1
|
Pan T, Liu F, Hao X, Wang S, Wasi M, Song JH, Lewis VO, Lin PP, Moon B, Bird JE, Panaretakis T, Lin SH, Wu D, Farach-Carson MC, Wang L, Zhang N, An Z, Zhang XHF, Satcher RL. BIGH3 mediates apoptosis and gap junction failure in osteocytes during renal cell carcinoma bone metastasis progression. Cancer Lett 2024; 596:217009. [PMID: 38849015 DOI: 10.1016/j.canlet.2024.217009] [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: 03/17/2024] [Revised: 05/26/2024] [Accepted: 05/30/2024] [Indexed: 06/09/2024]
Abstract
Renal cell carcinoma (RCC) bone metastatis progression is driven by crosstalk between tumor cells and the bone microenvironment, which includes osteoblasts, osteoclasts, and osteocytes. RCC bone metastases (RCCBM) are predominantly osteolytic and resistant to antiresorptive therapy. The molecular mechanisms underlying pathologic osteolysis and disruption of bone homeostasis remain incompletely understood. We previously reported that BIGH3/TGFBI (transforming growth factor-beta-induced protein ig-h3, shortened to BIGH3 henceforth) secreted by colonizing RCC cells drives osteolysis by inhibiting osteoblast differentiation, impairing healing of osteolytic lesions, which is reversible with osteoanabolic agents. Here, we report that BIGH3 induces osteocyte apoptosis in both human RCCBM tissue specimens and in a preclinical mouse model. We also demonstrate that BIGH3 reduces Cx43 expression, blocking gap junction (GJ) function and osteocyte network communication. BIGH3-mediated GJ inhibition is blocked by the lysosomal inhibitor hydroxychloroquine (HCQ), but not osteoanabolic agents. Our results broaden the understanding of pathologic osteolysis in RCCBM and indicate that targeting the BIGH3 mechanism could be a combinational strategy for the treatment of RCCBM-induced bone disease that overcomes the limited efficacy of antiresorptives that target osteoclasts.
Collapse
Affiliation(s)
- Tianhong Pan
- Departments of Orthopedic Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Fengshuo Liu
- Departments of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Xiaoxin Hao
- Departments of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Shubo Wang
- Department of Mechanical Engineering, University of Delaware, Newark, DE, USA
| | - Murtaza Wasi
- Department of Mechanical Engineering, University of Delaware, Newark, DE, USA
| | - Jian H Song
- Departments of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Valerae O Lewis
- Departments of Orthopedic Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Patrick P Lin
- Departments of Orthopedic Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bryan Moon
- Departments of Orthopedic Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Justin E Bird
- Departments of Orthopedic Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Theocharis Panaretakis
- Departments of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sue-Hwa Lin
- Departments of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Departments of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Danielle Wu
- Department of Diagnostic and Biomedical Sciences, The University of Texas Health Science Center at Houston, School of Dentistry, Houston, TX, USA; Departments of Bioengineering, Rice University, Houston, TX, USA
| | - Mary C Farach-Carson
- Department of Diagnostic and Biomedical Sciences, The University of Texas Health Science Center at Houston, School of Dentistry, Houston, TX, USA; Departments of BioSciences, Rice University, Houston, TX, USA; Departments of Bioengineering, Rice University, Houston, TX, USA
| | - Liyun Wang
- Department of Mechanical Engineering, University of Delaware, Newark, DE, USA
| | - Ningyan Zhang
- The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, USA
| | - Zhiqiang An
- The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, USA
| | - Xiang H-F Zhang
- Departments of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA; Departments of Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA; Departments of Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA; Departments of McNair Medical Institute, Baylor College of Medicine, Houston, TX, USA
| | - Robert L Satcher
- Departments of Orthopedic Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
2
|
Nasimi Shad A, Moghbeli M. Integrins as the pivotal regulators of cisplatin response in tumor cells. Cell Commun Signal 2024; 22:265. [PMID: 38741195 DOI: 10.1186/s12964-024-01648-0] [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: 01/24/2024] [Accepted: 05/04/2024] [Indexed: 05/16/2024] Open
Abstract
Cisplatin (CDDP) is a widely used first-line chemotherapeutic drug in various cancers. However, CDDP resistance is frequently observed in cancer patients. Therefore, it is required to evaluate the molecular mechanisms associated with CDDP resistance to improve prognosis among cancer patients. Integrins are critical factors involved in tumor metastasis that regulate cell-matrix and cell-cell interactions. They modulate several cellular mechanisms including proliferation, invasion, angiogenesis, polarity, and chemo resistance. Modification of integrin expression levels can be associated with both tumor progression and inhibition. Integrins are also involved in drug resistance of various solid tumors through modulation of the tumor cell interactions with interstitial matrix and extracellular matrix (ECM). Therefore, in the present review we discussed the role of integrin protein family in regulation of CDDP response in tumor cells. It has been reported that integrins mainly promoted the CDDP resistance through interaction with PI3K/AKT, MAPK, and WNT signaling pathways. They also regulated the CDDP mediated apoptosis in tumor cells. This review paves the way to suggest the integrins as the reliable therapeutic targets to improve CDDP response in tumor cells.
Collapse
Affiliation(s)
- Arya Nasimi Shad
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Meysam Moghbeli
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
3
|
Miyako S, Koma YI, Nakanishi T, Tsukamoto S, Yamanaka K, Ishihara N, Azumi Y, Urakami S, Shimizu M, Kodama T, Nishio M, Shigeoka M, Kakeji Y, Yokozaki H. Periostin in Cancer-Associated Fibroblasts Promotes Esophageal Squamous Cell Carcinoma Progression by Enhancing Cancer and Stromal Cell Migration. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:828-848. [PMID: 38320632 DOI: 10.1016/j.ajpath.2023.12.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 12/15/2023] [Accepted: 12/27/2023] [Indexed: 02/08/2024]
Abstract
Cancer-associated fibroblasts (CAFs) in the tumor microenvironment are involved in the progression of various cancers, including esophageal squamous cell carcinoma (ESCC). CAF-like cells were generated through direct co-culture of human bone marrow-derived mesenchymal stem cells, one of CAF origins, with ESCC cells. Periostin (POSTN) was found to be highly expressed in CAF-like cells. After direct co-culture, ESCC cells showed increased malignant phenotypes, such as survival, growth, and migration, as well as increased phosphorylation of Akt and extracellular signal-regulated kinase (Erk). Recombinant human POSTN activated Akt and Erk signaling pathways in ESCC cells, enhancing survival and migration. The suppression of POSTN in CAF-like cells by siRNA during direct co-culture also suppressed enhanced survival and migration in ESCC cells. In ESCC cells, knockdown of POSTN receptor integrin β4 inhibited Akt and Erk phosphorylation, and survival and migration increased by POSTN. POSTN also enhanced mesenchymal stem cell and macrophage migration and endowed macrophages with tumor-associated macrophage-like properties. Immunohistochemistry showed that high POSTN expression in the cancer stroma was significantly associated with tumor invasion depth, lymphatic and blood vessel invasion, higher pathologic stage, CAF marker expression, and infiltrating tumor-associated macrophage numbers. Moreover, patients with ESCC with high POSTN expression exhibited poor postoperative outcomes. Thus, CAF-secreted POSTN contributed to tumor microenvironment development. These results indicate that POSTN may be a novel therapeutic target for ESCC.
Collapse
Affiliation(s)
- Shoji Miyako
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe, Japan; Division of Gastro-intestinal Surgery, Department of Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yu-Ichiro Koma
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe, Japan.
| | - Takashi Nakanishi
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe, Japan; Division of Gastro-intestinal Surgery, Department of Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Shuichi Tsukamoto
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Keitaro Yamanaka
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe, Japan; Division of Obstetrics and Gynecology, Department of Surgery Related, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Nobuaki Ishihara
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe, Japan; Division of Hepato-Biliary-Pancreatic Surgery, Department of Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yuki Azumi
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe, Japan; Division of Gastro-intestinal Surgery, Department of Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Satoshi Urakami
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe, Japan; Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Masaki Shimizu
- Division of Gastro-intestinal Surgery, Department of Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Takayuki Kodama
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Mari Nishio
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Manabu Shigeoka
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yoshihiro Kakeji
- Division of Gastro-intestinal Surgery, Department of Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Hiroshi Yokozaki
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
4
|
Gong Y, Pang H, Yu Z, Wang X, Li P, Zhang Q. Construction of inflammatory associated risk gene prognostic model of NSCLC and its correlation with chemotherapy sensitivity. Ann Med 2023; 55:2200034. [PMID: 37083272 PMCID: PMC10124980 DOI: 10.1080/07853890.2023.2200034] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/22/2023] Open
Abstract
BACKGROUND Inflammation is an important pathogenic factor of most malignant tumors. It is essential to understand mechanism underlying inflammation and cancer development, so as to formulate and develop anti-cancer treatment strategies. However, inflammatory-related gene characterization as well as risk model construction in prognosis and response chemotherapy or immunotherapy in NSCLC are still remain unclear. METHODS A total of 1014 lung cancer samples with RNA-seqencing results were download from The Cancer Genome Atlas (TCGA) database. The patient cohort was randomized as a training and test cohorts, and 200 inflammatory-related genes were selected based on previously published data. Consensus clustering and Enrichment and immune function analyses base on Differential expression genes (DEGs) were performed. Prognosis Prediction Model were Constructed and Chemotherapy and immunotherapy sensitivity base on this model were performed. At last, H1299 and HCC827 cells were used to tested the mitoxantrone and oxal iplatin sensitivity after KRT6A knockdown. RESULTS We identified the inflammatory-related genes from NSCLC datasets to build one prognosis prediction signature based on cluster inflammatory-related genes to lay a certain foundation for distinguishing high-risk NSCLC cases with dismal prognostic outcome. The nomogram provides the AUC values for 1-, 3-, and 5-year overall survival were 0.831, 0.853, and 0.86 in validation cohort. Morover, different sensitivity of immunotherapy or chemotherapy also were classified base on the different risk groups in NSCLC patients, which provided potent clinical reference. At last, targeting KRT6A sensitive to mitoxantrone and oxaliplatin in H1299 and HCC827 cells. CONCLUSIONS Inflammatory-related gene risk-score is the potential chemotherapeutic and immunotherapeutic biomarker for NSCLC, and targeting KRT6A sensitive to mitoxantrone and oxaliplatin in NSCLC.HighlightsInflammatory-related genes can lay a certain foundation for distinguishing high-risk NSCLC cases with dismal prognostic outcome.Risk-score base on inflammatory-related genes is positive correlated with CD274, TGFBR1 and TGFB1 expression.Targeting KRT6A sensitive to mitoxantrone and oxaliplatin in H1299 and HCC827 cells.
Collapse
Affiliation(s)
- Yange Gong
- Department of pulmonary and Critical Care Medicine (PCCM) Ward II, Cangzhou Central Hospital, Cangzhou City, Hebei Province, China
| | - Hongyan Pang
- Department of pulmonary and Critical Care Medicine (PCCM) Ward II, Cangzhou Central Hospital, Cangzhou City, Hebei Province, China
| | - Zhiqiang Yu
- Department of pulmonary and Critical Care Medicine (PCCM) Ward II, Cangzhou Central Hospital, Cangzhou City, Hebei Province, China
| | - Xue Wang
- Department of pulmonary and Critical Care Medicine (PCCM) Ward II, Cangzhou Central Hospital, Cangzhou City, Hebei Province, China
| | - Ping Li
- Department of pulmonary and Critical Care Medicine (PCCM) Ward II, Cangzhou Central Hospital, Cangzhou City, Hebei Province, China
| | - Qianyun Zhang
- Department of pulmonary and Critical Care Medicine (PCCM) Ward II, Cangzhou Central Hospital, Cangzhou City, Hebei Province, China
| |
Collapse
|
5
|
Yuan Z, Li Y, Zhang S, Wang X, Dou H, Yu X, Zhang Z, Yang S, Xiao M. Extracellular matrix remodeling in tumor progression and immune escape: from mechanisms to treatments. Mol Cancer 2023; 22:48. [PMID: 36906534 PMCID: PMC10007858 DOI: 10.1186/s12943-023-01744-8] [Citation(s) in RCA: 221] [Impact Index Per Article: 110.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 02/11/2023] [Indexed: 03/13/2023] Open
Abstract
The malignant tumor is a multi-etiological, systemic and complex disease characterized by uncontrolled cell proliferation and distant metastasis. Anticancer treatments including adjuvant therapies and targeted therapies are effective in eliminating cancer cells but in a limited number of patients. Increasing evidence suggests that the extracellular matrix (ECM) plays an important role in tumor development through changes in macromolecule components, degradation enzymes and stiffness. These variations are under the control of cellular components in tumor tissue via the aberrant activation of signaling pathways, the interaction of the ECM components to multiple surface receptors, and mechanical impact. Additionally, the ECM shaped by cancer regulates immune cells which results in an immune suppressive microenvironment and hinders the efficacy of immunotherapies. Thus, the ECM acts as a barrier to protect cancer from treatments and supports tumor progression. Nevertheless, the profound regulatory network of the ECM remodeling hampers the design of individualized antitumor treatment. Here, we elaborate on the composition of the malignant ECM, and discuss the specific mechanisms of the ECM remodeling. Precisely, we highlight the impact of the ECM remodeling on tumor development, including proliferation, anoikis, metastasis, angiogenesis, lymphangiogenesis, and immune escape. Finally, we emphasize ECM "normalization" as a potential strategy for anti-malignant treatment.
Collapse
Affiliation(s)
- Zhennan Yuan
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Yingpu Li
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Sifan Zhang
- Department of Neurobiology, Harbin Medical University, Harbin, 150081, China
| | - Xueying Wang
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - He Dou
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Xi Yu
- Department of Gynecological Oncology, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Zhiren Zhang
- NHC Key Laboratory of Cell Transplantation, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China.,Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Heilongjiang Key Laboratory for Metabolic Disorder and Cancer Related Cardiovascular Diseases, Harbin, 150001, China
| | - Shanshan Yang
- Department of Gynecological Radiotherapy, Harbin Medical University Cancer Hospital, Harbin, 150000, China.
| | - Min Xiao
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China.
| |
Collapse
|
6
|
Zhu K, Cai Y, Si X, Ye Z, Gao Y, Liu C, Wang R, Ma Z, Zhu H, Zhang L, Li S, Zhang H, Yue J. The phosphorylation and dephosphorylation switch of VCP/p97 regulates the architecture of centrosome and spindle. Cell Death Differ 2022; 29:2070-2088. [PMID: 35430615 PMCID: PMC9525716 DOI: 10.1038/s41418-022-01000-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 01/30/2022] [Accepted: 03/28/2022] [Indexed: 01/27/2023] Open
Abstract
The proper orientation of centrosome and spindle is essential for genome stability; however, the mechanism that governs these processes remains elusive. Here, we demonstrated that polo-like kinase 1 (Plk1), a key mitotic kinase, phosphorylates residue Thr76 in VCP/p97 (an AAA-ATPase), at the centrosome from prophase to anaphase. This phosphorylation process recruits VCP to the centrosome and in this way, it regulates centrosome orientation. VCP exhibits strong co-localization with Eg5 (a mitotic kinesin motor), at the mitotic spindle, and the dephosphorylation of Thr76 in VCP is required for the enrichment of both VCP and Eg5 at the spindle, thus ensuring proper spindle architecture and chromosome segregation. We also showed that the phosphatase, PTEN, is responsible for the dephosphorylation of Thr76 in VCP; when PTEN was knocked down, the normal spread of VCP from the centrosome to the spindle was abolished. Cryo-EM structures of VCPT76A and VCPT76E, which represent dephosphorylated and phosphorylated states of VCP, respectively, revealed that the Thr76 phosphorylation modulates VCP by altering the inter-domain and inter-subunit interactions, and ultimately the nucleotide-binding pocket conformation. Interestingly, the tumor growth in nude mice implanted with VCPT76A-reconstituted cancer cells was significantly slower when compared with those implanted with VCPWT-reconstituted cancer cells. Collectively, our findings demonstrate that the phosphorylation and dephosphorylation switch of VCP regulates the architecture of centrosome and spindle for faithful chromosome segregation.
Collapse
Affiliation(s)
- Kaiyuan Zhu
- City University of Hong Kong Shenzhen Research Institute, Shenzhen, China
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Yang Cai
- Department of Biology, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Xiaotong Si
- City University of Hong Kong Shenzhen Research Institute, Shenzhen, China
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Zuodong Ye
- City University of Hong Kong Shenzhen Research Institute, Shenzhen, China
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Yuanzhu Gao
- Department of Biology, SUSTech Cryo-EM Centre, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Chuang Liu
- Department of Biology, SUSTech Cryo-EM Centre, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Rui Wang
- City University of Hong Kong Shenzhen Research Institute, Shenzhen, China
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Zhibin Ma
- City University of Hong Kong Shenzhen Research Institute, Shenzhen, China
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Huazhang Zhu
- City University of Hong Kong Shenzhen Research Institute, Shenzhen, China
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Liang Zhang
- City University of Hong Kong Shenzhen Research Institute, Shenzhen, China
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Shengjin Li
- The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Hongmin Zhang
- Department of Biology, Southern University of Science and Technology, Shenzhen, 518055, China.
| | - Jianbo Yue
- City University of Hong Kong Shenzhen Research Institute, Shenzhen, China.
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China.
| |
Collapse
|
7
|
Yang K, Huang N, Sun J, Dai W, Chen M, Zeng J. Transforming growth factor-β induced protein regulates pulmonary fibrosis via the G-protein signaling modulator 2 /Snail axis. Peptides 2022; 155:170842. [PMID: 35872259 DOI: 10.1016/j.peptides.2022.170842] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 07/19/2022] [Accepted: 07/20/2022] [Indexed: 12/01/2022]
Abstract
Pulmonary fibrosis, a severe condition that can progress to respiratory failure and death, is characterized by aberrant activation/proliferation of fibroblasts and excessive extracellular matrix (ECM) deposition and has limited therapeutic options. Identifying novel mediators of pulmonary fibrosis is currently needed to facilitate the development of more effective therapeutic strategies targeting pulmonary fibrosis. The present study was designed to investigate whether transforming growth factor-β (TGF-β) induced protein (TGFBI), an extracellular matrix protein, regulates pulmonary fibrosis in vitro and in vivo and the possible mechanism of actions. It was found that protein expressions of TGFBI were significantly upregulated and G-protein signaling modulator 2 (GPSM2) expression downregulated in fibrotic lung tissues from bleomycin (BLM)-induced rats and TGF-β1-stimulated human lung IMR-90 fibroblasts. Either silencing TGFBI with specific siRNA or treatment with the TGF-β signaling inhibitor SB431542 significantly inhibited TGF-β1-induced fibrotic effects and dysregulation of GPSM2 and Snail expressions in IMR-90 fibroblasts. Moreover, GPSM2 overexpression also inhibited TGF-β1-induced fibrotic effects and Snail upregulation in IMR-90 fibroblasts. Silencing Snail with specific siRNA attenuated TGF-β1-induced fibrotic effects. Therefore, our findings suggest that the extracellular matrix protein TGFBI mediates pulmonary fibrosis through regulation of the GPSM2/Snail axis, which identifies TGFBI as a novel mediator of pulmonary fibrosis and may be a potential therapeutic target for the treatment of pulmonary fibrosis.
Collapse
Affiliation(s)
- Kai Yang
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Chengdu Medical College, No. 278, Baoguang Avenue, Xindu District, Chengdu, Sichuan 610500, China; Chengdu Medical College, No. 783, Xindu Avenue, Xindu District, Chengdu, Sichuan 610500, China
| | - Na Huang
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Chengdu Medical College, No. 278, Baoguang Avenue, Xindu District, Chengdu, Sichuan 610500, China; Chengdu Medical College, No. 783, Xindu Avenue, Xindu District, Chengdu, Sichuan 610500, China
| | - Jian Sun
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Chengdu Medical College, No. 278, Baoguang Avenue, Xindu District, Chengdu, Sichuan 610500, China
| | - Wenjing Dai
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Chengdu Medical College, No. 278, Baoguang Avenue, Xindu District, Chengdu, Sichuan 610500, China
| | - Meifeng Chen
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Chengdu Medical College, No. 278, Baoguang Avenue, Xindu District, Chengdu, Sichuan 610500, China
| | - Jun Zeng
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Chengdu Medical College, No. 278, Baoguang Avenue, Xindu District, Chengdu, Sichuan 610500, China; Chengdu Medical College, No. 783, Xindu Avenue, Xindu District, Chengdu, Sichuan 610500, China.
| |
Collapse
|
8
|
Nicolescu C, Vaidya A, Schilb A, Lu ZR. Regulating Oncogenic LncRNA DANCR with Targeted ECO/siRNA Nanoparticles for Non-Small Cell Lung Cancer Therapy. ACS OMEGA 2022; 7:22743-22753. [PMID: 35811871 PMCID: PMC9260776 DOI: 10.1021/acsomega.2c02260] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 05/04/2022] [Indexed: 05/07/2023]
Abstract
Long noncoding RNA (lncRNA) differentiation antagonizing noncoding RNA (DANCR) is a proven oncogenic lncRNA across multiple cancer types. Its effects on cancer cell migration and invasion position it as a potential target for therapy on multiple levels of gene regulation. DANCR is overexpressed in non-small cell lung cancer (NSCLC), the most common lung cancer subtype with poor patient survival. To effectively deliver small interfering RNA (siRNA) against DANCR for NSCLC therapy, we used arginine-glycine-aspartic acid (RGD)-poly(ethylene glycol) (PEG)-(1-aminoethyl)-iminobis[N-oleicylcysteinyl-1-aminoethyl)propionamide] (ECO)/small interfering RNA against DANCR (siDANCR) nanoparticles to transfect A549 and NCI-H1299 cells. Over 90% DANCR silencing was observed along with inhibition of cell migration, invasion, and spheroid formation relative to transfection with negative control siRNA in RGD-PEG-ECO nanoparticles. DANCR knockdown further showed efficacy in reducing migration and invasion of epidermal growth factor receptor (EGFR)-inhibitor resistant NSCLC along with resensitization to the inhibitor. RGD-PEG-ECO/siDANCR demonstrated silencing for up to 7 d following a single transfection. The results suggest nanoparticle-mediated RNA interference against DANCR as a potential approach for NSCLC treatment by regulating cell migration and invasion in addition to improving EGFR inhibitor response.
Collapse
Affiliation(s)
- Calin Nicolescu
- Department
of Biomedical Engineering, Case Western
Reserve University, Cleveland, Ohio 44106, United States
| | - Amita Vaidya
- Department
of Biomedical Engineering, Case Western
Reserve University, Cleveland, Ohio 44106, United States
| | - Andrew Schilb
- Department
of Biomedical Engineering, Case Western
Reserve University, Cleveland, Ohio 44106, United States
| | - Zheng-Rong Lu
- Department
of Biomedical Engineering, Case Western
Reserve University, Cleveland, Ohio 44106, United States
- Case
Comprehensive Cancer Center, Case Western
Reserve University, Cleveland, Ohio 44106, United States
| |
Collapse
|
9
|
Chen Y, Zhao H, Feng Y, Ye Q, Hu J, Guo Y, Feng Y. Pan-Cancer Analysis of the Associations of TGFBI Expression With Prognosis and Immune Characteristics. Front Mol Biosci 2021; 8:745649. [PMID: 34671645 PMCID: PMC8521171 DOI: 10.3389/fmolb.2021.745649] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 09/17/2021] [Indexed: 01/25/2023] Open
Abstract
Transforming growth factor-beta-induced (TGFBI) protein has important roles in tumor growth, metastasis, and immunity. However, there is currently no pan-cancer evidence regarding TGFBI. In this study, we conducted a pan-cancer analysis of TGFBI mRNA and protein expression and prognoses of various cancer types using public databases. We also investigated the associations of TGFBI expression with tumor microenvironment (TME) components, immune cell infiltration, tumor mutational burden (TMB), and microsatellite instability (MSI), along with the TGFBI genetic alteration types. The results showed that TGFBI expression varied among different cancer types, and it was positively or negatively related to prognosis in various cancers. TGFBI expression was also significantly correlated with TME components, TMB, MSI, immune cell infiltration, and immunoinhibitory and immunostimulatory gene subsets. These findings indicate that TGFBI participates in various immune responses and it may function as a prognostic marker in various cancers. The findings may be useful for developing immunotherapies that target TGFBI.
Collapse
Affiliation(s)
- Yun Chen
- Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Han Zhao
- Department of Ophthalmology, Eye, Ear, Nose, and Throat Hospital of Fudan University, Shanghai, China
- Laboratory of Myopia, NHC Key Laboratory of Myopia (Fudan University), Chinese Academy of Medical Sciences, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai, China
| | - Yao Feng
- Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Qin Ye
- Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jing Hu
- Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yue Guo
- Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yunzhi Feng
- Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
10
|
Zheng B, Wu Q, Jiang Y, Hou M, Zhang P, Liu M, Zhang L, Li B, Zhang C. One-pot synthesis of 68Ga-doped ultrasmall gold nanoclusters for PET/CT imaging of tumors. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 128:112291. [PMID: 34474842 DOI: 10.1016/j.msec.2021.112291] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 06/19/2021] [Accepted: 06/28/2021] [Indexed: 10/20/2022]
Abstract
Gold nanoclusters (AuNCs) have attracted much attention for tumor theranostics in recent years because of their ability of renal clearance and to escape the reticuloendothelial system (RES) sequestration. In this study, we presented a novel method to synthesize 68Ga-doped (labeled) AuNCs by simultaneous reduction of 68GaCl3 and HAuCl4 by glutathione. As synthesized 68Ga-doped, glutathione-coated AuNCs (68Ga-GSH@AuNCs) were ultrasmall in size (<2 nm), highly stable under physiological conditions and renally clearable, and had high efficiency for tumor targeting. To demonstrate the universality of this 68Ga labeling method and further enhance tumor targeting efficiency, arginine-glycine-aspartate (RGD)-containing peptide was introduced as co-reductant to synthesize RGD peptide and glutathione co-coated, 68Ga-labeled AuNCs (68Ga-RGD-GSH@AuNCs). Introduction of RGD peptide did not interfere the synthesis process but significantly enhanced the tumor targeting efficiency of the AuNCs. Our study demonstrated that it was feasible to label AuNCs with gallium-68 by direct reduction of the radioisotope and HAuCl4 with reductant peptides, holding a great potential for clinical translation for PET/CT detection of tumors.
Collapse
Affiliation(s)
- Benchao Zheng
- Department of Nuclear Medicine, Rui Jin Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Qinghe Wu
- Department of Nuclear Medicine, Rui Jin Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Yifei Jiang
- Department of Nuclear Medicine, Rui Jin Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Mengfei Hou
- Department of Nuclear Medicine, Rui Jin Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Pengli Zhang
- Department of Nuclear Medicine, Rui Jin Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Meirong Liu
- Department of Nuclear Medicine, Rui Jin Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Lu Zhang
- Department of Nuclear Medicine, Rui Jin Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Biao Li
- Department of Nuclear Medicine, Rui Jin Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Chunfu Zhang
- Department of Nuclear Medicine, Rui Jin Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China.
| |
Collapse
|
11
|
Sedlář A, Trávníčková M, Bojarová P, Vlachová M, Slámová K, Křen V, Bačáková L. Interaction between Galectin-3 and Integrins Mediates Cell-Matrix Adhesion in Endothelial Cells and Mesenchymal Stem Cells. Int J Mol Sci 2021; 22:ijms22105144. [PMID: 34067978 PMCID: PMC8152275 DOI: 10.3390/ijms22105144] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 05/07/2021] [Accepted: 05/09/2021] [Indexed: 12/20/2022] Open
Abstract
Galectin-3 (Gal-3) is a β-galactoside-binding protein that influences various cell functions, including cell adhesion. We focused on the role of Gal-3 as an extracellular ligand mediating cell-matrix adhesion. We used human adipose tissue-derived stem cells and human umbilical vein endothelial cells that are promising for vascular tissue engineering. We found that these cells naturally contained Gal-3 on their surface and inside the cells. Moreover, they were able to associate with exogenous Gal-3 added to the culture medium. This association was reduced with a β-galactoside LacdiNAc (GalNAcβ1,4GlcNAc), a selective ligand of Gal-3, which binds to the carbohydrate recognition domain (CRD) in the Gal-3 molecule. This ligand was also able to detach Gal-3 newly associated with cells but not Gal-3 naturally present on cells. In addition, Gal-3 preadsorbed on plastic surfaces acted as an adhesion ligand for both cell types, and the cell adhesion was resistant to blocking with LacdiNAc. This result suggests that the adhesion was mediated by a binding site different from the CRD. The blocking of integrin adhesion receptors on cells with specific antibodies revealed that the cell adhesion to the preadsorbed Gal-3 was mediated, at least partially, by β1 and αV integrins-namely α5β1, αVβ3, and αVβ1 integrins.
Collapse
Affiliation(s)
- Antonín Sedlář
- Laboratory of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Vídeňská 1083, CZ 142 20 Prague 4, Czech Republic; (A.S.); (M.T.)
- Department of Physiology, Faculty of Science, Charles University, Viničná 7, CZ 128 44 Prague 2, Czech Republic
| | - Martina Trávníčková
- Laboratory of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Vídeňská 1083, CZ 142 20 Prague 4, Czech Republic; (A.S.); (M.T.)
| | - Pavla Bojarová
- Laboratory of Biotransformation, Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, CZ 142 20 Prague 4, Czech Republic; (M.V.); (K.S.); (V.K.)
- Department of Health Care Disciplines and Population Protection, Faculty of Biomedical Engineering, Czech Technical University in Prague, Nám. Sítná, CZ 272 01 Kladno, Czech Republic
- Correspondence: (P.B.); (L.B.); Tel.: +420-296442360 (P.B.); +420-296443743 (L.B.)
| | - Miluše Vlachová
- Laboratory of Biotransformation, Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, CZ 142 20 Prague 4, Czech Republic; (M.V.); (K.S.); (V.K.)
| | - Kristýna Slámová
- Laboratory of Biotransformation, Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, CZ 142 20 Prague 4, Czech Republic; (M.V.); (K.S.); (V.K.)
| | - Vladimír Křen
- Laboratory of Biotransformation, Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, CZ 142 20 Prague 4, Czech Republic; (M.V.); (K.S.); (V.K.)
| | - Lucie Bačáková
- Laboratory of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Vídeňská 1083, CZ 142 20 Prague 4, Czech Republic; (A.S.); (M.T.)
- Correspondence: (P.B.); (L.B.); Tel.: +420-296442360 (P.B.); +420-296443743 (L.B.)
| |
Collapse
|
12
|
Corona A, Blobe GC. The role of the extracellular matrix protein TGFBI in cancer. Cell Signal 2021; 84:110028. [PMID: 33940163 DOI: 10.1016/j.cellsig.2021.110028] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 04/23/2021] [Accepted: 04/24/2021] [Indexed: 02/07/2023]
Abstract
The secreted extracellular protein, transforming growth factor beta induced (TGFBI or βIGH3), has roles in regulating numerous biological functions, including cell adhesion and bone formation, both during embryonic development and during the pathogenesis of human disease. TGFBI has been most studied in the context of hereditary corneal dystrophies, where mutations in TGFBI result in accumulation of TGFBI in the cornea. In cancer, early studies focused on TGFBI as a tumor suppressor, in part by promoting chemotherapy sensitivity. However, in established tumors, TGFBI largely has a role in promoting tumor progression, with elevated levels correlating to poorer clinical outcomes. As an important regulator of cancer progression, TGFBI expression and function is tightly regulated by numerous mechanisms including epigenetic silencing through promoter methylation and microRNAs. Mechanisms to target TGFBI have potential clinical utility in treating advanced cancers, while assessing TGFBI levels could be a biomarker for chemotherapy resistance and tumor progression.
Collapse
Affiliation(s)
- Armando Corona
- Department of Pharmacology and Cancer Biology, Duke University Medical center, USA
| | - Gerard C Blobe
- Department of Pharmacology and Cancer Biology, Duke University Medical center, USA; Department of Medicine, Duke University Medical Center, USA.
| |
Collapse
|
13
|
Wang Y, Wei Q, Chen Y, Long S, Yao Y, Fu K. Identification of Hub Genes Associated With Sensitivity of 5-Fluorouracil Based Chemotherapy for Colorectal Cancer by Integrated Bioinformatics Analysis. Front Oncol 2021; 11:604315. [PMID: 33912443 PMCID: PMC8071956 DOI: 10.3389/fonc.2021.604315] [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: 09/09/2020] [Accepted: 03/16/2021] [Indexed: 12/24/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most common malignant tumors. 5-fluorouracil (5-FU) has been used for the standard first-line treatment for CRC patients for several decades. Although 5-FU based chemotherapy has increased overall survival (OS) of CRC patients, the resistance of CRC to 5-FU based chemotherapy is the principal cause for treatment failure. Thus, identifying novel biomarkers to predict response to 5-FU based chemotherapy is urgently needed. In the present study, the gene expression profile of GSE3964 from the Gene Expression Omnibus database was used to explore the potential genes related to intrinsic resistance to 5-FU. A gene module containing 81 genes was found to have the highest correlation with chemotherapy response using Weighted Gene Co-expression Network Analysis (WGCNA). Then a protein-protein interaction (PPI) network was constructed and ten hub genes (TGFBI, NID, LEPREL2, COL11A1, CYR61, PCOLCE, IGFBP7, COL4A2, CSPG2, and VTN) were identified using the CytoHubba plugin of Cytoscape. Seven of these hub genes showed significant differences in expression between chemotherapy-sensitive and chemotherapy-resistant samples. The prognostic value of these seven genes was evaluated using TCGA COAD (Colorectal Adenocarcinoma) data. The results showed that TGFBI was highly expressed in chemotherapy-sensitive patients, and patients with high TGFBI expression have better survival.
Collapse
Affiliation(s)
- Ya Wang
- Institute of Molecular Precision Medicine and Hunan Key Laboratory of Molecular Precision Medicine, Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Qunhui Wei
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - Yuqiao Chen
- Institute of Molecular Precision Medicine and Hunan Key Laboratory of Molecular Precision Medicine, Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Shichao Long
- Institute of Molecular Precision Medicine and Hunan Key Laboratory of Molecular Precision Medicine, Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Yuanbing Yao
- Institute of Molecular Precision Medicine and Hunan Key Laboratory of Molecular Precision Medicine, Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Kai Fu
- Institute of Molecular Precision Medicine and Hunan Key Laboratory of Molecular Precision Medicine, Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China.,Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, China.,Hunan Key Laboratory of Animal Models for Human Diseases, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Changsha, China.,Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
14
|
Jiang R, Chen X, Ge S, Wang Q, Liu Y, Chen H, Xu J, Wu J. MiR-21-5p Induces Pyroptosis in Colorectal Cancer via TGFBI. Front Oncol 2021; 10:610545. [PMID: 33614494 PMCID: PMC7892456 DOI: 10.3389/fonc.2020.610545] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 12/18/2020] [Indexed: 12/24/2022] Open
Abstract
Pyroptosis is a distinct form of programmed cell death in eukaryotic cells that has garnered increasing attention in cancer-related research. Moreover, although miR-21 has been reported as abnormally expressed in colorectal cancer, due to a lack of in-depth research on the transcriptional regulation mechanisms of miR-21, its clinical usage remains limited. Our study is the first, to our knowledge, to compare the clinical manifestations and laboratory phenotypes associated with miR-21-3p and miR-21-5p. Morphologically, the transfection of miR-21-3p or miR-21-5p inhibitors, as well as miR-21-5p mimics into HCT-116 and HT-29 cell lines, induced cell death. Surprisingly, overexpression of miR-21-5p induced cell death more strongly than its knockdown. Mechanistic studies of miR-21-5p overexpression revealed that various inflammatory factors including IL-1β and IL-18 were released, while pyroptosis-associated mRNAs were upregulated and proteins were activated. Moreover, miR-21-5p was found to act as a downstream factor to significantly and directly regulate transforming growth factor beta-induced (TGFB1). Specifically, miR-21-5p overexpression caused downregulation of TGFBI, which may have led to pyroptosis. Collectively, we revealed that miR-21-5p induces pyroptosis in colorectal cancer via TGFBI regulation, thereby providing important mechanistic insights into its antitumor effects and expanding its potential for clinical applications.
Collapse
Affiliation(s)
- Rilei Jiang
- School of Basic Medicine Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaolei Chen
- School of Basic Medicine Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shaohua Ge
- Department of Oncology, Kunshan Hospital of Traditional Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Kunshan, China
| | - Qin Wang
- First Clinical Medical College, Nanjing Medical University, Nanjing, China
| | - Yichang Liu
- Department of Oncology, Kunshan Hospital of Traditional Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Kunshan, China
| | - Haijun Chen
- Department of Oncology, Kunshan Hospital of Traditional Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Kunshan, China
| | - Jiatuo Xu
- School of Basic Medicine Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jiang Wu
- Department of Oncology, Kunshan Hospital of Traditional Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Kunshan, China
| |
Collapse
|
15
|
Sima M, Vrbova K, Zavodna T, Honkova K, Chvojkova I, Ambroz A, Klema J, Rossnerova A, Polakova K, Malina T, Belza J, Topinka J, Rossner P. The Differential Effect of Carbon Dots on Gene Expression and DNA Methylation of Human Embryonic Lung Fibroblasts as a Function of Surface Charge and Dose. Int J Mol Sci 2020; 21:E4763. [PMID: 32635498 PMCID: PMC7369946 DOI: 10.3390/ijms21134763] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 06/18/2020] [Accepted: 07/02/2020] [Indexed: 01/01/2023] Open
Abstract
This study presents a toxicological evaluation of two types of carbon dots (CD), similar in size (<10 nm) but differing in surface charge. Whole-genome mRNA and miRNA expression (RNAseq), as well as gene-specific DNA methylation changes, were analyzed in human embryonic lung fibroblasts (HEL 12469) after 4 h and 24 h exposure to concentrations of 10 and 50 µg/mL (for positive charged CD; pCD) or 10 and 100 µg/mL (for negative charged CD, nCD). The results showed a distinct response for the tested nanomaterials (NMs). The exposure to pCD induced the expression of a substantially lower number of mRNAs than those to nCD, with few commonly differentially expressed genes between the two CDs. For both CDs, the number of deregulated mRNAs increased with the dose and exposure time. The pathway analysis revealed a deregulation of processes associated with immune response, tumorigenesis and cell cycle regulation, after exposure to pCD. For nCD treatment, pathways relating to cell proliferation, apoptosis, oxidative stress, gene expression, and cycle regulation were detected. The expression of miRNAs followed a similar pattern: more pronounced changes after nCD exposure and few commonly differentially expressed miRNAs between the two CDs. For both CDs the pathway analysis based on miRNA-mRNA interactions, showed a deregulation of cancer-related pathways, immune processes and processes involved in extracellular matrix interactions. DNA methylation was not affected by exposure to any of the two CDs. In summary, although the tested CDs induced distinct responses on the level of mRNA and miRNA expression, pathway analyses revealed a potential common biological impact of both NMs independent of their surface charge.
Collapse
Affiliation(s)
- Michal Sima
- Department of Nanotoxicology and Molecular Epidemiology, Institute of Experimental Medicine of the Czech Academy of Sciences, 14220 Prague, Czech Republic; (M.S.); (K.V.); (A.A.)
| | - Kristyna Vrbova
- Department of Nanotoxicology and Molecular Epidemiology, Institute of Experimental Medicine of the Czech Academy of Sciences, 14220 Prague, Czech Republic; (M.S.); (K.V.); (A.A.)
| | - Tana Zavodna
- Department of Genetic Toxicology and Epigenetics, Institute of Experimental Medicine of the Czech Academy of Sciences, 14220 Prague, Czech Republic; (T.Z.); (K.H.); (I.C.); (A.R.); (J.T.)
| | - Katerina Honkova
- Department of Genetic Toxicology and Epigenetics, Institute of Experimental Medicine of the Czech Academy of Sciences, 14220 Prague, Czech Republic; (T.Z.); (K.H.); (I.C.); (A.R.); (J.T.)
| | - Irena Chvojkova
- Department of Genetic Toxicology and Epigenetics, Institute of Experimental Medicine of the Czech Academy of Sciences, 14220 Prague, Czech Republic; (T.Z.); (K.H.); (I.C.); (A.R.); (J.T.)
| | - Antonin Ambroz
- Department of Nanotoxicology and Molecular Epidemiology, Institute of Experimental Medicine of the Czech Academy of Sciences, 14220 Prague, Czech Republic; (M.S.); (K.V.); (A.A.)
| | - Jiri Klema
- Department of Computer Science, Czech Technical University in Prague, 12135 Prague, Czech Republic;
| | - Andrea Rossnerova
- Department of Genetic Toxicology and Epigenetics, Institute of Experimental Medicine of the Czech Academy of Sciences, 14220 Prague, Czech Republic; (T.Z.); (K.H.); (I.C.); (A.R.); (J.T.)
| | - Katerina Polakova
- Regional Centre of Advanced Technologies and Materials, Faculty of Science, Palacký University Olomouc, 77146 Olomouc, Czech Republic; (K.P.); (T.M.); (J.B.)
| | - Tomas Malina
- Regional Centre of Advanced Technologies and Materials, Faculty of Science, Palacký University Olomouc, 77146 Olomouc, Czech Republic; (K.P.); (T.M.); (J.B.)
- Department of Physical Chemistry, Faculty of Science, Palacký University Olomouc, 77146 Olomouc, Czech Republic
| | - Jan Belza
- Regional Centre of Advanced Technologies and Materials, Faculty of Science, Palacký University Olomouc, 77146 Olomouc, Czech Republic; (K.P.); (T.M.); (J.B.)
- Department of Physical Chemistry, Faculty of Science, Palacký University Olomouc, 77146 Olomouc, Czech Republic
| | - Jan Topinka
- Department of Genetic Toxicology and Epigenetics, Institute of Experimental Medicine of the Czech Academy of Sciences, 14220 Prague, Czech Republic; (T.Z.); (K.H.); (I.C.); (A.R.); (J.T.)
| | - Pavel Rossner
- Department of Nanotoxicology and Molecular Epidemiology, Institute of Experimental Medicine of the Czech Academy of Sciences, 14220 Prague, Czech Republic; (M.S.); (K.V.); (A.A.)
| |
Collapse
|
16
|
CADM1 inhibits ovarian cancer cell proliferation and migration by potentially regulating the PI3K/Akt/mTOR pathway. Biomed Pharmacother 2019; 123:109717. [PMID: 31865146 DOI: 10.1016/j.biopha.2019.109717] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 11/27/2019] [Accepted: 12/04/2019] [Indexed: 12/19/2022] Open
Abstract
Previous studies have shown that cell adhesion molecule 1 (CADM1), an immunoglobulin superfamily member, is frequently inactivated but functions as a tumor suppressor in many solid tumors. However, the characterization of CADM1 expression in ovarian cancer cells and the mechanisms of its tumor suppressor function are not fully understood. We generated ovarian cancer cell lines in which CADM1 was stably upregulated or downregulated. CADM1 expression was significantly decreased in ovarian cancer tissue and cells lines. Functionally, knockdown of CADM1 promoted the growth, migration and invasion of ovarian cancer cells. Conversely, further experimental evidence indicated that overexpression of CADM1 inhibited the migration and invasion of ovarian cancer cells potentially through inhibition of the PI3K/Akt/mTOR signaling pathway by regulating upstream regulators (LXR/RXR, IGF1, IFI44L and C4BPA) and downstream effectors (APP, EDN1, TGFBI and Rap1A). In conclusion, CADM1 inhibits ovarian cancer cell proliferation and migration by potentially regulating the PI3K/Akt/mTOR signaling pathway. CADM1 could be a potential therapeutic target for ovarian cancer.
Collapse
|
17
|
Zhang L, Su H, Wang H, Li Q, Li X, Zhou C, Xu J, Chai Y, Liang X, Xiong L, Zhang C. Tumor Chemo-Radiotherapy with Rod-Shaped and Spherical Gold Nano Probes: Shape and Active Targeting Both Matter. Am J Cancer Res 2019; 9:1893-1908. [PMID: 31037146 PMCID: PMC6485290 DOI: 10.7150/thno.30523] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 01/20/2019] [Indexed: 12/13/2022] Open
Abstract
The morphologies of gold nanoparticles (NPs) affect their tumor accumulation through enhanced permeability and retention effect. However, detailed information and mechanisms of NPs' characteristics affecting tumor accumulation are limited. The aim of this study is to evaluate the effects of shape and active targeting ligands of theranostic NPs on tumor accumulation and therapeutic efficacy, and to elucidate the underlying mechanism. Methods:αvβ3 integrin-targeted, cisplatin-loaded and radioisotope iodine-125 labeled spherical and rod-shaped gold nano theranostic probes (RGD-125IPt-AuNPs and RGD-125IPt-AuNRs) with similar sizes were fabricated and characterized. The in vivo distribution and chemo-radio therapeutic efficacy against tumors of these newly developed probes were subsequently evaluated. Moreover, a physiologically based pharmacokinetic (PBPK) model was developed to characterize the in vivo kinetics of these probes at the sub-organ level, and to reveal the mechanism of NPs' shape and active targeting ligands effects on tumor accumulation. Result: Cisplatin and iodine-125 were loaded sequentially onto the NPs through a thin polydopamine coating layer on the NPs. Both RGD-125IPt-AuNPs and RGD-125IPt-AuNRs exhibited high specificity for αvβ3 in vitro, with the rod-shaped probe being more efficient. The PBPK model revealed that rod-shaped gold NPs diffused more rapidly in tumor interstitial than the spherical ones. Tumor accumulations of non-targeted and rod-shaped RAD-125IPt-AuNRs was higher in short term (1 h post injection), but not pronounced and similar to that of non-targeted spherical RAD-125IPt-AuNPs in 24 h after intravenous injection, revealing that the NPs' shape did not have a significant impact on tumor accumulations through enhanced permeability and retention (EPR) effect in long-term. While for actively targeted NPs, in addition to a higher distribution coefficient, RGD-125IPt-AuNRs also had a much higher tumor maximum uptake rate constant than RGD-125IPt-AuNPs, indicating both the shape and active targeting ligands affected the tumor uptake of rod-shaped NPs. As a result, RGD-125IPt-AuNRs had a more effective inhibition of tumor growth than RGD-125IPt-AuNPs by chemo-radiationtherapy. Conclusion: Our study suggests that both the shape and active targeting ligands of gold NPs play important roles on tumor accumulation and chemo-radio therapeutic effect.
Collapse
|
18
|
Seok Y, Lee WK, Park JY, Kim DS. TGFBI Promoter Methylation is Associated with Poor Prognosis in Lung Adenocarcinoma Patients. Mol Cells 2019; 42:161-165. [PMID: 30726660 PMCID: PMC6399005 DOI: 10.14348/molcells.2018.0322] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 11/01/2018] [Accepted: 12/10/2018] [Indexed: 11/27/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) is the leading cause of cancer-related deaths worldwide and has high rates of metastasis. Transforming growth factor beta-inducible protein (TGFBI) is an extracellular matrix component involved in tumour growth and metastasis. However, the exact role of TGFBI in NSCLC remains controversial. Gene silencing via DNA methylation of the promoter region is common in lung tumorigenesis and could thus be used for the development of molecular biomarkers. We analysed the methylation status of the TGFBI promoter in 138 NSCLC specimens via methylation-specific PCR and evaluated the correlation between TGFBI methylation and patient survival. TGFBI promoter methylation was detected in 25 (18.1%) of the tumours and was demonstrated to be associated with gene silencing. We observed no statistical correlation between TGFBI methylation and clinicopathological characteristics. Univariate and multivariate analyses showed that TGFBI methylation is significantly associated with poor survival outcomes in adenocarcinoma cases (adjusted hazard ratio = 2.88, 95% confidence interval = 1.19-6.99, P = 0.019), but not in squamous cell cases. Our findings suggest that methylation in the TGFBI promoter may be associated with pathogenesis of NSCLC and can be used as a predictive marker for lung adenocarcinoma prognosis. Further large-scale studies are needed to confirm these findings.
Collapse
Affiliation(s)
- Yangki Seok
- Department of Thoracic Surgery, School of Medicine, Kyungpook National University, Daegu 702-422,
Korea
| | - Won Kee Lee
- Department of Preventive Medicine, School of Medicine, Kyungpook National University, Daegu 702-422,
Korea
| | - Jae Yong Park
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu 702-422,
Korea
| | - Dong Sun Kim
- Department of Anatomy, School of Medicine, Kyungpook National University, Daegu 702-422,
Korea
| |
Collapse
|
19
|
Yan L, Ma J, Wang Y, Zan J, Wang Z, Zhu Y, Zhu Y, Ling L, Cao L, Liu X, Li S, Xu L, Qi Z, Nie L, Zhang Y. miR-21-5p induces cell proliferation by targeting TGFBI in non-small cell lung cancer cells. Exp Ther Med 2018; 16:4655-4663. [PMID: 30542417 PMCID: PMC6257667 DOI: 10.3892/etm.2018.6752] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 08/24/2018] [Indexed: 12/17/2022] Open
Abstract
The mortality rate of non-small cell lung cancer (NSCLC) remains high worldwide. miR-21-5p plays an important part in many cancer types, including NSCLC. However, the effect of miR-21-5p in NSCLC tumorigenesis remains poorly understood. The present study investigated whether miR-21-5p promoted NSCLC cell proliferation in vitro. In order to study the molecular mechanism by which miR-21-5p contributes to NSCLC progression, three bioinformatics algorithms were used to predict the genes which miR-21-5p targeted. TGFBI was identfieid as a putative direct target in NSCLC cells via the luciferase reporter assay. Furthermore, miR-21-5p downregulated TGFBI protein expression by a post-transcriptional mechanism via western blotting and a reverse transcription-quantitative polymerase chain reaction analysis. Finally, TGFBI exhibited opposing effects to those of miR-21-5p on NSCLC cells, suggesting that miR-21-5p may promote cell proliferation by negative regulation of TGFBI. These results suggest miR-21-5p promote the proliferation of NSCLC cells via inhibiting TGFBI expression.
Collapse
Affiliation(s)
- Liang Yan
- Department of Biopharmaceuticals, College of Life Sciences, Anhui Normal University, Wuhu, Anhui 241002, P.R. China.,Department of Biochemistry, Provincial Key Laboratory of Biological Macro-molecules Research, Wannan Medical College, Wuhu, Anhui 241002, P.R. China
| | - Jinzhu Ma
- Department of Biochemistry, Provincial Key Laboratory of Biological Macro-molecules Research, Wannan Medical College, Wuhu, Anhui 241002, P.R. China
| | - Yi Wang
- Department of Clinical Teaching, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, Jiangsu 210023, P.R. China
| | - Jiawei Zan
- Department of Biopharmaceuticals, College of Life Sciences, Anhui Normal University, Wuhu, Anhui 241002, P.R. China
| | - Zhen Wang
- Department of Chemistry, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui 241002, P.R. China
| | - Yu Zhu
- Department of Biochemistry, Provincial Key Laboratory of Biological Macro-molecules Research, Wannan Medical College, Wuhu, Anhui 241002, P.R. China
| | - Yiping Zhu
- Department of Chemistry, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui 241002, P.R. China
| | - Liefeng Ling
- Department of Biopharmaceuticals, College of Life Sciences, Anhui Normal University, Wuhu, Anhui 241002, P.R. China
| | - Long Cao
- Department of Biochemistry, Provincial Key Laboratory of Biological Macro-molecules Research, Wannan Medical College, Wuhu, Anhui 241002, P.R. China
| | - Xin Liu
- Department of Biochemistry, Provincial Key Laboratory of Biological Macro-molecules Research, Wannan Medical College, Wuhu, Anhui 241002, P.R. China
| | - Shu Li
- Department of Biochemistry, Provincial Key Laboratory of Biological Macro-molecules Research, Wannan Medical College, Wuhu, Anhui 241002, P.R. China
| | - Lei Xu
- Department of Biochemistry, Provincial Key Laboratory of Biological Macro-molecules Research, Wannan Medical College, Wuhu, Anhui 241002, P.R. China
| | - Zhilin Qi
- Department of Biochemistry, Provincial Key Laboratory of Biological Macro-molecules Research, Wannan Medical College, Wuhu, Anhui 241002, P.R. China
| | - Liuwang Nie
- Department of Biopharmaceuticals, College of Life Sciences, Anhui Normal University, Wuhu, Anhui 241002, P.R. China
| | - Yao Zhang
- Department of Biochemistry, Provincial Key Laboratory of Biological Macro-molecules Research, Wannan Medical College, Wuhu, Anhui 241002, P.R. China
| |
Collapse
|
20
|
Bissey PA, Law JH, Bruce JP, Shi W, Renoult A, Chua MLK, Yip KW, Liu FF. Dysregulation of the MiR-449b target TGFBI alters the TGFβ pathway to induce cisplatin resistance in nasopharyngeal carcinoma. Oncogenesis 2018; 7:40. [PMID: 29795279 PMCID: PMC5966388 DOI: 10.1038/s41389-018-0050-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 02/26/2018] [Accepted: 04/11/2018] [Indexed: 01/07/2023] Open
Abstract
Despite the improvement in locoregional control of nasopharyngeal carcinoma (NPC), distant metastasis (DM), and chemoresistance persist as major causes of mortality. This study identified a novel role for miR-449b, an overexpressed gene in a validated four-miRNA signature for NPC DM, leading to chemoresistance via the direct targeting of transforming growth factor beta-induced (TGFBI). In vitro shRNA-mediated downregulation of TGFBI induced phosphorylation of PTEN and AKT, increasing cisplatin resistance. Conversely, the overexpression of TGFBI sensitized the NPC cells to cisplatin. In NPC patients treated with concurrent chemoradiotherapy (CRT), the overall survival (OS) was significantly inversely correlated with miR-449b, and directly correlated with both TGFBI mRNA and protein expression, as assessed by RNA sequencing and immunohistochemistry (IHC). Mechanistically, co-immunoprecipitation demonstrated that TGFBI competes with pro-TGFβ1 for integrin receptor binding. Decreased TGFBI led to increased pro-TGFβ1 activation and TGFβ1 canonical/noncanonical pathway-induced cisplatin resistance. Thus, overexpression of miR-449b decreases TGFBI, thereby altering the balance between TGFBI and pro-TGFβ1, revealing a novel mechanism of chemoresistance in NPC.
Collapse
Affiliation(s)
| | - Jacqueline H Law
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Jeff P Bruce
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Wei Shi
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Aline Renoult
- LabEx DEVweCAN, Université de Lyon, F-69000, Lyon, France
| | - Melvin L K Chua
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada.,Department of Radiation Oncology, Princess Margaret Cancer Centre and University of Toronto, Toronto, ON, Canada.,Division of Radiation Oncology, National Cancer Centre, Singapore, Singapore.,Duke-NUS Graduate School, Singapore, Singapore
| | - Kenneth W Yip
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Fei-Fei Liu
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada. .,Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada. .,Department of Radiation Oncology, Princess Margaret Cancer Centre and University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
21
|
Shang D, Song B, Liu Y. Epirubicin suppresses proliferative and metastatic potential by downregulating transforming growth factor-β-induced expression in urothelial carcinoma. Cancer Sci 2018; 109:980-987. [PMID: 28940965 PMCID: PMC5891197 DOI: 10.1111/cas.13403] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 09/13/2017] [Accepted: 09/18/2017] [Indexed: 12/29/2022] Open
Abstract
Transforming growth factor‐β‐induced (TGFΒI) is considered to be a vital gene in several carcinomas. In this study we determined the effect of TGFBI on the proliferative and metastatic potential of human urothelial carcinoma (UC) cells as well as its mRNA and protein expression, which were detected by RT‐PCR and western blot, respectively. UC cell proliferation was analyzed by WST‐1 assay and Hoechst 33258 staining. The effect of TGFBI on UC cell metastasis was analyzed using adhesion, migration and invasion assays. We found that TGFBI increased the proliferation of UC cells. Moreover, TGFBI enhanced the adhesion, migration and invasion of UC cells by upregulating MMP‐2, MMP‐9 and calpain‐2 expression. We evaluated the effect of Epirubicin (EPI) on the regulation of TGFBI expression and found that TGFBI acts as a downstream target of EPI and is suppressed by EPI in UC cells. EPI is more effective in inhibiting the proliferation and metastasis of UC cells with high TGFBI expression. This study demonstrates that TGFBI might lead to tumorigenesis and progression of UC and those cells with high TGFBI expression may be vulnerable to relapse. EPI could prove to be a therapeutic option in patients with high TGFBI expressing UC cells.
Collapse
Affiliation(s)
- Donghao Shang
- Department of Urology, Friendship Hospital, Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Capital Medical University, Beijing, China
| | - Bo Song
- Department of Urology, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Yuting Liu
- Department of Pathology, Capital Medical University, Beijing, China
| |
Collapse
|
22
|
Pan YB, Zhang CH, Wang SQ, Ai PH, Chen K, Zhu L, Sun ZL, Feng DF. Transforming growth factor beta induced (TGFBI) is a potential signature gene for mesenchymal subtype high-grade glioma. J Neurooncol 2018; 137:395-407. [PMID: 29294230 DOI: 10.1007/s11060-017-2729-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 12/24/2017] [Indexed: 12/19/2022]
Abstract
Previous study revealed that higher expression of transforming growth factor beta induced (TGFBI) is correlated to poorer cancer-specific survival and higher proportion of tumor necrosis and Fuhrman grades III and IV in clear cell renal cell carcinomas. However, the relationships between TGFBI expression and malignant phenotypes of gliomas remain unclear. We downloaded and analyzed data from seven GEO datasets (GSE68848, GSE4290, GSE13041, GSE4271, GSE83300, GSE34824 and GSE84010), the TCGA database and the REMBRANDT database to investigate whether TGFBI could be a biomarker of glioma. From microarray data (GSE68848, GSE4290) and RNA-seq data (TCGA), TGFBI expression levels were observed to correlate positively with pathological grade, and TGFBI expression levels were significantly higher in gliomas than in normal brain tissues. Furthermore, in GSE13041, GSE4271 and the TCGA cohort, TGFBI expression in the mesenchymal (Mes) subtype high-grade glioma (HGG) was significantly higher than that in the proneural subtype. Kaplan-Meier survival analysis of GBM patients in the GSE83300 dataset, REMBRANDT and TCGA cohort revealed that patients in the top 50% TGFBI expression group survived for markedly shorter periods than those in the bottom 50%. Analysis of grade III gliomas showed that the median survival time was significantly shorter in the TGFBI high expression group than in the TGFBI low expression group. In addition, we found that TGFBI expression levels might relate to several classical molecular characterizations of glioma, such as, IDH mutation, TP53 mutation, EGFR amplification, etc. These results suggest that TGFBI expression positively correlates with glioma pathological grades and that TGFBI is a potential signature gene for Mes subtype HGG and a potential prognostic molecule.
Collapse
Affiliation(s)
- Yuan-Bo Pan
- Department of Neurosurgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 201999, China
| | - Chi-Hao Zhang
- Department of General Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 201999, China
| | - Si-Qi Wang
- Department of Radiology, Affiliated Hospital of Medical School of Ningbo University, Ningbo, 315020, Zhejiang Province, China
| | - Peng-Hui Ai
- Department of Neurology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China
| | - Kui Chen
- Department of Neurosurgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 201999, China
| | - Liang Zhu
- Department of Neurosurgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 201999, China
| | - Zhao-Liang Sun
- Department of Neurosurgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 201999, China
| | - Dong-Fu Feng
- Department of Neurosurgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 201999, China.
| |
Collapse
|
23
|
Bigh3 silencing increases retinoblastoma tumor growth in the murine SV40-TAg-Rb model. Oncotarget 2017; 8:15490-15506. [PMID: 28099942 PMCID: PMC5362501 DOI: 10.18632/oncotarget.14659] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 12/24/2016] [Indexed: 11/25/2022] Open
Abstract
BIGH3, a secreted protein of the extracellular matrix interacts with collagen and integrins on the cell surface. BIGH3 can have opposing functions in cancer, acting either as tumor suppressor or promoter by enhancing tumor progression and angiogenesis. In the eye, BIGH3 is expressed in the cornea and the retinal pigment epithelium and could impact on the development of retinoblastoma, the most common paediatric intraocular neoplasm. Retinoblastoma initiation requires the inactivation of both alleles of the RB1 tumor suppressor gene in the developing retina and tumor progression involves additional genomic changes. To determine whether BIGH3 affects retinoblastoma development, we generated a retinoblastoma mouse model with disruption of the Bigh3 genomic locus. Bigh3 silencing in these mice resulted in enhanced tumor development in the retina. A decrease in apoptosis is involved in the initial events of tumorigenesis, followed by an increased activity of the pro-survival ERK pathway as well as an upregulation of cyclin-dependent kinases (CDKs). Taken together, these data suggest that BIGH3 acts as a tumor suppressor in the retina.
Collapse
|
24
|
Peláez R, Morales X, Salvo E, Garasa S, Ortiz de Solórzano C, Martínez A, Larrayoz IM, Rouzaut A. β3 integrin expression is required for invadopodia-mediated ECM degradation in lung carcinoma cells. PLoS One 2017; 12:e0181579. [PMID: 28767724 PMCID: PMC5540285 DOI: 10.1371/journal.pone.0181579] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 07/03/2017] [Indexed: 01/17/2023] Open
Abstract
Cancer related deaths are primarily due to tumor metastasis. To facilitate their dissemination to distant sites, cancer cells develop invadopodia, actin-rich protrusions capable of degrading the surrounding extracellular matrix (ECM). We aimed to determine whether β3 integrin participates in invadopodia formed by lung carcinoma cells, based on our previous findings of specific TGF-β induction of β3 integrin dependent metastasis in animal models of lung carcinoma. In this study, we demonstrate that lung carcinoma cells form invadopodia in response to TGF-β exposure. Invadopodia formation and degradation activity is dependent on β3 integrin expression since β3 integrin deficient cells are not able to degrade gelatin-coated surfaces. Even more, transient over-expression of SRC did not restore invadopodia formation in β3 integrin deficient cells. Finally, we observed that blockade of PLC-dependent signaling leads to more intense labeling for β3 integrin in invadopodia. Our results suggest that β3 integrin function, and location, in lung cancer cells are essential for invadopodia formation, and this integrin regulates the activation of different signal pathways necessary for the invasive structure. β3 integrin has been associated with poor prognosis and increased metastasis in several carcinoma types, including lung cancer. Our findings provide new evidence to support the use of targeted therapies against this integrin to combat the onset of metastases.
Collapse
Affiliation(s)
- Rafael Peláez
- Department of Oncology, Center for Applied Medical Research CIMA, Pamplona, Spain.,Biomarkers and Molecular Signaling Group, Neurodegenerative Diseases Area, Center for Biomedical Research of La Rioja, CIBIR, Logroño, Spain
| | - Xabier Morales
- Department of Oncology, Center for Applied Medical Research CIMA, Pamplona, Spain.,Laboratory of Preclinical Models and Analytical Tools, Division of Solid Tumors and Biomarkers, Center for Applied Medical Research and CIBERONC, Pamplona, Navarra, Spain
| | - Elizabeth Salvo
- Department of Oncology, Center for Applied Medical Research CIMA, Pamplona, Spain
| | - Saray Garasa
- Department of Oncology, Center for Applied Medical Research CIMA, Pamplona, Spain.,Department of Immunology and Immunotherapy, CIMA, Pamplona, Navarra, Spain
| | - Carlos Ortiz de Solórzano
- Laboratory of Preclinical Models and Analytical Tools, Division of Solid Tumors and Biomarkers, Center for Applied Medical Research and CIBERONC, Pamplona, Navarra, Spain
| | - Alfredo Martínez
- Oncology Area, Center for Biomedical Research of La Rioja, CIBIR, Logroño, Spain
| | - Ignacio M Larrayoz
- Biomarkers and Molecular Signaling Group, Neurodegenerative Diseases Area, Center for Biomedical Research of La Rioja, CIBIR, Logroño, Spain
| | - Ana Rouzaut
- Department of Oncology, Center for Applied Medical Research CIMA, Pamplona, Spain.,Department of Immunology and Immunotherapy, CIMA, Pamplona, Navarra, Spain.,Department of Biochemistry and Genetics, University of Navarra, Pamplona, Spain
| |
Collapse
|
25
|
Yokobori T, Nishiyama M. TGF-β Signaling in Gastrointestinal Cancers: Progress in Basic and Clinical Research. J Clin Med 2017; 6:jcm6010011. [PMID: 28106769 PMCID: PMC5294964 DOI: 10.3390/jcm6010011] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 12/31/2016] [Accepted: 01/16/2017] [Indexed: 12/18/2022] Open
Abstract
Transforming growth factor (TGF)-β superfamily proteins have many important biological functions, including regulation of tissue differentiation, cell proliferation, and migration in both normal and cancer cells. Many studies have reported that TGF-β signaling is associated with disease progression and therapeutic resistance in several cancers. Similarly, TGF-β-induced protein (TGFBI)—a downstream component of the TGF-β signaling pathway—has been shown to promote and/or inhibit cancer. Here, we review the state of basic and clinical research on the roles of TGF-β and TGFBI in gastrointestinal cancers.
Collapse
Affiliation(s)
- Takehiko Yokobori
- Research Program for Omics-based Medical Science, Division of Integrated Oncology Research, Gunma University Initiative for Advanced Research, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan.
| | - Masahiko Nishiyama
- Research Program for Omics-based Medical Science, Division of Integrated Oncology Research, Gunma University Initiative for Advanced Research, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan.
- Department of Molecular Pharmacology and Oncology, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan.
| |
Collapse
|
26
|
Liu Z, Cai J, Su H, Yang J, Sun W, Ma Y, Liu S, Zhang C. Feasibility of USPIOs for T1-weighted MR molecular imaging of tumor receptors. RSC Adv 2017. [DOI: 10.1039/c7ra04903j] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
USPIOs having a superior T1 contrast effect could only be used for T2-weighted, but not for T1-weighted MR tumor receptor imaging.
Collapse
Affiliation(s)
- Zhetao Liu
- State Key Laboratory of Oncogenes and Related Genes
- Shanghai Cancer Institute
- School of Biomedical Engineering
- Shanghai Jiao Tong University
- Shanghai 200030
| | - Jiali Cai
- Changzheng Hospital
- Secondary Military Medical University
- Shanghai 200003
- China
| | - Huilan Su
- State Key Laboratory of Metal Matrix Composites
- Shanghai Jiao Tong University
- Shanghai 200240
- China
| | - Jingxing Yang
- State Key Laboratory of Oncogenes and Related Genes
- Shanghai Cancer Institute
- School of Biomedical Engineering
- Shanghai Jiao Tong University
- Shanghai 200030
| | - Wenshe Sun
- State Key Laboratory of Oncogenes and Related Genes
- Shanghai Cancer Institute
- School of Biomedical Engineering
- Shanghai Jiao Tong University
- Shanghai 200030
| | - Yongjie Ma
- State Key Laboratory of Oncogenes and Related Genes
- Shanghai Cancer Institute
- School of Biomedical Engineering
- Shanghai Jiao Tong University
- Shanghai 200030
| | - Shiyuan Liu
- Changzheng Hospital
- Secondary Military Medical University
- Shanghai 200003
- China
| | - Chunfu Zhang
- State Key Laboratory of Oncogenes and Related Genes
- Shanghai Cancer Institute
- School of Biomedical Engineering
- Shanghai Jiao Tong University
- Shanghai 200030
| |
Collapse
|
27
|
Kriegsmann M, Longuespée R, Wandernoth P, Mohanu C, Lisenko K, Weichert W, Warth A, Dienemann H, De Pauw E, Katzenberger T, Aust D, Baretton G, Kriegsmann J, Casadonte R. Typing of colon and lung adenocarcinoma by high throughput imaging mass spectrometry. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2016; 1865:858-864. [PMID: 27939606 DOI: 10.1016/j.bbapap.2016.11.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 11/21/2016] [Accepted: 11/23/2016] [Indexed: 10/20/2022]
Abstract
In advanced tumor stages, diagnosis is frequently made from metastatic tumor tissue. In some cases, the identification of the tumor of origin may be difficult by histology alone. In this setting, immunohistochemical and molecular biological methods are often required. In a subset of tumors definite diagnosis cannot be achieved. Thus, additional new diagnostic methods are required for precise tumor subtyping. Mass spectrometric methods are of special interest for the discrimination of different tumor types. We investigated whether it is possible to discern adenocarcinomas of colon and lung using high-throughput imaging mass spectrometry on formalin-fixed paraffin-embedded tissue microarrays. 101 primary adenocarcinoma of the colon and 91 primary adenocarcinoma of the lung were used to train a Linear Discriminant Analysis model. Results were validated on an independent set of 116 colonic and 75 lung adenocarcinomas. In the validation cohort 109 of 116 patients with colonic and 67 of 75 patients with lung adenocarcinomas were correctly classified. The ability to define proteomic profiles capable to discern different tumor types promises a valuable tool in cancer diagnostics and might complement current approaches. This article is part of a Special Issue entitled: MALDI Imaging, edited by Dr. Corinna Henkel and Prof. Peter Hoffmann.
Collapse
Affiliation(s)
- Mark Kriegsmann
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany.
| | | | | | | | - Katharina Lisenko
- Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany.
| | | | - Arne Warth
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany; Translational Lung Research Center Heidelberg, Member of the German Center for Lung Research, Germany.
| | - Hendrik Dienemann
- Department of Thoracic Surgery, Thoraxklinik at Heidelberg University, Heidelberg, Germany.
| | - Edwin De Pauw
- Mass Spectrometry Laboratory, Systems Biology and Chemical Biology, GIGA-Research, University of Liège, Belgium.
| | | | - Daniela Aust
- Institute of Pathology, University Hospital Carl Gustav Carus, Dresden, Germany.
| | - Gustavo Baretton
- Institute of Pathology, University Hospital Carl Gustav Carus, Dresden, Germany.
| | - Joerg Kriegsmann
- Proteopath GmbH, Trier, Germany; Center for Histology, Cytology and Molecular Diagnostics Trier, Trier, Germany.
| | | |
Collapse
|
28
|
Ricciardelli C, Lokman NA, Ween MP, Oehler MK. WOMEN IN CANCER THEMATIC REVIEW: Ovarian cancer-peritoneal cell interactions promote extracellular matrix processing. Endocr Relat Cancer 2016; 23:T155-T168. [PMID: 27578826 DOI: 10.1530/erc-16-0320] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 08/30/2016] [Indexed: 12/13/2022]
Abstract
Ovarian cancer has a distinct tendency for metastasising via shedding of cancerous cells into the peritoneal cavity and implanting onto the peritoneum that lines the pelvic organs. Once ovarian cancer cells adhere to the peritoneal cells, they migrate through the peritoneal layer and invade the local organs. Alterations in the extracellular environment are critical for tumour initiation, progression and intra-peritoneal dissemination. To increase our understanding of the molecular mechanisms involved in ovarian cancer metastasis and to identify novel therapeutic targets, we recently studied the interaction of ovarian cancer and peritoneal cells using a proteomic approach. We identified several extracellular matrix (ECM) proteins including, fibronectin, TGFBI, periostin, annexin A2 and PAI-1 that were processed as a result of the ovarian cancer-peritoneal cell interaction. This review focuses on the functional role of these proteins in ovarian cancer metastasis. Our findings together with published literature support the notion that ECM processing via the plasminogen-plasmin pathway promotes the colonisation and attachment of ovarian cancer cells to the peritoneum and actively contributes to the early steps of ovarian cancer metastasis.
Collapse
Affiliation(s)
- C Ricciardelli
- Discipline of Obstetrics and GynaecologyAdelaide Medical School, Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - N A Lokman
- Discipline of Obstetrics and GynaecologyAdelaide Medical School, Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - M P Ween
- Lung Research LaboratoryHanson Institute, Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, South Australia, Australia
| | - M K Oehler
- Discipline of Obstetrics and GynaecologyAdelaide Medical School, Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
- Department of Gynaecological OncologyRoyal Adelaide Hospital, Adelaide, South Australia, Australia
| |
Collapse
|
29
|
Hung MS, Chen IC, You L, Jablons DM, Li YC, Mao JH, Xu Z, Lung JH, Yang CT, Liu ST. Knockdown of cullin 4A inhibits growth and increases chemosensitivity in lung cancer cells. J Cell Mol Med 2016; 20:1295-306. [PMID: 26969027 PMCID: PMC4929302 DOI: 10.1111/jcmm.12811] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Accepted: 01/15/2016] [Indexed: 12/31/2022] Open
Abstract
Cullin 4A (Cul4A) has been observed to be overexpressed in various cancers. In this study, the role of Cul4A in the growth and chemosensitivity in lung cancer cells were studied. We showed that Cul4A is overexpressed in lung cancer cells and tissues. Knockdown of the Cul4A expression by shRNA in lung cancer cells resulted in decreased cellular proliferation and growth in lung cancer cells. Increased sensitivity to gemcitabine, a chemotherapy drug, was also noted in those Cul4A knockdown lung cancer cells. Moreover, increased expression of p21, transforming growth factor (TGF)-β inducible early gene-1 (TIEG1) and TGF beta-induced (TGFBI) was observed in lung cancer cells after Cul4A knockdown, which may be partially related to increased chemosensitivity to gemcitabine. G0/G1 cell cycle arrest was also noted after Cul4A knockdown. Notably, decreased tumour growth and increased chemosensitivity to gemcitabine were also noted after Cul4A knockdown in lung cancer xenograft nude mice models. In summary, our study showed that targeting Cul4A with RNAi or other techniques may provide a possible insight to the development of lung cancer therapy in the future.
Collapse
Affiliation(s)
- Ming-Szu Hung
- Division of Thoracic Oncology, Department of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Chiayi, Taiwan.,Department of Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Department of Respiratory Care, Chang Gung University of Science and Technology, Chiayi, Taiwan
| | - I-Chuan Chen
- Department of Emergency Medicine, Chang Gung Memorial Hospital, Chiayi, Taiwan.,Department of Nursing, Chang Gung University of Science and Technology, Chiayi, Taiwan
| | - Liang You
- Thoracic Oncology Laboratory, Department of Surgery, Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - David M Jablons
- Thoracic Oncology Laboratory, Department of Surgery, Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Ya-Chin Li
- Division of Thoracic Oncology, Department of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Jian-Hua Mao
- Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Zhidong Xu
- Thoracic Oncology Laboratory, Department of Surgery, Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Jr-Hau Lung
- Department of Medical Research and Development, Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Cheng-Ta Yang
- Department of Respiratory Care, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Department of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Shih-Tung Liu
- Department of Microbiology and Immunology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
30
|
Tumor suppressor genes and their underlying interactions in paclitaxel resistance in cancer therapy. Cancer Cell Int 2016; 16:13. [PMID: 26900348 PMCID: PMC4761208 DOI: 10.1186/s12935-016-0290-9] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 02/12/2016] [Indexed: 01/01/2023] Open
Abstract
Objectives Paclitaxel (PTX) is frequently used in the clinical treatment of solid tumors. But the PTX-resistance is a great obstacle in cancer treatment. Exploration of the mechanisms of drug resistance suggests that tumor suppressor genes (TSGs) play a key role in the response of chemotherapeutic drugs. TSGs, a set of genes that are often inactivated in cancers, can regulate various biological processes. In this study, an overview of the contribution of TSGs to PTX resistance and their underlying relationship in cancers are reported by using GeneMANIA, a web-based tool for gene/protein function prediction. Methods Using PubMed online database and Google web site, the terms “paclitaxel resistance” or “taxol resistance” or “drug resistance” or “chemotherapy resistance”, and “cancer” or “carcinoma”, and “tumor suppressor genes” or “TSGs” or “negative regulated protein” or “antioncogenes” were searched and analyzed. GeneMANIA data base was used to predict gene/protein interactions and functions. Results We identified 22 TSGs involved in PTX resistance, including BRCA1, TP53, PTEN, APC, CDKN1A, CDKN2A, HIN-1, RASSF1, YAP, ING4, PLK2, FBW7, BLU, LZTS1, REST, FADD, PDCD4, TGFBI, ING1, Bax, PinX1 and hEx. The TSGs were found to have direct and indirect relationships with each other, and thus they could contribute to PTX resistance as a group. The varied expression status and regulation function of the TSGs on cell cycle in different cancers might play an important role in PTX resistance. Conclusion A further understanding of the roles of tumor suppressor genes in drug resistance is an important step to overcome chemotherapy tolerance. Tumor suppressor gene therapy targets the altered genes and signaling pathways and can be a new strategy to reverse chemotherapy resistance.
Collapse
|
31
|
Antonângelo L, Tuma T, Fabro A, Acencio M, Terra R, Parra E, Vargas F, Takagaki T, Capelozzi V. Id-1, Id-2, and Id-3 co-expression correlates with prognosis in stage I and II lung adenocarcinoma patients treated with surgery and adjuvant chemotherapy. Exp Biol Med (Maywood) 2016; 241:1159-68. [PMID: 26869608 DOI: 10.1177/1535370216632623] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 01/22/2016] [Indexed: 12/25/2022] Open
Abstract
Inhibitors of DNA binding/inhibitors of differentiation (Id) protein family have been shown to be involved in carcinogenesis. However, the roles of Id during lung adenocarcinoma (ADC) progression remain unclear. Eighty-eight ADC samples were evaluated for Id-1,2,3 level and angiogenesis (CD 34 and VEGF microvessel density) by immunohistochemistry and morphometry. The impact of these markers was tested on follow-up until death or recurrence. A significant difference between tumor and normal tissue was found for Id-1,2,3 expression (P < 0.01). In addition, high levels of nuclear Id-1 were associated with higher angiogenesis in the tumor stroma (P < 0.01). Equally significant was the association between patients in T1-stage and low cytoplasmic Id-2, as well as patients in stage-IIb and low Id-3. High cytoplasm Id-3 expression was also directly associated to lymph nodes metastasis (P = 0.05). Patients at stages I to III, with low Id-1 and Id-3 cytoplasm histoscores showed significant long metastasis-free survival time than those with high Id-1 or Id-3 expression (P = 0.04). Furthermore, high MVD-CD34 and MVD-VEGF expression were associated with short recurrence-free survival compared to low MVD-CD34 and MVD-VEGF expressions (P = 0.04). Cox model analyses controlled for age, lymph node metastasis, and adjuvant treatments showed that nuclear Id-1, cytoplasmic Id-3, and MVD-CD34 were significantly associated with survival time. Median score for nuclear Id-1 and cytoplasmic Id-3 divided patients in two groups, being that those with increased Id-1 and Id-3 presented higher risk of death. Ids showed an independent prognostic value in patients with lung ADC, regardless of disease stage. Id-1 and Id-3 should be considered new target candidates in the development of personalized therapy in lung ADC.
Collapse
Affiliation(s)
- Leila Antonângelo
- Department of Pathology, University of Sao Paulo, Sao Paulo 01246903,Brazil
| | - Taila Tuma
- Department of Pathology, University of Sao Paulo, Sao Paulo 01246903,Brazil
| | - Alexandre Fabro
- Department of Pathology, University of Sao Paulo, Sao Paulo 01246903,Brazil
| | - Milena Acencio
- Pulmonary Division, Heart Institute Clinics Hospital, University of Sao Paulo, Sao Paulo 01246903, Brazil
| | - Ricardo Terra
- Pulmonary Division, Heart Institute Clinics Hospital, University of Sao Paulo, Sao Paulo 01246903, Brazil
| | - Edwin Parra
- Department of Pathology, University of Sao Paulo, Sao Paulo 01246903,Brazil
| | - Francisco Vargas
- Pulmonary Division, Heart Institute Clinics Hospital, University of Sao Paulo, Sao Paulo 01246903, Brazil
| | - Teresa Takagaki
- Pulmonary Division, Heart Institute Clinics Hospital, University of Sao Paulo, Sao Paulo 01246903, Brazil
| | - Vera Capelozzi
- Department of Pathology, University of Sao Paulo, Sao Paulo 01246903,Brazil
| |
Collapse
|
32
|
Qin X, Yan M, Zhang J, Wang X, Shen Z, Lv Z, Li Z, Wei W, Chen W. TGFβ3-mediated induction of Periostin facilitates head and neck cancer growth and is associated with metastasis. Sci Rep 2016; 6:20587. [PMID: 26857387 PMCID: PMC4746667 DOI: 10.1038/srep20587] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 01/07/2016] [Indexed: 12/18/2022] Open
Abstract
The matrix-specific protein periostin (POSTN) is up-regulated in human cancers and associated with cancer growth, metastasis and angiogenesis. Although the stroma of cancer tissues is the main source of POSTN, it is still unclear how POSTN plays a role to facilitate the interplay between cancer cells and cancer-associated fibroblasts (CAFs) in head and neck cancer (HNC), thereby promoting tumorigenesis via modifying the tumor microenvironment. Herein, we have performed studies to investigate POSTN and its role in HNC microenvironment. Our results indicated that POSTN was significantly up-regulated in HNCs, especially in the tissues with lymph node metastasis. Moreover, POSTN was highly enriched in the stroma of cancer tissues and produced mainly by CAFs. More importantly, we have pinpointed TGF-β3 as the major upstream molecular that triggers the induction of POSTN in CAFs. As such, during the interaction between fibroblasts and cancer cells, the increased stromal POSTN induced by TGF-β3 directly accelerated the growth, migration and invasion of cancer cells. Hence, our study has provided a novel modulative role for POSTN on HNC progression and further reveals POSTN as an effective biomarker to predict metastasis as well as a potential cancer therapeutic target.
Collapse
Affiliation(s)
- Xing Qin
- Department of Oral and Maxillofacial-Head &Neck Oncology and Faculty of Oral and Maxillofacial Surgery, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.,Shanghai Research Institute of Stomatology and Shanghai Key Laboratory of Stomatology, Shanghai, 200011, China
| | - Ming Yan
- Department of Oral and Maxillofacial-Head &Neck Oncology and Faculty of Oral and Maxillofacial Surgery, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.,Shanghai Research Institute of Stomatology and Shanghai Key Laboratory of Stomatology, Shanghai, 200011, China
| | - Jianjun Zhang
- Department of Oral and Maxillofacial-Head &Neck Oncology and Faculty of Oral and Maxillofacial Surgery, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.,Shanghai Research Institute of Stomatology and Shanghai Key Laboratory of Stomatology, Shanghai, 200011, China
| | - Xu Wang
- Department of Oral and Maxillofacial-Head &Neck Oncology and Faculty of Oral and Maxillofacial Surgery, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.,Shanghai Research Institute of Stomatology and Shanghai Key Laboratory of Stomatology, Shanghai, 200011, China
| | - Zongze Shen
- Department of Oral and Maxillofacial-Head &Neck Oncology and Faculty of Oral and Maxillofacial Surgery, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.,Shanghai Research Institute of Stomatology and Shanghai Key Laboratory of Stomatology, Shanghai, 200011, China
| | - Zhongjing Lv
- Department of Oral and Maxillofacial-Head &Neck Oncology and Faculty of Oral and Maxillofacial Surgery, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.,Shanghai Research Institute of Stomatology and Shanghai Key Laboratory of Stomatology, Shanghai, 200011, China
| | - Zhihui Li
- Department of Oral and Maxillofacial-Head &Neck Oncology and Faculty of Oral and Maxillofacial Surgery, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.,Shanghai Research Institute of Stomatology and Shanghai Key Laboratory of Stomatology, Shanghai, 200011, China
| | - Wenyi Wei
- Department of Oral and Maxillofacial-Head &Neck Oncology and Faculty of Oral and Maxillofacial Surgery, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.,Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, Massachusetts 02215, USA
| | - Wantao Chen
- Department of Oral and Maxillofacial-Head &Neck Oncology and Faculty of Oral and Maxillofacial Surgery, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.,Shanghai Research Institute of Stomatology and Shanghai Key Laboratory of Stomatology, Shanghai, 200011, China
| |
Collapse
|
33
|
Yang Y, Zhang L, Cai J, Li X, Cheng D, Su H, Zhang J, Liu S, Shi H, Zhang Y, Zhang C. Tumor Angiogenesis Targeted Radiosensitization Therapy Using Gold Nanoprobes Guided by MRI/SPECT Imaging. ACS APPLIED MATERIALS & INTERFACES 2016; 8:1718-1732. [PMID: 26731347 DOI: 10.1021/acsami.5b09274] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Gold nanoparticles (AuNPs) have recently garnered great interest as potential radiosensitizers in tumor therapy. However, major challenges facing their application in this regard are further enhancement of tumor accumulation of the particles in addition to enhanced permeability retention (EPR) effect and an understanding of the optimal particle size and time for applying radiotherapy after the particle administration. In this study, we fabricated novel cyclic c(RGDyC)-peptide-conjugated, Gd- and 99 mTc-labeled AuNPs (RGD@AuNPs-Gd99 mTc) probes with different sizes (29, 51, and 80 nm) and evaluated their potential as radiosensitization therapy both in vitro and in vivo. We found that these probes have a high specificity for αvβ3 integrin positive cells, which resulted in their high cellular uptake and thereby enhanced radiosensitization. Imaging in vivo with MRI and SPECT/CT directly showed that the RGD@AuNPs-Gd99 mTc probes specifically target tumors and exhibit greater accumulation within tumors than the RAD@AuNPs-Gd99 mTc probes. Interestingly, we found that the 80 nm RGD@AuNPs-Gd99 mTc probes exhibit the greatest effects in vitro; however, the 29 nm RGD@AuNPs-Gd99 mTc probes were clearly most efficient in vivo. As a result, radiotherapy of tumors with the 29 nm probe was the most potent. Our study demonstrates that RGD@AuNPs-Gd99 mTc probes are highly useful radiosensitizers capable of guiding and enhancing radiation therapy of tumors.
Collapse
Affiliation(s)
- Yi Yang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, School of Biomedical Engineering, Shanghai Jiao Tong University , Shanghai 200030, China
| | - Lu Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, School of Biomedical Engineering, Shanghai Jiao Tong University , Shanghai 200030, China
| | - Jiali Cai
- Changzheng Hospital, Secondary Military Medical University , Shanghai 200003, China
| | - Xiao Li
- Department of Nuclear Medicine, Zhongshan Hospital, Shanghai Medical College, Fudan University , Shanghai 200032, China
| | - Dengfeng Cheng
- Department of Nuclear Medicine, Zhongshan Hospital, Shanghai Medical College, Fudan University , Shanghai 200032, China
| | - Huilan Su
- State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University , Shanghai 200240, China
| | - Jianping Zhang
- Department of Nuclear Medicine, Shanghai Cancer Center, Fudan University , Shanghai 200032, China
| | - Shiyuan Liu
- Changzheng Hospital, Secondary Military Medical University , Shanghai 200003, China
| | - Hongcheng Shi
- Department of Nuclear Medicine, Zhongshan Hospital, Shanghai Medical College, Fudan University , Shanghai 200032, China
| | - Yingjian Zhang
- Department of Nuclear Medicine, Shanghai Cancer Center, Fudan University , Shanghai 200032, China
| | - Chunfu Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, School of Biomedical Engineering, Shanghai Jiao Tong University , Shanghai 200030, China
- Department of Nuclear Medicine, Rui Jin Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai 200025, China
| |
Collapse
|
34
|
HUNG MINGSZU, CHEN ICHUAN, YOU LIANG, JABLONS DAVIDM, LI YACHIN, MAO JIANHUA, XU ZHIDONG, HSIEH MENGJER, LIN YUCHING, YANG CHENGTA, LIU SHIHTUNG, TSAI YINGHUANG. Knockdown of Cul4A increases chemosensitivity to gemcitabine through upregulation of TGFBI in lung cancer cells. Oncol Rep 2015; 34:3187-95. [PMID: 26503734 DOI: 10.3892/or.2015.4324] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2015] [Accepted: 08/11/2015] [Indexed: 11/06/2022] Open
|
35
|
Han B, Cai H, Chen Y, Hu B, Luo H, Wu Y, Wu J. The role of TGFBI (βig-H3) in gastrointestinal tract tumorigenesis. Mol Cancer 2015; 14:64. [PMID: 25889002 PMCID: PMC4435624 DOI: 10.1186/s12943-015-0335-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2014] [Accepted: 03/09/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND TGFβ-induced (TGFBI/βig-H3) is a protein inducible by TGFβ1 and secreted by many types of cells. It binds to collagen, forms part of the extracellular matrix (ECM), and interacts with integrins on cell surfaces. In this study, we investigated the role of TGFBI in tumorigenesis and the underlying mechanisms. METHODS Patient serum TGFBI levels were determined by ELISA. TGFBI transgenic and gene knockout mice and TGFBI-overexpressing liver cells were used for mechanistic studies. RESULTS We demonstrated that patients with cholangiocarcinomas, hepatic carcinomas or gastric carcinomas presented significantly elevated serum TGFBI levels, and the excess TGFBI was derived from the tumor masses. TGFBI overexpression in mice resulted in increased incidence of spontaneous tumors and N,N-diethylnitrosamine (DEN)-induced liver tumor nodules, compared to that in wild type (WT) mice, while TGFBI knockout mice were comparable to WT controls in these 2 aspects. TGFBI promoted the survival of Aml-12 liver cells with DNA damage after irradiation, and augmented their post-irradiation proliferation. It activated the FAK/AKT/AKT1S1/PRS6/EIF4EBP pathway, which is known to modulate cell survival and proliferation. CONCLUSIONS Our data suggest that TGFBI functions as a promoter of certain gastrointestinal tract cancers. It provides a survival advantage to cells with DNA damage. Over a long time span, this advantage could translate into increased tumor risks.
Collapse
Affiliation(s)
- Bing Han
- Laboratory of Immunology and Cardiovascular Research, Centre hospitalier de l'Université de Montréal (CRCHUM), 900 Stain-Denis Street, Montreal, Quebec, Canada.
| | - Haolei Cai
- Department of Surgery, The Second Affiliated Hospital of Zhejiang University, 88 Jiefang Road, Hangzhou, China.
| | - Ying Chen
- Department of Surgery, The Second Affiliated Hospital of Zhejiang University, 88 Jiefang Road, Hangzhou, China.
| | - Bing Hu
- Anatomic Pathology, AmeriPath Central Florida, 8150 Chancellor Dr, Orlando, FL, USA.
| | - Hongyu Luo
- Laboratory of Immunology and Cardiovascular Research, Centre hospitalier de l'Université de Montréal (CRCHUM), 900 Stain-Denis Street, Montreal, Quebec, Canada.
| | - Yulian Wu
- Department of Surgery, The Second Affiliated Hospital of Zhejiang University, 88 Jiefang Road, Hangzhou, China.
| | - Jiangping Wu
- Laboratory of Immunology and Cardiovascular Research, Centre hospitalier de l'Université de Montréal (CRCHUM), 900 Stain-Denis Street, Montreal, Quebec, Canada. .,Nephrology Service, Research Centre, Centre hospitalier de l'Université de Montréal (CRCHUM), 900 Stain-Denis Street, Montreal, Quebec, Canada.
| |
Collapse
|
36
|
Epigenetic regulation of inflammatory cytokines and associated genes in human malignancies. Mediators Inflamm 2015; 2015:201703. [PMID: 25814785 PMCID: PMC4359879 DOI: 10.1155/2015/201703] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 02/18/2015] [Indexed: 12/12/2022] Open
Abstract
Inflammation is a multifaceted defense response of immune system against infection. Chronic inflammation has been implicated as an imminent threat for major human malignancies and is directly linked to various steps involved in tumorigenesis. Inflammatory cytokines, interleukins, interferons, transforming growth factors, chemokines, and adhesion molecules have been associated with chronic inflammation. Numerous cytokines are reported to be aberrantly regulated by different epigenetic mechanisms like DNA methylation and histone modifications in tumor tissues, contributing to pathogenesis of tumor in multiple ways. Some of these cytokines also work as epigenetic regulators of other crucial genes in tumor biology, either directly or indirectly. Such regulations are reported in lung, breast, cervical, gastric, colorectal, pancreatic, prostate, and head and neck cancers. Epigenetics of inflammatory mediators in cancer is currently subject of extensive research. These investigations may help in understanding cancer biology and to develop effective therapeutic strategies. The purpose of this paper is to have a brief view of the aberrant regulation of inflammatory cytokines in human malignancies.
Collapse
|
37
|
Rudra-Ganguly N, Lowe C, Mattie M, Chang MS, Satpayev D, Verlinsky A, An Z, Hu L, Yang P, Challita-Eid P, Stover DR, Pereira DS. Discoidin domain receptor 1 contributes to tumorigenesis through modulation of TGFBI expression. PLoS One 2014; 9:e111515. [PMID: 25369402 PMCID: PMC4219757 DOI: 10.1371/journal.pone.0111515] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Accepted: 09/26/2014] [Indexed: 12/18/2022] Open
Abstract
Discoidin domain receptor 1 (DDR1) is a member of the receptor tyrosine kinase family. The receptor is activated upon binding to its ligand, collagen, and plays a crucial role in many fundamental processes such as cell differentiation, adhesion, migration and invasion. Although DDR1 is expressed in many normal tissues, upregulated expression of DDR1 in a variety of human cancers such as lung, colon and brain cancers is known to be associated with poor prognosis. Using shRNA silencing, we assessed the oncogenic potential of DDR1. DDR1 knockdown impaired tumor cell proliferation and migration in vitro and tumor growth in vivo. Microarray analysis of tumor cells demonstrated upregulation of TGFBI expression upon DDR1 knockdown, which was subsequently confirmed at the protein level. TGFBI is a TGFβ-induced extracellular matrix protein secreted by the tumor cells and is known to act either as a tumor promoter or tumor suppressor, depending on the tumor environment. Here, we show that exogenous addition of recombinant TGFBI to BXPC3 tumor cells inhibited clonogenic growth and migration, thus recapitulating the phenotypic effect observed from DDR1 silencing. BXPC3 tumor xenografts demonstrated reduced growth with DDR1 knockdown, and the same xenograft tumors exhibited an increase in TGFBI expression level. Together, these data suggest that DDR1 expression level influences tumor growth in part via modulation of TGFBI expression. The reciprocal expression of DDR1 and TGFBI may help to elucidate the contribution of DDR1 in tumorigenesis and TGFBI may also be used as a biomarker for the therapeutic development of DDR1 specific inhibitors.
Collapse
Affiliation(s)
- Nandini Rudra-Ganguly
- Agensys Inc., an affiliate of Astellas Pharma Inc, Santa Monica, CA, United States of America
- * E-mail:
| | - Christine Lowe
- Agensys Inc., an affiliate of Astellas Pharma Inc, Santa Monica, CA, United States of America
| | - Michael Mattie
- Agensys Inc., an affiliate of Astellas Pharma Inc, Santa Monica, CA, United States of America
| | - Mi Sook Chang
- Agensys Inc., an affiliate of Astellas Pharma Inc, Santa Monica, CA, United States of America
| | | | - Alla Verlinsky
- Agensys Inc., an affiliate of Astellas Pharma Inc, Santa Monica, CA, United States of America
| | - Zili An
- Agensys Inc., an affiliate of Astellas Pharma Inc, Santa Monica, CA, United States of America
| | - Liping Hu
- Agensys Inc., an affiliate of Astellas Pharma Inc, Santa Monica, CA, United States of America
| | - Peng Yang
- Agensys Inc., an affiliate of Astellas Pharma Inc, Santa Monica, CA, United States of America
| | - Pia Challita-Eid
- Agensys Inc., an affiliate of Astellas Pharma Inc, Santa Monica, CA, United States of America
| | - David R. Stover
- Agensys Inc., an affiliate of Astellas Pharma Inc, Santa Monica, CA, United States of America
| | - Daniel S. Pereira
- Agensys Inc., an affiliate of Astellas Pharma Inc, Santa Monica, CA, United States of America
| |
Collapse
|
38
|
Salvo E, Garasa S, Dotor J, Morales X, Peláez R, Altevogt P, Rouzaut A. Combined targeting of TGF-β1 and integrin β3 impairs lymph node metastasis in a mouse model of non-small-cell lung cancer. Mol Cancer 2014; 13:112. [PMID: 24884715 PMCID: PMC4049383 DOI: 10.1186/1476-4598-13-112] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Accepted: 05/09/2014] [Indexed: 12/31/2022] Open
Abstract
Background Transforming Growth Factor beta (TGF-β) acts as a tumor suppressor early in carcinogenesis but turns into tumor promoter in later disease stages. In fact, TGF-β is a known inducer of integrin expression by tumor cells which contributes to cancer metastatic spread and TGF-β inhibition has been shown to attenuate metastasis in mouse models. However, carcinoma cells often become refractory to TGF-β-mediated growth inhibition. Therefore identifying patients that may benefit from anti-TGF-β therapy requires careful selection. Methods We performed in vitro analysis of the effects of exposure to TGF-β in NSCLC cell chemotaxis and adhesion to lymphatic endothelial cells. We also studied in an orthotopic model of NSCLC the incidence of metastases to the lymph nodes after inhibition of TGF-β signaling, β3 integrin expression or both. Results We offer evidences of increased β3-integrin dependent NSCLC adhesion to lymphatic endothelium after TGF-β exposure. In vivo experiments show that targeting of TGF-β and β3 integrin significantly reduces the incidence of lymph node metastasis. Even more, blockade of β3 integrin expression in tumors that did not respond to TGF-β inhibition severely impaired the ability of the tumor to metastasize towards the lymph nodes. Conclusion These findings suggest that lung cancer tumors refractory to TGF-β monotherapy can be effectively treated using dual therapy that combines the inhibition of tumor cell adhesion to lymphatic vessels with stromal TGF-β inhibition.
Collapse
|
39
|
Zhao T, Sun Q, del Rincon SV, Lovato A, Marques M, Witcher M. Gallotannin imposes S phase arrest in breast cancer cells and suppresses the growth of triple-negative tumors in vivo. PLoS One 2014; 9:e92853. [PMID: 24658335 PMCID: PMC3962455 DOI: 10.1371/journal.pone.0092853] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Accepted: 02/27/2014] [Indexed: 12/18/2022] Open
Abstract
Triple-negative breast cancers are associated with poor clinical outcomes and new therapeutic strategies are clearly needed. Gallotannin (Gltn) has been previously demonstrated to have potent anti-tumor properties against cholangiocarcinoma in mice, but little is known regarding its capacity to suppress tumor outgrowth in breast cancer models. We tested Gltn for potential growth inhibitory properties against a variety of breast cancer cell lines in vitro. In particular, triple-negative breast cancer cells display higher levels of sensitivity to Gltn. The loss of proliferative capacity in Gltn exposed cells is associated with slowed cell cycle progression and S phase arrest, dependent on Chk2 phosphorylation and further characterized by changes to proliferation related genes, such as cyclin D1 (CcnD1) as determined by Nanostring technology. Importantly, Gltn administered orally or via intraperitoneal (IP) injections greatly reduced tumor outgrowth of triple-negative breast cells from mammary fat pads without signs of toxicity. In conclusion, these data strongly suggest that Gltn represents a novel approach to treat triple-negative breast carcinomas.
Collapse
Affiliation(s)
- Tiejun Zhao
- The Lady Davis Institute and Segal Cancer Center of the Jewish General Hospital, McGill University, Montreal, Quebec, Canada
| | - Qiang Sun
- The Lady Davis Institute and Segal Cancer Center of the Jewish General Hospital, McGill University, Montreal, Quebec, Canada
| | - Sonia V. del Rincon
- The Lady Davis Institute and Segal Cancer Center of the Jewish General Hospital, McGill University, Montreal, Quebec, Canada
| | - Amanda Lovato
- The Lady Davis Institute and Segal Cancer Center of the Jewish General Hospital, McGill University, Montreal, Quebec, Canada
| | - Maud Marques
- The Lady Davis Institute and Segal Cancer Center of the Jewish General Hospital, McGill University, Montreal, Quebec, Canada
| | - Michael Witcher
- The Lady Davis Institute and Segal Cancer Center of the Jewish General Hospital, McGill University, Montreal, Quebec, Canada
- * E-mail:
| |
Collapse
|
40
|
Pajares MJ, Agorreta J, Salvo E, Behrens C, Wistuba II, Montuenga LM, Pio R, Rouzaut A. TGFBI expression is an independent predictor of survival in adjuvant-treated lung squamous cell carcinoma patients. Br J Cancer 2014; 110:1545-51. [PMID: 24481402 PMCID: PMC3960613 DOI: 10.1038/bjc.2014.33] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Revised: 12/23/2013] [Accepted: 01/08/2014] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Transforming growth factor β-induced protein (TGFBI) is a secreted protein that mediates cell anchoring to the extracellular matrix. This protein is downregulated in lung cancer, and when overexpressed, contributes to apoptotic cell death. Using a small series of stage IV non-small cell lung cancer (NSCLC) patients, we previously suggested the usefulness of TGFBI as a prognostic and predictive factor in chemotherapy-treated late-stage NSCLC. In order to validate and extend these results, we broaden the analysis and studied TGFBI expression in a large series of samples obtained from stage I-IV NSCLC patients. METHODS TGFBI expression was assessed by immunohistochemistry in 364 completely resected primary NSCLC samples: 242 adenocarcinomas (ADCs) and 122 squamous cell carcinomas (SCCs). Kaplan-Meier curves, log-rank tests and the Cox proportional hazards model were used to analyse the association between TGFBI expression and survival. RESULTS High TGFBI levels were associated with longer overall survival (OS, P<0.001) and progression-free survival (PFS, P<0.001) in SCC patients who received adjuvant platinium-based chemotherapy. Moreover, multivariate analysis demonstrated that high TGFBI expression is an independent predictor of better survival in patients (OS: P=0.030 and PFS: P=0.026). CONCLUSIONS TGFBI may be useful for the identification of a subset of NSCLC who may benefit from adjuvant therapy.
Collapse
Affiliation(s)
- M J Pajares
- 1] Division of Oncology, Center for Applied Medical Research, University of Navarra, 31080 Pamplona, Spain [2] Department of Histology and Pathology, University of Navarra, 31080 Pamplona, Spain
| | - J Agorreta
- 1] Division of Oncology, Center for Applied Medical Research, University of Navarra, 31080 Pamplona, Spain [2] Department of Histology and Pathology, University of Navarra, 31080 Pamplona, Spain
| | - E Salvo
- 1] Division of Oncology, Center for Applied Medical Research, University of Navarra, 31080 Pamplona, Spain [2] Department of Biochemistry and Genetics, University of Navarra, 31080 Pamplona, Spain
| | - C Behrens
- Department of Pathology and Thoracic/Head and Neck Medical Oncology, The University of Texas, M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - I I Wistuba
- Department of Pathology and Thoracic/Head and Neck Medical Oncology, The University of Texas, M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - L M Montuenga
- 1] Division of Oncology, Center for Applied Medical Research, University of Navarra, 31080 Pamplona, Spain [2] Department of Biochemistry and Genetics, University of Navarra, 31080 Pamplona, Spain
| | - R Pio
- 1] Department of Biochemistry and Genetics, University of Navarra, 31080 Pamplona, Spain [2] Department of Pathology and Thoracic/Head and Neck Medical Oncology, The University of Texas, M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - A Rouzaut
- 1] Department of Biochemistry and Genetics, University of Navarra, 31080 Pamplona, Spain [2] Department of Pathology and Thoracic/Head and Neck Medical Oncology, The University of Texas, M.D. Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
41
|
Integrative Analysis of Gene Expression and Promoter Methylation during Reprogramming of a Non-Small-Cell Lung Cancer Cell Line Using Principal Component Analysis-Based Unsupervised Feature Extraction. INTELLIGENT COMPUTING IN BIOINFORMATICS 2014. [DOI: 10.1007/978-3-319-09330-7_52] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
|
42
|
Zhang HH, Zhang ZY, Che CL, Mei YF, Shi YZ. Array analysis for potential biomarker of gemcitabine identification in non-small cell lung cancer cell lines. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2013; 6:1734-1746. [PMID: 24040438 PMCID: PMC3759480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Accepted: 08/05/2013] [Indexed: 06/02/2023]
Abstract
Gemcitabine is one of the most widely used drugs for the treatment of advanced Non-small cell lung cancer (NSCLC), but modest objective response rate of patients to gemcitabine makes it necessary to identify novel biomarkers for patients who can benefit from gemcitabine-based therapy and to improve the effect of clinical therapy. In this work, 3 NSCLC cell lines displaying different sensitivities to gemcitabine were applied for mRNA and microRNA (miR) expression chips to figure out the biomarkers for gemcitabine sensitivity. Genes whose expression increased dramatically in sensitive cell lines were mainly enriched in cell adhesion (NRP2, CXCR3, CDK5R1, IL32 and CDH2) and secretory granule (SLC11A1, GP5, CD36 and IGF1), while genes with significantly upregulated expression in resistant cell line were mainly clustered in methylation modification (HIST1H2BF, RAB23 and TP53) and oxidoreductase (TP53I3, CYP27B1 and SOD3). The most intriguing is the activation of Wnt/β-catenin signaling in gemcitabine resistant NSCLC cell lines. The miR-155, miR-10a, miR-30a, miR-24-2* and miR-30c-2* were upregulated in sensitive cell lines, while expression of miR-200c, miR-203, miR-885-5p, miR-195 and miR-25* was increased in resistant cell line. Genes with significantly altered expression and putatively mediated by the expression-changed miRs were mainly enriched in chromatin assembly (MAF, HLF, BCL2, and IGSF3), anti-apoptosis (BCL2, IGF1 and IKBKB), protein kinase (NRP2, PAK7 and CDK5R1) (all the above genes were upregulated in sensitive cells) and small GTPase mediated signal transduction (GNA13, RAP2A, ARHGAP5 and RAB23, down-regulated in sensitive cells). Our results might provide potential biomarkers for gemcitabine sensitivity prediction and putative targets to overcome gemcitabine resistance in NSCLC patients.
Collapse
MESH Headings
- Antimetabolites, Antineoplastic/pharmacology
- Biomarkers, Tumor/genetics
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/pathology
- Cell Line, Tumor
- Cell Survival/drug effects
- Deoxycytidine/analogs & derivatives
- Deoxycytidine/pharmacology
- Dose-Response Relationship, Drug
- Drug Resistance, Neoplasm/genetics
- Drug Screening Assays, Antitumor
- Gene Expression Profiling/methods
- Gene Expression Regulation, Neoplastic/drug effects
- Gene Regulatory Networks/drug effects
- Genotype
- Humans
- Inhibitory Concentration 50
- Lung Neoplasms/genetics
- Lung Neoplasms/pathology
- MicroRNAs/metabolism
- Oligonucleotide Array Sequence Analysis
- Patient Selection
- Phenotype
- RNA, Messenger/metabolism
- Gemcitabine
Collapse
Affiliation(s)
- Hai-Hong Zhang
- Department of rheumatism and immunology, First Clinical Medical College affiliated to Harbin Medical UniversityHarbin, China
| | - Zhi-Yi Zhang
- Department of rheumatism and immunology, First Clinical Medical College affiliated to Harbin Medical UniversityHarbin, China
| | - Chun-Li Che
- Department of respiratory medicine, First Clinical Medical College affiliated to Harbin Medical UniversityHarbin, China
| | - Yi-Fang Mei
- Department of respiratory medicine, First Clinical Medical College affiliated to Harbin Medical UniversityHarbin, China
| | - Yu-Zhi Shi
- Department of respiratory medicine, First Clinical Medical College affiliated to Harbin Medical UniversityHarbin, China
| |
Collapse
|
43
|
The Xenopus Tgfbi is required for embryogenesis through regulation of canonical Wnt signalling. Dev Biol 2013; 379:16-27. [DOI: 10.1016/j.ydbio.2012.11.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2011] [Revised: 09/18/2012] [Accepted: 11/12/2012] [Indexed: 10/27/2022]
|
44
|
Salem S, Harris T, Mok JSL, Li MYS, Keenan CR, Schuliga MJ, Stewart AG. Transforming growth factor-β impairs glucocorticoid activity in the A549 lung adenocarcinoma cell line. Br J Pharmacol 2012; 166:2036-48. [PMID: 22300324 DOI: 10.1111/j.1476-5381.2012.01885.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND AND PURPOSE The lung adenocarcinoma cell line, A549, undergoes epithelial-mesenchymal cell transition (EMT) in response to TGF-β. Glucocorticoids do not prevent the EMT response, but TGF-β induced resistance to the cytokine-regulatory action of glucocorticoids. We sought to characterize the impairment of glucocorticoid response in A549 cells. EXPERIMENTAL APPROACH A549 cells were exposed to TGF-β for up to 96 h before glucocorticoid treatment and challenge with IL-1α to assess glucocorticoid regulation of IL-6 and CXCL8 production. Nuclear localization of the glucocorticoid receptor α (GRα) was ascertained by immunofluorescence and Western blotting. Transactivation of the glucocorticoid response element (GRE) was measured with a transfected GRE-secreted human placental alkaline phosphatase reporter. KEY RESULTS TGF-β (40-400 pM) reduced the maximum inhibitory effect of dexamethasone on IL-1α-induced IL-6 and CXCL8 production. The impaired glucocorticoid response was detected with 4 h of TGF-β (40 pM) exposure (and 4 h IL-1α to induce CXCL8 expression) and therefore was not secondary to EMT, a process that requires longer incubation periods and higher concentrations of TGF-β. TGF-β also impaired dexamethasone regulation of granulocyte-macrophage colony-stimulating factor in thrombin-stimulated BEAS-2B epithelial cells. Impaired regulation of CXCL8 was associated with markedly reduced GRE transactivation and reduced induction of mRNA for IκBα, the glucocorticoid-inducible leucine zipper and the epithelial sodium channel (SCNN1A). The expression, cellular levels and nuclear localization of GRα were reduced by TGF-β. CONCLUSIONS AND IMPLICATIONS We have identified mechanisms underlying the impairment of responses to glucocorticoids by TGF-β in the A549 and BEAS-2B cell lines.
Collapse
Affiliation(s)
- S Salem
- Department of Pharmacology, University of Melbourne, Vic., Australia
| | | | | | | | | | | | | |
Collapse
|
45
|
Oliemuller E, Peláez R, Garasa S, Pajares MJ, Agorreta J, Pío R, Montuenga LM, Teijeira A, Llanos S, Rouzaut A. Phosphorylated tubulin adaptor protein CRMP-2 as prognostic marker and candidate therapeutic target for NSCLC. Int J Cancer 2012; 132:1986-95. [PMID: 23023514 DOI: 10.1002/ijc.27881] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2012] [Accepted: 09/18/2012] [Indexed: 11/08/2022]
Abstract
Collapsin response mediator protein-2 (CRMP-2) is the first described and most studied member of a family of proteins that mediate the addition of tubulin dimers to the growing microtubule. CRMPs have mainly been studied in the nervous system, but recently, they have been described in other tissues where they participate in vesicle transport, migration and mitosis. In this work, we aimed at studying the role of CRMP-2 in lung cancer cell division. We first explored the expression of CRMP-2 and phosphorylated (Thr 514) CRMP-2 in 91 samples obtained from patients with localized nonsmall cell lung cancer. We observed a significant correlation between high levels of nuclear phosphorylated CRMP-2 and poor prognosis in those patients. Interestingly, this association was only positive for untreated patients. To provide a mechanistic explanation to these findings, we used in vitro models to analyze the role of CRMP-2 and its phosphorylated forms in cell division. Thus, we observed by confocal microscopy and immunoprecipitation assays that CRMP-2 differentially colocalizes with the mitotic spindle during cell division. The use of phosphodefective or phosphomimetic mutants of CRMP-2 allowed us to prove that anomalies in the phosphorylation status of CRMP-2 result in changes in the mitotic tempo, and increments in the number of multinucleated cells. Finally, here we demonstrate that CRMP-2 phosphorylation impairment, or silencing induces p53 expression and promotes apoptosis through caspase 3 activation. These results pointed to CRMP-2 phosphorylation as a prognostic marker and potential new target to be explored in cancer therapy.
Collapse
Affiliation(s)
- Erik Oliemuller
- Oncology Division, Center for Applied Medical Research (CIMA), University of Navarra, 55 Pamplona, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Yi M. Systems analysis of a mouse xenograft model reveals annexin A1 as a regulator of gene expression in tumor stroma. PLoS One 2012; 7:e43551. [PMID: 23077482 PMCID: PMC3471933 DOI: 10.1371/journal.pone.0043551] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2012] [Accepted: 07/23/2012] [Indexed: 12/26/2022] Open
Abstract
Annexin A1 is a multi functional molecule which is involved in inflammation, innate and adaptive immune systems, tumor progression and metastasis. We have previously showed the impaired tumor growth, metastasis, angiogenesis and wound healing in annexin A1 knockout mice. While tumor is a piece of heterogeneous mass including not only malignant tumor cells but also the stroma, the importance of the tumor stroma for tumor progression and metastasis is becoming increasingly clear. The tumor stroma is comprised by various components including extracellular matrix and non-malignant cells in the tumor, such as endothelial cells, fibroblasts, immune cells, inflammatory cells. Based on our previous finding of pro-angiogenic functions for annexin A1 in vascular endothelial cell sprouting, wound healing, tumor growth and metastasis, and the previously known properties for annexin A1 in immune cells and inflammation, this study hypothesized that annexin A1 is a key functional player in tumor development, linking the various components in tumor stroma by its actions in endothelial cells and immune cells. Using systems analysis programs commercially available, this paper further compared the gene expression between tumors from annexin A1 wild type mice and annexin A1 knockout mice and found a list of genes that significantly changed in the tumor stroma that lacked annexin A1. This revealed annexin A1 to be an effective regulator in tumor stroma and suggested a mechanism that annexin A1 affects tumor development and metastasis through interaction with the various components in the microenvironment surrounding the tumor cells.
Collapse
Affiliation(s)
- Ming Yi
- Sidney Kimmel Cancer Center and Proteogenomics Research Institute for Systems Medicine, San Diego, California, USA.
| |
Collapse
|
47
|
Transforming growth Factor-Beta-Induced Protein (TGFBI)/(βig-H3): a matrix protein with dual functions in ovarian cancer. Int J Mol Sci 2012; 13:10461-10477. [PMID: 22949874 PMCID: PMC3431872 DOI: 10.3390/ijms130810461] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 08/03/2012] [Accepted: 08/16/2012] [Indexed: 02/04/2023] Open
Abstract
Transforming growth factor-beta-induced protein (TGFBI, also known as βig-H3 and keratoepithelin) is an extracellular matrix protein that plays a role in a wide range of physiological and pathological conditions including diabetes, corneal dystrophy and tumorigenesis. Many reports indicate that βig-H3 functions as a tumor suppressor. Loss of βig-H3 expression has been described in several cancers including ovarian cancer and promoter hypermethylation has been identified as an important mechanism for the silencing of the TGFBI gene. Our recent findings that βig-H3 is down-regulated in ovarian cancer and that high concentrations of βig-H3 can induce ovarian cancer cell death support a tumor suppressor role. However, there is also convincing data in the literature reporting a tumor-promoting role for βig-H3. We have shown βig-H3 to be abundantly expressed by peritoneal cells and increase the metastatic potential of ovarian cancer cells by promoting cell motility, invasion, and adhesion to peritoneal cells. Our findings suggest that βig-H3 has dual functions and can act both as a tumor suppressor or tumor promoter depending on the tumor microenvironment. This article reviews the current understanding of βig-H3 function in cancer cells with particular focus on ovarian cancer.
Collapse
|
48
|
Zhang C, Xie X, Liang S, Li M, Liu Y, Gu H. Mono-dispersed high magnetic resonance sensitive magnetite nanocluster probe for detection of nascent tumors by magnetic resonance molecular imaging. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2012; 8:996-1006. [DOI: 10.1016/j.nano.2011.11.013] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2011] [Revised: 11/02/2011] [Accepted: 11/22/2011] [Indexed: 12/27/2022]
|
49
|
Tumbarello DA, Temple J, Brenton JD. ß3 integrin modulates transforming growth factor beta induced (TGFBI) function and paclitaxel response in ovarian cancer cells. Mol Cancer 2012; 11:36. [PMID: 22640878 PMCID: PMC3442987 DOI: 10.1186/1476-4598-11-36] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2011] [Accepted: 05/28/2012] [Indexed: 02/05/2023] Open
Abstract
Background The extracellular matrix (ECM) has a key role in facilitating the progression of ovarian cancer and we have shown recently that the secreted ECM protein TGFBI modulates the response of ovarian cancer to paclitaxel-induced cell death. Results We have determined TGFBI signaling from the extracellular environment is preferential for the cell surface αvß3 integrin heterodimer, in contrast to periostin, a TGFBI paralogue, which signals primarily via a ß1 integrin-mediated pathway. We demonstrate that suppression of ß1 integrin expression, in ß3 integrin-expressing ovarian cancer cells, increases adhesion to rTGFBI. In addition, Syndecan-1 and −4 expression is dispensable for adhesion to rTGFBI and loss of Syndecan-1 cooperates with the loss of ß1 integrin to further enhance adhesion to rTGFBI. The RGD motif present in the carboxy-terminus of TGFBI is necessary, but not sufficient, for SKOV3 cell adhesion and is dispensable for adhesion of ovarian cancer cells lacking ß3 integrin expression. In contrast to TGFBI, the carboxy-terminus of periostin, lacking a RGD motif, is unable to support adhesion of ovarian cancer cells. Suppression of ß3 integrin in SKOV3 cells increases resistance to paclitaxel-induced cell death while suppression of ß1 integrin has no effect. Furthermore, suppression of TGFBI expression stimulates a paclitaxel resistant phenotype while suppression of fibronectin expression, which primarily signals through a ß1 integrin-mediated pathway, increases paclitaxel sensitivity. Conclusions Therefore, different ECM components use distinct signaling mechanisms in ovarian cancer cells and in particular, TGFBI preferentially interacts through a ß3 integrin receptor mediated mechanism to regulate the response of cells to paclitaxel-induced cell death.
Collapse
Affiliation(s)
- David A Tumbarello
- Cancer Research UK, Cambridge Research Institute, Robinson Way, Cambridge CB2 0RE, United Kingdom
| | | | | |
Collapse
|
50
|
Holmberg C, Quante M, Steele I, Kumar JD, Balabanova S, Duval C, Czepan M, Rakonczay Z, Tiszlavicz L, Nemeth I, Lazar G, Simonka Z, Jenkins R, Hegyi P, Wang TC, Dockray GJ, Varro A. Release of TGFβig-h3 by gastric myofibroblasts slows tumor growth and is decreased with cancer progression. Carcinogenesis 2012; 33:1553-62. [PMID: 22610072 DOI: 10.1093/carcin/bgs180] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Tumor progression has been linked to changes in the stromal environment. Myofibroblasts are stromal cells that are often increased in tumors but their contribution to cancer progression is not well understood. Here, we show that the secretomes of myofibroblasts derived from gastric cancers [cancer-associated myofibroblasts (CAMs)] differ in a functionally significant manner from those derived from adjacent tissue [adjacent tissue myofibroblasts (ATMs)]. CAMs showed increased rates of migration and proliferation compared with ATMs or normal tissue myofibroblasts (NTMs). Moreover, conditioned medium (CM) from CAMs significantly stimulated migration, invasion and proliferation of gastric cancer cells compared with CM from ATMs or NTMs. Proteomic analysis of myofibroblast secretomes revealed decreased abundance of the extracellular matrix (ECM) adaptor protein like transforming growth factor-β-induced gene-h3 (TGFβig-h3) in CAMs, which was correlated with lymph node involvement and shorter survival. TGFβig-h3 inhibited IGF-II-stimulated migration and proliferation of both cancer cells and myofibroblasts, and suppressed IGF-II activation of p42/44 MAPkinase; TGFβig-h3 knockdown increased IGF-II- and CM-stimulated migration. Furthermore, administration of TGFβig-h3 inhibited myofibroblast-stimulated growth of gastric cancer xenografts. We conclude that stromal cells exert inhibitory as well as stimulatory effects on tumor cells; TGFβig-h3 is a stromal inhibitory factor that is decreased with progression of gastric cancers.
Collapse
Affiliation(s)
- Chris Holmberg
- Department of Cellular & Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|