1
|
Toti A, Lucarini E, Ferrara V, Parisio C, Ciampi C, Gerace E, Micheli L, Margiotta F, Venturi D, Mello T, Lacal PM, Graziani G, Mannaioni G, Ghelardini C, Di Cesare Mannelli L. The dual role of VEGF-A in a complex in vitro model of oxaliplatin-induced neurotoxicity: Pain-related and neuroprotective effects. Neurotherapeutics 2025; 22:e00532. [PMID: 39939241 PMCID: PMC12014407 DOI: 10.1016/j.neurot.2025.e00532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 01/15/2025] [Accepted: 01/15/2025] [Indexed: 02/14/2025] Open
Abstract
Vascular endothelial growth factor (VEGF)-A is a main player in the development of neuropathic pain induced by chemotherapy and the pharmacological blockade of VEGF receptor (VEGFR) subtype 1 is a pain killer strategy. Interestingly, VEGF-A has been demonstrated to have also neuroprotective properties. The aim of the study was to investigate the neuroprotective role of VEGF-A against oxaliplatin neurotoxicity, attempting to discriminate pain-related and restorative signaling pathways. We used rat organotypic spinal cord slices treated with oxaliplatin, as an in vitro model to study chemotherapy-induced toxicity. In this model, 10 μM oxaliplatin caused a time-dependent release of VEGF-A, which was reduced by the astrocyte inhibitor fluorocitrate. Moreover, glia inhibition exacerbated oxaliplatin-induced cytotoxicity in a VEGF-A sensitive manner. Treatment with VEGF165b, the main isoform of VEGF-A, prevented the oxaliplatin-induced neuronal damage (indicated by NeuN staining) and astrocyte activation (indicated by GFAP staining). In addition, the blockade of VEGFR-2 by the selective antibody DC101 blunted the protective action of VEGF165b. In the same model, VEGF165b increased the release of molecules relevant in pain signaling, like substance P and CGRP, as well as the mRNA expression of glutamate transporters (EAAT1 and EAAT2), similarly to oxaliplatin and these effects were prevented by the selective VEGFR-1 blocker antibody D16F7. In conclusion, VEGF-A plays a dichotomic role in an in vitro model of chemotherapy-induced toxicity, either promoting neuroprotection or triggering pain mediators release, depending on which of its two receptors is activated. The selective management of VEGF-A signaling is suggested as a therapeutic approach.
Collapse
Affiliation(s)
- A Toti
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - E Lucarini
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - V Ferrara
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - C Parisio
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - C Ciampi
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - E Gerace
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Florence, Italy; Department of Health Sciences, University of Florence, Florence, Italy
| | - L Micheli
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Florence, Italy.
| | - F Margiotta
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - D Venturi
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - T Mello
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - P M Lacal
- Laboratory of Molecular Oncology, IDI-IRCCS, Rome, Italy
| | - G Graziani
- Department of Systems Medicine, Pharmacology Section, University of Rome Tor Vergata, Rome, Italy
| | - G Mannaioni
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - C Ghelardini
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - L Di Cesare Mannelli
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| |
Collapse
|
2
|
Liu Y, Ma Q, Yang K, Zhang D, Li F, Chen J, Zhou F, Wang H, Li N, Wang Y, Cao Y, Zhang C, Li X, Zhang H, Wang W, Li Y. Reprogramming macrophage by targeting VEGF and CD40 potentiates OX40 immunotherapy. Biochem Biophys Res Commun 2024; 698:149546. [PMID: 38266314 DOI: 10.1016/j.bbrc.2024.149546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 01/17/2024] [Indexed: 01/26/2024]
Abstract
The low clinical response rate of checkpoint blockades, such as PD-1 and CTLA-4, highlighted the requirements of agonistic antibodies to boost optimal T cell responses. OX40, a co-stimulatory receptor on the T cells, plays a crucial role in promoting T cell survival and differentiation. However, the clinical efficacy of anti-OX40 agonistic antibodies was unimpressive. To explore the mechanism underlying the action of anti-OX40 agonists to improve the anti-tumor efficacy, we analyzed the dynamic changes of tumor-infiltrating immune cells at different days post-treatments using single-cell RNA-sequencing (scRNA-seq). In this study, we found that tumor-infiltrating regulatory T (Treg) cells were reduced after two rounds of anti-OX40 treatment, but the increase of infiltration and activation of CD8+ effector T cells, as well as M1 polarization in the tumor were only observed after three rounds of treatments. Moreover, our group first analyzed the antitumor effect of anti-OX40 treatments on regulating the macrophages and discovered the dynamic changes of vascular endothelial growth factor (VEGF) and CD40 signaling pathways on macrophages, indicating their possibility to being potential combination targets to improve the anti-OX40 agonists efficacy. The combination of VEGFR inhibitors or anti-CD40 agonist antibody with anti-OX40 agonists exhibited more remarkable inhibition of tumor growth. Therefore, the mechanism-driven combination of anti-OX40 agonists with VEGFR inhibitors or anti-CD40 agonists represented promising strategies.
Collapse
Affiliation(s)
- Yanqin Liu
- State Key Laboratory of Medicinal Chemical Biology and College of Life Science, Nankai University, Tianjin, China
| | - Qiongqiong Ma
- State Key Laboratory of Medicinal Chemical Biology and College of Life Science, Nankai University, Tianjin, China
| | - Kailu Yang
- State Key Laboratory of Medicinal Chemical Biology and College of Life Science, Nankai University, Tianjin, China
| | - Dongping Zhang
- State Key Laboratory of Medicinal Chemical Biology and College of Life Science, Nankai University, Tianjin, China
| | - Fan Li
- State Key Laboratory of Medicinal Chemical Biology and College of Life Science, Nankai University, Tianjin, China
| | - Jingru Chen
- State Key Laboratory of Medicinal Chemical Biology and College of Life Science, Nankai University, Tianjin, China
| | - Feilong Zhou
- State Key Laboratory of Medicinal Chemical Biology and College of Life Science, Nankai University, Tianjin, China
| | - Han Wang
- State Key Laboratory of Medicinal Chemical Biology and College of Life Science, Nankai University, Tianjin, China
| | - Na Li
- State Key Laboratory of Medicinal Chemical Biology and College of Life Science, Nankai University, Tianjin, China
| | - Yuan Wang
- Institutes of Biomedical Sciences, Inner Mongolia University, Hohhot, 010020, China
| | - Youjia Cao
- State Key Laboratory of Medicinal Chemical Biology and College of Life Science, Nankai University, Tianjin, China
| | - Cuizhu Zhang
- State Key Laboratory of Medicinal Chemical Biology and College of Life Science, Nankai University, Tianjin, China
| | - Xin Li
- State Key Laboratory of Medicinal Chemical Biology and College of Life Science, Nankai University, Tianjin, China
| | - Hongkai Zhang
- State Key Laboratory of Medicinal Chemical Biology and College of Life Science, Nankai University, Tianjin, China
| | - Wei Wang
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, China
| | - Yuanke Li
- State Key Laboratory of Medicinal Chemical Biology and College of Life Science, Nankai University, Tianjin, China.
| |
Collapse
|
3
|
Chen J, Lin Y, Zheng S, Chen Q, Tang S, Zhong X. CBX3 promotes clear cell renal carcinoma through PI3K/AKT activation and aberrant immunity. J Transl Med 2023; 21:600. [PMID: 37674204 PMCID: PMC10483741 DOI: 10.1186/s12967-023-04478-9] [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: 06/18/2023] [Accepted: 08/27/2023] [Indexed: 09/08/2023] Open
Abstract
BACKGROUND A chromobox homologue 3 (CBX3) is elevated in various cancers and significantly contributes to the promotion of malignant behavior; despite this, its exact involvement in clear cell renal cell carcinoma (ccRCC) is yet unknown. METHODS The Cancer Genome Atlas database served to evaluate CBX3 production and its connection to survival in patients with ccRCC. Our team evaluated the effects of knockdown of CBX3 levels in ccRCC cell populations using in vitro together with in vivo models. CBX3, proteins related to death, and epithelial-to-mesenchymal transition (EMT)-related proteins were measured in ccRCC cells using western blotting and immunohistochemical assays. Through the analysis of Kyoto Encyclopedia of Genes and Genomes (KEGG) and GeneOntology (GO) and Gene Set Enrichment Analysis (GSEA), the biological processes and signal pathways related to CBX3 expression were identified. Immune-related activity reduced by CBX3 was assessed using various online tools. RESULTS Both genomic and protein expression showed that CBX3 was upregulated in ccRCC. Further functional analyses revealed that CBX3 played a crucial role in enhancing cell growth, migration, and EMT in vitro along with in vivo. Moreover, the study results provided distinct mechanistic evidence that CBX3 exerts its pathological functions in ccRCC by activating the PI3K/AKT pathway. Finally, immunoassays revealed that CBX3, a possible biomarker of ccRCC, was significantly associated with immunity. CONCLUSIONS Our results suggest that the overexpression of CBX3 promotes ccRCC advancement through PI3K/AKT activation and even immunological dysregulation, making it a potentially viable and beneficial therapeutic target.
Collapse
Affiliation(s)
- Jiasheng Chen
- Department of Burns and Plastic Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, People's Republic of China
| | - Yuxin Lin
- Department of Burns and Plastic Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, People's Republic of China
| | - Shukai Zheng
- Department of Burns and Plastic Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, People's Republic of China
| | - Qingshan Chen
- Department of Burns and Plastic Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, People's Republic of China
| | - Shijie Tang
- Department of Burns and Plastic Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, People's Republic of China
| | - Xiaoping Zhong
- Department of Burns and Plastic Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, People's Republic of China.
| |
Collapse
|
4
|
Shi C, Li J, Fan G, Liu Y. Blocking CD47 Shows Superior Anti-tumor Therapeutic Effects of Bevacizumab in Gastric Cancer. Front Pharmacol 2022; 13:880139. [PMID: 35694254 PMCID: PMC9175199 DOI: 10.3389/fphar.2022.880139] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 04/08/2022] [Indexed: 11/16/2022] Open
Abstract
Background: Bevacizumab (Avastin®), a humanized antiangiogenic monoclonal antibody, is widely used in the clinical treatment of tumour diseases. However, recent research has shown that the beneficial antiangiogenic effects of these agents have been limited in a number of patients due to complex immunosuppressive mechanisms. Here, we report a synergistic antitumour strategy through simultaneous blockade of VEGF and CD47 signalling to enhance the curative effect of advanced gastric cancer. Method: A BGC-823 gastric tumour model was chosen to evaluate antitumour efficacy. Macrophage migration and phagocytosis were evaluated to determine immune-related resistance to bevacizumab therapy. Synergistic antitumour activity was observed on the basis of tumour volume, tumour weight, tumour inhibition rate, tumour angiogenesis and tumour metastasis when bevacizumab was combined with an anti-CD47 monoclonal antibody. Results: Our study demonstrated that synergistic therapy targeting CD47 and VEGF reversed macrophage migration and phagocytosis, which were inhibited by antiangiogenic therapy and enhanced antitumour effects. Moreover, blockade of CD47 induced by antiangiogenic therapy inhibited tumour metastasis. Conclusion: Our data provide an effective strategy to attenuate resistance to bevacizumab therapy, promoting clinical cancer treatment with antiangiogenic drugs in combination with CD47-targeting inhibitors.
Collapse
Affiliation(s)
- Chenyang Shi
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiaxin Li
- Department of Biochemistry, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Guorong Fan
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Guorong Fan, ; Yu Liu,
| | - Yu Liu
- Department of Biochemistry, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
- *Correspondence: Guorong Fan, ; Yu Liu,
| |
Collapse
|
5
|
Zhang H, Zhu G. Beyond Promoter: The Role of Macrophage in Invasion and Progression of Renal Cell Carcinoma. Curr Stem Cell Res Ther 2021; 15:588-596. [PMID: 32096752 DOI: 10.2174/1574888x15666200225093210] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 08/28/2019] [Accepted: 12/11/2019] [Indexed: 11/22/2022]
Abstract
Renal cell carcinoma (RCC) is one of the common urologic neoplasms, and its incidence has been increasing over the past several decades; however, its pathogenesis is still unknown up to now. Recent studies have found that in addition to tumor cells, other cells in the tumor microenvironment also affect the biological behavior of the tumor. Among them, macrophages exist in a large amount in tumor microenvironment, and they are generally considered to play a key role in promoting tumorigenesis. Therefore, we summarized the recent researches on macrophage in the invasiveness and progression of RCC in latest years, and we also introduced and discussed many studies about macrophage in RCC to promote angiogenesis by changing tumor microenvironment and inhibit immune response in order to activate tumor progression. Moreover, macrophage interactes with various cytokines to promote tumor proliferation, invasion and metastasis, and it also promotes tumor stem cell formation and induces drug resistance in the progression of RCC. The highlight of this review is to make a summary of the roles of macrophage in the invasion and progression of RCC; at the same time to raise some potential and possible targets for future RCC therapy.
Collapse
Affiliation(s)
- Haibao Zhang
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Guodong Zhu
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| |
Collapse
|
6
|
Lin E, Liu X, Liu Y, Zhang Z, Xie L, Tian K, Liu J, Yu Y. Roles of the Dynamic Tumor Immune Microenvironment in the Individualized Treatment of Advanced Clear Cell Renal Cell Carcinoma. Front Immunol 2021; 12:653358. [PMID: 33746989 PMCID: PMC7970116 DOI: 10.3389/fimmu.2021.653358] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 02/12/2021] [Indexed: 02/05/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) are currently a first-line treatment option for clear cell renal cell carcinoma (ccRCC). However, recent clinical studies have shown that a large number of patients do not respond to ICIs. Moreover, only a few patients achieve a stable and durable response even with combination therapy based on ICIs. Available studies have concluded that the response to immunotherapy and targeted therapy in patients with ccRCC is affected by the tumor immune microenvironment (TIME), which can be manipulated by targeted therapy and tumor genomic characteristics. Therefore, an in-depth understanding of the dynamic nature of the TIME is important for improving the efficacy of immunotherapy or combination therapy in patients with advanced ccRCC. Here, we explore the possible mechanisms by which the TIME affects the efficacy of immunotherapy and targeted therapy, as well as the factors that drive dynamic changes in the TIME in ccRCC, including the immunomodulatory effect of targeted therapy and genomic changes. We also describe the progress on novel therapeutic modalities for advanced ccRCC based on the TIME. Overall, this review provides valuable information on the optimization of combination therapy and development of individualized therapy for advanced ccRCC.
Collapse
MESH Headings
- Antineoplastic Combined Chemotherapy Protocols/pharmacology
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Biomarkers, Tumor/antagonists & inhibitors
- Biomarkers, Tumor/genetics
- Carcinoma, Renal Cell/drug therapy
- Carcinoma, Renal Cell/genetics
- Carcinoma, Renal Cell/immunology
- Carcinoma, Renal Cell/mortality
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- Gene Expression Regulation, Neoplastic/drug effects
- Gene Expression Regulation, Neoplastic/immunology
- Humans
- Immune Checkpoint Inhibitors/pharmacology
- Immune Checkpoint Inhibitors/therapeutic use
- Kidney Neoplasms/drug therapy
- Kidney Neoplasms/genetics
- Kidney Neoplasms/immunology
- Kidney Neoplasms/mortality
- Molecular Targeted Therapy/methods
- Precision Medicine/methods
- Progression-Free Survival
- Randomized Controlled Trials as Topic
- Tumor Microenvironment/drug effects
- Tumor Microenvironment/genetics
- Tumor Microenvironment/immunology
Collapse
Affiliation(s)
- Enyu Lin
- Department of Urology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Shantou University Medical College, Shantou, China
| | - Xuechao Liu
- Department of Gastrointestinal Surgery, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yanjun Liu
- Department of Immunology, School of Basic Medical Science, Southern Medical University, Guangzhou, China
| | - Zedan Zhang
- Department of Urology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Shantou University Medical College, Shantou, China
| | - Lu Xie
- Department of Urology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Kaiwen Tian
- Department of Urology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Jiumin Liu
- Department of Urology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yuming Yu
- Department of Urology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| |
Collapse
|
7
|
Santoni M, Massari F, Santoni G, Cimadamore A, Montironi R, Battelli N. Re: Human Chimeric Antigen Receptor Macrophages for Cancer Immunotherapy. Eur Urol 2021; 79:887-889. [PMID: 33549360 DOI: 10.1016/j.eururo.2021.01.025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 01/18/2021] [Indexed: 11/28/2022]
Affiliation(s)
| | - Francesco Massari
- Division of Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Italy
| | | | - Alessia Cimadamore
- Section of Pathological Anatomy, Polytechnic University of the Marche Region, School of Medicine, United Hospitals, Ancona, Italy
| | - Rodolfo Montironi
- Section of Pathological Anatomy, Polytechnic University of the Marche Region, School of Medicine, United Hospitals, Ancona, Italy
| | | |
Collapse
|
8
|
Lymphocytic microparticles suppress retinal angiogenesis via targeting Müller cells in the ischemic retinopathy mouse model. Exp Cell Res 2021; 399:112470. [PMID: 33434529 DOI: 10.1016/j.yexcr.2021.112470] [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: 08/06/2020] [Revised: 12/14/2020] [Accepted: 12/30/2020] [Indexed: 11/22/2022]
Abstract
Retinopathy of prematurity (ROP) is the primary cause of visual impairment and vision loss in premature infants, which results from the formation of aberrant retinal neovascularization (NV). An emerging body of evidence has shown that Müller cells are the predominant source of vascular endothelial growth factor (VEGF), which also serves as a chemoattractant for monocyte/macrophage lineage. The recruitment of macrophages is increased during retinal NV, and they exert a pro-angiogenic role in ROP. We have shown that lymphocytic microparticles (microvesicles; LMPs) derived from apoptotic human T lymphocytes possess strong angiogenesis-inhibiting properties. Here, we investigated the effect of LMPs on the chemotactic capacity of Müller cells in vitro using rat Müller cell rMC-1 and mouse macrophage RAW 264.7. In addition, the impact of LMPs was determined in vivo using a mouse model of oxygen-induced ischemic retinopathy (OIR). The results revealed that LMPs were internalized by rMC-1 and reduced their cell proliferation dose-dependently without inducing cell apoptosis. LMPs inhibited the chemotactic capacity of rMC-1 on RAW 264.7 via reducing the expression of VEGF. Moreover, LMPs attenuated pathological retinal NV and the infiltration of macrophages in vivo. LMPs downregulated ERK1/2 and HIF-1α both in vitro and in vivo. These findings expand our understanding of the effects of LMPs, providing evidence of LMPs as a promising therapeutic approach for the treatment of retinal NV diseases.
Collapse
|
9
|
Liu X, Guo A, Tu Y, Li W, Li L, Liu W, Ju Y, Zhou Y, Sang A, Zhu M. Fruquintinib inhibits VEGF/VEGFR2 axis of choroidal endothelial cells and M1-type macrophages to protect against mouse laser-induced choroidal neovascularization. Cell Death Dis 2020; 11:1016. [PMID: 33247124 PMCID: PMC7695853 DOI: 10.1038/s41419-020-03222-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 11/06/2020] [Accepted: 11/09/2020] [Indexed: 12/12/2022]
Abstract
Wet age-related macular degeneration, which is characterized by choroidal neovascularization (CNV) and induces obvious vision loss. Vascular endothelial growth factor (VEGF) family member VEGF-A (also named as VEGF) and its receptor VEGFR2 contribute to the pathogenesis of CNV. Choroidal endothelial cells (CECs) secret C–C motif chemokine ligand 2 (CCL2), which attracts macrophages to CNV lesion and promotes macrophage M1 polarization. Accordingly, infiltrating macrophages secret inflammatory cytokines to promote CNV. In vivo, intravitreal injection of fruquintinib (HMPL-013), an antitumor neovascularization drug, alleviated mouse CNV formation without obvious ocular toxicity. Meanwhile, HMPL-013 inhibited VEGF/VEGFR2 binding in CECs and macrophages, as well as macrophage M1 polarization. In vitro, noncontact coculture of human choroidal vascular endothelial cells (HCVECs) and macrophages under hypoxia conditions was established. HMPL-013 downregulated VEGF/VEGFR2/phosphoinositide-3-kinase/protein kinase B (AKT)/nuclear factor kappa B pathway and CCL2 secretion in HCVECs, as well as VEGF/VEGFR2-induced macrophage M1 polarization under hypoxia condition. In addition, HMPL-013 inhibited HCEVC derived CCL2-induced macrophage migration and M1 polarization, along with macrophage M1 polarization-induced HCVECs proliferation, migration, and tube formation. Altogether, HMPL-013 alleviated CNV formation might via breaking detrimental cross talk between CECs and macrophages.
Collapse
Affiliation(s)
- Xiaojuan Liu
- Department of Pathogen Biology, Medical College, Nantong University, Nantong, Jiangsu, China
| | - Aisong Guo
- Department of Traditional Chinese Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Yuanyuan Tu
- Department of Ophthalmology, Lixiang Eye Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Wendie Li
- Department of Ophthalmology, Ningbo Eye Hospital, Ningbo, Zhejiang, China
| | - Lele Li
- Department of Ophthalmology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Wangrui Liu
- Department of Neurosurgery, Affiliated Hospital of Youjiang Medical College for Nationalities, Baise, Guangxi, China
| | - Yuanyuan Ju
- Medical College, Nantong University, Nantong, Jiangsu, China
| | - Yamei Zhou
- Medical College, Nantong University, Nantong, Jiangsu, China
| | - Aimin Sang
- Department of Ophthalmology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China.
| | - Manhui Zhu
- Department of Ophthalmology, Lixiang Eye Hospital of Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
10
|
Chen W, Pan X, Cui X. RCC Immune Microenvironment Subsequent to Targeted Therapy: A Friend or a Foe? Front Oncol 2020; 10:573690. [PMID: 33117708 PMCID: PMC7561377 DOI: 10.3389/fonc.2020.573690] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 08/17/2020] [Indexed: 12/30/2022] Open
Abstract
Renal cell carcinoma (RCC) is composed of different subtypes with distinct molecular and histological tumor heterogeneity. Although the advent of various targeted therapies has improved the survival of patients with advanced RCC over the past 15 years (since 2006), few cases experienced complete response due to drug resistance. Recent studies have demonstrated that the outcomes following targeted therapies are potentially associated with intricate cross-links between immune responses and suppressors in the tumor microenvironment (TME). In addition, progress on drug research and development enhances our awareness and understanding about immunotherapy and combined treatment. In this review article, we intend to make a comprehensive summary about TME and its alterations following targeted therapies, provide valid evidence in this aspect, and discuss optimal matches between targeted therapy and immunotherapy.
Collapse
Affiliation(s)
- Wenjin Chen
- Department of Urology, The Third Affiliated Hospital of Second Military Medical University, Shanghai, China
| | - Xiuwu Pan
- Department of Urology, The Third Affiliated Hospital of Second Military Medical University, Shanghai, China
| | - Xingang Cui
- Department of Urology, The Third Affiliated Hospital of Second Military Medical University, Shanghai, China
| |
Collapse
|
11
|
Xu X, Qin L, Yan L. Changes of expression levels of serum cystatin C and soluble vascular endothelial growth factor receptor 1 in the treatment of patients with glomerulus nephritis. Exp Ther Med 2020; 20:1550-1556. [PMID: 32742386 PMCID: PMC7388255 DOI: 10.3892/etm.2020.8824] [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: 06/06/2019] [Accepted: 01/20/2020] [Indexed: 12/05/2022] Open
Abstract
Expression levels and changes of serum cystatin C (C's C) and soluble vascular endothelial growth factor receptor 1 (sVEGFR1) in treatment of patients with glomerulus nephritis (GN) were investigated. The medical records of 88 patients with GN who were diagnosed in Weifang People's Hospital from March 2014 to June 2017 were collected, and their medical records were considered as a study group. The study group was divided into secondary glomerulonephritis (SGN) group (52 cases) and primary glomerulonephritis (PGN) group (36 cases). Physical examination data of 50 healthy volunteers who were examined in the same hospital during the same period were considered as a control group. The correlation between expression of serum C's C and expression of sVEGFR1 of patients with GN was compared. Expression levels of serum C's C and sVEGFR1 of patients before treatment in the study group were higher than those in the control group (P<0.05). With the extension of the treatment cycle, C's C and sVEGFR1 expression levels in PGN and SGN groups reduced gradually (P<0.05). With the extension of the treatment cycle, the renal function indexes of the study group patients showed a downward trend (P<0.05). Expression of C's C was positively correlated with urea nitrogen and creatinine (P<0.05). In conclusion, C's C and sVEGFR1 are highly expressed in the serum of patients with GN. Expression of C's C and sVEGFR1 decrease as patients are treated. C's C and sVEGFR1 can be used as indicators for monitoring the condition of patients with GN. It is worthwhile to promote C's C and sVEGFR1 in clinical practice.
Collapse
Affiliation(s)
- Xinwei Xu
- Nephrology Department, Weifang People's Hospital, Weifang, Shandong 261041, P.R. China
| | - Lili Qin
- Nephrology Department, Weifang People's Hospital, Weifang, Shandong 261041, P.R. China
| | - Liping Yan
- Human Resources Department, Weifang People's Hospital, Weifang, Shandong 261041, P.R. China
| |
Collapse
|
12
|
Lee EK, Konstantinopoulos PA. PARP inhibition and immune modulation: scientific rationale and perspectives for the treatment of gynecologic cancers. Ther Adv Med Oncol 2020; 12:1758835920944116. [PMID: 32782491 PMCID: PMC7383615 DOI: 10.1177/1758835920944116] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 06/30/2020] [Indexed: 12/17/2022] Open
Abstract
Poly[adenosine diphosphate (ADP) ribose]polymerase (PARP) has multifaceted roles in the maintenance of genomic integrity, deoxyribonucleic acid (DNA) repair and replication, and the maintenance of immune-system homeostasis. PARP inhibitors are an attractive oncologic therapy, causing direct cancer cell cytotoxicity by propagating DNA damage and indirectly, by various mechanisms of immunostimulation, including activation of the cGAS/STING pathway, paracrine stimulation of dendritic cells, increased T-cell infiltration, and upregulation of death-ligand receptors to increase susceptibility to natural-killer-cell killing. However, these immunostimulatory effects are counterbalanced by PARPi-mediated upregulation of programmed cell-death-ligand 1 (PD-L1), which leads to immunosuppression. Combining PARP inhibition with immune-checkpoint blockade seeks to exploit the immune stimulatory effects of PARP inhibition while negating the immunosuppressive effects of PD-L1 upregulation.
Collapse
Affiliation(s)
- Elizabeth K Lee
- Department of Medical Oncology, Division of Gynecologic Oncology, Dana-Farber Cancer Institute, 450 Brookline Ave, Boston, MA 02115, USA
| | | |
Collapse
|
13
|
Sha Z, Lai R, Zhang X, Zhao Y, Wu J, Geng C, Guo Z. A Polymorphism at the microRNA Binding Site in the 3' Untranslated Region of KRT81 Is Associated with Breast Cancer. DNA Cell Biol 2020; 39:1886-1894. [PMID: 32678982 DOI: 10.1089/dna.2019.5179] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Single nucleotide polymorphisms in miRNA binding sites (miR-SNPs) are associated with cancer risk. We assessed the relationship between five miR-SNPs in the 3' untranslated region (3'-UTR) of RYR3 (rs1044129), KIAA0423 (rs1053667), C14orf101 (rs4901706), GOLGA7 (rs11337), and KRT81 (rs3660) and the risk of breast cancer (BC). The CC genotype of rs3660 located in the 3'-UTR of KRT81 was identified for its association with lower BC risk (odds ratio, 0.093; 95% confidence interval, 0.045-0.193; p = 0.000). Immunnochemical analysis and Renilla luciferase reporter assays indicated that the CC genotype of KRT81 was associated with lower expression of KRT81 (p < 0.05). The subsequently functional analysis showed that knockdown the KRT81 could inhibit proliferation and promote apoptosis of the MDA-MB-231 BC cells (p < 0.05) with monocyte chemotactic protein-1 (MCP-1) deregulation. Meanwhile, KRT81 overexpression could promote the proliferation and inhibit the apoptosis of MCF-7 BC cells (p < 0.05). Our data demonstrated that the KRT81 expressional change modulated by rs3660 miR-SNP could modify the carcinogenesis of BC, thereby KRT81 would be a new target for BC treatment.
Collapse
Affiliation(s)
- Ziyue Sha
- Department of Immunology and Rheumatology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, P.R. China
| | - Ruixue Lai
- Department of Immunology and Rheumatology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, P.R. China
| | - Xiaoyun Zhang
- Department of Immunology and Rheumatology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, P.R. China
| | - Yufei Zhao
- Department of Immunology and Rheumatology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, P.R. China
| | - Jianhua Wu
- Department of Animal Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, P.R. China
| | - Cuizhi Geng
- Department of Breast Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, P.R. China
| | - Zhanjun Guo
- Department of Immunology and Rheumatology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, P.R. China
| |
Collapse
|
14
|
Heidegger I, Pircher A, Pichler R. Targeting the Tumor Microenvironment in Renal Cell Cancer Biology and Therapy. Front Oncol 2019; 9:490. [PMID: 31259150 PMCID: PMC6587703 DOI: 10.3389/fonc.2019.00490] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 05/23/2019] [Indexed: 12/17/2022] Open
Abstract
Renal cell cancer (RCC) is a highly vascularized and immunogenic tumor type. The inhibition of vessel formation by anti-angiogenic therapies, as well as the stimulation of the immune system by immunotherapy has revolutionized the therapeutic landscape of RCC in recent years. Nevertheless, both therapies are associated with therapy resistance due to a highly dynamic, adaptive and heterogeneous tumor microenvironment (TME). The aim of this short review article is to provide an overview of the components of the RCC TME as well as to discuss their contribution to disease progression. In addition, we report on preclinical and clinical findings and how the different TME components can be modulated to impede treatment progression as well as to overcome therapy resistance to anti-angiogenic or immunomodulating therapy concepts. Furthermore, we discuss the predictive and prognostic role of the TME in RCC therapy. We also report on the concept of combinational targeting of anti-angiogenic therapies and immune checkpoint inhibitor therapy, also including the latest results of clinical studies discussed at recent oncological meetings. Finally, promising new therapeutic targets within the TME are mentioned.
Collapse
Affiliation(s)
- Isabel Heidegger
- Department of Urology, Medical University of Innsbruck, Innsbruck, Austria
| | - Andreas Pircher
- Department of Internal Medicine, Hematology and Oncology, Medical University of Innsbruck, Innsbruck, Austria
| | - Renate Pichler
- Department of Urology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
15
|
Xiao B, Wang W, Zhang D. Risk of bleeding associated with antiangiogenic monoclonal antibodies bevacizumab and ramucirumab: a meta-analysis of 85 randomized controlled trials. Onco Targets Ther 2018; 11:5059-5074. [PMID: 30174444 PMCID: PMC6110629 DOI: 10.2147/ott.s166151] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Aim Bevacizumab and ramucirumab are antiangiogenic monoclonal antibodies, which target vascular endothelial growth factor-A and vascular endothelial growth factor receptor-2, respectively, used in various cancers. Bleeding events have been described with these two agents. We conducted an up-to-date meta-analysis to determine the relative risk (RR) associated with the use of antiangiogenic monoclonal antibodies, bevacizumab and ramucirumab. Methods This meta-analysis of randomized controlled trials was performed after searching PubMed, American Society for Clinical Oncology Abstracts, European Society for Medical Oncology Abstracts, and the proceedings of major conferences for relevant clinical trials. RR and 95% CIs were calculated by random-effects or fixed-effects models for all-grade and high-grade bleeding events related to the angiogenesis inhibitors. Results Eighty-five randomized controlled trials were selected for the meta-analysis, covering 46,630 patients. The results showed that antiangiogenic monoclonal antibodies significantly increased the risk of all-grade (RR: 2.38, 95% CI: 2.09–2.71, p<0.00001) and high-grade (RR: 1.71, 95% CI: 1.48–1.97, p<0.00001) bleeding compared with control arms. In the subgroup analysis, bevacizumab significantly increased the risk of all-grade (RR: 2.73, 95% CI: 2.24–3.33, p<0.00001) and high-grade bleeding (RR: 1.98, 95% CI: 1.68–2.34, p<0.00001), but ramucirumab only increased the risk of all-grade bleeding (RR: 1.94, 95% CI: 1.76–2.13, p<0.00001) and no difference was observed for the risk of high-grade bleeding (RR: 1.04, 95% CI: 0.78–1.39, p=0.79) compared with the control group. For lung cancer patients, bevacizumab significantly increased the risk of all-grade (RR: 4.72, 95% CI: 1.99–11.19, p=0.0004) and high-grade pulmonary hemorrhage (RR: 3.97, 95% CI: 1.70–9.29, p=0.001), but no significant differences in the risk of all-grade (RR: 1.09, 95% CI: 0.76–1.57, p=0.64) and high-grade (RR: 1.22, 95% CI: 0.35–4.21, p=0.75) pulmonary hemorrhage were observed for ramucirumab. The increased risk of all-grade and high-grade bleeding was also observed in colorectal cancer or non-colorectal tumors and low-dose or high-dose angiogenesis inhibitors. Conclusion Antiangiogenic monoclonal antibodies are associated with a significant increase in the risk of all-grade and high-grade bleeding. Ramucirumab may be different from bevacizumab in terms of the risk of high-grade bleeding and the risk of all-grade and high-grade pulmonary hemorrhage in lung cancer patients.
Collapse
Affiliation(s)
- Bingkun Xiao
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, China
| | - Weilan Wang
- Department of Pharmacy, Chinese PLA General Hospital, Beijing, China,
| | - Dezhi Zhang
- Department of Pharmacy, The 264th Hospital of PLA, Taiyuan, Shanxi, China
| |
Collapse
|
16
|
Zizzari IG, Napoletano C, Botticelli A, Caponnetto S, Calabrò F, Gelibter A, Rughetti A, Ruscito I, Rahimi H, Rossi E, Schinzari G, Marchetti P, Nuti M. TK Inhibitor Pazopanib Primes DCs by Downregulation of the β-Catenin Pathway. Cancer Immunol Res 2018; 6:711-722. [PMID: 29700053 DOI: 10.1158/2326-6066.cir-17-0594] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 02/15/2018] [Accepted: 04/06/2018] [Indexed: 11/16/2022]
Abstract
Tyrosine kinase inhibitors (TKIs) target angiogenesis by affecting, for example, the VEGF receptors in tumors and have improved outcomes for patients with metastatic renal cell carcinoma (mRCC). Immune checkpoint inhibitors (ICIs) have also been proposed for treatment of mRCC with encouraging results. A better understanding of the activity of immune cells in mRCC, the immunomodulatory effects of TKIs, and the characteristics defining patients most likely to benefit from various therapies will help optimize immunotherapeutic approaches. In this study, we investigated the influence of the TKI pazopanib on dendritic cell (DC) performance and immune priming. Pazopanib improved DC differentiation and performance by promoting upregulation of the maturation markers HLA-DR, CD40, and CCR7; decreasing IL10 production and endocytosis; and increasing T-cell proliferation. PD-L1 expression was also downregulated. Our results demonstrate that pazopanib inhibits the Erk/β-catenin pathway, suggesting this pathway might be involved in increased DC activation. Similar results were confirmed in DCs differentiated from mRCC patients during pazopanib treatment. In treated patients pazopanib appeared to enhance a circulating CD4+ T-cell population that expresses CD137 (4-1BB). These results suggest that a potentially exploitable immunomodulatory effect induced by pazopanib could improve responses of patients with mRCC in customized protocols combining TKIs with ICI immunotherapy. Cancer Immunol Res; 6(6); 711-22. ©2018 AACR.
Collapse
Affiliation(s)
- Ilaria Grazia Zizzari
- Laboratory of Tumor Immunology and Cell Therapy, Department of Experimental Medicine, Policlinico Umberto I, "Sapienza" University of Rome, Rome, Italy.
| | - Chiara Napoletano
- Laboratory of Tumor Immunology and Cell Therapy, Department of Experimental Medicine, Policlinico Umberto I, "Sapienza" University of Rome, Rome, Italy
| | - Andrea Botticelli
- Division of Oncology, Department of Clinical and Molecular Medicine, Ospedale Sant'Andrea, "Sapienza" University of Rome, Rome, Italy
| | - Salvatore Caponnetto
- Division of Oncology, Department of Radiological, Oncological and Pathological Science, Policlinico Umberto I, "Sapienza" University of Rome, Rome, Italy
| | - Fabio Calabrò
- Division of Medical Oncology B, San Camillo Forlanini Hospital Rome, Rome, Italy
| | - Alain Gelibter
- Division of Oncology, Department of Radiological, Oncological and Pathological Science, Policlinico Umberto I, "Sapienza" University of Rome, Rome, Italy
| | - Aurelia Rughetti
- Laboratory of Tumor Immunology and Cell Therapy, Department of Experimental Medicine, Policlinico Umberto I, "Sapienza" University of Rome, Rome, Italy
| | - Ilary Ruscito
- Laboratory of Tumor Immunology and Cell Therapy, Department of Experimental Medicine, Policlinico Umberto I, "Sapienza" University of Rome, Rome, Italy
| | - Hassan Rahimi
- Laboratory of Tumor Immunology and Cell Therapy, Department of Experimental Medicine, Policlinico Umberto I, "Sapienza" University of Rome, Rome, Italy
| | - Ernesto Rossi
- Department of Medical Oncology, Fondazione Policlinico A. Gemelli Rome, Italy
| | - Giovanni Schinzari
- Department of Medical Oncology, Fondazione Policlinico A. Gemelli Rome, Italy
| | - Paolo Marchetti
- Division of Oncology, Department of Clinical and Molecular Medicine, Ospedale Sant'Andrea, "Sapienza" University of Rome, Rome, Italy
| | - Marianna Nuti
- Laboratory of Tumor Immunology and Cell Therapy, Department of Experimental Medicine, Policlinico Umberto I, "Sapienza" University of Rome, Rome, Italy
| |
Collapse
|
17
|
Graziani G, Ruffini F, Tentori L, Scimeca M, Dorio AS, Atzori MG, Failla CM, Morea V, Bonanno E, D'Atri S, Lacal PM. Antitumor activity of a novel anti-vascular endothelial growth factor receptor-1 monoclonal antibody that does not interfere with ligand binding. Oncotarget 2018; 7:72868-72885. [PMID: 27655684 PMCID: PMC5341950 DOI: 10.18632/oncotarget.12108] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 09/12/2016] [Indexed: 11/29/2022] Open
Abstract
Vascular endothelial growth factor receptor-1 (VEGFR-1) is a tyrosine kinase transmembrane receptor that has also a soluble isoform containing most of the extracellular ligand binding domain (sVEGFR-1). VEGF-A binds to both VEGFR-2 and VEGFR-1, whereas placenta growth factor (PlGF) interacts exclusively with VEGFR-1. In this study we generated an anti-VEGFR-1 mAb (D16F7) by immunizing BALB/C mice with a peptide that we had previously reported to inhibit angiogenesis and endothelial cell migration induced by PlGF. D16F7 did not affect binding of VEGF-A or PlGF to VEGFR-1, thus allowing sVEGFR-1 to act as decoy receptor for these growth factors, but it hampered receptor homodimerization and activation. D16F7 inhibited both the chemotactic response of human endothelial, myelomonocytic and melanoma cells to VEGFR-1 ligands and vasculogenic mimicry by tumor cells. Moreover, D16F7 exerted in vivo antiangiogenic effects in a matrigel plug assay. Importantly, D16F7 inhibited tumor growth and was well tolerated by B6D2F1 mice injected with syngeneic B16F10 melanoma cells. The antitumor effect was associated with melanoma cell apoptosis, vascular abnormalities and decrease of both monocyte/macrophage infiltration and myeloid progenitor mobilization. For all the above, D16F7 may be exploited in the therapy of metastatic melanoma and other tumors or pathological conditions involving VEGFR-1 activation.
Collapse
Affiliation(s)
- Grazia Graziani
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Federica Ruffini
- Laboratory of Molecular Oncology, "Istituto Dermopatico dell'Immacolata"-IRCCS, Rome, Italy
| | - Lucio Tentori
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Manuel Scimeca
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Annalisa S Dorio
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | | | - Cristina M Failla
- Laboratory of Experimental Immunology, "Istituto Dermopatico dell'Immacolata"-IRCCS, Rome, Italy
| | - Veronica Morea
- National Research Council of Italy (CNR), Institute of Molecular Biology and Pathology, Rome, Italy
| | - Elena Bonanno
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Stefania D'Atri
- Laboratory of Molecular Oncology, "Istituto Dermopatico dell'Immacolata"-IRCCS, Rome, Italy
| | - Pedro M Lacal
- Laboratory of Molecular Oncology, "Istituto Dermopatico dell'Immacolata"-IRCCS, Rome, Italy
| |
Collapse
|
18
|
Wheeler KC, Jena MK, Pradhan BS, Nayak N, Das S, Hsu CD, Wheeler DS, Chen K, Nayak NR. VEGF may contribute to macrophage recruitment and M2 polarization in the decidua. PLoS One 2018; 13:e0191040. [PMID: 29324807 PMCID: PMC5764356 DOI: 10.1371/journal.pone.0191040] [Citation(s) in RCA: 166] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 12/27/2017] [Indexed: 01/25/2023] Open
Abstract
It is increasingly evident that cytokines and growth factors produced in the decidua play a pivotal role in the regulation of the local immune microenvironment and the establishment of pregnancy. One of the major growth factors produced in the decidua is vascular endothelial growth factor (VEGF), which acts not only on endothelial cells, but also on multiple other cell types, including macrophages. We sought to determine whether decidua-derived VEGF affects macrophage recruitment and polarization using human endometrial/decidual tissue samples, primary human endometrial stromal cells (ESCs), and the human monocyte cell line THP1. In situ hybridization was used for assessment of local VEGF expression and immunohistochemistry was used for identification and localization of CD68-positive endometrial macrophages. Macrophage migration in culture was assessed using a transwell migration assay, and the various M1/M2 phenotypic markers and VEGF expression were assessed using quantitative real-time PCR (qRT-PCR). We found dramatic increases in both VEGF levels and macrophage numbers in the decidua during early pregnancy compared to the secretory phase endometrium (non-pregnant), with a significant increase in M2 macrophage markers, suggesting that M2 is the predominant macrophage phenotype in the decidua. However, decidual samples from preeclamptic pregnancies showed a significant shift in macrophage phenotype markers, with upregulation of M1 and downregulation of M2 markers. In THP1 cultures, VEGF treatment significantly enhanced macrophage migration and induced M1 macrophages to shift to an M2 phenotype. Moreover, treatment with conditioned media from decidualized ESCs induced changes in macrophage migration and polarization similar to that of VEGF treatment. These effects were abrogated by the addition of a potent VEGF inhibitor. Together these results suggest that decidual VEGF plays a significant role in macrophage recruitment and M2 polarization, and that inhibition of VEGF signaling may contribute to the shift in macrophage polarity observed in different pregnancy disorders, including preeclampsia.
Collapse
Affiliation(s)
- Karen C. Wheeler
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, Michigan, United States of America
| | - Manoj K. Jena
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, Michigan, United States of America
- Department of Biotechnology, School of Bioengineering and Biosciences, Lovely Professional University (LPU), Phagwara, Punjab, India
| | - Bhola S. Pradhan
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, Michigan, United States of America
| | - Neha Nayak
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, Michigan, United States of America
| | - Subhendu Das
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, Michigan, United States of America
| | - Chaur-Dong Hsu
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, Michigan, United States of America
| | - David S. Wheeler
- Division of Pulmonary and Critical Care Medicine, University of Michigan Health System, Ann Arbor, Michigan, United States of America
| | - Kang Chen
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, Michigan, United States of America
| | - Nihar R. Nayak
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, Michigan, United States of America
- * E-mail:
| |
Collapse
|
19
|
Atzori MG, Tentori L, Ruffini F, Ceci C, Bonanno E, Scimeca M, Lacal PM, Graziani G. The Anti-Vascular Endothelial Growth Factor Receptor-1 Monoclonal Antibody D16F7 Inhibits Glioma Growth and Angiogenesis In Vivo. J Pharmacol Exp Ther 2018; 364:77-86. [PMID: 29025978 DOI: 10.1124/jpet.117.244434] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 10/10/2017] [Indexed: 03/08/2025] Open
Abstract
The vascular endothelial growth factor (VEGF) receptor-1 (VEGFR-1) is a tyrosine kinase receptor that does not play a relevant role in physiologic angiogenesis in adults, whereas it is important in tumor angiogenesis. In high-grade glioma VEGFR-1 expression by tumor endothelium and neoplastic cells contributes to the aggressive phenotype. We recently generated an anti-VEGFR-1 monoclonal antibody (D16F7 mAb) characterized by a novel mechanism of action, since it hampers receptor activation without interfering with ligand binding. The mAb is able to inhibit chemotaxis and extracellular matrix invasion of glioma cells in vitro stimulated by VEGF-A and by the VEGFR-1-selective ligand placental growth factor (PlGF). In this study, we have investigated the influence of D16F7 on glioma growth and angiogenesis in vivo using C6 glioma cells transfected with the human VEGFR-1. D16F7 was able to inhibit receptor activation and migration and extracellular matrix invasion of C6 cells overexpressing the receptor in response to PlGF and VEGF-A. In nude mice, treatment with 10 and 20 mg/kg D16F7 as a single agent was well tolerated and significantly inhibited glioma growth (P < 0.001). Strikingly, in an intracranial orthotopic model, mice dosed with 20 mg/kg D16F7 demonstrated a 65% increase in median survival time compared with vehicle-treated controls (P < 0.001) with a high percentage of long-term survivors (46%). These effects were associated with glioma cell apoptosis and decreased tumor-associated vessel formation. Overall, these results highlight the therapeutic potential of D16F7 in glioma treatment, deserving further investigation after a humanization process as single agent or in combination therapies.
Collapse
Affiliation(s)
- Maria Grazia Atzori
- Departments of Systems Medicine (M.G.A., L.T., C.C., G.G.) and Experimental Medicine and Surgery (E.B., M.S.), University of Rome Tor Vergata, Rome, Italy; and Laboratory of Molecular Oncology, "Istituto Dermopatico dell'Immacolata"-Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy (F.R., P.M.L.)
| | - Lucio Tentori
- Departments of Systems Medicine (M.G.A., L.T., C.C., G.G.) and Experimental Medicine and Surgery (E.B., M.S.), University of Rome Tor Vergata, Rome, Italy; and Laboratory of Molecular Oncology, "Istituto Dermopatico dell'Immacolata"-Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy (F.R., P.M.L.)
| | - Federica Ruffini
- Departments of Systems Medicine (M.G.A., L.T., C.C., G.G.) and Experimental Medicine and Surgery (E.B., M.S.), University of Rome Tor Vergata, Rome, Italy; and Laboratory of Molecular Oncology, "Istituto Dermopatico dell'Immacolata"-Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy (F.R., P.M.L.)
| | - Claudia Ceci
- Departments of Systems Medicine (M.G.A., L.T., C.C., G.G.) and Experimental Medicine and Surgery (E.B., M.S.), University of Rome Tor Vergata, Rome, Italy; and Laboratory of Molecular Oncology, "Istituto Dermopatico dell'Immacolata"-Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy (F.R., P.M.L.)
| | - Elena Bonanno
- Departments of Systems Medicine (M.G.A., L.T., C.C., G.G.) and Experimental Medicine and Surgery (E.B., M.S.), University of Rome Tor Vergata, Rome, Italy; and Laboratory of Molecular Oncology, "Istituto Dermopatico dell'Immacolata"-Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy (F.R., P.M.L.)
| | - Manuel Scimeca
- Departments of Systems Medicine (M.G.A., L.T., C.C., G.G.) and Experimental Medicine and Surgery (E.B., M.S.), University of Rome Tor Vergata, Rome, Italy; and Laboratory of Molecular Oncology, "Istituto Dermopatico dell'Immacolata"-Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy (F.R., P.M.L.)
| | - Pedro Miguel Lacal
- Departments of Systems Medicine (M.G.A., L.T., C.C., G.G.) and Experimental Medicine and Surgery (E.B., M.S.), University of Rome Tor Vergata, Rome, Italy; and Laboratory of Molecular Oncology, "Istituto Dermopatico dell'Immacolata"-Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy (F.R., P.M.L.)
| | - Grazia Graziani
- Departments of Systems Medicine (M.G.A., L.T., C.C., G.G.) and Experimental Medicine and Surgery (E.B., M.S.), University of Rome Tor Vergata, Rome, Italy; and Laboratory of Molecular Oncology, "Istituto Dermopatico dell'Immacolata"-Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy (F.R., P.M.L.)
| |
Collapse
|
20
|
Jackaman C, Tomay F, Duong L, Abdol Razak NB, Pixley FJ, Metharom P, Nelson DJ. Aging and cancer: The role of macrophages and neutrophils. Ageing Res Rev 2017; 36:105-116. [PMID: 28390891 DOI: 10.1016/j.arr.2017.03.008] [Citation(s) in RCA: 175] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 03/30/2017] [Accepted: 03/30/2017] [Indexed: 12/12/2022]
Abstract
Impaired immune function has been implicated in the declining health and higher incidence of cancer in the elderly. However, age-related changes to immunity are not completely understood. Neutrophils and macrophages represent the first line of defence yet their ability to phagocytose pathogens decrease with aging. Cytotoxic T lymphocytes are critical in eliminating tumors, but T cell function is also compromised with aging. T cell responses can be regulated by macrophages and may depend on the functional phenotype macrophages adopt in response to microenvironmental signals. This can range from pro-inflammatory, anti-tumorigenic M1 to anti-inflammatory, pro-tumorigenic M2 macrophages. Macrophages in healthy elderly adipose and hepatic tissue exhibit a more pro-inflammatory M1 phenotype compared to young hosts whilst immunosuppressive M2 macrophages increase in elderly lymphoid tissues, lung and muscle. These M2-like macrophages demonstrate altered responses to stimuli. Recent studies suggest that neutrophils also regulate T cell function and, like macrophages, neutrophil function is modulated with aging. It is possible that age-modified tissue-specific macrophages and neutrophils contribute to chronic low-grade inflammation that is associated with dysregulated macrophage-mediated immunosuppression, which together are responsible for development of multiple pathologies, including cancer. This review discusses recent advances in macrophage and neutrophil biology in healthy aging and cancer.
Collapse
Affiliation(s)
- Connie Jackaman
- School of Biomedical Sciences, Curtin Health and Innovation Research Institute, Faculty of Health Sciences, Curtin University, Bentley, Western Australia, 6102, Australia.
| | - Federica Tomay
- School of Biomedical Sciences, Curtin Health and Innovation Research Institute, Faculty of Health Sciences, Curtin University, Bentley, Western Australia, 6102, Australia
| | - Lelinh Duong
- School of Biomedical Sciences, Curtin Health and Innovation Research Institute, Faculty of Health Sciences, Curtin University, Bentley, Western Australia, 6102, Australia
| | - Norbaini Bintu Abdol Razak
- School of Biomedical Sciences, Curtin Health and Innovation Research Institute, Faculty of Health Sciences, Curtin University, Bentley, Western Australia, 6102, Australia
| | - Fiona J Pixley
- School of Medicine and Pharmacology, University of Western Australia, Nedlands, Western Australia, 6009, Australia
| | - Pat Metharom
- School of Biomedical Sciences, Curtin Health and Innovation Research Institute, Faculty of Health Sciences, Curtin University, Bentley, Western Australia, 6102, Australia
| | - Delia J Nelson
- School of Biomedical Sciences, Curtin Health and Innovation Research Institute, Faculty of Health Sciences, Curtin University, Bentley, Western Australia, 6102, Australia
| |
Collapse
|
21
|
Cai B, Xiao Y, Li Y, Zheng S. CMTM5 inhibits renal cancer cell growth through inducing cell-cycle arrest and apoptosis. Oncol Lett 2017; 14:1536-1542. [PMID: 28789377 PMCID: PMC5529942 DOI: 10.3892/ol.2017.6350] [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: 09/26/2015] [Accepted: 04/04/2017] [Indexed: 12/31/2022] Open
Abstract
CKLF-like MARVEL transmembrane domain-containing 5 (CMTM5) has been reported to function as a potential tumor suppressor in several human cancers. However, the involvement of CMTM5 in human renal cell carcinoma (RCC) remains unclear. The current study aimed to detect its expression pattern in RCC tissues and cells, and to determine its anti-proliferative functions in this malignancy. The mRNA and protein expression levels of CMTM5 in RCC tissues and cells were detected by reverse transcription-quantitative polymerase chain reaction, immunohistochemistry and western blotting. Following the transfection with CMTM5 lentivirus or control lenti-EGFP lentivirus into the RCC cell line ACHN, the viability, migration, apoptosis and cell cycle of these cells were detected by Cell Counting kit-8 assay, Transwell assay and flow cytometry, respectively. Compared with the adjacent non-malignant kidney tissue samples, CMTM5 expression was significantly downregulated in RCC tissues (P<0.05). In addition, enforced expression of CMTM5 could efficiently inhibit the cell growth of ACHN cells, which were arrested in G0/G1 phase. Furthermore, the migration and invasion of ACHN cells were also inhibited by restoration of CMTM5 expression. The present data suggest that CMTM5 may function as a tumor suppressor in human RCC by suppressing the viability of RCC cells, implying its potential as a therapeutic target for this malignancy.
Collapse
Affiliation(s)
- Bing Cai
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China.,Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Yunbei Xiao
- Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Yeping Li
- Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Shaobin Zheng
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| |
Collapse
|
22
|
Tumor Associated Macrophages in Kidney Cancer. Anal Cell Pathol (Amst) 2016; 2016:9307549. [PMID: 27807511 PMCID: PMC5078639 DOI: 10.1155/2016/9307549] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2016] [Accepted: 09/20/2016] [Indexed: 11/17/2022] Open
Abstract
Tumor associated macrophages (TAMs) are an important element of tumor stroma. They originate from blood monocytes attracted by chemokines and cytokines produced by tumor cells and, being instructed by tumor microenvironment, develop into potent tumor-supporting cell population. TAMs were demonstrated to directly stimulate tumor cell proliferation and to promote angiogenesis. Further TAMs provide for efficient immune escape by producing immunosuppressive cytokines and facilitate tumor dissemination by producing extracellular matrix remodeling enzymes. In renal cell carcinoma (RCC), numerous studies were performed for elucidation of the role of TAM in tumor progression. Using pan-macrophages marker CD68 and type 2 macrophage (M2) markers CD163 and CD206, it was demonstrated that increased density of TAMs is associated with poor survival of patients. Although most of the studies are focused on M2 population in RCC, several markers rather typical for type 1 macrophages (M1) were also characterized. Macrophages isolated from RCC tumors were shown to produce proinflammatory cytokines TNFα, IL-1β, IL-6, and CCL2. It can be concluded that RCC is an excellent example of a tumor with hybrid phenotype of TAMs that share both M1 and M2 properties. Moreover, TAMs seem to be an attractive therapeutic target as well. Further investigations are needed for identification of RCC-specific TAM markers with high predictive capacity and/or suitable for therapeutic targeting.
Collapse
|
23
|
Klatt MG, Kowalewski DJ, Schuster H, Di Marco M, Hennenlotter J, Stenzl A, Rammensee HG, Stevanović S. Carcinogenesis of renal cell carcinoma reflected in HLA ligands: A novel approach for synergistic peptide vaccination design. Oncoimmunology 2016; 5:e1204504. [PMID: 27622074 PMCID: PMC5007970 DOI: 10.1080/2162402x.2016.1204504] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 06/15/2016] [Accepted: 06/17/2016] [Indexed: 01/08/2023] Open
Abstract
Despite recent advances in immunotherapy of renal cell carcinoma (RCC), peptide vaccination strategies still lack an approach for personalized peptide vaccination that takes intra- and inter-tumoral heterogeneity and biological characteristics into account. In this study, we use an immunoprecipitation and mass spectrometry-based approach supplemented by network analysis of HLA ligands to target this goal. By analyzing HLA-presented peptides for HLA class I and II of 11 malignant and 6 non-malignant kidney tissue samples, more than 2,700 peptides and 1,600 corresponding source proteins were identified. A high overlap with HLA ligands derived from peripheral blood mononuclear cells (PBMCs) was detected most likely due to tumor-infiltrating inflammatory cells and therefore excluded from network analysis. Subsequent biological function analysis of HLA ligands by the GeneMANIA online platform showed enrichment for well established, but also novel, pathways and biological processes involved in carcinogenesis of RCC almost exclusively in malignant tissue samples. By exploring source proteins involved in these overrepresented pathways, we verified various known tumor-associated antigens (TAAs) for RCC (e.g., CA9, EGLN3, IGFBP3, MMP7, PAX2, VEGFA, or EGFR) but could also detect novel TAAs for RCC (e.g., PLOD2, LOX, ENPEP, or TGFBI). Some of these new TAAs had already been shown to elicit a T cell response in cancer patients. Thus, network analysis of HLA ligands provided a new platform for implementing personalized, multipeptide vaccines with potentially synergistic antitumor effects.
Collapse
Affiliation(s)
- Martin G Klatt
- Department of Hematology and Oncology, University of Tübingen , Tübingen, Germany
| | - Daniel J Kowalewski
- Institute for Cell Biology, Department of Immunology, University of Tübingen, Tübingen, Germany; German Cancer Consortium (DKTK), DKFZ Partner Site, Tübingen, Germany
| | - Heiko Schuster
- Institute for Cell Biology, Department of Immunology, University of Tübingen, Tübingen, Germany; German Cancer Consortium (DKTK), DKFZ Partner Site, Tübingen, Germany
| | - Moreno Di Marco
- Institute for Cell Biology, Department of Immunology, University of Tübingen, Tübingen, Germany; German Cancer Consortium (DKTK), DKFZ Partner Site, Tübingen, Germany
| | | | - Arnulf Stenzl
- Department of Urology, University of Tübingen , Tübingen, Germany
| | - Hans-Georg Rammensee
- Institute for Cell Biology, Department of Immunology, University of Tübingen , Tübingen, Germany
| | - Stefan Stevanović
- Institute for Cell Biology, Department of Immunology, University of Tübingen , Tübingen, Germany
| |
Collapse
|
24
|
Abstract
Vascular endothelial growth factor (VEGF) is primarily known as a proangiogenic factor and is one of the most important growth and survival factors affecting the vascular endothelium. However, recent studies have shown that VEGF also plays a vital role in the immune environment. In addition to the traditional growth factor role of VEGF and VEGF receptors (VEGFRs), they have a complicated relationship with various immune cells. VEGF also reportedly inhibits the differentiation and function of immune cells during hematopoiesis. Dendritic cells (DCs), macrophages, and lymphocytes further express certain types of VEGF receptors. VEGF can be secreted as well by tumor cells through the autocrine pathway and can stimulate the function of cancer stemness. This review will provide a paradigm shift in our understanding of the role of VEGF/VEGFR signaling in the immune and cancer environment.
Collapse
Affiliation(s)
- Yu-Ling Li
- Department of Biotherapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China
| | - Hua Zhao
- Department of Biotherapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China; Key Laboratory of Cancer Immunology and Biotherapy, Research Center of Lung Cancer, Tianjin 300060, China
| | - Xiu-Bao Ren
- Department of Biotherapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China; Key Laboratory of Cancer Immunology and Biotherapy, Research Center of Lung Cancer, Tianjin 300060, China
| |
Collapse
|
25
|
Küper C, Beck FX, Neuhofer W. Autocrine MCP-1/CCR2 signaling stimulates proliferation and migration of renal carcinoma cells. Oncol Lett 2016; 12:2201-2209. [PMID: 27602164 DOI: 10.3892/ol.2016.4875] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 05/13/2016] [Indexed: 12/31/2022] Open
Abstract
The chemokine monocyte chemoattractant protein-1 [MCP-1; also known as chemokine (C-C motif) ligand 2] is an important mediator of monocyte recruitment during inflammatory processes. Pathologically high expression levels of MCP-1 by tumor cells have been observed in a variety of cancer types. In the majority of cases, high MCP-1 expression is associated with a poor prognosis, as infiltration of the tumor with inflammatory monocytes promotes tumor progression and metastasis. MCP-1 is also expressed in renal cell carcinoma (RCC). In the present study, the function and the regulation of MCP-1 was investigated in two RCC cell lines, CaKi-1 and 786-O. In both cell lines, expression of MCP-1 was significantly enhanced compared with non-cancerous control cells. As expected, secretion of MCP-1 into the medium facilitated the recruitment of peripheral blood monocytes via the chemokine (C-C motif) receptor type 2 (CCR2). As expression of CCR2 was also detected in 786-O and CaKi-1 cells, the effect of autocrine MCP-1/CCR2 signaling was evaluated in these cells. In proliferation assays, administration of an MCP-1 neutralizing antibody or of a CCR2 antagonist to CaKi-1 and 786-O cells significantly decreased cell growth; supplementation of the growth medium with recombinant human MCP-1 had no additional effect on proliferation. The migration ability of RCC cells was impaired by MCP-1 neutralization or pharmacological CCR2 inhibition, while it was stimulated by the addition of recombinant human MCP-1, compared with untreated control cells. Finally, substantial differences in the regulation of MCP-1 expression were observed between RCC cell lines. In CaKi-1 cells, expression of MCP-1 appears to be largely mediated by the transcription factor nuclear factor of activated T cells 5, while in 786-O cells, deletion of the tumor suppressor gene Von-Hippel-Lindau appeared to be responsible for MCP-1 upregulation, as suggested by previous studies. Taken together, the results of the current study indicate that expression of MCP-1 in RCC cells promotes tumor progression and metastasis not only by paracrine, but also by autocrine, MCP-1/CCR2 signaling events, enhancing cell proliferation and migration ability. Therefore, the present findings suggest the MCP-1/CCR2 axis is a potential target for future therapeutic strategies in the treatment of metastatic RCC.
Collapse
Affiliation(s)
- Christoph Küper
- Department of Physiology, University of Munich, D-80336 Munich, Germany
| | - Franz-Xaver Beck
- Department of Physiology, University of Munich, D-80336 Munich, Germany
| | - Wolfgang Neuhofer
- Division of Nephrology and Rheumatology, Clinical Center Traunstein, D-83278 Traunstein, Germany
| |
Collapse
|
26
|
Arantes RVN, Cestari TM, Viscelli BA, Dionísio TJ, Garlet GP, Santos CF, de Assis GF, Taga R. Meloxicam temporally inhibits the expression of vascular endothelial growth factor receptor (VEGFR)-1 and VEGFR-2 during alveolar bone repair in rats. J Periodontol 2016; 86:162-72. [PMID: 25327303 DOI: 10.1902/jop.2014.140259] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND Vascular endothelial growth factor (VEGF) plays an important role during angiogenesis and bone repair. This study investigated whether the use of meloxicam alters bone repair via downregulation of VEGF and receptor expression. METHODS One hundred twenty male Wistar rats had their maxillary right incisor extracted. Animals were divided into a control group (CG; n = 60) and a meloxicam-treated group (TG; n = 60) that received either a single daily intraperitoneal injection of 0.9% NaCl or meloxicam 3 mg/kg, respectively, for 7 consecutive days. Alveolar bone repair was evaluated histomorphometrically, whereas VEGF and its receptors were analyzed by immunohistochemistry and quantitative polymerase chain reaction (qPCR). Data were submitted to two-way analysis of variance and Tukey post hoc test with P < 0.05. RESULTS Bone volume density increased significantly (P = 0.001) in both groups with a strong correlation between treatment and periods (P = 0.003). In the TG, a small amount of bone formation occurred compared with the CG between 3 and 21 days. No significant differences in the number of VEGF-positive cells per square millimeter (P = 0.07) and VEGF messenger RNA (mRNA) expression (P = 0.49) were found between groups. Immunostained cells per square millimeter and mRNA expression for VEGF receptor (VEGFR)-1 (P = 0.04 and P < 0.001) and VEGFR-2 (P < 0.001 for both analysis) showed a strong interaction between treatment groups and periods. In the TG, immunostained cells per square millimeter and mRNA expression for VEGFR-1 were, respectively, 89% and 37% lower from 3 to 10 days compared with the CG, whereas for VEGFR-2, these values were 252% and 60%, respectively, from 3 to 7 days. CONCLUSION In rat alveolar bone repair, meloxicam did not affect VEGF expression but downregulated VEGFR expression, which may cause a delay in the bone repair/remodeling process.
Collapse
|
27
|
Parris GE. A Hypothesis Concerning the Biphasic Dose-response of Tumors to Angiostatin and Endostatin. Dose Response 2015; 13:10.2203_dose-response.14-020.Parris. [PMID: 26675544 PMCID: PMC4674172 DOI: 10.2203/dose-response.14-020.parris] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
This manuscript proposes a hypothesis to explain the U-shaped dose-response observed for angiostatin and other high-molecular-weight drugs in various anti-cancer bio-assays. The dose-response curves for angiostatin and endostatin (measured as suppression of tumor growth) go through an optimum (i.e., minimum tumor growth) and then becomes less effective at higher doses. The literature suggests that at lower doses the primary action of these high-molecular-weight drugs is to counteract the angiogenic effects of vascular endothelial growth factor (VEGF). To do this, the drugs must pass out of the blood vessel and enter the extra-cellular matrix (ECM) where VEGF induces the growth and fusion of tip cells. Ironically, VEGF actually facilitates access of the drugs to the ECM by making the vascular endothelium leaky. At higher doses, the high-molecular-weight drugs seem to reverse VEGF-induced permeability of the endothelium. Thus, at high dose rates, it is hypothesized that the drugs are not able to enter the ECM and block the angiogenic effects of VEGF there. As a result, high doses of the drugs do not suppress vascularization of the tumor or tumor growth. Moreover, if the permeability of the vessels is suppressed, the VEGF released by the stroma is concentrated in the ECM where it amplifies the angiogenic activity around the tumor.
Collapse
Affiliation(s)
- George E Parris
- Montgomery College, Department of Chemistry, Rockville, MD, USA
| |
Collapse
|
28
|
Mickley A, Kovaleva O, Kzhyshkowska J, Gratchev A. Molecular and immunologic markers of kidney cancer-potential applications in predictive, preventive and personalized medicine. EPMA J 2015; 6:20. [PMID: 26500709 PMCID: PMC4617448 DOI: 10.1186/s13167-015-0042-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 09/18/2015] [Indexed: 12/12/2022]
Abstract
Kidney cancer is one of the deadliest malignancies due to frequent late diagnosis (33 % or renal cell carcinoma are metastatic at diagnosis) and poor treatment options. There are two major subtypes of kidney cancer: renal cell carcinoma (RCC) and renal pelvis carcinoma. The risk factors for RCC, accounting for more than 90 % of all kidney cancers, are smoking, obesity, hypertension, misuse of pain medication, and some genetic diseases. The most common molecular markers of kidney cancer include mutations and epigenetic inactivation of von Hippel-Lindau (VHL) gene, genes of vascular endothelial growth factor (VEGF) pathway, and carbonic anhydrase IX (CIAX). The role of epigenetic pathways, including DNA methylation and chromatin structure remodeling, was also demonstrated. Immunologic properties of RCC enable this type of tumor to escape immune response effectively. An important role in this process is played by tumor-associated macrophages that demonstrate mixed M1/M2 phenotype. In this review, we discuss molecular and cellular aspects for RCC development and current state of knowledge allowing personalized approaches for diagnostics and prognostic prediction of this disease. A set of macrophage markers is suggested for the analysis of the association of macrophage phenotype and disease prognosis.
Collapse
Affiliation(s)
- Amanda Mickley
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Ruprecht-Karls University of Heidelberg, Mannheim, Germany
| | | | - Julia Kzhyshkowska
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Ruprecht-Karls University of Heidelberg, Mannheim, Germany ; Red Cross Blood Service Baden-Württemberg-Hessen, Mannheim, Germany ; Laboratory for Translational Cellular and Molecular Biomedicine, Tomsk State University, Tomsk, Russia
| | - Alexei Gratchev
- Blokhin Cancer Research Center, Moscow, Russia ; Laboratory for Translational Cellular and Molecular Biomedicine, Tomsk State University, Tomsk, Russia ; Laboratory of the Tumour Stromal Cells Biology, Institute of Carcinogenesis, Blokhin Cancer Research Center, Kashirskoye Shosse 24, Moscow, Russia
| |
Collapse
|
29
|
Haddad AQ, Margulis V. Tumour and patient factors in renal cell carcinoma-towards personalized therapy. Nat Rev Urol 2015; 12:253-62. [PMID: 25868564 DOI: 10.1038/nrurol.2015.71] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Renal cell carcinoma (RCC) comprises a heterogeneous group of histologically and molecularly distinct tumour subtypes. Current targeted therapies have improved survival in patients with advanced disease but complete response occurs rarely, if at all. The genomic characterization of RCC is central to the development of novel targeted therapies. Large-scale studies employing multiple 'omics' platforms have led to the identification of key driver genes and commonly altered pathways. Specific molecular alterations and signatures that correlate with tumour phenotype and clinical outcome have been identified and can be harnessed for patient management and counselling. RCC seems to be a remarkably diverse malignancy with significant intratumour and intertumour genetic heterogeneity. The tumour microenvironment is increasingly recognized as a vital regulator of RCC tumour biology. Patient factors, including immune response and drug metabolism, vary widely, which can lead to widely divergent responses to drug therapy. Intratumour heterogeneity poses a significant challenge to the development of personalized therapies in RCC as a single biopsy might not accurately represent the clonal population ultimately responsible for aggressive biologic behaviour. On the other hand, the diversity of genomic alterations in RCC could also afford opportunities for targeting unique pathways based on analysis of an individual tumour's molecular composition.
Collapse
Affiliation(s)
- Ahmed Q Haddad
- Department of Urology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Vitaly Margulis
- Department of Urology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| |
Collapse
|
30
|
Caronni N, Savino B, Bonecchi R. Myeloid cells in cancer-related inflammation. Immunobiology 2014; 220:249-53. [PMID: 25454487 DOI: 10.1016/j.imbio.2014.10.001] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Revised: 10/03/2014] [Accepted: 10/05/2014] [Indexed: 12/26/2022]
Abstract
Myeloid cells are key elements of the cancer-related inflammation with the potential to support not only tumor growth but also invasion and metastasis. Tumor-derived factors affect myeloid cell differentiation inducing a phenotype that supports tumor growth, inducing immunosuppression, angiogenesis and tissue remodeling. Soluble mediators, produced at primary tumor site, can also act in a remote mode inducing the release from bone marrow of myeloid cells that have immunosuppressive activities in tumor-draining lymphoid organs and can predispose to colonization when migrate to metastatic organs. We will here review current knowledge on the contribution of tumor-derived signals that affect polarized activation of myeloid cells, their bone marrow release and recruitment to metastatic sites with a particular focus on the role of chemokines.
Collapse
Affiliation(s)
- Nicoletta Caronni
- Humanitas Clinical and Research Center, Italy; Department of Medical Biotechnologies and Translational Medicine, Università degli Studi di Milano, Rozzano, Italy
| | - Benedetta Savino
- Humanitas Clinical and Research Center, Italy; Department of Medical Biotechnologies and Translational Medicine, Università degli Studi di Milano, Rozzano, Italy.
| | - Raffaella Bonecchi
- Humanitas Clinical and Research Center, Italy; Department of Medical Biotechnologies and Translational Medicine, Università degli Studi di Milano, Rozzano, Italy
| |
Collapse
|
31
|
Martínez-Bosch N, Iglesias M, Munné-Collado J, Martínez-Cáceres C, Moreno M, Guerra C, Yélamos J, Navarro P. Parp-1 genetic ablation in Ela-myc mice unveils novel roles for Parp-1 in pancreatic cancer. J Pathol 2014; 234:214-27. [PMID: 24889936 DOI: 10.1002/path.4384] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Revised: 05/15/2014] [Accepted: 05/28/2014] [Indexed: 12/21/2022]
Abstract
Pancreatic cancer has a dismal prognosis and is currently the fourth leading cause of cancer-related death in developed countries. The inhibition of poly(ADP-ribose) polymerase-1 (Parp-1), the major protein responsible for poly(ADP-ribosy)lation in response to DNA damage, has emerged as a promising treatment for several tumour types. Here we aimed to elucidate the involvement of Parp-1 in pancreatic tumour progression. We assessed Parp-1 protein expression in normal, preneoplastic and pancreatic tumour samples from humans and from K-Ras- and c-myc-driven mouse models of pancreatic cancer. Parp-1 was highly expressed in acinar cells in normal and cancer tissues. In contrast, ductal cells expressed very low or undetectable levels of this protein, both in a normal and in a tumour context. The Parp-1 expression pattern was similar in human and mouse samples, thereby validating the use of animal models for further studies. To determine the in vivo effects of Parp-1 depletion on pancreatic cancer progression, Ela-myc-driven pancreatic tumour development was analysed in a Parp-1 knock-out background. Loss of Parp-1 resulted in increased tumour necrosis and decreased proliferation, apoptosis and angiogenesis. Interestingly, Ela-myc:Parp-1(-/-) mice displayed fewer ductal tumours than their Ela-myc:Parp-1(+/+) counterparts, suggesting that Parp-1 participates in promoting acinar-to-ductal metaplasia, a key event in pancreatic cancer initiation. Moreover, impaired macrophage recruitment can be responsible for the ADM blockade found in the Ela-myc:Parp-1(-/-) mice. Finally, molecular analysis revealed that Parp-1 modulates ADM downstream of the Stat3-MMP7 axis and is also involved in transcriptional up-regulation of the MDM2, VEGFR1 and MMP28 cancer-related genes. In conclusion, the expression pattern of Parp-1 in normal and cancer tissue and the in vivo functional effects of Parp-1 depletion point to a novel role for this protein in pancreatic carcinogenesis and shed light into the clinical use of Parp-1 inhibitors.
Collapse
Affiliation(s)
- Neus Martínez-Bosch
- Cancer Research Programme, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Santoni M, Berardi R, Amantini C, Burattini L, Santini D, Santoni G, Cascinu S. Role of natural and adaptive immunity in renal cell carcinoma response to VEGFR-TKIs and mTOR inhibitor. Int J Cancer 2014; 134:2772-2777. [PMID: 24114790 DOI: 10.1002/ijc.28503] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Accepted: 09/12/2013] [Indexed: 12/15/2022]
Abstract
Angiogenesis and immunosuppression work hand-in-hand in the renal cell carcinoma (RCC) microenvironment. Tumor growth is associated with impaired antitumor immune response in RCC, which involves T cells, natural killer cells, dendritic cells (DCs) and macrophages. Vascular endothelial growth factor receptor (VEGFR), such as sorafenib, sunitinib, pazopanib and axitinib, and mammalian target of rapamycin (mTOR) inhibitors, such as temsirolimus and everolimus, do exert both antiangiogenic and immunomodulatory functions. Indeed, these agents affect neutrophil migration, as well as T lymphocyte-DC cross-talk, DC maturation and immune cell metabolism and reactivity. In this review, we overview the essential role of innate and adaptive immune response in RCC proliferation, invasion and metastasis and the relationship between tumor-associated immune cells and the response to targeted agents approved for the treatment of metastatic RCC.
Collapse
Affiliation(s)
- Matteo Santoni
- Medical Oncology AOU Ospedali Riuniti, Polytechnic University of the Marche Region, Ancona, Italy
| | | | | | | | | | | | | |
Collapse
|
33
|
Im YK, La Selva R, Gandin V, Ha JR, Sabourin V, Sonenberg N, Pawson T, Topisirovic I, Ursini-Siegel J. The ShcA adaptor activates AKT signaling to potentiate breast tumor angiogenesis by stimulating VEGF mRNA translation in a 4E-BP-dependent manner. Oncogene 2014; 34:1729-35. [PMID: 24837366 DOI: 10.1038/onc.2014.110] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2013] [Revised: 03/10/2014] [Accepted: 03/12/2014] [Indexed: 12/12/2022]
Abstract
The ShcA adaptor protein is engaged by numerous receptor tyrosine kinases (RTKs) in breast cancer cells. Once activated, RTKs phosphorylate three key tyrosine phosphorylation sites (Y239, Y240 and Y317) within ShcA that creates a docking site for Grb2/SOS and Grb2/Gab-containing complexes to activate the MAPK and AKT signaling pathways, respectively. We previously demonstrated that a tyrosine to phenylalanine substitution of the ShcA tyrosine phosphorylation sites (Shc3F-Y239/240/313F) significantly impairs breast tumor growth and angiogenesis in transgenic mouse models, in part, through the regulation of vascular endothelial growth factor (VEGF) production. Despite this fact, the underlying molecular mechanisms by which ShcA transduces pro-tumorigenic signals in breast cancer cells remain poorly defined. In this study, we demonstrate that ShcA-dependent activation of AKT, but not the RAS/MAPK pathway, induces VEGF production by bolstering VEGF mRNA translation. Accordingly, ShcA drives breast tumor growth and angiogenesis in vivo in a 4E-BP-dependent manner. These findings establish ShcA as a biological bridge that links AKT activation downstream of RTKs to cap-dependent VEGF mRNA translation in order to promote mammary tumorigenesis.
Collapse
Affiliation(s)
- Y K Im
- Department of Oncology, Lady Davis Institute for Medical Research, McGill University, Montreal, Quebec, Canada
| | - R La Selva
- Department of Oncology, Lady Davis Institute for Medical Research, McGill University, Montreal, Quebec, Canada
| | - V Gandin
- Department of Oncology, Lady Davis Institute for Medical Research, McGill University, Montreal, Quebec, Canada
| | - J R Ha
- Department of Oncology, Lady Davis Institute for Medical Research, McGill University, Montreal, Quebec, Canada
| | - V Sabourin
- Department of Oncology, Lady Davis Institute for Medical Research, McGill University, Montreal, Quebec, Canada
| | - N Sonenberg
- Goodman Cancer Research Centre, Montreal, Quebec, Canada
| | - T Pawson
- Samuel Lunenfeld Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada
| | - I Topisirovic
- Department of Oncology, Lady Davis Institute for Medical Research, McGill University, Montreal, Quebec, Canada
| | - J Ursini-Siegel
- Department of Oncology, Lady Davis Institute for Medical Research, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
34
|
Lkhagvadorj S, Oh SS, Lee MR, Jung JH, Chung HC, Cha SK, Eom M. VEGFR-1 Expression Relates to Fuhrman Nuclear Grade of Clear Cell Renal Cell Carcinoma. J Lifestyle Med 2014; 4:64-70. [PMID: 26064856 PMCID: PMC4390762 DOI: 10.15280/jlm.2014.4.1.64] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2014] [Accepted: 03/07/2014] [Indexed: 11/22/2022] Open
Abstract
Background: Increasing evidence suggests that vascular endothelial growth factor (VEGF) and VEGF receptor (VEGFR) 1 signaling may play an important role in the progression of pathological angiogenesis that occurs in many tumors, including renal cell carcinoma (RCC). Therapeutic targeting directed against VEGF and VEGFR-2 has been proven to be successful for metastatic clear cell RCC (CCRCC). However, the expression of VEGFR-1 and its association with prognostic parameters of CCRCC in the tumorigenesis of renal cancer remains unclear. Therefore, we examined the expression of VEGFR-1 and its prognostic significance in CCRCC. Methods: Immunohistochemical staining for VEGFR-1 was performed on 126 formalin-fixed paraffin-embedded CCRCC tissue samples. Six of these cases were available for Western blot analyses. The results were compared with various clinicopathologic parameters of CCRCC and patients’ survival. Results: VEGFR-1 expression was detected in 59 cases (46.8%) of CCRCC. Higher VEGFR-1 expression was significantly correlated with a lower Fuhrman nuclear grade and the absence of renal pelvis invasion, although it was not related to patients’ survival. Western blot analyses showed higher VEGFR-1 expression in low grade tumors. Conclusion: VEGFR-1 expression may be associated with favorable prognostic factors, particularly a lower Fuhrman nuclear grade in CCRCC.
Collapse
Affiliation(s)
- Sayamaa Lkhagvadorj
- Departments of Pathology, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Sung Soo Oh
- Occupational & Environmental Medicine, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Mi-Ra Lee
- Departments of Pathology, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Jae Hung Jung
- Urology, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Hyun Chul Chung
- Urology, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Seung-Kuy Cha
- Physiology, Yonsei University Wonju College of Medicine, Wonju, Korea ; Institute of Lifestyle Medicine, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Minseob Eom
- Departments of Pathology, Yonsei University Wonju College of Medicine, Wonju, Korea
| |
Collapse
|
35
|
Voronov E, Carmi Y, Apte RN. The role IL-1 in tumor-mediated angiogenesis. Front Physiol 2014; 5:114. [PMID: 24734023 PMCID: PMC3975103 DOI: 10.3389/fphys.2014.00114] [Citation(s) in RCA: 170] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Accepted: 03/07/2014] [Indexed: 12/12/2022] Open
Abstract
Tumor angiogenesis is one of the hallmarks of tumor progression and is essential for invasiveness and metastasis. Myeloid inflammatory cells, such as immature myeloid precursor cells, also termed myeloid-derived suppressor cells (MDSCs), neutrophils, and monocytes/macrophages, are recruited to the tumor microenvironment by factors released by the malignant cells that are subsequently “educated” in situ to acquire a pro-invasive, pro-angiogenic, and immunosuppressive phenotype. The proximity of myeloid cells to endothelial cells (ECs) lining blood vessels suggests that they play an important role in the angiogenic response, possibly by secreting a network of cytokines/chemokines and inflammatory mediators, as well as via activation of ECs for proliferation and secretion of pro-angiogenic factors. Interleukin-1 (IL-1) is an “alarm,” upstream, pro-inflammatory cytokine that is generated primarily by myeloid cells. IL-1 initiates and propagates inflammation, mainly by inducing a local cytokine network and enhancing inflammatory cell infiltration to affected sites and by augmenting adhesion molecule expression on ECs and leukocytes. Pro-inflammatory mediators were recently shown to play an important role in tumor-mediated angiogenesis and blocking their function may suppress tumor progression. In this review, we summarize the interactions between IL-1 and other pro-angiogenic factors during normal and pathological conditions. In addition, the feasibility of IL-1 neutralization approaches for anti-cancer therapy is discussed.
Collapse
Affiliation(s)
- Elena Voronov
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences and The Cancer Research Center, Ben-Gurion University of the Negev Beer-Sheva, Israel
| | - Yaron Carmi
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences and The Cancer Research Center, Ben-Gurion University of the Negev Beer-Sheva, Israel
| | - Ron N Apte
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences and The Cancer Research Center, Ben-Gurion University of the Negev Beer-Sheva, Israel
| |
Collapse
|
36
|
Bushway ME, Gerber SA, Fenton BM, Miller RK, Lord EM, Murphy SP. Morphological and phenotypic analyses of the human placenta using whole mount immunofluorescence. Biol Reprod 2014; 90:110. [PMID: 24648400 DOI: 10.1095/biolreprod.113.115915] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The human placenta performs multiple essential functions required for successful pregnancy. Alterations in the placental vasculature have been implicated in severe complications of pregnancy. Despite the importance of placental vascular function during pregnancy, there are gaps in our knowledge regarding the molecular pathways that control vessel development. Furthermore, there are limited tools available to simultaneously examine the morphology, phenotype, and spatial arrangement of cells within intact placental structures. To overcome these limitations, we developed whole mount immunofluorescence (WMIF) of the human placenta. Morphological analyses using WMIF revealed that blood vessel structures were consistent with an immature, angiogenic morphology in first-trimester placentas and mature, remodeled endothelium at term. To investigate placental expression of factors that control blood vessel development, we utilized WMIF to examine gestation age-specific expression of 1) the receptors for vascular endothelial growth factor (VEGFR-1, VEGFR-2, and VEGFR-3), which are required for placental vascular development in mice, and 2) activated, tyrosine phosphorylated STAT3 (pSTAT3), a transcription factor that mediates VEGFR2 signaling. We detected high levels of VEGFR2, VEGFR3, and pSTAT3 expression in early placental blood vessels that were significantly diminished by term. VEGFR1 was expressed primarily in trophoblast and Hofbauer cells throughout gestation. Based on our collective results, we propose that VEGFR2, VEGFR3, and STAT3 play essential roles in the development of the human placental vasculature. In addition, we anticipate that WMIF will provide a powerful approach for comparing placental morphology and protein expression in normal versus pathological pregnancies and for investigating the effects of environmental factors on placental function.
Collapse
Affiliation(s)
- Meghan E Bushway
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York
| | - Scott A Gerber
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York
| | - Bruce M Fenton
- Department of Radiation Oncology, University of Rochester School of Medicine and Dentistry, Rochester, New York
| | - Richard K Miller
- Department of Obstetrics and Gynecology, University of Rochester School of Medicine and Dentistry, Rochester, New York
| | - Edith M Lord
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York
| | - Shawn P Murphy
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York Department of Obstetrics and Gynecology, University of Rochester School of Medicine and Dentistry, Rochester, New York
| |
Collapse
|
37
|
Qu B, Liu BR, DU YJ, Chen J, Cheng YQ, Xu W, Wang XH. Wnt/β-catenin signaling pathway may regulate the expression of angiogenic growth factors in hepatocellular carcinoma. Oncol Lett 2014; 7:1175-1178. [PMID: 24944688 PMCID: PMC3961220 DOI: 10.3892/ol.2014.1828] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Accepted: 12/17/2013] [Indexed: 12/31/2022] Open
Abstract
The Wnt/β-catenin signaling pathway plays a key role during hepatocellular carcinoma (HCC) genesis and development. The present study aimed to investigate the effects of the Wnt/β-catenin signaling pathway on the expression of angiogenic growth factors involved in HCC. The HCC HepG2 cell line was transfected with small interfering RNA (siRNA) against β-catenin. After 72 and 96 h, protein was extracted and the expression levels of β-catenin, matrix metalloproteinase (MMP)-2, MMP-9, vascular endothelial growth factor (VEGF)-A, VEGF-C and basic fibroblast growth factor (bFGF) were detected by western blot analysis. β-catenin protein expression was inhibited at both time points. Notably, MMP-2, MMP-9, VEGF-A, VEGF-C and bFGF protein expression levels decreased at 72 h and then increased at 96 h after transfection. Our results demonstrated that in HCC cells, the Wnt/β-catenin signaling pathway may regulate the protein expression of the angiogenic factors, MMP-2, MMP-9, VEGF-A, VEGF-C and bFGF. These proteins were downstream of β-catenin signaling and were also regulated by other factors. In conclusion, the Wnt/β-catenin signaling pathway may contribute to the regulation of HCC angiogenesis, infiltration and metastasis through regulating the expression of these angiogenic factors.
Collapse
Affiliation(s)
- Bo Qu
- Department of Gastroenterology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Bing-Rong Liu
- Department of Gastroenterology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Ya-Ju DU
- Department of Gastroenterology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Jing Chen
- Department of Gastroenterology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Yan-Qiu Cheng
- Department of Gastroenterology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Wei Xu
- Department of Gastroenterology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Xin-Hong Wang
- Department of Gastroenterology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| |
Collapse
|
38
|
Wang D, Luo L, Chen W, Chen LZ, Zeng WT, Li W, Huang XH. Significance of the vascular endothelial growth factor and the macrophage migration inhibitory factor in the progression of hepatocellular carcinoma. Oncol Rep 2013; 31:1199-204. [PMID: 24366206 DOI: 10.3892/or.2013.2946] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Accepted: 12/16/2013] [Indexed: 12/26/2022] Open
Abstract
The aim of the present study was to investigate the expression of vascular endothelial growth factor (VEGF) and macrophage migration inhibitory factor (MIF) in HCC progression and their correlation with clinicopathological factors as well as the relationship between their expression levels. The expression of serum VEGF and MIF was evaluated in 150 patients with HCC and in 30 normal volunteers by enzyme-linked immunosorbent assay (ELISA). VEGF and MIF expression levels were evaluated by immunohistochemistry on tissue microarrays containing 150 HCCs with paired adjacent non-cancer liver tissues. VEGF and MIF mRNA levels were determined by quantitative PCR in another 48 HCCs. The correlation of VEGF and MIF with clinicopathological factors was analyzed in HCC. Serum VEGF and MIF concentrations were higher in HCC patients than the levels in the controls. The expression levels of VEGF and MIF in the HCC tissues were both higher than those in the adjacent non-tumor liver tissues. Overexpression of VEGF and MIF was significantly associated with tumor size (P=0.027 and 0.022, respectively), intrahepatic metastasis (P=0.032 and 0.027, respectively), vascular invasion (P=0.044 and 0.039, respectively) and TNM stage (P=0.028 and 0.013, respectively). Furthermore, VEGF and MIF mRNA levels were higher in HCC compared to levels in the paired non-cancer liver tissues. VEGF and MIF mRNA levels were correlated with tumor stage and metastasis. The expression of VEGF was positively related with MIF expression in HCC. The expression of MIF and VEGF in HCC was markedly positively correlated, which suggests that MIF and VEGF play an important role in the progression of HCC. Both factors may concomitantly accelerate the progression of HCC.
Collapse
Affiliation(s)
- Dong Wang
- Department of Laboratory Medicine, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, P.R. China
| | - Liang Luo
- Department of Medical Intensive Care Unit, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, P.R. China
| | - Wei Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, P.R. China
| | - Lian-Zhou Chen
- Department of General Surgical Laboratory, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, P.R. China
| | - Wen-Tao Zeng
- Department of General Surgical Laboratory, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, P.R. China
| | - Wen Li
- Department of General Surgical Laboratory, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, P.R. China
| | - Xiao-Hui Huang
- Department of General Surgical Laboratory, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, P.R. China
| |
Collapse
|
39
|
Kiguchi N, Kobayashi Y, Kadowaki Y, Fukazawa Y, Saika F, Kishioka S. Vascular endothelial growth factor signaling in injured nerves underlies peripheral sensitization in neuropathic pain. J Neurochem 2013; 129:169-78. [PMID: 24304382 DOI: 10.1111/jnc.12614] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Accepted: 11/18/2013] [Indexed: 12/11/2022]
Abstract
Chronic neuroinflammation may be a critical component of intractable inflammatory diseases, including neuropathic pain. Because angiogenesis as a result of vascular endothelial growth factor (VEGF) signaling plays a pivotal role in inflammation, we focused on the mechanisms of VEGF-regulated neuropathic pain in mice. The mRNA and protein expression of VEGFA were up-regulated in the injured sciatic nerve after partial sciatic nerve ligation (PSL). VEGFA was localized to accumulated macrophages and neutrophils derived from bone marrow. Up-regulation of VEGFA was mediated by histone H3 acetylation and trimethylation in its promoter region. VEGF receptors (VEGFR1 and VEGFR2) were localized to vascular endothelial cells or macrophages. By ex vivo fluorescence imaging and immunohistochemistry using DiI fluorescence, progression of angiogenesis was observed in the injured sciatic nerve after PSL. Perineural administration of pharmacological inhibitors of VEGFA and VEGFR tyrosine kinases prevented tactile allodynia and thermal hyperalgesia caused by PSL. Moreover, we determined the contribution of VEGF- and CXC-chemokine receptor 4-expressing angiogenic macrophages to neuropathic pain. Taken together, VEGFA is up-regulated in injured peripheral nerves and participates in angiogenesis and prolonged pain behaviors through its receptors. We propose that VEGFA-related components may underlie peripheral sensitization leading to neuropathic pain. Angiogenesis due to VEGF signaling is a key component of chronic inflammation. VEGFA up-regulation and pathological angiogenesis were observed in the injured nerves in mice. Pharmacological inhibition of VEGF signaling suppressed neuropathic pain behaviors. Therefore, VEGFA-related components may underlie peripheral neuroinflammation leading to neuropathic pain.
Collapse
Affiliation(s)
- Norikazu Kiguchi
- Department of Pharmacology, Wakayama Medical University, Wakayama, Japan
| | | | | | | | | | | |
Collapse
|
40
|
Santoni M, Massari F, Amantini C, Nabissi M, Maines F, Burattini L, Berardi R, Santoni G, Montironi R, Tortora G, Cascinu S. Emerging role of tumor-associated macrophages as therapeutic targets in patients with metastatic renal cell carcinoma. Cancer Immunol Immunother 2013; 62:1757-1768. [PMID: 24132754 PMCID: PMC11029754 DOI: 10.1007/s00262-013-1487-6] [Citation(s) in RCA: 99] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Accepted: 10/07/2013] [Indexed: 01/08/2023]
Abstract
Tumor-associated macrophages (TAMs) derived from peripheral blood monocytes recruited into the renal cell carcinoma (RCC) microenvironment. In response to inflammatory stimuli, macrophages undergo M1 (classical) or M2 (alternative) activation. M1 cells produce high levels of inflammatory cytokines, such as tumor necrosis factor-α, interleukin (IL)-12, IL-23 and IL-6, while M2 cells produce anti-inflammatory cytokines, such as IL-10, thus contributing to RCC-related immune dysfunction. The presence of extensive TAM infiltration in RCC microenvironment contributes to cancer progression and metastasis by stimulating angiogenesis, tumor growth, and cellular migration and invasion. Moreover, TAMs are involved in epithelial-mesenchymal transition of RCC cancer cells and in the development of tumor resistance to targeted agents. Interestingly, macrophage autophagy seems to play an important role in RCC. Based on this scenario, TAMs represent a promising and effective target for cancer therapy in RCC. Several strategies have been proposed to suppress TAM recruitment, to deplete their number, to switch M2 TAMs into antitumor M1 phenotype and to inhibit TAM-associated molecules. In this review, we summarize current data on the essential role of TAMs in RCC angiogenesis, invasion, impaired anti-tumor immune response and development of drug resistance, thus describing the emerging TAM-centered therapies for RCC patients.
Collapse
Affiliation(s)
- Matteo Santoni
- Medical Oncology, AOU Ospedali Riuniti, Polytechnic University of the Marche Region, via Conca 71, 60126, Ancona, Italy,
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Li FY, Zhu SX, Wang ZP, Wang H, Zhao Y, Chen GP. Consumption of hydrogen-rich water protects against ferric nitrilotriacetate-induced nephrotoxicity and early tumor promotional events in rats. Food Chem Toxicol 2013; 61:248-54. [PMID: 24140467 DOI: 10.1016/j.fct.2013.10.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Revised: 09/21/2013] [Accepted: 10/05/2013] [Indexed: 12/13/2022]
Abstract
The aim of this work was to test whether consumption with hydrogen-rich water (HW) alleviated renal injury and inhibited early tumor promotional events in Ferric nitrilotriacetate (Fe-NTA)-treated rats. Rats were injected with Fe-NTA solution (7.5mg Fe/kg body weight) intraperitoneally to induce renal injury and simultaneously treated with HW (1.3 ± 0.2mg/l). We found that consumption with HW ameliorated Fe-NTA-induced renal injuries including suppressing elevation of serum creatinine and blood urea nitrogen and inhibited early tumor promotional events including decreasing ornithine decarboxylase activity and incorporation of [3H]thymidine into renal DNA. Consumption with HW suppressed Fe-NTA-induced oxidative stress through decreasing formation of lipid peroxidation and peroxynitrite and activities of NADPH oxidase and xanthine oxidase, increasing activity of catalase, and restoring mitochondrial function in kidneys. Consumption with HW suppressed Fe-NTA-induced inflammation marked by reduced NF-κB, IL-6, and MCP-1 expression and macrophage accumulating in kidneys. In addition, consumption with HW suppressed VEGF expression, STAT3 phosphorylation and PCNA expression in kidneys of Fe-NTA-treated rats. Consumption with HW decreased the incidence of renal cell carcinoma and suppressed tumor growth in Fe-NTA-treated in rats. In conclusion, drinking with HW attenuated Fe-NTA-induced renal injury and inhibited early tumor promotional events in rats.
Collapse
Affiliation(s)
- Fang-Yin Li
- Department of Urology, Zhejiang Cancer Hospital, Hang Zhou 310022, Zhejiang Province, China
| | | | | | | | | | | |
Collapse
|
42
|
Carmi Y, Dotan S, Rider P, Kaplanov I, White MR, Baron R, Abutbul S, Huszar M, Dinarello CA, Apte RN, Voronov E. The role of IL-1β in the early tumor cell-induced angiogenic response. THE JOURNAL OF IMMUNOLOGY 2013; 190:3500-9. [PMID: 23475218 DOI: 10.4049/jimmunol.1202769] [Citation(s) in RCA: 150] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
In this study, we assessed the involvement of IL-1β in early angiogenic responses induced by malignant cells using Matrigel plugs supplemented with B16 melanoma cells. We found that during the angiogenic response, IL-1β and vascular endothelial growth factor (VEGF) interact in a newly described autoinduction circuit, in which each of these cytokines induces the other. The IL-1β and VEGF circuit acts through interactions between bone marrow-derived VEGF receptor 1(+)/IL-1R1(+) immature myeloid cells and tissue endothelial cells. Myeloid cells produce IL-1β and additional proinflammatory cytokines, which subsequently activate endothelial cells to produce VEGF and other proangiogenic factors and provide the inflammatory microenvironment for angiogenesis and tumor progression. These mechanisms were also observed in a nontumor early angiogenic response elicited in Matrigel plugs by either rIL-1β or recombinant VEGF. We have shown that IL-1β inhibition stably reduces tumor growth by limiting inflammation and inducing the maturation of immature myeloid cells into M1 macrophages. In sharp contrast, only transient inhibition of tumor growth was observed after VEGF neutralization, followed by tumor recurrence mediated by rebound angiogenesis. This occurs via the reprogramming of VEGF receptor 1(+)/IL-1R1(+) cells to express hypoxia inducible factor-1α, VEGF, and other angiogenic factors, thereby directly supporting proliferation of endothelial cells and blood vessel formation in a paracrine manner. We suggest using IL-1β inhibition as an effective antitumor therapy and are currently optimizing the conditions for its application in the clinic.
Collapse
Affiliation(s)
- Yaron Carmi
- The Shraga Segal Department of Microbiology, Immunology and Genetics and The Cancer Research Center, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Lee HW, Choi HJ, Ha SJ, Lee KT, Kwon YG. Recruitment of monocytes/macrophages in different tumor microenvironments. Biochim Biophys Acta Rev Cancer 2012; 1835:170-9. [PMID: 23287570 DOI: 10.1016/j.bbcan.2012.12.007] [Citation(s) in RCA: 120] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2012] [Revised: 12/16/2012] [Accepted: 12/20/2012] [Indexed: 01/21/2023]
Abstract
After emigration from the bone marrow into the peripheral blood, monocytes enter tissues and differentiate into macrophages. Monocytes/macrophages have many roles in immune regulation, angiogenesis, and tumor metastasis and invasion. In addition, studies have revealed that these cells are essential to tumor progression. Recently, an accumulation of evidence has indicated that macrophages in distinct regions of tumor masses have distinct origins. For instance, classical monocytes appear to be a major source of macrophages in tumor epithelial, perivascular, and hypoxic regions. In contrast, non-classical monocytes are an important source of macrophages in the tumor perivascular region. During the past century, it has been demonstrated that several chemoattractants can regulate the recruitment of monocytes/macrophages to tumor sites. Despite the importance of monocytes/macrophages in tumor progression, there had been, until recently, no efforts to summarize receptor-ligand pairs between tumor-derived chemokines and corresponding receptors in monocytes in different microenvironments. In this review, we present a cohesive view of the distinct expression patterns of chemokine receptors in two different monocyte subsets (classical and non-classical monocytes) and describe their roles in monocyte/macrophage recruitment into distinct tumor microenvironments. This review provides insight into the behavior of monocytes/macrophages in different tumor microenvironments.
Collapse
Affiliation(s)
- Heon-Woo Lee
- Department of Biochemistry, Yonsei University, Seoul, Republic of Korea
| | | | | | | | | |
Collapse
|
44
|
Role of bone marrow-derived cells in angiogenesis: focus on macrophages and pericytes. CANCER MICROENVIRONMENT 2012; 5:225-36. [PMID: 22528877 DOI: 10.1007/s12307-012-0106-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2012] [Accepted: 04/03/2012] [Indexed: 02/06/2023]
Abstract
Tumor growth relies on the formation of new blood vessels to receive an adequate supply of oxygen and nutrient. This process is facilitated by both the remodeling of the pre-existing vasculatures and the recruitment of the progenitor/stem cells originated from bone marrow-derived cells (BMDCs). Evidences from both animal studies and human trials have reported that these tumor-associated BMDCs differentiate into a series of stromal cells including macrophages and pericytes, and regulate tumor angiogenesis in various aspects. Macrophages constitute a large portion of the BMDCs infiltrated in the tumor microenvironment, and have been shown to disrupt the balance of pro- and anti-angiogenic signalings by the secretion of various cytokines. Pericytes, mainly derived from the subpopulation of PDGFRβ(+) BMDCs, can provide both pro-survival signaling and mechanical support to maintain the newly formed endothelium via the direct interactions with endothelial cells. In the current review, we summarize the recruitment mechanisms of BMDC-derived macrophages and pericytes within tumor microenvironment, and also review the contribution of these cells to the different aspects of angiogenesis, with particular emphasis on their therapeutic implications as potential targets for anti-tumor strategies.
Collapse
|