1
|
Zhou L, Cui M, Yu J, Liu Y, Zeng F, Liu Y. Identification of Versican as a target gene of the transcription Factor ZNF587B in ovarian cancer. Biochem Pharmacol 2025; 237:116946. [PMID: 40228636 DOI: 10.1016/j.bcp.2025.116946] [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: 12/05/2024] [Revised: 02/20/2025] [Accepted: 04/11/2025] [Indexed: 04/16/2025]
Abstract
Ovarian cancer is the most lethal malignancy affecting the female reproductive system, with its progression and metastasis being significant contributors to patient mortality. Our previous study identified the zinc finger protein ZNF587B as a potential tumor suppressor that inhibited the proliferation, migration and invasion of ovarian cancer cells, although the underlying mechanism remains elusive. In this study, ZNF587B was demonstrated to bind directly to the promoter region of Versican (VCAN), a high molecular weight chondroitin sulfate glycoprotein, and repress its transcription using Chromatin immunoprecipitation-qPCR (ChIP-qPCR), luciferase reporter assays, and immunofluorescence (IF). Moreover, in vivo and in vitro assays revealed that the effect of ZNF587B knockdown on ovarian cancer proliferation may be mediated through VCAN. Not only that, patients with reduced expression of ZNF587B and increased expression of VCAN exhibit a poorer prognosis. The potential mechanism behind this may involve its impact on the phosphorylation process of AKT.
Collapse
Affiliation(s)
- Lu Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha 410008, PR China; National Laboratory of Medical Genetics, Central South University, Changsha 410078, PR China
| | - Mengke Cui
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha 410008, PR China; National Laboratory of Medical Genetics, Central South University, Changsha 410078, PR China
| | - Jian Yu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha 410008, PR China; National Laboratory of Medical Genetics, Central South University, Changsha 410078, PR China
| | - Yujie Liu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha 410008, PR China; National Laboratory of Medical Genetics, Central South University, Changsha 410078, PR China
| | - Feiyue Zeng
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, PR China
| | - Yingzi Liu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha 410008, PR China; National Laboratory of Medical Genetics, Central South University, Changsha 410078, PR China.
| |
Collapse
|
2
|
Soucy AM, Brune JE, Jayaraman A, Shenoy AT, Korkmaz FT, Etesami NS, Hiller BE, Martin IM, Goltry WN, Ha CT, Crossland NA, Campbell JD, Beach TG, Traber KE, Jones MR, Quinton LJ, Bosmann M, Frevert CW, Mizgerd JP. Transcriptomic responses of lung mesenchymal cells during pneumonia. JCI Insight 2025; 10:e177084. [PMID: 39998887 PMCID: PMC11981624 DOI: 10.1172/jci.insight.177084] [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: 10/31/2023] [Accepted: 02/18/2025] [Indexed: 02/27/2025] Open
Abstract
The role of mesenchymal cells during respiratory infection is not well defined, including whether, which, and how the different types of mesenchymal cells respond. We collected all mesenchymal cells from lung single-cell suspensions of mice that were naive (after receiving only saline vehicle), pneumonic (after intratracheal instillation of pneumococcus 24 hours previously), or resolved from infection (after nonlethal pneumococcal infections 6 weeks previously) and performed single-cell RNA sequencing. Cells clustered into 5 well-separated groups based on their transcriptomes: matrix fibroblasts, myofibroblasts, pericytes, smooth muscle cells, and mesothelial cells. Fibroblasts were the most abundant and could be further segregated into Pdgfra+Npnt+Ces1d+Col13a1+ alveolar fibroblasts and Cd9+Pi16+Sca1+Col14a1+ adventitial fibroblasts. The cells from naive and resolved groups overlapped in dimension reduction plots, suggesting the mesenchymal cells returned to baseline transcriptomes after resolution. During pneumonia, all mesenchymal cells responded with altered transcriptomes, revealing a core response that had been conserved across cell types as well as distinct mesenchymal cell type-specific responses. The different subsets of fibroblasts induced similar gene sets, but the alveolar fibroblasts responded more strongly than the adventitial fibroblasts. These data demonstrated diverse and specialized immune activities of lung mesenchymal cells during pneumonia.
Collapse
Affiliation(s)
- Alicia M. Soucy
- Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
| | - Jourdan E. Brune
- Department of Comparative Medicine, University of Washington School of Medicine, Seattle, Washington, USA
- Center for Lung Biology, University of Washington, Seattle, Washington, USA
| | - Archana Jayaraman
- Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
| | - Anukul T. Shenoy
- Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Filiz T. Korkmaz
- Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
- University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Neelou S. Etesami
- Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
| | - Bradley E. Hiller
- Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
| | - Ian M.C. Martin
- Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
| | - Wesley N. Goltry
- Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
| | - Catherine T. Ha
- Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
| | - Nicholas A. Crossland
- National Emerging Infectious Diseases Laboratory, Boston University, Boston, Massachusetts, USA
- Department of Pathology and Laboratory Medicine
- Department of Virology, Immunology, & Microbiology; and
| | - Joshua D. Campbell
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
| | - Thomas G. Beach
- Banner Sun Health Research Institute Brain and Body Donation Program, Sun City, Arizona, USA
| | - Katrina E. Traber
- Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
| | - Matthew R. Jones
- Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
| | - Lee J. Quinton
- Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
- University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Markus Bosmann
- Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Charles W. Frevert
- Department of Comparative Medicine, University of Washington School of Medicine, Seattle, Washington, USA
- Center for Lung Biology, University of Washington, Seattle, Washington, USA
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, University of Washington, Seattle, Washington, USA
| | - Joseph P. Mizgerd
- Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
- Department of Virology, Immunology, & Microbiology; and
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
- Department of Biochemistry and Cell Biology, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
3
|
Xu Y, Benedikt J, Ye L. Hyaluronic Acid Interacting Molecules Mediated Crosstalk between Cancer Cells and Microenvironment from Primary Tumour to Distant Metastasis. Cancers (Basel) 2024; 16:1907. [PMID: 38791985 PMCID: PMC11119954 DOI: 10.3390/cancers16101907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 05/11/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024] Open
Abstract
Hyaluronic acid (HA) is a prominent component of the extracellular matrix, and its interactions with HA-interacting molecules (HAIMs) play a critical role in cancer development and disease progression. This review explores the multifaceted role of HAIMs in the context of cancer, focusing on their influence on disease progression by dissecting relevant cellular and molecular mechanisms in tumour cells and the tumour microenvironment. Cancer progression can be profoundly affected by the interactions between HA and HAIMs. They modulate critical processes such as cell adhesion, migration, invasion, and proliferation. The TME serves as a dynamic platform in which HAIMs contribute to the formation of a unique niche. The resulting changes in HA composition profoundly influence the biophysical properties of the TME. These modifications in the TME, in conjunction with HAIMs, impact angiogenesis, immune cell recruitment, and immune evasion. Therefore, understanding the intricate interplay between HAIMs and HA within the cancer context is essential for developing novel therapeutic strategies. Targeting these interactions offers promising avenues for cancer treatment, as they hold the potential to disrupt critical aspects of disease progression and the TME. Further research in this field is imperative for advancing our knowledge and the treatment of cancer.
Collapse
Affiliation(s)
- Yali Xu
- Cardiff China Medical Research Collaborative, Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff CF14 4XN, UK;
- School of Engineering, Cardiff University, Cardiff CF24 3AA, UK;
| | | | - Lin Ye
- Cardiff China Medical Research Collaborative, Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff CF14 4XN, UK;
| |
Collapse
|
4
|
Reis MBE, Maximo AI, Magno JM, de Lima Bellan D, Buzzo JLA, Simas FF, Rocha HAO, da Silva Trindade E, Camargo de Oliveira C. A Fucose-Containing Sulfated Polysaccharide from Spatoglossum schröederi Potentially Targets Tumor Growth Rather Than Cytotoxicity: Distinguishing Action on Human Melanoma Cell Lines. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2024; 26:181-198. [PMID: 38273163 DOI: 10.1007/s10126-024-10287-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 01/08/2024] [Indexed: 01/27/2024]
Abstract
Natural substances are strategic candidates for drug development in cancer research. Marine-derived molecules are of special interest due to their wide range of biological activities and sustainable large-scale production. Melanoma is a type of skin cancer that originates from genetic mutations in melanocytes. BRAF, RAS, and NF1 mutations are described as the major melanoma drivers, but approximately 20% of patients lack these mutations and are included in the triple wild-type (tripleWT) classification. Recent advances in targeted therapy directed at driver mutations along with immunotherapy have only partially improved patients' overall survival, and consequently, melanoma remains deadly when in advanced stages. Fucose-containing sulfated polysaccharides (FCSP) are potential candidates to treat melanoma; therefore, we investigated Fucan A, a FCSP from Spatoglossum schröederi brown seaweed, in vitro in human melanoma cell lines presenting different mutations. Up to 72 h Fucan A treatment was not cytotoxic either to normal melanocytes or melanoma cell lines. Interestingly, it was able to impair the tripleWT CHL-1 cell proliferation (57%), comparable to the chemotherapeutic cytotoxic drug cisplatin results, with the advantage of not causing cytotoxicity. Fucan A increased CHL-1 doubling time, an effect attributed to cell cycle arrest. Vascular mimicry, a close related angiogenesis process, was also impaired (73%). Fucan A mode of action could be related to gene expression modulation, in special β-catenin downregulation, a molecule with protagonist roles in important signaling pathways. Taken together, results indicate that Fucan A is a potential anticancer molecule and, therefore, deserves further investigation.
Collapse
Affiliation(s)
- Maíra Barbosa E Reis
- Cell Biology Department, Universidade Federal Do Paraná (UFPR), Curitiba, Paraná, Brazil
| | | | - Jessica Maria Magno
- Cell Biology Department, Universidade Federal Do Paraná (UFPR), Curitiba, Paraná, Brazil
| | - Daniel de Lima Bellan
- Cell Biology Department, Universidade Federal Do Paraná (UFPR), Curitiba, Paraná, Brazil
| | | | | | - Hugo Alexandre Oliveira Rocha
- Biochemistry Department, Centro de Biociências, Universidade Federal do Rio Grande do Norte (UFRN), Natal, Rio Grande do Norte, Brazil
| | | | | |
Collapse
|
5
|
Xiong J, Lai Y, Cheng N, Liu J, Wang F, Zheng X, Wang Y, Zhuang Q, Lin Y, Liu J, Yang Y, Zhao B, Yang X. Lnc-PLA2G4A-4 facilitates the progression of hepatocellular carcinoma by inducing versican expression via sponging miR-23b-3p. Heliyon 2023; 9:e18698. [PMID: 37554815 PMCID: PMC10405012 DOI: 10.1016/j.heliyon.2023.e18698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 07/20/2023] [Accepted: 07/25/2023] [Indexed: 08/10/2023] Open
Abstract
Aberrant expression of long non-coding RNAs (lncRNAs) is associated with progression of multiple human cancers including hepatocellular carcinoma (HCC). However, the role of lncRNAs in HCC is not been fully understood. Our study aimed to investigate the biological function and potential molecular mechanism of Lnc-PAL2G4A-4 in HCC. In the current study, we show that Lnc-PLA2G4A-4 was significantly up-regulated in HCC tissues and high Lnc-PLA2G4A-4 expression was remarkably associated with tumor size, microvascular invasion and poor prognosis of HCC patients. Functionally, Lnc-PLA2G4A-4 positively regulated cell proliferation, invasion and migration in vitro, and facilitated lung metastasis of HCC in vivo. Mechanistically, Lnc-PLA2G4A-4 functioned as a competing endogenous RNA (ceRNA) to bind to miR-23b-3p and subsequently facilitate miR-23b-3p's target gene versican (VCAN) expression in HCC cells. Over-expression of miR-23b-3p could reverse Lnc-PLA2G4A-4 induced cell phenotypes in HCC and suppress versican expression of by rescue analysis. Collectively, Lnc-PLA2G4A-4 promotes HCC progression by targeting the miR-23b-3p/versican axis, which may be a potential biomarker and therapeutic target for HCC.
Collapse
Affiliation(s)
- Jiahui Xiong
- Fuzhou Hospital of Traditional Chinese Medicine Affiliated to Fujian University of Traditional Chinese Medicine, Fuzhou 350001, PR China
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, PR China
- The School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350122, PR China
| | - Yongping Lai
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, PR China
- Fujian Provincial Clinical Research Center for Hepatobiliary and Pancreatic Tumors, Fuzhou 350025, PR China
| | - Niangmei Cheng
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, PR China
- Fujian Provincial Clinical Research Center for Hepatobiliary and Pancreatic Tumors, Fuzhou 350025, PR China
| | - Jizhe Liu
- College of Life Science, Fujian Agriculture and Forestry University, Fuzhou, Fujian, 350002, PR China
| | - Fei Wang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, PR China
- Fujian Provincial Clinical Research Center for Hepatobiliary and Pancreatic Tumors, Fuzhou 350025, PR China
| | - Xiaoyuan Zheng
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, PR China
- Fujian Provincial Clinical Research Center for Hepatobiliary and Pancreatic Tumors, Fuzhou 350025, PR China
| | - Yingchao Wang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, PR China
- Fujian Provincial Clinical Research Center for Hepatobiliary and Pancreatic Tumors, Fuzhou 350025, PR China
| | - Qiuyu Zhuang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, PR China
- Fujian Provincial Clinical Research Center for Hepatobiliary and Pancreatic Tumors, Fuzhou 350025, PR China
| | - Yantin Lin
- Fuzhou Hospital of Traditional Chinese Medicine Affiliated to Fujian University of Traditional Chinese Medicine, Fuzhou 350001, PR China
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, PR China
- The School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350122, PR China
| | - Jingfeng Liu
- Department of Hepatopancreatobiliary Surgery, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, Fujian, PR China
| | - Yixuan Yang
- Fuzhou Gezhi High School of Fujian, Fuzhou, PR China
| | - Bixing Zhao
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, PR China
- Fujian Provincial Clinical Research Center for Hepatobiliary and Pancreatic Tumors, Fuzhou 350025, PR China
| | - Xiaoyu Yang
- Fuzhou Hospital of Traditional Chinese Medicine Affiliated to Fujian University of Traditional Chinese Medicine, Fuzhou 350001, PR China
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, PR China
- The School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350122, PR China
| |
Collapse
|
6
|
Song J, Wei R, Huo S, Liu C, Liu X. Versican enrichment predicts poor prognosis and response to adjuvant therapy and immunotherapy in gastric cancer. Front Immunol 2022; 13:960570. [PMID: 36203562 PMCID: PMC9530562 DOI: 10.3389/fimmu.2022.960570] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 09/06/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundIncreasing evidence has revealed an important role of versican (VCAN) on various aspects of cancer progression. Here, we assessed the impact of VCAN expression on prognosis and the response to adjuvant therapy and immunotherapy in patients with gastric cancer (GC).MethodsFour independent cohorts containing 1353 patients with GC, were utilized to investigate the effect of VCAN expression on prognosis and response to adjuvant therapy in GC. Two cohorts treated with immune checkpoint blockades were included to assess the predict value of VCAN expression on response to immunotherapy. Moreover, the bulk RNA-seq and single-cell RNA-seq data were analyzed to illustrate the role of VCAN in tumor microenvironment. Clinical outcomes of patient subgroups were compared by Kaplan-Meier curves with the log-rank test.ResultHigh VCAN expression was associated with poor prognosis for patients with GC. Compared with patients with high VCAN expression, patients with low VCAN expression benefited more from adjuvant chemotherapy and adjuvant chemoradiotherapy. Moreover, patients with high VCAN expression tended to be resistant to immunotherapy, and VCAN could serve as a promising indicator for predicting the response to immunotherapy. VCANhigh tumors showed a specific microenvironment with more cancer associated fibroblasts infiltration and significant enrichment of stromal relevant signaling pathways.ConclusionVCAN could predict the response to adjuvant chemotherapy, adjuvant chemoradiotherapy and immunotherapy in GC, and designing new medicine target to VCAN might be an effective way to improve the efficacy of several treatment options for GC.
Collapse
Affiliation(s)
- Junquan Song
- Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
| | - Rongyuan Wei
- Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
| | - Shiying Huo
- Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
| | - Chenchen Liu
- Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
- *Correspondence: Chenchen Liu, ; Xiaowen Liu,
| | - Xiaowen Liu
- Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
- *Correspondence: Chenchen Liu, ; Xiaowen Liu,
| |
Collapse
|
7
|
Hu H, Chen Y, Tan S, Wu S, Huang Y, Fu S, Luo F, He J. The Research Progress of Antiangiogenic Therapy, Immune Therapy and Tumor Microenvironment. Front Immunol 2022; 13:802846. [PMID: 35281003 PMCID: PMC8905241 DOI: 10.3389/fimmu.2022.802846] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 02/01/2022] [Indexed: 02/05/2023] Open
Abstract
Anti-angiogenesis therapy, a promising strategy against cancer progression, is limited by drug-resistance, which could be attributed to changes within the tumor microenvironment. Studies have increasingly shown that combining anti-angiogenesis drugs with immunotherapy synergistically inhibits tumor growth and progression. Combination of anti-angiogenesis therapy and immunotherapy are well-established therapeutic options among solid tumors, such as non-small cell lung cancer, hepatic cell carcinoma, and renal cell carcinoma. However, this combination has achieved an unsatisfactory effect among some tumors, such as breast cancer, glioblastoma, and pancreatic ductal adenocarcinoma. Therefore, resistance to anti-angiogenesis agents, as well as a lack of biomarkers, remains a challenge. In this review, the current anti-angiogenesis therapies and corresponding drug-resistance, the relationship between tumor microenvironment and immunotherapy, and the latest progress on the combination of both therapeutic modalities are discussed. The aim of this review is to discuss whether the combination of anti-angiogenesis therapy and immunotherapy can exert synergistic antitumor effects, which can provide a basis to exploring new targets and developing more advanced strategies.
Collapse
Affiliation(s)
- Haoyue Hu
- Lung Cancer Center, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, China.,Department of Medical Oncology, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, Medicine School of University of Electronic Science and Technology, Chengdu, China
| | - Yue Chen
- Department of Pathology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Songtao Tan
- Lung Cancer Center, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, China
| | - Silin Wu
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Yan Huang
- Lung Cancer Center, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, China
| | - Shengya Fu
- Lung Cancer Center, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, China.,Second Department of Oncology, Sichuan Friendship Hospital, Chengdu, China
| | - Feng Luo
- Lung Cancer Center, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, China
| | - Jun He
- Department of Oncology, The Third Hospital of Mianyang (Sichuan Mental Health Center), Mianyang, China
| |
Collapse
|
8
|
Timms KP, Maurice SB. Context-dependent bioactivity of versican fragments. Glycobiology 2021; 30:365-373. [PMID: 31651027 DOI: 10.1093/glycob/cwz090] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 09/19/2019] [Accepted: 09/19/2019] [Indexed: 01/05/2023] Open
Abstract
Versican (VCAN) proteolysis and the accumulation of VCAN fragments occur in many developmental and disease processes, affecting extracellular matrix (ECM) structure and cell phenotype. Little is known about the significance of proteolysis and the roles of fragments, or how this ECM remodeling affects the microenvironment and phenotype of diseased cells. G1-DPEAAE fragments promote aspects of epithelial-mesenchymal transitioning in developing and diseased cells, resulting in cell migration. Enhanced proliferation and invasion of tumor and endothelial cells is directly associated with G1 domain deposition and G1-DPEAAE localization respectively. These tumorigenic and angiogenic roles could explain the disease exacerbating effect often associated with G1-containing fragments, however, the pathogenicity of G1 fragments depends entirely upon the context. Overall, VCAN fragments promote tumorigenesis and inflammation; however, the specific cleavage site, the extent of cleavage activity and the microenvironment in which cleavage occurs collectively determine how this pleiotropic molecule and its fragments influence cells.
Collapse
Affiliation(s)
- Katherine Payne Timms
- University of Northern British Columbia, 3333 University Way, Prince George, BC, V2N 4Z9, Canada
| | - Sean Bertram Maurice
- Northern Medical Program, University of Northern British Columbia, Dr. Donald Rix Northern Health Sciences Centre, 3333 University Way, Prince George, BC, V2N 4Z9, Canada.,Department of Cellular and Physiological Sciences, Faculty of Medicine, University of British Columbia, 2350 Health Sciences Mall Vancouver, BC, V6T 1Z3, Canada
| |
Collapse
|
9
|
Zhong A, Ding N, Zhou Y, Yang G, Peng Z, Zhang H, Chai X. Identification of Hub Genes Associated with the Pathogenesis of Intracranial Aneurysm via Integrated Bioinformatics Analysis. Int J Gen Med 2021; 14:4039-4050. [PMID: 34354366 PMCID: PMC8331219 DOI: 10.2147/ijgm.s320396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 07/09/2021] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND At present, the pathogenesis of intracranial aneurysms (IA) remains unclear, which significantly hinders the development of novel strategies for the clinical treatment. In this study, bioinformatics methods were used to identify the potential hub genes and pathways associated with the pathogenesis of IA. METHODS The gene expression datasets of patients with intracranial aneurysm were downloaded from the Gene Expression Database (GEO), and the different data sets were integrated by the robust rank aggregation (RRA) method to identify the differentially expressed genes between patients with intracranial aneurysm and the controls. The functional enrichment analyses of the significant differentially expressed genes (DEGs) were performed and the protein-protein interaction (PPI) network was constructed; thereafter, the hub genes were screened by cytoHubba plug-in of Cytoscape, and finally sequencing dataset GSE122897 was used to verify the hub genes. RESULTS The GSE15629, GSE75436, GSE26969, and GSE6551 expression profiles have been included in this study, including 34 intracranial aneurysm samples and 26 control samples. The four datasets obtained 136 significant DEGs (45 up-regulated, 91 down-regulated). Enrichment analysis showed that the extracellular matrix structural constituent and the ECM-receptor interaction were closely related to the occurrence of IA. It was finally determined that eight hub genes associated with the development of IA, including VCAN, COL1A1, COL11A1, COL5A1, COL5A2, POSTN, THBS2, and CDH2. CONCLUSION The discovery of potential hub genes and pathways could enhance the understanding of the molecular mechanisms associated with the development of IA. These hub genes may be potential therapeutic targets for the management and new biomarker for the diagnosis of IA.
Collapse
Affiliation(s)
- Aifang Zhong
- Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
- Trauma center, The Second Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Ning Ding
- Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
- Trauma center, The Second Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Yang Zhou
- Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
- Trauma center, The Second Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Guifang Yang
- Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
- Trauma center, The Second Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Zhenyu Peng
- Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
- Emergency Medicine and Difficult Disease Institute, The Second Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Hongliang Zhang
- Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
- Emergency Medicine and Difficult Disease Institute, The Second Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Xiangping Chai
- Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
- Trauma center, The Second Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| |
Collapse
|
10
|
Comments on the ambiguity of selected surface markers, signaling pathways and omics profiles hampering the identification of myeloid-derived suppressor cells. Cell Immunol 2021; 364:104347. [PMID: 33838447 DOI: 10.1016/j.cellimm.2021.104347] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 03/23/2021] [Accepted: 03/24/2021] [Indexed: 12/12/2022]
Abstract
Myeloid-derived suppressor cells (MDSC) are important immune-regulatory cells but their identification remains difficult. Here, we provide a critical view on selected surface markers, transcriptional and translational pathways commonly used to identify MDSC by specific, their developmental origin and new possibilities by transcriptional or proteomic profiling. Discrimination of MDSC from their non-suppressive counterparts is a prerequisite for the development of successful therapies. Understanding the switch mechanisms that direct granulocytic and monocytic development into a pro-inflammatory or anti-inflammatory direction will be crucial for therapeutic strategies. Manipulation of these myeloid checkpoints are exploited by tumors and pathogens, such as M. tuberculosis (Mtb), HIV or SARS-CoV-2, that induce MDSC for immune evasion. Thus, specific markers for MDSC identification may reveal also novel molecular candidates for therapeutic intervention at the level of MDSC.
Collapse
|
11
|
ADAMTS proteases and the tumor immune microenvironment: Lessons from substrates and pathologies. Matrix Biol Plus 2020; 9:100054. [PMID: 33718860 PMCID: PMC7930849 DOI: 10.1016/j.mbplus.2020.100054] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 11/25/2020] [Accepted: 11/25/2020] [Indexed: 02/07/2023] Open
Abstract
The relationship of ADAMTS (a disintegrin and metalloproteinase with thrombospondin motifs) proteases with inflammatory processes was anticipated since their discovery. Although knowledge of these extracellular proteases in different contexts continues to grow, many questions remain unanswered. In this review, we summarize the most important studies of ADAMTSs and their substrates in inflammation and in the immune system of non-oncological disorders. In addition, we update the findings on cancer and highlight their emerging role in the tumor immune microenvironment. Although the overall functions of extracellular molecules are known to be modulated by proteolysis, specific activities attributed to intact proteins and cleaved fragments in the context of inflammation are still subject to debate. A better understanding of ADAMTS activities will help to elucidate their contribution to the immune phenotype and to open up new therapeutic and diagnostic possibilities.
Collapse
|
12
|
Islam S, Watanabe H. Versican: A Dynamic Regulator of the Extracellular Matrix. J Histochem Cytochem 2020; 68:763-775. [PMID: 33131383 DOI: 10.1369/0022155420953922] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Versican is a large chondroitin sulfate/dermatan sulfate proteoglycan belonging to the aggrecan/lectican family. In adults, this proteoglycan serves as a structural macromolecule of the extracellular matrix in the brain and large blood vessels. In contrast, versican is transiently expressed at high levels during development and under pathological conditions when the extracellular matrix dramatically changes, including in the inflammation and repair process. There are many reports showing the upregulation of versican in cancer, which correlates with cancer aggressiveness. Versican has four classical splice variants, and all the variants contain G1 and G3 domains at N- and C-termini, respectively. There are two glycosaminoglycan attachment domains CSα and CSβ. The largest V0 variant contains both CSα and CSβ, V1 contains CSβ, V2 contains CSα, and the shortest G3 variant has neither of them. Versican degradation is initiated by cleavage at a site in the CSβ domain by ADAMTS (a disintegrin and metalloproteinase with thrombospondin motifs) proteinases. The N-terminal fragment containing the G1 domain has been reported to exert various biological functions, although its mechanisms of action have not yet been elucidated. In this review, we describe the role of versican in inflammation and cancer and also address the biological function of versikine.
Collapse
Affiliation(s)
- Shamima Islam
- Institute for Molecular Science of Medicine, Aichi Medical University, Nagakute, Japan
| | - Hideto Watanabe
- Institute for Molecular Science of Medicine, Aichi Medical University, Nagakute, Japan
| |
Collapse
|
13
|
Effect of endostatin on Wnt pathway of stem-like cells in bladder cancer in tumor microenvironment. Mol Biol Rep 2020; 47:3937-3948. [PMID: 32388699 DOI: 10.1007/s11033-020-05487-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 04/30/2020] [Indexed: 10/24/2022]
Abstract
Wnt/β-catenin signaling pathway modulates miscellaneous biological events in cells including gene expression, cell growth, apoptosis, metabolism and transition. The aim of this study was to investigate the effect of endostatin on Wnt signaling pathway of stem-like cells in bladder cancer in tumor microenvironment. The qRT-PCR assay and western blot were conducted to evaluate related factors expressions of Wnt signaling pathway in both bladder cancer 5637 cells and stem cells. Loss of function assays were carried out to detect the influence of endostatin on the proliferation, migration, cell proliferation and apoptosis of bladder cancer cells. We demonstrated that endostatin triggered the degradation of β-catenin, a key mediator of Wnt signaling. The activation of the endostatin blocked β-catenin function and inhibited cell growth and migration of bladder cancer. In order to verify that the Wnt/β-catenin signaling pathway was inhibited by endostain in 5637 bladder cancer cells and stem cells, the Wnt/β-catenin signaling pathway-associated molecules, including DKK1, LRP5, TCF4, β-catenin, cyclin D1, and c-Myc, were evaluated in 5637 bladder cancer cells and stem cells. The western blotting results showed that expressions of these molecules were remarkably increased in the 5637 bladder cancer cells and stem cells compared to the control group. In summary, our study demonstrated that endostatin inhibited angiogenesis. The downregulation of the Wnt/β-catenin pathway may be engaged in the suppression of angiogenesis by endostatin in bladder cancer cells and cancer stem cells.
Collapse
|
14
|
Green Tea Polyphenol EGCG Attenuates MDSCs-mediated Immunosuppression through Canonical and Non-Canonical Pathways in a 4T1 Murine Breast Cancer Model. Nutrients 2020; 12:nu12041042. [PMID: 32290071 PMCID: PMC7230934 DOI: 10.3390/nu12041042] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 03/31/2020] [Accepted: 04/06/2020] [Indexed: 01/12/2023] Open
Abstract
Several studies in the past decades have reported anti-tumor activity of the bioactive compounds extracted from tea leaves, with a focus on the compound epigallocatechin-3-gallate (EGCG). However, further investigations are required to unravel the underlying mechanisms behind the anti-tumor activity of EGCG. In this study, we demonstrate that EGCG significantly inhibits the growth of 4T1 breast cancer cells in vitro and in vivo. EGCG ameliorated immunosuppression by significantly decreasing the accumulation of myeloid-derived suppressor cells (MDSCs) and increasing the proportions of CD4+ and CD8+ T cells in spleen and tumor sites in 4T1 breast tumor-bearing mice. Surprisingly, a low dose of EGCG (0.5-5 μg/mL) effectively reduced the cell viability and increased the apoptosis rate of MDSCs in vitro. EGCG down-regulated the canonical pathways in MDSCs, mainly through the Arg-1/iNOS/Nox2/NF-κB/STAT3 signaling pathway. Moreover, transcriptomic analysis suggested that EGCG also affected the non-canonical pathways in MDSCs, such as ECM-receptor interaction and focal adhesion. qRT-PCR further validated that EGCG restored nine key genes in MDSCs, including Cxcl3, Vcan, Col4a1, Col8a1, Oasl2, Mmp12, Met, Itsnl and Acot1. Our results provide new insight into the mechanism of EGCG-associated key pathways/genes in MDSCs in the murine breast tumor model.
Collapse
|
15
|
Horn LA, Riskin J, Hempel HA, Fousek K, Lind H, Hamilton DH, McCampbell KK, Maeda DY, Zebala JA, Su Z, Schlom J, Palena C. Simultaneous inhibition of CXCR1/2, TGF-β, and PD-L1 remodels the tumor and its microenvironment to drive antitumor immunity. J Immunother Cancer 2020; 8:e000326. [PMID: 32188703 PMCID: PMC7078948 DOI: 10.1136/jitc-2019-000326] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/09/2020] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Despite the success of immune checkpoint blockade therapy in the treatment of certain cancer types, only a small percentage of patients with solid malignancies achieve a durable response. Consequently, there is a need to develop novel approaches that could overcome mechanisms of tumor resistance to checkpoint inhibition. Emerging evidence has implicated the phenomenon of cancer plasticity or acquisition of mesenchymal features by epithelial tumor cells, as an immune resistance mechanism. METHODS Two soluble factors that mediate tumor cell plasticity in the context of epithelial-mesenchymal transition are interleukin 8 (IL-8) and transforming growth factor beta (TGF-β). In an attempt to overcome escape mechanisms mediated by these cytokines, here we investigated the use of a small molecule inhibitor of the IL-8 receptors CXCR1/2, and a bifunctional agent that simultaneously blocks programmed death ligand 1 (PD-L1) and traps soluble TGF-β. RESULTS We demonstrate that simultaneous inhibition of CXCR1/2, TGF-β, and PD-L1 signaling synergizes to reduce mesenchymal tumor features in murine models of breast and lung cancer, and to markedly increase expression of tumor epithelial E-cadherin while reducing infiltration with suppressive granulocytic myeloid-derived suppressor cells, significantly enhancing T-cell infiltration and activation in tumors, and leading to improved antitumor activity. CONCLUSIONS This study highlights the potential benefit of combined blockade of CXCR1/2 and TGF-β signaling for modulation of tumor plasticity and potential enhancement of tumor responses to PD-L1 blockade. The data provide rationale for the evaluation of this novel approach in the clinic.
Collapse
Affiliation(s)
- Lucas A Horn
- Laboratory of Tumor Immunology and Biology, National Cancer Institute, Bethesda, Maryland, USA
| | - Jeffrey Riskin
- Laboratory of Tumor Immunology and Biology, National Cancer Institute, Bethesda, Maryland, USA
| | - Heidi A Hempel
- Laboratory of Tumor Immunology and Biology, National Cancer Institute, Bethesda, Maryland, USA
| | - Kristen Fousek
- Laboratory of Tumor Immunology and Biology, National Cancer Institute, Bethesda, Maryland, USA
| | - Hanne Lind
- Laboratory of Tumor Immunology and Biology, National Cancer Institute, Bethesda, Maryland, USA
| | - Duane H Hamilton
- Laboratory of Tumor Immunology and Biology, National Cancer Institute, Bethesda, Maryland, USA
| | - Kristen K McCampbell
- Laboratory of Tumor Immunology and Biology, National Cancer Institute, Bethesda, Maryland, USA
| | - Dean Y Maeda
- Syntrix Pharmaceuticals, Auburn, Washington, USA
| | | | - Zhen Su
- EMD Serono Research and Development Institute, Billerica, Massachusetts, USA
| | - Jeffrey Schlom
- Laboratory of Tumor Immunology and Biology, National Cancer Institute, Bethesda, Maryland, USA
| | - Claudia Palena
- Laboratory of Tumor Immunology and Biology, National Cancer Institute, Bethesda, Maryland, USA
| |
Collapse
|
16
|
Dos Reis DC, Damasceno KA, de Campos CB, Veloso ES, Pêgas GRA, Kraemer LR, Rodrigues MA, Mattos MS, Gomes DA, Campos PP, Ferreira E, Russo RC, Cassali GD. Versican and Tumor-Associated Macrophages Promotes Tumor Progression and Metastasis in Canine and Murine Models of Breast Carcinoma. Front Oncol 2019; 9:577. [PMID: 31334111 PMCID: PMC6616078 DOI: 10.3389/fonc.2019.00577] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 06/14/2019] [Indexed: 12/31/2022] Open
Abstract
Versican and tumor-associated macrophages (TAMs) are involved in growth and metastases in several cancers. Here, we investigated the potential role of versican, a matrix proteoglycan, and its correlation with TAMs infiltrates in different stages of two different breast cancer models: spontaneous canine mammary gland carcinomas and the murine 4T1 breast cancer model. The stromal versican expression was correlated with TAMs accumulation in tumors with an advanced stage from spontaneous canine mammary carcinoma samples. Versican expression in mice, identified in late stages of tumor progression, was associated to a high number of peri-tumoral infiltrating TAMs. Indeed, TAMs were related to a pro-inflammatory and pro-angiogenic state in the primary tumor. Furthermore, TAMs accumulation was related to versican expression in the lungs and an increased number of pulmonary metastatic nodules with pulmonary mechanical dysfunction, which was due to leukocyte influx in the airways and elevated growth factor levels in the microenvironment. Thus, we suggest that versican and TAMs as attractive targets for breast cancer therapy.
Collapse
Affiliation(s)
- Diego Carlos Dos Reis
- Department of General Pathology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.,Laboratory of Pulmonary Immunology and Mechanics, Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Cecília Bonolo de Campos
- Department of General Pathology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Emerson Soares Veloso
- Department of General Pathology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Gabriela Rafaela Arantes Pêgas
- Department of General Pathology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Lucas Rocha Kraemer
- Laboratory of Pulmonary Immunology and Mechanics, Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Michele Angela Rodrigues
- Department of General Pathology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Matheus Silvério Mattos
- Laboratory of Pulmonary Immunology and Mechanics, Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Dawidson Assis Gomes
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Paula Peixoto Campos
- Department of General Pathology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Enio Ferreira
- Department of General Pathology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Remo Castro Russo
- Laboratory of Pulmonary Immunology and Mechanics, Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Geovanni Dantas Cassali
- Department of General Pathology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
17
|
Clinical significance of the TNF-α receptors, TNFRSF2 and TNFRSF9, on cell migration molecules Fascin-1 and Versican in acute leukemia. Cytokine 2018; 111:523-529. [DOI: 10.1016/j.cyto.2018.05.025] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 05/23/2018] [Accepted: 05/29/2018] [Indexed: 02/07/2023]
|
18
|
Liu X, Han C, Liao X, Yu L, Zhu G, Su H, Qin W, Lu S, Ye X, Peng T. Genetic variants in the exon region of versican predict survival of patients with resected early-stage hepatitis B virus-associated hepatocellular carcinoma. Cancer Manag Res 2018; 10:1027-1036. [PMID: 29765250 PMCID: PMC5942399 DOI: 10.2147/cmar.s161906] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background The upregulated expression of versican (VCAN) promotes the proliferation, invasion, and metastasis of various types of human cancer cells, including hepatocellular carcinoma (HCC) cells. Patients and methods In this study, genetic variants in the exon region of VCAN were genotyped by DNA sequencing. Prognostic values of VCAN exon single nucleotide polymorphisms (SNPs) were assessed by Kaplan–Meier with the log-rank test, and uni- and multivariate Cox proportional hazard regression model. Results A total of 111 patients with resected hepatitis B virus-associated early-stage HCC were collected for genotyping VCAN exon SNPs using Sanger DNA sequencing. Haplotype analysis was performed using Haploview 4.2. Survival data were analyzed using Kaplan–Meier curves and Cox proportional hazards regression analyses. The rs2652098, rs309559, rs188703, rs160278, and rs160277 SNPs were significantly associated with overall patient survival (p<0.001, p=0.012, p=0.010, p=0.007, and p=0.007, respectively). Patients carrying the TAGTG haplotype had a poorer prognosis than those with the most common CGAAT haplotype, after adjusting for tumor size, tumor capsule, and regional invasion (adjusted hazard ratio [HR] =2.06, 95% CI: 1.27–3.34, p=0.003). Meanwhile, patients with the TAGTG haplotype and a larger tumor size or an incomplete tumor capsule had an increased risk of death, compared with the others (adjusted HR =3.00, 95% CI: 1.67–5.36, p<0.001; and adjusted HR = 1.99, 95% CI = 1.12–3.55, p = 0.02, respectively). The online database mining analysis showed that upregulated VCAN expression in HCC tissues was associated with a poor overall survival of 148 HCC patients. Conclusion Genetic variants in the exon region of VCAN were associated with overall survival in patients with resected early-stage hepatitis B virus-associated HCC, and may be a potential prognostic biomarker.
Collapse
Affiliation(s)
- Xiaoguang Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Chuangye Han
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Xiwen Liao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Long Yu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Guangzhi Zhu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Hao Su
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Wei Qin
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Sicong Lu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Xinping Ye
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Tao Peng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| |
Collapse
|
19
|
Gómez-Herrera Z, Molina-Frechero N, Damián-Matsumura P, Bologna-Molina R. Proteoglycans as potential biomarkers in odontogenic tumors. J Oral Maxillofac Pathol 2018; 22:98-103. [PMID: 29731564 PMCID: PMC5917551 DOI: 10.4103/jomfp.jomfp_151_17] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 02/12/2018] [Indexed: 12/31/2022] Open
Abstract
Proteoglycans (PGs) are essential for normal cellular development; however, alterations of their concentrations can promote tumor growth. To date, a limited number of studies report the presence of PGs in odontogenic tumors (OTs); therefore, the main purpose of this work is to gather the information published on the study of PGs. The search reported 26 articles referring to the presence of different PGs in distinct OTs from 1999 to May 2017. PGs seem to play an important role during OTs' development as they are involved in several tumor processes; however, the number of reports on the study of these molecules is low. Thus, more studies are necessary in order to gain a better understanding of the underlying pathophysiology of OTs.
Collapse
Affiliation(s)
- Zaira Gómez-Herrera
- Department of Health Care, Metropolitan Autonomous University, Xochimilco, 04960 Mexico City, Mexico
| | - Nelly Molina-Frechero
- Department of Health Care, Metropolitan Autonomous University, Xochimilco, 04960 Mexico City, Mexico
| | - Pablo Damián-Matsumura
- Department of Biology of Reproduction, Metropolitan Autonomous University, Iztapalapa, 09340 Mexico City, Mexico
| | - Ronell Bologna-Molina
- Molecular Pathology Department, School of Dentistry, University of the Republic, 19200 Montevideo, Uruguay
| |
Collapse
|
20
|
Chang MY, Kang I, Gale M, Manicone AM, Kinsella MG, Braun KR, Wigmosta T, Parks WC, Altemeier WA, Wight TN, Frevert CW. Versican is produced by Trif- and type I interferon-dependent signaling in macrophages and contributes to fine control of innate immunity in lungs. Am J Physiol Lung Cell Mol Physiol 2017; 313:L1069-L1086. [PMID: 28912382 PMCID: PMC5814701 DOI: 10.1152/ajplung.00353.2017] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 09/07/2017] [Accepted: 09/07/2017] [Indexed: 01/08/2023] Open
Abstract
Growing evidence suggests that versican is important in the innate immune response to lung infection. Our goal was to understand the regulation of macrophage-derived versican and the role it plays in innate immunity. We first defined the signaling events that regulate versican expression, using bone marrow-derived macrophages (BMDMs) from mice lacking specific Toll-like receptors (TLRs), TLR adaptor molecules, or the type I interferon receptor (IFNAR1). We show that LPS and polyinosinic-polycytidylic acid [poly(I:C)] trigger a signaling cascade involving TLR3 or TLR4, the Trif adaptor, type I interferons, and IFNAR1, leading to increased expression of versican by macrophages and implicating versican as an interferon-stimulated gene. The signaling events regulating versican are distinct from those for hyaluronan synthase 1 (HAS1) and syndecan-4 in macrophages. HAS1 expression requires TLR2 and MyD88. Syndecan-4 requires TLR2, TLR3, or TLR4 and both MyD88 and Trif. Neither HAS1 nor syndecan-4 is dependent on type I interferons. The importance of macrophage-derived versican in lungs was determined with LysM/Vcan-/- mice. These studies show increased recovery of inflammatory cells in the bronchoalveolar lavage fluid of poly(I:C)-treated LysM/Vcan-/- mice compared with control mice. IFN-β and IL-10, two important anti-inflammatory molecules, are significantly decreased in both poly(I:C)-treated BMDMs from LysM/Vcan-/- mice and bronchoalveolar lavage fluid from poly(I:C)-treated LysM/Vcan-/- mice compared with control mice. In short, type I interferon signaling regulates versican expression, and versican is necessary for type I interferon production. These findings suggest that macrophage-derived versican is an immunomodulatory molecule with anti-inflammatory properties in acute pulmonary inflammation.
Collapse
Affiliation(s)
- Mary Y Chang
- Comparative Pathology Program, Department of Comparative Medicine, University of Washington School of Medicine, Seattle, Washington
| | - Inkyung Kang
- Matrix Biology Program, Benaroya Research Institute at Virginia Mason, Seattle, Washington
| | - Michael Gale
- Department of Immunology, University of Washington School of Medicine, Seattle, Washington
| | - Anne M Manicone
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, Washington; and
| | - Michael G Kinsella
- Matrix Biology Program, Benaroya Research Institute at Virginia Mason, Seattle, Washington
| | - Kathleen R Braun
- Matrix Biology Program, Benaroya Research Institute at Virginia Mason, Seattle, Washington
| | - Tara Wigmosta
- Comparative Pathology Program, Department of Comparative Medicine, University of Washington School of Medicine, Seattle, Washington
| | - William C Parks
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, Washington; and
- Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - William A Altemeier
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, Washington; and
| | - Thomas N Wight
- Matrix Biology Program, Benaroya Research Institute at Virginia Mason, Seattle, Washington
| | - Charles W Frevert
- Comparative Pathology Program, Department of Comparative Medicine, University of Washington School of Medicine, Seattle, Washington;
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, Washington; and
| |
Collapse
|
21
|
Ricard-Blum S, Vallet SD. Fragments generated upon extracellular matrix remodeling: Biological regulators and potential drugs. Matrix Biol 2017; 75-76:170-189. [PMID: 29133183 DOI: 10.1016/j.matbio.2017.11.005] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 11/05/2017] [Accepted: 11/07/2017] [Indexed: 12/13/2022]
Abstract
The remodeling of the extracellular matrix (ECM) by several protease families releases a number of bioactive fragments, which regulate numerous biological processes such as autophagy, angiogenesis, adipogenesis, fibrosis, tumor growth, metastasis and wound healing. We review here the proteases which generate bioactive ECM fragments, their ECM substrates, the major bioactive ECM fragments, together with their biological properties and their receptors. The translation of ECM fragments into drugs is challenging and would take advantage of an integrative approach to optimize the design of pre-clinical and clinical studies. This could be done by building the contextualized interaction network of the ECM fragment repertoire including their parent proteins, remodeling proteinases, and their receptors, and by using mathematical disease models.
Collapse
Affiliation(s)
- Sylvie Ricard-Blum
- Univ Lyon, University Claude Bernard Lyon 1, CNRS, INSA Lyon, CPE, Institute of Molecular and Supramolecular Chemistry and Biochemistry, UMR 5246, F-69622 Villeurbanne cedex, France.
| | - Sylvain D Vallet
- Univ Lyon, University Claude Bernard Lyon 1, CNRS, INSA Lyon, CPE, Institute of Molecular and Supramolecular Chemistry and Biochemistry, UMR 5246, F-69622 Villeurbanne cedex, France.
| |
Collapse
|
22
|
Basso D, Gnatta E, Padoan A, Fogar P, Furlanello S, Aita A, Bozzato D, Zambon CF, Arrigoni G, Frasson C, Franchin C, Moz S, Brefort T, Laufer T, Navaglia F, Pedrazzoli S, Basso G, Plebani M. PDAC-derived exosomes enrich the microenvironment in MDSCs in a SMAD4-dependent manner through a new calcium related axis. Oncotarget 2017; 8:84928-84944. [PMID: 29156694 PMCID: PMC5689584 DOI: 10.18632/oncotarget.20863] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 08/04/2017] [Indexed: 02/04/2023] Open
Abstract
Tumor genetics and escape from immune surveillance concur in the poor prognosis of PDAC. In this study an experimental model was set up to verify whether SMAD4, deleted in about 55% PDAC and associated with poor prognosis, is involved in determining immunosuppression through Exosomes (Exo). Potential mechanisms and mediators underlying SMAD4-dependent immunosuppression were evaluated by studying intracellular calcium (Fluo-4), Exo-miRNAs (microarray) and Exo-proteins (SILAC). Two PDAC cell lines expressing (BxPC3-SMAD4+) or not-expressing (BxPC3) SMAD4 were used to prepare Exo-enriched conditioned media, employed in experiments with blood donors PBMCs. Exo expanded myeloid derived suppressor cells (gMDSC and mMDSC, flow cytometry) and altered intracellular calcium fluxes in an SMAD4 dependent manner. BxPC3-SMAD4+, but mainly BxPC3 Exo, increased calcium fluxes of PBMCs (p = 0.007) and this increased intracellular calcium trafficking characterized mMDSCs. The analysis of de-regulated Exo-miRNAs and transfection experiments revealed hsa-miR-494-3p and has-miR-1260a as potential mediators of SMAD4-associated de-regulated calcium fluxes. Eleven main biological processes were identified by the analysis of SMAD4-associated de-regulated Exo-proteins, including translation, cell adhesion, cell signaling and glycolysis. A reverse Warburg effect was observed by treating PBMCs with PDAC-derived Exo: BxPC3 Exo induced a higher glucose consumption and lactate production than BxPC3-SMAD4+ Exo. Conclusion: PDAC-derived Exo from cells with, but mainly from those without SMAD4 expression, create an immunosuppressive myeloid cell background by increasing calcium fluxes and glycolysis through the transfer of SMAD4-related differentially expressed miRNAs and proteins.
Collapse
Affiliation(s)
- Daniela Basso
- Department of Medicine - DIMED, University of Padova, Padova, Italy
| | - Elisa Gnatta
- Department of Medicine - DIMED, University of Padova, Padova, Italy
| | - Andrea Padoan
- Department of Medicine - DIMED, University of Padova, Padova, Italy
| | - Paola Fogar
- Department of Medicine - DIMED, University of Padova, Padova, Italy
| | - Sara Furlanello
- Department of Medicine - DIMED, University of Padova, Padova, Italy
| | - Ada Aita
- Department of Medicine - DIMED, University of Padova, Padova, Italy
| | - Dania Bozzato
- Department of Medicine - DIMED, University of Padova, Padova, Italy
| | | | - Giorgio Arrigoni
- Department of Biomedical Sciences, University of Padova, Padova, Italy.,Proteomic Center, University of Padova, Padova, Italy
| | - Chiara Frasson
- Department of Woman and Child Health, Oncohematology Laboratory, University of Padova, Padova, Italy
| | - Cinzia Franchin
- Department of Biomedical Sciences, University of Padova, Padova, Italy.,Proteomic Center, University of Padova, Padova, Italy
| | - Stefania Moz
- Department of Medicine - DIMED, University of Padova, Padova, Italy
| | - Thomas Brefort
- Eurofins Medigenomix GmbH, Ebersberg, Germany.,Comprehensive Biomarker Center GmbH (Recently re-named to Hummingbird Diagnostics GmbH), Heidelberg, Germany
| | - Thomas Laufer
- Comprehensive Biomarker Center GmbH (Recently re-named to Hummingbird Diagnostics GmbH), Heidelberg, Germany
| | - Filippo Navaglia
- Department of Medicine - DIMED, University of Padova, Padova, Italy
| | | | - Giuseppe Basso
- Department of Woman and Child Health, Oncohematology Laboratory, University of Padova, Padova, Italy
| | - Mario Plebani
- Department of Medicine - DIMED, University of Padova, Padova, Italy
| |
Collapse
|
23
|
Abstract
OPINION STATEMENT The survival rate for patients with advanced stages of squamous cell carcinoma of the head and neck (SCCHN) remains poor despite multimodal treatment options. Cetuximab, an anti-EGFR inhibitor, is the only FDA-approved targeted agent for this disease. Recent findings have implicated modifications of the microenvironment and, consequently, phenotypical modifications of the cancer cell, in treatment resistance mechanisms. For many years, cancer research has focused mainly on targetable sites on or inside the cancer cell. Nowadays, in preclinical and clinical studies, a greater emphasis is being placed on drugs that target the tumor microenvironment. Potential targets relate to tumor vascularization, immunology, extracellular matrix components, or cancer-associated fibroblasts. The combination of these new agents with standard treatment options is of particular interest to overcome resistance mechanisms and/or to increase treatment efficacy. Whereas antiangiogenic agents show poor clinical activity, immunotherapy seems to be a more promising tool with an objective response rate (ORR) of 20 % in patients with recurrent and/or metastatic squamous cell carcinoma (R/M SCC). Other targets, located inside the extracellular matrix or on cancer associated fibroblasts, are under preclinical investigation. These new agents all need to be tested in clinical trials alone, or in combination with standard treatment modalities, based on preclinical data. To increase our knowledge of the complex network between the cancer cell and its environment, preclinical studies should consider co-culture models, and clinical studies should incorporate a translational research objective.
Collapse
|
24
|
|
25
|
Zhang N, Liu Y, Wang Y, Zhao M, Tu L, Luo F. Decitabine reverses TGF-β1-induced epithelial-mesenchymal transition in non-small-cell lung cancer by regulating miR-200/ZEB axis. Drug Des Devel Ther 2017; 11:969-983. [PMID: 28405157 PMCID: PMC5378468 DOI: 10.2147/dddt.s129305] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVE Epithelial-mesenchymal transition (EMT) is a crucial driver of tumor progression. Tumor growth factor-beta 1 (TGF-β1) is an important factor in EMT induction in tumorigenesis. The targeting of EMT may, therefore, represent a promising approach in anticancer treatment. METHODS In this study, we determined the effect of decitabine, a DNA methyltransferase inhibitor, on TGF-β1-induced EMT in non-small-cell lung cancer (NSCLC) PC9 and A549 cells. We also assessed the involvement of the miR-200/ZEB axis. RESULTS Decitabine reversed TGF-β1-induced EMT in PC9 cells, but not in A549 cells. This phenomenon was associated with epigenetic changes in the miR-200 family, which regulated EMT by altering the expression of ZEB1 and ZEB2. TGF-β1 induced aberrant methylation in miR-200 promoters, leading to EMT in PC9 cells. Decitabine attenuated this effect and inhibited tumor cell migration in vitro and in vivo. In A549 cells, however, neither TGF-β1 nor decitabine exhibited an effect on miR-200 promoter methylation. CONCLUSION Our findings suggest that epigenetic regulation of the miR-200/ZEB axis is responsible for EMT induction by TGF-β1 in PC9 cells. Decitabine inhibits EMT in NSCLC cell PC9 through its epigenetic-based therapeutic activity.
Collapse
Affiliation(s)
- Nan Zhang
- Department of Medical Oncology, Cancer Center, Lung Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Yanyang Liu
- Department of Medical Oncology, Cancer Center, Lung Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Yuyi Wang
- Department of Medical Oncology, Cancer Center, Lung Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Maoyuan Zhao
- Department of Medical Oncology, Cancer Center, Lung Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Li Tu
- Department of Medical Oncology, Cancer Center, Lung Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Feng Luo
- Department of Medical Oncology, Cancer Center, Lung Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| |
Collapse
|
26
|
Wight TN, Frevert CW, Debley JS, Reeves SR, Parks WC, Ziegler SF. Interplay of extracellular matrix and leukocytes in lung inflammation. Cell Immunol 2017; 312:1-14. [PMID: 28077237 PMCID: PMC5290208 DOI: 10.1016/j.cellimm.2016.12.003] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 12/21/2016] [Accepted: 12/22/2016] [Indexed: 12/13/2022]
Abstract
During inflammation, leukocytes influx into lung compartments and interact with extracellular matrix (ECM). Two ECM components, versican and hyaluronan, increase in a range of lung diseases. The interaction of leukocytes with these ECM components controls leukocyte retention and accumulation, proliferation, migration, differentiation, and activation as part of the inflammatory phase of lung disease. In addition, bronchial epithelial cells from asthmatic children co-cultured with human lung fibroblasts generate an ECM that is adherent for monocytes/macrophages. Macrophages are present in both early and late lung inflammation. Matrix metalloproteinase 10 (MMP10) is induced in alveolar macrophages with injury and infection and modulates macrophage phenotype and their ability to degrade collagenous ECM components. Collectively, studies outlined in this review highlight the importance of specific ECM components in the regulation of inflammatory events in lung disease. The widespread involvement of these ECM components in the pathogenesis of lung inflammation make them attractive candidates for therapeutic intervention.
Collapse
Affiliation(s)
- Thomas N Wight
- Matrix Biology Program, Benaroya Research Institute at Virginia Mason, Seattle, WA, USA.
| | - Charles W Frevert
- Department of Comparative Medicine, University of Washington, Seattle, WA, USA
| | - Jason S Debley
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, and Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA
| | - Stephen R Reeves
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, and Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA
| | - William C Parks
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Steven F Ziegler
- Immunology Program, Benaroya Research Institute at Virginia Mason, Seattle, WA, USA
| |
Collapse
|
27
|
Liu Y, Tu L, Wang L, Long J, Wang J, Wang Y, Luo F, Cao D. The accumulation of macrophages attenuates the effect of recombinant human endostatin on lung cancer. Onco Targets Ther 2016; 9:6581-6595. [PMID: 27822063 PMCID: PMC5087788 DOI: 10.2147/ott.s114389] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Background Although anti-angiogenic therapy is widely applied clinically, its efficacy has been less than expected. Screening for regulatory factors and sensitive indicators to define the effectiveness of these drugs is required. Through a retrospective study of clinical data, we found that patients with a higher peripheral monocyte-to-lymphocyte ratio (MLR) obtained less benefit from recombinant human endostatin (rhES, Endostar®), an anti-angiogenic drug, in lung cancer. Because MLR is positively correlated with macrophage count in tumors, this result suggests that macrophages may influence the effectiveness of rhES therapy in lung cancer. Methods Clinical data from 72 lung cancer patients treated with rhES were collected. Animal study, flow cytometry, immunofluorescence, enzyme-linked immunosorbent assay, Western blot analysis, and transwell migration assays were carried on Lewis lung carcinoma (LLC) cells, bone marrow-derived macrophages, macrophage cell line RAW264.7, and ANA-1 cells. Results Clinical data showed that compared with the baseline MLR before rhES treatment, patients with progressive disease had higher MLRs than those of patients with partial response. Experimental results showed that more macrophages were recruited in the LLC tumors after rhES treatment and the majority of them displayed an M2-like phenotype. rhES aggravated hypoxia and the inflammatory response in the tumor microenvironment. Hypoxia promoted the expression of CCL2 by endothelial and fibroblast cells, which could induce macrophages recruitment, and increased levels of inflammatory cytokines (interleukin-4 [IL-4], IL-6, and IL-10) skewed macrophage polarization toward the M2-like phenotype. Hypoxia or inflammation cytokine-treated macrophages enhanced the progression of LLC in vitro and in vivo. Conclusion We found rhES could aggravate hypoxia and the inflammatory response in the tumor microenvironment. These changes were favorable for macrophage accumulation, and skewed their polarization toward the M2-like phenotype which could help LLC to escape from the anti-angiogenic therapy. Thus, these data indicate the accumulation of macrophages in the tumor microenvironment may adversely affect the efficacy of rhES on lung cancer.
Collapse
Affiliation(s)
- Yanyang Liu
- Department of Medical Oncology, Lung Cancer Center
| | - Li Tu
- Department of Medical Oncology, Lung Cancer Center
| | - Li Wang
- Department of Medical Oncology, Lung Cancer Center
| | - Jianlin Long
- Department of Medical Oncology, Lung Cancer Center
| | - Jiantao Wang
- Department of Medical Oncology, Lung Cancer Center
| | - Yuyi Wang
- Department of Medical Oncology, Lung Cancer Center
| | - Feng Luo
- Department of Medical Oncology, Lung Cancer Center
| | - Dan Cao
- Department of Abdominal Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| |
Collapse
|
28
|
Wang J, Qin Y, Mi X. The protective effects of bone marrow-derived mesenchymal stem cell (BMSC) on LPS-induced acute lung injury via TLR3-mediated IFNs, MAPK and NF-κB signaling pathways. Biomed Pharmacother 2016; 79:176-87. [PMID: 27044826 DOI: 10.1016/j.biopha.2016.02.037] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 02/16/2016] [Accepted: 02/16/2016] [Indexed: 12/29/2022] Open
Abstract
The study attempted to clarify the protective role of bone marrow-derived mesenchymal stem cell (BMSC) transplantation on LPS-induced acute lung injury (ALI) of rats. BMSC were obtained from bone marrow of rat, cultured and proliferated in vitro. Rats of ALI were established through lipopolysaccharide (LPS) administration. Male rats were allocated to control group, ALI group and BMSC, transplantation group. Rats were sacrificed after BMSC injection after 12h, 24h and 48h. Here we investigated the role of BMSC in LPS-induced alveolar macrophages to further demonstrate the mechanism of BMSC to lung injury. TLR3, a member of Toll-like receptor family, has been found in macrophages and the cell surface. In our study, first BMSC successfully reversed LPS-induced lung injury by hematoxylin-eosin (H&E) staining, ameliorated apoptosis via TUNEL and flow cytometer analysis, as well as improved cell structure. And then, western blot, quantitative real-time PCR, immunohistochemistry and immunofluorescence analysis were used to confirm that TLR3 was significantly down-regulated for BMSC treatment. Subsequently, TRIF and RIP1, down-streaming signals of TLR3, were inhibited greatly, leading to TRAF3, MAPK as well as NF-κB inactivity. Our results indicated that BMSC transplantation group displayed inhibitory effects on interferon (IFNs) levels via TLR3 in LPS-induced ALI and preventive effects on inflammation response via TLR3-regualted MAPK and NF-κB signaling pathway in LPS-induced lung injury. The present study indicated that BMSC could display protective effects on LPS-induced ALI and provide an experimental basis for clinical therapy.
Collapse
Affiliation(s)
- Jingcai Wang
- Department of Pediatrics, People's Hospital of Liaocheng, Shandong 252000, China
| | - Ying Qin
- General Hospital of Jinan Iron and Steel Group Co., Ltd., Shandong 252000, China.
| | - Xiuju Mi
- Department of Pediatrics, People's Hospital of Liaocheng, Shandong 252000, China
| |
Collapse
|
29
|
Ricard-Blum S, Vallet SD. Matricryptins Network with Matricellular Receptors at the Surface of Endothelial and Tumor Cells. Front Pharmacol 2016; 7:11. [PMID: 26869928 PMCID: PMC4740388 DOI: 10.3389/fphar.2016.00011] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 01/12/2016] [Indexed: 12/11/2022] Open
Abstract
The extracellular matrix (ECM) is a source of bioactive fragments called matricryptins or matrikines resulting from the proteolytic cleavage of extracellular proteins (e.g., collagens, elastin, and laminins) and proteoglycans (e.g., perlecan). Matrix metalloproteinases (MMPs), cathepsins, and bone-morphogenetic protein-1 release fragments, which regulate physiopathological processes including tumor growth, metastasis, and angiogenesis, a pre-requisite for tumor growth. A number of matricryptins, and/or synthetic peptides derived from them, are currently investigated as potential anti-cancer drugs both in vitro and in animal models. Modifications aiming at improving their efficiency and their delivery to their target cells are studied. However, their use as drugs is not straightforward. The biological activities of these fragments are mediated by several receptor families. Several matricryptins may bind to the same matricellular receptor, and a single matricryptin may bind to two different receptors belonging or not to the same family such as integrins and growth factor receptors. Furthermore, some matricryptins interact with each other, integrins and growth factor receptors crosstalk and a signaling pathway may be regulated by several matricryptins. This forms an intricate 3D interaction network at the surface of tumor and endothelial cells, which is tightly associated with other cell-surface associated molecules such as heparan sulfate, caveolin, and nucleolin. Deciphering the molecular mechanisms underlying the behavior of this network is required in order to optimize the development of matricryptins as anti-cancer agents.
Collapse
Affiliation(s)
- Sylvie Ricard-Blum
- University Claude Bernard Lyon 1, UMR 5246 Centre National de la Recherche Scientifique - University Lyon 1 - Institut National des Sciences Appliquées de Lyon - École Supérieure de Chimie Physique Électronique de Lyon Villeurbanne, France
| | - Sylvain D Vallet
- University Claude Bernard Lyon 1, UMR 5246 Centre National de la Recherche Scientifique - University Lyon 1 - Institut National des Sciences Appliquées de Lyon - École Supérieure de Chimie Physique Électronique de Lyon Villeurbanne, France
| |
Collapse
|
30
|
Wang Y, Jiang M, Li Z, Wang J, Du C, Yanyang L, Yu Y, Wang X, Zhang N, Zhao M, Wang L, Li M, Luo F. Hypoxia and TGF-β1 lead to endostatin resistance by cooperatively increasing cancer stem cells in A549 transplantation tumors. Cell Biosci 2015; 5:72. [PMID: 26705466 PMCID: PMC4690275 DOI: 10.1186/s13578-015-0064-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2015] [Accepted: 12/14/2015] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Lung cancer is the leading cause of cancer-related deaths worldwide, and treatments for lung cancer have a high failure rate. Anti-angiogenic therapy is also often ineffective because of refractory disease. Endostatin (ES) is one of the most widely-used anti-angiogenic drugs for lung cancer in China, and resistance to it is a barrier that needs to be resolved. It has been shown that myeloid-derived suppressor cells (MDSCs) are involved in resistance to ES. Whether other cells and/or cell factors in the tumor microenvironment that have been shown to be related to resistance to other anti-cancer drugs are also involved in ES resistance is unknown. RESULTS In this study, we showed that after continuously treatment with ES for 12 days, volumes of A549 transplantation tumors of mice reached the sizes of tumors which were borne by mice that were treated with normal saline and this meant that resistance to ES appeared. Cancer stem cells (CSCs), which have been widely accepted as one of reasons responsible for resistance to many anti-tumor drugs were also being discovered increased proportionally in A549 transplantation tumors after ES treatment for 12 days. During further exploration of reasons for this increase, we discovered that after ES treatment, microvessel density and vascular endothelial growth factor level was decreased in tumors, whereas transforming growth factor (TGF)-β1 level was elevated, and MDSCs, one of the sources of TGF-β1, were also increased. We speculate that hypoxia and TGF-β1 are responsible for the increased CSC number in A549 transplantation tumors. By using cobalt chloride to mimic hypoxia and human recombinant TGF-β1 in vitro, we found that hypoxia and TGF-β1can indeed enhance the stemness of A549 cells. In addition, the inductive effect of hypoxia is stronger than TGF-β1, and the combination of both is stronger than either alone, which is first report of such a finding, to our knowledge. CONCLUSIONS Increased TGF-β1 and strengthened hypoxia in A549 transplantation tumors, as a result of ES therapy, cooperatively increase proportion of CSCs that are involved in ES resistance which was revealed by failure of tumor volume repression after continuously treatment with ES for 12 days.
Collapse
Affiliation(s)
- Yuyi Wang
- Department of Medical Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan 61004 People's Republic of China
| | - Ming Jiang
- Department of Medical Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan 61004 People's Republic of China
| | - Zhixi Li
- Department of Medical Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan 61004 People's Republic of China
| | - Jiantao Wang
- Department of Medical Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan 61004 People's Republic of China
| | - Chi Du
- Department of Medical Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan 61004 People's Republic of China.,Department of Oncology, The Second People's Hospital of Neijiang, Neijiang, Sichuan 641000 People's Republic of China
| | - Liu Yanyang
- Department of Medical Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan 61004 People's Republic of China
| | - Yang Yu
- Department of Medical Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan 61004 People's Republic of China
| | - Xia Wang
- Department of Medical Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan 61004 People's Republic of China
| | - Nan Zhang
- Department of Medical Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan 61004 People's Republic of China
| | - Maoyuan Zhao
- Department of Medical Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan 61004 People's Republic of China
| | - Li Wang
- Department of Medical Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan 61004 People's Republic of China
| | - Mei Li
- Department of Medical Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan 61004 People's Republic of China
| | - Feng Luo
- Department of Medical Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan 61004 People's Republic of China
| |
Collapse
|
31
|
Novel delivery approaches for cancer therapeutics. J Control Release 2015; 219:248-268. [PMID: 26456750 DOI: 10.1016/j.jconrel.2015.09.067] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 09/09/2015] [Accepted: 09/30/2015] [Indexed: 02/07/2023]
Abstract
Currently, a majority of cancer treatment strategies are based on the removal of tumor mass mainly by surgery. Chemical and physical treatments such as chemo- and radiotherapies have also made a major contribution in inhibiting rapid growth of malignant cells. Furthermore, these approaches are often combined to enhance therapeutic indices. It is widely known that surgery, chemo- and radiotherapy also inhibit normal cells growth. In addition, these treatment modalities are associated with severe side effects and high toxicity which in turn lead to low quality of life. This review encompasses novel strategies for more effective chemotherapeutic delivery aiming to generate better prognosis. Currently, cancer treatment is a highly dynamic field and significant advances are being made in the development of novel cancer treatment strategies. In contrast to conventional cancer therapeutics, novel approaches such as ligand or receptor based targeting, triggered release, intracellular drug targeting, gene delivery, cancer stem cell therapy, magnetic drug targeting and ultrasound-mediated drug delivery, have added new modalities for cancer treatment. These approaches have led to selective detection of malignant cells leading to their eradication with minimal side effects. Lowering multi-drug resistance and involving influx transportation in targeted drug delivery to cancer cells can also contribute significantly in the therapeutic interventions in cancer.
Collapse
|