1
|
Xiao Y, Chen X, Li W, Li X, Zhou W. Impact of ACEI/ARB use on the survival of hypertensive patients with cancer: A meta‑analysis. Oncol Lett 2024; 28:534. [PMID: 39290956 PMCID: PMC11406588 DOI: 10.3892/ol.2024.14667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 07/01/2024] [Indexed: 09/19/2024] Open
Abstract
Angiotensin-converting enzyme inhibitors (ACEIs) and angiotensin receptor blockers (ARBs) are commonly used antihypertensive drugs. However, the impact that the use of ACEI and ARB drugs will have on the survival of patients with hypertension and cancer is still unclear. Therefore, the present study aimed to investigate the effects of ACEI and ARB use on the survival of patients with cancer. The Embase, PubMed and Web of Science databases were used to systematically analyze the survival of hypertensive patients with cancer treated with ACEIs or ARBs. Hazard ratios (HRs) and 95% confidence intervals (CIs) were calculated to evaluate the association between ACEI and ARB use and patient survival. The relationship between the survival of patients with certain types of cancer and ACEI and ARB use was evaluated using the calculated HRs. Patients with ovarian, pancreatic, prostate, hepatocellular, lung, esophageal, gastric, colon, nasopharyngeal, head and neck tumors, gallbladder and rectal cancers that used ACEI and ARB analogs had significantly increased survival times, except for patients with breast cancer (HR, 1.04; 95% CI, 0.90-1.19; P<0.01) and uroepithelial carcinoma (HR, 1.15; 95% CI, 0.69-1.94; P<0.01), who had significantly decreased survival times, when compared with patients who did not use these drugs. Analysis of the relationship between the use of ACEIs or ARBs alone or in combination on the overall survival of hypertensive patients with cancer demonstrated that the use of ACEIs alone (HR, 1.00; 95% CI, 0.93-1.08; P<0.01) did not have a significant effect on the survival of these patients. By contrast, the survival time was increased in hypertensive patients with cancer who used either ARBs alone (HR, 0.89; 95% CI, 0.84-0.94; P<0.01) or a combination of ACEIs and ARBs (HR, 0.84; 95% CI, 0.78-0.91; P<0.01). The present meta-analysis demonstrated the potential effects of ACEI and ARB use on the overall survival of patients with cancer. Therefore, investigation of the underlying mechanisms of action of ACEIs and ARBs, as well as the identification of specific groups of patients who may benefit from these interventions, could potentially lead to novel therapeutic options and improve the prognosis of patients with cancer in the future.
Collapse
Affiliation(s)
- Yao Xiao
- Department of General Surgery, The Second Clinical Medical College, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, P.R. China
| | - Xinlong Chen
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shanxi 710061, P.R. China
| | - Wancheng Li
- Department of General Surgery, The Second Clinical Medical College, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, P.R. China
| | - Xin Li
- Department of General Surgery, The Second Clinical Medical College, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, P.R. China
| | - Wence Zhou
- Department of General Surgery, The Second Clinical Medical College, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, P.R. China
| |
Collapse
|
2
|
An overview of kinin mediated events in cancer progression and therapeutic applications. Biochim Biophys Acta Rev Cancer 2022; 1877:188807. [PMID: 36167271 DOI: 10.1016/j.bbcan.2022.188807] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/12/2022] [Accepted: 09/21/2022] [Indexed: 11/22/2022]
Abstract
Kinins are bioactive peptides generated in the inflammatory milieu of the tissue microenvironment, which is involved in cancer progression and inflammatory response. Kinins signals through activation of two G-protein coupled receptors; inducible Bradykinin Receptor B1 (B1R) and constitutive receptor B2 (B2R). Activation of kinin receptors and its cross-talk with receptor tyrosine kinases activates multiple signaling pathways, including ERK/MAPK, PI3K, PKC, and p38 pathways regulating cancer hallmarks. Perturbations of the kinin-mediated events are implicated in various aspects of cancer invasion, matrix remodeling, and metastasis. In the tumor microenvironment, kinins initiate fibroblast activation, mesenchymal stem cell interactions, and recruitment of immune cells. Albeit the precise nature of kinin function in the metastasis and tumor microenvironment are not completely clear yet, several kinin receptor antagonists show anti-metastatic potential. Here, we showcase an overview of the complex biology of kinins and their role in cancer pathogenesis and therapeutic aspects.
Collapse
|
3
|
Zeman M, Skałba W, Szymański P, Hadasik G, Żaworonkow D, Walczak DA, Czarniecka A. Risk factors for long-term survival in patients with ypN+ M0 rectal cancer after radical anterior resection. BMC Gastroenterol 2022; 22:141. [PMID: 35346064 PMCID: PMC8961971 DOI: 10.1186/s12876-022-02226-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 03/21/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Regional lymph node metastases are the main adverse prognostic factor in patients with rectal cancer without distant metastases. There are discrepancies, however, regarding additional risk factors in the group of ypN + M0 patients. The purpose of the study was to assess clinical and pathological factors affecting long-term oncological outcomes in the group of ypN + M0 patients after radical rectal anterior resection.
Methods
112 patients with ypN + M0 rectal cancer after neoadjuvant therapy and radical anterior resection were subject to a retrospective analysis. The effect of potential factors on survival was assessed with the use of Kaplan–Meier curves together with a log-rank test and multiple factor Cox proportional hazards model.
Results
In the multiple factor Cox analysis, adverse factors affecting disease-free survival (DFS) were: the use of angiotensin-converting enzyme inhibitors (ACEIs) (hazard ratio HR: 3.11, 95% CI 1.01–9.56, p = 0.047), presence of perineural invasion (HR: 7.27, 95% CI 2.74–19.3, p < 0.001) and occurrence of postoperative complications (HR: 6.79, 95% CI 2.09–22.11, p = 0.001), while a positive factor was the negative lymph node (NLN) count > 7 (HR: 0.33, 95% CI 0.12–0.88, p = 0.026). In the disease-specific survival (DSS) analysis, an adverse factor was the use of ACEIs (HR: 4.275, 95% CI 1.44–12.694, p = 0.009), while a positive effect was caused by NLN > 5 (HR: 0.22, 95% CI 0.082–0.586, p = 0.002).
Conclusions
The use of ACEIs may have a negative effect on long-term treatment outcomes in patients with ypN + M0 rectal cancer. In this group of patients, the NLN count seems to be an important prognostic factor, as well.
Collapse
|
4
|
Jakwerth CA, Feuerherd M, Guerth FM, Oelsner M, Schellhammer L, Giglberger J, Pechtold L, Jerin C, Kugler L, Mogler C, Haller B, Erb A, Wollenberg B, Spinner CD, Buch T, Protzer U, Schmidt-Weber CB, Zissler UM, Chaker AM. Early reduction of SARS-CoV-2-replication in bronchial epithelium by kinin B 2 receptor antagonism. J Mol Med (Berl) 2022; 100:613-627. [PMID: 35247068 PMCID: PMC8897552 DOI: 10.1007/s00109-022-02182-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 02/07/2022] [Accepted: 02/15/2022] [Indexed: 12/14/2022]
Abstract
Abstract SARS-CoV-2 has evolved to enter the host via the ACE2 receptor which is part of the kinin-kallikrein pathway. This complex pathway is only poorly understood in context of immune regulation but critical to control infection. This study examines SARS-CoV-2-infection and epithelial mechanisms of the kinin-kallikrein-system at the kinin B2 receptor level in SARS-CoV-2-infection that is of direct translational relevance. From acute SARS-CoV-2-positive study participants and -negative controls, transcriptomes of nasal curettages were analyzed. Primary airway epithelial cells (NHBEs) were infected with SARS-CoV-2 and treated with the approved B2R-antagonist icatibant. SARS-CoV-2 RNA RT-qPCR, cytotoxicity assays, plaque assays, and transcriptome analyses were performed. The treatment effect was further studied in a murine airway inflammation model in vivo. Here, we report a broad and strong upregulation of kallikreins and the kinin B2 receptor (B2R) in the nasal mucosa of acutely symptomatic SARS-CoV-2-positive study participants. A B2R-antagonist impeded SARS-CoV-2 replication and spread in NHBEs, as determined in plaque assays on Vero-E6 cells. B2R-antagonism reduced the expression of SARS-CoV-2 entry receptor ACE2, G protein–coupled receptor signaling, and ion transport in vitro and in a murine airway inflammation in vivo model. In summary, this study provides evidence that treatment with B2R-antagonists protects airway epithelial cells from SARS-CoV-2 by inhibiting its replication and spread, through the reduction of ACE2 levels and the interference with several cellular signaling processes. Future clinical studies need to shed light on the airway protection potential of approved B2R-antagonists, like icatibant, in the treatment of early-stage COVID-19. Graphical Abstract ![]()
Key messages Induction of kinin B2 receptor in the nose of SARS-CoV-2-positive patients. Treatment with B2R-antagonist protects airway epithelial cells from SARS-CoV-2. B2R-antagonist reduces ACE2 levels in vivo and ex vivo. Protection by B2R-antagonist is mediated by inhibiting viral replication and spread.
Supplementary information The online version contains supplementary material available at 10.1007/s00109-022-02182-7.
Collapse
Affiliation(s)
- Constanze A Jakwerth
- Center of Allergy & Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, German, Research Center for Environmental Health, Member of the German Center for Lung Research (DZL), CPC-M, and Member of the Helmholtz I&I Initiative, Biedersteiner Str. 29, 80202, Munich, Germany
| | - Martin Feuerherd
- Institute of Virology, Technical University of Munich/Helmholtz Zentrum München, German Center of Infectiology Research (DZIF), Munich partner site, Munich, Germany
| | - Ferdinand M Guerth
- Center of Allergy & Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, German, Research Center for Environmental Health, Member of the German Center for Lung Research (DZL), CPC-M, and Member of the Helmholtz I&I Initiative, Biedersteiner Str. 29, 80202, Munich, Germany
| | - Madlen Oelsner
- Center of Allergy & Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, German, Research Center for Environmental Health, Member of the German Center for Lung Research (DZL), CPC-M, and Member of the Helmholtz I&I Initiative, Biedersteiner Str. 29, 80202, Munich, Germany
| | - Linda Schellhammer
- Institute of Laboratory Animal Science, University of Zurich, Zurich, Switzerland
| | - Johanna Giglberger
- Center of Allergy & Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, German, Research Center for Environmental Health, Member of the German Center for Lung Research (DZL), CPC-M, and Member of the Helmholtz I&I Initiative, Biedersteiner Str. 29, 80202, Munich, Germany.,Department of Otorhinolaryngology and Head and Neck Surgery, Medical School, Technical University of Munich, Munich, Germany
| | - Lisa Pechtold
- Department of Otorhinolaryngology and Head and Neck Surgery, Medical School, Technical University of Munich, Munich, Germany
| | - Claudia Jerin
- Center of Allergy & Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, German, Research Center for Environmental Health, Member of the German Center for Lung Research (DZL), CPC-M, and Member of the Helmholtz I&I Initiative, Biedersteiner Str. 29, 80202, Munich, Germany.,Department of Otorhinolaryngology and Head and Neck Surgery, Medical School, Technical University of Munich, Munich, Germany
| | - Luisa Kugler
- Department of Otorhinolaryngology and Head and Neck Surgery, Medical School, Technical University of Munich, Munich, Germany
| | - Carolin Mogler
- Institute of Pathology, Technical University Munich, Munich, Germany
| | - Bernhard Haller
- Institute of Medical Informatics, Statistics and Epidemiology, Medical School, Technical University of Munich, Munich, Germany
| | - Anna Erb
- Center of Allergy & Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, German, Research Center for Environmental Health, Member of the German Center for Lung Research (DZL), CPC-M, and Member of the Helmholtz I&I Initiative, Biedersteiner Str. 29, 80202, Munich, Germany
| | - Barbara Wollenberg
- Department of Otorhinolaryngology and Head and Neck Surgery, Medical School, Technical University of Munich, Munich, Germany
| | - Christoph D Spinner
- Department of Internal Medicine II, University Hospital Rechts Der Isar, Medical School, Technical University of Munich, Munich, Germany
| | - Thorsten Buch
- Institute of Laboratory Animal Science, University of Zurich, Zurich, Switzerland
| | - Ulrike Protzer
- Institute of Virology, Technical University of Munich/Helmholtz Zentrum München, German Center of Infectiology Research (DZIF), Munich partner site, Munich, Germany
| | - Carsten B Schmidt-Weber
- Center of Allergy & Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, German, Research Center for Environmental Health, Member of the German Center for Lung Research (DZL), CPC-M, and Member of the Helmholtz I&I Initiative, Biedersteiner Str. 29, 80202, Munich, Germany.
| | - Ulrich M Zissler
- Center of Allergy & Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, German, Research Center for Environmental Health, Member of the German Center for Lung Research (DZL), CPC-M, and Member of the Helmholtz I&I Initiative, Biedersteiner Str. 29, 80202, Munich, Germany
| | - Adam M Chaker
- Center of Allergy & Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, German, Research Center for Environmental Health, Member of the German Center for Lung Research (DZL), CPC-M, and Member of the Helmholtz I&I Initiative, Biedersteiner Str. 29, 80202, Munich, Germany.,Department of Otorhinolaryngology and Head and Neck Surgery, Medical School, Technical University of Munich, Munich, Germany
| |
Collapse
|
5
|
Li W, Xu J, Cheng L, Zhao C, Zhang L, Shao Q, Guo F. RelB promotes the migration and invasion of prostate cancer DU145 cells via exosomal ICAM1 in vitro. Cell Signal 2021; 91:110221. [PMID: 34933092 DOI: 10.1016/j.cellsig.2021.110221] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 12/10/2021] [Accepted: 12/14/2021] [Indexed: 12/24/2022]
Abstract
RelB confers the aggressiveness to prostate cancer (PC) cells. Exosomes modulate the oncogenesis and progression of PC. We aimed to identify the downstream molecule in the exosomes, by which RelB increases the aggressiveness of DU145. Totally, 137 upregulated and 55 downregulated exosomal proteins were identified from RelB-knockdown DU145 cells by Liquid Chromatography-Mass Spectrometry. UALCAN, GeneMANIA and tissue microarray analysis revealed that intercellular adhesion molecule-1 (ICAM1) was positively related to and co-expressed with RelB in PC. Luciferase reporter assay revealed that RelB bound directly to the promoter of ICAM1. ICAM1 overexpression enhanced the migration and invasion abilities of DU145 cells. Exposure to exosomes derived from ICAM1 overexpressing cells (hICAM1-exo) strengthened the aggressiveness of RelB-knockdown cells, especially the migration and invasion capabilities. Mechanistically, the expression of ICAM1, Integrin β1, MMP9 and uPA were upregulated in RelB-knockdown cells upon hICAM1-exo treatment. Exosomal ICAM1 is the key molecule regulated by RelB, which increased the aggressiveness of DU145. The study suggests that cell-cell communication via exosomal ICAM1 is a novel mechanism by which RelB promotes PC progression.
Collapse
Affiliation(s)
- Wenjing Li
- Department of Oncology, the First Affiliated Hospital of Soochow University, Suzhou, China; Department of Clinical Laboratory, the Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Jingjing Xu
- Department of Clinical Laboratory, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Li Cheng
- Department of Oncology, Shanghai East Hospital, Tongji Uiniversity School of Medicine, Shanghai, China
| | - Chenyi Zhao
- Department of Oncology, the Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Lianjun Zhang
- Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, China
| | - Qiang Shao
- Department of Urology, the Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China.
| | - Feng Guo
- Department of Oncology, the Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China.
| |
Collapse
|
6
|
Huang D, Sun L, Huang L, Chen Y. Nanodrug Delivery Systems Modulate Tumor Vessels to Increase the Enhanced Permeability and Retention Effect. J Pers Med 2021; 11:124. [PMID: 33672813 PMCID: PMC7917988 DOI: 10.3390/jpm11020124] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 02/07/2021] [Accepted: 02/08/2021] [Indexed: 12/24/2022] Open
Abstract
The use of nanomedicine for antitumor therapy has been extensively investigated for a long time. Enhanced permeability and retention (EPR) effect-mediated drug delivery is currently regarded as an effective way to bring drugs to tumors, especially macromolecular drugs and drug-loaded pharmaceutical nanocarriers. However, a disordered vessel network, and occluded or embolized tumor blood vessels seriously limit the EPR effect. To augment the EPR effect and improve curative effects, in this review, we focused on the perspective of tumor blood vessels, and analyzed the relationship among abnormal angiogenesis, abnormal vascular structure, irregular blood flow, extensive permeability of tumor vessels, and the EPR effect. In this commentary, nanoparticles including liposomes, micelles, and polymers extravasate through the tumor vasculature, which are based on modulating tumor vessels, to increase the EPR effect, thereby increasing their therapeutic effect.
Collapse
Affiliation(s)
- Dong Huang
- Shanghai Key Laboratory of Functional Materials Chemistry, East China University of Science and Technology, Shanghai 200237, China; (D.H.); (L.S.)
- Engineering Research Centre of Pharmaceutical Process Chemistry, Ministry of Education, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Lingna Sun
- Shanghai Key Laboratory of Functional Materials Chemistry, East China University of Science and Technology, Shanghai 200237, China; (D.H.); (L.S.)
- Engineering Research Centre of Pharmaceutical Process Chemistry, Ministry of Education, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Leaf Huang
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA;
| | - Yanzuo Chen
- Shanghai Key Laboratory of Functional Materials Chemistry, East China University of Science and Technology, Shanghai 200237, China; (D.H.); (L.S.)
- Engineering Research Centre of Pharmaceutical Process Chemistry, Ministry of Education, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| |
Collapse
|
7
|
Vanden Oever M, Muldoon D, Mathews W, Tolar J. Fludarabine modulates expression of type VII collagen during haematopoietic stem cell transplantation for recessive dystrophic epidermolysis bullosa. Br J Dermatol 2020; 185:380-390. [PMID: 33368156 DOI: 10.1111/bjd.19757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 12/16/2020] [Accepted: 12/19/2020] [Indexed: 11/28/2022]
Abstract
BACKGROUND Recessive dystrophic epidermolysis bullosa (RDEB) is a severe, complicated inherited blistering skin disease with few treatment options currently available. Recently, haematopoietic stem cell transplantation (HCT) has been used as an alternative therapy that can improve skin integrity, but it is not known if the preparative HCT regimen also contributes to the therapeutic response. OBJECTIVES To determine whether chemotherapy drugs used in the HCT preparative regimen influence type VII collagen (C7) expression, which is inherently reduced or absent in RDEB skin, and to explore the pathomechanisms of such responses, if present. METHODS Drugs from the HCT preparative regimen (busulfan, cyclophosphamide, ciclosporin A, fludarabine and mycophenolate) with inhibitors (PD98059, U0126, LY294002, SR11302, SIS3 and N-acetyl-l-cysteine) were added to normal human dermal and human RDEB fibroblasts. C7 expression was measured using reversetranscription polymerase chain reaction and immunoblotting. RESULTS We uncovered a previously unknown consequence of fludarabine whereby dermal fibroblasts exposed to fludarabine upregulate C7. This effect is mediated, in part, through activation of the mitogen-activated protein kinase/extracellular signal-regulated kinase, phosphoinositide 3-kinase/protein kinase B and transforming growth factor-β pathways. Activation of these pathways leads to activation of downstream transcription factors, including activator protein 1 (AP-1) and SMAD. Subsequently, both AP-1 and SMAD bind the COL7A1 promoter and increase COL7A1 expression. CONCLUSIONS Fludarabine influences the production of type VII collagen in RDEB fibroblasts.
Collapse
Affiliation(s)
- M Vanden Oever
- Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota, Minneapolis, MN, USA.,Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA
| | - D Muldoon
- Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota, Minneapolis, MN, USA.,Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA
| | - W Mathews
- Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota, Minneapolis, MN, USA
| | - J Tolar
- Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota, Minneapolis, MN, USA.,Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
8
|
Sökeland G, Schumacher U. The functional role of integrins during intra- and extravasation within the metastatic cascade. Mol Cancer 2019; 18:12. [PMID: 30657059 PMCID: PMC6337777 DOI: 10.1186/s12943-018-0937-3] [Citation(s) in RCA: 137] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 12/27/2018] [Indexed: 02/07/2023] Open
Abstract
Formation of distant metastases is by far the most common cause of cancer-related deaths. The process of metastasis formation is complex, and within this complex process the formation of migratory cells, the so called epithelial mesenchymal transition (EMT), which enables cancer cells to break loose from the primary tumor mass and to enter the bloodstream, is of particular importance. To break loose from the primary cancer, cancer cells have to down-regulate the cell-to-cell adhesion molecuIes (CAMs) which keep them attached to neighboring cancer cells. In contrast to this downregulation of CAMS in the primary tumor, cancer cells up-regulate other types of CAMs, that enable them to attach to the endothelium in the organ of the future metastasis. During EMT, the expression of cell-to-cell and cell-to-matrix adhesion molecules and their down- and upregulation is therefore critical for metastasis formation. Tumor cells mimic leukocytes to enable transmigration of the endothelial barrier at the metastatic site. The attachment of leukocytes/cancer cells to the endothelium are mediated by several CAMs different from those at the site of the primary tumor. These CAMs and their ligands are organized in a sequential row, the leukocyte adhesion cascade. In this adhesion process, integrins and their ligands are centrally involved in the molecular interactions governing the transmigration. This review discusses the integrin expression patterns found on primary tumor cells and studies whether their expression correlates with tumor progression, metastatic capacity and prognosis. Simultaneously, further possible, but so far unclearly characterized, alternative adhesion molecules and/or ligands, will be considered and emerging therapeutic possibilities reviewed.
Collapse
Affiliation(s)
- Greta Sökeland
- Institute of Anatomy and Experimental Morphology, University Cancer Center, University Medical Center Hamburg Eppendorf, Martinistraße 52, 20246, Hamburg, Germany.
| | - Udo Schumacher
- Institute of Anatomy and Experimental Morphology, University Cancer Center, University Medical Center Hamburg Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| |
Collapse
|
9
|
Kalyane D, Raval N, Maheshwari R, Tambe V, Kalia K, Tekade RK. Employment of enhanced permeability and retention effect (EPR): Nanoparticle-based precision tools for targeting of therapeutic and diagnostic agent in cancer. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 98:1252-1276. [PMID: 30813007 DOI: 10.1016/j.msec.2019.01.066] [Citation(s) in RCA: 531] [Impact Index Per Article: 88.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 01/02/2019] [Accepted: 01/15/2019] [Indexed: 02/07/2023]
Abstract
In tumorous tissues, the absence of vasculature supportive tissues intimates the formation of leaky vessels and pores (100 nm to 2 μm in diameter) and the poor lymphatic system offers great opportunity to treat cancer and the phenomenon is known as Enhanced permeability and retention (EPR) effect. The trends in treating cancer by making use of EPR effect is increasing day by day and generate multitudes of possibility to design novel anticancer therapeutics. This review aimed to present various factors affecting the EPR effect along with important things to know about EPR effect such as tumor perfusion, lymphatic function, interstitial penetration, vascular permeability, nanoparticle retention etc. This manuscript expounds the current advances and cross-talks the developments made in the of EPR effect-based therapeutics in cancer therapy along with a transactional view of its current clinical and industrial aspects.
Collapse
Affiliation(s)
- Dnyaneshwar Kalyane
- National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, An Institute of National Importance, Government of India, Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, Palaj, Opposite Air Force Station, Gandhinagar, Gujarat 382355, India
| | - Nidhi Raval
- National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, An Institute of National Importance, Government of India, Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, Palaj, Opposite Air Force Station, Gandhinagar, Gujarat 382355, India
| | - Rahul Maheshwari
- National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, An Institute of National Importance, Government of India, Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, Palaj, Opposite Air Force Station, Gandhinagar, Gujarat 382355, India
| | - Vishakha Tambe
- National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, An Institute of National Importance, Government of India, Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, Palaj, Opposite Air Force Station, Gandhinagar, Gujarat 382355, India
| | - Kiran Kalia
- National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, An Institute of National Importance, Government of India, Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, Palaj, Opposite Air Force Station, Gandhinagar, Gujarat 382355, India
| | - Rakesh K Tekade
- National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, An Institute of National Importance, Government of India, Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, Palaj, Opposite Air Force Station, Gandhinagar, Gujarat 382355, India.
| |
Collapse
|
10
|
Liu YS, Hsu JW, Lin HY, Lai SW, Huang BR, Tsai CF, Lu DY. Bradykinin B1 receptor contributes to interleukin-8 production and glioblastoma migration through interaction of STAT3 and SP-1. Neuropharmacology 2019; 144:143-154. [PMID: 30366000 DOI: 10.1016/j.neuropharm.2018.10.033] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 10/20/2018] [Accepted: 10/22/2018] [Indexed: 01/05/2023]
Abstract
Glioblastoma (GBM), the most aggressive brain tumor, has a poor prognosis due to the ease of migration to surrounding healthy brain tissue. Recent studies have shown that bradykinin receptors are involved in the progression of various cancers. However, the molecular mechanism and pathological role of bradykinin receptors remains unclear. We observed the expressions of two major bradykinin receptors, B1R and B2R, in two different human GBM cell lines, U87 and GBM8901. Cytokine array analysis showed that bradykinin increases the production of interleukin (IL)-8 in GBM via B1R. Higher B1R levels correlate with IL-8 expression in U87 and GBM8901. We observed increased levels of phosphorylated STAT3 and SP-1 in the nucleus as well. Using chromatin immunoprecipitation assay, we found that STAT3 and SP-1 mediate IL-8 expression, which gets abrogated by the inhibition of FAK and STAT3. We further demonstrated that IL-8 expression and cell migration are also regulated by the SP-1. In addition, expression levels of STAT3 and SP-1 positively correlate with clinicopathological grades of gliomas. Interestingly, our results found that inhibition of HDAC increases IL-8 expression. Moreover, stimulation with bradykinin caused increases in acetylated SP-1 and p300 complex formation, which are abrogated by inhibition of FAK and STAT3. Meanwhile, knockdown of SP-1 and p300 decreased the augmentation of bradykinin-induced IL-8 expression. These results indicate that bradykinin-induced IL-8 expression is dependent on B1R which causes phosphorylated STAT3 and acetylated SP-1 to translocate to the nucleus, hence resulting in GBM migration.
Collapse
Affiliation(s)
- Yu-Shu Liu
- Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan; Department of Biotechnology, Asia University, Taichung, Taiwan
| | - Jhih-Wen Hsu
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Hsiao-Yun Lin
- Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan
| | - Sheng-Wei Lai
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
| | - Bor-Ren Huang
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan; Department of Neurosurgery, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung, Taiwan; School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Cheng-Fang Tsai
- Department of Biotechnology, Asia University, Taichung, Taiwan
| | - Dah-Yuu Lu
- Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan; Department of Photonics and Communication Engineering, Asia University, Taichung, Taiwan.
| |
Collapse
|
11
|
Gillan V, Simpson DM, Kinnaird J, Maitland K, Shiels B, Devaney E. Characterisation of infection associated microRNA and protein cargo in extracellular vesicles of Theileria annulata infected leukocytes. Cell Microbiol 2018; 21:e12969. [PMID: 30370674 PMCID: PMC6492283 DOI: 10.1111/cmi.12969] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 10/19/2018] [Indexed: 12/11/2022]
Abstract
The protozoan parasites Theileria annulata and Theileria parva are unique amongst intracellular eukaryotic pathogens as they induce a transformation-like phenotype in their bovine host cell. T. annulata causes tropical theileriosis, which is frequently fatal, with infected leukocytes becoming metastatic and forming foci in multiple organs resulting in destruction of the lymphoid system. Exosomes, a subset of extracellular vesicles (EV), are critical in metastatic progression in many cancers. Here, we characterised the cargo of EV from a control bovine lymphosarcoma cell line (BL20) and BL20 infected with T. annulata (TBL20) by comparative mass spectrometry and microRNA (miRNA) profiling (data available via ProteomeXchange, identifier PXD010713 and NCBI GEO, accession number GSE118456, respectively). Ingenuity pathway analysis that many infection-associated proteins essential to migration and extracellular matrix digestion were upregulated in EV from TBL20 cells compared with BL20 controls. An altered repertoire of host miRNA, many with known roles in tumour and/or infection biology, was also observed. Focusing on the tumour suppressor miRNA, bta-miR-181a and bta-miR-181b, we identified putative messenger RNA targets and confirmed the interaction of bta-miR181a with ICAM-1. We propose that EV and their miRNA cargo play an important role in the manipulation of the host cell phenotype and the pathobiology of Theileria infection.
Collapse
Affiliation(s)
- Victoria Gillan
- Institute of Biodiversity, Animal Health and Comparative Medicine, University of Glasgow, Glasgow, UK
| | - Deborah M Simpson
- Institute of Integrative Biology, Centre for Proteome Research, University of Liverpool, Liverpool, UK
| | - Jane Kinnaird
- Institute of Biodiversity, Animal Health and Comparative Medicine, University of Glasgow, Glasgow, UK
| | - Kirsty Maitland
- Institute of Biodiversity, Animal Health and Comparative Medicine, University of Glasgow, Glasgow, UK
| | - Brian Shiels
- Institute of Biodiversity, Animal Health and Comparative Medicine, University of Glasgow, Glasgow, UK
| | - Eileen Devaney
- Institute of Biodiversity, Animal Health and Comparative Medicine, University of Glasgow, Glasgow, UK
| |
Collapse
|
12
|
ICAM3 mediates tumor metastasis via a LFA-1-ICAM3-ERM dependent manner. Biochim Biophys Acta Mol Basis Dis 2018; 1864:2566-2578. [PMID: 29729315 DOI: 10.1016/j.bbadis.2018.05.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 03/20/2018] [Accepted: 05/01/2018] [Indexed: 12/23/2022]
Abstract
ICAM3 was reported to promote metastasis in tumors. However, the underlying mechanism remains elusive. Here, we disclosed that the expression of ICAM3 was closely correlated with the TNM stage of human breast and lung cancer, as well as the dominant overexpression in high aggressive tumor cell lines (231 and A549 cells). Moreover, the knockdown of ICAM3 inhibited tumor metastasis whereas the ectopic expression of ICAM3 promoted tumor metastasis both in vitro and in vivo. In addition, exploration of the underlying mechanism demonstrated that ICAM3 not only binds to LFA-1 with its extracellular domain and structure protein ERM but also to lamellipodia with its intracellular domain which causes a tension that pulls cells apart (metastasis). Furthermore, ICAM3 extracellular or intracellular mutants alternatively abolished ICAM3 mediated tumor metastasis in vitro and in vivo. As a therapy strategy, LFA-1 antibody or Lifitegrast restrained tumor metastasis via targeting ICAM3-LFA-1 interaction. In summary, the aforementioned findings suggest a model of ICAM3 in mediating tumor metastasis. This may provide a promising target or strategy for the prevention of tumor metastasis.
Collapse
|
13
|
ICAM3 mediates inflammatory signaling to promote cancer cell stemness. Cancer Lett 2018; 422:29-43. [PMID: 29477378 DOI: 10.1016/j.canlet.2018.02.034] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 02/19/2018] [Accepted: 02/20/2018] [Indexed: 02/05/2023]
Abstract
In this study, we present a medium throughput siRNA screen platform to identify inflammation genes that regulate cancer cell stemness. We identified several novel candidates that decrease OCT4 expression and reduce the ALDH + subpopulation both of which are characteristic of stemness. Furthermore, one of the novel candidates ICAM3 up-regulates in the ALDH + subpopulation, the side population and the developed spheres. ICAM3 knockdown reduces the side population, sphere formation and chemo-resistance in MDA-MB-231 human breast cancer cells and A549 lung cancer cells. In addition, mice bearing MDA-MB-231-shICAM3 cells develop smaller tumors and fewer lung metastases versus control. Interestingly, ICAM3 recruits and binds to Src by the YLPL motif in its intracellular domain which further activates the PI3K-AKT phosphorylation cascades. The activated p-AKT enhances SOX2 and OCT4 activity and thereby maintains cancer cell stemness. Meanwhile, the p-AKT facilitated p50 nuclear translocation/activation enhances p50 feedback and thereby promotes ICAM3 expression by binding to the ICAM3 promoter region. On this basis, Src and PI3K inhibitors suppress ICAM3-mediated signaling pathways and reduce chemo-resistance which results in tumor growth suppression in vitro and in vivo. In summary, we identify a potential CSC regulator and suggest a novel mechanism by which ICAM3 governs cancer cell stemness and inflammation.
Collapse
|
14
|
Che X, Lu R, Fu Z, Sun Y, Zhu ZF, Li JP, Wang S, Jia J, Wang Q, Yao Z. Therapeutic effects of tyroserleutide on lung metastasis of human hepatocellular carcinoma SK-HEP-1 and its mechanism affecting ICAM-1 and MMP-2 and -9. Drug Des Devel Ther 2018; 12:3357-3368. [PMID: 30349190 PMCID: PMC6186774 DOI: 10.2147/dddt.s177126] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Tyroserleutide (YSL) inhibits the growth and metastasis of human hepatocellular carcinoma (HCC). This paper studied the effect of YSL on metastasis of human HCC and investigated its mechanisms. METHODS In vivo, experimental lung metastasis models of human HCC SK-HEP-1 cells in nude mice were established, and In vitro, the proliferation, adhesion and invasion of SK-HEP-1 cells were detected. RESULTS In vivo, YSL significantly inhibited the metastasis of human HCC. In vitro, YSL significantly inhibited the proliferation, adhesion and invasion of SK-HEP-1 cells. Through analyses with reverse transcription PCR (RT-PCR) and Western blot, we observed that YSL significantly inhibited the expressions of ICAM-1 in SK-HEP-1 cells. Through RT-PCR, Western blot and zymography methods, YSL was discovered to decrease the mRNA level, protein expression and activity of MMP-2 and -9 in SK-HEP-1 cells. CONCLUSION We concluded that YSL could inhibit tumor growth and metastasis of human HCC SK-HEP-1 cells.
Collapse
Affiliation(s)
- Xuchun Che
- Department of Immunology, Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Educational Ministry of China, School of Medical Sciences, Tianjin Medical University, Tianjin, People's Republic of China, ;
| | - Rong Lu
- Department of Immunology, Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Educational Ministry of China, School of Medical Sciences, Tianjin Medical University, Tianjin, People's Republic of China, ; .,Department of Drug Development, Tianjin Kangzhe Pharmaceutical Company, Ltd., Tianjin, People's Republic of China
| | - Zheng Fu
- Department of Drug Development, Tianjin Kangzhe Pharmaceutical Company, Ltd., Tianjin, People's Republic of China
| | - Yajun Sun
- Department of Blood Transfusion, General Hospital, Tianjin Medical University, Tianjin, People's Republic of China
| | - Zhi-Feng Zhu
- Department of Drug Development, Tianjin Kangzhe Pharmaceutical Company, Ltd., Tianjin, People's Republic of China
| | - Jin-Ping Li
- Department of Drug Development, Tianjin Kangzhe Pharmaceutical Company, Ltd., Tianjin, People's Republic of China
| | - Song Wang
- Department of Drug Development, Tianjin Kangzhe Pharmaceutical Company, Ltd., Tianjin, People's Republic of China
| | - Jing Jia
- Department of Drug Development, Tianjin Kangzhe Pharmaceutical Company, Ltd., Tianjin, People's Republic of China
| | - Qing Wang
- Department of Clinical Laboratory, General Hospital, Tianjin Medical University, Tianjin, People's Republic of China
| | - Zhi Yao
- Department of Immunology, Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Educational Ministry of China, School of Medical Sciences, Tianjin Medical University, Tianjin, People's Republic of China, ;
| |
Collapse
|
15
|
Kang JH, Choi MY, Cui YH, Kaushik N, Uddin N, Yoo KC, Kim MJ, Lee SJ. Regulation of FBXO4-mediated ICAM-1 protein stability in metastatic breast cancer. Oncotarget 2017; 8:83100-83113. [PMID: 29137327 PMCID: PMC5669953 DOI: 10.18632/oncotarget.20912] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 08/27/2017] [Indexed: 12/20/2022] Open
Abstract
Advanced or progressive cancers share common traits such as altered transcriptional modulation, genetic modification, and abnormal post-translational regulation. These processes influence protein stability and cellular activity. Intercellular adhesion molecule-1 (ICAM-1) is involved in the malignant progression of various human cancers, including breast, liver, renal, and pancreatic cancers, but protein stability has not been deal with in metastatic breast cancer. Additionally, the relevance of the stability maintenance of ICAM-1 protein remains obscure. Here, we identified a novel interaction of E3 ligase FBXO4 that is specifically presented to ICAM-1. To understand how FBXO4 modulates ICAM-1 stability, we investigated ICAM-1-overexpressing or knockdown metastatic/non-metastatic breast cancers. ICAM-1 was found to influence tumor progression and metastasis, whereas FBXO4 regulated aggressive tumorigenic conditions. These results demonstrate that FBXO4 is a major regulator of ICAM-1 stability and that alterations in the stability of ICAM-1 can influence therapeutic outcome in metastatic cancer.
Collapse
Affiliation(s)
- Jae-Hyeok Kang
- Department of Life Science, Research Institute for Natural Sciences, Hanyang University, Seoul, Korea
| | - Mi-Young Choi
- Department of Life Science, Research Institute for Natural Sciences, Hanyang University, Seoul, Korea
| | - Yan-Hong Cui
- Department of Life Science, Research Institute for Natural Sciences, Hanyang University, Seoul, Korea
| | - Neha Kaushik
- Department of Life Science, Research Institute for Natural Sciences, Hanyang University, Seoul, Korea
| | - Nizam Uddin
- Centre of Excellence in Molecular Biology (CEMB), University of The Panjab, Lahore, Pakistan
| | - Ki-Chun Yoo
- Department of Life Science, Research Institute for Natural Sciences, Hanyang University, Seoul, Korea
| | - Min-Jung Kim
- Laboratory of Radiation Exposure and Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - Su-Jae Lee
- Department of Life Science, Research Institute for Natural Sciences, Hanyang University, Seoul, Korea
| |
Collapse
|
16
|
Kuo HT, Pan J, Lau J, Zhang C, Zeisler J, Colpo N, Bénard F, Lin KS. Radiolabeled R954 Derivatives for Imaging Bradykinin B1 Receptor Expression with Positron Emission Tomography. Mol Pharm 2017; 14:821-829. [PMID: 28094956 DOI: 10.1021/acs.molpharmaceut.6b01055] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Peptide receptors have emerged as promising targets for diagnosis and therapy. The aberrant overexpression of these receptors in different cancer subtypes allows for the adoption of new treatment strategies that complement conventional chemotherapies. Bradykinin B1 receptor (B1R) is a G protein-coupled receptor that is overexpressed in many cancers, with limited expression in healthy tissues. Previously, we developed 68Ga- and 18F-labeled derivatives of B1R antagonist peptides B9858 and B9958, and successfully targeted B1R-expressing tumor xenografts in vivo. R954 (Ac-Orn-Arg-Oic-Pro-Gly-αMePhe-Ser-d-2-Nal-Ile), a potent B1R antagonist, is reportedly more stable than B9858 against peptidase degradation. We evaluated two radiolabeled derivatives of R954 (68Ga-HTK01083 and 18F-HTK01146) for B1R PET imaging. Peptides were synthesized via solid phase strategy. Nonradioactive standards were obtain by reacting GaCl3 with DOTA-dPEG2-R954 and by clicking N-propargyl-N,N-dimethylammoniomethyl-trifluoroborate with azidoacetyl-dPEG2-R954. Binding affinity for B1R was determined by an in vitro competition binding assay. 68Ga-HTK01083 was obtained by incubating DOTA-dPEG2-R954 with 68GaCl3 under acidic conditions, while 18F-HTK01146 was prepared via an 18F-19F isotope exchange reaction. Biodistribution and imaging studies were conducted at 1 h postinjection (p.i.) in mice inoculated with B1R-expressing (B1R+) and B1R-nonexpressing (B1R-) cells. HTK01083 and HTK01146 bound B1R with good affinity (Ki = 30.5 and 24.8 nM, respectively). 68Ga/18F-labeled R954 were obtained on average in ≥10% decay-corrected radiochemical yield with >99% radiochemical purity and ≥52 GBq/μmol specific activity. For both tracers, clearance was predominantly renal with minimal involvement of the hepatobiliary system. For PET images, B1R+ tumors, kidneys, and bladder were visible. At 1 h p.i., uptake in B1R+ tumor was comparable between 68Ga-HTK01083 (8.46 ± 1.44%ID/g) and 18F-HTK01146 (9.25 ± 0.69%ID/g). B1R+ tumor-to-blood and B1R+ tumor-to-muscle ratios were 6.32 ± 1.44 and 20.7 ± 3.58 for 68Ga-HTK01083, and 7.24 ± 2.56 and 19.5 ± 4.29 for 18F-HTK01146. Our results indicate R954 is a good lead sequence for optimization of B1R tracers for cancer imaging.
Collapse
Affiliation(s)
- Hsiou-Ting Kuo
- Department of Molecular Oncology, BC Cancer Agency , Vancouver, BC V5Z 1L3, Canada
| | - Jinhe Pan
- Department of Molecular Oncology, BC Cancer Agency , Vancouver, BC V5Z 1L3, Canada
| | - Joseph Lau
- Department of Molecular Oncology, BC Cancer Agency , Vancouver, BC V5Z 1L3, Canada
| | - Chengcheng Zhang
- Department of Molecular Oncology, BC Cancer Agency , Vancouver, BC V5Z 1L3, Canada
| | - Jutta Zeisler
- Department of Molecular Oncology, BC Cancer Agency , Vancouver, BC V5Z 1L3, Canada
| | - Nadine Colpo
- Department of Molecular Oncology, BC Cancer Agency , Vancouver, BC V5Z 1L3, Canada
| | - François Bénard
- Department of Molecular Oncology, BC Cancer Agency , Vancouver, BC V5Z 1L3, Canada.,Department of Functional Imaging, BC Cancer Agency , Vancouver, BC V5Z 4E6, Canada.,Department of Radiology, University of British Columbia , Vancouver, BC V5Z 4E3, Canada
| | - Kuo-Shyan Lin
- Department of Molecular Oncology, BC Cancer Agency , Vancouver, BC V5Z 1L3, Canada.,Department of Functional Imaging, BC Cancer Agency , Vancouver, BC V5Z 4E6, Canada.,Department of Radiology, University of British Columbia , Vancouver, BC V5Z 4E3, Canada
| |
Collapse
|
17
|
Chen Y, Yu Y, Sun S, Wang Z, Liu P, Liu S, Jiang J. Bradykinin promotes migration and invasion of hepatocellular carcinoma cells through TRPM7 and MMP2. Exp Cell Res 2016; 349:68-76. [DOI: 10.1016/j.yexcr.2016.09.022] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 09/26/2016] [Accepted: 09/28/2016] [Indexed: 12/26/2022]
|
18
|
Wang Y, Xu J, Gao G, Li J, Huang H, Jin H, Zhu J, Che X, Huang C. Tumor-suppressor NFκB2 p100 interacts with ERK2 and stabilizes PTEN mRNA via inhibition of miR-494. Oncogene 2016; 35:4080-90. [PMID: 26686085 PMCID: PMC4916044 DOI: 10.1038/onc.2015.470] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Revised: 10/19/2015] [Accepted: 11/14/2015] [Indexed: 12/19/2022]
Abstract
Emerging evidence from The Cancer Genome Atlas has revealed that nuclear factor κB2 (nfκb2) gene encoding p100 is genetically deleted or mutated in human cancers, implicating NFκB2 as a potential tumor suppressor. However, the molecular mechanism underlying the antitumorigenic action of p100 remains poorly understood. Here we report that p100 inhibits cancer cell anchorage-independent growth, a hallmark of cellular malignancy, by stabilizing the tumor-suppressor phosphatase and tensin homolog (PTEN) mRNA via a mechanism that is independent of p100's inhibitory role in NFκB activation. We further demonstrate that the regulatory effect of p100 on PTEN expression is mediated by its downregulation of miR-494 as a result of the inactivation of extracellular signal-regulated kinase 2 (ERK2), in turn leading to inhibition of c-Jun/activator protein-1-dependent transcriptional activity. Furthermore, we identify that p100 specifically interacts with non-phosphorylated ERK2 and prevents ERK2 phosphorylation and nuclear translocation. Moreover, the death domain at C-terminal of p100 is identified as being crucial and sufficient for its interaction with ERK2. Taken together, our findings provide novel mechanistic insights into the understanding of the tumor-suppressive role for NFκB2 p100.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Chuanshu Huang
- Corresponding author: Dr. Chuanshu Huang, Nelson Institute of Environmental Medicine, New York University School of Medicine, 57 Old Forge Road, Tuxedo, NY 10987, Tel: 845-731-3519, Fax: 845-351-2320,
| |
Collapse
|
19
|
Bai J, Adriani G, Dang TM, Tu TY, Penny HXL, Wong SC, Kamm RD, Thiery JP. Contact-dependent carcinoma aggregate dispersion by M2a macrophages via ICAM-1 and β2 integrin interactions. Oncotarget 2016; 6:25295-307. [PMID: 26231039 PMCID: PMC4694832 DOI: 10.18632/oncotarget.4716] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 07/17/2015] [Indexed: 12/20/2022] Open
Abstract
Tumor-associated macrophages (TAMs) can constitute up to 50% of the tumor mass and have strong implications in tumor progression and metastasis. Macrophages are plastic and can polarize to various subtypes that differ in terms of surface receptor expression as well as cytokine and chemokine production and effector function. Conventionally, macrophages are grouped into two major subtypes: the classically activated M1 macrophages and the alternatively activated M2 macrophages. M1 macrophages are pro-inflammatory, promote T helper (Th) 1 responses, and show tumoricidal activity, whereas M2 macrophages contribute to tissue repair and promote Th2 responses. Herein, we present a microfluidic system integrating tumor cell aggregates and subtypes of human monocyte-derived macrophages in a three-dimensional hydrogel scaffold, in close co-culture with an endothelial monolayer to create an in vitro tumor microenvironment. This platform was utilized to study the role of individual subtypes of macrophages (M0, M1, M2a, M2b and M2c) in human lung adenocarcinoma (A549) aggregate dispersion, as a representation of epithelial-mesenchymal transition (EMT). A significant difference was observed when M2a macrophages were in direct contact with or separated from A549 aggregates, suggesting a possible mechanism for proximity-induced, contact-dependent dissemination via ICAM-1 and integrin β2 interactions. Indeed, M2a macrophages tended to infiltrate and release cells from carcinoma cell aggregates. These findings may help in the development of immunotherapies based on enhancing the tumor-suppressive properties of TAMs.
Collapse
Affiliation(s)
- Jing Bai
- BioSystems and Micromechanics IRG, Singapore-MIT Alliance for Research and Technology, 138602, Singapore.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Giulia Adriani
- BioSystems and Micromechanics IRG, Singapore-MIT Alliance for Research and Technology, 138602, Singapore
| | - Truong-Minh Dang
- SIgN (Singapore Immunology Network), A*STAR (Agency for Science, Technology and Research), Biopolis, 138648, Singapore
| | - Ting-Yuan Tu
- BioSystems and Micromechanics IRG, Singapore-MIT Alliance for Research and Technology, 138602, Singapore
| | - Hwei-Xian Leong Penny
- SIgN (Singapore Immunology Network), A*STAR (Agency for Science, Technology and Research), Biopolis, 138648, Singapore
| | - Siew-Cheng Wong
- SIgN (Singapore Immunology Network), A*STAR (Agency for Science, Technology and Research), Biopolis, 138648, Singapore
| | - Roger D Kamm
- BioSystems and Micromechanics IRG, Singapore-MIT Alliance for Research and Technology, 138602, Singapore.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Jean-Paul Thiery
- BioSystems and Micromechanics IRG, Singapore-MIT Alliance for Research and Technology, 138602, Singapore.,Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, 117597, Singapore.,Institute of Molecular and Cell Biology, Proteos, 138673, Singapore
| |
Collapse
|
20
|
Xu Y, Zhang S, Niu H, Ye Y, Hu F, Chen S, Li X, Luo X, Jiang S, Liu Y, Chen Y, Li J, Xiang R, Li N. STIM1 accelerates cell senescence in a remodeled microenvironment but enhances the epithelial-to-mesenchymal transition in prostate cancer. Sci Rep 2015; 5:11754. [PMID: 26257076 PMCID: PMC4530453 DOI: 10.1038/srep11754] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 06/01/2015] [Indexed: 12/19/2022] Open
Abstract
The importance of store-operated Ca2+ entry (SOCE) and the role of its key molecular regulators, STIM1 and ORAI1, in the development of cancer are emerging. Here, we report an unexpected dual function of SOCE in prostate cancer progression by revealing a decrease in the expression of STIM1 in human hyperplasia and tumor tissues of high histological grade and by demonstrating that STIM1 and ORAI1 inhibit cell growth by arresting the G0/G1 phase and enhancing cell senescence in human prostate cancer cells. In addition, STIM1 and ORAI1 inhibited NF-κB signaling and remodeled the tumor microenvironment by reducing the formation of M2 phenotype macrophages, possibly creating an unfavorable tumor microenvironment and inhibiting cancer development. However, STIM1 also promoted cell migration and the epithelial-to-mesenchymal transition by activating TGF-β, Snail and Wnt/β-Catenin pathways. Thus, our study revealed novel regulatory effects and the mechanisms by which STIM1 affects cell senescence, tumor migration and the tumor microenvironment, revealing that STIM1 has multiple functions in prostate cancer cells.
Collapse
Affiliation(s)
- Yingxi Xu
- 1] School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China [2] State Key Lab of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences, Tianjin 300020, China
| | - Shu Zhang
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Haiying Niu
- Department of Obstetrics and Gynecology, First Central Hospital Clinic Institute, Tianjin Medical University, 24 Fukang Road, Tianjin 300192 China
| | - Yujie Ye
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Fen Hu
- School of Physics, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Si Chen
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Xuefei Li
- Beijing Health Vocational College, 94 Nanhengxijie Street, Beijing, 100053 China
| | - Xiaohe Luo
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Shan Jiang
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Yanhua Liu
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Yanan Chen
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Junying Li
- Department of Obstetrics and Gynecology, First Central Hospital Clinic Institute, Tianjin Medical University, 24 Fukang Road, Tianjin 300192 China
| | - Rong Xiang
- 1] School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China [2] Tianjin Key Laboratory of Tumor Microenvironment and Neurovascular Regulation, Tianjin 300071, China [3] Collaborative Innovation Center for Biotherapy, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Na Li
- 1] School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China [2] Tianjin Key Laboratory of Tumor Microenvironment and Neurovascular Regulation, Tianjin 300071, China [3] Collaborative Innovation Center for Biotherapy, Nankai University, 94 Weijin Road, Tianjin 300071, China
| |
Collapse
|
21
|
Lin CC, Lee IT, Hsu CH, Hsu CK, Chi PL, Hsiao LD, Yang CM. Sphingosine-1-phosphate mediates ICAM-1-dependent monocyte adhesion through p38 MAPK and p42/p44 MAPK-dependent Akt activation. PLoS One 2015; 10:e0118473. [PMID: 25734900 PMCID: PMC4348486 DOI: 10.1371/journal.pone.0118473] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Accepted: 01/19/2015] [Indexed: 11/25/2022] Open
Abstract
Up-regulation of intercellular adhesion molecule-1 (ICAM-1) is frequently implicated in lung inflammation. Sphingosine-1-phosphate (S1P) has been shown to play a key role in inflammation via adhesion molecules induction, and then causes lung injury. However, the mechanisms underlying S1P-induced ICAM-1 expression in human pulmonary alveolar epithelial cells (HPAEpiCs) remain unclear. The effect of S1P on ICAM-1 expression was determined by Western blot and real-time PCR. The involvement of signaling pathways in these responses was investigated by using the selective pharmacological inhibitors and transfection with siRNAs. S1P markedly induced ICAM-1 expression and monocyte adhesion which were attenuated by pretreatment with the inhibitor of S1PR1 (W123), S1PR3 (CAY10444), c-Src (PP1), EGFR (AG1478), PDGFR (AG1296), MEK1/2 (U0126), p38 MAPK (SB202190), JNK1/2 (SP600125), PI3K (LY294002), or AP-1 (Tanshinone IIA) and transfection with siRNA of S1PR1, S1PR3, c-Src, EGFR, PDGFR, p38, p42, JNK1, c-Jun, or c-Fos. We observed that S1P-stimulated p42/p44 MAPK and p38 MAPK activation was mediated via a c-Src/EGFR and PDGFR-dependent pathway. S1P caused the c-Src/EGFR/PDGFR complex formation. On the other hand, we demonstrated that S1P induced p42/p44 MAPK and p38 MAPK-dependent Akt activation. In addition, S1P-stimulated JNK1/2 phosphorylation was attenuated by SP600125 or PP1. Finally, S1P enhanced c-Fos mRNA levels and c-Jun phosphorylation. S1P-induced c-Jun activation was reduced by PP1, AG1478, AG1296, U0126, SP600125, SB202190, or LY294002. These results demonstrated that S1P-induced ICAM-1 expression and monocyte adhesion were mediated through S1PR1/3/c-Src/EGFR, PDGFR/p38 MAPK, p42/p44 MAPK/Akt-dependent AP-1 activation.
Collapse
Affiliation(s)
- Chih-Chung Lin
- Department of Anesthetics, Chang Gung Memorial Hospital at Lin-Kou and College of Medicine, Chang Gung University, Kwei-San, Tao-Yuan, Taiwan
| | - I-Ta Lee
- Department of Physiology and Pharmacology and Health Ageing Research Center, College of Medicine, Chang Gung University, Kwei-San, Tao-Yuan, Taiwan
| | - Chun-Hao Hsu
- Department of Physiology and Pharmacology and Health Ageing Research Center, College of Medicine, Chang Gung University, Kwei-San, Tao-Yuan, Taiwan
| | - Chih-Kai Hsu
- Department of Physiology and Pharmacology and Health Ageing Research Center, College of Medicine, Chang Gung University, Kwei-San, Tao-Yuan, Taiwan
| | - Pei-Ling Chi
- Department of Physiology and Pharmacology and Health Ageing Research Center, College of Medicine, Chang Gung University, Kwei-San, Tao-Yuan, Taiwan
| | - Li-Der Hsiao
- Department of Anesthetics, Chang Gung Memorial Hospital at Lin-Kou and College of Medicine, Chang Gung University, Kwei-San, Tao-Yuan, Taiwan
| | - Chuen-Mao Yang
- Department of Physiology and Pharmacology and Health Ageing Research Center, College of Medicine, Chang Gung University, Kwei-San, Tao-Yuan, Taiwan
- * E-mail:
| |
Collapse
|
22
|
Mou W, Xu Y, Ye Y, Chen S, Li X, Gong K, Liu Y, Chen Y, Li X, Tian Y, Xiang R, Li N. Expression of Sox2 in breast cancer cells promotes the recruitment of M2 macrophages to tumor microenvironment. Cancer Lett 2014; 358:115-123. [PMID: 25444903 DOI: 10.1016/j.canlet.2014.11.004] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Revised: 08/04/2014] [Accepted: 11/03/2014] [Indexed: 12/31/2022]
Abstract
Transcriptional factor Sox2 promotes tumor metastasis; however its regulatory effect on tumor-associated macrophages (TAMs, M2 phenotype) has not been defined. This study disclosed concomitant expression of TAMs marker-CD163 with SOX2 in human breast cancer and showed that Sox2 in breast cancer cells promotes recruitment of TAMs with altered expression of multiple chemokines, including MIP-1α, ICAM-1 etc. and activation of Stat3 and NF-κB signalings. In addition, TAMs rescued the compromised lung metastasis induced by Sox2 silencing in breast cancer cells. Together, this study documented that Sox2 plays an important role in recruiting TAMs and promotes tumor metastasis in a TAMs dependent manner.
Collapse
Affiliation(s)
- Wenjun Mou
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China; Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, Beijing 100045, China; Department of Biochemistry, Chinese PLA General Hospital, Beijing 100853, China
| | - Yingxi Xu
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Yujie Ye
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Si Chen
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Xuefei Li
- Beijing Health Vocational College, 94 Nanhengxijie Street, Beijing 100053, China
| | - Kangzi Gong
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Yanhua Liu
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Yanan Chen
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Xiru Li
- Department of General Surgery, Chinese PLA General Hospital, Beijing 100853, China
| | - Yaping Tian
- Department of Biochemistry, Chinese PLA General Hospital, Beijing 100853, China
| | - Rong Xiang
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China; Tianjin Key Laboratory of Tumor Microenvironment and Neurovascular Regulation, Tianjin 300071, China; Collaborative Innovation Center for Biotherapy, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Na Li
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China; Tianjin Key Laboratory of Tumor Microenvironment and Neurovascular Regulation, Tianjin 300071, China; Collaborative Innovation Center for Biotherapy, Nankai University, 94 Weijin Road, Tianjin 300071, China.
| |
Collapse
|
23
|
Wang D, Luo L, Guo J. miR-129-1-3p inhibits cell migration by targeting BDKRB2 in gastric cancer. Med Oncol 2014; 31:98. [DOI: 10.1007/s12032-014-0098-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Accepted: 06/23/2014] [Indexed: 12/20/2022]
|
24
|
Chen HT, Tsou HK, Chen JC, Shih JMK, Chen YJ, Tang CH. Adiponectin enhances intercellular adhesion molecule-1 expression and promotes monocyte adhesion in human synovial fibroblasts. PLoS One 2014; 9:e92741. [PMID: 24667577 PMCID: PMC3965461 DOI: 10.1371/journal.pone.0092741] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Accepted: 02/24/2014] [Indexed: 12/13/2022] Open
Abstract
Adiponectin is a protein hormone secreted predominantly by differentiated adipocytes and is involved in energy homeostasis. Adiponectin expression is significantly high in the synovial fluid of patients with osteoarthritis (OA). Intercellular adhesion molecule-1 (ICAM-1) is an important adhesion molecule that mediates monocyte adhesion and infiltration during OA pathogenesis. Adiponectin-induced expression of ICAM-1 in human OA synovial fibroblasts (OASFs) was examined by using qPCR, flow cytometry and western blotting. The intracellular signaling pathways were investigated by pretreated with inhibitors or transfection with siRNA. The monocyte THP-1 cell line was used for an adhesion assay with OASFs. Stimulation of OASFs with adiponectin induced ICAM-1 expression. Pretreatment with AMP-activated protein kinase (AMPK) inhibitors (AraA and compound C) or transfection with siRNA against AMPKα1 and two AMPK upstream activator- liver kinase B1 (LKB1) and calmodulin-dependent protein kinase II (CaMKII) diminished the adiponectin-induced ICAM-1 expression. Stimulation of OASFs with adiponectin increased phosphorylation of LKB1, CaMKII, AMPK, and c-Jun, resulting in c-Jun binding to AP-1 element of ICAM-1 promoter. In addition, adiponectin-induced activation of the LKB1/CaMKII, AMPK, and AP-1 pathway increased the adhesion of monocytes to the OASF monolayer. Our results suggest that adiponectin increases ICAM-1 expression in human OASFs via the LKB1/CaMKII, AMPK, c-Jun, and AP-1 signaling pathway. Adiponectin-induced ICAM-1 expression promoted the adhesion of monocytes to human OASFs. These findings may provide a better understanding of the pathogenesis of OA and can utilize this knowledge to design a new therapeutic strategy.
Collapse
Affiliation(s)
- Hsien-Te Chen
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
- Department of Orthopaedic Surgery, China Medical University Hospital, Taichung, Taiwan
| | - Hsi-Kai Tsou
- Department of Neurosurgery, Taichung Veterans General Hospital, Taichung, Taiwan
- Department of Early Childhood Care and Education, Jen-Teh Junior College of Medicine, Nursing and Management, Miaoli County, Taiwan
| | - Jui-Chieh Chen
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
- National Institute of Cancer Research, National Health Research Institutes, Miaoli County, Zhunan, Taiwan
| | | | - Yen-Jen Chen
- Department of Orthopaedic Surgery, China Medical University Hospital, Taichung, Taiwan
- School of Medicine, China Medical University, Taichung, Taiwan
| | - Chih-Hsin Tang
- School of Medicine, China Medical University, Taichung, Taiwan
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
- Department of Biotechnology, College of Health Science, Asia University, Taichung, Taiwan
| |
Collapse
|
25
|
da Costa PLN, Sirois P, Tannock IF, Chammas R. The role of kinin receptors in cancer and therapeutic opportunities. Cancer Lett 2013; 345:27-38. [PMID: 24333733 DOI: 10.1016/j.canlet.2013.12.009] [Citation(s) in RCA: 90] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Revised: 11/29/2013] [Accepted: 12/02/2013] [Indexed: 12/20/2022]
Abstract
Kinins are generated within inflammatory tissue microenvironments, where they exert diverse functions, including cell proliferation, leukocyte activation, cell migration, endothelial cell activation and nociception. These pleiotropic functions depend on signaling through two cross talking receptors, the constitutively expressed kinin receptor 2 (B2R) and the inducible kinin receptor 1 (B1R). We have reviewed evidence, which supports the concept that kinin receptors, especially kinin receptor 1, are promising targets for cancer therapy, since (1) many tumor cells express aberrantly high levels of these receptors; (2) some cancers produce kinins and use them as autocrine factors to stimulate their growth; (3) activation of kinin receptors leads to activation of macrophages, dendritic cells and other cells from the tumor microenvironment; (4) kinins have pro-angiogenic properties; (5) kinin receptors have been implicated in cancer migration, invasion and metastasis; and (6) selective antagonists for either B1R or B2R have shown anti-proliferative, anti-inflammatory, anti-angiogenic and anti-migratory properties. The multiple cross talks between kinin receptors and renin-angiotensin system (RAS) as well as its implications for targeting KKS or RAS for the treatment of malignancies are also discussed. It is expected that B1R antagonists would interfere less with housekeeping functions and therefore would be attractive compounds to treat selected types of cancer. Reliable clinical studies are needed to establish the translatability of these data to human settings and the usefulness of kinin receptor antagonists.
Collapse
Affiliation(s)
- Patrícia L N da Costa
- Laboratório de Oncologia Experimental, Faculdade de Medicina da Universidade de São Paulo and Instituto do Câncer do Estado de São Paulo, Brazil
| | - Pierre Sirois
- CHUL Research Center, Laval University, Quebec City, Canada
| | - Ian F Tannock
- Princess Margaret Cancer Centre and University of Toronto, Toronto, ON, Canada
| | - Roger Chammas
- Laboratório de Oncologia Experimental, Faculdade de Medicina da Universidade de São Paulo and Instituto do Câncer do Estado de São Paulo, Brazil.
| |
Collapse
|
26
|
Bradykinin promotes vascular endothelial growth factor expression and increases angiogenesis in human prostate cancer cells. Biochem Pharmacol 2013; 87:243-53. [PMID: 24225154 DOI: 10.1016/j.bcp.2013.10.016] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Revised: 10/16/2013] [Accepted: 10/17/2013] [Indexed: 12/15/2022]
Abstract
Prostate cancer is the most commonly diagnosed malignancy in men and shows a tendency for metastasis to distant organs. Angiogenesis is required for metastasis. Bradykinin (BK) is an inflammatory mediator involved in tumor growth and metastasis, but its role in vascular endothelial growth factor (VEGF) expression and angiogenesis in human prostate cancer remains unknown. The aim of this study was to examine whether BK promotes prostate cancer angiogenesis via VEGF expression. We found that exogenous BK increased VEGF expression in prostate cancer cells and further promoted tube formation in endothelial progenitor cells and human umbilical vein endothelial cells. Pretreatment of prostate cancer with B2 receptor antagonist or small interfering RNA (siRNA) reduced BK-mediated VEGF production. The Akt and mammalian target of rapamycin (mTOR) pathways were activated after BK treatment, and BK-induced VEGF expression was abolished by the specific inhibitor and siRNA of the Akt and mTOR cascades. BK also promoted nuclear factor-κB (NF-κB) and activator protein 1 (AP-1) activity. Importantly, BK knockdown reduced VEGF expression and abolished prostate cancer cell conditional medium-mediated angiogenesis. Taken together, these results indicate that BK operates through the B2 receptor, Akt, and mTOR, which in turn activate NF-κB and AP-1, activating VEGF expression and contributing to angiogenesis in human prostate cancer cells.
Collapse
|