1
|
Liu L, Liu X, Gao C, Liu M, Peng M, Wang L. Hsa-miR-21 promoted the progression of lung adenocarcinoma by regulating LRIG1 expression. BMC Pulm Med 2025; 25:189. [PMID: 40269842 PMCID: PMC12016109 DOI: 10.1186/s12890-025-03620-1] [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: 11/16/2024] [Accepted: 03/24/2025] [Indexed: 04/25/2025] Open
Abstract
Lung cancer is the foremost cause of cancer-related fatalities globally, and lung adenocarcinoma (LUAD) is one of the common types of lung cancer with significant molecular heterogeneity. Leucine rich repeats and immunoglobulin like domains 1 (LRIG1) has been demonstrated to be down-regulated in lung cancer and related to prognosis of patients. The purpose of this work is to explore the targeting miRNAs of LRIG1, and the related regulatory mechanisms in LUAD. The data of LUAD patients were collected from The Cancer Genome Atlas and Gene Expression Omnibus databases. The differential expression analysis and gene set enrichment analysis (GSEA) were performed using "limma" and "clusterProfiler" function package, respectively. The levels of hsa-miR-21 mRNA and LRIG1 mRNA and LRIG1 protein expressions were analyzed using RT-qPCR and western blot analysis. The infiltration of immune cells was determined using CIBERSORT software. In LUAD patients, hsa-miR-21 expression was observably related to LRIG1 expression. Hsa-miR-21 might negatively modulate the LRIG1 expression in LUAD. LUAD patients with hsa-miR-21 up-regulation exhibited inferior prognosis. In addition, those with LUAD who had high hsa-miR-21 expression but low LRIG1 expression had a worse prognosis, whereas those with low hsa-miR-21 expression but high LRIG1 expression had a better prognosis. Functional enrichment analysis indicated that metabolic related signaling pathways (EGFR tyrosine kinase inhibitor resistance) were significantly activated in LUAD patients with LRIG1 up-regulation. Finally, we found that relative content of naive B cells, plasma cells and resting CD4 + T cells were significantly increased and regulatory T cells and Macrophages M0 were decreased in LRIG1 high expression group and hsa-miR-21 low expression group. We firstly reported that hsa-miR-21 might regulate the LRIG1 expression in LUAD, thereby effecting the onset and progression of LUAD. Clinical trial number: Not applicable.
Collapse
Affiliation(s)
- Li Liu
- Department of Respiratory, Weifang People's Hospital, No. 151 Guangwen Street, Kuiwen District, Weifang, 261041, Shandong Province, P. R. China
| | - Xinhua Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Hangzhou Normal University, Yuhang District, Hangzhou, 311121, Zhejiang Province, P. R. China
| | - Chengpeng Gao
- Department of Respiratory, Weifang People's Hospital, No. 151 Guangwen Street, Kuiwen District, Weifang, 261041, Shandong Province, P. R. China
| | - Meijuan Liu
- Department of Respiratory, Weifang People's Hospital, No. 151 Guangwen Street, Kuiwen District, Weifang, 261041, Shandong Province, P. R. China.
| | - Mengmeng Peng
- Department of Respiratory, Weifang People's Hospital, No. 151 Guangwen Street, Kuiwen District, Weifang, 261041, Shandong Province, P. R. China
| | - Leqiang Wang
- Department of Respiratory, Weifang People's Hospital, No. 151 Guangwen Street, Kuiwen District, Weifang, 261041, Shandong Province, P. R. China
| |
Collapse
|
2
|
Shimizu S, Kondo J, Onuma K, Coppo R, Ota K, Kamada M, Harada Y, Tanaka Y, Nakazawa MA, Tamada Y, Okuno Y, Kawada K, Obama K, Coffey RJ, Fujiwara Y, Inoue M. Inhibition of the bone morphogenetic protein pathway suppresses tumor growth through downregulation of epidermal growth factor receptor in MEK/ERK-dependent colorectal cancer. Cancer Sci 2023; 114:3636-3648. [PMID: 37357017 PMCID: PMC10475764 DOI: 10.1111/cas.15882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 05/22/2023] [Accepted: 05/27/2023] [Indexed: 06/27/2023] Open
Abstract
The bone morphogenetic protein (BMP) pathway promotes differentiation and induces apoptosis in normal colorectal epithelial cells. However, its role in colorectal cancer (CRC) is controversial, where it can act as context-dependent tumor promoter or tumor suppressor. Here we have found that CRC cells reside in a BMP-rich environment based on curation of two publicly available RNA-sequencing databases. Suppression of BMP using a specific BMP inhibitor, LDN193189, suppresses the growth of select CRC organoids. Colorectal cancer organoids treated with LDN193189 showed a decrease in epidermal growth factor receptor, which was mediated by protein degradation induced by leucine-rich repeats and immunoglobulin-like domains protein 1 (LRIG1) expression. Among 18 molecularly characterized CRC organoids, suppression of growth by BMP inhibition correlated with induction of LRIG1 gene expression. Notably, knockdown of LRIG1 in organoids diminished the growth-suppressive effect of LDN193189. Furthermore, in CRC organoids, which are susceptible to growth suppression by LDN193189, simultaneous treatment with LDN193189 and trametinib, an FDA-approved MEK inhibitor, resulted in cooperative growth inhibition both in vitro and in vivo. Taken together, the simultaneous inhibition of BMP and MEK could be a novel treatment option in CRC cases, and evaluating in vitro growth suppression and LRIG1 induction by BMP inhibition using patient-derived organoids could offer functional biomarkers for predicting potential responders to this regimen.
Collapse
Affiliation(s)
- Shota Shimizu
- Department of Clinical Bioresource Research and DevelopmentKyoto University Graduate School of MedicineKyotoJapan
- Division of Gastrointestinal and Pediatric Surgery, Department of Surgery, School of MedicineTottori University Faculty of MedicineTottoriJapan
| | - Jumpei Kondo
- Department of Clinical Bioresource Research and DevelopmentKyoto University Graduate School of MedicineKyotoJapan
- Department of BiochemistryOsaka International Cancer InstituteOsakaJapan
- Department of Molecular Biochemistry and Clinical Investigation, Division of Health ScienceOsaka University Graduate School of MedicineOsakaJapan
- Epithelial Biology CenterVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Kunishige Onuma
- Department of Clinical Bioresource Research and DevelopmentKyoto University Graduate School of MedicineKyotoJapan
| | - Roberto Coppo
- Department of Clinical Bioresource Research and DevelopmentKyoto University Graduate School of MedicineKyotoJapan
| | - Kasumi Ota
- Graduate School of Medicine and Faculty of MedicineKyoto UniversityKyotoJapan
| | - Mayumi Kamada
- Graduate School of Medicine and Faculty of MedicineKyoto UniversityKyotoJapan
| | - Yohei Harada
- Graduate School of Medicine and Faculty of MedicineKyoto UniversityKyotoJapan
| | - Yoshihisa Tanaka
- Graduate School of Pharmaceutical SciencesKyoto UniversityKyotoJapan
- RIKEN Center for Computational Science(R‐CCS)HPC/HPC‐ and AI‐driven Drug Development Platform DivisionKobeJapan
| | - Mai Adachi Nakazawa
- Graduate School of Medicine and Faculty of MedicineKyoto UniversityKyotoJapan
- Department of Medical Data Intelligence and Data Analysis Division, Innovation Center for Health PromotionHirosaki UniversityHirosakiJapan
| | - Yoshinori Tamada
- Department of Medical Data Intelligence and Data Analysis Division, Innovation Center for Health PromotionHirosaki UniversityHirosakiJapan
| | - Yasushi Okuno
- Graduate School of Medicine and Faculty of MedicineKyoto UniversityKyotoJapan
- RIKEN Center for Computational Science(R‐CCS)HPC/HPC‐ and AI‐driven Drug Development Platform DivisionKobeJapan
| | - Kenji Kawada
- Department of Surgery, Graduate School of MedicineKyoto UniversityKyotoJapan
| | - Kazutaka Obama
- Department of Surgery, Graduate School of MedicineKyoto UniversityKyotoJapan
| | - Robert J. Coffey
- Epithelial Biology CenterVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Yoshiyuki Fujiwara
- Division of Gastrointestinal and Pediatric Surgery, Department of Surgery, School of MedicineTottori University Faculty of MedicineTottoriJapan
| | - Masahiro Inoue
- Department of Clinical Bioresource Research and DevelopmentKyoto University Graduate School of MedicineKyotoJapan
- Department of BiochemistryOsaka International Cancer InstituteOsakaJapan
| |
Collapse
|
3
|
Succony L, Gómez-López S, Pennycuick A, Alhendi ASN, Davies D, Clarke SE, Gowers KHC, Wright NA, Jensen KB, Janes SM. Lrig1 expression identifies airway basal cells with high proliferative capacity and restricts lung squamous cell carcinoma growth. Eur Respir J 2021; 59:13993003.00816-2020. [PMID: 34385275 PMCID: PMC8968013 DOI: 10.1183/13993003.00816-2020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 08/01/2021] [Indexed: 12/24/2022]
Abstract
Background Lung squamous cell carcinoma (LUSC) accounts for a significant proportion of cancer deaths worldwide, and is preceded by the appearance of progressively disorganised pre-invasive lesions in the airway epithelium. Yet the biological mechanisms underlying progression of pre-invasive lesions into invasive LUSC are not fully understood. LRIG1 (leucine-rich repeats and immunoglobulin-like domains 1) is downregulated in pre-invasive airway lesions and invasive LUSC tumours and this correlates with decreased lung cancer patient survival. Methods and results Using an Lrig1 knock-in reporter mouse and human airway epithelial cells collected at bronchoscopy, we show that during homeostasis LRIG1 is heterogeneously expressed in the airway epithelium. In basal airway epithelial cells, the suspected cell of origin of LUSC, LRIG1 identifies a subpopulation of progenitor cells with higher in vitro proliferative and self-renewal potential in both the mouse and human. Using the N-nitroso-tris-chloroethylurea (NTCU)-induced murine model of LUSC, we find that Lrig1 loss-of-function leads to abnormally high cell proliferation during the earliest stages of pre-invasive disease and to the formation of significantly larger invasive tumours, suggesting accelerated disease progression. Conclusion Together, our findings identify LRIG1 as a marker of basal airway progenitor cells with high proliferative potential and as a regulator of pre-invasive lung cancer progression. This work highlights the clinical relevance of LRIG1 and the potential of the NTCU-induced LUSC model for functional assessment of candidate tumour suppressors and oncogenes. LRIG1 is lost in development of squamous cell lung cancers. This study shows that LRIG1 marks basal airway progenitor cells with high proliferative potential and regulates progression of pre-invasive squamous cell lung cancer.https://bit.ly/3AbPtY3
Collapse
Affiliation(s)
- Laura Succony
- Lungs for Living Research Centre, UCL Respiratory, University College London, London, UK.,These authors contributed equally to this work
| | - Sandra Gómez-López
- Lungs for Living Research Centre, UCL Respiratory, University College London, London, UK.,These authors contributed equally to this work
| | - Adam Pennycuick
- Lungs for Living Research Centre, UCL Respiratory, University College London, London, UK
| | - Ahmed S N Alhendi
- Lungs for Living Research Centre, UCL Respiratory, University College London, London, UK
| | - Derek Davies
- Flow Cytometry Facility, Francis Crick Institute, London, UK
| | - Sarah E Clarke
- Lungs for Living Research Centre, UCL Respiratory, University College London, London, UK
| | - Kate H C Gowers
- Lungs for Living Research Centre, UCL Respiratory, University College London, London, UK
| | - Nicholas A Wright
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Kim B Jensen
- Biotech Research and Innovation Centre, University of Copenhagen; Novo Nordisk Foundation Center for Stem Cell Biology, DanStem, University of Copenhagen, Copenhagen, Denmark
| | - Sam M Janes
- Lungs for Living Research Centre, UCL Respiratory, University College London, London, UK
| |
Collapse
|
4
|
Ji Y, Kumar R, Gokhale A, Chao HP, Rycaj K, Chen X, Li Q, Tang DG. LRIG1, a regulator of stem cell quiescence and a pleiotropic feedback tumor suppressor. Semin Cancer Biol 2021; 82:120-133. [PMID: 33476721 PMCID: PMC8286266 DOI: 10.1016/j.semcancer.2020.12.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 12/04/2020] [Accepted: 12/16/2020] [Indexed: 12/14/2022]
Abstract
LRIG1, leucine-rich repeats and immunoglobulin-like domains protein 1, was discovered more than 20 years ago and has been shown to be downregulated or lost, and to function as a tumor suppressor in several cancers. Another well-reported biological function of LRIG1 is to regulate and help enforce the quiescence of adult stem cells (SCs). In both contexts, LRIG1 regulates SC quiescence and represses tumor growth via, primarily, antagonizing the expression and activities of ERBB and other receptor tyrosine kinases (RTKs). We have recently reported that in treatment-naïve human prostate cancer (PCa), LRIG1 is primarily regulated by androgen receptor (AR) and is prominently overexpressed. In castration-resistant PCa (CRPC), both LRIG1 and AR expression becomes heterogeneous and, frequently, discordant. Importantly, in both androgen-dependent PCa and CRPC models, LRIG1 exhibits tumor-suppressive functions. Moreover, LRIG1 induction inhibits the growth of pre-established AR+ and AR− PCa. Here, upon a brief introduction of the LRIG1 and the LRIG family, we provide an updated overview on LRIG1 functions in regulating SC quiescence and repressing tumor development. We further highlight the expression, regulation and functions of LRIG1 in treatment-naïve PCa and CRPC. We conclude by offering the perspectives of identifying novel cancer-specific LRIG1-interacting signaling partners and developing LRIG1-based anti-cancer therapeutics and diagnostic/prognostic biomarkers.
Collapse
Affiliation(s)
- Yibing Ji
- Department of Pharmacology & Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA.
| | - Rahul Kumar
- Department of Pharmacology & Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Abhiram Gokhale
- Department of Pharmacology & Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Hseu-Ping Chao
- Department of Epigenetics & Mol. Carcinogenesis, the University of Texas M.D Anderson Cancer Center, Smithville, TX 78957, USA
| | - Kiera Rycaj
- Department of Pharmacology & Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA; Department of Epigenetics & Mol. Carcinogenesis, the University of Texas M.D Anderson Cancer Center, Smithville, TX 78957, USA
| | - Xin Chen
- Department of Pharmacology & Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Qiuhui Li
- Department of Pharmacology & Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA.
| | - Dean G Tang
- Department of Pharmacology & Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA; Department of Epigenetics & Mol. Carcinogenesis, the University of Texas M.D Anderson Cancer Center, Smithville, TX 78957, USA.
| |
Collapse
|
5
|
Xiong D, Wang Y, You M. Tumor intrinsic immunity related proteins may be novel tumor suppressors in some types of cancer. Sci Rep 2019; 9:10918. [PMID: 31358815 PMCID: PMC6662687 DOI: 10.1038/s41598-019-47382-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 07/11/2019] [Indexed: 02/06/2023] Open
Abstract
Immune checkpoint blockade therapy (ICBT) can unleash T-cell responses against cancer. However, only a small fraction of patients exhibited responses to ICBT. The role of immune checkpoints in cancer cells is not well understood. In this study, we analyzed T-cell coinhibitory/costimulatory genes across more than 1100 samples of the Cancer Cell Line Encyclopedia (CCLE). Nearly 90% of such genes were not expressed or had low expression across the CCLE cancer cell lines. Cell line screening showed the enrichment of cancer cells deprived of the expression of CD27, CEACAM1, CTLA4, LRIG1, PDCD1LG2, or TNFRSF18, suggesting their role as tumor suppressor. The metagene expression signature derived from these six genes - Immu6Metagene was associated with prolonged survival phenotypes. A common set of five oncogenic pathways were significantly inhibited in different types of tumors of the cancer patients with good survival outcome and high Immu6Metagene signature expression. These pathways were TGF-β signaling, angiogenesis, EMT, hypoxia and mitotic process. Our study showed that oncoimmunology related molecules especially the six genes of the Immu6Metagene signature may play the tumor suppressor role in certain cancers. Therefore, the ICBT targeting them should be considered in such context to improve the efficacy.
Collapse
Affiliation(s)
- Donghai Xiong
- Center for Disease Prevention Research and Department of Pharmacology and Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Yian Wang
- Center for Disease Prevention Research and Department of Pharmacology and Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Ming You
- Center for Disease Prevention Research and Department of Pharmacology and Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.
| |
Collapse
|
6
|
LRIG1 acts as a critical regulator of melanoma cell invasion, migration, and vasculogenic mimicry upon hypoxia by regulating EGFR/ERK-triggered epithelial-mesenchymal transition. Biosci Rep 2019; 39:BSR20181165. [PMID: 30487162 PMCID: PMC6328857 DOI: 10.1042/bsr20181165] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Revised: 11/01/2018] [Accepted: 11/27/2018] [Indexed: 12/29/2022] Open
Abstract
Intratumoral hypoxia is a well-known feature of solid cancers and constitutes a major contributor to cancer metastasis and poor outcomes including melanoma. Leucine-rich repeats and Ig-like domains 1 (LRIG1) participate in the aggressive progression of several tumors, where its expression is frequently decreased. In the present study, hypoxia exposure aggravated melanoma cell invasion, migration, vasculogenic mimicry (VM), and epithelial–mesenchymal transition (EMT). During this process, LRIG1 expression was also decreased. Importantly, overexpression of LRIG1 notably counteracted hypoxia-induced invasion, migration, and VM, which was further augmented after LRIG1 inhibition. Mechanism analysis corroborated that LRIG1 elevation muted hypoxia-induced EMT by suppressing E-cadherin expression and increasing N-cadherin expression. Conversely, cessation of LRIG1 further potentiated hypoxia-triggered EMT. Additionally, hypoxia stimulation activated the epidermal growth factor receptor (EGFR)/ERK pathway, which was dampened by LRIG1 up-regulation but further activated by LRIG1 inhibition. More important, blocking this pathway with its antagonist erlotinib abrogated LRIG1 suppression-induced EMT, and subsequently cell invasion, migration, and VM of melanoma cells under hypoxia. Together, these findings suggest that LRIG1 overexpression can antagonize hypoxia-evoked aggressive metastatic phenotype by suppressing cell invasion, migration, and VM via regulating EGFR/ERK-mediated EMT process. Therefore, these findings may provide a promising target for melanoma therapy.
Collapse
|
7
|
Zhang Q, Shi W, Wang Q, Zhu Y, Zhai C, Wang J, Yan X, Chai L, Li M. Clinicopathological and prognostic significance of leucine-rich repeats and immunoglobulin-like domains protein 1 (LRIG1) in malignant tumors: A meta-analysis. J Cancer 2018; 9:2895-2909. [PMID: 30123358 PMCID: PMC6096372 DOI: 10.7150/jca.24749] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 06/09/2018] [Indexed: 01/11/2023] Open
Abstract
Background: Accumulating studies have demonstrated that the expression of leucine-rich repeats and immunoglobulin-like domains protein1 (LRIG1) is associated with various types of tumors. However, the conclusions of previous studies are not completely consistent. Thus, we conducted this meta-analysis to further explore the authentic value of LRIG1 in cancer outcome and clinical significance. Methods: We systematically searched electronic databases including PubMed, Web of Science, Embase, Chinese National Knowledge Infrastructure and Wanfang database. The hazard ratios (HRs), odds ratio (OR) and 95 % confidence intervals (CI) were calculated for effect measures. Results: 16 qualified studies involving 2043 patients with cancer were enrolled. High LRIG1 expression was associated with a good prognosis in malignant tumors (HR: 0.49, 95% CI=0.39-0.59). Furthermore, positive expression rate of LRIG1 was distinctly lower in cancer tissues than that in normal tissues (OR: 0.09, 95% CI=0.05-0.17). Positive LRIG1 expression was definitely related with smaller tumor size (OR: 1.64, 95% CI=1.11-2.42), early tumor stage (OR: 3.67, 95% CI=1.87-7.21), well degree of differentiation (OR: 4.35, 95% CI=2.12-8.93) and negative recurrence (OR: 0.29, 95% CI=0.16-0.53). Conclusions: LRIG1 expression was associated with a good prognosis in terms of overall survival (OS) and might act as a predictive factor for characteristics of cancer patients.
Collapse
Affiliation(s)
- Qianqian Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Wenhua Shi
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Qingting Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Yanting Zhu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Cui Zhai
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Jian Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Xin Yan
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Limin Chai
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Manxiang Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| |
Collapse
|
8
|
Expression of LRIG proteins as possible prognostic factors in primary vaginal carcinoma. PLoS One 2017; 12:e0183816. [PMID: 28841699 PMCID: PMC5571912 DOI: 10.1371/journal.pone.0183816] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 08/13/2017] [Indexed: 11/19/2022] Open
Abstract
Background Primary vaginal carcinoma (PVC) is a rare malignancy. Established prognostic factors include tumour stage and age at diagnosis. The leucine-rich repeats and immunoglobuline-like domains (LRIG)-1 protein functions as a tumour suppressor, but less is known about the functions of LRIG2 and LRIG3. The present study aimed to evaluate the expression of LRIG proteins and analyse their possible associations with clinical characteristics and survival in a cohort of PVC patients. Methods We used immunohistochemistry to investigate LRIG1, LRIG2, and LRIG3 expression in tumour samples from a consecutive cohort of 70 PVC patients. The association between LRIG protein expression and clinical characteristics and cancer-specific survival was investigated using univariate and multivariate analyses. Results The majority of PVC patients (72%) had >50% LRIG1- and LRIG2-positive cells, and no or low LRIG3-positive cells. HPV status was significantly correlated with LRIG1 expression (p = 0.0047). Having high LRIG1 expression was significantly correlated with superior cancer-specific survival in univariate and multivariate analyses. LRIG2 and LRIG3 expression did not significantly correlate with clinical characteristics or survival. Conclusion LRIG1 expression might be of interest as a prognostic marker in PVC patients, whereas the role of LRIG2 and LRIG3 expression remains to be clarified.
Collapse
|
9
|
Zhou M, Leung A, Echegaray S, Gentles A, Shrager JB, Jensen KC, Berry GJ, Plevritis SK, Rubin DL, Napel S, Gevaert O. Non-Small Cell Lung Cancer Radiogenomics Map Identifies Relationships between Molecular and Imaging Phenotypes with Prognostic Implications. Radiology 2017; 286:307-315. [PMID: 28727543 PMCID: PMC5749594 DOI: 10.1148/radiol.2017161845] [Citation(s) in RCA: 127] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Purpose To create a radiogenomic map linking computed tomographic (CT) image features and gene expression profiles generated by RNA sequencing for patients with non-small cell lung cancer (NSCLC). Materials and Methods A cohort of 113 patients with NSCLC diagnosed between April 2008 and September 2014 who had preoperative CT data and tumor tissue available was studied. For each tumor, a thoracic radiologist recorded 87 semantic image features, selected to reflect radiologic characteristics of nodule shape, margin, texture, tumor environment, and overall lung characteristics. Next, total RNA was extracted from the tissue and analyzed with RNA sequencing technology. Ten highly coexpressed gene clusters, termed metagenes, were identified, validated in publicly available gene-expression cohorts, and correlated with prognosis. Next, a radiogenomics map was built that linked semantic image features to metagenes by using the t statistic and the Spearman correlation metric with multiple testing correction. Results RNA sequencing analysis resulted in 10 metagenes that capture a variety of molecular pathways, including the epidermal growth factor (EGF) pathway. A radiogenomic map was created with 32 statistically significant correlations between semantic image features and metagenes. For example, nodule attenuation and margins are associated with the late cell-cycle genes, and a metagene that represents the EGF pathway was significantly correlated with the presence of ground-glass opacity and irregular nodules or nodules with poorly defined margins. Conclusion Radiogenomic analysis of NSCLC showed multiple associations between semantic image features and metagenes that represented canonical molecular pathways, and it can result in noninvasive identification of molecular properties of NSCLC. Online supplemental material is available for this article.
Collapse
Affiliation(s)
- Mu Zhou
- From the Stanford Center for Biomedical Informatics Research, Department of Medicine (M.Z., O.G.), Department of Radiology (A.L., S.E., A.G., S.K.P., D.L.R., S.N.), Division of Thoracic Surgery, Department of Cardiothoracic Surgery (J.B.S.), and Department of Pathology (K.C.J., G.J.B.), Stanford University, 1265 Welch Rd, Stanford, CA 94305-5479
| | - Ann Leung
- From the Stanford Center for Biomedical Informatics Research, Department of Medicine (M.Z., O.G.), Department of Radiology (A.L., S.E., A.G., S.K.P., D.L.R., S.N.), Division of Thoracic Surgery, Department of Cardiothoracic Surgery (J.B.S.), and Department of Pathology (K.C.J., G.J.B.), Stanford University, 1265 Welch Rd, Stanford, CA 94305-5479
| | - Sebastian Echegaray
- From the Stanford Center for Biomedical Informatics Research, Department of Medicine (M.Z., O.G.), Department of Radiology (A.L., S.E., A.G., S.K.P., D.L.R., S.N.), Division of Thoracic Surgery, Department of Cardiothoracic Surgery (J.B.S.), and Department of Pathology (K.C.J., G.J.B.), Stanford University, 1265 Welch Rd, Stanford, CA 94305-5479
| | - Andrew Gentles
- From the Stanford Center for Biomedical Informatics Research, Department of Medicine (M.Z., O.G.), Department of Radiology (A.L., S.E., A.G., S.K.P., D.L.R., S.N.), Division of Thoracic Surgery, Department of Cardiothoracic Surgery (J.B.S.), and Department of Pathology (K.C.J., G.J.B.), Stanford University, 1265 Welch Rd, Stanford, CA 94305-5479
| | - Joseph B Shrager
- From the Stanford Center for Biomedical Informatics Research, Department of Medicine (M.Z., O.G.), Department of Radiology (A.L., S.E., A.G., S.K.P., D.L.R., S.N.), Division of Thoracic Surgery, Department of Cardiothoracic Surgery (J.B.S.), and Department of Pathology (K.C.J., G.J.B.), Stanford University, 1265 Welch Rd, Stanford, CA 94305-5479
| | - Kristin C Jensen
- From the Stanford Center for Biomedical Informatics Research, Department of Medicine (M.Z., O.G.), Department of Radiology (A.L., S.E., A.G., S.K.P., D.L.R., S.N.), Division of Thoracic Surgery, Department of Cardiothoracic Surgery (J.B.S.), and Department of Pathology (K.C.J., G.J.B.), Stanford University, 1265 Welch Rd, Stanford, CA 94305-5479
| | - Gerald J Berry
- From the Stanford Center for Biomedical Informatics Research, Department of Medicine (M.Z., O.G.), Department of Radiology (A.L., S.E., A.G., S.K.P., D.L.R., S.N.), Division of Thoracic Surgery, Department of Cardiothoracic Surgery (J.B.S.), and Department of Pathology (K.C.J., G.J.B.), Stanford University, 1265 Welch Rd, Stanford, CA 94305-5479
| | - Sylvia K Plevritis
- From the Stanford Center for Biomedical Informatics Research, Department of Medicine (M.Z., O.G.), Department of Radiology (A.L., S.E., A.G., S.K.P., D.L.R., S.N.), Division of Thoracic Surgery, Department of Cardiothoracic Surgery (J.B.S.), and Department of Pathology (K.C.J., G.J.B.), Stanford University, 1265 Welch Rd, Stanford, CA 94305-5479
| | - Daniel L Rubin
- From the Stanford Center for Biomedical Informatics Research, Department of Medicine (M.Z., O.G.), Department of Radiology (A.L., S.E., A.G., S.K.P., D.L.R., S.N.), Division of Thoracic Surgery, Department of Cardiothoracic Surgery (J.B.S.), and Department of Pathology (K.C.J., G.J.B.), Stanford University, 1265 Welch Rd, Stanford, CA 94305-5479
| | - Sandy Napel
- From the Stanford Center for Biomedical Informatics Research, Department of Medicine (M.Z., O.G.), Department of Radiology (A.L., S.E., A.G., S.K.P., D.L.R., S.N.), Division of Thoracic Surgery, Department of Cardiothoracic Surgery (J.B.S.), and Department of Pathology (K.C.J., G.J.B.), Stanford University, 1265 Welch Rd, Stanford, CA 94305-5479
| | - Olivier Gevaert
- From the Stanford Center for Biomedical Informatics Research, Department of Medicine (M.Z., O.G.), Department of Radiology (A.L., S.E., A.G., S.K.P., D.L.R., S.N.), Division of Thoracic Surgery, Department of Cardiothoracic Surgery (J.B.S.), and Department of Pathology (K.C.J., G.J.B.), Stanford University, 1265 Welch Rd, Stanford, CA 94305-5479
| |
Collapse
|
10
|
Abstract
Background Sebaceous glands contribute significantly to the barrier functions of the skin. However, little is known about their homeostasis and tumorigenesis. Recently, increased expression of stem cell marker Lrig1 has been reported in sebaceous carcinoma-like tumors of K14ΔNLef1 transgenic mice. In this study, we analyzed the Lrig1 expression in human sebaceous tumors. Methods Twenty-eight formalin-fixed paraffin-embedded sebaceous tumor specimens (7 sebaceous hyperplasias, 7 sebaceous adenomas, 10 sebaceomas and 4 sebaceous carcinomas) were stained with anti-Lrig1, anti-CD44v3 and anti-Ki67 antibody. Results Four (100%) sebaceous carcinomas, 8 (80%) sebaceomas, 3 (43%) sebaceous adenomas and no sebaceous hyperplasia showed Lrig1 overexpression. Discussion and Conclusion Lrig1 is a known tumor suppressor gene and is usually considered to be an indicator of poorly aggressive tumors. In human sebaceous tumors, the stronger Lrig1 staining in sebaceous carcinoma compared to other sebaceous tumors might be a feature of an advanced stage in tumorigenesis and a bad prognosis. In our study, 100% of sebaceous carcinomas revealed Lrig1 overexpression. We propose that Lrig1 may be used as a possible new marker of poorly differentiated sebaceous carcinoma.
Collapse
Affiliation(s)
- Jöri Pünchera
- Department of Dermatology, University Hospital of Geneva, Geneva, Switzerland
| | - Laurent Barnes
- Department of Dermatology, University Hospital of Geneva, Geneva, Switzerland
| | - Gürkan Kaya
- Department of Dermatology, University Hospital of Geneva, Geneva, Switzerland
| |
Collapse
|