1
|
Jensen TSR, Olsen MH, Lelkaitis G, Kjaer A, Binderup T, Fugleholm K. Urokinase Plasminogen Activator Receptor: An Important Focal Player in Chronic Subdural Hematoma? Inflammation 2024; 47:1015-1027. [PMID: 38236383 PMCID: PMC11147925 DOI: 10.1007/s10753-023-01957-5] [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: 12/01/2023] [Revised: 12/18/2023] [Accepted: 12/23/2023] [Indexed: 01/19/2024]
Abstract
Chronic subdural hematoma (CSDH) development involves inflammatory, angiogenetic, and fibrinolytic mechanisms, several components of which are now unraveled through intensive research. The urokinase plasminogen activator receptor (uPAR) is part of the plasminogen activator system and possesses inflammatory, angiogenetic, and fibrinolytic capabilities. As a first, this study aims to identify uPAR in the hematoma fluid, hematoma membrane, dura mater, and systemic blood from patients with CSDH and, if present, to investigate if the uPAR level at the time of surgery may be a predictor for later developing recurrent CSDH. uPAR expression in the hematoma membrane and dura mater was analyzed using immunohistochemistry and presented as the H-score of the positive immunostaining. The uPAR levels in the hematoma fluid and systemic blood were determined using a multiplex antibody bead kit (Luminex). Samples were collected at the time of the first CSDH surgery, and in the case of recurrent CSDH within 90 days, the samples were again collected at reoperation. A comparison of uPAR expression between the hematoma membrane and dura mater, as well as uPAR levels in systemic blood and hematoma fluid, was performed using the Wilcoxon rank sum test. We included 112 patients, 26 of whom had recurrent CSDH. The median hematoma uPAR level was 22,125 (14,845-33,237) and significantly higher than the median systemic blood level of 789 pg/L (465-2,088) (p < 0.001). Similarly, the uPAR level of the hematoma membrane was 14.3 (7.54-44.8) and significantly higher than the dural uPAR level of 0.81 (0.3-1.98) (p < 0.001). For the first time, we identified uPAR in the subdural fluid, hematoma membrane, dura mater, and systemic blood from patients with CSDH. The high expression of uPAR in the subdural fluid and hematoma membrane indicates that the mechanisms of CSDH are predominantly in the subdural fluid collection and surrounding hematoma membrane.
Collapse
Affiliation(s)
- Thorbjørn Søren Rønn Jensen
- Department of Neurosurgery, The Neuroscience Center, Copenhagen University Hospital, Inge Lehmanns Vej 6, 2100, Rigshospitalet, Copenhagen, Denmark.
| | - Markus Harboe Olsen
- Department of Neuroanesthesiology, The Neuroscience Center, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Department of Anaesthesiology, Zealand University Hospital, Køge, Denmark
| | | | - Andreas Kjaer
- Department of Clinical Physiology, Nuclear Medicine and PET & Cluster for Molecular Imaging, Copenhagen University Hospital-Rigshospitalet & Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 9, 2100, Copenhagen, Denmark
| | - Tina Binderup
- Department of Clinical Physiology, Nuclear Medicine and PET & Cluster for Molecular Imaging, Copenhagen University Hospital-Rigshospitalet & Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 9, 2100, Copenhagen, Denmark
| | - Kåre Fugleholm
- Department of Neurosurgery, The Neuroscience Center, Copenhagen University Hospital, Inge Lehmanns Vej 6, 2100, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
2
|
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: 13] [Impact Index Per Article: 13.0] [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
|
3
|
Wang G, Hu JQ, Liu JY, Zhang XM. Angiogenesis-Related Gene Signature-Derived Risk Score for Glioblastoma: Prospects for Predicting Prognosis and Immune Heterogeneity in Glioblastoma. Front Cell Dev Biol 2022; 10:778286. [PMID: 35372355 PMCID: PMC8971933 DOI: 10.3389/fcell.2022.778286] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 02/15/2022] [Indexed: 12/16/2022] Open
Abstract
Background: Glioblastoma multiforme (GBM) is the most common malignant tumor in the central nervous system with poor prognosis and unsatisfactory therapeutic efficacy. Considering the high correlation between tumors and angiogenesis, we attempted to construct a more effective model with angiogenesis-related genes (ARGs) to better predict therapeutic response and prognosis. Methods: The ARG datasets were downloaded from the NCBI-Gene and Molecular Signatures Database. The gene expression data and clinical information were obtained from TCGA and CGGA databases. The differentially expressed angiogenesis-related genes (DE-ARGs) were screened with the R package “DESeq2”. Univariate Cox proportional hazards regression analysis was used to screen for ARGs related to overall survival. The redundant ARGs were removed by least absolute shrinkage and selection operator (LASSO) regression analysis. Based on the gene signature of DE-ARGs, a risk score model was established, and its effectiveness was estimated through Kaplan–Meier analysis, ROC analysis, etc. Results: A total of 626 DE-ARGs were explored between GBM and normal samples; 31 genes were identified as key DE-ARGs. Then, the risk score of ARG signature was established. Patients with high-risk score had poor survival outcomes. It was proved that the risk score could predict some medical treatments’ response, such as temozolomide chemotherapy, radiotherapy, and immunotherapy. Besides, the risk score could serve as a promising prognostic predictor. Three key prognostic genes (PLAUR, ITGA5, and FMOD) were selected and further discussed. Conclusion: The angiogenesis-related gene signature-derived risk score is a promising predictor of prognosis and treatment response in GBM and will help in making appropriate therapeutic strategies.
Collapse
Affiliation(s)
- Gang Wang
- Department of Neurosurgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Jin-Qu Hu
- Department of Neurosurgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Ji-Yuan Liu
- Department of Neurosurgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Xiao-Mei Zhang
- Department of Rheumatology and Immunology, ShengJing Hospital of China Medical University, Shenyang, China
- *Correspondence: Xiao-Mei Zhang,
| |
Collapse
|
4
|
Zhai BT, Tian H, Sun J, Zou JB, Zhang XF, Cheng JX, Shi YJ, Fan Y, Guo DY. Urokinase-type plasminogen activator receptor (uPAR) as a therapeutic target in cancer. J Transl Med 2022; 20:135. [PMID: 35303878 PMCID: PMC8932206 DOI: 10.1186/s12967-022-03329-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 03/03/2022] [Indexed: 12/22/2022] Open
Abstract
Urokinase-type plasminogen activator receptor (uPAR) is an attractive target for the treatment of cancer, because it is expressed at low levels in healthy tissues but at high levels in malignant tumours. uPAR is closely related to the invasion and metastasis of malignant tumours, plays important roles in the degradation of extracellular matrix (ECM), tumour angiogenesis, cell proliferation and apoptosis, and is associated with the multidrug resistance (MDR) of tumour cells, which has important guiding significance for the judgement of tumor malignancy and prognosis. Several uPAR-targeted antitumour therapeutic agents have been developed to suppress tumour growth, metastatic processes and drug resistance. Here, we review the recent advances in the development of uPAR-targeted antitumor therapeutic strategies, including nanoplatforms carrying therapeutic agents, photodynamic therapy (PDT)/photothermal therapy (PTT) platforms, oncolytic virotherapy, gene therapy technologies, monoclonal antibody therapy and tumour immunotherapy, to promote the translation of these therapeutic agents to clinical applications.
Collapse
Affiliation(s)
- Bing-Tao Zhai
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Huan Tian
- Xi'an Hospital of Traditional Chinese Medicine, Xi'an, 710021, China
| | - Jing Sun
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Jun-Bo Zou
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Xiao-Fei Zhang
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Jiang-Xue Cheng
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Ya-Jun Shi
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Yu Fan
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Dong-Yan Guo
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an, 712046, China.
| |
Collapse
|
5
|
Tagirasa R, Yoo E. Role of Serine Proteases at the Tumor-Stroma Interface. Front Immunol 2022; 13:832418. [PMID: 35222418 PMCID: PMC8873516 DOI: 10.3389/fimmu.2022.832418] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 01/24/2022] [Indexed: 01/19/2023] Open
Abstract
During tumor development, invasion and metastasis, the intimate interaction between tumor and stroma shapes the tumor microenvironment and dictates the fate of tumor cells. Stromal cells can also influence anti-tumor immunity and response to immunotherapy. Understanding the molecular mechanisms that govern this complex and dynamic interplay, thus is important for cancer diagnosis and therapy. Proteolytic enzymes that are expressed and secreted by both cancer and stromal cells play important roles in modulating tumor-stromal interaction. Among, several serine proteases such as fibroblast activation protein, urokinase-type plasminogen activator, kallikrein-related peptidases, and granzymes have attracted great attention owing to their elevated expression and dysregulated activity in the tumor microenvironment. This review highlights the role of serine proteases that are mainly derived from stromal cells in tumor progression and associated theranostic applications.
Collapse
|
6
|
Li J, Fan H, Zhou X, Xiang Y, Liu Y. Prognostic Significance and Gene Co-Expression Network of PLAU and PLAUR in Gliomas. Front Oncol 2022; 11:602321. [PMID: 35087738 PMCID: PMC8787124 DOI: 10.3389/fonc.2021.602321] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 12/16/2021] [Indexed: 02/05/2023] Open
Abstract
The urokinase-type plasminogen activator(PLAU) and its receptor PLAUR participate in a series of cell physiological activities on the extracellular surface. Abnormal expression of PLAU and PLAUR is associated with tumorigenesis. This study aims to evaluate the prognostic value of PLAU/PLAUR transcription expression in glioma and to explore how they affect the generation and progression of glioma. In this study, online databases are applied, such as Oncomine, GEPIA, CGGA, cBioPortal, and LinkedOmics. Overexpression of PLAU/PLAUR was found to be significantly associated with clinical variables including age, tumor type, WHO grade, histology, IDH-1 mutation, and 1p19q status. PLAU and PLAUR had a high correlation in transcriptional expression levels. High expression of PLAU and PLAUR predicted a poor prognosis in primary glioma and recurrent glioma patients, especially in lower grade gliomas. Cox regression analysis indicated that high expression of PLAU and PLAUR were independent prognostic factors for shorter overall survival in glioma patients. In gene co-expression network analysis PLAU and PLAUR and their co-expression genes were found to be involved in inflammatory activities and tumor-related signaling pathways. In conclusion, PLAU and PLAUR could be promising prognostic biomarkers and potential therapeutic targets of glioma patients.
Collapse
Affiliation(s)
- Junhong Li
- Department of Neurosurgery, West China Hospital of Sichuan University, Chengdu, China
| | - Huanhuan Fan
- West China Brain Research Center, West China Hospital of Sichuan University, Chengdu, China
| | - Xingwang Zhou
- Department of Neurosurgery, West China Hospital of Sichuan University, Chengdu, China
| | - Yufan Xiang
- Department of Neurosurgery, West China Hospital of Sichuan University, Chengdu, China
| | - Yanhui Liu
- Department of Neurosurgery, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
7
|
Hu P, Chiarini A, Wu J, Freddi G, Nie K, Armato U, Prà ID. Exosomes of adult human fibroblasts cultured on 3D silk fibroin nonwovens intensely stimulate neoangiogenesis. BURNS & TRAUMA 2021; 9:tkab003. [PMID: 34212056 PMCID: PMC8240536 DOI: 10.1093/burnst/tkab003] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 12/16/2020] [Indexed: 12/11/2022]
Abstract
Background Bombyx mori silk fibroin is a biomacromolecule that allows the assembly of scaffolds for tissue engineering and regeneration purposes due to its cellular adhesiveness, high biocompatibility and low immunogenicity. Earlier work showed that two types of 3D silk fibroin nonwovens (3D-SFnws) implanted into mouse subcutaneous tissue were promptly vascularized via undefined molecular mechanisms. The present study used nontumorigenic adult human dermal fibroblasts (HDFs) adhering to a third type of 3D-SFnws to assess whether HDFs release exosomes whose contents promote neoangiogenesis. Methods Electron microscopy imaging and physical tests defined the features of the novel carded/hydroentangled 3D-SFnws. HDFs were cultured on 3D-SFnws and polystyrene plates in an exosome-depleted medium. DNA amounts and D-glucose consumption revealed the growth and metabolic activities of HDFs on 3D-SFnws. CD9-expressing total exosome fractions were from conditioned media of 3D-SFnws and 2D polystyrene plates HDF cultures. Angiogenic growth factors (AGFs) in equal amounts of the two groups of exosomal proteins were analysed via double-antibody arrays. A tube formation assay using human dermal microvascular endothelial cells (HDMVECs) was used to evaluate the exosomes’ angiogenic power. Results The novel features of the 3D-SFnws met the biomechanical requirements typical of human soft tissues. By experimental day 15, 3D-SFnws-adhering HDFs had increased 4.5-fold in numbers and metabolized 5.4-fold more D-glucose than at day 3 in vitro. Compared to polystyrene-stuck HDFs, exosomes from 3D-SFnws-adhering HDFs carried significantly higher amounts of AGFs, such as interleukin (IL)-1α, IL-4 and IL-8; angiopoietin-1 and angiopoietin-2; angiopoietin-1 receptor (or Tie-2); growth-regulated oncogene (GRO)-α, GRO-β and GRO-γ; matrix metalloproteinase-1; tissue inhibitor metalloproteinase-1; and urokinase-type plasminogen activator surface receptor, but lesser amounts of anti-angiogenic tissue inhibitor metalloproteinase-2 and pro-inflammatory monocyte chemoattractant protein-1. At concentrations from 0.62 to 10 μg/ml, the exosomes from 3D-SFnws-cultured HDFs proved their angiogenic power by inducing HDMVECs to form significant amounts of tubes in vitro. Conclusions The structural and mechanical properties of carded/hydroentangled 3D-SFnws proved their suitability for tissue engineering and regeneration applications. Consistent with our hypothesis, 3D-SFnws-adhering HDFs released exosomes carrying several AGFs that induced HDMVECs to promptly assemble vascular tubes in vitro. Hence, we posit that once implanted in vivo, the 3D-SFnws/HDFs interactions could promote the vascularization and repair of extended skin wounds due to burns or other noxious agents in human and veterinary clinical settings.
Collapse
Affiliation(s)
- Peng Hu
- Human Histology & Embryology Section, Department of Surgery, Dentistry, Paediatrics & Gynaecology, University of Verona Medical School, Strada Le Grazie 8, I-37134, Verona, Venetia, Italy.,Department of Burns & Plastic Surgery, The Affiliated Hospital of ZunYi Medical University, 149 Dalian Road, ZunYi City, 563003 Guizhou Province, China
| | - Anna Chiarini
- Human Histology & Embryology Section, Department of Surgery, Dentistry, Paediatrics & Gynaecology, University of Verona Medical School, Strada Le Grazie 8, I-37134, Verona, Venetia, Italy
| | - Jun Wu
- Human Histology & Embryology Section, Department of Surgery, Dentistry, Paediatrics & Gynaecology, University of Verona Medical School, Strada Le Grazie 8, I-37134, Verona, Venetia, Italy.,Department of Burns and Plastic Surgery, Second People's Hospital, University of Shenzhen, 3002 Sungang West Road, Futian District, Shenzhen, 518000, Guangdong Province, China
| | - Giuliano Freddi
- Silk Biomaterials S.r.l., Via Cavour 2, I-22074, Lomazzo, Lombardy, Italy
| | - Kaiyu Nie
- Department of Burns & Plastic Surgery, The Affiliated Hospital of ZunYi Medical University, 149 Dalian Road, ZunYi City, 563003 Guizhou Province, China
| | - Ubaldo Armato
- Human Histology & Embryology Section, Department of Surgery, Dentistry, Paediatrics & Gynaecology, University of Verona Medical School, Strada Le Grazie 8, I-37134, Verona, Venetia, Italy.,Department of Burns and Plastic Surgery, Second People's Hospital, University of Shenzhen, 3002 Sungang West Road, Futian District, Shenzhen, 518000, Guangdong Province, China
| | - Ilaria Dal Prà
- Human Histology & Embryology Section, Department of Surgery, Dentistry, Paediatrics & Gynaecology, University of Verona Medical School, Strada Le Grazie 8, I-37134, Verona, Venetia, Italy.,Department of Burns and Plastic Surgery, Second People's Hospital, University of Shenzhen, 3002 Sungang West Road, Futian District, Shenzhen, 518000, Guangdong Province, China
| |
Collapse
|
8
|
Rysenkova KD, Klimovich PS, Shmakova AA, Karagyaur MN, Ivanova KA, Aleksandrushkina NA, Tkachuk VA, Rubina KA, Semina EV. Urokinase receptor deficiency results in EGFR-mediated failure to transmit signals for cell survival and neurite formation in mouse neuroblastoma cells. Cell Signal 2020; 75:109741. [PMID: 32822758 DOI: 10.1016/j.cellsig.2020.109741] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 08/14/2020] [Accepted: 08/14/2020] [Indexed: 12/26/2022]
Abstract
Urokinase-type plasminogen activator uPA and its receptor (uPAR) are the central players in extracellular matrix proteolysis, which facilitates cancer invasion and metastasis. EGFR is one of the important components of uPAR interactome. uPAR/EGFR interaction controls signaling pathways that regulate cell survival, proliferation and migration. We have previously established that uPA binding to uPAR stimulates neurite elongation in neuroblastoma cells, while blocking uPA/uPAR interaction induces neurite branching and new neurite formation. Here we demonstrate that blocking the uPA binding to uPAR with anti-uPAR antibody decreases the level of pEGFR and its downstream pERK1/2, but does increase phosphorylation of Akt, p38 and c-Src Since long-term uPAR blocking results in a severe DNA damage, accompanied by PARP-1 proteolysis and Neuro2a cell death, we surmise that Akt, p38 and c-Src activation transmits a pro-apoptotic signal, rather than a survival. Serum deprivation resulting in enhanced neuritogenesis is accompanied by an upregulated uPAR mRNA expression, while EGFR mRNA remains unchanged. EGFR activation by EGF stimulates neurite growth only in uPAR-overexpressing cells but not in control or uPAR-deficient cells. In addition, AG1478-mediated inhibition of EGFR activity impedes neurite growth in control and uPAR-deficient cells, but not in uPAR-overexpressing cells. Altogether these data implicate uPAR as an important regulator of EGFR and ERK1/2 signaling, representing a novel mechanism which implicates urokinase system in neuroblastoma cell survival and differentiation.
Collapse
Affiliation(s)
- K D Rysenkova
- Laboratory of Molecular Endocrinology, Institute of Experimental Cardiology, Federal State Budgetary Organization National Cardiology Research Center Ministry of Health of the Russian Federation, Moscow, Russia; Laboratory of Gene and Cell Technologies, Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - P S Klimovich
- Laboratory of Molecular Endocrinology, Institute of Experimental Cardiology, Federal State Budgetary Organization National Cardiology Research Center Ministry of Health of the Russian Federation, Moscow, Russia
| | - A A Shmakova
- Laboratory of Molecular Endocrinology, Institute of Experimental Cardiology, Federal State Budgetary Organization National Cardiology Research Center Ministry of Health of the Russian Federation, Moscow, Russia
| | - M N Karagyaur
- Institute of Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, Moscow, Russian Federation
| | - K A Ivanova
- Laboratory of Gene and Cell Technologies, Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - N A Aleksandrushkina
- Laboratory of Gene and Cell Technologies, Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia; Institute of Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, Moscow, Russian Federation
| | - V A Tkachuk
- Laboratory of Molecular Endocrinology, Institute of Experimental Cardiology, Federal State Budgetary Organization National Cardiology Research Center Ministry of Health of the Russian Federation, Moscow, Russia; Laboratory of Gene and Cell Technologies, Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - K A Rubina
- Laboratory of Morphogenesis and Tissue Reparation, Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia.
| | - E V Semina
- Laboratory of Molecular Endocrinology, Institute of Experimental Cardiology, Federal State Budgetary Organization National Cardiology Research Center Ministry of Health of the Russian Federation, Moscow, Russia; Laboratory of Gene and Cell Technologies, Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
9
|
Sharma A, Jha NK, Dahiya K, Singh VK, Chaurasiya K, Jha AN, Jha SK, Mishra PC, Dholpuria S, Astya R, Nand P, Kumar A, Ruokolainen J, Kesari KK. Nanoparticulate RNA delivery systems in cancer. Cancer Rep (Hoboken) 2020; 3:e1271. [PMID: 32729987 DOI: 10.1002/cnr2.1271] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 06/26/2020] [Accepted: 06/29/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Drug delivery system is a common practice in cancer treatment. RNA interference-mediated post-transcriptional gene silencing holds promise as an approach to knockdown in the expression of target genes responsible for cancer cell growth and metastasis. RNA interference (RNAi) can be achieved by delivering small interfering RNA (siRNA) and short hairpin RNA (shRNA) to target cells. Since neither interfering RNAs can be delivered in naked form due to poor stability, an efficient delivery system is required that protects, guides, and delivers the siRNA and shRNA to target cells as part of cancer therapy (chemotherapy). RECENT FINDINGS In this review, a discussion is presented about the different types of drug delivery system used to deliver siRNA and shRNA, together with an overview of the potential benefits associated with this sophisticated biomolecular therapy. Improved understanding of the different approaches used in nanoparticle (NP) fabrication, along with an enhanced appreciation of the biochemical properties of siRNA/shRNA, will assist in developing improved drug delivery strategies in basic and clinical research. CONCLUSION These novel delivery techniques are able to solve the problems that form an inevitable part of delivering genes in more efficient manner and as part of more effective treatment protocols. The present review concludes that the nanoparticulate RNA delivery system has great possibility for cancer treatment along with several other proposed methods. Several NPs or nanocarriers are already in use, but the methods proposed here could fulfill the missing gap in cancer research. It is the future technology, which unravels the mystery of resolving genomic diseases that is, especially genomic instability and its signaling cascades.
Collapse
Affiliation(s)
- Ankur Sharma
- Department of Life Science, School of Basic Science & Research, Sharda University, Greater Noida, India
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering & Technology, Sharda University, Greater Noida, India
| | - Kajal Dahiya
- Department of Life Science, School of Basic Science & Research, Sharda University, Greater Noida, India
| | - Vivek Kumar Singh
- Department of Biotechnology, School of Engineering & Technology, Sharda University, Greater Noida, India
| | - Kundan Chaurasiya
- Department of Biotechnology, School of Engineering & Technology, Sharda University, Greater Noida, India
| | - Aditya Narayan Jha
- Department of Biotechnology, School of Engineering & Technology, Sharda University, Greater Noida, India
| | - Saurabh Kumar Jha
- Department of Biotechnology, School of Engineering & Technology, Sharda University, Greater Noida, India
| | - Prabhu Chandra Mishra
- Department of Regenerative Medicine & Cellular Therapy, StemMax Research & Therapeutics Pvt Ltd., New Delhi, India
| | - Sunny Dholpuria
- Department of Life Science, School of Basic Science & Research, Sharda University, Greater Noida, India
| | - Rani Astya
- Department of Biotechnology, School of Engineering & Technology, Sharda University, Greater Noida, India
| | - Parma Nand
- Department of Biotechnology, School of Engineering & Technology, Sharda University, Greater Noida, India
| | - Amit Kumar
- Department of Zoology, Ram Krishna College, Lalit Narayan Mithila University, Darbhanga, India
| | | | | |
Collapse
|
10
|
Hsu FT, Chiang I, Wang W. Induction of apoptosis through extrinsic/intrinsic pathways and suppression of ERK/NF-κB signalling participate in anti-glioblastoma of imipramine. J Cell Mol Med 2020; 24:3982-4000. [PMID: 32149465 PMCID: PMC7171418 DOI: 10.1111/jcmm.15022] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Revised: 11/27/2019] [Accepted: 01/06/2020] [Indexed: 12/11/2022] Open
Abstract
Glioblastomas are the most aggressive type of brain tumour, with poor prognosis even after standard treatment such as surgical resection, temozolomide and radiation therapy. The overexpression of the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) in glioblastomas is recognized as an important treatment target. Thus, an urgent need regarding glioblastomas is the development of a new, suitable agent that may show potential for the inhibition of extracellular signal-regulated kinase (ERK)/NF-κB-mediated glioblastoma progression. Imipramine, a tricyclic antidepressant, has anti-inflammatory actions against inflamed glial cells; additionally, imipramine can induce glioblastoma toxicity via the activation of autophagy. However, whether imipramine can suppress glioblastoma progression via the induction of apoptosis and blockage of ERK/NF-κB signalling remains unclear. The main purpose of this study was to investigate the effects of imipramine on apoptotic signalling and ERK/NF-κB-mediated glioblastoma progression by using cell proliferation (3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide [MTT] assay), flow cytometry, Western blotting, and cell invasion/migration assay analysis in vitro. The ERK and NF-κB inhibitory capacity of imipramine is detected by NF-κB reporter gene assay and Western blotting. Additionally, a glioblastoma-bearing animal model was used to validate the therapeutic efficacy and general toxicity of imipramine. Our results demonstrated that imipramine successfully triggered apoptosis through extrinsic/intrinsic pathways and suppressed the invasion/migration ability of glioblastoma cells. Furthermore, imipramine effectively suppressed glioblastoma progression in vivo via the inhibition of the ERK/NF-κB pathway. In summary, imipramine is a potential anti-glioblastoma drug which induces apoptosis and has the capacity to inhibit ERK/NF-κB signalling.
Collapse
Affiliation(s)
- Fei-Ting Hsu
- Department of Biological Science and TechnologyChina Medical UniversityTaichungTaiwan
| | - I‐Tsang Chiang
- Department of Radiation OncologyShow Chwan Memorial HospitalChanghuaTaiwan
- Department of Radiation OncologyChang Bing Show Chwan Memorial HospitalLukangTaiwan
- Department of Medical Imaging and Radiological SciencesCentral Taiwan University of Science and TechnologyTaichungTaiwan
| | - Wei‐Shu Wang
- Department of MedicineNational Yang‐Ming University HospitalYilanTaiwan
- School of MedicineNational Yang‐Ming UniversityTaipeiTaiwan
| |
Collapse
|
11
|
Zhang D, Yang Y, Liang C, Liu J, Wang H, Liu S, Yan Q. poFUT1 promotes uterine angiogenesis and vascular remodeling via enhancing the O-fucosylation on uPA. Cell Death Dis 2019; 10:775. [PMID: 31601791 PMCID: PMC6787057 DOI: 10.1038/s41419-019-2005-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 09/12/2019] [Accepted: 09/17/2019] [Indexed: 12/17/2022]
Abstract
Uterine angiogenesis and vascular remodeling play critical roles in determing the normal menstrual cycle and successful pregnancy. Poor uterine angiogenesis usually results in pregnancy failure. Protein O-fucosyltransferase 1 (poFUT1) is the key enzyme responsible for O-fucosylated glycan biosynthesis on glycoproteins. However, the dynamic expression and regulation of poFUT1 on the uterine angiogenesis and vascular remodeling remain unknown. Here, we showed that the enlargement of the vascular lumen in the secretory phase was greater than that in the proliferative phase of the uterine endometrium during menstrual cycle; whereas there was a narrower vessel lumen and fewer blood vessels in the decidua from miscarriage patients than in that from healthy pregnancy women. Additionally, the expression of poFUT1 was increased in the uterine endometrium during the secretory phase compared with that in the proliferation phase, and its expression was decreased in the uterus of miscarriage patients compared with that of the healthy pregnancy women. Using hESCs and a mouse model, we demonstrated that poFUT1 increased the O-fucosylation on uPA, and activated of the RhoA signaling pathway, thus facilitating uterine angiogenesis and vascular remodeling. We also provide evidence that poFUT1 promotes hESCs angiogenesis by the decreased stemness of hESCs. These findings reveal a new insight into the uterine angiogenesis and vascular remodeling. The study suggests that poFUT1 could be seen as a novel potential diagnostic and therapeutic target for miscarriage.
Collapse
Affiliation(s)
- Dandan Zhang
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Liaoning Provincial Core Lab of Glycobiology and Glycoengineering, 116044, Dalian, China
| | - Yu Yang
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Liaoning Provincial Core Lab of Glycobiology and Glycoengineering, 116044, Dalian, China
| | - Caixia Liang
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Liaoning Provincial Core Lab of Glycobiology and Glycoengineering, 116044, Dalian, China
| | - Jianwei Liu
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Liaoning Provincial Core Lab of Glycobiology and Glycoengineering, 116044, Dalian, China
| | - Hao Wang
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Liaoning Provincial Core Lab of Glycobiology and Glycoengineering, 116044, Dalian, China
| | - Shuai Liu
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Liaoning Provincial Core Lab of Glycobiology and Glycoengineering, 116044, Dalian, China.
| | - Qiu Yan
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Liaoning Provincial Core Lab of Glycobiology and Glycoengineering, 116044, Dalian, China.
| |
Collapse
|
12
|
Dimitrova I, Tacheva T, Mindov I, Petrov B, Aleksandrova E, Valkanov S, Gulubova M, Vlaykova T. Serum levels of MMP-7 in primary brain cancers and brain metastases. BIOTECHNOL BIOTEC EQ 2019. [DOI: 10.1080/13102818.2019.1626282] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Affiliation(s)
- Ivelina Dimitrova
- Department of Surgery and Neurosurgery, Medical Faculty, Trakia University, Stara Zagora, Bulgaria
| | - Tanya Tacheva
- Department of Chemistry and Biochemistry, Medical Faculty, Trakia University, Stara Zagora, Bulgaria
| | - Ivan Mindov
- Department of Surgery and Neurosurgery, Medical Faculty, Trakia University, Stara Zagora, Bulgaria
| | - Bozhidar Petrov
- Department of Surgery and Neurosurgery, Medical Faculty, Trakia University, Stara Zagora, Bulgaria
| | - Elina Aleksandrova
- Department of General and Clinical Pathology, Medical Faculty, Trakia University, Stara Zagora, Bulgaria
| | - Stefan Valkanov
- Department of Surgery and Neurosurgery, Medical Faculty, Trakia University, Stara Zagora, Bulgaria
| | - Maya Gulubova
- Department of General and Clinical Pathology, Medical Faculty, Trakia University, Stara Zagora, Bulgaria
| | - Tatyana Vlaykova
- Department of Chemistry and Biochemistry, Medical Faculty, Trakia University, Stara Zagora, Bulgaria
| |
Collapse
|
13
|
Rysenkova KD, Semina EV, Karagyaur MN, Shmakova AA, Dyikanov DT, Vasiluev PA, Rubtsov YP, Rubina KA, Tkachuk VA. CRISPR/Cas9 nickase mediated targeting of urokinase receptor gene inhibits neuroblastoma cell proliferation. Oncotarget 2018; 9:29414-29430. [PMID: 30034627 PMCID: PMC6047682 DOI: 10.18632/oncotarget.25647] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 06/05/2018] [Indexed: 12/26/2022] Open
Abstract
Neuroblastoma is a tumor arising from pluripotent sympathoadrenal precursor cells of neural cell origin. Neuroblastoma is one of the most aggressive childhood tumors with highly invasive and metastatic potential. The increased expression of urokinase and its receptor is often associated with a negative prognosis in neuroblastoma patients. We have shown that targeting of the Plaur gene in mouse neuroblastoma Neuro 2A cells by CRISPR/Cas9n results in ~60% decrease in cell proliferation (p<0.05), reduction in the number of Ki-67 positive cells, caspase 3 activation and PARP-1 cleavage. Knockout of uPAR leads to downregulation of mRNA encoding full-length TrkC receptor, which is involved in p38MAPK and Akt signalling pathways. This finding provides a rationale to study a role of uPAR in neuroblastoma progression, since uPAR could be considered a potential therapeutic target in neuroblastoma treatment.
Collapse
Affiliation(s)
- Karina D Rysenkova
- Lomonosov Moscow State University, Faculty of Medicine, Laboratory of Gene and Cell Technologies, 119991, Moscow, Russian Federation
| | - Ekaterina V Semina
- Lomonosov Moscow State University, Faculty of Medicine, Laboratory of Gene and Cell Technologies, 119991, Moscow, Russian Federation.,Federal State Budgetary Organization National Cardiology Research Center Ministry of Health of the Russian Federation, Institute of Experimental Cardiology, 121552, Moscow, Russian Federation
| | - Maxim N Karagyaur
- Institute of Regenerative Medicine, Lomonosov Moscow State University, 119991, Moscow, Russian Federation
| | - Anna A Shmakova
- Lomonosov Moscow State University, Faculty of Medicine, Laboratory of Gene and Cell Technologies, 119991, Moscow, Russian Federation
| | - Daniyar T Dyikanov
- Lomonosov Moscow State University, Faculty of Medicine, Laboratory of Gene and Cell Technologies, 119991, Moscow, Russian Federation
| | - Petr A Vasiluev
- Lomonosov Moscow State University, Faculty of Medicine, Laboratory of Gene and Cell Technologies, 119991, Moscow, Russian Federation
| | - Yury P Rubtsov
- Lomonosov Moscow State University, Faculty of Medicine, Laboratory of Gene and Cell Technologies, 119991, Moscow, Russian Federation.,Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997, Moscow, Russian Federation
| | - Kseniya A Rubina
- Lomonosov Moscow State University, Faculty of Medicine, Laboratory of Gene and Cell Technologies, 119991, Moscow, Russian Federation
| | - Vsevolod A Tkachuk
- Lomonosov Moscow State University, Faculty of Medicine, Laboratory of Gene and Cell Technologies, 119991, Moscow, Russian Federation.,Federal State Budgetary Organization National Cardiology Research Center Ministry of Health of the Russian Federation, Institute of Experimental Cardiology, 121552, Moscow, Russian Federation
| |
Collapse
|
14
|
Mahmood N, Mihalcioiu C, Rabbani SA. Multifaceted Role of the Urokinase-Type Plasminogen Activator (uPA) and Its Receptor (uPAR): Diagnostic, Prognostic, and Therapeutic Applications. Front Oncol 2018; 8:24. [PMID: 29484286 PMCID: PMC5816037 DOI: 10.3389/fonc.2018.00024] [Citation(s) in RCA: 283] [Impact Index Per Article: 47.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 01/24/2018] [Indexed: 01/01/2023] Open
Abstract
The plasminogen activator (PA) system is an extracellular proteolytic enzyme system associated with various physiological and pathophysiological processes. A large body of evidence support that among the various components of the PA system, urokinase-type plasminogen activator (uPA), its receptor (uPAR), and plasminogen activator inhibitor-1 and -2 (PAI-1 and PAI-2) play a major role in tumor progression and metastasis. The binding of uPA with uPAR is instrumental for the activation of plasminogen to plasmin, which in turn initiates a series of proteolytic cascade to degrade the components of the extracellular matrix, and thereby, cause tumor cell migration from the primary site of origin to a distant secondary organ. The components of the PA system show altered expression patterns in several common malignancies, which have identified them as ideal diagnostic, prognostic, and therapeutic targets to reduce cancer-associated morbidity and mortality. This review summarizes the various components of the PA system and focuses on the role of uPA-uPAR in different biological processes especially in the context of malignancy. We also discuss the current state of knowledge of uPA-uPAR-targeted diagnostic and therapeutic strategies for various malignancies.
Collapse
Affiliation(s)
- Niaz Mahmood
- Department of Medicine, McGill University Health Centre, Montreal, QC, Canada
| | - Catalin Mihalcioiu
- Department of Oncology, McGill University Health Centre, Montreal, QC, Canada
| | - Shafaat A. Rabbani
- Department of Medicine, McGill University Health Centre, Montreal, QC, Canada
- Department of Oncology, McGill University Health Centre, Montreal, QC, Canada
| |
Collapse
|
15
|
Bibes R, Gobron S, Vincent F, Mélin C, Vedrenne N, Perraud A, Labrousse F, Jauberteau MO, Lalloué F. SCO-spondin oligopeptide inhibits angiogenesis in glioblastoma. Oncotarget 2017; 8:85969-85983. [PMID: 29156770 PMCID: PMC5689660 DOI: 10.18632/oncotarget.20837] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 08/04/2017] [Indexed: 11/25/2022] Open
Abstract
Angiogenesis plays a critical role in glioblastoma growth and progression. We therefore aimed at evaluating the anti-angiogenic properties of an oligopeptide originating from SCO-spondin (NX) on a model of human glioblastoma. To this end, we studied the impact of NX treatment on human brain endothelial cells (HBMECs) alone or co-cultured with glioblastoma cells (U87-MG) on apoptosis, proliferation, migration and release of angiogenic factors. We further investigated the anti-angiogenic potential of NX on human glioblastoma cells grown on chorio-allantoic membrane (CAM) or in glioblastoma xenografts. The results of our experiments showed that NX treatment impaired the microvascular network and induced a decrease in cell proliferation, vascularization and tumor growth in the CAM model as well as in xenotransplants. Interestingly, our in vitro experiments showed that NX impairs HBMECs migration but also regulates the release of angiogenic factors from U87-MG. These results are confirmed by the profiling of NX-treated U87-MG grown on CAM that highlighted modifications of several genes involved in angiogenesis. In conclusion, NX inhibits tumorigenesis by impairing the ability of glioblastoma cells to induce angiogenesis and by inhibiting endothelial cell migration. This molecule might therefore be an interesting candidate for future cancer therapies.
Collapse
Affiliation(s)
- Romain Bibes
- EA3842 Cellular Homeostasis and Diseases, University of Limoges, Faculty of Medicine, 87025 Limoges Cedex, France
| | - Stéphane Gobron
- Neuronax, Biopôle Clermont-Limagne, 63360 Saint-Beauzire, France
| | - François Vincent
- EA3842 Cellular Homeostasis and Diseases, University of Limoges, Faculty of Medicine, 87025 Limoges Cedex, France.,Limoges University Hospital, Department of Physiological Functional Investigation, 87042 Limoges Cedex, France
| | - Carole Mélin
- EA3842 Cellular Homeostasis and Diseases, University of Limoges, Faculty of Medicine, 87025 Limoges Cedex, France
| | - Nicolas Vedrenne
- EA3842 Cellular Homeostasis and Diseases, University of Limoges, Faculty of Medicine, 87025 Limoges Cedex, France
| | - Aurélie Perraud
- EA3842 Cellular Homeostasis and Diseases, University of Limoges, Faculty of Medicine, 87025 Limoges Cedex, France.,Limoges University Hospital, Department of Digestive Surgery, 87042 Limoges Cedex, France
| | - Francois Labrousse
- EA3842 Cellular Homeostasis and Diseases, University of Limoges, Faculty of Medicine, 87025 Limoges Cedex, France.,Limoges University Hospital, Department of Pathology, 87042 Limoges Cedex, France
| | - Marie-Odile Jauberteau
- EA3842 Cellular Homeostasis and Diseases, University of Limoges, Faculty of Medicine, 87025 Limoges Cedex, France.,Limoges University Hospital, Department of Immunology, 87042 Limoges Cedex, France
| | - Fabrice Lalloué
- EA3842 Cellular Homeostasis and Diseases, University of Limoges, Faculty of Medicine, 87025 Limoges Cedex, France
| |
Collapse
|
16
|
Woolf EC, Curley KL, Liu Q, Turner GH, Charlton JA, Preul MC, Scheck AC. The Ketogenic Diet Alters the Hypoxic Response and Affects Expression of Proteins Associated with Angiogenesis, Invasive Potential and Vascular Permeability in a Mouse Glioma Model. PLoS One 2015; 10:e0130357. [PMID: 26083629 PMCID: PMC4470583 DOI: 10.1371/journal.pone.0130357] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 05/18/2015] [Indexed: 01/09/2023] Open
Abstract
Background The successful treatment of malignant gliomas remains a challenge despite the current standard of care, which consists of surgery, radiation and temozolomide. Advances in the survival of brain cancer patients require the design of new therapeutic approaches that take advantage of common phenotypes such as the altered metabolism found in cancer cells. It has therefore been postulated that the high-fat, low-carbohydrate, adequate protein ketogenic diet (KD) may be useful in the treatment of brain tumors. We have demonstrated that the KD enhances survival and potentiates standard therapy in a mouse model of malignant glioma, yet the mechanisms are not fully understood. Methods To explore the effects of the KD on various aspects of tumor growth and progression, we used the immunocompetent, syngeneic GL261-Luc2 mouse model of malignant glioma. Results Tumors from animals maintained on KD showed reduced expression of the hypoxia marker carbonic anhydrase 9, hypoxia inducible factor 1-alpha, and decreased activation of nuclear factor kappa B. Additionally, tumors from animals maintained on KD had reduced tumor microvasculature and decreased expression of vascular endothelial growth factor receptor 2, matrix metalloproteinase-2 and vimentin. Peritumoral edema was significantly reduced in animals fed the KD and protein analyses showed altered expression of zona occludens-1 and aquaporin-4. Conclusions The KD directly or indirectly alters the expression of several proteins involved in malignant progression and may be a useful tool for the treatment of gliomas.
Collapse
MESH Headings
- Animals
- Aquaporin 4/genetics
- Aquaporin 4/metabolism
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Brain Neoplasms/blood supply
- Brain Neoplasms/diet therapy
- Brain Neoplasms/metabolism
- Brain Neoplasms/pathology
- Carbonic Anhydrase IX
- Carbonic Anhydrases/genetics
- Carbonic Anhydrases/metabolism
- Cell Membrane Permeability
- Diet, Ketogenic
- Disease Models, Animal
- Female
- Glioma/blood supply
- Glioma/diet therapy
- Glioma/metabolism
- Glioma/pathology
- Hypoxia/diet therapy
- Hypoxia/metabolism
- Hypoxia/pathology
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Immunoenzyme Techniques
- Matrix Metalloproteinase 2/genetics
- Matrix Metalloproteinase 2/metabolism
- Mice
- Mice, Inbred C57BL
- NF-kappa B/genetics
- NF-kappa B/metabolism
- Neoplasm Invasiveness
- Neovascularization, Pathologic/diet therapy
- Neovascularization, Pathologic/metabolism
- Neovascularization, Pathologic/pathology
- RNA, Messenger/genetics
- Real-Time Polymerase Chain Reaction
- Reverse Transcriptase Polymerase Chain Reaction
- Vascular Endothelial Growth Factor A/genetics
- Vascular Endothelial Growth Factor A/metabolism
- Zonula Occludens-1 Protein/genetics
- Zonula Occludens-1 Protein/metabolism
Collapse
Affiliation(s)
- Eric C. Woolf
- Neuro-Oncology Research, Barrow Brain Tumor Research Center, Barrow Neurological Institute dba St. Joseph’s Hospital and Medical Center, Phoenix, Arizona, 85013, United States of America
- School of Life Sciences, Arizona State University, Tempe, Arizona, 85281, United States of America
| | - Kara L. Curley
- Neuro-Oncology Research, Barrow Brain Tumor Research Center, Barrow Neurological Institute dba St. Joseph’s Hospital and Medical Center, Phoenix, Arizona, 85013, United States of America
| | - Qingwei Liu
- BNI-ASU Center for Preclinical Imaging, Barrow Neurological Institute dba St. Joseph’s Hospital and Medical Center, Phoenix, Arizona, 85013, United States of America
| | - Gregory H. Turner
- BNI-ASU Center for Preclinical Imaging, Barrow Neurological Institute dba St. Joseph’s Hospital and Medical Center, Phoenix, Arizona, 85013, United States of America
| | - Julie A. Charlton
- Neuro-Oncology Research, Barrow Brain Tumor Research Center, Barrow Neurological Institute dba St. Joseph’s Hospital and Medical Center, Phoenix, Arizona, 85013, United States of America
| | - Mark C. Preul
- Neurosurgery Research, Barrow Neurological Institute dba St. Joseph's Hospital and Medical Center, Phoenix, Arizona, 85013, United States of America
| | - Adrienne C. Scheck
- Neuro-Oncology Research, Barrow Brain Tumor Research Center, Barrow Neurological Institute dba St. Joseph’s Hospital and Medical Center, Phoenix, Arizona, 85013, United States of America
- School of Life Sciences, Arizona State University, Tempe, Arizona, 85281, United States of America
- Neurosurgery Research, Barrow Neurological Institute dba St. Joseph's Hospital and Medical Center, Phoenix, Arizona, 85013, United States of America
- * E-mail:
| |
Collapse
|
17
|
Identification of Glioblastoma Phosphotyrosine-Containing Proteins with Two-Dimensional Western Blotting and Tandem Mass Spectrometry. BIOMED RESEARCH INTERNATIONAL 2015; 2015:134050. [PMID: 26090378 PMCID: PMC4450212 DOI: 10.1155/2015/134050] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Revised: 08/25/2014] [Accepted: 08/25/2014] [Indexed: 12/24/2022]
Abstract
To investigate the presence of, and the potential biological roles of, protein tyrosine phosphorylation in the glioblastoma pathogenesis, two-dimensional gel electrophoresis- (2DGE-) based Western blotting coupled with liquid chromatography-electrospray ionization-tandem mass spectrometry (LC-ESI-MS/MS) analysis was used to detect and identify the phosphotyrosine immunoreaction-positive proteins in a glioblastoma tissue. MS/MS and Mascot analyses were used to determine the phosphotyrosine sites of each phosphopeptide. Protein domain and motif analysis and systems pathway analysis were used to determine the protein domains/motifs that contained phosphotyrosine residue and signal pathway networks to clarify the potential biological functions of protein tyrosine phosphorylation. A total of 24 phosphotyrosine-containing proteins were identified. Each phosphotyrosine-containing protein contained at least one tyrosine kinase phosphorylation motif and a certain structural and functional domains. Those phosphotyrosine-containing proteins were involved in the multiple signal pathway systems such as oxidative stress, stress response, and cell migration. Those data show 2DGE-based Western blotting, MS/MS, and bioinformatics are a set of effective approaches to detect and identify glioblastoma tyrosine-phosphorylated proteome and to effectively rationalize the biological roles of tyrosine phosphorylation in the glioblastoma biological systems. It provides novel insights regarding tyrosine phosphorylation and its potential role in the molecular mechanism of a glioblastoma.
Collapse
|
18
|
TONG YU, YUE JUN, MAO MENG, LIU QINGQING, ZHOU JING, YANG JIYUN. Recombinant nematode anticoagulant protein c2 inhibits cell invasion by decreasing uPA expression in NSCLC cells. Oncol Rep 2015; 33:1815-22. [DOI: 10.3892/or.2015.3795] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 01/23/2015] [Indexed: 11/06/2022] Open
|
19
|
CHEN HONG, ZOU YANG, YANG HONG, WANG JINGJING, PAN HONG. Downregulation of FoxM1 inhibits proliferation, invasion and angiogenesis of HeLa cells in vitro and in vivo. Int J Oncol 2014; 45:2355-64. [DOI: 10.3892/ijo.2014.2645] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 08/14/2014] [Indexed: 11/05/2022] Open
|
20
|
Li D, Wei P, Peng Z, Huang C, Tang H, Jia Z, Cui J, Le X, Huang S, Xie K. The critical role of dysregulated FOXM1-PLAUR signaling in human colon cancer progression and metastasis. Clin Cancer Res 2012; 19:62-72. [PMID: 23136192 DOI: 10.1158/1078-0432.ccr-12-1588] [Citation(s) in RCA: 90] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
PURPOSE The mammalian Forkhead Box (Fox) transcription factor FOXM1 is implicated in tumorigenesis including mouse intestinal cancer. However, the clinical significance of FOXM1 signaling in human colorectal cancer pathogenesis remains unknown. EXPERIMENTAL DESIGN We investigated FOXM1 expression in 203 cases of primary colon cancer and matched normal colon tissue specimens and explored the underlying mechanisms of altered FOXM1 expression and the impact of this altered expression on colon cancer growth and metastasis using in vitro and animal models of colon cancer. RESULTS We found weak expression of FOXM1 protein in the colon mucosa, whereas we observed strong FOXM1 expression in tumor-cell nuclei of colon cancer and lymph node metastases. A Cox proportional hazards model revealed that FOXM1 expression was an independent prognostic factor in multivariate analysis. Experimentally, overexpression of FOXM1 by gene transfer significantly promoted the growth and metastasis of colon cancer cells in orthotopic mouse models, whereas knockdown of FOXM1 expression by siRNA did the opposite. Promotion of colon tumorigenesis by FOXM1 directly and significantly correlated with activation of urokinase-type plasminogen activator receptor (PLAUR) expression and elevation of invasion and metastasis. CONCLUSIONS Given the importance of FOXM1 in regulation of the expression of genes key to cancer biology, dysregulated expression and activation of FOXM1 may play important roles in colon cancer progression and metastasis.
Collapse
Affiliation(s)
- Dawei Li
- Department of General Surgery, Shanghai Jiaotong University Affiliated First People's Hospital, Shanghai, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Breuss JM, Uhrin P. VEGF-initiated angiogenesis and the uPA/uPAR system. Cell Adh Migr 2012; 6:535-615. [PMID: 23076133 DOI: 10.4161/cam.22243] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Angiogenesis involves a series of tightly regulated cellular processes initiated primarily by the vascular endothelial growth factor (VEGF). The urokinase-type plasminogen activator system, consisting of the urokinase-type plasminogen activator (uPA), its cellular receptor uPAR and its inhibitor PAI-1, participates in the realization of these VEGF-induced processes by activating pericellular proteolysis, increasing vascular permeability and by supporting endothelial cell proliferation and migration.
Collapse
Affiliation(s)
- Johannes M Breuss
- Institute of Vascular Biology and Thrombosis Research, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria.
| | | |
Collapse
|
22
|
Differential roles of uPAR in peritoneal ovarian carcinomatosis. Neoplasia 2012; 14:259-70. [PMID: 22577342 DOI: 10.1593/neo.12442] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2012] [Revised: 03/19/2012] [Accepted: 03/29/2012] [Indexed: 01/10/2023] Open
Abstract
Epithelial ovarian cancer is the fourth leading cause of death from gynecologic malignancies in the United States. Most cases are diagnosed at late stages, with the solid tumor masses growing as peritoneal implants, or floating within the ascitic fluid (peritoneal ovarian carcinomatosis). Despite aggressive surgical "debulking," recurrence of recalcitrant disease is frequent with poor patient survival. Efforts to improve survival rates are hindered by lack of biomarkers that can detect and effectively treat ovarian cancer in its early stages. Urokinase plasminogen activator receptor (uPAR) is a multifunctional receptor involved in a myriad of tumor cell processes. However, the role of host uPAR in ovarian cancer is still elusive. To define the potential proinflammatory role of uPAR in ovarian cancer, first, using a syngeneic murine model in uPAR(-/-) mice, we found that ablation of uPAR restrained tumor take and peritoneal implants and prolonged the survival of uPAR(-/-) mice compared with their uPAR(+/+) counterparts. Ascitic fluid accumulation was significantly decreased in uPAR(-/-) mice with decreased macrophage infiltration. Second, in vitro mechanistic studies revealed that host uPAR is involved in the multiple steps of peritoneal metastatic cascade. Third, we evaluated the prognostic utility of tumor and stromal uPAR in human ovarian cancer tissue microarray. In summary, our studies indicated that uPAR plays a significant role in ovarian cancer cell-stromal crosstalk and contributes to increased vascular permeability and inflammatory ovarian cancer microenvironment. This provides a rationale for targeting the uPAR with either specific neutralizing antibodies or targeting its downstream inflammatory effectors in patients with ovarian cancer.
Collapse
|
23
|
Raghu H, Nalla AK, Gondi CS, Gujrati M, Dinh DH, Rao JS. uPA and uPAR shRNA inhibit angiogenesis via enhanced secretion of SVEGFR1 independent of GM-CSF but dependent on TIMP-1 in endothelial and glioblastoma cells. Mol Oncol 2011; 6:33-47. [PMID: 22177802 DOI: 10.1016/j.molonc.2011.11.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2011] [Revised: 11/22/2011] [Accepted: 11/23/2011] [Indexed: 12/31/2022] Open
Abstract
The uPA/uPAR system is known to play a critical role in angiogenesis of glioblastoma. Previously, we have shown that shRNA against uPA and uPAR attenuates angiogenesis by blocking nuclear translocation of angiogenin, inhibition of angiopoietin/Tie2 signaling, and regulating several other pro-angiogenic, angiostatic and anti-angiogenic molecules. Further analysis revealed that GM-CSF, a pleiotropic cytokine, was significantly inhibited in U87MG and 4910 co-cultures with endothelial cells transfected with shRNA against uPA and uPAR. The role of the uPA/uPAR system in this process is not completely understood. Analysis of tumor conditioned medium of U87MG, 4910 and HMECs transfected with shRNA against uPA or uPAR alone or in combination (pU2) revealed inhibition of GM-CSF-enhanced secretion of SVEGFR1 as shown by Western blotting and ELISA. Moreover, phosphorylation of JAK2 and STAT5, the downstream effectors of GM-CSF signaling, was also inhibited in all three cell lines. Phosphorylation at Tyr 166 position of the GM-CSFRβ subunit, the signal activating subunit of the GM-CSF receptor, was inhibited in HMEC, U87MG and 4910 cells. Further analysis revealed that shRNA against uPA and/or uPAR increased secretion of TIMP-1, which is known to enhance SVEGFR1 secretion in endothelial cells. Moreover, addition of purified uPA (with and without GM-CSF) activated JAK2/STAT5 signaling in HMEC. Exogenous addition of SVEGFR1 to pU2 tumor conditioned medium enhanced inhibition of VEGF-induced endothelial capillary tube formation as assessed by an in vitro angiogenesis assay. To determine the significance of these events in vivo, nude mice with pre-established tumors treated with shRNA against uPA and/or uPAR showed decreased levels of GM-CSF and increased levels of SVEGFR1 and TIMP-1 when compared with controls. Enhanced secretion of SVEGFR1 by puPA, puPAR and pU2 in endothelial and GBM cells was mediated indirectly by MMP-7 and augmented by ectodomain shedding of VEGFr1 by tyrosine phosphorylation at the 1213 position. Taken together, these results suggest that the uPA/uPAR system could prove beneficial as an indirect target for inhibition of angiogenesis in glioblastoma.
Collapse
Affiliation(s)
- Hari Raghu
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine, Peoria, IL 61605, USA
| | | | | | | | | | | |
Collapse
|