1
|
Israelsson P, Oda H, Öfverman C, Stefansson K, Lindquist D. Immunoreactivity of LMO7 and other molecular markers as potential prognostic factors in oropharyngeal squamous cell carcinoma. BMC Oral Health 2024; 24:729. [PMID: 38918827 PMCID: PMC11197244 DOI: 10.1186/s12903-024-04510-4] [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: 12/19/2023] [Accepted: 06/20/2024] [Indexed: 06/27/2024] Open
Abstract
BACKGROUND Despite the better prognosis associated with human papillomavirus (HPV)-positive oropharyngeal squamous cell carcinoma (OPSCC), some patients experience relapse and succumb to the disease; thus, there is a need for biomarkers identifying these patients for intensified treatment. Leucine-rich repeats and immunoglobulin-like domain (LRIG) protein 1 is a negative regulator of receptor tyrosine kinase signaling and a positive prognostic factor in OPSCC. Studies indicate that LRIG1 interacts with the LIM domain 7 protein (LMO7), a stabilizer of adherence junctions. Its role in OPSCC has not been studied before. METHODS A total of 145 patients diagnosed with OPSCC were enrolled. Immunohistochemical LMO7 expression and staining intensity were evaluated in the tumors and correlated with known clinical and pathological prognostic factors, such as HPV status and LRIG1, CD44, Ki67, and p53 expression. RESULTS Our results show that high LMO7 expression is associated with significantly longer overall survival (OS) (p = 0.044). LMO7 was a positive prognostic factor for OS in univariate analysis (HR 0.515, 95% CI: 0.267-0.994, p = 0.048) but not in multivariate analysis. The LMO7 expression correlated with LRIG1 expression (p = 0.048), consistent with previous findings. Interestingly, strong LRIG1 staining intensity was an independent negative prognostic factor in the HPV-driven group of tumors (HR 2.847, 95% Cl: 1.036-7.825, p = 0.043). CONCLUSIONS We show for the first time that high LMO7 expression is a positive prognostic factor in OPSCC, and we propose that LMO7 should be further explored as a biomarker. In contrast to previous reports, LRIG1 expression was shown to be an independent negative prognostic factor in HPV-driven OPSCC.
Collapse
Affiliation(s)
- Pernilla Israelsson
- Department of Diagnostics and Intervention, Oncology, Umeå University, Umeå, 90185, Sweden.
| | - Husam Oda
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, 90185, Sweden
| | - Charlotte Öfverman
- Department of Diagnostics and Intervention, Oncology, Umeå University, Umeå, 90185, Sweden
| | - Kristina Stefansson
- Department of Medical Biosciences, Clinical Chemistry, Umeå University, Umeå, 90185, Sweden
| | - David Lindquist
- Department of Clinical Sciences, Professional Development, Umeå University, Umeå, 90185, Sweden
| |
Collapse
|
2
|
Cheng S, Li C, Liu L, Liu X, Li M, Zhuo J, Wang J, Zheng W, Wang Z. Dysregulation and antimetastatic function of circLRIG1 modulated by miR-214-3p/LRIG1 axis in bladder carcinoma. Biol Direct 2024; 19:20. [PMID: 38454507 PMCID: PMC10918934 DOI: 10.1186/s13062-023-00446-x] [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: 09/12/2023] [Accepted: 12/19/2023] [Indexed: 03/09/2024] Open
Abstract
CircLRIG1, a newly discovered circRNA, has yet to have its potential function and biological processes reported. This study explored the role of circLRIG1 in the development and progression of bladder carcinoma and its potential molecular mechanisms. Techniques such as qRT-PCR, Western blot, various cellular assays, and in vivo models were used to investigate mRNA and protein levels, cell behavior, molecular interactions, and tumor growth. The results showed that both circLRIG1 and LRIG1 were significantly reduced in bladder carcinoma tissues and cell lines. Low circLRIG1 expression was associated with poor patient prognosis. Overexpressing circLRIG1 inhibited bladder carcinoma cell growth, migration, and invasion, promoted apoptosis, and decreased tumor growth and metastasis in vivo. Importantly, circLRIG1 was found to sponge miR-214-3p, enhancing LRIG1 expression, and its overexpression also modulated protein levels of E-cadherin, N-cadherin, Vimentin, and LRIG1. Similar effects were observed with LRIG1 overexpression. Notably, a positive correlation was found between circLRIG1 and LRIG1 expression in bladder carcinoma tissues. Additionally, the tumor-suppressing effect of circLRIG1 was reversed by overexpressing miR-214-3p or silencing LRIG1. The study concludes that circLRIG1 suppresses bladder carcinoma progression by enhancing LRIG1 expression via sponging miR-214-3p, providing a potential strategy for early diagnosis and treatment of bladder carcinoma.
Collapse
Affiliation(s)
- Shiliang Cheng
- Department of Clinical Laboratory, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan Xingqi Medical Laboratory Co., Ltd., 12 Wuyingshan Middle Road, Jinan, 250000, Shandong, China.
| | - Chunguang Li
- Department of Digestive Oncology, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital and Institute, 44 Xiaoheyan Road, ShenyangLiaoning, 110042, China
| | - Lu Liu
- Department of Digestive Oncology, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital and Institute, 44 Xiaoheyan Road, ShenyangLiaoning, 110042, China
| | - Xinli Liu
- Department of Clinical Laboratory, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, 12 Wuyingshan Middle Road, Jinan, 250000, Shandong, China
| | - Meng Li
- Department of Clinical Laboratory, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, 12 Wuyingshan Middle Road, Jinan, 250000, Shandong, China
| | - Jinhua Zhuo
- Department of Clinical Laboratory, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, 12 Wuyingshan Middle Road, Jinan, 250000, Shandong, China
| | - Jue Wang
- Department of Clinical Laboratory, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, 12 Wuyingshan Middle Road, Jinan, 250000, Shandong, China
| | - Wen Zheng
- Department of Emergency, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, 12 Wuyingshan Middle Road, Jinan, 250000, Shandong, China.
| | - Zhongmin Wang
- Department of Pharmacy, Shanghai Eastern Hepatobiliary Surgery Hospital, Navy Military Medical University, 225 Changhai Road, Shanghai, 200000, China.
| |
Collapse
|
3
|
Krop IE, Mittempergher L, Paulson JN, Andre F, Bonnefoi H, Loi S, Loibl S, Gelber RD, Caballero C, Bhaskaran R, Dreezen C, Menicucci AR, Bernards R, van 't Veer LJ, Piccart MJ. Prediction of Benefit From Adjuvant Pertuzumab by 80-Gene Signature in the APHINITY (BIG 4-11) Trial. JCO Precis Oncol 2024; 8:e2200667. [PMID: 38237097 DOI: 10.1200/po.22.00667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 03/30/2023] [Accepted: 05/04/2023] [Indexed: 01/23/2024] Open
Abstract
PURPOSE At the primary analysis, the APHINITY trial reported a statistically significant but modest benefit of adding pertuzumab to standard adjuvant chemotherapy plus trastuzumab in patients with histologically confirmed human epidermal growth factor receptor 2 (HER2)-positive early-stage breast cancer. This study evaluated whether the 80-gene molecular subtyping signature (80-GS) could identify patients within the APHINITY population who derive the most benefit from dual anti-HER2 therapy. METHODS In a nested case-control study design of 1,023 patients (matched event to control ratio of 3:1), the 80-GS classified breast tumors into functional luminal type, HER2 type, or basal type. Additionally, 80-GS distinguished tumor subtypes that exhibited a single-dominant functional pathway versus tumors with multiple activated pathways. The primary end point was invasive disease-free survival (IDFS). Hazard ratios (HRs) were evaluated by Cox regression. After excluding patients without appropriate consent and those with missing data, 964 patients were included. RESULTS The 80-GS classified 50% (n = 479) of tumors as luminal type, 28% (n = 275) as HER2 type, and 22% (n = 209) as basal type. Most luminal-type tumors (86%) displayed a single-activated pathway, whereas 49% of HER2-type and 42% of basal-type tumors were dual activated. There was no significant difference in IDFS among different conventional 80-GS subtypes (single- and dual-activated subtypes combined). However, basal single-subtype tumors were significantly more likely to have an IDFS event (hazard ratio, 1.69 [95% CI, 1.12 to 2.54]) compared with other subtypes. HER2 single-subtype tumors displayed a trend toward greater beneficial effect on the addition of pertuzumab (hazard ratio, 0.56 [95% CI, 0.27 to 1.16]) compared with all other subtypes. CONCLUSION The 80-GS identified subgroups of histologically confirmed HER2-positive tumors with distinct biological characteristics. Basal single-subtype tumors exhibit an inferior prognosis compared with other subgroups and may be candidates for additional therapeutic strategies. Preliminary results suggest patients with HER2-positive, genomically HER2 single-subtype tumors may particularly benefit from added pertuzumab, which warrants further investigation.
Collapse
Affiliation(s)
| | | | | | | | | | - Sherene Loi
- Peter MacCallum Cancer Centre, Melbourne, Australia
| | | | - Richard D Gelber
- Dana-Farber Cancer Institute, Harvard Medical School, Harvard TH Chan School of Public Health, and Frontier Science Foundation, Boston, MA
| | | | | | | | | | | | | | | |
Collapse
|
4
|
Umeh-Garcia M, O'Geen H, Simion C, Gephart MH, Segal DJ, Sweeney CA. Aberrant promoter methylation contributes to LRIG1 silencing in basal/triple-negative breast cancer. Br J Cancer 2022; 127:436-448. [PMID: 35440669 PMCID: PMC9346006 DOI: 10.1038/s41416-022-01812-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 03/16/2022] [Accepted: 03/29/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND LRIG1, the founding member of the LRIG (leucine-rich repeat and immunoglobulin-like domain) family of transmembrane proteins, is a negative regulator of receptor tyrosine kinases and a tumour suppressor. Decreased LRIG1 expression is consistently observed in cancer, across diverse tumour types, and is linked to poor patient prognosis. However, mechanisms by which LRIG1 is repressed are not fully understood. Silencing of LRIG1 through promoter CpG island methylation has been reported in colorectal and cervical cancer but studies in breast cancer remain limited. METHODS In silico analysis of human breast cancer patient data were used to demonstrate a correlation between DNA methylation and LRIG1 silencing in basal/triple-negative breast cancer, and its impact on patient survival. LRIG1 gene expression, protein abundance, and methylation enrichment were examined by quantitative reverse-transcription PCR, immunoblotting, and methylation immunoprecipitation, respectively, in breast cancer cell lines in vitro. We examined the impact of global demethylation on LRIG1 expression and methylation enrichment using 5-aza-2'-deoxycytidine. We also examined the effects of targeted demethylation of the LRIG1 CpG island, and transcriptional activation of LRIG1 expression, using the RNA guided deadCas9 transactivation system. RESULTS Across breast cancer subtypes, LRIG1 expression is lowest in the basal/triple-negative subtype so we investigated whether differential methylation may contribute to this. Indeed, we find that LRIG1 CpG island methylation is most prominent in basal/triple-negative cell lines and patient samples. Use of the global demethylating agent 5-aza-2'-deoxycytidine decreases methylation leading to increased LRIG1 transcript expression in basal/triple-negative cell lines, while having no effect on LRIG1 expression in luminal/ER-positive cell lines. Using a CRISPR/deadCas9 (dCas9)-based targeting approach, we demonstrate that TET1-mediated demethylation (Tet1-dCas9) along with VP64-mediated transcriptional activation (VP64-dCas9) at the CpG island, increased endogenous LRIG1 expression in basal/triple-negative breast cancer cells, without transcriptional upregulation at predicted off-target sites. Activation of LRIG1 by the dCas9 transactivation system significantly increased LRIG1 protein abundance, reduced site-specific methylation, and reduced cancer cell viability. Our findings suggest that CRISPR-mediated targeted activation may be a feasible way to restore LRIG1 expression in cancer. CONCLUSIONS Our study contributes novel insight into mechanisms which repress LRIG1 in triple-negative breast cancer and demonstrates for the first time that targeted de-repression of LRIG1 in cancer cells is possible. Understanding the epigenetic mechanisms associated with repression of tumour suppressor genes holds potential for the advancement of therapeutic approaches.
Collapse
Affiliation(s)
- Maxine Umeh-Garcia
- Department of Biochemistry and Molecular Medicine, University of California, Davis, CA, USA.
- Department Neurosurgery, Stanford University, Stanford, CA, USA.
| | | | - Catalina Simion
- Department of Biochemistry and Molecular Medicine, University of California, Davis, CA, USA
| | | | - David J Segal
- Department of Biochemistry and Molecular Medicine, University of California, Davis, CA, USA
- Genome Center, University of California, Davis, CA, USA
| | - Colleen A Sweeney
- Department of Biochemistry and Molecular Medicine, University of California, Davis, CA, USA.
| |
Collapse
|
5
|
Bakherad M, Salimi M, Angaji SA, Mahjoubi F, Majidizadeh T. LRIG1 expression and colorectal cancer prognosis. BMC Med Genomics 2021; 14:20. [PMID: 33461538 PMCID: PMC7814534 DOI: 10.1186/s12920-020-00846-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 12/02/2020] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND To make the right treatment decisions about colorectal cancer (CRC) patients reliable predictive and prognostic data are needed. However, in many cases this data is not enough. Some studies suggest that LRIG1 gene (leucine-rich repeats and immunoglobulin-like domains1) has prognostic implications in different kinds of cancers. METHODS One hundred and two patients with colorectal cancer were retrospectively analyzed for LRIG1 expression at both mRNA and protein levels. SYBR Green Real-Time RT-PCR technique was used for mRNA expression analyses and Glyceraldehyde-3-Phosphate Dehydrogenase gene (GAPDH) was considered as a reference gene for data normalization. LRIG1 protein expression was analyzed using Immunohistochemistry. Additionally, appropriate statistic analyses were used to assess the expression of LRIG1 in test and control groups. The prognostic significance of LRIG1 expression was analyzed using the univariate and multivariate analyses. RESULTS The data revealed that the expression of LRIG1 in both mRNA and protein levels was down regulated in colorectal tumor tissues (P < 0.01) but is not clinically relevant prognostic indicator in CRC. CONCLUSIONS Therefore, it is suggested that LRIG1 expression analyses may not be considered as an important issue when making informed and individualized clinical decisions regarding the management of colorectal cancer patients.
Collapse
Affiliation(s)
- Maryam Bakherad
- Department of Cell and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Mahdieh Salimi
- Department of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Seyed Abdolhamid Angaji
- Department of Cell and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Frouzandeh Mahjoubi
- Department of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Tayebeh Majidizadeh
- Department of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| |
Collapse
|
6
|
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
|
7
|
Faraz M, Tellström A, Ardnor CE, Grankvist K, Huminiecki L, Tavelin B, Henriksson R, Hedman H, Ljuslinder I. LRIG1 gene copy number analysis by ddPCR and correlations to clinical factors in breast cancer. BMC Cancer 2020; 20:459. [PMID: 32448168 PMCID: PMC7245921 DOI: 10.1186/s12885-020-06919-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 04/30/2020] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Leucine-rich repeats and immunoglobulin-like domains 1 (LRIG1) copy number alterations and unbalanced gene recombination events have been reported to occur in breast cancer. Importantly, LRIG1 loss was recently shown to predict early and late relapse in stage I-II breast cancer. METHODS We developed droplet digital PCR (ddPCR) assays for the determination of relative LRIG1 copy numbers and used these assays to analyze LRIG1 in twelve healthy individuals, 34 breast tumor samples previously analyzed by fluorescence in situ hybridization (FISH), and 423 breast tumor cytosols. RESULTS Four of the LRIG1/reference gene assays were found to be precise and robust, showing copy number ratios close to 1 (mean, 0.984; standard deviation, +/- 0.031) among the healthy control population. The correlation between the ddPCR assays and previous FISH results was low, possibly because of the different normalization strategies used. One in 34 breast tumors (2.9%) showed an unbalanced LRIG1 recombination event. LRIG1 copy number ratios were associated with the breast cancer subtype, steroid receptor status, ERBB2 status, tumor grade, and nodal status. Both LRIG1 loss and gain were associated with unfavorable metastasis-free survival; however, they did not remain significant prognostic factors after adjustment for common risk factors in the Cox regression analysis. Furthermore, LRIG1 loss was not significantly associated with survival in stage I and II cases. CONCLUSIONS Although LRIG1 gene aberrations may be important determinants of breast cancer biology, and prognostic markers, the results of this study do not verify an important role for LRIG1 copy number analyses in predicting the risk of relapse in early-stage breast cancer.
Collapse
Affiliation(s)
- Mahmood Faraz
- Department of Radiation Sciences, Oncology, Umeå University, SE-90187, Umeå, Sweden
| | - Andreas Tellström
- Department of Radiation Sciences, Oncology, Umeå University, SE-90187, Umeå, Sweden
| | | | - Kjell Grankvist
- Department of Medical Biosciences, Umeå University, SE-90187, Umeå, Sweden
| | - Lukasz Huminiecki
- National Bioinformatics Infrastructure Sweden, SciLifeLab, Uppsala, Sweden.,Current address: Instytut Genetyki i Hodowli Zwierząt Polskiej Akademii Nauk, ul. Postępu 36A, 05-552, Jastrzębiec, Magdalenka, Poland
| | - Björn Tavelin
- Department of Radiation Sciences, Oncology, Umeå University, SE-90187, Umeå, Sweden
| | - Roger Henriksson
- Department of Radiation Sciences, Oncology, Umeå University, SE-90187, Umeå, Sweden
| | - Håkan Hedman
- Department of Radiation Sciences, Oncology, Umeå University, SE-90187, Umeå, Sweden
| | - Ingrid Ljuslinder
- Department of Radiation Sciences, Oncology, Umeå University, SE-90187, Umeå, Sweden.
| |
Collapse
|
8
|
Li Q, Liu B, Chao HP, Ji Y, Lu Y, Mehmood R, Jeter C, Chen T, Moore JR, Li W, Liu C, Rycaj K, Tracz A, Kirk J, Calhoun-Davis T, Xiong J, Deng Q, Huang J, Foster BA, Gokhale A, Chen X, Tang DG. LRIG1 is a pleiotropic androgen receptor-regulated feedback tumor suppressor in prostate cancer. Nat Commun 2019; 10:5494. [PMID: 31792211 PMCID: PMC6889295 DOI: 10.1038/s41467-019-13532-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Accepted: 11/06/2019] [Indexed: 12/13/2022] Open
Abstract
LRIG1 has been reported to be a tumor suppressor in gastrointestinal tract and epidermis. However, little is known about the expression, regulation and biological functions of LRIG1 in prostate cancer (PCa). We find that LRIG1 is overexpressed in PCa, but its expression correlates with better patient survival. Functional studies reveal strong tumor-suppressive functions of LRIG1 in both AR+ and AR- xenograft models, and transgenic expression of LRIG1 inhibits tumor development in Hi-Myc and TRAMP models. LRIG1 also inhibits castration-resistant PCa and exhibits therapeutic efficacy in pre-established tumors. We further show that 1) AR directly transactivates LRIG1 through binding to several AR-binding sites in LRIG1 locus, and 2) LRIG1 dampens ERBB expression in a cell type-dependent manner and inhibits ERBB2-driven tumor growth. Collectively, our study indicates that LRIG1 represents a pleiotropic AR-regulated feedback tumor suppressor that functions to restrict oncogenic signaling from AR, Myc, ERBBs, and, likely, other oncogenic drivers.
Collapse
Affiliation(s)
- Qiuhui Li
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory for Oral Biomedicine of Ministry of Education (KLOBM), School and Hospital of Stomatology, Wuhan University, 430079, Wuhan, China
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Science Park, Smithville, TX, 78957, USA
| | - Bigang Liu
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Science Park, Smithville, TX, 78957, USA
| | - Hsueh-Ping Chao
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Science Park, Smithville, TX, 78957, USA
| | - Yibing Ji
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Yue Lu
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Science Park, Smithville, TX, 78957, USA
| | - Rashid Mehmood
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Collene Jeter
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Science Park, Smithville, TX, 78957, USA
| | - Taiping Chen
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Science Park, Smithville, TX, 78957, USA
| | - John R Moore
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Science Park, Smithville, TX, 78957, USA
| | - Wenqian Li
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Science Park, Smithville, TX, 78957, USA
| | - Can Liu
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Science Park, Smithville, TX, 78957, USA
| | - Kiera Rycaj
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Science Park, Smithville, TX, 78957, USA
| | - Amanda Tracz
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Jason Kirk
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Tammy Calhoun-Davis
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Science Park, Smithville, TX, 78957, USA
| | - Jie Xiong
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Science Park, Smithville, TX, 78957, USA
| | - Qu Deng
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Science Park, Smithville, TX, 78957, USA
| | - Jiaoti Huang
- Department of Pathology, Duke University of School of Medicine, Durham, NC, 27710, USA
| | - Barbara A Foster
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Abhiram Gokhale
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Xin Chen
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA.
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Science Park, Smithville, TX, 78957, USA.
- Department of Oncology, Tongji Hospital, Tongji Medical School, Huazhong University of Science and Technology (HUST), 430030, Wuhan, China.
| | - Dean G Tang
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA.
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Science Park, Smithville, TX, 78957, USA.
- Cancer Stem Cell Institute, Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, 200120, Shanghai, China.
| |
Collapse
|
9
|
Kim J, Jo YH, Jang M, Nguyen NNY, Yun HR, Ko SH, Shin Y, Lee JS, Kang I, Ha J, Choi TG, Kim SS. PAC-5 Gene Expression Signature for Predicting Prognosis of Patients with Pancreatic Adenocarcinoma. Cancers (Basel) 2019; 11:cancers11111749. [PMID: 31703415 PMCID: PMC6896100 DOI: 10.3390/cancers11111749] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 10/24/2019] [Accepted: 11/04/2019] [Indexed: 01/05/2023] Open
Abstract
Pancreatic adenocarcinoma (PAC) is one of the most aggressive malignancies. Intratumoural molecular heterogeneity impedes improvement of the overall survival rate. Current pathological staging system is not sufficient to accurately predict prognostic outcomes. Thus, accurate prognostic model for patient survival and treatment decision is demanded. Using differentially expressed gene analysis between normal pancreas and PAC tissues, the cancer-specific genes were identified. A prognostic gene expression model was computed by LASSO regression analysis. The PAC-5 signature (LAMA3, E2F7, IFI44, SLC12A2, and LRIG1) that had significant prognostic value in the overall dataset was established, independently of the pathological stage. We provided evidence that the PAC-5 signature further refined the selection of the PAC patients who might benefit from postoperative therapies. SLC12A2 and LRIG1 interacted with the proteins that were implicated in resistance of EGFR kinase inhibitor. DNA methylation was significantly involved in the gene regulations of the PAC-5 signature. The PAC-5 signature provides new possibilities for improving the personalised therapeutic strategies. We suggest that the PAC-5 genes might be potential drug targets for PAC.
Collapse
Affiliation(s)
- Jieun Kim
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Korea; (J.K.); (N.N.Y.N.); (H.R.Y.); (Y.S.); (I.K.); (J.H.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Korea; (Y.H.J.); (M.J.)
| | - Yong Hwa Jo
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Korea; (Y.H.J.); (M.J.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Korea
| | - Miran Jang
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Korea; (Y.H.J.); (M.J.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Korea
| | - Ngoc Ngo Yen Nguyen
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Korea; (J.K.); (N.N.Y.N.); (H.R.Y.); (Y.S.); (I.K.); (J.H.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Korea; (Y.H.J.); (M.J.)
| | - Hyeong Rok Yun
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Korea; (J.K.); (N.N.Y.N.); (H.R.Y.); (Y.S.); (I.K.); (J.H.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Korea; (Y.H.J.); (M.J.)
| | - Seok Hoon Ko
- Department of Emergency Medicine, School of Medicine, Kyung Hee University, Seoul 02447, Korea
| | - Yoonhwa Shin
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Korea; (J.K.); (N.N.Y.N.); (H.R.Y.); (Y.S.); (I.K.); (J.H.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Korea; (Y.H.J.); (M.J.)
| | - Ju-Seog Lee
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Insug Kang
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Korea; (J.K.); (N.N.Y.N.); (H.R.Y.); (Y.S.); (I.K.); (J.H.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Korea; (Y.H.J.); (M.J.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Korea
| | - Joohun Ha
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Korea; (J.K.); (N.N.Y.N.); (H.R.Y.); (Y.S.); (I.K.); (J.H.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Korea; (Y.H.J.); (M.J.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Korea
| | - Tae Gyu Choi
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Korea; (Y.H.J.); (M.J.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Korea
- Correspondence: (T.G.C.); (S.S.K.); Tel.: +82-961-028-7 (T.G.C.); +82-961-052-4 (S.S.K.)
| | - Sung Soo Kim
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Korea; (J.K.); (N.N.Y.N.); (H.R.Y.); (Y.S.); (I.K.); (J.H.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Korea; (Y.H.J.); (M.J.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Korea
- Correspondence: (T.G.C.); (S.S.K.); Tel.: +82-961-028-7 (T.G.C.); +82-961-052-4 (S.S.K.)
| |
Collapse
|
10
|
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
|
11
|
Karlsson T, Kvarnbrink S, Holmlund C, Botling J, Micke P, Henriksson R, Johansson M, Hedman H. LMO7 and LIMCH1 interact with LRIG proteins in lung cancer, with prognostic implications for early-stage disease. Lung Cancer 2018; 125:174-184. [PMID: 30429017 DOI: 10.1016/j.lungcan.2018.09.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 09/20/2018] [Accepted: 09/21/2018] [Indexed: 10/28/2022]
Abstract
OBJECTIVES The human leucine-rich repeats and immunoglobulin-like domains (LRIG) protein family comprises the integral membrane proteins LRIG1, LRIG2 and LRIG3. LRIG1 is frequently down-regulated in human cancer, and high levels of LRIG1 in tumor tissue are associated with favorable clinical outcomes in several tumor types including non-small cell lung cancer (NSCLC). Mechanistically, LRIG1 negatively regulates receptor tyrosine kinases and functions as a tumor suppressor. However, the details of the molecular mechanisms involved are poorly understood, and even less is known about the functions of LRIG2 and LRIG3. The aim of this study was to further elucidate the functions and molecular interactions of the LRIG proteins. MATERIALS AND METHODS A yeast two-hybrid screen was performed using a cytosolic LRIG3 peptide as bait. In transfected human cells, co-immunoprecipitation and co-localization experiments were performed. Proximity ligation assay was performed to investigate interactions between endogenously expressed proteins. Expression levels of LMO7 and LIMCH1 in normal and malignant lung tissue were investigated using qRT-PCR and through in silico analyses of public data sets. Finally, a clinical cohort comprising 355 surgically treated NSCLC cases was immunostained for LMO7. RESULTS In the yeast two-hybrid screen, the two paralogous proteins LMO7 and LIMCH1 were identified as interaction partners to LRIG3. LMO7 and LIMCH1 co-localized and co-immunoprecipitated with both LRIG1 and LRIG3. Endogenously expressed LMO7 was in close proximity of both LRIG1 and LRIG3. LMO7 and LIMCH1 were highly expressed in normal lung tissue and down-regulated in malignant lung tissue. LMO7 immunoreactivity was shown to be a negative prognostic factor in LRIG1 positive tumors, predicting poor patient survival. CONCLUSION These findings suggest that LMO7 and LIMCH1 physically interact with LRIG proteins and that expression of LMO7 is of clinical importance in NSCLC.
Collapse
Affiliation(s)
- Terese Karlsson
- Department of Radiation Sciences, Oncology, Umeå University, SE-901 87, Umeå, Sweden
| | - Samuel Kvarnbrink
- Department of Radiation Sciences, Oncology, Umeå University, SE-901 87, Umeå, Sweden.
| | - Camilla Holmlund
- Department of Radiation Sciences, Oncology, Umeå University, SE-901 87, Umeå, Sweden
| | - Johan Botling
- Department of Immunology, Genetics and Pathology, Molecular and Morphological Pathology, Uppsala University, SE-751 85, Uppsala, Sweden
| | - Patrick Micke
- Department of Immunology, Genetics and Pathology, Molecular and Morphological Pathology, Uppsala University, SE-751 85, Uppsala, Sweden
| | - Roger Henriksson
- Department of Radiation Sciences, Oncology, Umeå University, SE-901 87, Umeå, Sweden
| | - Mikael Johansson
- Department of Radiation Sciences, Oncology, Umeå University, SE-901 87, Umeå, Sweden
| | - Håkan Hedman
- Department of Radiation Sciences, Oncology, Umeå University, SE-901 87, Umeå, Sweden
| |
Collapse
|
12
|
Mizuguchi A, Takai A, Shimizu T, Matsumoto T, Kumagai K, Miyamoto S, Seno H, Marusawa H. Genetic features of multicentric/multifocal intramucosal gastric carcinoma. Int J Cancer 2018; 143:1923-1934. [DOI: 10.1002/ijc.31578] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 04/06/2018] [Accepted: 04/18/2018] [Indexed: 02/06/2023]
Affiliation(s)
- Aya Mizuguchi
- Department of Gastroenterology and HepatologyGraduate School of Medicine, Kyoto UniversityKyoto Japan
| | - Atsushi Takai
- Department of Gastroenterology and HepatologyGraduate School of Medicine, Kyoto UniversityKyoto Japan
| | - Takahiro Shimizu
- Department of Gastroenterology and HepatologyGraduate School of Medicine, Kyoto UniversityKyoto Japan
| | - Tomonori Matsumoto
- Department of Gastroenterology and HepatologyGraduate School of Medicine, Kyoto UniversityKyoto Japan
| | - Ken Kumagai
- Department of Gastroenterology and HepatologyGraduate School of Medicine, Kyoto UniversityKyoto Japan
| | - Shin'ichi Miyamoto
- Department of Gastroenterology and HepatologyGraduate School of Medicine, Kyoto UniversityKyoto Japan
| | - Hiroshi Seno
- Department of Gastroenterology and HepatologyGraduate School of Medicine, Kyoto UniversityKyoto Japan
| | - Hiroyuki Marusawa
- Department of Gastroenterology and HepatologyGraduate School of Medicine, Kyoto UniversityKyoto Japan
| |
Collapse
|
13
|
Zhou L, Li X, Zhou F, Jin Z, Chen D, Wang P, Zhang S, Zhuge Y, Shang Y, Zou X. Downregulation of leucine-rich repeats and immunoglobulin-like domains 1 by microRNA-20a modulates gastric cancer multidrug resistance. Cancer Sci 2018; 109:1044-1054. [PMID: 29450946 PMCID: PMC5891193 DOI: 10.1111/cas.13538] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 01/28/2018] [Accepted: 02/11/2018] [Indexed: 12/13/2022] Open
Abstract
Multidrug resistance (MDR) significantly restricts the clinical efficacy of gastric cancer (GC) chemotherapy, and it is critical to search novel targets to predict and overcome MDR. Leucine-rich repeats and immunoglobulin-like domains 1 (LRIG1) has been proved to be correlated with drug resistance in several cancers. The present study revealed that LRIG1 was overexpressed in chemosensitive GC tissues and decreased expression of LRIG1 predicted poor survival in GC patients. We observed that upregulation of LRIG1 enhanced chemosensitivity in GC cells. Interestingly, miR-20a, which was overexpressed in GC MDR cell lines and tissues, was identified to regulate LRIG1 expression by directly targeting its 3' untranslated region. We also found that inhibition of miR-20a suppressed GC MDR, and upregulation showed opposite effects. Moreover, we demonstrated that the miR-20a/LRIG1 axis regulated GC cell MDR through epidermal growth factor receptor (EGFR)-mediated PI3K/AKT and MAPK/ERK signaling pathways. Finally, LRIG1 expression in human GC tissues is inversely correlated with miR-20a and EGFR. Taken together, the newly identified miR-20a/LRIG1/EGFR link provides insight into the MDR process of GC, and targeting this axis represents a novel potential therapeutic strategy to block GC chemoresistance.
Collapse
Affiliation(s)
- Lin Zhou
- Department of GastroenterologyNanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNanjingChina
- Jiangsu Clinical Medical Center of Digestive DiseaseNanjingChina
| | - Xiaowei Li
- State Key Laboratory of Cancer Biology & Xijing Hospital of Digestive DiseasesXijing HospitalThe Fourth Military Medical UniversityXi'anChina
| | - Fan Zhou
- Department of GastroenterologyNanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNanjingChina
- Jiangsu Clinical Medical Center of Digestive DiseaseNanjingChina
| | - Zhi'an Jin
- The Second Outpatient Department of Chengdu Army Region AuthorityChengduChina
| | - Di Chen
- State Key Laboratory of Cancer Biology & Xijing Hospital of Digestive DiseasesXijing HospitalThe Fourth Military Medical UniversityXi'anChina
| | - Pin Wang
- Department of GastroenterologyNanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNanjingChina
- Jiangsu Clinical Medical Center of Digestive DiseaseNanjingChina
| | - Shu Zhang
- Department of GastroenterologyNanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNanjingChina
- Jiangsu Clinical Medical Center of Digestive DiseaseNanjingChina
| | - Yuzheng Zhuge
- Department of GastroenterologyNanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNanjingChina
- Jiangsu Clinical Medical Center of Digestive DiseaseNanjingChina
| | - Yulong Shang
- State Key Laboratory of Cancer Biology & Xijing Hospital of Digestive DiseasesXijing HospitalThe Fourth Military Medical UniversityXi'anChina
| | - Xiaoping Zou
- Department of GastroenterologyNanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNanjingChina
- Jiangsu Clinical Medical Center of Digestive DiseaseNanjingChina
| |
Collapse
|
14
|
Faraz M, Herdenberg C, Holmlund C, Henriksson R, Hedman H. A protein interaction network centered on leucine-rich repeats and immunoglobulin-like domains 1 (LRIG1) regulates growth factor receptors. J Biol Chem 2018; 293:3421-3435. [PMID: 29317492 PMCID: PMC5836135 DOI: 10.1074/jbc.m117.807487] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 12/29/2017] [Indexed: 12/11/2022] Open
Abstract
Leucine-rich repeats and immunoglobulin-like domains 1 (LRIG1) is a tumor suppressor and a negative regulator of several receptor tyrosine kinases. The molecular mechanisms by which LRIG1 mediates its tumor suppressor effects and regulates receptor tyrosine kinases remain incompletely understood. Here, we performed a yeast two-hybrid screen to identify novel LRIG1-interacting proteins and mined data from the BioPlex (biophysical interactions of ORFeome-based complexes) protein interaction data repository. The putative LRIG1 interactors identified in the screen were functionally evaluated using a triple co-transfection system in which HEK293 cells were co-transfected with platelet-derived growth factor receptor α, LRIG1, and shRNAs against the identified LRIG1 interactors. The effects of the shRNAs on the ability of LRIG1 to down-regulate platelet-derived growth factor receptor α expression were evaluated. On the basis of these results, we present an LRIG1 protein interaction network with many newly identified components. The network contains the apparently functionally important LRIG1-interacting proteins RAB4A, PON2, GAL3ST1, ZBTB16, LRIG2, CNPY3, HLA-DRA, GML, CNPY4, LRRC40, and LRIG3, together with GLRX3, PTPRK, and other proteins. In silico analyses of The Cancer Genome Atlas data sets revealed consistent correlations between the expression of the transcripts encoding LRIG1 and its interactors ZBTB16 and PTPRK and inverse correlations between the transcripts encoding LRIG1 and GLRX3. We further studied the LRIG1 function–promoting paraoxonase PON2 and found that it co-localized with LRIG1 in LRIG1-transfected cells. The proposed LRIG1 protein interaction network will provide leads for future studies aiming to understand the molecular functions of LRIG1 and the regulation of growth factor signaling.
Collapse
Affiliation(s)
- Mahmood Faraz
- From the Oncology Research Laboratory, Department of Radiation Sciences, Umeå University, SE-90187 Umeå, Sweden
| | - Carl Herdenberg
- From the Oncology Research Laboratory, Department of Radiation Sciences, Umeå University, SE-90187 Umeå, Sweden
| | - Camilla Holmlund
- From the Oncology Research Laboratory, Department of Radiation Sciences, Umeå University, SE-90187 Umeå, Sweden
| | - Roger Henriksson
- From the Oncology Research Laboratory, Department of Radiation Sciences, Umeå University, SE-90187 Umeå, Sweden
| | - Håkan Hedman
- From the Oncology Research Laboratory, Department of Radiation Sciences, Umeå University, SE-90187 Umeå, Sweden
| |
Collapse
|
15
|
Zhang Y, Liu Z, Yu S. Role and mechanism of action of LRIG1 in ovarian cancer cell line and VP16 drug-resistant cell line. Oncol Lett 2017; 14:4619-4624. [PMID: 28943962 PMCID: PMC5592861 DOI: 10.3892/ol.2017.6730] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 08/01/2017] [Indexed: 11/05/2022] Open
Abstract
We investigated the role of leucine-rich repeats and immunoglobulin-like domains (LRIG)-1 in ovarian cancer cell line and VP16 drug-resistant cell line to explore the possible mechanism of action. Human ovarian cancer cell line SKOV3 and the VP16 drug-resistant cell line SKOV3/VP16 were used to investigate whether LRIG1 affects the sensitivity of SKOV3 to drugs. RT-qPCR was used to detect the difference in LRIG1 expression between drug-resistant and wild-type cell lines. siRNA LRIG1 was designed and transfected to silence LRIG1 to investigate the mechanism by which LRIG1 affects the sensitivity of SKOV3 to drugs. Wild-type cells were transfected with SKOV3. The cells were divided into 3 groups (VP16, NC + VP16 and siRNA LRIG1 + VP16 treatment group). VP16 (IC50 value) was added 24 h after transfection. The CCK-8 method was used to detect the proliferation of each group at multiple time points (0, 24, 48 and 72 h). A colony-forming assay was used to detect cell proliferation and flow cytometry was used to detect cell apoptosis. The expression of LRIG1 was lower in the drug resistant cell line than that of the wild-type cell line. The expression of LRIG1 significantly decreased with the increase of VP16 concentration (P<0.05). The apoptotic rate was decreased but there was an increase on cell clones in the siLRIG1 + VP16-treated group as compared to VP16- and NC+ VP16-treated groups (P<0.05). The LRIG1 gene affects the sensitivity of SKOV3 cells to drug in a dose-related manner, indicating that the reduced expression of LRIG1 can inhibit cell apoptosis.
Collapse
Affiliation(s)
- Yaqi Zhang
- Department of Gynecology, Yidu Central Hospital of Weifang, Weifang, Shandong 262500, P.R. China
| | - Zhizhen Liu
- Department of Gynecology, Yidu Central Hospital of Weifang, Weifang, Shandong 262500, P.R. China
| | - Shunrui Yu
- Department of Gynecology, Yidu Central Hospital of Weifang, Weifang, Shandong 262500, P.R. China
| |
Collapse
|
16
|
Yang B, Dai C, Tan R, Zhang B, Meng X, Ye J, Wang X, Wei L, He F, Chen Z. Lrig1 is a positive prognostic marker in hepatocellular carcinoma. Onco Targets Ther 2016; 9:7071-7079. [PMID: 27895499 PMCID: PMC5117876 DOI: 10.2147/ott.s112534] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Background The prevalence of hepatocellular carcinoma (HCC) is increasing worldwide. As a consequence, there is an urgent need for identifying molecular markers of HCC development and progression. Recently, several studies have suggested that the Lrig1 may have prognostic implications in various cancer types, but its clinical value in HCC is not well evaluated. Materials and methods In this study, the expression level of Lrig1 was examined in 133 HCC tissues and adjacent normal tissues by immunohistochemistry. Furthermore, potential associations between Lrig1 expression and the carcinoma clinical parameters were investigated, including recurrence and survival rate. We silenced the Lrig1 in the normal liver cell line (LO2) and liver cancer cell line (Hep-G2) in vitro by the small interference RNA and detected its influence on proliferation, migration, and invasion. Results The expression of Lrig1 was significantly downregulated in liver cancer tissues and cell lines, and its expression levels were related to tumor size, tumor–node–metastasis staging and tumor recurrence. Furthermore, analysis of 6-year survival of 133 HCC patients showed that those with stronger Lrig1 expression had significantly longer overall survival time than those with weaker Lrig1 expression. In addition, decreased expression of Lrig1 in vitro promoted the growth, migration, or invasion of normal liver cells and cancer cells. Conclusion Our findings demonstrate that Lrig1 could serve as a potential marker in the prognosis of patients with HCC. We also revealed that Lrig1 might be involved in the metastatic progression of liver cancer. However, its clinical value should be further investigated in the future.
Collapse
Affiliation(s)
- Bo Yang
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China; Key Laboratory of Ministry of Health and Key Laboratory of Ministry of Education, Wuhan, Hubei, People's Republic of China
| | - Chen Dai
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China; Key Laboratory of Ministry of Health and Key Laboratory of Ministry of Education, Wuhan, Hubei, People's Republic of China
| | - Rumeng Tan
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China; Key Laboratory of Ministry of Health and Key Laboratory of Ministry of Education, Wuhan, Hubei, People's Republic of China
| | - Bo Zhang
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China; Key Laboratory of Ministry of Health and Key Laboratory of Ministry of Education, Wuhan, Hubei, People's Republic of China
| | - Xiao Meng
- Department of Pathology, Liaocheng People's Hospital, Liaocheng, Shandong, People's Republic of China
| | - Jing Ye
- Department of Pathology, Liaocheng People's Hospital, Liaocheng, Shandong, People's Republic of China
| | - Xinqiang Wang
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China; Key Laboratory of Ministry of Health and Key Laboratory of Ministry of Education, Wuhan, Hubei, People's Republic of China
| | - Lai Wei
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China; Key Laboratory of Ministry of Health and Key Laboratory of Ministry of Education, Wuhan, Hubei, People's Republic of China
| | - Fan He
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Zhishui Chen
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China; Key Laboratory of Ministry of Health and Key Laboratory of Ministry of Education, Wuhan, Hubei, People's Republic of China
| |
Collapse
|
17
|
Lando M, Fjeldbo CS, Wilting SM, C Snoek B, Aarnes EK, Forsberg MF, Kristensen GB, Steenbergen RD, Lyng H. Interplay between promoter methylation and chromosomal loss in gene silencing at 3p11-p14 in cervical cancer. Epigenetics 2016; 10:970-80. [PMID: 26291246 DOI: 10.1080/15592294.2015.1085140] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Loss of 3p11-p14 is a frequent event in epithelial cancer and a candidate prognostic biomarker in cervical cancer. In addition to loss, promoter methylation can participate in gene silencing and promote tumor aggressiveness. We have performed a complete mapping of promoter methylation at 3p11-p14 in two independent cohorts of cervical cancer patients (n = 149, n = 121), using Illumina 450K methylation arrays. The aim was to investigate whether hyperm-ethylation was frequent and could contribute to gene silencing and disease aggressiveness either alone or combined with loss. By comparing the methylation level of individual CpG sites with corresponding data of normal cervical tissue, 26 out of 41 genes were found to be hypermethylated in both cohorts. The frequency of patients with hypermethylation of these genes was found to be higher at tumor stages of 3 and 4 than in stage 1 tumors. Seventeen of the 26 genes were transcriptionally downregulated in cancer compared to normal tissue, whereof 6 genes showed a significant correlation between methylation and expression. Integrated analysis of methylation, gene dosage, and expression of the 26 hypermethylated genes identified 3 regulation patterns encompassing 8 hypermethylated genes; a methylation driven pattern (C3orf14, GPR27, ZNF717), a gene dosage driven pattern (THOC7, PSMD6), and a combined methylation and gene dosage driven pattern (FHIT, ADAMTS9, LRIG1). In survival analysis, patients with both hypermethylation and loss of LRIG1 had a worse outcome compared to those harboring only hypermethylation or none of the events. C3orf14 emerged as a novel methylation regulated suppressor gene, for which knockdown was found to promote invasive growth in human papilloma virus (HPV)-transformed keratinocytes. In conclusion, hypermethylation at 3p11-p14 is common in cervical cancer and may exert a selection pressure during carcinogenesis alone or combined with loss. Information on both events could lead to improved prognostic markers.
Collapse
Affiliation(s)
- Malin Lando
- a Department of Radiation Biology ; Norwegian Radium Hospital; Oslo University Hospital ; Oslo , Norway
| | - Christina S Fjeldbo
- a Department of Radiation Biology ; Norwegian Radium Hospital; Oslo University Hospital ; Oslo , Norway
| | - Saskia M Wilting
- b Department of Pathology ; VU University Medical Center ; Amsterdam , the Netherlands
| | - Barbara C Snoek
- b Department of Pathology ; VU University Medical Center ; Amsterdam , the Netherlands
| | - Eva-Katrine Aarnes
- a Department of Radiation Biology ; Norwegian Radium Hospital; Oslo University Hospital ; Oslo , Norway
| | - Malin F Forsberg
- a Department of Radiation Biology ; Norwegian Radium Hospital; Oslo University Hospital ; Oslo , Norway
| | - Gunnar B Kristensen
- c Department of Gynecologic Oncology ; Norwegian Radium Hospital; Oslo University Hospital ; Oslo , Norway.,d Institute for Cancer Genetics and Informatics; Oslo University Hospital ; Oslo , Norway.,e Faculty of Medicine; University of Oslo ; Oslo , Norway
| | - Renske Dm Steenbergen
- b Department of Pathology ; VU University Medical Center ; Amsterdam , the Netherlands
| | - Heidi Lyng
- a Department of Radiation Biology ; Norwegian Radium Hospital; Oslo University Hospital ; Oslo , Norway
| |
Collapse
|
18
|
Joe S, Nam H. Prognostic factor analysis for breast cancer using gene expression profiles. BMC Med Inform Decis Mak 2016; 16 Suppl 1:56. [PMID: 27454576 PMCID: PMC4959370 DOI: 10.1186/s12911-016-0292-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The survival of patients with breast cancer is highly sporadic, from a few months to more than 15 years. In recent studies, the gene expression profiling of tumors has been used as a promising means of predicting prognosis factors. METHODS In this study, we used gene expression datasets of tumors to identify prognostic factors in breast cancer. We conducted log-rank tests and used unsupervised clustering methods to find reciprocally expressed gene sets associated with worse survival rates. Prognosis prediction scores were determined as the ratio of gene expressions. RESULTS As a result, four prognosis prediction gene set modules were constructed. The four prognostic gene sets predicted worse survival rates in three independent gene expression data sets. In addition, we found that cancer patient with poor prognosis, i.e., triple-negative cancer, HER2-enriched, TP53 mutated and high-graded patients had higher prognosis prediction scores than those with other types of breast cancer. CONCLUSIONS In conclusion, based on a gene expression analysis, we suggest that our well-defined scoring method of the prediction of survival outcome may be useful for developing prognostic factors in breast cancer.
Collapse
Affiliation(s)
- Soobok Joe
- School of Electrical Engineering and Computer Science, Gwangju Institute of Science and Technology, 123 Cheomdangwagi-ro, Buk-gu, Gwangju, Republic of Korea
| | - Hojung Nam
- School of Electrical Engineering and Computer Science, Gwangju Institute of Science and Technology, 123 Cheomdangwagi-ro, Buk-gu, Gwangju, Republic of Korea.
| |
Collapse
|
19
|
Singh RR, Mehrotra M, Chen H, Almohammedsalim AA, Sahin A, Bosamra A, Patel KP, Routbort MJ, Lu X, Ronald A, Mishra BM, Virani S, Medeiros LJ, Luthra R. Comprehensive Screening of Gene Copy Number Aberrations in Formalin-Fixed, Paraffin-Embedded Solid Tumors Using Molecular Inversion Probe-Based Single-Nucleotide Polymorphism Array. J Mol Diagn 2016; 18:676-687. [PMID: 27392636 DOI: 10.1016/j.jmoldx.2016.03.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Revised: 02/23/2016] [Accepted: 03/29/2016] [Indexed: 02/08/2023] Open
Abstract
Gene copy number aberrations (CNAs) represent a major class of cancer-related genomic alterations that drive solid tumors. Comprehensive and sensitive detection of CNAs is challenging because of often low quality and quantity of DNA isolated from the formalin-fixed, paraffin-embedded (FFPE) solid tumor samples. Here, in a clinical molecular diagnostic laboratory, we tested the utility and validated a molecular inversion probe-based (MIP) array to routinely screen for CNAs in solid tumors. Using low-input FFPE DNA, the array detects genome-wide CNAs with a special focus on 900 cancer-related genes. A cohort of 76 solid tumors of various types and tumor cellularity (20% to 100%), and four cancer cell lines were used. These harbored CNAs in clinically important genes (ERBB2, EGFR, FGFR1, KRAS, MYC) as detected by orthogonal techniques like next-generation sequencing or fluorescence in situ hybridization. Results of the MIP array were concordant with results from orthogonal techniques, and also provided additional information regarding the allelic nature of the CNAs. Limit-of-detection and assay reproducibility studies showed a high degree of sensitivity and reproducibility of detection, respectively. FFPE compatibility, ability to detect CNAs with high sensitivity, accuracy, and provide valuable information such as loss of heterozygosity along with relatively short turnaround times makes the MIP array a desirable clinical platform for routine screening of solid tumors in a clinical laboratory.
Collapse
Affiliation(s)
- Rajesh R Singh
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Meenakshi Mehrotra
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hui Chen
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Alaa A Almohammedsalim
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ayesagul Sahin
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Alex Bosamra
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Keyur P Patel
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mark J Routbort
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Xinyan Lu
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Abraham Ronald
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Bal Mukund Mishra
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Shumaila Virani
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - L Jeffrey Medeiros
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Rajyalakshmi Luthra
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
20
|
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
|
21
|
Wang Y, Shi C, Lu Y, Poulin EJ, Franklin JL, Coffey RJ. Loss of Lrig1 leads to expansion of Brunner glands followed by duodenal adenomas with gastric metaplasia. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 185:1123-34. [PMID: 25794708 DOI: 10.1016/j.ajpath.2014.12.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Revised: 12/16/2014] [Accepted: 12/23/2014] [Indexed: 01/15/2023]
Abstract
Leucine-rich repeats and immunoglobulin-like domains 1 (LRIG1) is a pan-ErbB negative regulator and intestinal stem cell marker down-regulated in many malignancies. We previously reported that 14 of 16 Lrig1-CreERT2/CreERT2 (Lrig1(-/-)) mice developed duodenal adenomas, providing the first in vivo evidence that Lrig1 acts as a tumor suppressor. We extended this study to a larger cohort and found that 49 of 54 Lrig1(-/-) mice develop duodenal adenomas beginning at 3 months. Most adenomas were histologically low grade and overlaid expanded Brunner glands. There was morphologic and biochemical blurring of the boundary between the epithelium and Brunner glands with glandular coexpression of ErbB2, which is normally restricted to the epithelium, and the Brunner gland marker Mucin6. Some adenomas were high grade with reduced Brunner glands. At age 4 to 5 weeks, before adenoma formation, we observed enhanced proliferation in Brunner glands and, at 2 months, an increase in the size of the Brunner gland compartment. Elevated expression of the epidermal growth factor receptor (Egfr) ligands amphiregulin and β-cellulin, as well as Egfr and phosphorylated Egfr, was detected in adenomas compared with adjacent normal tissue. These adenomas expressed the gastric-specific genes gastrokine1 and mucin5ac, indicating gastric metaplasia. Moreover, we found that a subset of human duodenal tumors exhibited features of LRIG1(-/-) adenomas, including loss of LRIG1, gastric metaplasia (MUCIN5AC and MUCIN6), and increased amphiregulin and Egfr activity.
Collapse
Affiliation(s)
- Yang Wang
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Chanjuan Shi
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Yuanyuan Lu
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Emily J Poulin
- Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Jeffery L Franklin
- Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Robert J Coffey
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee; Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, Tennessee; Department of Veterans Affairs Medical Center, Nashville, Tennessee.
| |
Collapse
|
22
|
Yokdang N, Hatakeyama J, Wald JH, Simion C, Tellez JD, Chang DZ, Swamynathan MM, Chen M, Murphy WJ, Carraway Iii KL, Sweeney C. LRIG1 opposes epithelial-to-mesenchymal transition and inhibits invasion of basal-like breast cancer cells. Oncogene 2015; 35:2932-47. [PMID: 26387542 PMCID: PMC4805527 DOI: 10.1038/onc.2015.345] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Revised: 06/24/2015] [Accepted: 08/04/2015] [Indexed: 01/04/2023]
Abstract
LRIG1, a member of the LRIG family of transmembrane leucine rich repeat-containing proteins, is a negative regulator of receptor tyrosine kinase signaling and a tumor suppressor. LRIG1 expression is broadly decreased in human cancer and in breast cancer, low expression of LRIG1 has been linked to decreased relapse-free survival. Recently, low expression of LRIG1 was revealed to be an independent risk factor for breast cancer metastasis and death. These findings suggest that LRIG1 may oppose breast cancer cell motility and invasion, cellular processes which are fundamental to metastasis. However, very little is known of LRIG1 function in this regard. In this study, we demonstrate that LRIG1 is down-regulated during epithelial to mesenchymal transition (EMT) of human mammary epithelial cells, suggesting that LRIG1 expression may represent a barrier to EMT. Indeed, depletion of endogenous LRIG1 in human mammary epithelial cells expands the stem cell population, augments mammosphere formation and accelerates EMT. Conversely, expression of LRIG1 in highly invasive Basal B breast cancer cells provokes a mesenchymal to epithelial transition accompanied by a dramatic suppression of tumorsphere formation and a striking loss of invasive growth in three-dimensional culture. LRIG1 expression perturbs multiple signaling pathways and represses markers and effectors of the mesenchymal state. Furthermore, LRIG1 expression in MDA-MB-231 breast cancer cells significantly slows their growth as tumors, providing the first in vivo evidence that LRIG1 functions as a growth suppressor in breast cancer.
Collapse
Affiliation(s)
- N Yokdang
- Department of Biochemistry and Molecular Medicine, University of California, Davis, Sacramento, CA, USA
| | - J Hatakeyama
- Department of Biochemistry and Molecular Medicine, University of California, Davis, Sacramento, CA, USA
| | - J H Wald
- Department of Biochemistry and Molecular Medicine, University of California, Davis, Sacramento, CA, USA
| | - C Simion
- Department of Biochemistry and Molecular Medicine, University of California, Davis, Sacramento, CA, USA
| | - J D Tellez
- Department of Pathology and Laboratory Medicine, University of California, Davis, Sacramento, CA, USA
| | - D Z Chang
- Department of Biochemistry and Molecular Medicine, University of California, Davis, Sacramento, CA, USA
| | - M M Swamynathan
- Department of Biochemistry and Molecular Medicine, University of California, Davis, Sacramento, CA, USA
| | - M Chen
- Department of Pathology and Laboratory Medicine, University of California, Davis, Sacramento, CA, USA
| | - W J Murphy
- Department of Dermatology, University of California, Davis, Sacramento, CA, USA
| | - K L Carraway Iii
- Department of Biochemistry and Molecular Medicine, University of California, Davis, Sacramento, CA, USA
| | - C Sweeney
- Department of Biochemistry and Molecular Medicine, University of California, Davis, Sacramento, CA, USA
| |
Collapse
|
23
|
Kvarnbrink S, Karlsson T, Edlund K, Botling J, Lindquist D, Jirström K, Micke P, Henriksson R, Johansson M, Hedman H. LRIG1 is a prognostic biomarker in non-small cell lung cancer. Acta Oncol 2015; 54:1113-9. [PMID: 25813475 DOI: 10.3109/0284186x.2015.1021427] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND The leucine-rich repeats and immunoglobulin-like domains (LRIG) family of transmembrane proteins are involved in the regulation of cellular signal transduction. LRIG1 is an endogenous inhibitor of receptor tyrosine kinases (RTKs) and an emerging tumor suppressor. In the lung epithelium, the expression of LRIG1 is downregulated by tobacco smoking, and further downregulated in lung squamous cell carcinoma. MATERIAL AND METHODS The expression of LRIG proteins were analyzed in 347 cases of non-small cell lung cancer (NSCLC) by immunohistochemistry, and LRIG1 mRNA expression was evaluated in 807 lung cancer samples in silico in the Oncomine database. Potential associations between the expression data and the clinical parameters, including patient survival, were investigated. RESULTS Expression of the LRIG1 protein was found to be an independent prognostic factor in NSCLC, whereas expression of LRIG2 or LRIG3 did not correlate with patient survival. The levels of LRIG1 mRNA also correlated with the survival of NSCLC patients. CONCLUSION These findings demonstrate that LRIG1 is an independent prognostic factor in patients with NSCLC that could be important in future decision-making algorithms for adjuvant lung cancer treatment.
Collapse
Affiliation(s)
- Samuel Kvarnbrink
- a Department of Radiation Sciences , Oncology, Umeå University , Umeå , Sweden
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Hu Z, Zhu L, Tan M, Cai M, Deng L, Yu G, Liu D, Liu J, Lin B. The expression and correlation between the transcription factor FOXP1 and estrogen receptors in epithelial ovarian cancer. Biochimie 2014; 109:42-8. [PMID: 25500588 DOI: 10.1016/j.biochi.2014.12.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 12/03/2014] [Indexed: 01/08/2023]
Abstract
BACKGROUND Estrogen plays an important role in the progression of ovarian cancer in humans. FOXP1 belongs to the forkhead/winged-helix transcription factor family, and previous research indicated that FOXP1 functioned as a tumor suppressor gene. FOXP1 may be similar to FOXA1 and is closely related to steroid hormone receptors, but the relationship between FOXP1 and ER currently remains unclear. METHODS Ovarian tumors (60 malignant cases, 26 borderline cases, and 13 benign cases) and 14 normal ovarian tissues were collected retrospectively. Immunohistochemistry, western blotting and real-time PCR were used to characterize the expression patterns of FOXP1, ERα, and ERβ both at the mRNA and protein levels. We also used co-immunoprecipitation and immunofluorescent colocalization to investigate whether a correlation exists between FOXP1 and ERα/ERβ in ovarian cancer tissues. RESULTS The mRNA level for FOXP1 and ERβ in ovarian carcinoma tissues decreased, while the expression level of ERα mRNA increased compared with normal ovarian tissues. With an increase in the degree of ovarian carcinoma malignancy, the ERα expression level also increased. The expression pattern of ERβ in ovarian neoplasms was similar to that of the FOXP1 protein; presenting nuclear staining decreased, while cytoplasmic expression increased. Colocalization of FOXP1, ERα, and ERβ was present in the cytoplasm, with ERβ specific co-localization with FOXP1 in the perinuclear area. While immunoprecipitates created with FOXP1 mouse anti-human monoclonal antibody showed a positive reaction to an anti-ER antibody, immunoprecipitates containing anti-ER antibody and react to anti-FOXP1 antibody. CONCLUSION Interactions between FOXP1 and ER may play a pivotal role in the progression of ovarian cancer, and the activation or induction of FOXP1 and ERβ expression in cancer cells may inhibit tumor proliferation.
Collapse
Affiliation(s)
- Zhenhua Hu
- Department of Obstetrics and Gynecology, Shengjing Hospital Affiliated to China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, Liaoning Province 110004, PR China; Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province 450052, PR China
| | - Liancheng Zhu
- Department of Obstetrics and Gynecology, Shengjing Hospital Affiliated to China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, Liaoning Province 110004, PR China
| | - Mingzi Tan
- Department of Obstetrics and Gynecology, Shengjing Hospital Affiliated to China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, Liaoning Province 110004, PR China
| | - Mingbo Cai
- Department of Obstetrics and Gynecology, Shengjing Hospital Affiliated to China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, Liaoning Province 110004, PR China
| | - Lu Deng
- Department of Obstetrics and Gynecology, Shengjing Hospital Affiliated to China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, Liaoning Province 110004, PR China
| | - Guannan Yu
- Department of Obstetrics and Gynecology, Shengjing Hospital Affiliated to China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, Liaoning Province 110004, PR China
| | - Dawo Liu
- Department of Obstetrics and Gynecology, Shengjing Hospital Affiliated to China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, Liaoning Province 110004, PR China
| | - Juanjuan Liu
- Department of Obstetrics and Gynecology, Shengjing Hospital Affiliated to China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, Liaoning Province 110004, PR China
| | - Bei Lin
- Department of Obstetrics and Gynecology, Shengjing Hospital Affiliated to China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, Liaoning Province 110004, PR China.
| |
Collapse
|
25
|
Abstract
BACKGROUND Optimal treatment decisions for cancer patients require reliable prognostic and predictive information. However, this information is inadequate in many cases. Several recent studies suggest that the leucine-rich repeats and immunoglobulin-like domains (LRIG) genes, transcripts, and proteins have prognostic implications in various cancer types. MATERIAL AND METHODS Relevant literature was identified on PubMed using the key words lrig1, lrig2, and lrig3. LRIG mRNA expression in cancer versus normal tissues was investigated using the Oncomine database. RESULTS The three human LRIG genes, LRIG1, LRIG2, and LRIG3, encode single-pass transmembrane proteins. LRIG1 is a negative regulator of growth factor signaling that has been shown to function as a tumor suppressor in vitro and in vivo in mice. The functions of LRIG2 and LRIG3 are less well defined. LRIG gene and protein expression are commonly dysregulated in human cancer. In early stage breast cancer, LRIG1 copy number was recently shown to predict early and late relapse in addition to overall survival; in nasopharyngeal carcinoma, loss of LRIG1 is also associated with poor survival. LRIG gene and protein expression have prognostic value in breast cancer, uterine cervical cancer, head-and-neck cancer, glioma, non-small cell lung cancer, prostate cancer, and cutaneous squamous cell carcinoma. In general, expression of LRIG1 and LRIG3 is associated with good survival, whereas expression of LRIG2 is associated with poor survival. Additionally, LRIG1 regulates cellular sensitivity to anti-cancer drugs, which indicates a possible role as a predictive marker. CONCLUSIONS LRIG gene statuses and mRNA and protein expression are clinically relevant prognostic indicators in several types of human cancer. We propose that LRIG analyses could become important when making informed and individualized clinical decisions regarding the management of cancer patients.
Collapse
MESH Headings
- Animals
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Breast Neoplasms/genetics
- Breast Neoplasms/metabolism
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/metabolism
- Carcinoma, Non-Small-Cell Lung/mortality
- Carcinoma, Squamous Cell/genetics
- Carcinoma, Squamous Cell/metabolism
- Carcinoma, Squamous Cell/mortality
- Down-Regulation
- Female
- Gene Expression
- Genes, Tumor Suppressor
- Glioma/genetics
- Glioma/metabolism
- Glioma/mortality
- Head and Neck Neoplasms/genetics
- Head and Neck Neoplasms/metabolism
- Humans
- Lung Neoplasms/genetics
- Lung Neoplasms/metabolism
- Lung Neoplasms/mortality
- Male
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/metabolism
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Mice
- Nasopharyngeal Neoplasms/genetics
- Nasopharyngeal Neoplasms/metabolism
- Neoplasm Proteins/genetics
- Neoplasm Proteins/metabolism
- Neoplasm Recurrence, Local
- Neoplasms/genetics
- Neoplasms/metabolism
- Neoplasms/mortality
- Prognosis
- RNA, Messenger/metabolism
- Up-Regulation
- Uterine Cervical Neoplasms/genetics
- Uterine Cervical Neoplasms/metabolism
Collapse
Affiliation(s)
- David Lindquist
- Oncology Research Laboratory, Department of Radiation Sciences, Umeå University, Umeå, Sweden
| | - Samuel Kvarnbrink
- Oncology Research Laboratory, Department of Radiation Sciences, Umeå University, Umeå, Sweden
| | - Roger Henriksson
- Oncology Research Laboratory, Department of Radiation Sciences, Umeå University, Umeå, Sweden
| | - Håkan Hedman
- Oncology Research Laboratory, Department of Radiation Sciences, Umeå University, Umeå, Sweden
- Correspondence: H. Hedman, Oncology Research Laboratory, Department of Radiation Sciences, Umeå University, SE-90187 Umeå, Sweden. Tel: + 46 90 785 2881. E-mail:
| |
Collapse
|
26
|
Simion C, Cedano-Prieto ME, Sweeney C. The LRIG family: enigmatic regulators of growth factor receptor signaling. Endocr Relat Cancer 2014; 21:R431-43. [PMID: 25183430 PMCID: PMC4182143 DOI: 10.1530/erc-14-0179] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The leucine-rich repeats and immunoglobulin-like domains (LRIG) family of transmembrane proteins contains three vertebrate members (LRIG1, LRIG2 and LRIG3) and one member each in flies (Lambik) and worms (Sma-10). LRIGs have stepped into the spotlight as essential regulators of growth factor receptors, including receptor tyrosine and serine/threonine kinases. LRIGs have been found to both negatively (LRIG1 and LRIG3) and positively (Sma-10 and LRIG3) regulate growth factor receptor expression and signaling, although the precise molecular mechanisms by which LRIGs function are not yet understood. The most is known about LRIG1, which was recently demonstrated to be a tumor suppressor. Indeed, in vivo experiments reinforce the essential link between LRIG1 and repression of its targets for tissue homeostasis. LRIG1 has also been identified as a stem cell marker and regulator of stem cell quiescence in a variety of tissues, discussed within. Comparably, less is known about LRIG2 and LRIG3, although studies to date suggest that their functions are largely distinct from that of LRIG1 and that they likely do not serve as growth/tumor suppressors. Finally, the translational applications of expressing soluble forms of LRIG1 in LRIG1-deficient tumors are being explored and hold tremendous promise.
Collapse
Affiliation(s)
- Catalina Simion
- Department of Biochemistry and Molecular MedicineUniversity of California Davis School of Medicine, 4645 2nd Avenue, Sacramento, California 95817, USA
| | - Maria Elvira Cedano-Prieto
- Department of Biochemistry and Molecular MedicineUniversity of California Davis School of Medicine, 4645 2nd Avenue, Sacramento, California 95817, USA
| | - Colleen Sweeney
- Department of Biochemistry and Molecular MedicineUniversity of California Davis School of Medicine, 4645 2nd Avenue, Sacramento, California 95817, USA
| |
Collapse
|