1
|
Barcellos-Hoff MH, Yom SS. Revisiting the TGFβ paradox: insights from HPV-driven cancer and the DNA damage response. Nat Rev Cancer 2025:10.1038/s41568-025-00819-6. [PMID: 40389543 DOI: 10.1038/s41568-025-00819-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/01/2025] [Indexed: 05/21/2025]
Abstract
The transforming growth factor-β (TGFβ) paradox refers to the well-established role of TGFβ in suppressing cancer in healthy tissues yet promoting malignancy in established cancers. Although this positioned TGFβ inhibitors as a potential therapeutic strategy for malignancy, therapuetic blockade has failed in multiple clinical trials. The general lack of selection principles for defining which patients would most benefit from the addition of a TGFβ inhibitor has probably hindered its deployment. Here, we highlight the therapeutic potential in TGFβ regulation of DNA repair using human papillomavirus (HPV)-driven head and neck squamous cell carcinoma (HNSCC) as an illustrative example. HPV inhibits TGFβ signalling, which in turn reduces DNA damage repair, ultimately conferring sensitivity to cancer treatments and thus contributing to the favourable prognosis of HPV-positive HNSCC. Here, we review the DNA repair deficit caused by a loss of TGFβ signalling and how this could be targeted to induce synthetic lethality. Moreover, we explore its role in predicting response to immune checkpoint inhibitors and the potential of biomarkers to select which patients with cancer could ultimately benefit from TGFβ inhibition.
Collapse
Affiliation(s)
| | - Sue S Yom
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
2
|
Xue X, Zhou Y, Liu H, Gao Y, Ma X, Xiao Z, Zhang W, Li P, Tian Z, Luan Y, Zhao X. A novel missense mutation Smad4 V354L enhances the efficacy of docetaxel in non-small cell lung cancer. Biomed Pharmacother 2025; 184:117899. [PMID: 39923405 DOI: 10.1016/j.biopha.2025.117899] [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: 10/08/2024] [Revised: 01/31/2025] [Accepted: 02/03/2025] [Indexed: 02/11/2025] Open
Abstract
NSCLC is a heterogeneous disease with unique combinations of somatic molecular alterations in individual patients. The different mutations in tumor oncogene and suppressors might be associated with the response to therapy. However, little is known about how Smad4 genomic alterations cause the therapeutic effect of docetaxel. The retrospective analysis was conducted on 49 patients with stage IIB or IIIA non-small cell lung cancer receiving docetaxel chemotherapy. One novel missense variant, c.1060 G > C in Smad4 was identified by next-generation sequencing. The Smad4c.1060 G > C variant results in the substitution of valine with leucine at amino acid 354 (p.Val354Leu, V354L). The clinical analysis showed that the patients with Smad4 V354L mutation receiving docetaxel treatment manifested better prognosis with prolonged disease-free survival and overall survival compared with patients with the wild-type. Smad4 V354L cells demonstrated increased sensitivity to docetaxel with apoptosis and G2/M cell cycle arrest. Furthermore, the cell-cycle related protein expression of CDK2 was remarkably decreased, while the expression of cyclin-dependent kinase inhibitor p21 Waf1 and p27 Kip1 was significantly increased. In vivo experiments further demonstrated the increased inhibitory effects of docetaxel in the nude mice inoculated with Smad4 V354L cells compared to the mice inoculated with wild-type cells group. The novel V354L missense mutation of Smad4 gene enhances the efficacy of docetaxel in non-small cell lung cancer, which would provide new opportunities for precise clinical therapy of NSCLC.
Collapse
Affiliation(s)
- Xia Xue
- Department of Pharmacy, The Second Hospital, Cheeloo College of Medicine, Shandong University, 250033, China
| | - Yongjia Zhou
- Department of Thoracic Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, 250033, China
| | - Huiping Liu
- School of Pharmacy, Yantai University, Yantai 264005, China
| | - Yan Gao
- Department of Pharmacy, The Second Hospital, Cheeloo College of Medicine, Shandong University, 250033, China
| | - Xinyu Ma
- Department of Pharmacy, The Second Hospital, Cheeloo College of Medicine, Shandong University, 250033, China
| | - Zhaohua Xiao
- Department of Thoracic Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, 250033, China
| | - Wenhao Zhang
- Department of Thoracic Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, 250033, China
| | - Peichao Li
- Department of Thoracic Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, 250033, China; Key Laboratory of Thoracic Cancer in Universities of Shandong, The Second Hospital, Cheeloo College of Medicine, Shandong University, 250033, China
| | - Zhongxian Tian
- Department of Thoracic Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, 250033, China; Key Laboratory of Thoracic Cancer in Universities of Shandong, The Second Hospital, Cheeloo College of Medicine, Shandong University, 250033, China
| | - Yun Luan
- Central Laboratory, Institute of Medical Science, The Second Hospital, Cheeloo College of Medicine, Shandong University, 250033, China
| | - Xiaogang Zhao
- Department of Thoracic Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, 250033, China; Key Laboratory of Thoracic Cancer in Universities of Shandong, The Second Hospital, Cheeloo College of Medicine, Shandong University, 250033, China.
| |
Collapse
|
3
|
Citro S, Miccolo C, Medda A, Ghiani L, Tagliabue M, Ansarin M, Chiocca S. HPV-mediated regulation of SMAD4 modulates the DNA damage response in head and neck cancer. J Exp Clin Cancer Res 2022; 41:59. [PMID: 35144669 PMCID: PMC8830113 DOI: 10.1186/s13046-022-02258-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 01/17/2022] [Indexed: 01/17/2023] Open
Abstract
Background Head and Neck cancer (HNC) is a fatal malignancy with poor prognosis. Human Papillomavirus (HPV) infection is becoming the prominent cause of HNC in the western world, and studying the molecular mechanisms underlying its action in cancers is key towards targeted therapy. To replicate, HPV regulates the host DNA damage repair (DDR) pathway. SMAD4 is also involved in the regulation of the DDR machinery and likely plays important role in maintaining cell viability upon genotoxic stress. In this study, we investigated the role of HPV in the upregulation of SMAD4 to control the DDR response and facilitate its lifecycle. Methods SMAD4, Rad51 and CHK1 expression was assessed in HPV-positive and HPV-negative HNC using TCGA data, a panel of 14 HNC cell lines and 8 fresh tumour tissue samples from HNC patients. HPV16 expression was modulated by E6/E7 siRNA knock-down or transduction in HPV-positive HNC cell lines and Human Primary keratinocytes respectively. SMAD4 half-life was assessed by cycloheximide treatment in HNC cell lines, together with βTRCP1-dependent SMAD4 ubiquitination. SMAD4 siRNA knock-down was used to determine its role in HPV-mediated regulation of DDR machinery and to assess cisplatin sensitivity in HPV-positive HNC cell lines. Results We found that HPV increases SMAD4 expression is both HPV-positive HNC tumours and cell lines, impairing its degradation which is mediated by the E3 ubiquitin ligase βTRCP1. SMAD4 expression highly correlates with the expression of two main players of the DDR pathway, CHK1 and Rad51, which expression is also upregulated by the presence of HPV. In particular, we demonstrate that HPV stabilizes SMAD4 to increase CHK1 and Rad51 expression. In addition, SMAD4-deficient HPV-positive cells have increased sensitivity to cisplatin treatment. Conclusions Our results give a clear molecular mechanism at the basis of HPV regulation of the DDR pathway. In particular, we show how HPV stabilizes SMAD4 to promote DDR protein expression, which may be used to facilitate viral replication and HNC onset. Moreover, we found that SMAD4 silencing in HPV-positive HNC cell lines increases sensitivity to cisplatin treatment, suggesting that HPV-positive HNC with low SMAD4 expression may be preferentially susceptible to similar treatments. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-022-02258-9.
Collapse
Affiliation(s)
- Simona Citro
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, IEO Campus, Via Adamello 16, 20139, Milan, Italy.
| | - Claudia Miccolo
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, IEO Campus, Via Adamello 16, 20139, Milan, Italy
| | - Alessandro Medda
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, IEO Campus, Via Adamello 16, 20139, Milan, Italy
| | - Lavinia Ghiani
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, IEO Campus, Via Adamello 16, 20139, Milan, Italy
| | - Marta Tagliabue
- Division of Otolaryngology Head & Neck Surgery, IEO, European Institute of Oncology IRCCS, 20141, Milan, Italy.,Department of Biomedical Sciences, University of Sassari, Sassari, Italy, University of Sassari, Sassari, Italy
| | - Mohssen Ansarin
- Division of Otolaryngology Head & Neck Surgery, IEO, European Institute of Oncology IRCCS, 20141, Milan, Italy
| | - Susanna Chiocca
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, IEO Campus, Via Adamello 16, 20139, Milan, Italy.
| |
Collapse
|
4
|
Barbirou M, Miller A, Manjunath Y, Ramirez AB, Ericson NG, Staveley-O’Carroll KF, Mitchem JB, Warren WC, Chaudhuri AA, Huang Y, Li G, Tonellato PJ, Kaifi JT. Single Circulating-Tumor-Cell-Targeted Sequencing to Identify Somatic Variants in Liquid Biopsies in Non-Small-Cell Lung Cancer Patients. Curr Issues Mol Biol 2022; 44:750-763. [PMID: 35723337 PMCID: PMC8928994 DOI: 10.3390/cimb44020052] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 01/30/2022] [Accepted: 01/31/2022] [Indexed: 12/12/2022] Open
Abstract
Non-small-cell lung cancer (NSCLC) accounts for most cancer-related deaths worldwide. Liquid biopsy by a blood draw to detect circulating tumor cells (CTCs) is a tool for molecular profiling of cancer using single-cell and next-generation sequencing (NGS) technologies. The aim of the study was to identify somatic variants in single CTCs isolated from NSCLC patients by targeted NGS. Thirty-one subjects (20 NSCLC patients, 11 smokers without cancer) were enrolled for blood draws (7.5 mL). CTCs were identified by immunofluorescence, individually retrieved, and DNA-extracted. Targeted NGS was performed to detect somatic variants (single-nucleotide variants (SNVs) and insertions/deletions (Indels)) across 65 oncogenes and tumor suppressor genes. Cancer-associated variants were classified using OncoKB database. NSCLC patients had significantly higher CTC counts than control smokers (p = 0.0132; Mann–Whitney test). Analyzing 23 CTCs and 13 white blood cells across seven patients revealed a total of 644 somatic variants that occurred in all CTCs within the same subject, ranging from 1 to 137 per patient. The highest number of variants detected in ≥1 CTC within a patient was 441. A total of 18/65 (27.7%) genes were highly mutated. Mutations with oncogenic impact were identified in functional domains of seven oncogenes/tumor suppressor genes (NF1, PTCH1, TP53, SMARCB1, SMAD4, KRAS, and ERBB2). Single CTC-targeted NGS detects heterogeneous and shared mutational signatures within and between NSCLC patients. CTC single-cell genomics have potential for integration in NSCLC precision oncology.
Collapse
Affiliation(s)
- Mouadh Barbirou
- Center for Biomedical Informatics, Department of Health Management and Informatics, School of Medicine, University of Missouri, Columbia, MO 65212, USA; (M.B.); (A.M.); (P.J.T.)
| | - Amanda Miller
- Center for Biomedical Informatics, Department of Health Management and Informatics, School of Medicine, University of Missouri, Columbia, MO 65212, USA; (M.B.); (A.M.); (P.J.T.)
| | - Yariswamy Manjunath
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO 65212, USA; (Y.M.); (K.F.S.-O.); (J.B.M.); (G.L.)
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO 65201, USA
| | | | | | - Kevin F. Staveley-O’Carroll
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO 65212, USA; (Y.M.); (K.F.S.-O.); (J.B.M.); (G.L.)
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO 65201, USA
- Siteman Cancer Center, St. Louis, MO 63110, USA; (W.C.W.); (A.A.C.); (Y.H.)
| | - Jonathan B. Mitchem
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO 65212, USA; (Y.M.); (K.F.S.-O.); (J.B.M.); (G.L.)
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO 65201, USA
- Siteman Cancer Center, St. Louis, MO 63110, USA; (W.C.W.); (A.A.C.); (Y.H.)
| | - Wesley C. Warren
- Siteman Cancer Center, St. Louis, MO 63110, USA; (W.C.W.); (A.A.C.); (Y.H.)
- Department of Animal Sciences and Surgery, Informatics and Data Sciences Institute, Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA
| | - Aadel A. Chaudhuri
- Siteman Cancer Center, St. Louis, MO 63110, USA; (W.C.W.); (A.A.C.); (Y.H.)
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Yi Huang
- Siteman Cancer Center, St. Louis, MO 63110, USA; (W.C.W.); (A.A.C.); (Y.H.)
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Guangfu Li
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO 65212, USA; (Y.M.); (K.F.S.-O.); (J.B.M.); (G.L.)
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO 65201, USA
- Siteman Cancer Center, St. Louis, MO 63110, USA; (W.C.W.); (A.A.C.); (Y.H.)
| | - Peter J. Tonellato
- Center for Biomedical Informatics, Department of Health Management and Informatics, School of Medicine, University of Missouri, Columbia, MO 65212, USA; (M.B.); (A.M.); (P.J.T.)
| | - Jussuf T. Kaifi
- Center for Biomedical Informatics, Department of Health Management and Informatics, School of Medicine, University of Missouri, Columbia, MO 65212, USA; (M.B.); (A.M.); (P.J.T.)
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO 65212, USA; (Y.M.); (K.F.S.-O.); (J.B.M.); (G.L.)
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO 65201, USA
- Siteman Cancer Center, St. Louis, MO 63110, USA; (W.C.W.); (A.A.C.); (Y.H.)
- Correspondence:
| |
Collapse
|
5
|
Zhang J, Zhang M, Liang Y, Liu M, Huang Z. Downregulation of Smad4 expression confers chemoresistance against imatinib mesylate to chronic myeloid leukemia K562 cells. Hematology 2021; 27:43-52. [PMID: 34957936 DOI: 10.1080/16078454.2021.2010331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
Objective: Imatinib mesylate (IM), a tyrosine kinase inhibitor, exhibits clinically prominent effects against chronic myeloid leukemia (CML); however, a few patients have shown resistance to IM treatment, resulting in disease progression. Smad4 is a tumor inhibitor that transduces TGF-β signaling and modulates genomic stability. Previous studies have indicated that decreased Smad4 expression played a bidirectional role in chemosensitivity in many types of cancers. Therefore, this study aims to evaluate the association between IM sensitivity and decreased Smad4 expression in human CML K562 cells.Methods: Bone marrow (BM) samples were acquired from the patients prior to treatment. qRT-PCR, Western Blotting (WB), colony formation assay (CFA), and apoptosis assay were used to detect relevant indices.Results: Smad4 expression was downregulated in the bone marrow and plasma of patients with multidrug-resistant CML as well as IM-resistant K562 (K562R) cells compared with samples collected from CML patients and K562 cells. Smad4 overexpression inhibited IM-treated K562R cell proliferation and augmented apoptosis, whereas Smad4 silencing promoted viability and inhibited apoptosis in IM-treated K562 cells. In addition, Smad4 expression was inversely correlated with laminin subunit gamma 1 (LAMC1) expression. The upregulation or downregulation of LAMC1 expression partially abolished the effect of Smad4 overexpression or silencing on the IM resistance of CML cells.Conclusion: The downregulation of Smad4 expression might induce drug resistance in CML cells and displayed a possible mechanism through which Smad4 modulates CML cell survival and apoptosis upon IM treatment.
Collapse
Affiliation(s)
- Jiangzhao Zhang
- Department of Hematology, Jingzhou Central Hospital, Institute of Hematology, Yangtze University, Jingzhou, People's Republic of China
| | - Min Zhang
- Department of Nephrology, Jingzhou Central Hospital, Jingzhou, People's Republic of China
| | - Yan Liang
- Department of Hematology, Jingzhou Central Hospital, Jingzhou, People's Republic of China
| | - Min Liu
- Department of Hematology, Jingzhou Central Hospital, Jingzhou, People's Republic of China
| | - Zhiping Huang
- Department of Hematology, Jingzhou Central Hospital, Institute of Hematology, Yangtze University, Jingzhou, People's Republic of China
| |
Collapse
|
6
|
Preoperative clinical and tumor genomic features associated with pathologic lymph node metastasis in clinical stage I and II lung adenocarcinoma. NPJ Precis Oncol 2021; 5:70. [PMID: 34290393 PMCID: PMC8295366 DOI: 10.1038/s41698-021-00210-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 06/29/2021] [Indexed: 11/08/2022] Open
Abstract
While next-generation sequencing (NGS) is used to guide therapy in patients with metastatic lung adenocarcinoma (LUAD), use of NGS to determine pathologic LN metastasis prior to surgery has not been assessed. To bridge this knowledge gap, we performed NGS using MSK-IMPACT in 426 treatment-naive patients with clinical N2-negative LUAD. A multivariable logistic regression model that considered preoperative clinical and genomic variables was constructed. Most patients had cN0 disease (85%) with pN0, pN1, and pN2 rates of 80%, 11%, and 9%, respectively. Genes altered at higher rates in pN-positive than in pN-negative tumors were STK11 (p = 0.024), SMARCA4 (p = 0.006), and SMAD4 (p = 0.011). Fraction of genome altered (p = 0.037), copy number amplifications (p = 0.001), and whole-genome doubling (p = 0.028) were higher in pN-positive tumors. Multivariable analysis revealed solid tumor morphology, tumor SUVmax, clinical stage, SMARCA4 and SMAD4 alterations were independently associated with pathologic LN metastasis. Incorporation of clinical and tumor genomic features can identify patients at risk of pathologic LN metastasis; this may guide therapy decisions before surgical resection.
Collapse
|
7
|
Guo X, Li M, Wang X, Pan Y, Li J. Correlation between loss of Smad4 and clinical parameters of non-small cell lung cancer: an observational cohort study. BMC Pulm Med 2021; 21:111. [PMID: 33794845 PMCID: PMC8017835 DOI: 10.1186/s12890-021-01480-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 03/24/2021] [Indexed: 02/08/2023] Open
Abstract
Background SMAD4 has been found to be inactivated to varying degrees in many types of cancer; the purpose of this study was to investigate the correlation between SMAD4 expression in non-small cell lung cancer (NSCLC) and clinical pathological parameters. Methods The serum concentration of SMAD4 was measured by enzyme-linked immunosorbent assay and its histological expression was quantified by immunohistochemistry. Results The serum concentration of Smad4 in patients with NSCLC was lower than that in benign lung disease patients and healthy individuals (P < 0.001) and its concentration was related to the histological classification, pathological differentiation, lymphatic metastasis and clinical stage of NSCLC. The sensitivity and specificity of serum Smad4 were 91.56% and 61.56% for screening NSCLC from healthy individuals and 84.55% and 60.36% for screening NSCLC from patients with benign lung disease. Logistic regression analysis showed that the degree of cell differentiation (P < 0.001), lymph node metastasis (P < 0.001) and clinical stage of NSCLC (P = 0.007) affected the expression of Smad4, and had a strong correlation with the expression of Smad4. The expression of Smad4 in NSCLC tissues was lower than that in normal lung tissues (P = 0.009) and its expression was related to the degree of tissue differentiation, lymph node metastasis and clinical stage (P < 0.05). Conclusions The downregulation or deletion of Smad4 is related to the malignant biological behavior of NSCLC and serum Smad4 could be considered as a potential molecular indicator for diagnosis and evaluation of NSCLC.
Collapse
Affiliation(s)
- Xiangjun Guo
- Department of Respiratory and Critical Care Medicine, The First People's Hospital of Lianyungang City, Lianyungang, Jiangsu, China
| | - Mengmeng Li
- Department of Respiratory and Critical Care Medicine, The First People's Hospital of Lianyungang City, Lianyungang, Jiangsu, China
| | - Xin Wang
- Department of Respiratory and Critical Care Medicine, The First People's Hospital of Lianyungang City, Lianyungang, Jiangsu, China
| | - Yun Pan
- Department of Respiratory and Critical Care Medicine, The First People's Hospital of Lianyungang City, Lianyungang, Jiangsu, China
| | - Jiashu Li
- Department of Respiratory and Critical Care Medicine, The First People's Hospital of Lianyungang City, Lianyungang, Jiangsu, China.
| |
Collapse
|
8
|
Detection of Genetic Mutations by Next-Generation Sequencing for Predicting Prognosis of Extensive-Stage Small-Cell Lung Cancer. JOURNAL OF ONCOLOGY 2021; 2020:8811487. [PMID: 33643409 PMCID: PMC7901041 DOI: 10.1155/2020/8811487] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 11/02/2020] [Accepted: 11/06/2020] [Indexed: 01/22/2023]
Abstract
Some studies have revealed that specific genetic mutations could be associated with chemotherapy response or even survival in small-cell lung cancer (SCLC). Our retrospective study aimed to identify the correlation between genetic mutations and progression-free survival (PFS) in extensive-stage SCLC after first-line chemotherapy. A total of 75 patients with extensive-stage SCLC confirmed by histopathology from February 2018 to February 2019 were retrospectively analyzed. The biopsy specimens of all patients were analyzed by Next-Generation Sequencing (NGS). All patients received first-line chemotherapy and follow-up at Shanghai Chest Hospital. Eleven genes were mutated in, at least, 10% of the 75 patients, including TP53 (96%), RB1 (77%), SMAD4 (32%), NOTCH1 (21%), PTEN (16%), FGFR1 (16%), KDR (15%), PIK3CA (15%), ROS1 (15%), BRCA2 (13%), and ERBB4 (10%). The median number of mutated genes among all patients was 5. Patients with more than 5 mutated genes (PFS = 6.7 months, P=0.004), mutant TP53 (PFS = 5.0 months, P=0.011), and mutant BRCA2 (PFS = 6.7 months, P=0.046) had better PFS after first-line chemotherapy than other patients. Multivariate Cox regression analysis showed that patients who achieved a PR (HR 3.729, 95% CI 2.038–6.822), had more than 5 mutated genes (HR 1.929, 95% CI 1.096–3.396), had BRCA2 mutations (HR 4.581, 95% CI 1.721–12.195), and had no liver metastasis (HR 0.415, 95% CI 0.181–0.951) showed improvements in PFS after first-line chemotherapy. In conclusion, the number of mutated genes and BRCA2 mutation status in extensive-stage SCLC were significantly related to PFS after first-line chemotherapy.
Collapse
|
9
|
Lu C, Ning G, Si P, Zhang C, Liu W, Ge W, Cui K, Zhang R, Ge S. E3 ubiquitin ligase HECW1 promotes the metastasis of non-small cell lung cancer cells through mediating the ubiquitination of Smad4. Biochem Cell Biol 2021; 99:675-681. [PMID: 33529121 DOI: 10.1139/bcb-2020-0505] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Lung cancer is the leading cause of cancer-related death globally. Ubiquitin modification plays a crucial role in the regulation of gene expression, and is closely associated with cancer pathogenesis. The aim of our study was to clarify the role and mechanisms of action for HECT, C2 and WW domain containing E3 ubiquitin protein ligase 1 (HECW1) in non-small cell lung cancer (NSCLC). Herein, we demonstrate that the expression of HECW1 was significantly increased in NSCLC cell lines and tissues. Upregulation of HECW1 markedly enhanced the proliferation of NSCLC cells, whereas downregulation of HECW1 significantly inhibited proliferation. Moreover, the expression levels of HECW1 positively correlated with the migration and invasiveness of NSCLC cells. Upregulation or downregulation of HECW1 only affected the protein expression levels of SMAD family member 4 (Smad4), but had no effect on the mRNA expression levels. Furthermore, after treatment with MG-132, the relative protein level of Smad4 significantly increased in NSCLC cells. HECW1 promoted the proliferation, migration, and invasiveness of NSCLC cells by inducing the ubiquitination and degradation of Smad4, thus our data provide a novel target for NSCLC treatment.
Collapse
Affiliation(s)
- Chen Lu
- Department of Thoracic Surgery, The First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Shushan District, Hefei 230022, Anhui, China
| | - Guangyao Ning
- Department of Thoracic Surgery, The First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Shushan District, Hefei 230022, Anhui, China
| | - Panpan Si
- Department of Thoracic Surgery, The First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Shushan District, Hefei 230022, Anhui, China
| | - Chunsheng Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Shushan District, Hefei 230022, Anhui, China
| | - Wenjian Liu
- Department of Thoracic Surgery, The First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Shushan District, Hefei 230022, Anhui, China
| | - Wei Ge
- Department of Thoracic Surgery, The First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Shushan District, Hefei 230022, Anhui, China
| | - Kai Cui
- Department of Thoracic Surgery, The First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Shushan District, Hefei 230022, Anhui, China
| | - Renquan Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Shushan District, Hefei 230022, Anhui, China
| | - Shenglin Ge
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Shushan District, Hefei 230022, Anhui, China
| |
Collapse
|
10
|
张 昆, 戴 朝, 刘 思, 李 丹, 杨 达, 崔 赛. [Clinical Value of Cerebrospinal Fluid ctDNA in Patients
with Non-small Cell Lung Cancer Meningeal Metastasis]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2020; 23:1039-1048. [PMID: 33357310 PMCID: PMC7786234 DOI: 10.3779/j.issn.1009-3419.2020.102.42] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 09/17/2020] [Accepted: 09/18/2020] [Indexed: 11/25/2022]
Abstract
BACKGROUND The mortality rate of lung cancer meningeal metastasis is extremely high. Circulating tumor DNA (ctDNA) has been confirmed to be contain the genomic alterations present in tumors and has been used to monitor tumor progression and response to treatments. Due to the presence of blood-brain barrier and other factors, peripheral blood ctDNA cannot reflect the information of brain lesions for patients with meningeal metastases. However, cerebrospinal fluid ctDNA as a test sample can better reflect the genetic status of intracranial tumors and guide clinical targeted treatment of intracranial lesions. This study explored the feasibility of cerebrospinal fluid ctNDA for evaluating non-small cell lung cancer (NSCLC) meningeal metastasis and the potential clinical value of cerebrospinal fluid ctDNA detection in NSCLC meningeal metastasis. METHODS A total of 21 patients with NSCLC meningeal metastasis were included. Tumor genomic variation was performed on the cerebrospinal fluid and peripheral blood samples of patients by second-generation gene sequencing technology. The situation was examined, and pathological evaluation of cerebrospinal fluid cytology and head magnetic resonance imaging (MRI) enhanced examination were performed. RESULTS ctDNA was detected in the cerebrospinal fluid of 21 patients. The sensitivity of cerebrospinal fluid ctDNA detection was superior to cytology in the diagnosis of meningeal metastasis (P<0.001). The detection rate and gene mutation abundance of cerebrospinal fluid were higher than plasma (P<0.001). Cerebro-spinal fluid had a unique genetic profile. In 6 patients with dynamic detection, changes of ctDNA allele fraction occurred at the same time or earlier than clinical disease changes, which could timely monitor drug resistance mechanism and relapse trend. CONCLUSIONS The detection rate of ctDNA in cerebrospinal fluid is higher than that in cytology and imaging. The detection of ctDNA in cerebrospinal fluid can reveal the specific mutation map of meningeal metastasis lesions. The dynamic monitoring of ctDNA in cerebrospinal fluid has hint significance for clinical response of lung cancer patients.
Collapse
Affiliation(s)
- 昆煜 张
- />116021 大连,大连医科大学附属第二医院The Second Affiliated Hospital of Dalian Medical University, Dalian 116021, China
| | - 朝霞 戴
- />116021 大连,大连医科大学附属第二医院The Second Affiliated Hospital of Dalian Medical University, Dalian 116021, China
| | - 思雅 刘
- />116021 大连,大连医科大学附属第二医院The Second Affiliated Hospital of Dalian Medical University, Dalian 116021, China
| | - 丹 李
- />116021 大连,大连医科大学附属第二医院The Second Affiliated Hospital of Dalian Medical University, Dalian 116021, China
| | - 达夫 杨
- />116021 大连,大连医科大学附属第二医院The Second Affiliated Hospital of Dalian Medical University, Dalian 116021, China
| | - 赛琼 崔
- />116021 大连,大连医科大学附属第二医院The Second Affiliated Hospital of Dalian Medical University, Dalian 116021, China
| |
Collapse
|
11
|
Pan S, Zhou G, Hu W, Pei H. SMAD-6, -7 and -9 are potential molecular biomarkers for the prognosis in human lung cancer. Oncol Lett 2020; 20:2633-2644. [PMID: 32782581 PMCID: PMC7401007 DOI: 10.3892/ol.2020.11851] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 05/13/2020] [Indexed: 12/24/2022] Open
Abstract
SMADs, a family of proteins that function as signal transducers and transcriptional regulators to regulate various signaling pathways, including the transforming growth factor-β signaling pathway, are similar to the mothers against decapentaplegic family of genes and the sma gene family in Caenorhabditis elegans. SMADs generate context-dependent modulation by interacting with various sequence-specific transcription factors, such as E2F4/5, c-Fos, GATA3, YY1 and SRF, which have been found to serve a key role in lung carcinoma oncogenesis and progression. However, the prognostic values of the eight SMADs in lung cancer have not been fully understood. In the present study, the expression levels and survival data of SMADs in patients with lung carcinoma from the Oncomine, Gene Expression Profiling Interactive Analysis, Kaplan-Meier plotter and cBioPortal databases were downloaded and analyzed. It was found that the mRNA expression levels of SMAD-6, -7 and -9 were decreased in lung adenocarcinoma and squamous cell carcinoma compared with that in adjacent normal tissues, while there was no significant difference in SMADs 1-5. Survival analysis revealed that not only were low transcriptional levels of SMAD-6, -7 and -9 associated with low overall survival but they also had prognostic role for progression-free survival and post-progression survival (P<0.05) in patients with lung carcinoma. In conclusion, the present study demonstrated that SMAD-6, -7 and -9 are potential biomarkers for the prognosis of patients with lung carcinoma.
Collapse
Affiliation(s)
- Shuxian Pan
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, P.R. China
- Department of Radiation Oncology, Yancheng Third People's Hospital, Yancheng, Jiangsu 224000, P.R. China
| | - Guangming Zhou
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, P.R. China
| | - Wentao Hu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, P.R. China
| | - Hailong Pei
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, P.R. China
| |
Collapse
|
12
|
Nolan K, Verzosa G, Cleaver T, Tippimanchai D, DePledge LN, Wang XJ, Young C, Le A, Doebele R, Li H, Malkoski SP. Development of syngeneic murine cell lines for use in immunocompetent orthotopic lung cancer models. Cancer Cell Int 2020; 20:417. [PMID: 32874131 PMCID: PMC7455907 DOI: 10.1186/s12935-020-01503-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 08/18/2020] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Immunocompetent animal models are required to study tumor-host interactions, immunotherapy, and immunotherapeutic combinations, however the currently available immunocompetent lung cancer models have substantial limitations. While orthotopic models potentially help fill this gap, the utility of these models has been limited by the very small number of murine lung cancer cell lines capable of forming orthotopic tumors in immunocompetent C57BL/6 hosts. METHODS Primary lung tumors with specific genetic alterations were created in C57BL/6 background mice. These tumors were then passaged through other animals to increase tumorigenicity and select for the ability to grow in a non-self animal. Once tumors demonstrated growth in a non-self host, cell lines were established. Successful cell lines were evaluated for the ability to produce orthotopic lung tumors in immunocompetent hosts. RESULTS We produced six murine lung cancer lines capable of orthotopic lung tumor formation in immunocompetent C57BL/6 animals. These lines demonstrate the expected genetic alterations based on their primary tumor genetics. CONCLUSIONS These novel cell lines will be useful for evaluating tumor-host interactions, the impact of specific oncogenic alterations on the tumor microenvironment, and immunotherapeutic approaches. This method of generating murine lines capable of orthotopic growth can likely be applied to other tumors and will broaden the applicability of pre-clinical testing of immunotherapeutic treatment regimens.
Collapse
Affiliation(s)
- Kyle Nolan
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Denver Anschutz Medical Campus, 12700 E. 19th Avenue, RC2, Room #9112, Mail stop C272, Aurora, CO 80045 USA
| | - Gregory Verzosa
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Denver Anschutz Medical Campus, 12700 E. 19th Avenue, RC2, Room #9112, Mail stop C272, Aurora, CO 80045 USA
| | - Tim Cleaver
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Denver Anschutz Medical Campus, 12700 E. 19th Avenue, RC2, Room #9112, Mail stop C272, Aurora, CO 80045 USA
| | - Darinee Tippimanchai
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Denver Anschutz Medical Campus, 12700 E. 19th Avenue, RC2, Room #9112, Mail stop C272, Aurora, CO 80045 USA
| | - Lisa N. DePledge
- Department of Pathology, University of Colorado Denver Anschutz Medical Campus, Aurora, CO USA
| | - Xiao-Jing Wang
- Department of Pathology, University of Colorado Denver Anschutz Medical Campus, Aurora, CO USA
| | - Christian Young
- Department of Pathology, University of Colorado Denver Anschutz Medical Campus, Aurora, CO USA
| | - Anh Le
- Division of Medical Oncology, University of Colorado Denver Anschutz Medical Campus, Aurora, CO USA
| | - Robert Doebele
- Division of Medical Oncology, University of Colorado Denver Anschutz Medical Campus, Aurora, CO USA
| | - Howard Li
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Denver Anschutz Medical Campus, 12700 E. 19th Avenue, RC2, Room #9112, Mail stop C272, Aurora, CO 80045 USA
- Division of Pulmonary Disease and Critical Care Medicine, Virginia Commonwealth University, Richmond, VA USA
| | - Stephen P. Malkoski
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Denver Anschutz Medical Campus, 12700 E. 19th Avenue, RC2, Room #9112, Mail stop C272, Aurora, CO 80045 USA
- Sound Critical Care, Sacred Heart Medical Center, Spokane, WA USA
| |
Collapse
|
13
|
Belluti S, Rigillo G, Imbriano C. Transcription Factors in Cancer: When Alternative Splicing Determines Opposite Cell Fates. Cells 2020; 9:E760. [PMID: 32244895 PMCID: PMC7140685 DOI: 10.3390/cells9030760] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 03/05/2020] [Accepted: 03/17/2020] [Indexed: 02/08/2023] Open
Abstract
Alternative splicing (AS) is a finely regulated mechanism for transcriptome and proteome diversification in eukaryotic cells. Correct balance between AS isoforms takes part in molecular mechanisms that properly define spatiotemporal and tissue specific transcriptional programs in physiological conditions. However, several diseases are associated to or even caused by AS alterations. In particular, multiple AS changes occur in cancer cells and sustain the oncogenic transcriptional program. Transcription factors (TFs) represent a key class of proteins that control gene expression by direct binding to DNA regulatory elements. AS events can generate cancer-associated TF isoforms with altered activity, leading to sustained proliferative signaling, differentiation block and apoptosis resistance, all well-known hallmarks of cancer. In this review, we focus on how AS can produce TFs isoforms with opposite transcriptional activities or antagonistic functions that severely impact on cancer biology. This summary points the attention to the relevance of the analysis of TFs splice variants in cancer, which can allow patients stratification despite the presence of interindividual genetic heterogeneity. Recurrent TFs variants that give advantage to specific cancer types not only open the opportunity to use AS transcripts as clinical biomarkers but also guide the development of new anti-cancer strategies in personalized medicine.
Collapse
Affiliation(s)
| | | | - Carol Imbriano
- Department of Life Sciences, University of Modena and Reggio Emilia, via Campi 213/D, 41125 Modena, Italy; (S.B.); (G.R.)
| |
Collapse
|
14
|
Zheng H, Jarvis IWH, Bottai M, Dreij K, Stenius U. TGF beta promotes repair of bulky DNA damage through increased ERCC1/XPF and ERCC1/XPA interaction. Carcinogenesis 2020; 40:580-591. [PMID: 30418489 DOI: 10.1093/carcin/bgy156] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 10/30/2018] [Accepted: 11/07/2018] [Indexed: 12/15/2022] Open
Abstract
Transforming growth factor beta (TGFβ) is multifunctional cytokine that is involved in the coordination and regulation of many cellular homeostatic processes. Compromised TGFβ activity has been attributed to promotion of human cancers. Recent studies have identified a role for TGFβ in response to radiation-induced DNA damage, suggesting a link between TGFβ and the DNA damage response with implications for cancer development. In this study, the effects of TGFβ on promoting the repair of bulky DNA damage, through modulation of nucleotide excision repair (NER), were investigated. We show that treatment of cells with exogenous TGFβ leads to enhanced repair of DNA damage formed by polycyclic aromatic hydrocarbons and ultraviolet-C radiation; similarly, cells with constitutively activated endogenous TGFβ signaling show comparable responses. This effect of TGFβ is independent of the cell cycle. The response to TGFβ is decreased in cells that have compromised TGFβ signaling through RNA interference of Smad4 and is decreased in NER-deficient cells and cells with compromised NER through RNA interference of excision repair cross-complementing group 1 (ERCC1). Increased interaction and nuclear localization of ERCC1/xeroderma pigmentosum (XP) F and ERCC1/XPA proteins is observed after TGFβ treatment. Our study represents the first experimental evidence of a role for TGFβ in the repair of bulky DNA damage resulting from promotion of the interaction and localization of repair protein complexes involved in the incision step of NER.
Collapse
Affiliation(s)
- Huiyuan Zheng
- Unit of Biochemical Toxicology, Karolinska Institutet, Stockholm, Sweden
| | - Ian W H Jarvis
- Unit of Biochemical Toxicology, Karolinska Institutet, Stockholm, Sweden
| | - Matteo Bottai
- Unit of Biostatistics, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Kristian Dreij
- Unit of Biochemical Toxicology, Karolinska Institutet, Stockholm, Sweden
| | - Ulla Stenius
- Unit of Biochemical Toxicology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
15
|
Wong CK, Lambert AW, Ozturk S, Papageorgis P, Lopez D, Shen N, Sen Z, Abdolmaleky HM, Győrffy B, Feng H, Thiagalingam S. Targeting RICTOR Sensitizes SMAD4-Negative Colon Cancer to Irinotecan. Mol Cancer Res 2020; 18:414-423. [PMID: 31932471 DOI: 10.1158/1541-7786.mcr-19-0525] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 11/04/2019] [Accepted: 12/16/2019] [Indexed: 12/15/2022]
Abstract
Deciphering molecular targets to enhance sensitivity to chemotherapy is becoming a priority for effectively treating cancers. Loss of function mutations of SMAD4 in colon cancer are associated with metastatic progression and resistance to 5-fluorouracil (5-FU), the most extensively used drug of almost all chemotherapy combinations used in the treatment of metastatic colon cancer. Here, we report that SMAD4 deficiency also confers resistance to irinotecan, another common chemotherapeutic frequently used alone or in combination with 5-FU against colon cancer. Mechanistically, we find that SMAD4 interacts with and inhibits RICTOR, a component of the mTORC2 complex, resulting in suppression of downstream effector phosphorylation of AKT at Serine 473. In silico meta-analysis of publicly available gene expression datasets derived from tumors indicates that lower levels of SMAD4 or higher levels of RICTOR/AKT, irrespective of the SMAD4 status, correlate with poor survival, suggesting them as strong prognostic biomarkers and targets for therapeutic intervention. Moreover, we find that overexpression of SMAD4 or depletion of RICTOR suppresses AKT signaling and increases sensitivity to irinotecan in SMAD4-deficient colon cancer cells. Consistent with these observations, pharmacologic inhibition of AKT sensitizes SMAD4-negative colon cancer cells to irinotecan in vitro and in vivo. Overall, our study suggests that hyperactivation of the mTORC2 pathway is a therapeutic vulnerability that could be exploited to sensitize SMAD4-negative colon cancer to irinotecan. IMPLICATIONS: Hyperactivation of the mTORC2 pathway in SMAD4-negative colon cancer provides a mechanistic rationale for targeted inhibition of mTORC2 or AKT as a distinctive combinatorial therapeutic opportunity with chemotherapy for colon cancer.
Collapse
Affiliation(s)
- Chen Khuan Wong
- Graduate Program in Genetics and Genomics, Boston University School of Medicine, Boston, Massachusetts.,Biomedical Genetics Section, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
| | - Arthur W Lambert
- Graduate Program in Molecular and Translational Medicine, Boston University School of Medicine, Boston, Massachusetts
| | - Sait Ozturk
- Graduate Program in Molecular and Translational Medicine, Boston University School of Medicine, Boston, Massachusetts
| | - Panagiotis Papageorgis
- Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia, Cyprus
| | - Delia Lopez
- Graduate Program in Molecular and Translational Medicine, Boston University School of Medicine, Boston, Massachusetts
| | - Ning Shen
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, Massachusetts
| | - Zaina Sen
- Biomedical Genetics Section, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
| | - Hamid M Abdolmaleky
- Biomedical Genetics Section, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
| | - Balázs Győrffy
- MTA TTK Lendület Cancer Biomarker Research Group, Institute of Enzymology, Budapest, Hungary.,Semmelweis University 2nd Department of Pediatrics, Budapest, Hungary
| | - Hui Feng
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, Massachusetts
| | - Sam Thiagalingam
- Graduate Program in Genetics and Genomics, Boston University School of Medicine, Boston, Massachusetts. .,Biomedical Genetics Section, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts.,Graduate Program in Molecular and Translational Medicine, Boston University School of Medicine, Boston, Massachusetts.,Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, Massachusetts.,Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts
| |
Collapse
|
16
|
Lin LH, Chang KW, Cheng HW, Liu CJ. SMAD4 Somatic Mutations in Head and Neck Carcinoma Are Associated With Tumor Progression. Front Oncol 2019; 9:1379. [PMID: 31867281 PMCID: PMC6909744 DOI: 10.3389/fonc.2019.01379] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 11/22/2019] [Indexed: 12/19/2022] Open
Abstract
As the incidence and the mortality rate of head and neck squamous cell carcinoma (HNSCC) is increasing worldwide, gaining knowledge about the genomic changes which happen in the carcinogenesis of HNSCC is essential for the diagnosis and therapy of the disease. SMAD4 (DPC4) is a tumor suppressor gene. It is located at chromosome 18q21.1 and a member of the SMAD family. Which mediates the TGF-β signaling pathway, thereby controlling the growth of epithelial cells. In the study presented here, we analyzed tumor samples by multiplex PCR-based next-generation sequencing (NGS) and found deleterious mutations of SMAD4 in 4.1% of the tumors. Knock-down experiments of endogenous and exogenous SMAD4 expression demonstrated that SMAD4 is involved in the migration and invasion of HNSCC cells. Functional analysis of a missense mutation in the MH1 domain of SMAD4 may be responsible for the loss of function in suppressing tumor progression. Missense SMAD4 mutations, therefore, could be useful prognostic determinants for patients affected by HNSCCs. This report is the first study where NGS analysis based on multiplex-PCR is used to demonstrate the imminent occurrence of missense SMAD4 mutations in HNSCC cells. The gene analysis that we performed may support the identification of SMAD4 mutations as a diagnostic marker or even as a potential therapeutic target in head and neck cancer. Moreover, the analytic strategy proposed for the detection of mutations in the SMAD4 gene may be validated as a platform to assist mutation screening.
Collapse
Affiliation(s)
- Li-Han Lin
- Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan
| | - Kuo-Wei Chang
- Department of Stomatology, Taipei Veterans General Hospital, Taipei, Taiwan.,School of Dentistry, Institute of Oral Biology, National Yang-Ming University, Taipei, Taiwan
| | - Hui-Wen Cheng
- Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan
| | - Chung-Ji Liu
- School of Dentistry, Institute of Oral Biology, National Yang-Ming University, Taipei, Taiwan.,Department of Oral and Maxillofacial Surgery, Taipei MacKay Memorial Hospital, Taipei, Taiwan
| |
Collapse
|
17
|
Immunological consequences of chemotherapy: Single drugs, combination therapies and nanoparticle-based treatments. J Control Release 2019; 305:130-154. [DOI: 10.1016/j.jconrel.2019.04.020] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 04/09/2019] [Accepted: 04/14/2019] [Indexed: 02/07/2023]
|
18
|
Wang Y, Tan X, Tang Y, Zhang C, Xu J, Zhou J, Cheng X, Hou N, Liu W, Yang G, Teng Y, Yang X. Dysregulated Tgfbr2/ERK-Smad4/SOX2 Signaling Promotes Lung Squamous Cell Carcinoma Formation. Cancer Res 2019; 79:4466-4479. [PMID: 31209059 DOI: 10.1158/0008-5472.can-19-0161] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 05/05/2019] [Accepted: 06/10/2019] [Indexed: 11/16/2022]
Abstract
Lung squamous cell carcinoma (SCC) is a common type of lung cancer. There is limited information on the genes and pathways that initiate lung SCC. Here, we report that loss of TGFβ type II receptor (Tgfbr2), frequently deleted in human lung cancer, led to predominant lung SCC development in KrasG12D mice with a short latency, high penetrance, and extensive metastases. Tgfbr2-loss-driven lung SCCs resembled the salient features of human lung SCC, including histopathology, inflammatory microenvironment, and biomarker expression. Surprisingly, loss of Smad4, a key mediator of Tgfbr2, failed to drive lung SCC; instead, low levels of phosphorylated ERK1/2, a Smad-independent downstream effector of Tgfbr2, were tightly associated with lung SCC in both mouse and human. Mechanistically, inhibition of phosphorylated ERK1/2 significantly upregulated the expression of SOX2, an oncogenic driver of lung SCC, and cooperated with SMAD4 repression to elevate SOX2. Inhibition of ERK1/2 in Smad4fl/fl ;KrasG12D mice led to extensive lung SCC formation that resembled the SCC phenotype of Tgfbr2-deficient mice. Overall, we reveal a key role of ERK1/2 in suppressing SCC formation and demonstrate that dysregulated Tgfbr2/ERK-Smad4/SOX2 signaling drives lung SCC formation. We also present a mouse model of metastatic lung SCC that may be valuable for screening therapeutic targets. SIGNIFICANCE: This study sheds new light on the mechanisms underlying lung SCC formation driven by mutated Kras.
Collapse
Affiliation(s)
- Yanxiao Wang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Beijing Institute of Lifeomics, Beijing, China
| | - Xiaohong Tan
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Beijing Institute of Lifeomics, Beijing, China
| | - Yuling Tang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Beijing Institute of Lifeomics, Beijing, China
| | - Chong Zhang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Beijing Institute of Lifeomics, Beijing, China
| | - Jiaqian Xu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Beijing Institute of Lifeomics, Beijing, China
| | - Jian Zhou
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Beijing Institute of Lifeomics, Beijing, China
| | - Xuan Cheng
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Beijing Institute of Lifeomics, Beijing, China
| | - Ning Hou
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Beijing Institute of Lifeomics, Beijing, China
| | - Wenjia Liu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Beijing Institute of Lifeomics, Beijing, China
| | - Guan Yang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Beijing Institute of Lifeomics, Beijing, China
| | - Yan Teng
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Beijing Institute of Lifeomics, Beijing, China.
| | - Xiao Yang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Beijing Institute of Lifeomics, Beijing, China.
| |
Collapse
|
19
|
Hernandez AL, Young CD, Wang JH, Wang XJ. Lessons learned from SMAD4 loss in squamous cell carcinomas. Mol Carcinog 2019; 58:1648-1655. [PMID: 31140647 DOI: 10.1002/mc.23049] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 04/30/2019] [Accepted: 05/10/2019] [Indexed: 12/13/2022]
Abstract
SMAD4 is a potent tumor suppressor and a central mediator of the TGFß signaling pathway. SMAD4 genetic loss is frequent in squamous cell carcinomas (SCCs). Reports of SMAD4 expression in SCCs vary significantly possibly due to inter-tumor heterogeneity or technical reasons. SMAD4 loss is an initiation event for SCCs. In tumor epithelial cells, SMAD4 loss causes increased proliferation, decreased apoptosis, and "Brca-like" genomic instability associated with DNA repair defects. SMAD4 loss also plays a role in the expansion of cancer stem cells. Epithelial SMAD4 loss causes overexpression of TGFß that is released into the tumor microenvironment and contributes to SCC progression through proinflammatory and immune evasive mechanisms. SMAD4 loss, while not a direct therapeutic target, is associated with multiple targetable pathways that require further therapeutic studies. Altogether, SMAD4 loss is a potential biomarker in SCCs that should be further studied for its values in prognostic and therapeutic predictions. Such information will potentially guide future biomarker-driven clinical trial designs and improve SCC patient outcomes.
Collapse
Affiliation(s)
- Ariel L Hernandez
- Department of Pathology, School of Medicine, University of Colorado, Aurora, Colorado
| | - Christian D Young
- Department of Pathology, School of Medicine, University of Colorado, Aurora, Colorado
| | - Jing H Wang
- Department of Immunology and Microbiology, University of Colorado, Aurora, Colorado.,Department of Biomedical Research, National Jewish Health, Denver, Colorado
| | - Xiao-Jing Wang
- Department of Pathology, School of Medicine, University of Colorado, Aurora, Colorado.,Research Service, Veterans Affairs Medical Center, VA Eastern Colorado Health Care System, Aurora, Colorado
| |
Collapse
|
20
|
MiR-144 inhibits growth and metastasis in colon cancer by down-regulating SMAD4. Biosci Rep 2019; 39:BSR20181895. [PMID: 30745456 PMCID: PMC6395301 DOI: 10.1042/bsr20181895] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 01/10/2019] [Accepted: 01/30/2019] [Indexed: 12/12/2022] Open
Abstract
MicroRNAs (MiRs) are thought to display regulator action in tumor suppression and oncogenesis. miR-144 plays an important role in the development of various cancers, such as colorectal cancer, breast cancer, and lung cancer, by targetting different molecules potentially involved in many signaling pathways. SMAD4 is a common signaling during tumor progression, and it can inhibit cell proliferation and promote cell motility in most epithelial cells. The present study focused on the effect of miR-144 and SMAD4 on colon cancer in order to find the novel gene therapy target for the treatment of colon cancer. Quantitative real-time polymerase chain reaction was used to assess the expression level of miR-144 in colon cancer tissues and SW620 cells. MTT assay, scratch test, and transwell assay were used to evaluate cell proliferation, migration, and invasion, respectively. Moreover, luciferase assays were utilized to identify the predictive effect of miR-144 on SMAD4. Western blotting was performed to determine the relative expression of protein related to SMAD4. We found miR-144 level was significantly lower in colon cancer tissues and SW620 cells. Moreover, SMAD4 level, both in mRNA and protein, was obviously elevated in colon cancer tissues. Further, miR-144 mimics treatment inhibited cells proliferation, invasion, and migration. Fluorescence intensity of miR-144 mimics group in wild type cells was decreased. MiR-144 mimics repressed the SMAD4 expression both in mRNA and protein. These findings about miR-144/SMAD4 pair provide a novel therapeutic method for colon cancer patients.
Collapse
|
21
|
Luo P, Ding Y, Lei X, Wu FX. deepDriver: Predicting Cancer Driver Genes Based on Somatic Mutations Using Deep Convolutional Neural Networks. Front Genet 2019; 10:13. [PMID: 30761181 PMCID: PMC6361806 DOI: 10.3389/fgene.2019.00013] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 01/11/2019] [Indexed: 12/16/2022] Open
Abstract
With the advances in high-throughput technologies, millions of somatic mutations have been reported in the past decade. Identifying driver genes with oncogenic mutations from these data is a critical and challenging problem. Many computational methods have been proposed to predict driver genes. Among them, machine learning-based methods usually train a classifier with representations that concatenate various types of features extracted from different kinds of data. Although successful, simply concatenating different types of features may not be the best way to fuse these data. We notice that a few types of data characterize the similarities of genes, to better integrate them with other data and improve the accuracy of driver gene prediction, in this study, a deep learning-based method (deepDriver) is proposed by performing convolution on mutation-based features of genes and their neighbors in the similarity networks. The method allows the convolutional neural network to learn information within mutation data and similarity networks simultaneously, which enhances the prediction of driver genes. deepDriver achieves AUC scores of 0.984 and 0.976 on breast cancer and colorectal cancer, which are superior to the competing algorithms. Further evaluations of the top 10 predictions also demonstrate that deepDriver is valuable for predicting new driver genes.
Collapse
Affiliation(s)
- Ping Luo
- Division of Biomedical Engineering, University of Saskatchewan, Saskatoon, SK, Canada
| | - Yulian Ding
- Division of Biomedical Engineering, University of Saskatchewan, Saskatoon, SK, Canada
| | - Xiujuan Lei
- School of Computer Science, Shaanxi Normal University, Xian, China
| | - Fang-Xiang Wu
- Division of Biomedical Engineering, University of Saskatchewan, Saskatoon, SK, Canada.,School of Mathematics and Statistics, Hainan Normal University, Haikou, China.,Department of Mechanical Engineering, University of Saskatchewan, Saskatoon, SK, Canada.,Department of Computer Science, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
22
|
Hernandez AL, Wang Y, Somerset HL, Keysar SB, Aisner DL, Marshall C, Bowles DW, Karam SD, Raben D, Jimeno A, Varella-Garcia M, Wang XJ. Inter- and intra-tumor heterogeneity of SMAD4 loss in head and neck squamous cell carcinomas. Mol Carcinog 2019; 58:666-673. [PMID: 30575147 DOI: 10.1002/mc.22958] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Revised: 12/04/2018] [Accepted: 12/15/2018] [Indexed: 12/16/2022]
Abstract
Reports regarding the frequency of SMAD4 loss in human head and neck squamous cell carcinoma (HNSCC) vary significantly. We have shown that SMAD4 deletion contributes to HNSCC initiation and progression. Therefore, accurately detecting genetic SMAD4 loss is critical to determine prognosis and therapeutic interventions in personalized medicine. We developed a SMAD4 fluorescence in situ hybridization (FISH) assay to identify chromosomal SMAD4 loss at the single cell level of primary HNSCC specimens and patient derived xenograft (PDX) tumors derived from HNSCCs. SMAD4 heterozygous loss was detected in 35% of primary HNSCCs and 41.3% of PDX tumors. Additionally, 4.3% of PDX tumors had SMAD4 homozygous loss. These frequencies of SMAD4 loss were similar to those in The Cancer Genome Atlas (TCGA). However, we identified significant heterogeneities of SMAD4 loss (partial or complete) among cells within each tumor. We also found that aneuploidy (monosomy and polysomy) contributed greatly to how to define chromosomal SMAD4 deletion. Furthermore, in cultured PDX tumors, SMAD4 mutant cells outcompeted SMAD4 wildtype cells, resulting in establishing homogenous SMAD4 mutant HNSCC cell lines with partial or complete genomic SMAD4 loss, suggesting a survival advantage of SMAD4 mutant cells. Taken together, our study reveals inter- and intra-tumor heterogeneities of SMAD4 chromosomal loss in HNSCCs. Further, SMAD4 FISH assay provides a platform for future clinical diagnosis of SMAD4 chromosomal loss that potentially serves as a molecular marker for prognosis and therapeutic intervention in cancer patients.
Collapse
Affiliation(s)
- Ariel L Hernandez
- Department of Pathology, School of Medicine, Molecular Biology Program, Medical Scientist Training Program, University of Colorado, Aurora, Colorado
| | - Ying Wang
- University of Colorado Cancer Center, Molecular Pathology/Cytogenetics Shared Resource, University of Colorado, Aurora, Colorado
| | - Hilary L Somerset
- Department of Pathology, School of Medicine, Molecular Biology Program, Medical Scientist Training Program, University of Colorado, Aurora, Colorado
| | - Stephen B Keysar
- Department of Medical Oncology, University of Colorado, Aurora, Colorado
| | - Dara L Aisner
- Department of Pathology, School of Medicine, Molecular Biology Program, Medical Scientist Training Program, University of Colorado, Aurora, Colorado
| | - Carrie Marshall
- Department of Pathology, School of Medicine, Molecular Biology Program, Medical Scientist Training Program, University of Colorado, Aurora, Colorado
| | - Daniel W Bowles
- Veterans Affairs Medical Center, VA Eastern Colorado Health Care System, Aurora, Colorado
| | - Sana D Karam
- Department of Radiation Oncology, University of Colorado, Aurora, Colorado
| | - David Raben
- Department of Radiation Oncology, University of Colorado, Aurora, Colorado
| | - Antonio Jimeno
- Department of Medical Oncology, University of Colorado, Aurora, Colorado
| | - Marileila Varella-Garcia
- University of Colorado Cancer Center, Molecular Pathology/Cytogenetics Shared Resource, University of Colorado, Aurora, Colorado
- Department of Medical Oncology, University of Colorado, Aurora, Colorado
| | - Xiao-Jing Wang
- Department of Pathology, School of Medicine, Molecular Biology Program, Medical Scientist Training Program, University of Colorado, Aurora, Colorado
- Veterans Affairs Medical Center, VA Eastern Colorado Health Care System, Aurora, Colorado
| |
Collapse
|
23
|
Selvaraj G, Kaliamurthi S, Kaushik AC, Khan A, Wei YK, Cho WC, Gu K, Wei DQ. Identification of target gene and prognostic evaluation for lung adenocarcinoma using gene expression meta-analysis, network analysis and neural network algorithms. J Biomed Inform 2018; 86:120-134. [PMID: 30195659 DOI: 10.1016/j.jbi.2018.09.004] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 08/11/2018] [Accepted: 09/05/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND Lung adenocarcinoma (LUAD) is a heterogeneous disease with poor survival in the advanced stage and a high incidence rate in the world. Novel drug targets are urgently required to improve patient treatment. Therefore, we aimed to identify therapeutic targets for LUAD based on protein-protein and protein-drug interaction network analysis with neural network algorithms using mRNA expression profiles. RESULTS A comprehensive meta-analysis of selective non-small cell lung cancer (NSCLC) mRNA expression profile datasets from Gene Expression Omnibus were used to identify potential biomarkers and the molecular mechanisms related to the prognosis of NSCLC patients. Using the Network Analyst tool, based on combined effect size (ES) methods, we recognized 6566 differentially expressed genes (DEGs), which included 3036 downregulated and 3530 upregulated genes linked to NSCLC patient survival. ClueGO, a Cytoscape plugin, was exploited to complete the function and pathway enrichment analysis, which disclosed "regulated exocytosis", "purine nucleotide binding", "pathways in cancer", and "cell cycle" between exceptionally supplemented terms. Enrichr, a web tool examination, demonstrated "early growth response protein 1 (EGR-1)", "hepatocyte nuclear factor 4α (HNF4A)", "mitogen-activated protein kinase 14 (MAP3K14)", and "cyclin-dependent kinase 1 (CDK1)" to be among the most prevalent TFs and kinases associated with NSCLC. Our meta-analysis identified that MAPK1 and aurora kinase (AURKA) are the most obvious class of hub nodes. Furthermore, protein-drug interaction network and neural network algorithms identified candidate drugs such as phosphothreonine and 4-(4-methylpiperazin-1-yl)-n-[5-(2-thienylacetyl)-1,5-dihydropyrrolo[3,4-c]pyrazol-3-yl] benzamide and for the targets MAPK1 and AURKA, respectively. CONCLUSION Our study has identified novel candidate biomarkers, pathways, transcription factors (TFs), and kinases associated with NSCLC prognosis, as well as drug candidates, which may assist treatment strategy for NSCLC patients.
Collapse
Affiliation(s)
- Gurudeeban Selvaraj
- Center of Interdisciplinary Sciences-Computational Life Sciences, College of Food Science and Engineering, Henan University of Technology, Zhengzhou, China; College of Chemistry, Chemical Engineering, and Environment, Henan University of Technology, Zhengzhou, China
| | - Satyavani Kaliamurthi
- Center of Interdisciplinary Sciences-Computational Life Sciences, College of Food Science and Engineering, Henan University of Technology, Zhengzhou, China; College of Chemistry, Chemical Engineering, and Environment, Henan University of Technology, Zhengzhou, China
| | - Aman Chandra Kaushik
- Department of Bioinformatics, The State Key Laboratory of Microbial Metabolism, College of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Abbas Khan
- Department of Bioinformatics, The State Key Laboratory of Microbial Metabolism, College of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Yong-Kai Wei
- College of Science, Henan University of Technology, Zhengzhou, China
| | - William C Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong
| | - Keren Gu
- Center of Interdisciplinary Sciences-Computational Life Sciences, College of Food Science and Engineering, Henan University of Technology, Zhengzhou, China; College of Chemistry, Chemical Engineering, and Environment, Henan University of Technology, Zhengzhou, China
| | - Dong-Qing Wei
- Center of Interdisciplinary Sciences-Computational Life Sciences, College of Food Science and Engineering, Henan University of Technology, Zhengzhou, China; College of Science, Henan University of Technology, Zhengzhou, China; Department of Bioinformatics, The State Key Laboratory of Microbial Metabolism, College of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
24
|
Son BK, Kim DH, Min KW, Kim EK, Kwon MJ. Smad4/Fascin index is highly prognostic in patients with diffuse type EBV-associated gastric cancer. Pathol Res Pract 2018; 214:475-481. [PMID: 29572117 DOI: 10.1016/j.prp.2018.03.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 03/06/2018] [Accepted: 03/13/2018] [Indexed: 01/25/2023]
Abstract
Gastric cancer is a heterogeneous disorder for which predicting clinical outcomes is challenging, although various biomarkers have been suggested. The Smad4 and Fascin proteins are known prognostic indicators of different types of malignancy. Smad4 primarily functions as a key regulator of tumor suppression, whereas Fascin exhibits oncogenic function by enhancing tumor infiltration. A combined marker based on these opposing roles may improve prognostic accuracy in gastric cancer. Smad4 and Fascin expression was assessed in tissue microarrays obtained from 285 primary gastric adenocarcinoma, 201 normal tissue, and 51 metastatic adenocarcinoma samples. A Smad4/Fascin index based on the relative expression of each protein was divided into low- and high-expression groups using receiver operating characteristic curves. We compared normal tissue, primary adenocarcinoma, and metastatic adenocarcinoma in Smad4 and Fascin expression and the differences in clinicopathological findings between low Smad4/Fascin and high Smad4/Fascin expression in gastric adenocarcinoma. High Smad4/Fascin expression was significantly associated with worse outcomes, such as old age, advanced T and N category, large tumor size, high histological grade, lymphatic and vascular invasion, and presence of Epstein-Barr virus (EBV) (all p < 0.05). Univariate and multivariate analyses revealed a significant relationship between disease-free or overall survival and Smad4/Fascin index in diffuse-type or EBV-associated gastric cancer (all p < 0.05). A dual marker system using Smad4 and Fascin may be a reliable indicator for predicting clinical outcomes in patients with diffuse-type or EBV-associated gastric cancer.
Collapse
Affiliation(s)
- Byoung Kwan Son
- Department of Internal Medicine, Eulji Hospital, Eulji University School of medicine, Seoul, Republic of Korea
| | - Dong-Hoon Kim
- Department of Pathology, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Kyueng-Whan Min
- Department of Pathology, Hanyang University Guri Hospital, Hanyang University College of Medicine, Guri, Gyeonggi-do, Republic of Korea.
| | - Eun-Kyung Kim
- Department of Pathology, Eulji Hospital, Eulji University School of Medicine, Seoul, Republic of Korea
| | - Mi Jung Kwon
- Department of Pathology, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang, Gyeonggi-do, Republic of Korea
| |
Collapse
|
25
|
Yang XH, Guo F. miR‑3147 serves as an oncomiR in vulvar squamous cell cancer via Smad4 suppression. Mol Med Rep 2018; 17:6397-6404. [PMID: 29512734 PMCID: PMC5928616 DOI: 10.3892/mmr.2018.8697] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2017] [Accepted: 03/01/2018] [Indexed: 02/06/2023] Open
Abstract
The incidence of vulvar squamous cell carcinoma (VSCC) has increased annually over the last decade. MicroRNAs (miRNAs/miRs) serve an important role in tumor progression and development. Our previous microarray studies have revealed that miR-3147 was overexpressed in VSCC. However, its function and underlying mechanism in VSCC remain unknown. In the present study, it was confirmed by reverse transcription-quantitative polymerase chain reaction that the expression of miR-3147 was markedly upregulated in VSCC tissues. The increased expression of miR-3147 was positively associated with the depth of invasion. The overexpression of miR-3147 resulted in the promotion of vulvar cancer cell proliferation, migration, invasion, G1/S progression and invasion-associated gene expression. miR-3147 may participate in the process of epithelial-mesenchymal transition and reduce the expressions of downstream target genes in the transforming growth factor-β/Smad signaling pathway in A431 cells. The knockdown of Smad4 by small interfering RNA promoted malignant behaviours in A431 cells. In addition, miR-3147 regulated Smad4 by directly binding to its 3′ untranslated region. In conclusion, the results indicated that miR-3147 may serve an oncogenic role in VSCC by targeting Smad4. miR-3147 may represent a novel potential therapeutic target marker for VSCC.
Collapse
Affiliation(s)
- Xiu-Hua Yang
- Department of Obstetrics, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Feng Guo
- Department of Emergency, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| |
Collapse
|
26
|
Tippimanchai DD, Nolan K, Poczobutt J, Verzosa G, Li H, Scarborough H, Huang J, Young C, DeGregori J, Nemenoff RA, Malkoski SP. Adenoviral vectors transduce alveolar macrophages in lung cancer models. Oncoimmunology 2018; 7:e1438105. [PMID: 29872579 PMCID: PMC5980415 DOI: 10.1080/2162402x.2018.1438105] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 01/29/2018] [Accepted: 02/02/2018] [Indexed: 11/09/2022] Open
Abstract
Adenoviral vectors expressing Cre recombinase are commonly used to initiate tumor formation in murine lung cancer models. While these vectors are designed to target genetic recombination to lung epithelial cells, adenoviruses can infect additional cell types that potentially influence tumor development. Our goal was to explore the consequences of adenoviral-mediated alveolar macrophage (AM) transduction in a Kras-initiated lung tumor model. As expected, treatment of animals harboring the KrasLSL-G12D allele and an inducible green fluorescence protein (GFP) tracking allele with an adenoviral vector expressing Cre recombinase under the control of the cytomegalovirus (CMV) promoter (Ad5-CMV-Cre), caused GFP-positive lung adenocarcinomas. Surprisingly, however, up to 70% of the total GFP+ cells were AM, and GFP+ AM could be detected 6 months after tumor initiation, and transduced AM demonstrated Kras activation and increased proliferation. In contrast, recombination was not detected in other immune cell populations and AM recombination could be eliminated by tumor initiation with an adenovirus expressing Cre recombinase under the control of the surfactant protein C (SPC) promoter. In addition, AM isolated from KrasLSL-G12D animals and transduced by Ad5-CMV-Cre ex vivo displayed prolonged survival in vitro and increased the growth of murine lung adenocarcinoma CMT/167 cells when co-injected in an orthotopic flank model. Given the importance of the immune system in tumor development and progression, inadvertent AM transduction by Ad5-CMV-Cre merits careful consideration during lung cancer model selection particularly if studies evaluating the tumor-immune interactions are planned.
Collapse
Affiliation(s)
- Darinee D Tippimanchai
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Denver Anschutz Medical Campus, Aurora, CO
| | - Kyle Nolan
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Denver Anschutz Medical Campus, Aurora, CO
| | - Joanna Poczobutt
- Division of Renal Disease and Hypertension, University of Colorado Denver Anschutz Medical Campus, Aurora, CO
| | - Gregory Verzosa
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Denver Anschutz Medical Campus, Aurora, CO
| | - Howard Li
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Denver Anschutz Medical Campus, Aurora, CO
| | - Hannah Scarborough
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver Anschutz Medical Campus, Aurora, CO
| | - Jing Huang
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Denver Anschutz Medical Campus, Aurora, CO
| | - Christian Young
- Department of Pathology, University of Colorado Denver Anschutz Medical Campus, Aurora, CO
| | - James DeGregori
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver Anschutz Medical Campus, Aurora, CO
| | - Raphael A Nemenoff
- Division of Renal Disease and Hypertension, University of Colorado Denver Anschutz Medical Campus, Aurora, CO
| | - Stephen P Malkoski
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Denver Anschutz Medical Campus, Aurora, CO
| |
Collapse
|
27
|
Nikolova O, Moser R, Kemp C, Gönen M, Margolin AA. Modeling gene-wise dependencies improves the identification of drug response biomarkers in cancer studies. Bioinformatics 2018; 33:1362-1369. [PMID: 28082455 DOI: 10.1093/bioinformatics/btw836] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 12/29/2016] [Indexed: 01/07/2023] Open
Abstract
Motivation In recent years, vast advances in biomedical technologies and comprehensive sequencing have revealed the genomic landscape of common forms of human cancer in unprecedented detail. The broad heterogeneity of the disease calls for rapid development of personalized therapies. Translating the readily available genomic data into useful knowledge that can be applied in the clinic remains a challenge. Computational methods are needed to aid these efforts by robustly analyzing genome-scale data from distinct experimental platforms for prioritization of targets and treatments. Results We propose a novel, biologically motivated, Bayesian multitask approach, which explicitly models gene-centric dependencies across multiple and distinct genomic platforms. We introduce a gene-wise prior and present a fully Bayesian formulation of a group factor analysis model. In supervised prediction applications, our multitask approach leverages similarities in response profiles of groups of drugs that are more likely to be related to true biological signal, which leads to more robust performance and improved generalization ability. We evaluate the performance of our method on molecularly characterized collections of cell lines profiled against two compound panels, namely the Cancer Cell Line Encyclopedia and the Cancer Therapeutics Response Portal. We demonstrate that accounting for the gene-centric dependencies enables leveraging information from multi-omic input data and improves prediction and feature selection performance. We further demonstrate the applicability of our method in an unsupervised dimensionality reduction application by inferring genes essential to tumorigenesis in the pancreatic ductal adenocarcinoma and lung adenocarcinoma patient cohorts from The Cancer Genome Atlas. Availability and Implementation : The code for this work is available at https://github.com/olganikolova/gbgfa. Contact : nikolova@ohsu.edu or margolin@ohsu.edu. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Olga Nikolova
- Computational Biology Program, Oregon Health and Science University, Portland, OR 97239, USA
| | - Russell Moser
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Christopher Kemp
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Mehmet Gönen
- Computational Biology Program, Oregon Health and Science University, Portland, OR 97239, USA.,Department of Industrial Engineering, Koç University, İstanbul, Turkey
| | - Adam A Margolin
- Computational Biology Program, Oregon Health and Science University, Portland, OR 97239, USA
| |
Collapse
|
28
|
Chung Y, Wi YC, Kim Y, Bang SS, Yang JH, Jang K, Min KW, Paik SS. The Smad4/PTEN Expression Pattern Predicts Clinical Outcomes in Colorectal Adenocarcinoma. J Pathol Transl Med 2017; 52:37-44. [PMID: 29056035 PMCID: PMC5784229 DOI: 10.4132/jptm.2017.10.20] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 09/19/2017] [Accepted: 10/16/2017] [Indexed: 02/07/2023] Open
Abstract
Background Smad4 and PTEN are prognostic indicators for various tumor types. Smad4 regulates tumor suppression, whereas PTEN inhibits cell proliferation. We analyzed and compared the performance of Smad4 and PTEN for predicting the prognosis of patients with colorectal adenocarcinoma. Methods Combined expression patterns based on Smad4+/– and PTEN+/– status were evaluated by immunostaining using a tissue microarray of colorectal adenocarcinoma. The relationships between the protein expression and clinicopathological variables were analyzed. Results Smad4–/PTEN– status was most frequently observed in metastatic adenocarcinoma, followed by primary adenocarcinoma and tubular adenoma (p<.001). When Smad4–/PTEN– and Smad4+/PTEN+ groups were compared, Smad4–/PTEN– status was associated with high N stage (p=.018) and defective mismatch repair proteins (p=.006). Significant differences in diseasefree survival and overall survival were observed among the three groups (Smad4+/PTEN+, Smad4–/PTEN+ or Smad4+/PTEN–, and Smad4–/PTEN–) (all p<.05). Conclusions Concurrent loss of Smad4 and PTEN may lead to more aggressive disease and poor prognosis in patients with colorectal adenocarcinoma compared to the loss of Smad4 or PTEN alone.
Collapse
Affiliation(s)
- Yumin Chung
- Department of Pathology, Hanyang University College of Medicine, Seoul, Korea
| | - Young Chan Wi
- Department of Pathology, Hanyang University College of Medicine, Seoul, Korea
| | - Yeseul Kim
- Department of Pathology, Hanyang University College of Medicine, Seoul, Korea
| | - Seong Sik Bang
- Department of Pathology, Hanyang University College of Medicine, Seoul, Korea
| | - Jung-Ho Yang
- Department of Pathology, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Kiseok Jang
- Department of Pathology, Hanyang University College of Medicine, Seoul, Korea
| | - Kyueng-Whan Min
- Department of Pathology, Hanyang University College of Medicine, Seoul, Korea
| | - Seung Sam Paik
- Department of Pathology, Hanyang University College of Medicine, Seoul, Korea
| |
Collapse
|
29
|
Zhou S, Zhang Z, Zheng P, Zhao W, Han N. MicroRNA-1285-5p influences the proliferation and metastasis of non-small-cell lung carcinoma cells via downregulating CDH1 and Smad4. Tumour Biol 2017. [PMID: 28631567 DOI: 10.1177/1010428317705513] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Abnormal expression of microRNAs has been reported to regulate gene expression and cancer cell growth, invasion, and migration. Recently, upregulation of hsa-miR-1285 was demonstrated in bronchoalveolar lavage fluid samples from patients with lung cancer and downregulation in plasma level of stage-I lung cancer patients. However, the function and the underlying mechanism of miR-1285 in non-small-cell lung carcinoma have not been elucidated. In this study, we found that miR-1285-5p, the mature form of miR-1285, was significantly upregulated in human non-small-cell lung carcinoma cell lines A549 and SK-MES-1. Additionally, cells transfected with the miR-1285-5p inhibitor LV-anti-miR-1285-5p demonstrated significantly inhibited proliferation and invasion and depressed migration. Further analysis demonstrated that the miR-1285-5p precursor LV-miR-1285-5p attenuated the expression of Smad4 and cadherin-1 (CDH1) but that LV-anti-miR-1285-5p showed opposite results. A luciferase reporter assay confirmed that miR-1285-5p targeted Smad4 and CDH1. Mechanism analyses revealed that silence of Smad4 and CDH1 significantly attenuated the inhibitory effects of LV-anti-miR-1285-5p on non-small-cell lung carcinoma growth and invasion. Taken together, our data suggest that miR-1285-5p functions as a tumor promoter in the development of non-small-cell lung carcinoma by targeting Smad4 and CDH1, indicating a novel therapeutic strategy for non-small-cell lung carcinoma patients.
Collapse
Affiliation(s)
- Shixia Zhou
- 1 Department of Oncology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhongmian Zhang
- 1 Department of Oncology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Pengyuan Zheng
- 2 Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wenchao Zhao
- 3 Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Na Han
- 1 Department of Oncology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
30
|
Ozawa H, Ranaweera RS, Izumchenko E, Makarev E, Zhavoronkov A, Fertig EJ, Howard JD, Markovic A, Bedi A, Ravi R, Perez J, Le QT, Kong CS, Jordan RC, Wang H, Kang H, Quon H, Sidransky D, Chung CH. SMAD4 Loss Is Associated with Cetuximab Resistance and Induction of MAPK/JNK Activation in Head and Neck Cancer Cells. Clin Cancer Res 2017; 23:5162-5175. [PMID: 28522603 DOI: 10.1158/1078-0432.ccr-16-1686] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2016] [Revised: 12/01/2016] [Accepted: 05/15/2017] [Indexed: 01/01/2023]
Abstract
Purpose: We previously demonstrated an association between decreased SMAD4 expression and cetuximab resistance in head and neck squamous cell carcinoma (HNSCC). The purpose of this study was to further elucidate the clinical relevance of SMAD4 loss in HNSCC.Experimental Design: SMAD4 expression was assessed by IHC in 130 newly diagnosed and 43 patients with recurrent HNSCC. Correlative statistical analysis with clinicopathologic data was also performed. OncoFinder, a bioinformatics tool, was used to analyze molecular signaling in TCGA tumors with low or high SMAD4 mRNA levels. The role of SMAD4 was investigated by shRNA knockdown and gene reconstitution of HPV-negative HNSCC cell lines in vitro and in vivoResults: Our analysis revealed that SMAD4 loss was associated with an aggressive, HPV-negative, cetuximab-resistant phenotype. We found a signature of prosurvival and antiapoptotic pathways that were commonly dysregulated in SMAD4-low cases derived from TCGA-HNSCC dataset and an independent oral cavity squamous cell carcinoma (OSCC) cohort obtained from GEO. We show that SMAD4 depletion in an HNSCC cell line induces cetuximab resistance and results in worse survival in an orthotopic mouse model in vivo We implicate JNK and MAPK activation as mediators of cetuximab resistance and provide the foundation for the concomitant EGFR and JNK/MAPK inhibition as a potential strategy for overcoming cetuximab resistance in HNSCCs with SMAD4 loss.Conclusions: Our study demonstrates that loss of SMAD4 expression is a signature characterizing the cetuximab-resistant phenotype and suggests that SMAD4 expression may be a determinant of sensitivity/resistance to EGFR/MAPK or EGFR/JNK inhibition in HPV-negative HNSCC tumors. Clin Cancer Res; 23(17); 5162-75. ©2017 AACR.
Collapse
Affiliation(s)
- Hiroyuki Ozawa
- Department of Oncology, School of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Ruchira S Ranaweera
- Department of Oncology, School of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Evgeny Izumchenko
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Eugene Makarev
- Insilico Medicine, Inc, ETC, Johns Hopkins University, Baltimore, Maryland
| | - Alex Zhavoronkov
- Insilico Medicine, Inc, ETC, Johns Hopkins University, Baltimore, Maryland
| | - Elana J Fertig
- Department of Oncology, School of Medicine, Johns Hopkins University, Baltimore, Maryland
- Department of Health Science Informatics, Johns Hopkins University, Baltimore, Maryland
| | - Jason D Howard
- Department of Oncology, School of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Ana Markovic
- Department of Oncology, School of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Atul Bedi
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Rajani Ravi
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jimena Perez
- Department of Oncology, School of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Quynh-Thu Le
- Department of Pathology, Stanford University School of Medicine Stanford, California
| | - Christina S Kong
- Department of Pathology, Stanford University School of Medicine Stanford, California
| | - Richard C Jordan
- Departments of Orofacial Sciences and Pathology, University of California, San Francisco, San Francisco, CA, USA
| | - Hao Wang
- Department of Oncology, School of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Hyunseok Kang
- Department of Oncology, School of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Harry Quon
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University, Baltimore, Maryland
| | - David Sidransky
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Christine H Chung
- Department of Oncology, School of Medicine, Johns Hopkins University, Baltimore, Maryland.
- Department of Head and Neck-Endocrine Oncology, Moffitt Cancer Center, Tampa, Florida
| |
Collapse
|
31
|
Ziemke M, Patil T, Nolan K, Tippimanchai D, Malkoski SP. Reduced Smad4 expression and DNA topoisomerase inhibitor chemosensitivity in non-small cell lung cancer. Lung Cancer 2017; 109:28-35. [PMID: 28577946 DOI: 10.1016/j.lungcan.2017.04.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 04/13/2017] [Accepted: 04/22/2017] [Indexed: 01/13/2023]
Abstract
OBJECTIVE Smad4 is a tumor suppressor that transduces transforming growth factor beta signaling and regulates genomic stability. We previously found that Smad4 knockdown in vitro inhibited DNA repair and increased sensitivity to DNA topoisomerase inhibitors. In this study, we assessed the association between reduced Smad4 expression and DNA topoisomerase inhibitor sensitivity in human non-small cell lung cancer (NSCLC) patients and evaluated the relationship between genomic alterations of Smad4 and molecular alterations in DNA repair molecules. MATERIALS AND METHODS We retrospectively identified NSCLC patients who received etoposide or gemcitabine. Chemotherapeutic response was quantified by RECIST 1.1 criteria and Smad4 expression was assessed by immunohistochemistry. Relationships between Smad4 mutation and DNA repair molecule mutations were evaluated using publically available datasets. RESULTS We identified 28 individuals who received 30 treatments with gemcitabine or etoposide containing regimens for NSCLC. Reduced Smad4 expression was seen in 13/28 patients and was not associated with significant differences in clinical or pathologic parameters. Patients with reduced Smad4 expression had a larger response to DNA topoisomerase inhibitor containing regimens then patients with high Smad4 expression (-25.7% vs. -6.8% in lesion size, p=0.03); this relationship was more pronounced with gemcitabine containing regimens. The overall treatment response was higher in patients with reduced Smad4 expression (8/14 vs 2/16 p=0.02). Analysis of data from The Cancer Genome Atlas revealed that Smad4 mutation or homozygous loss was mutually exclusive with genomic alterations in DNA repair molecules. CONCLUSIONS Reduced Smad4 expression may predict responsiveness to regimens that contain DNA topoisomerase inhibitors. That Smad4 signaling alterations are mutually exclusive with alterations in DNA repair machinery is consistent with an important role of Smad4 in regulating DNA repair.
Collapse
Affiliation(s)
- Michael Ziemke
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, United States
| | - Tejas Patil
- Division of General Internal Medicine and Divison of Medical Oncology, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, United States
| | - Kyle Nolan
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, United States
| | - Darinee Tippimanchai
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, United States
| | - Stephen P Malkoski
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, United States; Department of Pathology, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, United States.
| |
Collapse
|
32
|
Gan TQ, Xie ZC, Tang RX, Zhang TT, Li DY, Li ZY, Chen G. Clinical value of miR-145-5p in NSCLC and potential molecular mechanism exploration: A retrospective study based on GEO, qRT-PCR, and TCGA data. Tumour Biol 2017; 39:1010428317691683. [PMID: 28347234 DOI: 10.1177/1010428317691683] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
MicroRNAs have been reported to be involved in various biological processes. Here, we performed a systematic analysis to explore the clinical value and potential molecular mechanism of miR-145-5p in non-small cell lung cancer. First, a meta-analysis was performed with eligible literature, followed by microRNA microarrays in the Gene Expression Omnibus database, to verify the diagnostic and prognostic values of miR-145-5p. A cohort of 125 clinical paired non-small cell lung cancer samples was next used to detect the level of miR-145-5p and to explore the relationship of miR-145-5p with clinicopathological parameters. The Cancer Genome Atlas database was additionally applied to investigate the role of miR-145-5p in non-small cell lung cancer. The potential targets of miR-145-5p were predicted using 12 online prediction databases to explore the prospective molecular mechanism of miR-145-5p in non-small cell lung cancer. The expression of miR-145-5p in non-small cell lung cancer was significantly lower than that in healthy tissues. And miR-145-5p tended to show better diagnostic performance in lung squamous cell carcinoma than in lung adenocarcinoma. Furthermore, the expression of miR-145-5p was closely associated with lymph node metastasis in non-small cell lung cancer. Gene ontology enrichment and Kyoto Encyclopedia of Genes and Genomes pathway analysis revealed that the target genes were mainly enriched with enzyme-linked receptor protein signaling pathways, SH3 domain binding, cell leading edge, and adherens junction. The protein-protein interaction network showed that eight hub genes (SMAD4, SMAD2, IRS1, FOXO1, ERBB4, NRAS, ACTB, and ACTG1) might be the key target genes of miR-145-5p in non-small cell lung cancer. The information we obtained might offer new perspectives for clinical diagnosis and treatment for non-small cell lung cancer.
Collapse
Affiliation(s)
- Ting-Qing Gan
- 1 Department of Medical Oncology, First Affiliated Hospital of Guangxi Medical University, Nanning, P.R. China
| | - Zu-Cheng Xie
- 2 Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, P.R. China
| | - Rui-Xue Tang
- 2 Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, P.R. China
| | - Tong-Tong Zhang
- 2 Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, P.R. China
| | - Dong-Yao Li
- 3 Department of Thoracic and Cardiovascular Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, P.R. China
| | - Zu-Yun Li
- 2 Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, P.R. China
| | - Gang Chen
- 2 Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, P.R. China
| |
Collapse
|
33
|
Zheng L, Jiao W, Song H, Qu H, Li D, Mei H, Chen Y, Yang F, Li H, Huang K, Tong Q. miRNA-558 promotes gastric cancer progression through attenuating Smad4-mediated repression of heparanase expression. Cell Death Dis 2016; 7:e2382. [PMID: 27685626 PMCID: PMC5059886 DOI: 10.1038/cddis.2016.293] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 08/20/2016] [Accepted: 08/23/2016] [Indexed: 12/25/2022]
Abstract
Previous studies have indicated that as the only mammalian endo-β-D-glucuronidase, heparanase (HPSE) is up-regulated and associated with poor prognosis in gastric cancer, while the underlying mechanisms still remain to be determined. Herein, through integrative analysis of public datasets, we found microRNA-558 (miR-558) and SMAD family member 4 (Smad4) as the crucial transcription regulators of HPSE expression in gastric cancer, with their adjacent target sites within the promoter of HPSE. We identified that endogenous miR-558 activated the transcription and expression of HPSE in gastric cancer cell lines. In contrast, Smad4 suppressed the nascent transcription and expression of HPSE via directly binding to its promoter. Mechanistically, miR-558 recognized its complementary site within HPSE promoter to decrease the binding of Smad4 in an Argonaute 1-dependent manner. Ectopic expression or knockdown experiments indicated that miR-558 promoted the in vitro and in vivo tumorigenesis and aggressiveness of gastric cancer cell lines via attenuating Smad4-mediated repression of HPSE expression. In clinical gastric cancer specimens, up-regulation of miR-558 and down-regulation of Smad4 were positively correlated with HPSE expression. Kaplan–Meier survival analysis revealed that miR-558 and Smad4 were associated with unfavourable and favourable outcome of gastric cancer patients, respectively. Therefore, these findings demonstrate that miR-558 facilitates the progression of gastric cancer through directly targeting the HPSE promoter to attenuate Smad4-mediated repression of HPSE expression.
Collapse
Affiliation(s)
- Liduan Zheng
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei Province, P. R. China.,Clinical Center of Human Genomic Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei Province, P. R. China
| | - Wanju Jiao
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei Province, P. R. China
| | - Huajie Song
- Department of Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei Province, P. R. China
| | - Hongxia Qu
- Department of Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei Province, P. R. China
| | - Dan Li
- Department of Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei Province, P. R. China
| | - Hong Mei
- Department of Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei Province, P. R. China
| | - Yajun Chen
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei Province, P. R. China
| | - Feng Yang
- Department of Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei Province, P. R. China
| | - Huanhuan Li
- Department of Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei Province, P. R. China
| | - Kai Huang
- Clinical Center of Human Genomic Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei Province, P. R. China
| | - Qiangsong Tong
- Clinical Center of Human Genomic Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei Province, P. R. China.,Department of Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei Province, P. R. China
| |
Collapse
|
34
|
Virtual microdissection identifies distinct tumor- and stroma-specific subtypes of pancreatic ductal adenocarcinoma. Nat Genet 2015; 47:1168-78. [PMID: 26343385 PMCID: PMC4912058 DOI: 10.1038/ng.3398] [Citation(s) in RCA: 1441] [Impact Index Per Article: 144.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 08/17/2015] [Indexed: 12/11/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains a lethal disease with a 5-year survival rate of 4%. A key hallmark of PDAC is extensive stromal involvement, which makes capturing precise tumor-specific molecular information difficult. Here we have overcome this problem by applying blind source separation to a diverse collection of PDAC gene expression microarray data, including data from primary tumor, metastatic and normal samples. By digitally separating tumor, stromal and normal gene expression, we have identified and validated two tumor subtypes, including a 'basal-like' subtype that has worse outcome and is molecularly similar to basal tumors in bladder and breast cancers. Furthermore, we define 'normal' and 'activated' stromal subtypes, which are independently prognostic. Our results provide new insights into the molecular composition of PDAC, which may be used to tailor therapies or provide decision support in a clinical setting where the choice and timing of therapies are critical.
Collapse
|