - Mario Del Rosso
- Department of Experimental and Clinical Biomedical SciencesUniversity of FlorenceFlorence50134Italy
| | - Gabriella Fibbi
- Department of Experimental and Clinical Biomedical SciencesUniversity of FlorenceFlorence50134Italy
| | - Anna Laurenzana
- Department of Experimental and Clinical Biomedical SciencesUniversity of FlorenceFlorence50134Italy
| |
Collapse
|
11
|
Andreucci E, Margheri F, Peppicelli S, Bianchini F, Ruzzolini J, Laurenzana A, Fibbi G, Bruni C, Bellando-Randone S, Guiducci S, Romano E, Manetti M, Matucci-Cerinic M, Calorini L. Glycolysis-derived acidic microenvironment as a driver of endothelial dysfunction in systemic sclerosis. Rheumatology (Oxford) 2021; 60:4508-4519. [PMID: 33471123 DOI: 10.1093/rheumatology/keab022] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 01/06/2021] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVES Systemic sclerosis (SSc) is an autoimmune disease characterized by peripheral vasculopathy and skin and internal organ fibrosis. Accumulating evidence underlines a close association between a metabolic reprogramming of activated fibroblasts and fibrosis. This prompted us to determine the metabolism of SSc dermal fibroblasts and the effect on the vasculopathy characterizing the disease. METHODS Seahorse XF96 Extracellular Flux Analyzer was exploited to evaluate SSc fibroblast metabolism. In vitro invasion and capillary morphogenesis assays were used to determine the angiogenic ability of endothelial cells (EC). Immunofluorescence, flow cytometer and real time PCR techniques provided evidence of the molecular mechanism behind the impaired vascularization that characterizes SSc patients. RESULTS SSc fibroblasts, compared with control, showed a boosted glycolytic metabolism with increased lactic acid release and subsequent extracellular acidification, that in turn was found to impair EC invasion and organization in capillary-like networks without altering cell viability. A molecular link between extracellular acidosis and endothelial dysfunction was identified as acidic EC up-regulated MMP-12 which cleaves and inactivates uPAR, impairing angiogenesis in SSc. Moreover, the acidic environment was found to induce the loss of endothelial markers and the acquisition of mesenchymal-like features in EC, thus promoting the endothelial-to-mesenchymal transition (EndoMT) process that contributes to both capillary rarefaction and tissue fibrosis in SSc. CONCLUSION This study disclosed a liaison among the metabolic reprogramming of SSc dermal fibroblasts, extracellular acidosis and endothelial dysfunction that may contribute to the impairment and loss of peripheral capillary networks in SSc disease.
Collapse
Affiliation(s)
- Elena Andreucci
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Section of Experimental Pathology and Oncology, University of Florence, Florence, Italy
| | - Francesca Margheri
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Section of Experimental Pathology and Oncology, University of Florence, Florence, Italy
| | - Silvia Peppicelli
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Section of Experimental Pathology and Oncology, University of Florence, Florence, Italy
| | - Francesca Bianchini
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Section of Experimental Pathology and Oncology, University of Florence, Florence, Italy
| | - Jessica Ruzzolini
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Section of Experimental Pathology and Oncology, University of Florence, Florence, Italy
| | - Anna Laurenzana
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Section of Experimental Pathology and Oncology, University of Florence, Florence, Italy
| | - Gabriella Fibbi
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Section of Experimental Pathology and Oncology, University of Florence, Florence, Italy
| | - Cosimo Bruni
- Department of Experimental and Clinical Medicine, Division of Rheumatology, University of Florence, Florence, Italy
| | - Silvia Bellando-Randone
- Department of Experimental and Clinical Medicine, Division of Rheumatology, University of Florence, Florence, Italy
| | - Serena Guiducci
- Department of Experimental and Clinical Medicine, Division of Rheumatology, University of Florence, Florence, Italy
| | - Eloisa Romano
- Department of Experimental and Clinical Medicine, Division of Rheumatology, University of Florence, Florence, Italy
| | - Mirko Manetti
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, Florence, Italy
| | - Marco Matucci-Cerinic
- Department of Experimental and Clinical Medicine, Division of Rheumatology, University of Florence, Florence, Italy
| | - Lido Calorini
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Section of Experimental Pathology and Oncology, University of Florence, Florence, Italy.,Center of Excellence for Research, Transfer and High Education DenoTHE University of Florence, Florence, Italy
| |
Collapse
|
12
|
Prokopiou E D, Pissas M, Fibbi G, Margheri F, Kalska-Szostko B, Papanastasiou G, Jansen M, Wang J, Laurenzana A, Efthimiadou K E. Synthesis and characterization of modified magnetic nanoparticles as theranostic agents: in vitro safety assessment in healthy cells. Toxicol In Vitro 2021; 72:105094. [PMID: 33460736 DOI: 10.1016/j.tiv.2021.105094] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 12/24/2020] [Accepted: 01/11/2021] [Indexed: 12/27/2022]
Abstract
Over the past few decades nanotechnology has paved its way into cancer treatment procedures with the use of nanoparticles (NPs) for contrast media and therapeutic agents. Iron based NPs are the most investigated since they can be used for drug delivery, imaging and when magnetically activate employed as local heat sources in cancer hyperthermia. In this work, was performed synthesis, characterization and biological evaluation of different types of iron oxide nanoparticles (mNPs'), as promising material for tumor hyperthermia. The surface of mNPs' has modified with inorganic stabilizing agents to particularly improve characteristics such as their magnetic properties, colloidal stability and biocompatibility. The successful coating of mNPs' was confirmed by morphological and structural characterization by transmission electron microscopy (TEM) and Fourier-Transform Infra-Red spectroscopy (FT-IR), while their hydrodynamic diameter was studied by using Dynamic light scattering (DLS). X-ray Diffraction (XRD) proved that the crystallite phase of mNPs' is the same with the pattern of magnetite. Superparamagnetic behavior and mNPs' response under the application of alternating magnetic field (AMF) were also thoroughly investigated and showed good heating efficiency in magnetic hyperthermia experiments. The contrast ability in magnetic resonance imaging (MRI) is also discussed indicating that mNPs are negative MRI contrast types. Nonetheless the effects of mNPs on cell viability was performed by MTT on human keratinocytes, human embryonic kidney cells, endothelial cells and by hemolytic assay on erythrocytes. In healthy keratinocytes wound healing assay in different time intervals was performed, assessing both the cell migration and wound closure. Endothelial cells have also been studied in functional activity performing capillary morphogenesis. In vitro studies showed that mNPs are safely taken by the healthy cells and do not interfere with the biological processes such as cell migration and motility.
Collapse
Affiliation(s)
- Danai Prokopiou E
- Inorganic Chemistry Laboratory, Chemistry Department, National and Kapodistrian University of Athens, Panepistimiopolis, Zografou 157 71, Greece; Institute of Nanoscience and Nanotechnology, NCSR "Demokritos", 153 41 Aghia Paraskevi Attikis, Greece
| | - Michael Pissas
- Institute of Nanoscience and Nanotechnology, NCSR "Demokritos", 153 41 Aghia Paraskevi Attikis, Greece
| | - Gabriella Fibbi
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale G.B. Morgagni, 50, Florence 50134, Italy
| | - Francesca Margheri
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale G.B. Morgagni, 50, Florence 50134, Italy
| | - Beata Kalska-Szostko
- Institute of Chemistry, Faculty of Biology and Chemistry, University of Bialystok, Ciołkowskiego 1K, Białystok 15-245, Poland
| | - Giorgos Papanastasiou
- Edinburgh Imaging, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Maurits Jansen
- Edinburgh Imaging, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Jansen Wang
- Edinburgh Imaging, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Anna Laurenzana
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale G.B. Morgagni, 50, Florence 50134, Italy.
| | - Eleni Efthimiadou K
- Inorganic Chemistry Laboratory, Chemistry Department, National and Kapodistrian University of Athens, Panepistimiopolis, Zografou 157 71, Greece; Institute of Nanoscience and Nanotechnology, NCSR "Demokritos", 153 41 Aghia Paraskevi Attikis, Greece.
| |
Collapse
|
13
|
uPAR-expressing melanoma exosomes promote angiogenesis by VE-Cadherin, EGFR and uPAR overexpression and rise of ERK1,2 signaling in endothelial cells. Cell Mol Life Sci 2020; 78:3057-3072. [PMID: 33237352 PMCID: PMC8004497 DOI: 10.1007/s00018-020-03707-4] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 11/02/2020] [Accepted: 11/06/2020] [Indexed: 12/19/2022]
Abstract
Exosomes (Exos) have been reported to promote pre-metastatic niche formation, proliferation, angiogenesis and metastasis. We have investigated the role of uPAR in melanoma cell lines-derived Exos and their pro-angiogenic effects on human microvascular endothelial cells (HMVECs) and endothelial colony-forming cells (ECFCs). Melanoma Exos were isolated from conditioned media of A375 and M6 cells by differential centrifugation and filtration. Tunable Resistive Pulse Sensing (TRPS) and Nanoparticle tracking analysis were performed to analyze dimension and concentration of Exos. The CRISPR–Cas 9 technology was exploited to obtain a robust uPAR knockout. uPAR is expressed in melanoma Exos that are internalized by HMVECs and ECFCs, enhancing VE-Cadherin, EGFR and uPAR expression in endothelial cells that undergo a complete angiogenic program, including proliferation, migration and tube formation. uPAR loss reduced the pro-angiogenic effects of melanoma Exos in vitro and in vivo by inhibition of VE-Cadherin, EGFR and uPAR expression and of ERK1,2 signaling in endothelial cells. A similar effect was obtained with a peptide that inhibits uPAR–EGFR interaction and with the EGFR inhibitor Gefitinib, which also inhibited melanoma Exos-dependent EGFR phosphorylation. This study suggests that uPAR is required for the pro-angiogenic activity of melanoma Exos. We propose the identification of uPAR-expressing Exos as a potentially useful biomarker for assessing pro-angiogenic propensity and eventually monitoring the response to treatment in metastatic melanoma patients.
Collapse
|
14
|
New Pieces in the Puzzle of uPAR Role in Cell Migration Mechanisms. Cells 2020; 9:cells9122531. [PMID: 33255171 PMCID: PMC7761155 DOI: 10.3390/cells9122531] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 11/16/2020] [Accepted: 11/20/2020] [Indexed: 12/13/2022] Open
Abstract
The urokinase (uPA) receptor (uPAR) plays a key role in cell migration. We previously showed that uPAR-negative HEK-293 cells efficiently migrate toward serum but, after uPAR ectopic expression, migrate only in a uPAR-dependent manner. In fact, migration of uPAR-transfected HEK-293 (uPAR-293) cells is impaired by anti-uPAR antibodies, without recovery of the uPAR-independent migration mechanisms formerly active. Prostate carcinoma PC3 cells, which express high endogenous uPAR levels, migrated only through a uPAR-dependent mechanism; in fact, the silencing of uPAR expression inhibited their migration. We hypothesize a crucial role of the uPAR glycosyl-phosphatidyl-inositol (GPI) tail, which promotes uPAR partitioning to lipid rafts, in uPAR-controlled cell migration. Here, we show that removal of the uPAR GPI-tail, or lipid rafts disruption by methyl-beta-cyclodextrin impairs migration of PC3 cells, incapable of uPAR-independent migration, whereas it restores uPAR-independent migration in uPAR-293 cells. We then show that, in PC3 cells, both uPAR signaling partners, β1 integrins and receptors for formylated peptides (FPRs), partly associate with lipid rafts. Inhibition of their interaction with uPAR impairs this association and impairs cell migration. Interestingly, blocking uPAR association with FPRs also impairs β1 integrin partitioning to lipid rafts, whereas blocking its association with β1 integrins has no effect on FPRs partitioning. On these bases, we propose that uPAR controls cell migration by connecting β1 integrins and FPRs and, through its GPI tail, by driving them into lipid rafts, thus promoting pro-migratory signals. uPAR-mediated partitioning of integrins to lipid rafts is strictly dependent on uPAR association with FPRs.
Collapse
|
15
|
Chillà A, Margheri F, Biagioni A, Del Rosso T, Fibbi G, Del Rosso M, Laurenzana A. Cell-Mediated Release of Nanoparticles as a Preferential Option for Future Treatment of Melanoma. Cancers (Basel) 2020; 12:cancers12071771. [PMID: 32630815 PMCID: PMC7408438 DOI: 10.3390/cancers12071771] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 06/22/2020] [Accepted: 06/30/2020] [Indexed: 01/15/2023] Open
Abstract
Targeted and immune therapies have unquestionably improved the prognosis of melanoma patients. However the treatment of this neoplasm still requires approaches with a higher therapeutic index, in order to reduce shortcomings related to toxic effects and aspecific targeting. This means developing therapeutic tools derived with high affinity molecules for tumor components differentially expressed in melanoma cells with respect to their normal counterpart. Nanomedicine has sought to address this problem owing to the high modulability of nanoparticles. This approach exploits not only the enhanced permeability and retention effect typical of the tumor microenvironment (passive targeting), but also the use of specific "molecular antennas" that recognize some tumor-overexpressed molecules (active targeting). This line of research has given rise to the so-called "smart nanoparticles," some of which have already passed the preclinical phase and are under clinical trials in melanoma patients. To further improve nanoparticles partition within tumors, for some years now a line of thought is exploiting the molecular systems that regulate the innate tumor-homing activity of platelets, granulocytes, monocytes/macrophages, stem cells, endothelial-colony-forming cells, and red blood cells loaded with nanoparticles. This new vision springs from the results obtained with some of these cells in regenerative medicine, an approach called "cell therapy." This review takes into consideration the advantages of cell therapy as the only one capable of overcoming the limits of targeting imposed by the increased interstitial pressure of tumors.
Collapse
Affiliation(s)
- Anastasia Chillà
- Department of Experimental and Clinical Biomedical Sciences School of Health Sciences, University of Florence-Viale G.B. Morgagni, 50–50134 Florence, Italy; (A.C.); (F.M.); (A.B.); (G.F.)
| | - Francesca Margheri
- Department of Experimental and Clinical Biomedical Sciences School of Health Sciences, University of Florence-Viale G.B. Morgagni, 50–50134 Florence, Italy; (A.C.); (F.M.); (A.B.); (G.F.)
| | - Alessio Biagioni
- Department of Experimental and Clinical Biomedical Sciences School of Health Sciences, University of Florence-Viale G.B. Morgagni, 50–50134 Florence, Italy; (A.C.); (F.M.); (A.B.); (G.F.)
| | - Tommaso Del Rosso
- Department of Physics, Pontifical Catholic University of Rio de Janeiro, 22451-900 Rio de Janeiro-RJ, Brazil;
| | - Gabriella Fibbi
- Department of Experimental and Clinical Biomedical Sciences School of Health Sciences, University of Florence-Viale G.B. Morgagni, 50–50134 Florence, Italy; (A.C.); (F.M.); (A.B.); (G.F.)
| | - Mario Del Rosso
- Department of Experimental and Clinical Biomedical Sciences School of Health Sciences, University of Florence-Viale G.B. Morgagni, 50–50134 Florence, Italy; (A.C.); (F.M.); (A.B.); (G.F.)
- Correspondence: (M.D.R.); (A.L.)
| | - Anna Laurenzana
- Department of Experimental and Clinical Biomedical Sciences School of Health Sciences, University of Florence-Viale G.B. Morgagni, 50–50134 Florence, Italy; (A.C.); (F.M.); (A.B.); (G.F.)
- Correspondence: (M.D.R.); (A.L.)
| |
Collapse
|
16
|
Menicacci B, Margheri F, Laurenzana A, Chillà A, Del Rosso M, Giovannelli L, Fibbi G, Mocali A. Chronic Resveratrol Treatment Reduces the Pro-angiogenic Effect of Human Fibroblast "Senescent-Associated Secretory Phenotype" on Endothelial Colony-Forming Cells: The Role of IL8. J Gerontol A Biol Sci Med Sci 2020; 74:625-633. [PMID: 30084946 DOI: 10.1093/gerona/gly175] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Indexed: 12/12/2022] Open
Abstract
Senescent cells are characterized by an increased secretion of inflammatory and growth factors, known as the "senescence-associated secretory phenotype" (SASP), producing a pro-tumoral and pro-angiogenic microenvironment. This work proposes chronic resveratrol treatment (5 µM for 5 weeks, termed R5) of senescent MRC5 fibroblasts as a mean to mimic and target the angiogenic trait of stromal fibroblast SASP. Senescent fibroblast conditioned medium (CM sen) was effective in enhancing the angiogenic properties of endothelial colony-forming cells (ECFCs), that is, invasive activity and capillary morphogenesis capability in vitro, that were significantly reduced when conditioned media were collected after resveratrol pretreatment (CM senR5). The attenuation of ECFC angiogenic phenotype induced by CM senR5 was accompanied by reduced protein levels of epidermal growth factor and urokinase plasminogen activator receptors (EGFR, uPAR), and by a related decreased activation of receptor-tyrosine-kinase signaling pathways. IL8 levels were found reduced in CM senR5 compared to CM sen, with the associated reduction of IL8-CXCR2 binding in ECFCs. IL8-subtraction mitigated the pro-angiogenic features of CM sen and the associated intracellular signaling in ECFCs, indicating a prominent role of IL8 in the pro-angiogenic effects of CM sen. IL8 modulation is an important mechanism underlying the antiangiogenic activity of resveratrol on MRC5 SASP.
Collapse
Affiliation(s)
- Beatrice Menicacci
- Department of Experimental and Clinical Biomedical Science "Mario Serio", Section of Experimental Pathology and Oncology, University of Florence.,Department of Medical Biotechnologies, University of Siena
| | - Francesca Margheri
- Department of Experimental and Clinical Biomedical Science "Mario Serio", Section of Experimental Pathology and Oncology, University of Florence
| | - Anna Laurenzana
- Department of Experimental and Clinical Biomedical Science "Mario Serio", Section of Experimental Pathology and Oncology, University of Florence
| | - Anastasia Chillà
- Department of Experimental and Clinical Biomedical Science "Mario Serio", Section of Experimental Pathology and Oncology, University of Florence
| | - Mario Del Rosso
- Department of Experimental and Clinical Biomedical Science "Mario Serio", Section of Experimental Pathology and Oncology, University of Florence
| | - Lisa Giovannelli
- Department NeuroFarBa, Section of Pharmacology and Toxicology, University of Florence, Italy
| | - Gabriella Fibbi
- Department of Experimental and Clinical Biomedical Science "Mario Serio", Section of Experimental Pathology and Oncology, University of Florence
| | - Alessandra Mocali
- Department of Experimental and Clinical Biomedical Science "Mario Serio", Section of Experimental Pathology and Oncology, University of Florence
| |
Collapse
|
17
|
Wang CY, Zhang CP, Li BJ, Jiang SS, He WH, Long SY, Tian Y. MMP-12 as a potential biomarker to forecast ischemic stroke in obese patients. Med Hypotheses 2019; 136:109524. [PMID: 31862687 DOI: 10.1016/j.mehy.2019.109524] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 11/29/2019] [Accepted: 12/08/2019] [Indexed: 11/18/2022]
Abstract
Human health is threatened by obesity which causes the increasing incidence of various diseases, especially stroke. Ischemic stroke (IS) is mostly caused by the rupture of arterial plaque, whose instability is positively associated with matrix metalloproteinases (MMPs) that degrades extracellular matrix components. Studies have shown that matrix metalloproteinase-12 (MMP-12) may be involved in the pathogenesis of IS. Because of the higher incidence of stroke in obese patients than that in normal weight people, it is urgent for obesity to forecast stroke early. Considering high levels MMP-12 in obesity, we put forward that MMP-12 may be a potential biomarker for IS in obese patients.
Collapse
Affiliation(s)
- Chu-Yao Wang
- Department of Biochemistry & Molecular Biology, Medical College, University of South China, Hengyang 421001, Hunan, China
| | - Cai-Ping Zhang
- Department of Biochemistry & Molecular Biology, Medical College, University of South China, Hengyang 421001, Hunan, China
| | - Bo-Jie Li
- Department of Biochemistry & Molecular Biology, Medical College, University of South China, Hengyang 421001, Hunan, China
| | - Su-Su Jiang
- Department of Biochemistry & Molecular Biology, Medical College, University of South China, Hengyang 421001, Hunan, China
| | - Wei-He He
- Department of Pharmacology, College of Pharmacy, Hunan University of Chinese Medicine, 300# Xueshi Rd, Hanpu Science & Education District, Changsha 410208, Hunan, China
| | - Shi-Yin Long
- Department of Biochemistry & Molecular Biology, Medical College, University of South China, Hengyang 421001, Hunan, China.
| | - Ying Tian
- Affiliated Nanhua Hospital, University of South China, 336# S Dongfeng Rd., Hengyang 421001, Hunan, China.
| |
Collapse
|
18
|
Zheng C, Terreni M, Sollogoub M, Zhang Y. Ganglioside GM3 and Its Role in Cancer. Curr Med Chem 2019; 26:2933-2947. [PMID: 29376491 DOI: 10.2174/0929867325666180129100619] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 01/03/2018] [Accepted: 01/25/2018] [Indexed: 11/22/2022]
Abstract
Ganglioside GM3 is strongly related with human tumors, such as lung, brain cancers and melanomas, and more and more evidences have revealed that GM3 possesses powerful effects on cancer development and progression. GM3 is over expressed on several types of cancers, and can be as a tumor-associated carbohydrate antigen, used for immunotherapy of cancers. GM3 can also inhibit tumor cells growth by anti-angiogenesis or motility and so on. Especially, GM3 has effects on the EGFR tyrosine kinase signaling, uPAR-related signaling and glycolipid-enriched microdomains, which are essential for cancer signaling conduction. It is obvious that GM3 will be a promising target for cancer treatment.
Collapse
Affiliation(s)
- Changping Zheng
- Sorbonne Universite, CNRS, Institut Parisien de Chimie Moleculaire (UMR 8232), 4 Place Jussieu, 75005 Paris, France
| | - Marco Terreni
- Drug Sciences Department, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy
| | - Matthieu Sollogoub
- Sorbonne Universite, CNRS, Institut Parisien de Chimie Moleculaire (UMR 8232), 4 Place Jussieu, 75005 Paris, France
| | - Yongmin Zhang
- Sorbonne Universite, CNRS, Institut Parisien de Chimie Moleculaire (UMR 8232), 4 Place Jussieu, 75005 Paris, France.,Institute for Interdisciplinary Research, Jianghan University, Wuhan Economic and Technological Development Zone, 430056 Wuhan, China
| |
Collapse
|
19
|
Cimmino I, Margheri F, Prisco F, Perruolo G, D'Esposito V, Laurenzana A, Fibbi G, Paciello O, Doti N, Ruvo M, Miele C, Beguinot F, Formisano P, Oriente F. Prep1 regulates angiogenesis through a PGC-1α-mediated mechanism. FASEB J 2019; 33:13893-13904. [PMID: 31618597 DOI: 10.1096/fj.201901230rr] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Angiogenesis depends on a delicate balance between the different transcription factors, and their control should be considered necessary for preventing or treating diseases. Pre-B-cell leukemia transcription factor regulating protein 1 (Prep1) is a homeodomain transcription factor that plays a primary role in organogenesis and metabolism. Observations performed in a Prep1 hypomorphic mouse model, expressing 3-5% of the protein, show an increase of embryonic lethality due, in part, to defects in angiogenesis. In this study, we provide evidence that overexpression of Prep1 in mouse aortic endothelial cells (MAECs) stimulates migration, proliferation, and tube formation. These effects are paralleled by an increase of several proangiogenic factors and by a decrease of the antiangiogenic gene neurogenic locus notch homolog protein 1 (Notch1). Prep1-mediated angiogenesis involves the activation of the p160 Myb-binding protein (p160)/peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α) pathway. Indeed, Prep1 overexpression increases its binding with p160 and induces a 4-fold increase of p160 and 70% reduction of PGC-1α compared with control cells. Incubation of MAECs with a synthetic Prep1(54-72) peptide, mimicking the Prep1 region involved in the interaction with p160, reverts the proangiogenic effects mediated by Prep1. In addition, Prep1 levels increase by 3.2-fold during the fibroblast growth factor β (bFGF)-mediated endothelial colony-forming cells' activation, whereas Prep1(54-72) peptide reduces the capability of these cells to generate tubular-like structures in response to bFGF, suggesting a possible role of Prep1 both in angiogenesis from preexisting vessels and in postnatal vasculogenesis. Finally, Prep1 hypomorphic heterozygous mice, expressing low levels of Prep1, show attenuated placental angiogenesis and vessel formation within Matrigel plugs. All of these observations indicate that Prep1, complexing with p160, decreases PGC-1α and stimulates angiogenesis.-Cimmino, I., Margheri, F., Prisco, F., Perruolo, G., D'Esposito, V., Laurenzana, A., Fibbi, G., Paciello, O., Doti, N., Ruvo, M., Miele, C., Beguinot, F., Formisano, P., Oriente, F. Prep1 regulates angiogenesis through a PGC-1α-mediated mechanism.
Collapse
Affiliation(s)
- Ilaria Cimmino
- Department of Translational Medicine, Research Unit (URT) Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Council of Research (CNR), University of Naples Federico II, Naples, Italy
| | - Francesca Margheri
- Department of Experimental and Clinical Biomedical Sciences Mario Serio, University of Florence, Florence, Italy
| | - Francesco Prisco
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, Naples, Italy
| | - Giuseppe Perruolo
- Department of Translational Medicine, Research Unit (URT) Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Council of Research (CNR), University of Naples Federico II, Naples, Italy
| | - Vittoria D'Esposito
- Department of Translational Medicine, Research Unit (URT) Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Council of Research (CNR), University of Naples Federico II, Naples, Italy
| | - Anna Laurenzana
- Department of Experimental and Clinical Biomedical Sciences Mario Serio, University of Florence, Florence, Italy
| | - Gabriella Fibbi
- Department of Experimental and Clinical Biomedical Sciences Mario Serio, University of Florence, Florence, Italy
| | - Orlando Paciello
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, Naples, Italy
| | - Nunzianna Doti
- Institute of Biostructure and Bioimaging, National Research Council-Interuniversity Research Centre on Bioactive Peptides, Naples, Italy
| | - Menotti Ruvo
- Institute of Biostructure and Bioimaging, National Research Council-Interuniversity Research Centre on Bioactive Peptides, Naples, Italy
| | - Claudia Miele
- Department of Translational Medicine, Research Unit (URT) Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Council of Research (CNR), University of Naples Federico II, Naples, Italy
| | - Francesco Beguinot
- Department of Translational Medicine, Research Unit (URT) Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Council of Research (CNR), University of Naples Federico II, Naples, Italy
| | - Pietro Formisano
- Department of Translational Medicine, Research Unit (URT) Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Council of Research (CNR), University of Naples Federico II, Naples, Italy
| | - Francesco Oriente
- Department of Translational Medicine, Research Unit (URT) Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Council of Research (CNR), University of Naples Federico II, Naples, Italy
| |
Collapse
|
20
|
Fujimura T, Okabe T, Tanita K, Sato Y, Lyu C, Kambayashi Y, Maruyama S, Aiba S. A novel technique to diagnose non‐melanoma skin cancer by thermal conductivity measurements: Correlations with cancer stromal factors. Exp Dermatol 2019; 28:1029-1035. [DOI: 10.1111/exd.13997] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Revised: 06/04/2019] [Accepted: 06/26/2019] [Indexed: 12/13/2022]
Affiliation(s)
- Taku Fujimura
- Graduate School of Medicine Tohoku University Sendai Japan
| | - Takahiro Okabe
- Graduate School of Science and Technology Hirosaki University Hirosaki Japan
| | - Kayo Tanita
- Graduate School of Medicine Tohoku University Sendai Japan
| | - Yota Sato
- Graduate School of Medicine Tohoku University Sendai Japan
| | - Chunbing Lyu
- Graduate School of Medicine Tohoku University Sendai Japan
| | | | | | - Setsuya Aiba
- Graduate School of Medicine Tohoku University Sendai Japan
| |
Collapse
|
21
|
Potential Role of Caveolin-1 in Regulating the Function of Endothelial Progenitor Cells from Experimental MODS Model. Mediators Inflamm 2019; 2019:8297391. [PMID: 31148948 PMCID: PMC6501138 DOI: 10.1155/2019/8297391] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 02/27/2019] [Accepted: 03/14/2019] [Indexed: 01/04/2023] Open
Abstract
Multiple organ dysfunction syndrome (MODS) remains a great challenge in critical care because of its common occurrence, high cost of care, and high mortality. Vascular endothelial injury is the initiation step in the development of MODS, and EPCs are essential for the process of organ repair. It is unclear whether and how caveolin-1 (Cav-1) in EPCs contributes to the pathogenesis of MODS. The present study is aimed at investigating the potential role of Cav-1 in EPCs during MODS. We established a MODS model in pigs, isolated and characterized EPCs from the MODS model, and tracked Cav-1 expression and various in vitro behaviors of EPCs from the MODS model. Then, we knockdown Cav-1 expression with siRNA or induce Cav-1 expression with proinflammatory factors to evaluate potential effects on EPCs. Our results suggest that Cav-1 expression correlated with EPC functions during MODS and Cav-1 regulates the function of endothelial progenitor cells via PI3K/Akt/eNOS signaling during MODS. Thus, Cav-1 in EPCs could be an attractive target for the treatment of MODS.
Collapse
|
22
|
Bianchini F, De Santis A, Portioli E, Russo Krauss I, Battistini L, Curti C, Peppicelli S, Calorini L, D'Errico G, Zanardi F, Sartori A. Integrin-targeted AmpRGD sunitinib liposomes as integrated antiangiogenic tools. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2019; 18:135-145. [PMID: 30849548 DOI: 10.1016/j.nano.2019.02.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 02/08/2019] [Accepted: 02/19/2019] [Indexed: 01/01/2023]
Abstract
We report here the preparation, physico-chemical characterization, and biological evaluation of a new liposome formulation as a tool for tumor angiogenesis inhibition. Liposomes are loaded with sunitinib, a tyrosine kinase inhibitor, and decorated with cyclo-aminoprolineRGD units (cAmpRGD), efficient and selective ligands for integrin αVβ3. The RGD units play multiple roles since they target the nanovehicles at the integrin αVβ3-overexpressing cells (e.g. activated endothelial cells), favor their active cell internalization, providing drug accumulation in the cytoplasm, and likely take part in the angiogenesis inhibition by interfering in the αVβ3-VEGFR2 cross-talk. Both in vitro and in vivo studies show a better efficacy of this integrated antiangiogenic tool with respect to the free sunitinib and untargeted sunitinib-loaded liposomes. This system could allow a lower administration of the drug and, by increasing the vector specificity, reduce side-effects in a prolonged antiangiogenic therapy.
Collapse
Affiliation(s)
- Francesca Bianchini
- Dipartimento di Scienze Biomediche, Sperimentali e Cliniche "Mario Serio", Università degli Studi di Firenze, Firenze, Italy
| | - Augusta De Santis
- Dipartimento di Scienze Chimiche, Università degli Studi di Napoli "Federico II", Napoli, Italy
| | - Elisabetta Portioli
- Dipartimento di Scienze degli Alimenti e del Farmaco, Università di Parma, Parma, Italy
| | - Irene Russo Krauss
- Dipartimento di Scienze Chimiche, Università degli Studi di Napoli "Federico II", Napoli, Italy
| | - Lucia Battistini
- Dipartimento di Scienze degli Alimenti e del Farmaco, Università di Parma, Parma, Italy
| | - Claudio Curti
- Dipartimento di Scienze degli Alimenti e del Farmaco, Università di Parma, Parma, Italy
| | - Silvia Peppicelli
- Dipartimento di Scienze Biomediche, Sperimentali e Cliniche "Mario Serio", Università degli Studi di Firenze, Firenze, Italy
| | - Lido Calorini
- Dipartimento di Scienze Biomediche, Sperimentali e Cliniche "Mario Serio", Università degli Studi di Firenze, Firenze, Italy
| | - Gerardino D'Errico
- Dipartimento di Scienze Chimiche, Università degli Studi di Napoli "Federico II", Napoli, Italy
| | - Franca Zanardi
- Dipartimento di Scienze degli Alimenti e del Farmaco, Università di Parma, Parma, Italy.
| | - Andrea Sartori
- Dipartimento di Scienze degli Alimenti e del Farmaco, Università di Parma, Parma, Italy.
| |
Collapse
|
23
|
Oleuropein aglycone attenuates the pro-angiogenic phenotype of senescent fibroblasts: A functional study in endothelial cells. J Funct Foods 2019. [DOI: 10.1016/j.jff.2018.12.026] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
24
|
Kanno Y. The Role of Fibrinolytic Regulators in Vascular Dysfunction of Systemic Sclerosis. Int J Mol Sci 2019; 20:ijms20030619. [PMID: 30709025 PMCID: PMC6387418 DOI: 10.3390/ijms20030619] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 01/28/2019] [Accepted: 01/29/2019] [Indexed: 02/08/2023] Open
Abstract
Systemic sclerosis (SSc) is a connective tissue disease of autoimmune origin characterized by vascular dysfunction and extensive fibrosis of the skin and visceral organs. Vascular dysfunction is caused by endothelial cell (EC) apoptosis, defective angiogenesis, defective vasculogenesis, endothelial-to-mesenchymal transition (EndoMT), and coagulation abnormalities, and exacerbates the disease. Fibrinolytic regulators, such as plasminogen (Plg), plasmin, α2-antiplasmin (α2AP), tissue-type plasminogen activator (tPA), urokinase-type plasminogen activator (uPA) and its receptor (uPAR), plasminogen activator inhibitor 1 (PAI-1), and angiostatin, are considered to play an important role in the maintenance of endothelial homeostasis, and are associated with the endothelial dysfunction of SSc. This review considers the roles of fibrinolytic factors in vascular dysfunction of SSc.
Collapse
Affiliation(s)
- Yosuke Kanno
- Department of Clinical Pathological Biochemistry, Faculty of Pharmaceutical Science, Doshisha Women's College of Liberal Arts, 97-1 Kodo Kyo-tanabe, Kyoto 610-0395, Japan.
| |
Collapse
|
25
|
Liu J, Chen G, Xu H, Hu K, Sun J, Liu M, Zhang F, Gu N. Pre-vascularization in fibrin Gel/PLGA microsphere scaffolds designed for bone regeneration. NPG ASIA MATERIALS 2018; 10:827-839. [DOI: 10.1038/s41427-018-0076-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2017] [Revised: 06/13/2018] [Accepted: 07/02/2018] [Indexed: 01/14/2025]
|
26
|
Chillà A, Margheri F, Biagioni A, Del Rosso M, Fibbi G, Laurenzana A. Mature and progenitor endothelial cells perform angiogenesis also under protease inhibition: the amoeboid angiogenesis. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:74. [PMID: 29615071 PMCID: PMC5883600 DOI: 10.1186/s13046-018-0742-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 03/19/2018] [Indexed: 01/05/2023]
Abstract
Background Controlling vascular growth is a challenging aim for the inhibition of tumor growth and metastasis. The amoeboid and mesenchymal types of invasiveness are two modes of migration interchangeable in cancer cells: the Rac-dependent mesenchymal migration requires the activity of proteases; the Rho-ROCK-dependent amoeboid motility is protease-independent and has never been described in endothelial cells. Methods A cocktail of physiologic inhibitors (Ph-C) of serine-proteases, metallo-proteases and cysteine-proteases, mimicking the physiological environment that cells encounter during their migration within the angiogenesis sites was used to induce amoeboid style migration of Endothelial colony forming cells (ECFCs) and mature endothelial cells (ECs). To evaluate the mesenchymal-ameboid transition RhoA and Rac1 activation assays were performed along with immunofluorescence analysis of proteins involved in cytoskeleton organization. Cell invasion was studied in Boyden chambers and Matrigel plug assay for the in vivo angiogenesis. Results In the present study we showed in both ECFCs and ECs, a decrease of activated Rac1 and an increase of activated RhoA upon shifting of cells to the amoeboid conditions. In presence of Ph-C inhibitors both cell lines acquired a round morphology and Matrigel invasion was greatly enhanced with respect to that observed in the absence of protease inhibition. We also observed that the urokinase-plasminogen-activator (uPAR) receptor silencing and uPAR-integrin uncoupling with the M25 peptide abolished both mesenchymal and amoeboid angiogenesis of ECFCs and ECs in vitro and in vivo, indicating a role of the uPAR-integrin-actin axis in the regulation of amoeboid angiogenesis. Furthermore, under amoeboid conditions endothelial cells seem to be indifferent to VEGF stimulation, which induces an amoeboid signaling pattern also in mesenchymal conditions. Conclusion Here we first provide a data set disclosing that endothelial cells can move and differentiate into vascular structures in vitro and in vivo also in the absence of proteases activity, performing a new type of neovascularization: the “amoeboid angiogenesis”. uPAR is indispensable for ECs and ECFCs to perform an efficient amoeboid angiogenesis. Therefore, uPAR silencing or the block of its integrin-interaction, together with standard treatment against VEGF, could be a possible solution for angiogenesis inhibition. Electronic supplementary material The online version of this article (10.1186/s13046-018-0742-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Anastasia Chillà
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Section of Experimental Pathology and Oncology, Viale G.B. Morgagni, 50-50134, Florence, Italy.
| | - Francesca Margheri
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Section of Experimental Pathology and Oncology, Viale G.B. Morgagni, 50-50134, Florence, Italy
| | - Alessio Biagioni
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Section of Experimental Pathology and Oncology, Viale G.B. Morgagni, 50-50134, Florence, Italy
| | - Mario Del Rosso
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Section of Experimental Pathology and Oncology, Viale G.B. Morgagni, 50-50134, Florence, Italy.
| | - Gabriella Fibbi
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Section of Experimental Pathology and Oncology, Viale G.B. Morgagni, 50-50134, Florence, Italy
| | - Anna Laurenzana
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Section of Experimental Pathology and Oncology, Viale G.B. Morgagni, 50-50134, Florence, Italy
| |
Collapse
|
27
|
Comito G, Pons Segura C, Taddei ML, Lanciotti M, Serni S, Morandi A, Chiarugi P, Giannoni E. Zoledronic acid impairs stromal reactivity by inhibiting M2-macrophages polarization and prostate cancer-associated fibroblasts. Oncotarget 2018; 8:118-132. [PMID: 27223431 PMCID: PMC5352046 DOI: 10.18632/oncotarget.9497] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 05/05/2016] [Indexed: 01/11/2023] Open
Abstract
Zoledronic acid (ZA) is a biphosphonate used for osteoporosis treatment and also proved to be effective to reduce the pain induced by bone metastases when used as adjuvant therapy in solid cancers. However, it has been recently proposed that ZA could have direct anti-tumour effects, although the molecular mechanism is unknown. We herein unravel a novel anti-tumour activity of ZA in prostate cancer (PCa), by targeting the pro-tumorigenic properties of both stromal and immune cells. Particularly, we demonstrate that ZA impairs PCa-induced M2-macrophages polarization, reducing their pro-invasive effect on tumour cells and their pro-angiogenic features. Crucially, ZA administration reverts cancer associated fibroblasts (CAFs) activation by targeting the mevalonate pathway and RhoA geranyl-geranylation, thereby impairing smooth muscle actin-α fibers organization, a prerequisite of fibroblast activation. Moreover, ZA prevents the M2 macrophages-mediated activation of normal fibroblast, highlighting the broad efficacy of this drug on tumour microenvironment. These results are confirmed in a metastatic xenograft PCa mouse model in which ZA-induced stromal normalization impairs cancer-stromal cells crosstalk, resulting in a significant reduction of primary tumour growth and metastases. Overall these findings reinforce the efficacy of ZA as a potential therapeutic approach to reduce cancer aggressiveness, by abrogating the supportive role of tumour microenvironment.
Collapse
Affiliation(s)
- Giuseppina Comito
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134 Florence, Italy
| | - Coral Pons Segura
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134 Florence, Italy
| | - Maria Letizia Taddei
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134 Florence, Italy
| | - Michele Lanciotti
- Department of Urology, University of Florence, Careggi Hospital, Urologic Clinic San Luca, 50100 Florence, Italy
| | - Sergio Serni
- Department of Urology, University of Florence, Careggi Hospital, Urologic Clinic San Luca, 50100 Florence, Italy
| | - Andrea Morandi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134 Florence, Italy
| | - Paola Chiarugi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134 Florence, Italy.,Tuscany Tumor Institute and "Center for Research, Transfer and High Education DenoTHE", 50134 Florence, Italy
| | - Elisa Giannoni
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134 Florence, Italy
| |
Collapse
|
28
|
Laurenzana A, Margheri F, Chillà A, Biagioni A, Margheri G, Calorini L, Fibbi G, Del Rosso M. Endothelial Progenitor Cells as Shuttle of Anticancer Agents. Hum Gene Ther 2018; 27:784-791. [PMID: 27502560 DOI: 10.1089/hum.2016.066] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Cell therapies are treatments in which stem or progenitor cells are stimulated to differentiate into specialized cells able to home to and repair damaged tissues. After their discovery, endothelial progenitor cells (EPCs) stimulated worldwide interest as possible vehicles to perform autologous cell therapy of tumors. Taking into account the tumor-homing properties of EPCs, two different approaches to control cancer progression have been pursued by combining cell-based therapy with gene therapy or with nanomedicine. The first approach is based on the possibility of engineering EPCs to express different transgenes, and the second is based on the capacity of EPCs to take up nanomaterials. Here we review the most important progress covering the following issues: the characterization of bona fide endothelial progenitor cells, their role in tumor vascularization and metastasis, and preclinical data about their use in cell-based tumor therapy, considering antiangiogenic, suicide, immune-stimulating, and oncolytic virus gene therapy. The mixed approach of EPC cell therapy and nanomedicine is discussed in terms of plasmonic-dependent thermoablation and molecular imaging.
Collapse
Affiliation(s)
- Anna Laurenzana
- 1 Department of Clinical and Experimental Biomedical Sciences, University of Florence , Florence, Italy
| | - Francesca Margheri
- 1 Department of Clinical and Experimental Biomedical Sciences, University of Florence , Florence, Italy
| | - Anastasia Chillà
- 1 Department of Clinical and Experimental Biomedical Sciences, University of Florence , Florence, Italy
| | - Alessio Biagioni
- 1 Department of Clinical and Experimental Biomedical Sciences, University of Florence , Florence, Italy
| | - Giancarlo Margheri
- 2 Institute for Complex Systems , National Research Council, Sesto Fiorentino, Italy
| | - Lido Calorini
- 1 Department of Clinical and Experimental Biomedical Sciences, University of Florence , Florence, Italy.,3 Center of Excellence for the Study at Molecular and Clinical Levels of Chronic, Degenerative, and Neoplastic Diseases to Develop Novel Therapies (DENOTHE) , Florence, Italy
| | - Gabriella Fibbi
- 1 Department of Clinical and Experimental Biomedical Sciences, University of Florence , Florence, Italy
| | - Mario Del Rosso
- 1 Department of Clinical and Experimental Biomedical Sciences, University of Florence , Florence, Italy.,3 Center of Excellence for the Study at Molecular and Clinical Levels of Chronic, Degenerative, and Neoplastic Diseases to Develop Novel Therapies (DENOTHE) , Florence, Italy
| |
Collapse
|
29
|
Margheri G, Zoppi A, Olmi R, Trigari S, Traversi R, Severi M, Bani D, Bianchini F, Torre E, Margheri F, Chillà A, Biagioni A, Calorini L, Laurenzana A, Fibbi G, Del Rosso M. Tumor-tropic endothelial colony forming cells (ECFCs) loaded with near-infrared sensitive Au nanoparticles: A "cellular stove" approach to the photoablation of melanoma. Oncotarget 2018; 7:39846-39860. [PMID: 27223433 PMCID: PMC5129975 DOI: 10.18632/oncotarget.9511] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 05/08/2016] [Indexed: 12/19/2022] Open
Abstract
In the photothermal treatments (PTs) of tumor, the localization of a high number of near-infrared (NIR) absorbing gold nanoparticles in the tumor mass is still a challenging issue. Here, we propose a promising strategy to deliver therapeutic chitosan-coated gold nanoparticles to tumor cells as hidden cargo of Endothelial Colony Forming Cells (ECFCs) endowed with an innate tumor-tropism. Remarkably, ECFC gold enrichement doesn't affect cell viability and preserves the endothelial lineage characteristics such as capillary morphogenesis and cell migration. We demonstrate that heavily Au-doped ECFCs are able to efficiently warm up the tumor environment, and kill the cancer cells via hyperthermic heating both in vitro as well as in vivo. Thus, we show an excellent thermotransductive property of gold enriched ECFCs and their capability to kill melanoma cells at moderate NIR light intensities.
Collapse
Affiliation(s)
- Giancarlo Margheri
- Institute for Complex Systems, National Research Council, Sesto Fiorentino, Italy
| | - Angela Zoppi
- Department of Physics "Enrico Fermi", University of Pisa, Italy.,Present address: Plasmatech, Department of Physics "Enrico Fermi", University of Pisa, Pisa, Italy
| | - Roberto Olmi
- Institute of Applied Physics "Nello Carrara", National Research Council, Sesto Fiorentino, Italy
| | - Silvana Trigari
- Institute for Complex Systems, National Research Council, Sesto Fiorentino, Italy
| | - Rita Traversi
- Department of Chemistry "Ugo Schiff", University of Florence, Sesto Fiorentino, Italy
| | - Mirko Severi
- Department of Chemistry "Ugo Schiff", University of Florence, Sesto Fiorentino, Italy
| | - Daniele Bani
- Department of Clinical and Experimental Medicine, University of Florence, Florence, Italy
| | - Francesca Bianchini
- Department of Experimental and Clinical Biomedical Science, University of Florence, Florence, Italy
| | - Eugenio Torre
- Department of Experimental and Clinical Biomedical Science, University of Florence, Florence, Italy
| | - Francesca Margheri
- Department of Experimental and Clinical Biomedical Science, University of Florence, Florence, Italy
| | - Anastasia Chillà
- Department of Experimental and Clinical Biomedical Science, University of Florence, Florence, Italy
| | - Alessio Biagioni
- Department of Experimental and Clinical Biomedical Science, University of Florence, Florence, Italy
| | - Lido Calorini
- Department of Experimental and Clinical Biomedical Science, University of Florence, Florence, Italy.,Excellence Center for Research, Transfer and High Education 'Study at Molecular and Clinical Level of Chronic, Inflammatory, Degenerative and Neoplastic Disorders for the Development on Novel Therapies', Florence, Italy
| | - Anna Laurenzana
- Department of Experimental and Clinical Biomedical Science, University of Florence, Florence, Italy
| | - Gabriella Fibbi
- Department of Experimental and Clinical Biomedical Science, University of Florence, Florence, Italy
| | - Mario Del Rosso
- Department of Experimental and Clinical Biomedical Science, University of Florence, Florence, Italy.,Excellence Center for Research, Transfer and High Education 'Study at Molecular and Clinical Level of Chronic, Inflammatory, Degenerative and Neoplastic Disorders for the Development on Novel Therapies', Florence, Italy
| |
Collapse
|
30
|
Zhang J, Li C, Zheng Y, Lin Z, Zhang Y, Zhang Z. Inhibition of angiogenesis by arsenic trioxide via TSP-1-TGF-β1-CTGF-VEGF functional module in rheumatoid arthritis. Oncotarget 2017; 8:73529-73546. [PMID: 29088724 PMCID: PMC5650279 DOI: 10.18632/oncotarget.19867] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 07/11/2017] [Indexed: 01/25/2023] Open
Abstract
Angiogenesis is a critical factor for rheumatoid arthritis (RA). Although anti-TNF biologics work effectively on some RA patients, concerns have been raised about the possible increased development of malignancies alongside such treatments. Arsenic trioxide (As2O3) has attracted worldwide attention and has been reported to treat some cancers. However, the effects of As2O3 on angiogenesis in the RA synovium remain unclear. Here, we report a systematic increased expression of TSP-1, TGF-β1, CTGF and VEGF in supernatants of a RA fibroblast-like synoviocytes (RA-FLS) and human dermal microvascular endothelial cells (HDMECs) co-culture compared with those from a normal human fibroblast-like synoviocytes (NH-FLS) and HDMECs co-culture. This increased expression may up-regulate endothelial tube formation and transwell migration, as well as microvessel sprouting in ex vivo aortic ring assay. These networked angiogenic factors mainly form a functional module regulating angiogenesis in the RA synovium. We show that As2O3 inhibits angiogenesis in the collagen-induced arthritis (CIA) synovium and consequently arthritis severity via significant suppression of TSP-1, TGF-β1, CTGF and VEGF expression in the CIA synovium, plus in the RA-FLS and HDMECs co-culture as well as NH-FLS and HDMECs co-culture system along with the presence or absence of TNF-α treatment. Thus As2O3 has a significant anti-angiogenesis effect on the RA-FLS and CIA synovium via its inhibition of the RA angiogenic functional module of TSP-1, TGF-β1, CTGF and VEGF and may have a potential for treating RA beyond cancer therapy.
Collapse
Affiliation(s)
- Juan Zhang
- Department of Rheumatology, The First Affiliated Hospital, Harbin Medical University, Nan Gang, Harbin, China
| | - Chunling Li
- Department of Rheumatology, The First Affiliated Hospital, Harbin Medical University, Nan Gang, Harbin, China
| | - Yining Zheng
- Department of Rheumatology, The First Affiliated Hospital, Harbin Medical University, Nan Gang, Harbin, China
| | - Zhiguo Lin
- Department of Rheumatology, The First Affiliated Hospital, Harbin Medical University, Nan Gang, Harbin, China
| | - Yue Zhang
- Department of Rheumatology, The First Affiliated Hospital, Harbin Medical University, Nan Gang, Harbin, China
| | - Zhiyi Zhang
- Department of Rheumatology, The First Affiliated Hospital, Harbin Medical University, Nan Gang, Harbin, China
| |
Collapse
|
31
|
Laurenzana A, Chillà A, Luciani C, Peppicelli S, Biagioni A, Bianchini F, Tenedini E, Torre E, Mocali A, Calorini L, Margheri F, Fibbi G, Del Rosso M. uPA/uPAR system activation drives a glycolytic phenotype in melanoma cells. Int J Cancer 2017; 141:1190-1200. [DOI: 10.1002/ijc.30817] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 04/21/2017] [Accepted: 05/24/2017] [Indexed: 12/22/2022]
Affiliation(s)
- Anna Laurenzana
- Department of Experimental and Clinical Biomedical Sciences, Section of Experimental Pathology and Oncology; University of Florence; Florence 50134 Italy
| | - Anastasia Chillà
- Department of Experimental and Clinical Biomedical Sciences, Section of Experimental Pathology and Oncology; University of Florence; Florence 50134 Italy
| | - Cristina Luciani
- Department of Experimental and Clinical Biomedical Sciences, Section of Experimental Pathology and Oncology; University of Florence; Florence 50134 Italy
| | - Silvia Peppicelli
- Department of Experimental and Clinical Biomedical Sciences, Section of Experimental Pathology and Oncology; University of Florence; Florence 50134 Italy
| | - Alessio Biagioni
- Department of Experimental and Clinical Biomedical Sciences, Section of Experimental Pathology and Oncology; University of Florence; Florence 50134 Italy
| | - Francesca Bianchini
- Department of Experimental and Clinical Biomedical Sciences, Section of Experimental Pathology and Oncology; University of Florence; Florence 50134 Italy
| | - Elena Tenedini
- Center for Genome Research, Life Sciences Department; University of Modena; Modena 41125 Italy
| | - Eugenio Torre
- Department of Experimental and Clinical Biomedical Sciences, Section of Experimental Pathology and Oncology; University of Florence; Florence 50134 Italy
| | - Alessandra Mocali
- Department of Experimental and Clinical Biomedical Sciences, Section of Experimental Pathology and Oncology; University of Florence; Florence 50134 Italy
| | - Lido Calorini
- Department of Experimental and Clinical Biomedical Sciences, Section of Experimental Pathology and Oncology; University of Florence; Florence 50134 Italy
| | - Francesca Margheri
- Department of Experimental and Clinical Biomedical Sciences, Section of Experimental Pathology and Oncology; University of Florence; Florence 50134 Italy
| | - Gabriella Fibbi
- Department of Experimental and Clinical Biomedical Sciences, Section of Experimental Pathology and Oncology; University of Florence; Florence 50134 Italy
| | - Mario Del Rosso
- Department of Experimental and Clinical Biomedical Sciences, Section of Experimental Pathology and Oncology; University of Florence; Florence 50134 Italy
| |
Collapse
|
32
|
Lv FZ, Wang JL, Wu Y, Chen HF, Shen XY. Knockdown of MMP12 inhibits the growth and invasion of lung adenocarcinoma cells. Int J Immunopathol Pharmacol 2017; 28:77-84. [PMID: 25816409 DOI: 10.1177/0394632015572557] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Matrix metalloproteinase-12 (MMP12) is involved in many pathological processes including cancer. The expression and function of MMP12 in lung adenocarcinoma (LAC) remain unclear. The present study aimed to investigate the correlation of MMP12 expression with LAC patients and clarify its role in growth and invasion of LAC cells. The expression of MMP12 in human LAC was examined by immunohistochemical assay using a tissue microarray procedure. A loss-of-function experiment was used for observing the effects of lentiviral vector-mediated MMP12 shRNA (shMMP12) on cell growth and invasion in LAC cell lines (A549), indicated by MTT and Transwell assays. We found that the expression of MMP12 protein was significantly increased in LAC tissues compared with that in adjacent non-cancerous tissues (ANCT) (57.69% vs. 32.69%, P = 0.019), and was closely correlated with the pathological stage and lymph node metastasis of LAC patients (P = 0.01; P = 0.003). Knockdown of MMP12 inhibited proliferation and invasion of LAC cells followed by the downregulation of proliferating cell nuclear antigen (PCNA) and vascular endothelial growth factor (VEGF). In conclusion, our findings show that high expression of MMP12 is correlated with the pathological stage and tumor metastasis of LAC patients, and knockdown of MMP12 suppresses the development of LAC cells, suggesting that MMP12 may be a promising therapeutic target for the treatment of LAC.
Collapse
Affiliation(s)
- F-Z Lv
- Department of Thoracic Surgery, The Huadong Hospital, Shanghai Fudan University, Shanghai, PR China
| | - J-L Wang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, PR China
| | - Y Wu
- Department of Thoracic Surgery, The Huadong Hospital, Shanghai Fudan University, Shanghai, PR China
| | - H-F Chen
- Department of Respiration medicine, The Huadong Hospital, Shanghai Fudan University, Shanghai, PR China
| | - X-Y Shen
- Department of Thoracic Surgery, The Huadong Hospital, Shanghai Fudan University, Shanghai, PR China
| |
Collapse
|
33
|
Sartori A, Portioli E, Battistini L, Calorini L, Pupi A, Vacondio F, Arosio D, Bianchini F, Zanardi F. Synthesis of Novel c(AmpRGD)-Sunitinib Dual Conjugates as Molecular Tools Targeting the α vβ 3 Integrin/VEGFR2 Couple and Impairing Tumor-Associated Angiogenesis. J Med Chem 2016; 60:248-262. [PMID: 27997164 DOI: 10.1021/acs.jmedchem.6b01266] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
On the basis of a previously discovered anti-αVβ3 integrin peptidomimetic (c(AmpRGD)) and the clinically approved antiangiogenic kinase inhibitor sunitinib, three novel dual conjugates were synthesized (compounds 1-3), featuring the covalent and robust linkage between these two active modules. In all conjugates, the ligand binding competence toward αVβ3 (using both isolated receptors and αVβ3-overexpressing endothelial progenitor EP cells) and the kinase inhibitory activity (toward both isolated kinases and EPCs) remained almost untouched and comparable to the activity of the single active units. Compounds 1-3 showed interesting antiangiogenesis properties in an in vitro tubulogenic assay; furthermore, dimeric-RGD conjugate 3 strongly inhibited in vivo angiogenesis in Matrigel plug assays in FVB mice. These results offer proof-of-concept of how the covalent conjugation of two angiogenesis-related small modules may result in novel and stable molecules, which impair tumor-related angiogenesis with equal or even superior ability as compared to the single modules or their simple combinations.
Collapse
Affiliation(s)
- Andrea Sartori
- Dipartimento di Farmacia, Università degli Studi di Parma , Parco Area delle Scienze 27A, 43124 Parma, Italy
| | - Elisabetta Portioli
- Dipartimento di Farmacia, Università degli Studi di Parma , Parco Area delle Scienze 27A, 43124 Parma, Italy
| | - Lucia Battistini
- Dipartimento di Farmacia, Università degli Studi di Parma , Parco Area delle Scienze 27A, 43124 Parma, Italy
| | - Lido Calorini
- Dipartimento di Scienze Biomediche, Sperimentali e Cliniche "Mario Serio", Università degli Studi di Firenze , Viale G. B. Morgagni 50, 50134 Firenze, Italy
| | - Alberto Pupi
- Dipartimento di Scienze Biomediche, Sperimentali e Cliniche "Mario Serio", Università degli Studi di Firenze , Viale G. B. Morgagni 50, 50134 Firenze, Italy.,Centro Interdipartimentale per lo Sviluppo Preclinico dell'Imaging Molecolare (CISPIM), Università degli Studi di Firenze , Viale G. B. Morgagni 50, 50134 Firenze, Italy
| | - Federica Vacondio
- Dipartimento di Farmacia, Università degli Studi di Parma , Parco Area delle Scienze 27A, 43124 Parma, Italy
| | - Daniela Arosio
- Istituto di Scienze e Tecnologie Molecolari, Consiglio Nazionale delle Ricerche , Via Golgi 19, 20133 Milano, Italy
| | - Francesca Bianchini
- Dipartimento di Scienze Biomediche, Sperimentali e Cliniche "Mario Serio", Università degli Studi di Firenze , Viale G. B. Morgagni 50, 50134 Firenze, Italy.,Centro Interdipartimentale per lo Sviluppo Preclinico dell'Imaging Molecolare (CISPIM), Università degli Studi di Firenze , Viale G. B. Morgagni 50, 50134 Firenze, Italy
| | - Franca Zanardi
- Dipartimento di Farmacia, Università degli Studi di Parma , Parco Area delle Scienze 27A, 43124 Parma, Italy
| |
Collapse
|
34
|
Bianchini F, Peppicelli S, Fabbrizzi P, Biagioni A, Mazzanti B, Menchi G, Calorini L, Pupi A, Trabocchi A. Triazole RGD antagonist reverts TGFβ1-induced endothelial-to-mesenchymal transition in endothelial precursor cells. Mol Cell Biochem 2016; 424:99-110. [PMID: 27761847 PMCID: PMC5219041 DOI: 10.1007/s11010-016-2847-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 10/08/2016] [Indexed: 12/26/2022]
Abstract
Fibrosis is the dramatic consequence of a dysregulated reparative process in which activated fibroblasts (myofibroblasts) and Transforming Growth Factor β1 (TGFβ1) play a central role. When exposed to TGFβ1, fibroblast and epithelial cells differentiate in myofibroblasts; in addition, endothelial cells may undergo endothelial-to-mesenchymal transition (EndoMT) and actively participate to the progression of fibrosis. Recently, the role of αv integrins, which recognize the Arg-Gly-Asp (RGD) tripeptide, in the release and signal transduction activation of TGFβ1 became evident. In this study, we present a class of triazole-derived RGD antagonists that interact with αvβ3 integrin. Above different compounds, the RGD-2 specifically interferes with integrin-dependent TGFβ1 EndoMT in Endothelial Colony-Forming Cells (ECPCs) derived from circulating Endothelial Precursor Cells (ECPCs). The RGD-2 decreases the amount of membrane-associated TGFβ1, and reduces both ALK5/TGFβ1 type I receptor expression and Smad2 phosphorylation in ECPCs. We found that RGD-2 antagonist reverts EndoMT, reducing α-smooth muscle actin (α-SMA) and vimentin expression in differentiated ECPCs. Our results outline the critical role of integrin in fibrosis progression and account for the opportunity of using integrins as target for anti-fibrotic therapeutic treatment.
Collapse
Affiliation(s)
- Francesca Bianchini
- Department of Clinical and Experimental Biomedical Science "Mario Serio", University of Florence, Florence, Italy.
| | - Silvia Peppicelli
- Department of Clinical and Experimental Biomedical Science "Mario Serio", University of Florence, Florence, Italy
| | | | - Alessio Biagioni
- Department of Clinical and Experimental Biomedical Science "Mario Serio", University of Florence, Florence, Italy
| | | | - Gloria Menchi
- Department of Chemistry "Ugo Schiff", University of Florence, Florence, Italy.,Interdepartmental Center for Preclinical Development of Molecular Imaging (CISPIM), University of Florence, Florence, Italy
| | - Lido Calorini
- Department of Clinical and Experimental Biomedical Science "Mario Serio", University of Florence, Florence, Italy
| | - Alberto Pupi
- Department of Clinical and Experimental Biomedical Science "Mario Serio", University of Florence, Florence, Italy.,Interdepartmental Center for Preclinical Development of Molecular Imaging (CISPIM), University of Florence, Florence, Italy
| | - Andrea Trabocchi
- Department of Chemistry "Ugo Schiff", University of Florence, Florence, Italy.,Interdepartmental Center for Preclinical Development of Molecular Imaging (CISPIM), University of Florence, Florence, Italy
| |
Collapse
|
35
|
Endothelial progenitor cells accelerate the resolution of deep vein thrombosis. Vascul Pharmacol 2016; 83:10-6. [DOI: 10.1016/j.vph.2015.07.007] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 06/27/2015] [Accepted: 07/11/2015] [Indexed: 11/23/2022]
|
36
|
Mrkonjic S, Destaing O, Albiges-Rizo C. Mechanotransduction pulls the strings of matrix degradation at invadosome. Matrix Biol 2016; 57-58:190-203. [PMID: 27392543 DOI: 10.1016/j.matbio.2016.06.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 06/16/2016] [Accepted: 06/28/2016] [Indexed: 02/07/2023]
Abstract
Degradation of the extracellular matrix is a critical step of tumor cell invasion. Both protease-dependent and -independent mechanisms have been described as alternate processes in cancer cell motility. Interestingly, some effectors of protease-dependent degradation are focalized at invadosomes and are directly coupled with contractile and adhesive machineries composed of multiple mechanosensitive proteins. This review presents recent findings in protease-dependent mechanisms elucidating the ways the force affects extracellular matrix degradation by targeting protease expression and activity at invadosome. The aim is to highlight mechanosensing and mechanotransduction processes to direct the degradative activity at invadosomes, with the focus on membrane tension, proteases and mechanosensitive ion channels.
Collapse
Affiliation(s)
- Sanela Mrkonjic
- INSERM U1209, Grenoble F-38042, France; Université Grenoble Alpes, Institut Albert Bonniot, F-38042 Grenoble, France; CNRS UMR 5309, F-38042 Grenoble, France
| | - Olivier Destaing
- INSERM U1209, Grenoble F-38042, France; Université Grenoble Alpes, Institut Albert Bonniot, F-38042 Grenoble, France; CNRS UMR 5309, F-38042 Grenoble, France.
| | - Corinne Albiges-Rizo
- INSERM U1209, Grenoble F-38042, France; Université Grenoble Alpes, Institut Albert Bonniot, F-38042 Grenoble, France; CNRS UMR 5309, F-38042 Grenoble, France.
| |
Collapse
|
37
|
Herkenne S, Paques C, Nivelles O, Lion M, Bajou K, Pollenus T, Fontaine M, Carmeliet P, Martial JA, Nguyen NQN, Struman I. The interaction of uPAR with VEGFR2 promotes VEGF-induced angiogenesis. Sci Signal 2015; 8:ra117. [PMID: 26577922 DOI: 10.1126/scisignal.aaa2403] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In endothelial cells, binding of vascular endothelial growth factor (VEGF) to the receptor VEGFR2 activates multiple signaling pathways that trigger processes such as proliferation, survival, and migration that are necessary for angiogenesis. VEGF-bound VEGFR2 becomes internalized, which is a key step in the proangiogenic signal. We showed that the urokinase plasminogen activator receptor (uPAR) interacted with VEGFR2 and described the mechanism by which this interaction mediated VEGF signaling and promoted angiogenesis. Knockdown of uPAR in human umbilical vein endothelial cells (HUVECs) impaired VEGFR2 signaling, and uPAR deficiency in mice prevented VEGF-induced angiogenesis. Upon exposure of HUVECs to VEGF, uPAR recruited the low-density lipoprotein receptor-related protein 1 (LRP-1) to VEGFR2, which induced VEGFR2 internalization. Thus, the uPAR-VEGFR2 interaction is crucial for VEGF signaling in endothelial cells.
Collapse
Affiliation(s)
- Stéphanie Herkenne
- Molecular Angiogenesis Laboratory, GIGA Research, University of Liège, Avenue de l'Hôpital, 1, 4000 Liège, Belgium. Dulbecco-Telethon Institute, Venetian Institute of Molecular Medicine, Via Orus 2, 35129 Padova, Italy. Department of Biology, University of Padova, Via U. Bassi 58B, 35121 Padova, Italy
| | - Cécile Paques
- Molecular Angiogenesis Laboratory, GIGA Research, University of Liège, Avenue de l'Hôpital, 1, 4000 Liège, Belgium
| | - Olivier Nivelles
- Molecular Angiogenesis Laboratory, GIGA Research, University of Liège, Avenue de l'Hôpital, 1, 4000 Liège, Belgium
| | - Michelle Lion
- Molecular Angiogenesis Laboratory, GIGA Research, University of Liège, Avenue de l'Hôpital, 1, 4000 Liège, Belgium
| | - Khalid Bajou
- Molecular Angiogenesis Laboratory, GIGA Research, University of Liège, Avenue de l'Hôpital, 1, 4000 Liège, Belgium. Department of Applied Biology, College of Sciences, University of Sharjah, P.O. Box 27272, Emirates of Sharjah, United Arab Emirates
| | - Thomas Pollenus
- Molecular Angiogenesis Laboratory, GIGA Research, University of Liège, Avenue de l'Hôpital, 1, 4000 Liège, Belgium
| | - Marie Fontaine
- Molecular Angiogenesis Laboratory, GIGA Research, University of Liège, Avenue de l'Hôpital, 1, 4000 Liège, Belgium
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center (VRC), Vlaams Instituut Biotechnologie, 3000 Leuven, Belgium. Laboratory of Angiogenesis and Neurovascular Link, VRC, Department of Oncology, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Joseph A Martial
- Molecular Angiogenesis Laboratory, GIGA Research, University of Liège, Avenue de l'Hôpital, 1, 4000 Liège, Belgium
| | - Ngoc-Quynh-Nhu Nguyen
- Molecular Angiogenesis Laboratory, GIGA Research, University of Liège, Avenue de l'Hôpital, 1, 4000 Liège, Belgium
| | - Ingrid Struman
- Molecular Angiogenesis Laboratory, GIGA Research, University of Liège, Avenue de l'Hôpital, 1, 4000 Liège, Belgium.
| |
Collapse
|
38
|
Joo HJ, Song S, Seo HR, Shin JH, Choi SC, Park JH, Yu CW, Hong SJ, Lim DS. Human endothelial colony forming cells from adult peripheral blood have enhanced sprouting angiogenic potential through up-regulating VEGFR2 signaling. Int J Cardiol 2015; 197:33-43. [DOI: 10.1016/j.ijcard.2015.06.013] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Revised: 05/03/2015] [Accepted: 06/12/2015] [Indexed: 12/27/2022]
|
39
|
Laurenzana A, Cencetti F, Serratì S, Bruno G, Japtok L, Bianchini F, Torre E, Fibbi G, Del Rosso M, Bruni P, Donati C. Endothelial sphingosine kinase/SPNS2 axis is critical for vessel-like formation by human mesoangioblasts. J Mol Med (Berl) 2015; 93:1145-57. [PMID: 25952146 DOI: 10.1007/s00109-015-1292-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Revised: 04/10/2015] [Accepted: 04/15/2015] [Indexed: 10/23/2022]
Abstract
UNLABELLED The interaction between endothelial cells and pericytes is crucial for the stabilization of newly formed vessels in angiogenesis. The comprehension of the mechanisms regulating pericyte recruitment might open therapeutical perspectives on vascular-related pathologies. Sphingosine 1-phosphate (S1P) is a bioactive sphingolipid that derives from sphingomyelin catabolism and regulates biological functions in cell survival, proliferation, and differentiation. In this study, we aimed to identify the role of S1P axis in the intercellular communication between human mesenchymal progenitor mesoangioblasts (MAB) and endothelial cells (human microvascular endothelial cells (H-MVEC)) in the formation of capillary-like structures. We demonstrated that the S1P biosynthetic pathway brought about by sphingosine kinases (SK) SK1 and SK2 as well as spinster homolog 2 (SPNS2) transporter in H-MVEC is crucial for MAB migration measured by Boyden chambers and for the formation and stabilization of capillary-like structures in a 3D Matrigel culture. Moreover, the conditioned medium (CM) harvested from H-MVEC, where SK1, SK2, and SPNS2 were down-regulated, exerted a significantly diminished effect on MAB capillary morphogenesis and migration. Notably, we demonstrated that S1P1 and S1P3 receptors were positively involved in CM-induced capillary-like formation and migration, while S1P2 exerted a negative role on CM-induced migratory action of MAB. Finally, SK inhibition as well as MAB S1P1 and S1P3 down-regulation impaired H-MVEC-MAB cross-talk significantly reducing in vivo angiogenesis evaluated by Matrigel plug assay. These findings individuate novel targets for the employment of MAB in vascular-related pathologic conditions. KEY MESSAGE • Down-regulation of SK1/2 in H-MVEC impaired vessel formation when cultured with MAB. • H-MVEC SPNS2 is critical for morphogenesis and migration induced by H-MVEC CM of MAB. • CM from SK1- and SK2-siRNA H-MVEC impaired morphogenesis and migration of MAB. • S1P1/3 were involved on CM-induced morphogenesis and migration of MAB. • Matrigel plug assay showed the role of S1P axis in MAB-endothelial cell interaction.
Collapse
Affiliation(s)
- Anna Laurenzana
- Dipartimento di Scienze Biomediche Sperimentali e Cliniche "Mario Serio", Università di Firenze, Viale G.B. Morgagni 50, 50134, Florence, Italy
| | - Francesca Cencetti
- Dipartimento di Scienze Biomediche Sperimentali e Cliniche "Mario Serio", Università di Firenze, Viale G.B. Morgagni 50, 50134, Florence, Italy
| | - Simona Serratì
- Dipartimento di Scienze Biomediche Sperimentali e Cliniche "Mario Serio", Università di Firenze, Viale G.B. Morgagni 50, 50134, Florence, Italy.,Department of Experimental Oncology, Hematology Unit, Advanced Cellular Therapy Centre, Bari, Italy
| | - Gennaro Bruno
- Dipartimento di Scienze Biomediche Sperimentali e Cliniche "Mario Serio", Università di Firenze, Viale G.B. Morgagni 50, 50134, Florence, Italy
| | - Lukasz Japtok
- Faculty of Mathematics and Natural Science, Institute of Nutritional Science, Department of Toxicology, University of Potsdam, Arthur-Scheunert Allee 114-116, 14558, Potsdam, Nuthetal, Germany
| | - Francesca Bianchini
- Dipartimento di Scienze Biomediche Sperimentali e Cliniche "Mario Serio", Università di Firenze, Viale G.B. Morgagni 50, 50134, Florence, Italy
| | - Eugenio Torre
- Dipartimento di Scienze Biomediche Sperimentali e Cliniche "Mario Serio", Università di Firenze, Viale G.B. Morgagni 50, 50134, Florence, Italy
| | - Gabriella Fibbi
- Dipartimento di Scienze Biomediche Sperimentali e Cliniche "Mario Serio", Università di Firenze, Viale G.B. Morgagni 50, 50134, Florence, Italy
| | - Mario Del Rosso
- Dipartimento di Scienze Biomediche Sperimentali e Cliniche "Mario Serio", Università di Firenze, Viale G.B. Morgagni 50, 50134, Florence, Italy
| | - Paola Bruni
- Dipartimento di Scienze Biomediche Sperimentali e Cliniche "Mario Serio", Università di Firenze, Viale G.B. Morgagni 50, 50134, Florence, Italy
| | - Chiara Donati
- Dipartimento di Scienze Biomediche Sperimentali e Cliniche "Mario Serio", Università di Firenze, Viale G.B. Morgagni 50, 50134, Florence, Italy.
| |
Collapse
|
40
|
Melanoma cell therapy: Endothelial progenitor cells as shuttle of the MMP12 uPAR-degrading enzyme. Oncotarget 2015; 5:3711-27. [PMID: 25003596 PMCID: PMC4116515 DOI: 10.18632/oncotarget.1987] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The receptor for the urokinase-type plasminogen activator (uPAR) accounts for many features of cancer progression, and is therefore considered a target for anti-tumoral therapy. Only full length uPAR mediates tumor progression. Matrix-metallo-proteinase-12 (MMP12)-dependent uPAR cleavage results into the loss of invasion properties and angiogenesis. MMP12 can be employed in the field of “targeted therapies” as a biological drug to be delivered directly in patient's tumor mass. Endothelial Progenitor Cells (EPCs) are selectively recruited within the tumor and could be used as cellular vehicles for delivering anti-cancer molecules. The aim of our study is to inhibit cancer progression by engeneering ECFCs, a subset of EPC, with a lentivirus encoding the anti-tumor uPAR-degrading enzyme MMP12. Ex vivo manipulated ECFCs lost the capacity to perform capillary morphogenesis and acquired the anti-tumor and anti-angiogenetic activity. In vivo MMP12-engineered ECFCs cleaved uPAR within the tumor mass and strongly inhibited tumor growth, tumor angiogenesis and development of lung metastasis. The possibility to exploit tumor homing and activity of autologous MMP12-engineered ECFCs represents a novel way to combat melanoma by a “personalized therapy”, without rejection risk. The i.v. injection of radiolabelled MMP12-ECFCs can thus provide a new theranostic approach to control melanoma progression and metastasis.
Collapse
|
41
|
Laurenzana A, Fibbi G, Chillà A, Margheri G, Del Rosso T, Rovida E, Del Rosso M, Margheri F. Lipid rafts: integrated platforms for vascular organization offering therapeutic opportunities. Cell Mol Life Sci 2015; 72:1537-57. [PMID: 25552244 PMCID: PMC11113367 DOI: 10.1007/s00018-014-1814-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Revised: 12/12/2014] [Accepted: 12/19/2014] [Indexed: 02/07/2023]
Abstract
Research on the nanoscale membrane structures known as lipid rafts is relevant to the fields of cancer biology, inflammation and ischaemia. Lipid rafts recruit molecules critical to signalling and regulation of the invasion process in malignant cells, the leukocytes that provide immunity in inflammation and the endothelial cells that build blood and lymphatic vessels, as well as the patterning of neural networks. As angiogenesis is a common denominator, regulation of receptors and signalling molecules critical to angiogenesis is central to the design of new approaches aimed at reducing, promoting or normalizing the angiogenic process. The goal of this review is to highlight some of the key issues that indicate the involvement of endothelial cell lipid rafts at each step of so-called 'sprouting angiogenesis', from stimulation of the vascular endothelial growth factor to the choice of tip cells, activation of migratory and invasion pathways, recruitment of molecules that guide axons in vascular patterning and maturation of blood vessels. Finally, the review addresses opportunities for future studies to define how these lipid domains (and their constituents) may be manipulated to stimulate the so-called 'normalization' of vascular networks within tumors, and be identified as the main target, enabling the development of more efficient chemotherapeutics and cancer immunotherapies.
Collapse
Affiliation(s)
- Anna Laurenzana
- Section of Experimental Pathology and Oncology, Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale GB Morgagni 50, 50134 Florence, Italy
| | - Gabriella Fibbi
- Section of Experimental Pathology and Oncology, Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale GB Morgagni 50, 50134 Florence, Italy
| | - Anastasia Chillà
- Section of Experimental Pathology and Oncology, Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale GB Morgagni 50, 50134 Florence, Italy
| | - Giancarlo Margheri
- Institute of Complex Systems (ISC), Consiglio Nazionale delle Ricerche (CNR), Florence, Italy
| | - Tommaso Del Rosso
- Department of Physics, Pontificia Universidade Catolica do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Elisabetta Rovida
- Section of Experimental Pathology and Oncology, Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale GB Morgagni 50, 50134 Florence, Italy
| | - Mario Del Rosso
- Section of Experimental Pathology and Oncology, Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale GB Morgagni 50, 50134 Florence, Italy
- Istituto Toscano Tumori, Florence, Italy
| | - Francesca Margheri
- Section of Experimental Pathology and Oncology, Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale GB Morgagni 50, 50134 Florence, Italy
| |
Collapse
|
42
|
Calvani M, Pelon F, Comito G, Taddei ML, Moretti S, Innocenti S, Nassini R, Gerlini G, Borgognoni L, Bambi F, Giannoni E, Filippi L, Chiarugi P. Norepinephrine promotes tumor microenvironment reactivity through β3-adrenoreceptors during melanoma progression. Oncotarget 2015; 6:4615-32. [PMID: 25474135 PMCID: PMC4467103 DOI: 10.18632/oncotarget.2652] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Accepted: 10/27/2014] [Indexed: 11/25/2022] Open
Abstract
Stress has an emerging role in cancer and targeting stress-related β-adrenergic receptors (AR) has been proposed as a potential therapeutic approach in melanoma. Here we report that β3-AR expression correlates with melanoma aggressiveness. In addition, we highlight that β3-AR expression is not only restricted to cancer cells, but it is also expressed in vivo in stromal, inflammatory and vascular cells of the melanoma microenvironment. Particularly, we demonstrated that β3-AR can (i) instruct melanoma cells to respond to environmental stimuli, (ii) enhance melanoma cells response to stromal fibroblasts and macrophages, (iii) increase melanoma cell motility and (iv) induce stem-like traits. Noteworthy, β3-AR activation in melanoma accessory cells drives stromal reactivity by inducing pro-inflammatory cytokines secretion and de novo angiogenesis, sustaining tumor growth and melanoma aggressiveness. β3-ARs also play a mandatory role in the recruitment to tumor sites of circulating stromal cells precursors, in the differentiation of these cells towards different lineages, further favoring tumor inflammation, angiogenesis and ultimately melanoma malignancy. Our findings validate selective β3-AR antagonists as potential promising anti-metastatic agents. These could be used to complement current therapeutic approaches for melanoma patients (e.g. propranolol) by targeting non-neoplastic stromal cells, hence reducing therapy resistance of melanoma.
Collapse
Affiliation(s)
- Maura Calvani
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Tuscany Tumor Institute and “Center for Research, Transfer and High Education DenoTHE”, Florence 50134, Italy
| | - Floriane Pelon
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Tuscany Tumor Institute and “Center for Research, Transfer and High Education DenoTHE”, Florence 50134, Italy
| | - Giuseppina Comito
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Tuscany Tumor Institute and “Center for Research, Transfer and High Education DenoTHE”, Florence 50134, Italy
| | - Maria Letizia Taddei
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Tuscany Tumor Institute and “Center for Research, Transfer and High Education DenoTHE”, Florence 50134, Italy
| | - Silvia Moretti
- Department of Surgery and Translational Medicine, Dermatology Section University of Florence, Florence, Italy
| | | | - Romina Nassini
- Department of Health Sciences, Clinical Pharmacology and Oncology Unit, University of Florence, Florence, Italy
| | - Gianni Gerlini
- Plastic Surgery Unit, Regional Melanoma Referral Center, Tuscan Tumor Institute, Santa Maria Annunziata Hospital, Florence 50012, Italy
| | - Lorenzo Borgognoni
- Plastic Surgery Unit, Regional Melanoma Referral Center, Tuscan Tumor Institute, Santa Maria Annunziata Hospital, Florence 50012, Italy
| | - Franco Bambi
- Transfusion Medicine and Cell Therapy “A. Meyer” University Children's Hospital, Florence, Italy
| | - Elisa Giannoni
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Tuscany Tumor Institute and “Center for Research, Transfer and High Education DenoTHE”, Florence 50134, Italy
| | - Luca Filippi
- Neonatal Intensive Care Unit, Medical Surgical Fetal-Neonatal Department, “A. Meyer” University Children's Hospital, Florence, Italy
| | - Paola Chiarugi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Tuscany Tumor Institute and “Center for Research, Transfer and High Education DenoTHE”, Florence 50134, Italy
| |
Collapse
|
43
|
Laurenzana A, Biagioni A, Bianchini F, Peppicelli S, Chillà A, Margheri F, Luciani C, Pimpinelli N, Del Rosso M, Calorini L, Fibbi G. Inhibition of uPAR-TGFβ crosstalk blocks MSC-dependent EMT in melanoma cells. J Mol Med (Berl) 2015; 93:783-94. [PMID: 25694039 DOI: 10.1007/s00109-015-1266-2] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Revised: 01/29/2015] [Accepted: 02/03/2015] [Indexed: 12/26/2022]
Abstract
UNLABELLED The capacity of cancer cells to undergo epithelial-to-mesenchymal transition (EMT) is now considered a hallmark of tumor progression, and it is known that interactions between cancer cells and mesenchymal stem cells (MSCs) of tumor microenvironment may promote this program. Herein, we demonstrate that MSC-conditioned medium (MSC-CM) is a potent inducer of EMT in melanoma cells. The EMT profile acquired by MSC-CM-exposed melanoma cells is characterized by an enhanced level of mesenchymal markers, including TGFβ/TGFβ-receptors system upregulation, by increased invasiveness and uPAR expression, and in vivo tumor growth. Silencing TGFβ in MSC is found to abrogate ability of MSC to promote EMT characteristics in melanoma cells, together with uPAR expression, and this finding is strengthened using an antagonist peptide of TGFβRIII, the so-called P17. Finally, we demonstrate that the uPAR antisense oligonucleotide (uPAR aODN) may inhibit EMT of melanoma cells either stimulated by exogenous TGFβ or MSC-CM. Thus, uPAR upregulation in melanoma cells exposed to MSC-medium drives TGFβ-mediated EMT. On the whole, TGFβ/uPAR dangerous liaison in cancer cell/MSC interactions may disclose a new strategy to abrogate melanoma progression. KEY MESSAGE Mesenchymal stem cell (MSC)-conditioned medium induces EMT-like profile in melanoma. MSC-derived TGFβ promotes uPAR and TGFβ/TGFβ-receptor upregulation in melanoma. TGFβ gene silencing in MSCs downregulates uPAR expression and EMT in melanoma. uPAR downregulation prevents MSC-induced EMT-like profile in melanoma cells. Inhibition of the dangerous TGFβ/uPAR relationship might abrogate melanoma progression.
Collapse
Affiliation(s)
- Anna Laurenzana
- Department of Experimental and Clinical Biomedical Science, University of Florence, Viale G.B. Morgagni, 50, 50134, Florence, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Margheri F, Luciani C, Taddei ML, Giannoni E, Laurenzana A, Biagioni A, Chillà A, Chiarugi P, Fibbi G, Del Rosso M. The receptor for urokinase-plasminogen activator (uPAR) controls plasticity of cancer cell movement in mesenchymal and amoeboid migration style. Oncotarget 2015; 5:1538-53. [PMID: 24681666 PMCID: PMC4039230 DOI: 10.18632/oncotarget.1754] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The receptor for the urokinase plasminogen activator (uPAR) is up-regulated in malignant tumors. Historically the function of uPAR in cancer cell invasion is strictly related to its property to promote uPA-dependent proteolysis of extracellular matrix and to open a path to malignant cells. These features are typical of mesenchymal motility. Here we show that the full-length form of uPAR is required when prostate and melanoma cancer cells convert their migration style from the “path generating” mesenchymal to the “path finding” amoeboid one, thus conferring a plasticity to tumor cell invasiveness across three-dimensional matrices. Indeed, in response to a protease inhibitors-rich milieu, prostate and melanoma cells activated an amoeboid invasion program connoted by retraction of cell protrusions, RhoA-mediated rounding of the cell body, formation of a cortical ring of actin and a reduction of Rac-1 activation. While the mesenchymal movement was reduced upon silencing of uPAR expression, the amoeboid one was almost completely abolished, in parallel with a deregulation of small Rho-GTPases activity. In melanoma and prostate cancer cells we have shown uPAR colocalization with β1/β3 integrins and actin cytoskeleton, as well integrins-actin co-localization under both mesenchymal and amoeboid conditions. Such co-localizations were lost upon treatment of cells with a peptide that inhibits uPAR-integrin interactions. Similarly to uPAR silencing, the peptide reduced mesenchymal invasion and almost abolished the amoeboid one. These results indicate that full-length uPAR bridges the mesenchymal and amoeboid style of movement by an inward-oriented activity based on its property to promote integrin-actin interactions and the following cytoskeleton assembly.
Collapse
Affiliation(s)
- Francesca Margheri
- Department of Experimental and Clinical Biomedical Sciences, University of FlorenceIstituto Toscano Tumori
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Margheri F, Papucci L, Schiavone N, D'Agostino R, Trigari S, Serratì S, Laurenzana A, Biagioni A, Luciani C, Chillà A, Andreucci E, Del Rosso T, Margheri G, Del Rosso M, Fibbi G. Differential uPAR recruitment in caveolar-lipid rafts by GM1 and GM3 gangliosides regulates endothelial progenitor cells angiogenesis. J Cell Mol Med 2014; 19:113-23. [PMID: 25313007 PMCID: PMC4288355 DOI: 10.1111/jcmm.12410] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Accepted: 07/24/2014] [Indexed: 12/13/2022] Open
Abstract
Gangliosides and the urokinase plasminogen activator receptor (uPAR) tipically partition in specialized membrane microdomains called lipid-rafts. uPAR becomes functionally important in fostering angiogenesis in endothelial progenitor cells (EPCs) upon recruitment in caveolar-lipid rafts. Moreover, cell membrane enrichment with exogenous GM1 ganglioside is pro-angiogenic and opposite to the activity of GM3 ganglioside. On these basis, we first checked the interaction of uPAR with membrane models enriched with GM1 or GM3, relying on the adoption of solid-supported mobile bilayer lipid membranes with raft-like composition formed onto solid hydrophilic surfaces, and evaluated by surface plasmon resonance (SPR) the extent of uPAR recruitment. We estimated the apparent dissociation constants of uPAR-GM1/GM3 complexes. These preliminary observations, indicating that uPAR binds preferentially to GM1-enriched biomimetic membranes, were validated by identifying a pro-angiogenic activity of GM1-enriched EPCs, based on GM1-dependent uPAR recruitment in caveolar rafts. We have observed that addition of GM1 to EPCs culture medium promotes matrigel invasion and capillary morphogenesis, as opposed to the anti-angiogenesis activity of GM3. Moreover, GM1 also stimulates MAPKinases signalling pathways, typically associated with an angiogenesis program. Caveolar-raft isolation and Western blotting of uPAR showed that GM1 promotes caveolar-raft partitioning of uPAR, as opposed to control and GM3-challenged EPCs. By confocal microscopy, we have shown that in EPCs uPAR is present on the surface in at least three compartments, respectively, associated to GM1, GM3 and caveolar rafts. Following GM1 exogenous addition, the GM3 compartment is depleted of uPAR which is recruited within caveolar rafts thereby triggering angiogenesis.
Collapse
Affiliation(s)
- Francesca Margheri
- Department of Experimental and Clinical Biomedical Sciences, Section of Experimental Pathology and Oncology, University of Florence, Florence, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Taddei ML, Cavallini L, Comito G, Giannoni E, Folini M, Marini A, Gandellini P, Morandi A, Pintus G, Raspollini MR, Zaffaroni N, Chiarugi P. Senescent stroma promotes prostate cancer progression: the role of miR-210. Mol Oncol 2014; 8:1729-46. [PMID: 25091736 DOI: 10.1016/j.molonc.2014.07.009] [Citation(s) in RCA: 100] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Revised: 07/10/2014] [Accepted: 07/11/2014] [Indexed: 12/30/2022] Open
Abstract
We focused our interest on senescent human-derived fibroblasts in the progression of prostate cancer. Hypoxic senescent fibroblasts promote prostate cancer aggressiveness by inducing epithelial to mesenchymal transition (EMT) and by secreting energy-rich compounds to support cancer cell growth. Hypoxic senescent fibroblasts additionally increase: i) the recruitment of monocytes and their M2-macrophage polarization, ii) the recruitment of bone marrow-derived endothelial precursor cells, facilitating their vasculogenic ability and iii) capillary morphogenesis, proliferation and invasion of human mature endothelial cells. In addition, we highlight that overexpression of the hypoxia-induced miR-210 in young fibroblasts increases their senescence-associated features and converts them into cancer associated fibroblast (CAF)-like cells, able to promote cancer cells EMT, to support angiogenesis and to recruit endothelial precursor cells and monocytes/macrophages.
Collapse
Affiliation(s)
- Maria Letizia Taddei
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134 Florence, Italy.
| | - Lorenzo Cavallini
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134 Florence, Italy
| | - Giuseppina Comito
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134 Florence, Italy
| | - Elisa Giannoni
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134 Florence, Italy
| | - Marco Folini
- Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Via G. Amadeo 42, 20133 Milan, Italy
| | - Alberto Marini
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B 07100 Sassari, Italy
| | - Paolo Gandellini
- Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Via G. Amadeo 42, 20133 Milan, Italy
| | - Andrea Morandi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134 Florence, Italy
| | - Gianfranco Pintus
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B 07100 Sassari, Italy
| | - Maria Rosaria Raspollini
- Histology and Molecular Diagnostic, University Hospital Careggi, Viale G.B. Morgagni 85, 50134 Florence, Italy
| | - Nadia Zaffaroni
- Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Via G. Amadeo 42, 20133 Milan, Italy
| | - Paola Chiarugi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134 Florence, Italy; Center for Research, Transfer and High Education 'Study at Molecular and Clinical Level of Chronic, Inflammatory, Degenerative and Neoplastic Disorders for the Development on Novel Therapies', Italy
| |
Collapse
|
47
|
Parri M, Pietrovito L, Grandi A, Campagnoli S, De Camilli E, Bianchini F, Marchiò S, Bussolino F, Jin B, Sarmientos P, Grandi G, Viale G, Pileri P, Chiarugi P, Grifantini R. Angiopoietin-like 7, a novel pro-angiogenetic factor over-expressed in cancer. Angiogenesis 2014; 17:881-96. [PMID: 24903490 DOI: 10.1007/s10456-014-9435-4] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Accepted: 05/22/2014] [Indexed: 12/21/2022]
Abstract
Angiopoietin-like (ANGPTL) proteins are secreted proteins showing structural similarity to members of the angiopoietin family. Some ANGPTL proteins possess pleiotropic activities, being involved in cancer lipid, glucose energy metabolisms, and angiogenesis. ANGPTL7 is the less characterized member of the family whose functional role is only marginally known. In this study, we provide experimental evidences that ANGPTL7 is over-expressed in different human cancers. To understand the role played by ANGPTL7 in tumor biology, we asked whether ANGPTL7 is endogenously expressed by malignant cells or in response to environmental stimuli. We found that ANGPTL7 is marginally expressed under standard growth condition while it is specifically up-regulated by hypoxia. Interestingly, the protein is secreted and partially associated with the exosomal fraction, suggesting that it could be found in the systemic circulation of oncologic patients and act in an endocrine way. Moreover, we found that ANGPTL7 exerts a pro-angiogenetic effect on human differentiated endothelial cells by stimulating their proliferation, motility, invasiveness, and capability to form capillary-like networks while it does not stimulate progenitor endothelial cells. Finally, we showed that ANGPTL7 promotes vascularization in vivo in the mouse Matrigel sponge assay, thereby accrediting this molecule as a pro-angiogenic factor.
Collapse
Affiliation(s)
- Matteo Parri
- Externautics SpA, Via Fiorentina 1, 53100, Siena, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Sipos PI, Rens W, Schlecht H, Fan X, Wareing M, Hayward C, Hubel CA, Bourque S, Baker PN, Davidge ST, Sibley CP, Crocker IP. Uterine vasculature remodeling in human pregnancy involves functional macrochimerism by endothelial colony forming cells of fetal origin. Stem Cells 2014; 31:1363-70. [PMID: 23554274 PMCID: PMC3813980 DOI: 10.1002/stem.1385] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Revised: 02/26/2013] [Accepted: 03/04/2013] [Indexed: 12/19/2022]
Abstract
The potency of adult-derived circulating progenitor endothelial colony forming cells (ECFCs) is drastically surpassed by their fetal counterparts. Human pregnancy is associated with robust intensification of blood flow and vascular expansion in the uterus, crucial for placental perfusion and fetal supply. Here, we investigate whether fetal ECFCs transmigrate to maternal bloodstream and home to locations of maternal vasculogenesis, primarily the pregnant uterus. In the first instance, endothelial-like cells, originating from mouse fetuses expressing paternal eGFP, were identified within uterine endothelia. Subsequently, LacZ or enhanced green fluorescent protein (eGFP)-labeled human fetal ECFCs, transplanted into immunodeficient (NOD/SCID) fetuses on D15.5 pregnancy, showed similar integration into the mouse uterus by term. Mature endothelial controls (human umbilical vein endothelial cells), similarly introduced, were unequivocally absent. In humans, SRY was detected in 6 of 12 myometrial microvessels obtained from women delivering male babies. The copy number was calculated at 175 [IQR 149-471] fetal cells per millimeter square endothelium, constituting 12.5% of maternal vessel lumina. Cross-sections of similar human vessels, hybridized for Y-chromosome, positively identified endothelial-associated fetal cells. It appears that through ECFC donation, fetuses assist maternal uterine vascular expansion in pregnancy, potentiating placental perfusion and consequently their own fetal supply. In addition to fetal growth, this cellular mechanism holds implications for materno-fetal immune interactions and long-term maternal vascular health.
Collapse
Affiliation(s)
- Peter I Sipos
- Maternal and Fetal Health Research Centre, University of Manchester, Manchester, UK
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Mesenchymal to amoeboid transition is associated with stem-like features of melanoma cells. Cell Commun Signal 2014; 12:24. [PMID: 24690323 PMCID: PMC4022383 DOI: 10.1186/1478-811x-12-24] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Accepted: 03/11/2014] [Indexed: 12/11/2022] Open
Abstract
Background Cellular plasticity confers cancer cells the ability to adapt to microenvironmental changes, a fundamental requirement for tumour progression and metastasis. The epithelial to mesenchymal transition (EMT) is a transcriptional programme associated with increased cell motility and stemness. Besides EMT, the mesenchymal to amoeboid transition (MAT) has been described during tumour progression but to date, little is known about its transcriptional control and involvement in stemness. The aim of this manuscript is to investigate (i) the transcriptional profile associated with the MAT programme and (ii) to study whether MAT acquisition in melanoma cancer cells correlates with clonogenic potential to promote tumour growth. Results By using a multidisciplinary approach, we identified four different treatments able to induce MAT in melanoma cells: EphA2 overexpression, Rac1 functional inhibition using its RacN17 dominant negative mutant, stimulation with Ilomastat or treatment with the RhoA activator Calpeptin. First, gene expression profiling identified the transcriptional pathways associated with MAT, independently of the stimulus that induces the MAT programme. Notably, gene sets associated with the repression of mesenchymal traits, decrease in the secretion of extracellular matrix components as well as increase of cellular stemness positively correlate with MAT. Second, the link between MAT and stemness has been investigated in vitro by analysing stemness markers and clonogenic potential of melanoma cells undergoing MAT. Finally, the link between MAT inducing treatments and tumour initiating capability has been validated in vivo. Conclusion Taken together, our results demonstrate that MAT programme in melanoma is characterised by increased stemness and clonogenic features of cancer cells, thus sustaining tumour progression. Furthermore, these data suggest that stemness is not an exclusive feature of cells undergoing EMT, but more generally is associated with an increase in cellular plasticity of cancer cells.
Collapse
|
50
|
Asuthkar S, Gogineni VR, Rao JS, Velpula KK. Nuclear Translocation of Hand-1 Acts as a Molecular Switch to Regulate Vascular Radiosensitivity in Medulloblastoma Tumors: The Protein uPAR Is a Cytoplasmic Sequestration Factor for Hand-1. Mol Cancer Ther 2014; 13:1309-22. [DOI: 10.1158/1535-7163.mct-13-0892] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|