1
|
Li Santi A, Alfieri M, Meo L, Ragno P. Let-7 Family microRNAs Regulate the Expression of the Urokinase-Receptor in Acute Myeloid Leukemia Cells. Cells 2025; 14:623. [PMID: 40358147 DOI: 10.3390/cells14090623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 03/31/2025] [Accepted: 04/16/2025] [Indexed: 05/15/2025] Open
Abstract
The urokinase-receptor (uPAR) exerts multiple functions supporting most cancer hallmarks. Increased uPAR expression is associated with an unfavorable prognosis in several cancer types, including hematologic malignancies. We previously reported that three oncosuppressor microRNAs (miRNAs) can target the 3'untranslated region (3'UTR) of the uPAR mRNA and that uPAR mRNA is a competitive endogenous RNA (ceRNA) able to recruit oncosuppressor miRs, thus impairing their activity. We now show that uPAR mRNA can also be targeted by oncosuppressor members of the let-7 miRNA family in acute myeloid leukemia (AML) cell lines. Indeed, let-7a, let7d and let-7g directly target the 3'UTR of uPAR mRNA, thus down-regulating uPAR expression. These let-7 miRNAs are expressed in KG1 and U937 AML cells; their levels are high in KG1 cells, which express low uPAR levels, and low in the U937 cell line, expressing high levels of uPAR. Overexpression of these miRNAs down-regulates uPAR expression and impairs the adhesion to fibronectin and migration of U937 cells, without affecting their proliferation. Accordingly, the overexpression of specific inhibitors targeting these let-7 miRNAs efficiently increases uPAR expression in KG1 cells. These results indicate that selected let-7 miRNAs regulate uPAR expression and impair the adhesion and migration of AML cells.
Collapse
MESH Headings
- Humans
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/pathology
- Receptors, Urokinase Plasminogen Activator/genetics
- Receptors, Urokinase Plasminogen Activator/metabolism
- Cell Line, Tumor
- 3' Untranslated Regions/genetics
- Gene Expression Regulation, Leukemic
- Cell Movement/genetics
- Cell Proliferation/genetics
- U937 Cells
- Cell Adhesion/genetics
- Gene Expression Regulation, Neoplastic
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Down-Regulation/genetics
Collapse
Affiliation(s)
- Anna Li Santi
- Department of Chemistry and Biology, University of Salerno, 84084 Fisciano (Salerno), Italy
| | - Mariaevelina Alfieri
- Clinical Pathology, Pausilipon Hospital, A.O.R.N Santobono-Pausilipon, 80129 Naples, Italy
| | - Luigia Meo
- Department of Chemistry and Biology, University of Salerno, 84084 Fisciano (Salerno), Italy
| | - Pia Ragno
- Department of Chemistry and Biology, University of Salerno, 84084 Fisciano (Salerno), Italy
| |
Collapse
|
2
|
Nagarajan Y, Chandrasekaran N, Deepa Parvathi V. Functionalized Nanomaterials In Pancreatic Cancer Theranostics And Molecular Imaging. ChemistryOpen 2025; 14:e202400232. [PMID: 39434498 PMCID: PMC11726697 DOI: 10.1002/open.202400232] [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: 07/09/2024] [Revised: 08/13/2024] [Indexed: 10/23/2024] Open
Abstract
Pancreatic cancer (PC) is one of the most fatal malignancies in the world. This lethality persists due to lack of effective and efficient treatment strategies. Pancreatic ductal adenocarcinoma (PDAC) is an aggressive epithelial malignancy which has a high incidence rate and contributes to overall cancer fatalities. As of 2022, pancreatic cancer contributes to about 3 % of all cancers globally. Over the years, research has characterised germline predisposition, the origin cell, precursor lesions, genetic alterations, structural alterations, transcriptional changes, tumour heterogeneity, metastatic progression, and the tumour microenvironment, which has improved the understanding of PDAC carcinogenesis. By using molecular-based target therapies, these fundamental advancements support primary prevention, screening, early detection, and treatment. The focus of this review is the use of targeted nanoparticles as an alternative to conventional pancreatic cancer treatment due to the various side effects of the latter. The principles of nanoparticle based cancer therapy is efficient targeting of tumour cells via enhanced permeability and retention (EPR) effects and decrease the chemotherapy side effects due to their non-specificity. To increase the efficiency of existing therapies and modify target nanoparticles, several molecular markers of pancreatic cancer cells have been identified. Thus pancreatic cancer cells can be detected using appropriately functionalized nanoparticles with specific signalling molecules. Once cancer has been identified, these nanoparticles can kill the tumour by inducing hyperthermia, medication delivery, immunotherapy or gene therapy. As potent co-delivery methods for adjuvants and tumor-associated antigens; nanoparticles (NPs) have demonstrated significant promise as delivery vehicles in cancer therapy. This ensures the precise internalization of the functionalized nanoparticle and thus also activates the immune system effectively against tumor cells. This review also discusses the immunological factors behind the uptake of functionalized nanoparticles in cancer therapies. Theranostics, which combine imaging and therapeutic chemicals in a single nanocarrier, are the next generation of medicines. Pancreatic cancer treatment may be revolutionised by the development of a tailored nanocarrier with diagnostic, therapeutic, and imaging capabilities. It is extremely difficult to incorporate various therapeutic modalities into a single nanocarrier without compromising the individual functionalities. Surface modification of nanocarriers with antibodies or proteins will enable to attain multifunctionality which increases the efficiency of pancreatic cancer therapy.
Collapse
Affiliation(s)
- Yoghalakshmi Nagarajan
- Department of Biomedical SciencesFaculty of Biomedical Sciences & TechnologySri Ramachandra Institute of Higher Education and Research (SRIHER)Tamil NaduChennai600116India
| | - Natarajan Chandrasekaran
- Senior Professor & Former DirectorCentre for NanobiotechnologyVellore Institute of Technology (VIT)Vellore Campus, Tiruvalam roadTamil NaduKatpadiVellore 632014
| | - Venkatachalam Deepa Parvathi
- Department of Biomedical SciencesFaculty of Biomedical Sciences & TechnologySri Ramachandra Institute of Higher Education and Research (SRIHER)Tamil NaduChennai600116India
| |
Collapse
|
3
|
Jamshidi S, Tavangar M, Shojaei S, Taherkhani A. Malignant Transformation of Normal Oral Tissue to Dysplasia and Early Oral Squamous Cell Carcinoma: An In Silico Transcriptomics Approach. Anal Cell Pathol (Amst) 2024; 2024:6260651. [PMID: 39376501 PMCID: PMC11458300 DOI: 10.1155/2024/6260651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 06/22/2024] [Accepted: 08/19/2024] [Indexed: 10/09/2024] Open
Abstract
Background: Oral squamous cell carcinoma (OSCC) is a prevalent and aggressive form of head and neck cancer, often diagnosed at advanced stages. Elucidating the molecular mechanisms involved in the malignant transformation from normal oral tissue to oral preinvasive lesions (OPL) and primary OSCC could facilitate early diagnosis and improve therapeutic strategies. Methods: Differentially expressed genes (DEGs) were identified from the GSE30784 dataset by comparing normal oral tissue, oral dysplasia, and primary OSCC samples. Cross-validation was performed using an independent RNA-seq dataset, GSE186775. Protein-protein interaction (PPI) network analysis, gene ontology annotation, and pathway enrichment analysis were conducted on the common DEGs. Hub genes were identified, and their prognostic significance was evaluated using survival analysis. Transcription factor (TF) enrichment analysis, cross-validation, and immunohistochemistry analyses were also performed. Results: A total of 226 proteins and 677 interactions were identified in the PPI network, with 34 hub genes, including FN1, SERPINE1, PLAUR, THBS1, and ITGA6. Pathways such as "Formation of the cornified envelope," "Keratinization," and "Developmental biology" were enriched. Overexpression of SERPINE1, PLAUR, THBS1, and ITGA6 correlated with poor prognosis, while upregulation of CALML5 and SPINK5 was associated with favorable outcomes. NFIB emerged as the most significant TF-regulating hub genes. Immunohistochemistry validated ITGA6 overexpression in primary OSCC. Cross-validation using the RNA-seq dataset supported the involvement of critical genes in the malignant transformation process. Conclusion: This study identified vital genes, pathways, and prognostic markers involved in the malignant transformation from normal oral tissue to OPL and primary OSCC, providing insights for early diagnosis and targeted therapy development. Cross-validation with an independent RNA-seq dataset and immunohistochemistry reinforced the findings, supporting the robustness of the identified molecular signatures.
Collapse
Affiliation(s)
- Shokoofeh Jamshidi
- Department of Oral and Maxillofacial Pathology, School of Dentistry, Dental Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Matina Tavangar
- Department of Oral and Maxillofacial Pathology, School of Dentistry, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Setareh Shojaei
- Department of Oral and Maxillofacial Pathology, School of Dentistry, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Amir Taherkhani
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
4
|
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
|
5
|
Camedda R, Frantellizzi V, Danieli R, De Vincentis G, Filippi L. Positron emission computed tomography targeting urokinase plasminogen activator receptor (uPAR) in cancer: a systematic review. Expert Rev Anticancer Ther 2024; 24:137-145. [PMID: 38451196 DOI: 10.1080/14737140.2024.2328167] [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: 11/19/2023] [Accepted: 03/05/2024] [Indexed: 03/08/2024]
Abstract
INTRODUCTION To provide an overview of the available literature data on clinical applications of positron emission tomography (PET) targeting the urokinase-type plasminogen activator receptor in oncology. METHODS A literature search was conducted in PubMed, Web of Science and Scopus databases up to June 2023. The results were presented according to the PRISMA guidelines. The quality of the studies was assessed using the Critical Appraisal Skill Program checklist. RESULTS Seven papers were selected for final analysis, involving 266 patients with solid tumors who underwent PET with uPAR-ligands. Thematic areas identified include feasibility studies (n = 2) on the safety, pharmacokinetics, and dosimetry of uPAR-targeting radiopharmaceuticals; uPAR-directed imaging in head and neck cancer (n = 2); uPAR PET in prostate cancer (n = 2); and the investigation of uPAR in neuroendocrine neoplasms (n = 1). Six of the seven studies used the radiopharmaceutical [68Ga]Ga-NOTA-AE105 while one study used [64Cu]Cu-DOTA-AE105. The studies showed protocol homogeneity, with static PET imaging at 20 minutes. The quality assessment revealed limitations such as small cohorts and the fact that all studies were performed by a single research group. CONCLUSIONS uPAR-PET appears to be a promising imaging tool in well-selected oncological settings, but it needs to be validated by multicentre collaboration.
Collapse
Affiliation(s)
- Riccardo Camedda
- Department of Biomedicine and Prevention, University Tor Vergata, Rome, Italy
| | - Viviana Frantellizzi
- Department of Radiological Sciences, Oncology and Anatomo-Pathology, Sapienza University of Rome, Rome, Italy
| | - Roberta Danieli
- Department of Human Sciences and Promotion of the Quality of Life, University San Raffaele, Rome, Italy
| | - Giuseppe De Vincentis
- Department of Radiological Sciences, Oncology and Anatomo-Pathology, Sapienza University of Rome, Rome, Italy
| | - Luca Filippi
- Nuclear Medicine Unit, Department of Oncohaematology, Fondazione PTV Policlinico Tor Vergata University Hospital, Rome, Italy
| |
Collapse
|
6
|
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
|
7
|
Matsuoka T, Yashiro M. Molecular Insight into Gastric Cancer Invasion-Current Status and Future Directions. Cancers (Basel) 2023; 16:54. [PMID: 38201481 PMCID: PMC10778111 DOI: 10.3390/cancers16010054] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 12/15/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
Gastric cancer (GC) is one of the most common malignancies worldwide. There has been no efficient therapy for stage IV GC patients due to this disease's heterogeneity and dissemination ability. Despite the rapid advancement of molecular targeted therapies, such as HER2 and immune checkpoint inhibitors, survival of GC patients is still unsatisfactory because the understanding of the mechanism of GC progression is still incomplete. Invasion is the most important feature of GC metastasis, which causes poor mortality in patients. Recently, genomic research has critically deepened our knowledge of which gene products are dysregulated in invasive GC. Furthermore, the study of the interaction of GC cells with the tumor microenvironment has emerged as a principal subject in driving invasion and metastasis. These results are expected to provide a profound knowledge of how biological molecules are implicated in GC development. This review summarizes the advances in our current understanding of the molecular mechanism of GC invasion. We also highlight the future directions of the invasion therapeutics of GC. Compared to conventional therapy using protease or molecular inhibitors alone, multi-therapy targeting invasion plasticity may seem to be an assuring direction for the progression of novel strategies.
Collapse
Affiliation(s)
| | - Masakazu Yashiro
- Molecular Oncology and Therapeutics, Osaka Metropolitan University Graduate School of Medicine, Osaka 5458585, Japan;
| |
Collapse
|
8
|
Azawi N, Mosholt KSS, Fryd ND, Lund L, Brignone JI, Hvid N, Wulf-Johansson H, Pedersen OBV, Saekmose SG, Dabestani S. suPARnostic: an advanced predictive tool for detecting recurrence in renal cell carcinoma. BMC Urol 2023; 23:168. [PMID: 37875832 PMCID: PMC10594785 DOI: 10.1186/s12894-023-01337-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 10/06/2023] [Indexed: 10/26/2023] Open
Abstract
BACKGROUND Plasma soluble urokinase-type Plasminogen Activator Receptor (suPAR) predicts disease aggressiveness in renal cell carcinoma (ccRCC), but its prognostic accuracy has not been investigated. To investigate the prognostic accuracy of preoperative plasma suPAR in patients who received curative treatment for initially localized ccRCC. METHODS We retrospectively analyzed plasma samples stored in the Danish National Biobank between 2010 and 2015 from 235 patients with ccRCC at any stage. Relationships with outcome analyzed using univariate and multiple logistic Cox regression analysis. RESULTS There were 235 patients with ccRCC. The median follow-up period was 7.7 years. In univariate analysis suPAR ≥ 6 ng/mL was significantly associated with overall survival (OS) and recurrence-free survival (RFS). Patients with elevated suPAR were more likely to recur, with a Hazard Ratio (HR) of 2.3 for RFS. In multiple logistic regression, suPAR ≥ 6 ng/mL remained a negative predictor of OS and RFS. Limitations include retrospective study design, wide confidence intervals, and tumor subtype heterogeneity bias. CONCLUSIONS ccRCC patients with high plasma suPAR concentrations are at an elevated risk of disease recurrence and see lower OS. suPAR is a promising surveillance tool to more precisely follow up with ccRCC patients and detect future recurrences. In this study, we showed that new type of liquid marker in blood plasma, called suPAR, is associated to a higher risk of kidney cancer recurrence when elevated above 6ng/mL. We also showed suPAR to independently be able to predict patients overall and recurrence free survival in patient with any stage of kidney cancer.
Collapse
Affiliation(s)
- Nessn Azawi
- Department of Urology, Zealand University Hospital, Sygehusvej 6, Roskilde, 4000, Denmark.
- Institute of Clinical Medicine, University of Copenhagen, Norregade 10, copenhagen, Copenhagen, 1165, Denmark.
| | | | - Nathalie Demuth Fryd
- Deparment of Urology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200 Aarhus N, Aarhus, Denmark
| | - Lars Lund
- Deparment of Urology, Odense University Hospital, J. B. Winslows Vej 4, Odense, 5000, Denmark
| | - Juan Ignacio Brignone
- Department of Urology, Aalborg University hospital, Hobrovej 18-22, Aalborg, Aalborg, 9100, Denmark
| | - Nanna Hvid
- Deparment of Urology, Odense University Hospital, J. B. Winslows Vej 4, Odense, 5000, Denmark
| | - Helle Wulf-Johansson
- Department of Clinical Immunology, Naestved Hospital, Ringstedgade 61, Naestved, Naestved, 4700, Denmark
| | | | - Susanne Gjørup Saekmose
- Department of Clinical Immunology, Naestved Hospital, Ringstedgade 61, Naestved, Naestved, 4700, Denmark
| | - Saeed Dabestani
- Department of Urology, Kristianstad Central Hospital, Region Skane, Kristianstad, Sweden
- Department of Translational Medicine, Division of Urological Cancers, Lund University, Lund, Sweden
| |
Collapse
|
9
|
Napolitano F, Giudice V, Selleri C, Montuori N. Plasminogen System in the Pathophysiology of Sepsis: Upcoming Biomarkers. Int J Mol Sci 2023; 24:12376. [PMID: 37569751 PMCID: PMC10418678 DOI: 10.3390/ijms241512376] [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: 06/27/2023] [Revised: 07/21/2023] [Accepted: 08/01/2023] [Indexed: 08/13/2023] Open
Abstract
Severe hemostatic disturbances and impaired fibrinolysis occur in sepsis. In the most serious cases, the dysregulation of fibrinolysis contributes to septic shock, disseminated intravascular coagulation (DIC), and death. Therefore, an analysis of circulating concentrations of pro- and anti-fibrinolytic mediators could be a winning strategy in both the diagnosis and the treatment of sepsis. However, the optimal cutoff value, the timing of the measurements, and their combination with coagulation indicators should be further investigated. The purpose of this review is to summarize all relevant publications regarding the role of the main components of the plasminogen activation system (PAS) in the pathophysiology of sepsis. In addition, the clinical value of PAS-associated biomarkers in the diagnosis and the outcomes of patients with septic syndrome will be explored. In particular, experimental and clinical trials performed in emergency departments highlight the validity of soluble urokinase plasminogen activator receptor (suPAR) as a predictive and prognostic biomarker in patients with sepsis. The measurements of PAI-I may also be useful, as its increase is an early manifestation of sepsis and may precede the development of thrombocytopenia. The upcoming years will undoubtedly see progress in the use of PAS-associated laboratory parameters.
Collapse
Affiliation(s)
- Filomena Napolitano
- Department of Translational Medical Sciences, University of Naples “Federico II”, 80138 Naples, Italy;
| | - Valentina Giudice
- Hematology and Transplant Center, University Hospital “San Giovanni di Dio e Ruggi d’Aragona”, 84131 Salerno, Italy; (V.G.); (C.S.)
- Department of Medicine and Surgery, University of Salerno, 84081 Baronissi, Italy
| | - Carmine Selleri
- Hematology and Transplant Center, University Hospital “San Giovanni di Dio e Ruggi d’Aragona”, 84131 Salerno, Italy; (V.G.); (C.S.)
- Department of Medicine and Surgery, University of Salerno, 84081 Baronissi, Italy
| | - Nunzia Montuori
- Department of Translational Medical Sciences, University of Naples “Federico II”, 80138 Naples, Italy;
- Center for Basic and Clinical Immunology Research (CISI), WAO Center of Excellence, University of Naples “Federico II”, 80138 Naples, Italy
| |
Collapse
|
10
|
Manzo P, Giudice V, Napolitano F, De Novellis D, Serio B, Moscato P, Montuori N, Selleri C. Macrophages and Urokinase Plasminogen Activator Receptor System in Multiple Myeloma: Case Series and Literature Review. Int J Mol Sci 2023; 24:10519. [PMID: 37445697 DOI: 10.3390/ijms241310519] [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: 05/05/2023] [Revised: 06/10/2023] [Accepted: 06/20/2023] [Indexed: 07/15/2023] Open
Abstract
The microenvironment plays an essential role in multiple myeloma (MM) development, progression, cell proliferation, survival, immunological escape, and drug resistance. Mesenchymal stromal cells and macrophages release tolerogenic cytokines and favor anti-apoptotic signaling pathway activation, while the urokinase plasminogen activator receptor (uPAR) system contributes to migration through an extracellular matrix. Here, we first summarized the role of macrophages and the uPAR system in MM pathogenesis, and then we reported the potential therapeutic effects of uPAR inhibitors in a case series of primary MM-derived adherent cells. Our preliminary results showed that after uPAR inhibitor treatments, interleukein-6 (mean ± SD, 8734.95 ± 4169.2 pg/mL vs. 359.26 ± 393.8 pg/mL, pre- vs. post-treatment; p = 0.0012) and DKK-1 levels (mean ± SD, 7005.41 ± 6393.4 pg/mL vs. 61.74 ± 55.2 pg/mL, pre- vs. post-treatment; p = 0.0043) in culture medium were almost completely abolished, supporting further investigation of uPAR blockade as a therapeutic strategy for MM treatment. Therefore, uPAR inhibitors could exert both anti-inflammatory and pro-immunosurveillance activity. However, our preliminary results need further validation in additional in vitro and in vivo studies.
Collapse
Affiliation(s)
- Paola Manzo
- Hematology and Transplant Center, University Hospital "San Giovanni di Dio e Ruggi d'Aragona", 84131 Salerno, Italy
| | - Valentina Giudice
- Hematology and Transplant Center, University Hospital "San Giovanni di Dio e Ruggi d'Aragona", 84131 Salerno, Italy
- Department of Medicine and Surgery, University of Salerno, 84081 Baronissi, Italy
| | - Filomena Napolitano
- Department of Translational Medical Sciences, University of Naples "Federico II", 80138 Naples, Italy
| | - Danilo De Novellis
- Hematology and Transplant Center, University Hospital "San Giovanni di Dio e Ruggi d'Aragona", 84131 Salerno, Italy
- Department of Medicine and Surgery, University of Salerno, 84081 Baronissi, Italy
| | - Bianca Serio
- Hematology and Transplant Center, University Hospital "San Giovanni di Dio e Ruggi d'Aragona", 84131 Salerno, Italy
| | - Paolo Moscato
- Rheumatology Unit, University Hospital "San Giovanni di Dio e Ruggi d'Aragona", 84131 Salerno, Italy
| | - Nunzia Montuori
- Department of Translational Medical Sciences, University of Naples "Federico II", 80138 Naples, Italy
| | - Carmine Selleri
- Hematology and Transplant Center, University Hospital "San Giovanni di Dio e Ruggi d'Aragona", 84131 Salerno, Italy
- Department of Medicine and Surgery, University of Salerno, 84081 Baronissi, Italy
| |
Collapse
|
11
|
Koistinen H, Kovanen RM, Hollenberg MD, Dufour A, Radisky ES, Stenman UH, Batra J, Clements J, Hooper JD, Diamandis E, Schilling O, Rannikko A, Mirtti T. The roles of proteases in prostate cancer. IUBMB Life 2023; 75:493-513. [PMID: 36598826 PMCID: PMC10159896 DOI: 10.1002/iub.2700] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 11/22/2022] [Indexed: 01/05/2023]
Abstract
Since the proposition of the pro-invasive activity of proteolytic enzymes over 70 years ago, several roles for proteases in cancer progression have been established. About half of the 473 active human proteases are expressed in the prostate and many of the most well-characterized members of this enzyme family are regulated by androgens, hormones essential for development of prostate cancer. Most notably, several kallikrein-related peptidases, including KLK3 (prostate-specific antigen, PSA), the most well-known prostate cancer marker, and type II transmembrane serine proteases, such as TMPRSS2 and matriptase, have been extensively studied and found to promote prostate cancer progression. Recent findings also suggest a critical role for proteases in the development of advanced and aggressive castration-resistant prostate cancer (CRPC). Perhaps the most intriguing evidence for this role comes from studies showing that the protease-activated transmembrane proteins, Notch and CDCP1, are associated with the development of CRPC. Here, we review the roles of proteases in prostate cancer, with a special focus on their regulation by androgens.
Collapse
Affiliation(s)
- Hannu Koistinen
- Department of Clinical Chemistry and Haematology, Faculty of Medicine, University of Helsinki and Helsinki University Hospital, Finland
| | - Ruusu-Maaria Kovanen
- Department of Clinical Chemistry and Haematology, Faculty of Medicine, University of Helsinki and Helsinki University Hospital, Finland
- Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, Finland
- Department of Pathology, HUS Diagnostic Centre, Helsinki University Hospital, Helsinki, Finland
| | - Morley D Hollenberg
- Department of Physiology & Pharmacology and Department of Medicine, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Antoine Dufour
- Department of Physiology & Pharmacology and Department of Medicine, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Evette S. Radisky
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, U.S.A
| | - Ulf-Håkan Stenman
- Department of Clinical Chemistry and Haematology, Faculty of Medicine, University of Helsinki and Helsinki University Hospital, Finland
| | - Jyotsna Batra
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Australia
- Translational Research Institute, Queensland University of Technology, Brisbane, Australia
| | - Judith Clements
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Australia
- Translational Research Institute, Queensland University of Technology, Brisbane, Australia
| | - John D. Hooper
- Mater Research Institute, The University of Queensland, Brisbane, Australia
| | - Eleftherios Diamandis
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Oliver Schilling
- Institute for Surgical Pathology, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Antti Rannikko
- Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, Finland
- Department of Urology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Tuomas Mirtti
- Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, Finland
- Department of Pathology, HUS Diagnostic Centre, Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
12
|
The uPA/uPAR System Orchestrates the Inflammatory Response, Vascular Homeostasis, and Immune System in Fibrosis Progression. Int J Mol Sci 2023; 24:ijms24021796. [PMID: 36675310 PMCID: PMC9866279 DOI: 10.3390/ijms24021796] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 01/11/2023] [Accepted: 01/13/2023] [Indexed: 01/18/2023] Open
Abstract
Fibrotic diseases, such as systemic sclerosis (SSc), idiopathic pulmonary fibrosis, renal fibrosis and liver cirrhosis are characterized by tissue overgrowth due to excessive extracellular matrix (ECM) deposition. Fibrosis progression is caused by ECM overproduction and the inhibition of ECM degradation due to several events, including inflammation, vascular endothelial dysfunction, and immune abnormalities. Recently, it has been reported that urokinase plasminogen activator (uPA) and its receptor (uPAR), known to be fibrinolytic factors, orchestrate the inflammatory response, vascular homeostasis, and immune homeostasis system. The uPA/uPAR system may show promise as a potential therapeutic target for fibrotic diseases. This review considers the role of the uPA/uPAR system in the progression of fibrotic diseases.
Collapse
|
13
|
Alfieri M, Meo L, Ragno P. Posttranscriptional Regulation of the Plasminogen Activation System by Non-Coding RNA in Cancer. Int J Mol Sci 2023; 24:ijms24020962. [PMID: 36674481 PMCID: PMC9860977 DOI: 10.3390/ijms24020962] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 12/30/2022] [Accepted: 01/01/2023] [Indexed: 01/07/2023] Open
Abstract
Various species of non-coding RNAs (ncRNAs) may act as functional molecules regulating diverse biological processes. In cancer cell biology, ncRNAs include RNAs that regulate the expression of oncogenes and tumor suppressor genes through various mechanisms. The urokinase (uPA)-mediated plasminogen activation system (PAS) includes uPA, its inhibitors PAI-1 and PAI-2 and its specific cellular receptor uPAR; their increased expression represents a negative prognostic factor in several cancers. Here, we will briefly describe the main uPA-mediated PAS components and ncRNA species; then, we will review more recent evidence of the roles that ncRNAs may play in regulating the expression and functions of uPA-mediated PAS components in cancer.
Collapse
Affiliation(s)
- Mariaevelina Alfieri
- Clinical Pathology, Pausilipon Hospital, A.O.R.N Santobono-Pausilipon, 80123 Naples, Italy
| | - Luigia Meo
- Department of Chemistry and Biology, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy
| | - Pia Ragno
- Department of Chemistry and Biology, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy
- Correspondence: ; Tel.: +39-089-969456
| |
Collapse
|
14
|
Shen J, Zhao C, Zhang H, Zhou P, Li Z. Classification of tuberculosis-related programmed cell death-related patient subgroups and associated immune cell profiling. Front Immunol 2023; 14:1159713. [PMID: 37205113 PMCID: PMC10185908 DOI: 10.3389/fimmu.2023.1159713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 04/12/2023] [Indexed: 05/21/2023] Open
Abstract
Background Tuberculosis (TB) is the deadliest communicable disease in the world with the exception of the ongoing COVID-19 pandemic. Programmed cell death (PCD) patterns play key roles in the development and progression of many disease states such that they may offer value as effective biomarkers or therapeutic targets that can aid in identifying and treating TB patients. Materials and methods The Gene Expression Omnibus (GEO) was used to gather TB-related datasets after which immune cell profiles in these data were analyzed to examine the potential TB-related loss of immune homeostasis. Profiling of differentially expressed PCD-related genes was performed, after which candidate hub PCD-associated genes were selected via a machine learning approach. TB patients were then stratified into two subsets based on the expression of PCD-related genes via consensus clustering. The potential roles of these PCD-associated genes in other TB-related diseases were further examined. Results In total, 14 PCD-related differentially expressed genes (DEGs) were identified and highly expressed in TB patient samples and significantly correlated with the abundance of many immune cell types. Machine learning algorithms enabled the selection of seven hub PCD-related genes that were used to establish PCD-associated patient subgroups, followed by the validation of these subgroups in independent datasets. These findings, together with GSVA results, indicated that immune-related pathways were significantly enriched in TB patients exhibiting high levels of PCD-related gene expression, whereas metabolic pathways were significantly enriched in the other patient group. Single cell RNA-seq (scRNA-seq) further highlighted significant differences in the immune status of these different TB patient samples. Furthermore, we used CMap to predict five potential drugs for TB-related diseases. Conclusion These results highlight clear enrichment of PCD-related gene expression in TB patients and suggest that this PCD activity is closely associated with immune cell abundance. This thus indicates that PCD may play a role in TB progression through the induction or dysregulation of an immune response. These findings provide a foundation for further research aimed at clarifying the molecular drivers of TB, the selection of appropriate diagnostic biomarkers, and the design of novel therapeutic interventions aimed at treating this deadly infectious disease.
Collapse
Affiliation(s)
- Jie Shen
- School of Medical Laboratory, Weifang Medical University, Weifang, China
| | - Chao Zhao
- Office of Academic Affairs, Weifang Medical University, Weifang, China
| | - Hong Zhang
- School of Public Health, Weifang Medical University, Weifang, China
| | - Peipei Zhou
- School of Medical Laboratory, Weifang Medical University, Weifang, China
| | - Zhenpeng Li
- School of Medical Laboratory, Weifang Medical University, Weifang, China
- *Correspondence: Zhenpeng Li,
| |
Collapse
|
15
|
He B, Huang Z, Huang C, Nice EC. Clinical applications of plasma proteomics and peptidomics: Towards precision medicine. Proteomics Clin Appl 2022; 16:e2100097. [PMID: 35490333 DOI: 10.1002/prca.202100097] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 04/16/2022] [Accepted: 04/28/2022] [Indexed: 02/05/2023]
Abstract
In the context of precision medicine, disease treatment requires individualized strategies based on the underlying molecular characteristics to overcome therapeutic challenges posed by heterogeneity. For this purpose, it is essential to develop new biomarkers to diagnose, stratify, or possibly prevent diseases. Plasma is an available source of biomarkers that greatly reflects the physiological and pathological conditions of the body. An increasing number of studies are focusing on proteins and peptides, including many involving the Human Proteome Project (HPP) of the Human Proteome Organization (HUPO), and proteomics and peptidomics techniques are emerging as critical tools for developing novel precision medicine preventative measures. Excitingly, the emerging plasma proteomics and peptidomics toolbox exhibits a huge potential for studying pathogenesis of diseases (e.g., COVID-19 and cancer), identifying valuable biomarkers and improving clinical management. However, the enormous complexity and wide dynamic range of plasma proteins makes plasma proteome profiling challenging. Herein, we summarize the recent advances in plasma proteomics and peptidomics with a focus on their emerging roles in COVID-19 and cancer research, aiming to emphasize the significance of plasma proteomics and peptidomics in clinical applications and precision medicine.
Collapse
Affiliation(s)
- Bo He
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, P. R. China
| | - Zhao Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, P. R. China
| | - Canhua Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, P. R. China.,Department of Pharmacology, and Provincial Key Laboratory of Pathophysiology in Ningbo University School of Medicine, Ningbo, Zhejiang, China
| | - Edouard C Nice
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
16
|
Carlsen EA, Loft M, Loft A, Berthelsen AK, Langer SW, Knigge U, Kjaer A. Prospective Phase II Trial of Prognostication by 68Ga-NOTA-AE105 uPAR PET in Patients with Neuroendocrine Neoplasms: Implications for uPAR-Targeted Therapy. J Nucl Med 2022; 63:1371-1377. [PMID: 35058319 PMCID: PMC9454472 DOI: 10.2967/jnumed.121.263177] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 01/12/2022] [Indexed: 01/26/2023] Open
Abstract
The clinical course for patients with neuroendocrine neoplasms (NENs) ranges from indolent to highly aggressive. Noninvasive tools to improve prognostication and guide decisions on treatment are warranted. Expression of urokinase plasminogen activator receptor (uPAR) is present in many cancer types and associated with a poor outcome. Therefore, using an in-house-developed uPAR PET tracer [68Ga]Ga-NOTA-Asp-Cha-Phe-D-Ser-D-Arg-Tyr-Leu-Trp-Ser-OH (68Ga-NOTA-AE105), we aimed to assess uPAR expression in NENs. We hypothesized that uPAR expression was detectable in a significant proportion of patients and associated with a poorer outcome. In addition, as uPAR-targeted radionuclide therapy has previously proven effective in preclinical models, the study would also indicate the potential for uPAR-targeted radionuclide therapy in NEN patients. Methods: In a prospective clinical phase II trial, we included 116 patients with NENs of all grades, of whom 96 subsequently had uPAR PET/CT performed with evaluable lesions. PET/CT was performed 20 min after injection of approximately 200 MBq of 68Ga-NOTA-AE105. uPAR target-to-liver ratio was used to define lesions as uPAR-positive when lesion SUVmax-to-liver SUVmean ratio was at least 2. Patients were followed for at least 1 y to assess progression-free and overall survival. Results: Most patients had small intestinal NENs (n = 61) and metastatic disease (n = 86). uPAR-positive lesions were seen in 68% (n = 65) of all patients and in 75% (n = 18) of patients with high-grade (grade 3) NENs. During follow-up (median, 28 mo), 59 patients (62%) experienced progressive disease and 28 patients (30%) died. High uPAR expression, defined as a uPAR target-to-liver ratio above median, had a hazard ratio of 1.87 (95% CI, 1.11-3.17) and 2.64 (95% CI, 1.19-5.88) for progression-free and overall survival, respectively (P < 0.05 for both). Conclusion: When 68Ga-NOTA-AE105 PET was used to image uPAR in patients with NENs, uPAR-positive lesions were seen in most patients, notably in patients with both low-grade and high-grade NENs. Furthermore, uPAR expression was associated with a worse prognosis. We suggest that uPAR PET is relevant for risk stratification and that uPAR may be a promising target for therapy in patients with NENs.
Collapse
Affiliation(s)
- Esben Andreas Carlsen
- Department of Clinical Physiology and Nuclear Medicine & Cluster for Molecular Imaging, Copenhagen University Hospital – Rigshospitalet & Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark;,ENETS Neuroendocrine Tumor Center of Excellence, Copenhagen University Hospital–Rigshospitalet, Copenhagen, Denmark
| | - Mathias Loft
- Department of Clinical Physiology and Nuclear Medicine & Cluster for Molecular Imaging, Copenhagen University Hospital – Rigshospitalet & Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark;,ENETS Neuroendocrine Tumor Center of Excellence, Copenhagen University Hospital–Rigshospitalet, Copenhagen, Denmark
| | - Annika Loft
- Department of Clinical Physiology and Nuclear Medicine & Cluster for Molecular Imaging, Copenhagen University Hospital – Rigshospitalet & Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark;,ENETS Neuroendocrine Tumor Center of Excellence, Copenhagen University Hospital–Rigshospitalet, Copenhagen, Denmark
| | - Anne Kiil Berthelsen
- Department of Clinical Physiology and Nuclear Medicine & Cluster for Molecular Imaging, Copenhagen University Hospital – Rigshospitalet & Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark;,ENETS Neuroendocrine Tumor Center of Excellence, Copenhagen University Hospital–Rigshospitalet, Copenhagen, Denmark
| | - Seppo W. Langer
- ENETS Neuroendocrine Tumor Center of Excellence, Copenhagen University Hospital–Rigshospitalet, Copenhagen, Denmark;,Department of Oncology, Copenhagen University Hospital – Rigshospitalet, Copenhagen, Denmark;,Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark; and
| | - Ulrich Knigge
- ENETS Neuroendocrine Tumor Center of Excellence, Copenhagen University Hospital–Rigshospitalet, Copenhagen, Denmark;,Departments of Clinical Endocrinology and Surgical Gastroenterology, Copenhagen University Hospital–Rigshospitalet, Copenhagen, Denmark
| | - Andreas Kjaer
- Department of Clinical Physiology and Nuclear Medicine & Cluster for Molecular Imaging, Copenhagen University Hospital - Rigshospitalet & Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark; .,ENETS Neuroendocrine Tumor Center of Excellence, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
17
|
Zhang T, Wang B, Su F, Gu B, Xiang L, Gao L, Zheng P, Li XM, Chen H. TCF7L2 promotes anoikis resistance and metastasis of gastric cancer by transcriptionally activating PLAUR. Int J Biol Sci 2022; 18:4560-4577. [PMID: 35864968 PMCID: PMC9295057 DOI: 10.7150/ijbs.69933] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 06/14/2022] [Indexed: 12/04/2022] Open
Abstract
Gastric cancer (GC) is the most common gastrointestinal malignant tumor, and distant metastasis is a critical factor in the prognosis of patients with GC. Understanding the mechanism of GC metastasis will help improve patient prognosis. Studies have confirmed that urokinase-type plasminogen activator receptor (PLAUR) promotes GC metastasis; however, its relationship with anoikis resistance and associated mechanisms remains unclear. In this study, we demonstrated that PLAUR promotes the anoikis resistance and metastasis of GC cells and identified transcription Factor 7 Like 2 (TCF7L2) as an important transcriptional regulator of PLAUR. We also revealed that TCF7L2 is highly expressed in GC and promotes the anoikis resistance and metastasis of GC cells. Moreover, we found that TCF7L2 transcription activates PLAUR. Finally, we confirmed that TCF7L2 is an independent risk factor for poor prognosis of patients with GC. Our results show that TCF7L2 and PLAUR are candidate targets for developing therapeutic strategies for GC metastasis.
Collapse
Affiliation(s)
- Tao Zhang
- Department of oncology, The First Hospital of Lanzhou University, Lanzhou, Gansu, China.,The second clinical medical college of Lanzhou university, Lanzhou , Gansu, China.,Key laboratory of digestive system tumors, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Bofang Wang
- The second clinical medical college of Lanzhou university, Lanzhou , Gansu, China.,Key laboratory of digestive system tumors, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Fei Su
- Department of oncology, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Baohong Gu
- The second clinical medical college of Lanzhou university, Lanzhou , Gansu, China.,Key laboratory of digestive system tumors, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Lin Xiang
- The second clinical medical college of Lanzhou university, Lanzhou , Gansu, China.,Key laboratory of digestive system tumors, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Lei Gao
- The second clinical medical college of Lanzhou university, Lanzhou , Gansu, China.,Key laboratory of digestive system tumors, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Peng Zheng
- The second clinical medical college of Lanzhou university, Lanzhou , Gansu, China.,Key laboratory of digestive system tumors, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Xue-Mei Li
- The second clinical medical college of Lanzhou university, Lanzhou , Gansu, China.,Key laboratory of digestive system tumors, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Hao Chen
- The second clinical medical college of Lanzhou university, Lanzhou , Gansu, China.,Key laboratory of digestive system tumors, Lanzhou University Second Hospital, Lanzhou, Gansu, China.,Cancer center, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| |
Collapse
|
18
|
DSCAM-AS1 Long Non-Coding RNA Exerts Oncogenic Functions in Endometrial Adenocarcinoma via Activation of a Tumor-Promoting Transcriptome Profile. Biomedicines 2022; 10:biomedicines10071727. [PMID: 35885035 PMCID: PMC9313190 DOI: 10.3390/biomedicines10071727] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 07/12/2022] [Accepted: 07/13/2022] [Indexed: 11/16/2022] Open
Abstract
Accumulating evidence suggests that lncRNA DSCAM-AS1 acts tumor-promoting in various cancer entities. In breast cancer, DSCAM-AS1 was shown to be the lncRNA being most responsive to induction by estrogen receptor α (ERα). In this study, we examined the function of DSCAM-AS1 in endometrial adenocarcinoma using in silico and different in vitro approaches. Initial analysis of open-source data revealed DSCAM-AS1 overexpression in endometrial cancer (EC) (p < 0.01) and a significant association with shorter overall survival of EC patients (HR = 1.78, p < 0.01). In EC, DSCAM-AS1 was associated with endometrial tumor promotor gene PRL and with expression of ERα and its target genes TFF1 and PGR. Silencing of this lncRNA by RNAi in two EC cell lines was more efficient in ERα-negative HEC-1B cells and reduced their growth and the expression of proliferation activators like NOTCH1, PTK2 and EGR1. DSCAM-AS1 knockdown triggered an anti-tumoral transcriptome response as revealed by Affymetrix microarray analysis, emerging from down-regulation of tumor-promoting genes and induction of tumor-suppressive networks. Finally, several genes regulated upon DSCAM-AS1 silencing in vitro were found to be inversely correlated with this lncRNA in EC tissues. This study clearly suggests an oncogenic function of DSCAM-AS1 in endometrial adenocarcinoma via activation of a tumor-promoting transcriptome profile.
Collapse
|
19
|
Clinical Efficacy of Xueshuantong plus Urokinase in the Treatment of Sudden Deafness. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:7775556. [PMID: 35815266 PMCID: PMC9259216 DOI: 10.1155/2022/7775556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 05/25/2022] [Accepted: 05/30/2022] [Indexed: 11/17/2022]
Abstract
Objective To investigate the clinical effect of Xueshuantong combined with urokinase in the treatment of sudden deafness. Methods A total of 90 patients with sudden deafness who were treated in South China Hospital affiliated to Shenzhen University from June 2019 to August 2020 were recruited and assigned (1 : 1) into the control group (n = 45, urokinase) and the experimental group (n = 45, Xueshuantong plus urokinase) according to the different treatment methods. The clinical treatment effect, the degree of tinnitus, the average auditory valve of the damaged frequency, and the changes in hemorheology (plasma viscosity, whole blood high-shear reduced viscosity, whole blood low-shear reduced viscosity, hematocrit, and fibrinogen) were compared between the two groups of patients. Results The treatment with urokinase and Xueshuangtong injection in the experimental group resulted in a significantly higher clinical treatment effect when compared with the treatment in the control group (P < 0.05). After treatment, the degree of tinnitus and the average auditory valve of the damaged frequency in the experimental group were significantly lower than those in the control group (P < 0.05). The levels of hemorheology (plasma viscosity, whole blood high-shear reduced viscosity, whole blood low-shear reduced viscosity, hematocrit, and fibrinogen) in the experimental group after treatment were significantly lower than those in the control group (P < 0.05). Conclusion The clinical effect of Xueshuantong combined with urokinase in the treatment of patients with sudden deafness is remarkable, and it can effectively improve the hearing level and hemorheology-related indexes of patients, and it thus merits clinical application.
Collapse
|
20
|
Vaghari-Tabari M, Hassanpour P, Sadeghsoltani F, Malakoti F, Alemi F, Qujeq D, Asemi Z, Yousefi B. CRISPR/Cas9 gene editing: a new approach for overcoming drug resistance in cancer. Cell Mol Biol Lett 2022; 27:49. [PMID: 35715750 PMCID: PMC9204876 DOI: 10.1186/s11658-022-00348-2] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 05/24/2022] [Indexed: 12/18/2022] Open
Abstract
The CRISPR/Cas9 system is an RNA-based adaptive immune system in bacteria and archaea. Various studies have shown that it is possible to target a wide range of human genes and treat some human diseases, including cancers, by the CRISPR/Cas9 system. In fact, CRISPR/Cas9 gene editing is one of the most efficient genome manipulation techniques. Studies have shown that CRISPR/Cas9 technology, in addition to having the potential to be used as a new therapeutic approach in the treatment of cancers, can also be used to enhance the effectiveness of existing treatments. Undoubtedly, the issue of drug resistance is one of the main obstacles in the treatment of cancers. Cancer cells resist anticancer drugs by a variety of mechanisms, such as enhancing anticancer drugs efflux, enhancing DNA repair, enhancing stemness, and attenuating apoptosis. Mutations in some proteins of different cellular signaling pathways are associated with these events and drug resistance. Recent studies have shown that the CRISPR/Cas9 technique can be used to target important genes involved in these mechanisms, thereby increasing the effectiveness of anticancer drugs. In this review article, studies related to the applications of this technique in overcoming drug resistance in cancer cells will be reviewed. In addition, we will give a brief overview of the limitations of the CRISP/Cas9 gene-editing technique.
Collapse
Affiliation(s)
- Mostafa Vaghari-Tabari
- Department of Clinical Biochemistry and Laboratory Medicine, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parisa Hassanpour
- Department of Clinical Biochemistry and Laboratory Medicine, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fatemeh Sadeghsoltani
- Department of Clinical Biochemistry and Laboratory Medicine, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Faezeh Malakoti
- Department of Clinical Biochemistry and Laboratory Medicine, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Forough Alemi
- Department of Clinical Biochemistry and Laboratory Medicine, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Durdi Qujeq
- Cellular and Molecular Biology Research Center (CMBRC), Health Research Institute, Babol University of Medical Sciences, Babol, Iran.,Department of Clinical Biochemistry, Babol University of Medical Sciences, Babol, Iran
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran.
| | - Bahman Yousefi
- Department of Clinical Biochemistry and Laboratory Medicine, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran. .,Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
21
|
Nascente EDP, Amorim RL, Fonseca-Alves CE, de Moura VMBD. Comparative Pathobiology of Canine and Human Prostate Cancer: State of the Art and Future Directions. Cancers (Basel) 2022; 14:2727. [PMID: 35681707 PMCID: PMC9179314 DOI: 10.3390/cancers14112727] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 05/14/2022] [Accepted: 05/15/2022] [Indexed: 02/01/2023] Open
Abstract
First described in 1817, prostate cancer is considered a complex neoplastic entity, and one of the main causes of death in men in the western world. In dogs, prostatic carcinoma (PC) exhibits undifferentiated morphology with different phenotypes, is hormonally independent of aggressive character, and has high rates of metastasis to different organs. Although in humans, the risk factors for tumor development are known, in dogs, this scenario is still unclear, especially regarding castration. Therefore, with the advent of molecular biology, studies were and are carried out with the aim of identifying the main molecular mechanisms and signaling pathways involved in the carcinogenesis and progression of canine PC, aiming to identify potential biomarkers for diagnosis, prognosis, and targeted treatment. However, there are extensive gaps to be filled, especially when considering the dog as experimental model for the study of this neoplasm in humans. Thus, due to the complexity of the subject, the objective of this review is to present the main pathobiological aspects of canine PC from a comparative point of view to the same neoplasm in the human species, addressing the historical context and current understanding in the scientific field.
Collapse
Affiliation(s)
- Eduardo de Paula Nascente
- School of Veterinary Medicine and Animal Science, Federal University of Goiás, Goiânia 74001-970, Brazil;
| | - Renée Laufer Amorim
- Veterinary Clinic Department, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), Botucatu 18618-970, Brazil;
| | - Carlos Eduardo Fonseca-Alves
- Department of Veterinary Surgery and Anesthesiology, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), Botucatu 18618-970, Brazil;
| | | |
Collapse
|
22
|
Napolitano F, Montuori N. Role of Plasminogen Activation System in Platelet Pathophysiology: Emerging Concepts for Translational Applications. Int J Mol Sci 2022; 23:ijms23116065. [PMID: 35682744 PMCID: PMC9181697 DOI: 10.3390/ijms23116065] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 05/25/2022] [Accepted: 05/26/2022] [Indexed: 12/13/2022] Open
Abstract
Traditionally, platelets have been exclusively considered for their procoagulant and antifibrinolytic effects during normal activation of hemostasis. Effectively, activated platelets secrete coagulation factors, expose phosphatidylserine, and promote thrombin and fibrin production. In addition to procoagulant activities, platelets confer resistance of thrombi to fibrinolysis by inducing clot retraction of the fibrin network and release of huge amounts of plasminogen activator inhibitor-1, which is the major physiologic inhibitor of the fibrinolytic cascade. However, the discovery of multiple relations with the fibrinolytic system, also termed Plasminogen Activation System (PAS), has introduced new perspectives on the platelet role in fibrinolysis. Indeed, the activated membrane surface of platelets provides binding sites on which fibrinolytic enzymes can be activated. This review discusses the evidence of the profibrinolytic properties of platelets through the description of PAS components and related proteins that are contained in or bind to platelets. Our analyses of literature data lead to the conclusion that in the initial phase of the hemostatic process, antifibrinolytic effects prevail over profibrinolytic activity, but at later stages, platelets might enhance fibrinolysis through the engagement of PAS components. A better understanding of spatial and temporal characteristics of platelet-mediated fibrinolysis during normal hemostasis could improve therapeutic options for bleeding and thrombotic disorders.
Collapse
|
23
|
Qin L, Wang L, Zhang J, Zhou H, Yang Z, Wang Y, Cai W, Wen F, Jiang X, Zhang T, Ye H, Long B, Qin J, Shi W, Guan X, Yu Z, Yang J, Wang Q, Jiao Z. Therapeutic strategies targeting uPAR potentiate anti-PD-1 efficacy in diffuse-type gastric cancer. SCIENCE ADVANCES 2022; 8:eabn3774. [PMID: 35613265 PMCID: PMC9132454 DOI: 10.1126/sciadv.abn3774] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
The diffuse-type gastric cancer (DGC) is a subtype of gastric cancer (GC) associated with low HER2 positivity rate and insensitivity to chemotherapy and immune checkpoint inhibitors. Here, we identify urokinase-type plasminogen activator receptor (uPAR) as a potential therapeutic target for DGC. We have developed a novel anti-uPAR monoclonal antibody, which targets the domains II and III of uPAR and blocks the binding of urokinase-type plasminogen activator to uPAR. We show that the combination of anti-uPAR and anti-Programmed cell death protein 1 (PD-1) remarkably inhibits tumor growth and prolongs survival via multiple mechanisms, using cell line-derived xenograft and patient-derived xenograft mouse models. Furthermore, uPAR chimeric antigen receptor-expressing T cells based on the novel anti-uPAR effectively kill DGC patient-derived organoids and exhibit impressive survival benefit in the established mouse models, especially when combined with PD-1 blockade therapy. Our study provides a new possibility of DGC treatment by targeting uPAR in a unique manner.
Collapse
Affiliation(s)
- Long Qin
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, China
| | - Long Wang
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, China
| | - Junchang Zhang
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, China
| | - Huinian Zhou
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, China
| | - Zhiliang Yang
- Lanzhou Huazhitiancheng Biotechnologies Co., Ltd, Lanzhou, Gansu 730000, China
| | - Yan Wang
- Lanzhou Huazhitiancheng Biotechnologies Co., Ltd, Lanzhou, Gansu 730000, China
| | - Weiwen Cai
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, China
| | - Fei Wen
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, China
| | - Xiangyan Jiang
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, China
| | - Tiansheng Zhang
- Lanzhou Huazhitiancheng Biotechnologies Co., Ltd, Lanzhou, Gansu 730000, China
| | - Huili Ye
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, China
| | - Bo Long
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, China
| | - Junjie Qin
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, China
| | - Wengui Shi
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, China
| | - Xiaoying Guan
- Department of Pathology, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, China
| | - Zeyuan Yu
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, China
| | - Jing Yang
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, China
- Corresponding author. (Z.J.); (Q.W.); (J.Y.)
| | - Qi Wang
- Lanzhou Huazhitiancheng Biotechnologies Co., Ltd, Lanzhou, Gansu 730000, China
- Corresponding author. (Z.J.); (Q.W.); (J.Y.)
| | - Zuoyi Jiao
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, China
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, China
- Corresponding author. (Z.J.); (Q.W.); (J.Y.)
| |
Collapse
|
24
|
Inhibition of Human Urokinase-Type Plasminogen Activator (uPA) Enzyme Activity and Receptor Binding by DNA Aptamers as Potential Therapeutics through Binding to the Different Forms of uPA. Int J Mol Sci 2022; 23:ijms23094890. [PMID: 35563278 PMCID: PMC9100121 DOI: 10.3390/ijms23094890] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 04/25/2022] [Accepted: 04/26/2022] [Indexed: 02/06/2023] Open
Abstract
Urokinase-type plasminogen activator is widely discussed as a marker for cancer prognosis and diagnosis and as a target for cancer therapies. Together with its receptor, uPA plays an important role in tumorigenesis, tumor progression and metastasis. In the present study, systematic evolution of ligands by exponential enrichment (SELEX) was used to select single-stranded DNA aptamers targeting different forms of human uPA. Selected aptamers allowed the distinction between HMW-uPA and LMW-uPA, and therefore, presumably, have different binding regions. Here, uPAapt-02-FR showed highly affine binding with a KD of 0.7 nM for HMW-uPA and 21 nM for LMW-uPA and was also able to bind to pro-uPA with a KD of 14 nM. Furthermore, no cross-reactivity to mouse uPA or tissue-type plasminogen activator (tPA) was measured, demonstrating high specificity. Suppression of the catalytic activity of uPA and inhibition of uPAR-binding could be demonstrated through binding with different aptamers and several of their truncated variants. Since RNA aptamers are already known to inhibit uPA-uPAR binding and other pathological functions of the uPA system, these aptamers represent a novel, promising tool not only for detection of uPA but also for interfering with the pathological functions of the uPA system by additionally inhibiting uPA activity.
Collapse
|
25
|
Identification of uPAR Variants Acting as ceRNAs in Leukaemia Cells. Cancers (Basel) 2022; 14:cancers14081980. [PMID: 35454884 PMCID: PMC9025028 DOI: 10.3390/cancers14081980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 04/03/2022] [Accepted: 04/11/2022] [Indexed: 12/10/2022] Open
Abstract
Simple Summary The urokinase (uPA) receptor (uPAR) concentrates proteolytic activities on the cell surface and is an adhesion receptor for vitronectin. Urokinase/Vitronectin binding to uPAR activates intracellular signals promoting cell adhesion, migration, proliferation and survival. Thus, uPAR can sustain most activities of malignant cells and, accordingly, increased uPAR expression is associated with poor prognosis in several malignancies. We previously demonstrated that, in leukaemia cells, the uPAR 3′untranslated region (3′UTR) up-regulates the expression of pro-tumoral factors by recruiting microRNAs targeting their mRNAs, thus acting as competitive endogenous RNA (ceRNA). Here, we identify 3′UTR-containing variants of uPAR mRNA in leukaemia cells and demonstrate that the over-expression of uPAR Δ5-variant mRNA promotes expression of pro-tumoral factors and increase in biological activities, probably by its ceRNA activity. On this basis, we propose that uPAR may play a crucial role in cancer biology also at mRNA level, through the ceRNA activity of its variants. Abstract The 3′untranslated region (3′UTR) of the urokinase (uPA) receptor (uPAR) mRNA can act as a competitive endogenous RNA (ceRNA) in acute myeloid leukaemia (AML) cells, promoting the expression of pro-tumoral targets, including uPAR. Here, we identified three variants of uPAR mRNA containing the 3′UTR, in KG1 and U937 leukaemia cells expressing low and high uPAR levels, respectively. Identified variants lack exon 5 (uPAR Δ5) or exon 6 (uPAR Δ6) or part of exon 6, exon 7 and part of 3′UTR (uPAR Δ6/7). uPAR Δ5 and uPAR Δ6 transcript levels were higher in U937 cells compared to KG1 cells. Both uPAR variants were expressed also in AML blasts, at higher levels as compared to CD34 hematopoietic cells from healthy donors. The presence of the 3′UTR conferred high instability to the uPAR Δ5 variant transcript, preventing its translation in protein. Overexpression of the uPAR Δ5-3′UTR variant regulated the expression of some pro-tumoral factors previously reported to be regulated by the 3′UTR of uPAR and increased KG1 cell adhesion, migration and proliferation. These results demonstrate the expression of uPAR mRNA variants containing the 3′UTR in AML cells and the ceRNA activity and the biological effects of the uPAR Δ5-3′UTR variant.
Collapse
|
26
|
Alfano D, Franco P, Stoppelli MP. Modulation of Cellular Function by the Urokinase Receptor Signalling: A Mechanistic View. Front Cell Dev Biol 2022; 10:818616. [PMID: 35493073 PMCID: PMC9045800 DOI: 10.3389/fcell.2022.818616] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 03/15/2022] [Indexed: 12/15/2022] Open
Abstract
Urokinase-type plasminogen activator receptor (uPAR or CD87) is a glycosyl-phosphatidyl-inositol anchored (GPI) membrane protein. The uPAR primary ligand is the serine protease urokinase (uPA), converting plasminogen into plasmin, a broad spectrum protease, active on most extracellular matrix components. Besides uPA, the uPAR binds specifically also to the matrix protein vitronectin and, therefore, is regarded also as an adhesion receptor. Complex formation of the uPAR with diverse transmembrane proteins, including integrins, formyl peptide receptors, G protein-coupled receptors and epidermal growth factor receptor results in intracellular signalling. Thus, the uPAR is a multifunctional receptor coordinating surface-associated pericellular proteolysis and signal transduction, thereby affecting physiological and pathological mechanisms. The uPAR-initiated signalling leads to remarkable cellular effects, that include increased cell migration, adhesion, survival, proliferation and invasion. Although this is beyond the scope of this review, the uPA/uPAR system is of great interest to cancer research, as it is associated to aggressive cancers and poor patient survival. Increasing evidence links the uPA/uPAR axis to epithelial to mesenchymal transition, a highly dynamic process, by which epithelial cells can convert into a mesenchymal phenotype. Furthermore, many reports indicate that the uPAR is involved in the maintenance of the stem-like phenotype and in the differentiation process of different cell types. Moreover, the levels of anchor-less, soluble form of uPAR, respond to a variety of inflammatory stimuli, including tumorigenesis and viral infections. Finally, the role of uPAR in virus infection has received increasing attention, in view of the Covid-19 pandemics and new information is becoming available. In this review, we provide a mechanistic perspective, via the detailed examination of consolidated and recent studies on the cellular responses to the multiple uPAR activities.
Collapse
|
27
|
Miles LA, Krajewski S, Baik N, Parmer RJ, Mueller BM. Plg-RKT Expression in Human Breast Cancer Tissues. Biomolecules 2022; 12:biom12040503. [PMID: 35454092 PMCID: PMC9028288 DOI: 10.3390/biom12040503] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 03/21/2022] [Accepted: 03/23/2022] [Indexed: 12/12/2022] Open
Abstract
The plasminogen activation system regulates the activity of the serine protease, plasmin. The role of plasminogen receptors in cancer progression is being increasingly appreciated as key players in modulation of the tumor microenvironment. The interaction of plasminogen with cells to promote plasminogen activation requires the presence of proteins exposing C-terminal lysines on the cell surface. Plg-RKT is a structurally unique plasminogen receptor because it is an integral membrane protein that is synthesized with and binds plasminogen via a C-terminal lysine exposed on the cell surface. Here, we have investigated the expression of Plg-RKT in human breast tumors and human breast cancer cell lines. Breast cancer progression tissue microarrays were probed with anti-Plg-RKT mAB and we found that Plg-RKT is widely expressed in human breast tumors, that its expression is increased in tumors that have spread to draining lymph nodes and distant organs, and that Plg-RKT expression is most pronounced in hormone receptor (HR)-positive tumors. Plg-RKT was detected by Western blotting in human breast cancer cell lines. By flow cytometry, Plg-RKT cell surface expression was highest on the most aggressive tumor cell line. Future studies are warranted to address the functions of Plg-RKT in breast cancer.
Collapse
Affiliation(s)
- Lindsey A. Miles
- Department of Molecular Medicine, Scripps Research Institute, La Jolla, CA 92037, USA; (L.A.M.); (N.B.)
| | | | - Nagyung Baik
- Department of Molecular Medicine, Scripps Research Institute, La Jolla, CA 92037, USA; (L.A.M.); (N.B.)
| | - Robert J. Parmer
- Department of Medicine, Veterans Administration San Diego Healthcare System, University of California San Diego, San Diego, CA 92161, USA;
| | - Barbara M. Mueller
- San Diego Biomedical Research Institute, San Diego, CA 92121, USA
- Correspondence:
| |
Collapse
|
28
|
Shmakova AA, Klimovich PS, Rysenkova KD, Popov VS, Gorbunova AS, Karpukhina AA, Karagyaur MN, Rubina KA, Tkachuk VA, Semina EV. Urokinase Receptor uPAR Downregulation in Neuroblastoma Leads to Dormancy, Chemoresistance and Metastasis. Cancers (Basel) 2022; 14:cancers14040994. [PMID: 35205745 PMCID: PMC8870350 DOI: 10.3390/cancers14040994] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Revised: 02/05/2022] [Accepted: 02/12/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary uPAR is a membrane receptor that contributes to extracellular matrix remodeling and controls cellular adhesion, proliferation, survival, and migration. We demonstrate that the initially high uPAR expression predicts poor survival in neuroblastoma. However, relapsed neuroblastomas have a significantly decreased uPAR expression. uPAR downregulation in neuroblastoma cells leads to dormancy and resistance to chemotherapeutic drugs. In mice, low uPAR-expressing neuroblastoma cells formed smaller primary tumors but more frequent metastasis. Abstract uPAR is a membrane receptor that binds extracellular protease urokinase, contributes to matrix remodeling and plays a crucial role in cellular adhesion, proliferation, survival, and migration. uPAR overexpression in tumor cells promotes mitogenesis, opening a prospective avenue for targeted therapy. However, uPAR targeting in cancer has potential risks. We have recently shown that uPAR downregulation in neuroblastoma promotes epithelial-mesenchymal transition (EMT), potentially associated with metastasis and chemoresistance. We used data mining to evaluate the role of uPAR expression in primary and relapsed human neuroblastomas. To model the decreased uPAR expression, we targeted uPAR using CRISPR/Cas9 and shRNA in neuroblastoma Neuro2a cells and evaluated their chemosensitivity in vitro as well as tumor growth and metastasis in vivo. We demonstrate that the initially high PLAUR expression predicts poor survival in human neuroblastoma. However, relapsed neuroblastomas have a significantly decreased PLAUR expression. uPAR targeting in neuroblastoma Neuro2a cells leads to p38 activation and an increased p21 expression (suggesting a dormant phenotype). The dormancy in neuroblastoma cells can be triggered by the disruption of uPAR-integrin interaction. uPAR-deficient cells are less sensitive to cisplatin and doxorubicin treatment and exhibit lower p53 activation. Finally, low uPAR-expressing Neuro2a cells formed smaller primary tumors, but more frequent metastasis in mice. To the best of our knowledge, this is the first study revealing the pathological role of dormant uPAR-deficient cancer cells having a chemoresistant and motile phenotype.
Collapse
Affiliation(s)
- Anna A. Shmakova
- National Cardiology Research Center of the Ministry of Health of the Russian Federation, Institute of Experimental Cardiology, 121552 Moscow, Russia; (A.A.S.); (P.S.K.); (K.D.R.); (V.A.T.)
- Faculty of Medicine, Lomonosov Moscow State University, 119192 Moscow, Russia; (V.S.P.); (A.S.G.); (M.N.K.); (K.A.R.)
| | - Polina S. Klimovich
- National Cardiology Research Center of the Ministry of Health of the Russian Federation, Institute of Experimental Cardiology, 121552 Moscow, Russia; (A.A.S.); (P.S.K.); (K.D.R.); (V.A.T.)
- Faculty of Medicine, Lomonosov Moscow State University, 119192 Moscow, Russia; (V.S.P.); (A.S.G.); (M.N.K.); (K.A.R.)
| | - Karina D. Rysenkova
- National Cardiology Research Center of the Ministry of Health of the Russian Federation, Institute of Experimental Cardiology, 121552 Moscow, Russia; (A.A.S.); (P.S.K.); (K.D.R.); (V.A.T.)
- Faculty of Medicine, Lomonosov Moscow State University, 119192 Moscow, Russia; (V.S.P.); (A.S.G.); (M.N.K.); (K.A.R.)
| | - Vladimir S. Popov
- Faculty of Medicine, Lomonosov Moscow State University, 119192 Moscow, Russia; (V.S.P.); (A.S.G.); (M.N.K.); (K.A.R.)
| | - Anna S. Gorbunova
- Faculty of Medicine, Lomonosov Moscow State University, 119192 Moscow, Russia; (V.S.P.); (A.S.G.); (M.N.K.); (K.A.R.)
| | - Anna A. Karpukhina
- Koltzov Institute of Developmental Biology, Russian Academy of Science, 117334 Moscow, Russia;
| | - Maxim N. Karagyaur
- Faculty of Medicine, Lomonosov Moscow State University, 119192 Moscow, Russia; (V.S.P.); (A.S.G.); (M.N.K.); (K.A.R.)
| | - Kseniya A. Rubina
- Faculty of Medicine, Lomonosov Moscow State University, 119192 Moscow, Russia; (V.S.P.); (A.S.G.); (M.N.K.); (K.A.R.)
| | - Vsevolod A. Tkachuk
- National Cardiology Research Center of the Ministry of Health of the Russian Federation, Institute of Experimental Cardiology, 121552 Moscow, Russia; (A.A.S.); (P.S.K.); (K.D.R.); (V.A.T.)
- Faculty of Medicine, Lomonosov Moscow State University, 119192 Moscow, Russia; (V.S.P.); (A.S.G.); (M.N.K.); (K.A.R.)
| | - Ekaterina V. Semina
- National Cardiology Research Center of the Ministry of Health of the Russian Federation, Institute of Experimental Cardiology, 121552 Moscow, Russia; (A.A.S.); (P.S.K.); (K.D.R.); (V.A.T.)
- Faculty of Medicine, Lomonosov Moscow State University, 119192 Moscow, Russia; (V.S.P.); (A.S.G.); (M.N.K.); (K.A.R.)
- Correspondence:
| |
Collapse
|
29
|
Therapeutic Strategies Targeting Urokinase and Its Receptor in Cancer. Cancers (Basel) 2022; 14:cancers14030498. [PMID: 35158766 PMCID: PMC8833673 DOI: 10.3390/cancers14030498] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 01/11/2022] [Accepted: 01/15/2022] [Indexed: 01/19/2023] Open
Abstract
Several studies have ascertained that uPA and uPAR do participate in tumor progression and metastasis and are involved in cell adhesion, migration, invasion and survival, as well as angiogenesis. Increased levels of uPA and uPAR in tumor tissues, stroma and biological fluids correlate with adverse clinic-pathologic features and poor patient outcomes. After binding to uPAR, uPA activates plasminogen to plasmin, a broad-spectrum matrix- and fibrin-degrading enzyme able to facilitate tumor cell invasion and dissemination to distant sites. Moreover, uPAR activated by uPA regulates most cancer cell activities by interacting with a broad range of cell membrane receptors. These findings make uPA and uPAR not only promising diagnostic and prognostic markers but also attractive targets for developing anticancer therapies. In this review, we debate the uPA/uPAR structure-function relationship as well as give an update on the molecules that interfere with or inhibit uPA/uPAR functions. Additionally, the possible clinical development of these compounds is discussed.
Collapse
|
30
|
Sinotsko AE, Bespalov AV, Pashchevskaya NV, Dotsenko VV, Aksenov NA, Aksenova IV. N, N'-Diphenyldithiomalonodiamide: Structural Features, Acidic Properties, and In Silico Estimation of Biological Activity. RUSS J GEN CHEM+ 2021; 91:2136-2150. [PMID: 34934303 PMCID: PMC8680065 DOI: 10.1134/s1070363221110037] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 08/14/2021] [Accepted: 09/18/2021] [Indexed: 11/25/2022]
Abstract
The spectral characteristics of dithiomalondianilide (N,N'-diphenyldithiomalonodiamide) were studied, and the dissociation constant was determined by potentiometric titration. Quantum-chemical methods at the B3LYP-D3BJ/6-311+G (2d,p) level were used to calculate the molecular geometry and vibrational spectra of the most stable tautomeric forms of dithiomalondianilide. The bioavailability parameters were calculated, and possible protein targets were predicted by the protein ligand docking method.
Collapse
Affiliation(s)
| | | | | | - V. V. Dotsenko
- Kuban State University, 350040 Krasnodar, Russia
- North Caucasus Federal University, 355009 Stavropol, Russia
| | - N. A. Aksenov
- North Caucasus Federal University, 355009 Stavropol, Russia
| | - I. V. Aksenova
- North Caucasus Federal University, 355009 Stavropol, Russia
| |
Collapse
|
31
|
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
|
32
|
Metrangolo V, Ploug M, Engelholm LH. The Urokinase Receptor (uPAR) as a "Trojan Horse" in Targeted Cancer Therapy: Challenges and Opportunities. Cancers (Basel) 2021; 13:cancers13215376. [PMID: 34771541 PMCID: PMC8582577 DOI: 10.3390/cancers13215376] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/15/2021] [Accepted: 10/19/2021] [Indexed: 12/23/2022] Open
Abstract
Simple Summary Discovered more than three decades ago, the urokinase-type plasminogen activator receptor (uPAR) has now firmly established itself as a versatile molecular target holding promise for the treatment of aggressive malignancies. The copious abundance of uPAR in virtually all human cancerous tissues versus their healthy counterparts has fostered a gradual shift in the therapeutic landscape targeting this receptor from function inhibition to cytotoxic approaches to selectively eradicate the uPAR-expressing cells by delivering a targeted cytotoxic insult. Multiple avenues are being explored in a preclinical setting, including the more innovative immune- or stroma targeting therapies. This review discusses the current state of these strategies, their potentialities, and challenges, along with future directions in the field of uPAR targeting. Abstract One of the largest challenges to the implementation of precision oncology is identifying and validating selective tumor-driving targets to enhance the therapeutic efficacy while limiting off-target toxicity. In this context, the urokinase-type plasminogen activator receptor (uPAR) has progressively emerged as a promising therapeutic target in the management of aggressive malignancies. By focalizing the plasminogen activation cascade and subsequent extracellular proteolysis on the cell surface of migrating cells, uPAR endows malignant cells with a high proteolytic and migratory potential to dissolve the restraining extracellular matrix (ECM) barriers and metastasize to distant sites. uPAR is also assumed to choreograph multiple other neoplastic stages via a complex molecular interplay with distinct cancer-associated signaling pathways. Accordingly, high uPAR expression is observed in virtually all human cancers and is frequently associated with poor patient prognosis and survival. The promising therapeutic potential unveiled by the pleiotropic nature of this receptor has prompted the development of distinct targeted intervention strategies. The present review will focus on recently emerged cytotoxic approaches emphasizing the novel technologies and related limits hindering their application in the clinical setting. Finally, future research directions and emerging opportunities in the field of uPAR targeting are also discussed.
Collapse
Affiliation(s)
- Virginia Metrangolo
- The Finsen Laboratory, Rigshospitalet, DK-2200 Copenhagen, Denmark; (V.M.); (M.P.)
- Biotech Research & Innovation Centre (BRIC), Department of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Michael Ploug
- The Finsen Laboratory, Rigshospitalet, DK-2200 Copenhagen, Denmark; (V.M.); (M.P.)
- Biotech Research & Innovation Centre (BRIC), Department of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Lars H. Engelholm
- The Finsen Laboratory, Rigshospitalet, DK-2200 Copenhagen, Denmark; (V.M.); (M.P.)
- Biotech Research & Innovation Centre (BRIC), Department of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
- Correspondence: ; Tel.: +45-31-43-20-77
| |
Collapse
|
33
|
Liu M, Chen S, Zhang A, Zheng Q, Fu J. PLAUR as a Potential Biomarker Associated with Immune Infiltration in Bladder Urothelial Carcinoma. J Inflamm Res 2021; 14:4629-4641. [PMID: 34552345 PMCID: PMC8450190 DOI: 10.2147/jir.s326559] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 08/20/2021] [Indexed: 12/16/2022] Open
Abstract
Background Bladder urothelial carcinoma (BLCA) is one of the most lethal and aggressive malignancies of genitourinary system that affects human health. The urokinase plasminogen activator receptor (PLAUR) plays essential roles in tumorigenesis and immune modulation, and its aberrant expression is closely correlated with cancer progression. However, whether PLAUR has the potential to be one promising biomarker or immunotherapy target for BLCA is unknown. Methodology Various online databases were applied to assess the expression profile and prognostic value of PLAUR, as well as its correlation with immune infiltration in BLCA, including Oncomine, PrognoScan, TCGA, cBioPortal, TIMER, TISIDB, UALCAN, and MethSurv. The expression of PLAUR in BLCA was confirmed with ELISA assay for serum samples and immunohistochemistry for tissue samples. Results The results showed that the expression of PLAUR was elevated in BLCA, which was further confirmed by ELISA and immunohistochemistry. Patients with higher PLAUR level were predicted to have lower overall survival and disease specific survival rates, which were not impacted by the genetic alterations of PLAUR. In addition, the expression of PLAUR was positively associated with immune infiltration, and also the expression levels of gene markers of various immune cells. The negative correlation between PLAUR expression and PLAUR methylation level was observed, among which PLAUR expression was positively correlated with the abundance of 28 kinds of tumor-infiltrating lymphocytes, while PLAUR methylation level was negatively correlated with the abundance of 11 types of tumor-infiltrating lymphocytes. Moreover, the methylation level of PLAUR was closely correlated with patients’ clinicopathological features, and hypomethylation of PLAUR was associated with better outcomes of BLCA patients. Conclusion These findings suggested that PLAUR had the potential to serve as a valuable detection and prognostic biomarker or immunotherapeutic target for BLCA.
Collapse
Affiliation(s)
- Mulin Liu
- Department of Clinical Laboratory, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, 116011, People's Republic of China
| | - Siyi Chen
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Dalian Medical University, Dalian, Liaoning Province, 116044, People's Republic of China
| | - Aihui Zhang
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Dalian Medical University, Dalian, Liaoning Province, 116044, People's Republic of China
| | - Qin Zheng
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Dalian Medical University, Dalian, Liaoning Province, 116044, People's Republic of China
| | - Juan Fu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, 116011, People's Republic of China
| |
Collapse
|
34
|
Leth JM, Ploug M. Targeting the Urokinase-Type Plasminogen Activator Receptor (uPAR) in Human Diseases With a View to Non-invasive Imaging and Therapeutic Intervention. Front Cell Dev Biol 2021; 9:732015. [PMID: 34490277 PMCID: PMC8417595 DOI: 10.3389/fcell.2021.732015] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 07/26/2021] [Indexed: 12/31/2022] Open
Abstract
The interaction between the serine protease urokinase-type plasminogen activator (uPA) and its glycolipid-anchored receptor (uPAR) focalizes plasminogen activation to cell surfaces, thereby regulating extravascular fibrinolysis, cell adhesion, and migration. uPAR belongs to the Ly6/uPAR (LU) gene superfamily and the high-affinity binding site for uPA is assembled by a dynamic association of its three consecutive LU domains. In most human solid cancers, uPAR is expressed at the invasive areas of the tumor-stromal microenvironment. High levels of uPAR in resected tumors or shed to the plasma of cancer patients are robustly associated with poor prognosis and increased risk of relapse and metastasis. Over the years, a plethora of different strategies to inhibit uPA and uPAR function have been designed and investigated in vitro and in vivo in mouse models, but so far none have been implemented in the clinics. In recent years, uPAR-targeting with the intent of cytotoxic eradication of uPAR-expressing cells have nonetheless gained increasing momentum. Another avenue that is currently being explored is non-invasive imaging with specific uPAR-targeted reporter-molecules containing positron emitting radionuclides or near-infrared (NIR) florescence probes with the overarching aim of being able to: (i) localize disease dissemination using positron emission tomography (PET) and (ii) assist fluorescence guided surgery using optical imaging. In this review, we will discuss these advancements with special emphasis on applications using a small 9-mer peptide antagonist that targets uPAR with high affinity.
Collapse
Affiliation(s)
- Julie Maja Leth
- Finsen Laboratory, Rigshospitalet, Copenhagen, Denmark.,Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Michael Ploug
- Finsen Laboratory, Rigshospitalet, Copenhagen, Denmark.,Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
35
|
Park JY, Shin Y, Won WR, Lim C, Kim JC, Kang K, Husni P, Lee ES, Youn YS, Oh KT. Development of AE147 Peptide-Conjugated Nanocarriers for Targeting uPAR-Overexpressing Cancer Cells. Int J Nanomedicine 2021; 16:5437-5449. [PMID: 34408417 PMCID: PMC8367088 DOI: 10.2147/ijn.s315619] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 07/02/2021] [Indexed: 12/12/2022] Open
Abstract
Purpose An AE147 peptide-conjugated nanocarrier based on PEGylated liposomes was developed in order to target the metastatic tumors overexpressing urokinase-type plasminogen activator receptor (uPAR), which cancer progression via uPA signaling. Therefore, the AE147 peptide-conjugated nanocarrier system may hold the potential for active targeting of metastatic tumors. Methods The AE147 peptide, an antagonist of uPAR, was conjugated to the PEGylated liposomes for targeting metastatic tumors overexpressing uPAR. Docetaxel (DTX), an anticancer drug, was incorporated into the nanocarriers. The structure of the AE147-conjugated nanocarrier, its physicochemical properties, and in vivo biodistribution were evaluated. Results The DTX-loaded nanocarrier showed a spherical structure, a high drug-loading capacity, and a high colloidal stability. Drug carrying AE147 conjugates were actively taken up by the uPAR-overexpressing MDA-MB-231 cancer cells. In vivo animal imaging confirmed that the AE147-conjugated nanoparticles effectively accumulated at the sites of tumor metastasis. Conclusion The AE147-nanocarrier showed potential for targeting metastatic tumor cells overexpressing uPAR and as a nanomedicine platform for theragnosis applications. These results suggest that this novel nano-platform will facilitate further advancements in cancer therapy.
Collapse
Affiliation(s)
- June Yong Park
- Department of Global Innovative Drugs, The Graduate School of Chung-Ang University, Dongjak-gu, Seoul, 06974, Republic of Korea
| | - Yuseon Shin
- Department of Global Innovative Drugs, The Graduate School of Chung-Ang University, Dongjak-gu, Seoul, 06974, Republic of Korea
| | - Woong Roeck Won
- Department of Global Innovative Drugs, The Graduate School of Chung-Ang University, Dongjak-gu, Seoul, 06974, Republic of Korea
| | - Chaemin Lim
- Department of Global Innovative Drugs, The Graduate School of Chung-Ang University, Dongjak-gu, Seoul, 06974, Republic of Korea.,Center for Nanotechnology in Drug Delivery and Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Jae Chang Kim
- Department of Global Innovative Drugs, The Graduate School of Chung-Ang University, Dongjak-gu, Seoul, 06974, Republic of Korea
| | - Kioh Kang
- Department of Global Innovative Drugs, The Graduate School of Chung-Ang University, Dongjak-gu, Seoul, 06974, Republic of Korea
| | - Patihul Husni
- Department of Global Innovative Drugs, The Graduate School of Chung-Ang University, Dongjak-gu, Seoul, 06974, Republic of Korea
| | - Eun Seong Lee
- Division of Biotechnology, The Catholic University of Korea, Bucheon, Gyeonggi-do, 14662, Republic of Korea
| | - Yu Seok Youn
- School of Pharmacy, Sungkyunkwan University, Suwon, Gyeonggi-do, 16419, Republic of Korea
| | - Kyung Taek Oh
- Department of Global Innovative Drugs, The Graduate School of Chung-Ang University, Dongjak-gu, Seoul, 06974, Republic of Korea.,College of Pharmacy, Chung-Ang University, Dongjak-gu, Seoul, 06974, Republic of Korea
| |
Collapse
|
36
|
Montemagno C, Cassim S, De Leiris N, Durivault J, Faraggi M, Pagès G. Pancreatic Ductal Adenocarcinoma: The Dawn of the Era of Nuclear Medicine? Int J Mol Sci 2021; 22:6413. [PMID: 34203923 PMCID: PMC8232627 DOI: 10.3390/ijms22126413] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/18/2021] [Accepted: 05/20/2021] [Indexed: 12/12/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC), accounting for 90-95% of all pancreatic tumors, is a highly devastating disease associated with poor prognosis. The lack of accurate diagnostic tests and failure of conventional therapies contribute to this pejorative issue. Over the last decade, the advent of theranostics in nuclear medicine has opened great opportunities for the diagnosis and treatment of several solid tumors. Several radiotracers dedicated to PDAC imaging or internal vectorized radiotherapy have been developed and some of them are currently under clinical consideration. The functional information provided by Positron Emission Tomography (PET) or Single Photon Emission Computed Tomography (SPECT) could indeed provide an additive diagnostic value and thus help in the selection of patients for targeted therapies. Moreover, the therapeutic potential of β-- and α-emitter-radiolabeled agents could also overcome the resistance to conventional therapies. This review summarizes the current knowledge concerning the recent developments in the nuclear medicine field for the management of PDAC patients.
Collapse
Affiliation(s)
- Christopher Montemagno
- Département de Biologie Médicale, Centre Scientifique de Monaco, 98000 Monaco, Monaco; (S.C.); (J.D.); (G.P.)
- Institute for Research on Cancer and Aging of Nice, Centre Antoine Lacassagne, CNRS UMR 7284 and IN-SERM U1081, Université Cote d’Azur, 06200 Nice, France
- LIA ROPSE, Laboratoire International Associé Université Côte d’Azur—Centre Scientifique de Monaco, 98000 Monaco, Monaco
| | - Shamir Cassim
- Département de Biologie Médicale, Centre Scientifique de Monaco, 98000 Monaco, Monaco; (S.C.); (J.D.); (G.P.)
- LIA ROPSE, Laboratoire International Associé Université Côte d’Azur—Centre Scientifique de Monaco, 98000 Monaco, Monaco
| | - Nicolas De Leiris
- Nuclear Medicine Department, Grenoble-Alpes University Hospital, 38000 Grenoble, France;
- Laboratoire Radiopharmaceutiques Biocliniques, Univ. Grenoble Alpes, INSERM, CHU Grenoble Alpes, 38000 Grenoble, France
| | - Jérôme Durivault
- Département de Biologie Médicale, Centre Scientifique de Monaco, 98000 Monaco, Monaco; (S.C.); (J.D.); (G.P.)
- LIA ROPSE, Laboratoire International Associé Université Côte d’Azur—Centre Scientifique de Monaco, 98000 Monaco, Monaco
| | - Marc Faraggi
- Centre Hospitalier Princesse Grace, Nuclear Medicine Department, 98000 Monaco, Monaco;
| | - Gilles Pagès
- Département de Biologie Médicale, Centre Scientifique de Monaco, 98000 Monaco, Monaco; (S.C.); (J.D.); (G.P.)
- Institute for Research on Cancer and Aging of Nice, Centre Antoine Lacassagne, CNRS UMR 7284 and IN-SERM U1081, Université Cote d’Azur, 06200 Nice, France
- LIA ROPSE, Laboratoire International Associé Université Côte d’Azur—Centre Scientifique de Monaco, 98000 Monaco, Monaco
| |
Collapse
|