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
|
Piracha ZZ, Saeed U. Leucine-rich repeats and immunoglobulin-like domains protein 1 (LRIG1) is downregulated in Invasive ductal carcinoma and potential prognostic marker of breast cancer. J Cancer Res Ther 2023; 19:1870-1879. [PMID: 38376291 DOI: 10.4103/jcrt.jcrt_105_22] [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: 01/13/2022] [Accepted: 02/11/2022] [Indexed: 02/21/2024]
Abstract
BACKGROUND LRIG1 belongs to the family of transmembrane proteins containing leucine-rich repeats. LRIGs are considered as tumor suppressors as they negatively regulate receptor tyrosine kinases. The role of LRIG1 as an EGFR regulator makes it an important marker to be studied in various epithelial-derived cancers. METHODS LRIG1 expression was determined in Erbb2 + cell lines by western blotting, and cell motility was examined by cell migration assay. The AKT/GSK3-β/β-catenin pathway was determined in the presence of LRIG1 and Erbb2 by using western blotting. RESULTS So far, no study has reported the expression of LRIG1 in benign forms of tumor such as fibroadenoma. The current study aims to analyze LRIG1 expression in fibroadenoma and invasive ductal carcinoma (IDC) tissues. In this study, we compared the LRIG1 expression with different clinicopathological parameters of patients having IDC or fibroadenoma. LRIG1 expression was low in Erbb2+ cell lines, and more cell motility was observed. The AKT/GSK3-β/β-catenin pathway was activated when LRIG1 was downregulated; consequently, Erbb2 was upregulated. Our results indicated that LRIG1 expression can be significantly correlated with age, Nottingham index, and Her2/neu status of cancer. The expression of LRIG1 in IDC and fibroadenoma were found to be significantly different. CONCLUSION The fibroadenoma tissue sections were found to express LRIG1 more intensely as compared to the IDC sections, which are in line with the studies reporting reduced copy number of the gene either due to gene deletion or transcriptional inhibition. This further supports that the downregulation of LRIG1 may lead to malignant tumor acting as a tumor suppressor.
Collapse
Affiliation(s)
- Zahra Zahid Piracha
- International Center of Medical Sciences Research, Islamabad (44000) Pakistan
- Department of Microbiology, School of Medicine, AJOU University, San 5, Woncheon-dong, Yeongtong-gu, Suwon-si 16222-16713, Gyeonggi-do, South Korea
| | - Umar Saeed
- International Center of Medical Sciences Research, Islamabad (44000) Pakistan
- Department of Microbiology, School of Medicine, AJOU University, San 5, Woncheon-dong, Yeongtong-gu, Suwon-si 16222-16713, Gyeonggi-do, South Korea
| |
Collapse
|
4
|
Monti N, Verna R, Piombarolo A, Querqui A, Bizzarri M, Fedeli V. Paradoxical Behavior of Oncogenes Undermines the Somatic Mutation Theory. Biomolecules 2022; 12:662. [PMID: 35625590 PMCID: PMC9138429 DOI: 10.3390/biom12050662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/21/2022] [Accepted: 04/26/2022] [Indexed: 12/04/2022] Open
Abstract
The currently accepted theory on the influence of DNA mutations on carcinogenesis (the Somatic Mutation Theory, SMT) is facing an increasing number of controversial results that undermine the explanatory power of mutated genes considered as "causative" factors. Intriguing results have demonstrated that several critical genes may act differently, as oncogenes or tumor suppressors, while phenotypic reversion of cancerous cells/tissues can be achieved by modifying the microenvironment, the mutations they are carrying notwithstanding. Furthermore, a high burden of mutations has been identified in many non-cancerous tissues without any apparent pathological consequence. All things considered, a relevant body of unexplained inconsistencies calls for an in depth rewiring of our theoretical models. Ignoring these paradoxes is no longer sustainable. By avoiding these conundrums, the scientific community will deprive itself of the opportunity to achieve real progress in this important biomedical field. To remedy this situation, we need to embrace new theoretical perspectives, taking the cell-microenvironment interplay as the privileged pathogenetic level of observation, and by assuming new explanatory models based on truly different premises. New theoretical frameworks dawned in the last two decades principally focus on the complex interaction between cells and their microenvironment, which is thought to be the critical level from which carcinogenesis arises. Indeed, both molecular and biophysical components of the stroma can dramatically drive cell fate commitment and cell outcome in opposite directions, even in the presence of the same stimulus. Therefore, such a novel approach can help in solving apparently inextricable paradoxes that are increasingly observed in cancer biology.
Collapse
Affiliation(s)
| | | | | | | | | | - Valeria Fedeli
- Systems Biology Group Lab, Department of Experimental Medicine, “Sapienza” University of Rome, Viale Regina Elena 324, 00161 Rome, Italy; (N.M.); (R.V.); (A.P.); (A.Q.); (M.B.)
| |
Collapse
|
5
|
Özturan D, Morova T, Lack NA. Androgen Receptor-Mediated Transcription in Prostate Cancer. Cells 2022; 11:898. [PMID: 35269520 PMCID: PMC8909478 DOI: 10.3390/cells11050898] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 02/25/2022] [Accepted: 03/01/2022] [Indexed: 11/16/2022] Open
Abstract
Androgen receptor (AR)-mediated transcription is critical in almost all stages of prostate cancer (PCa) growth and differentiation. This process involves a complex interplay of coregulatory proteins, chromatin remodeling complexes, and other transcription factors that work with AR at cis-regulatory enhancer regions to induce the spatiotemporal transcription of target genes. This enhancer-driven mechanism is remarkably dynamic and undergoes significant alterations during PCa progression. In this review, we discuss the AR mechanism of action in PCa with a focus on how cis-regulatory elements modulate gene expression. We explore emerging evidence of genetic variants that can impact AR regulatory regions and alter gene transcription in PCa. Finally, we highlight several outstanding questions and discuss potential mechanisms of this critical transcription factor.
Collapse
Affiliation(s)
- Doğancan Özturan
- School of Medicine, Koç University, Istanbul 34450, Turkey;
- Koç University Research Centre for Translational Medicine (KUTTAM), Koç University, Istanbul 34450, Turkey
| | - Tunç Morova
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, BC V6H 3Z6, Canada;
| | - Nathan A. Lack
- School of Medicine, Koç University, Istanbul 34450, Turkey;
- Koç University Research Centre for Translational Medicine (KUTTAM), Koç University, Istanbul 34450, Turkey
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, BC V6H 3Z6, Canada;
| |
Collapse
|
6
|
M Serag El-Dien M, Fathy Mahmoud S, Alhanafy AM, Mohamed Zanaty F, Shawky Holah N. Prognostic significance of LRIG2 and LRIG3 proteins in urothelial bladder carcinoma. J Immunoassay Immunochem 2021; 43:308-332. [PMID: 34839782 DOI: 10.1080/15321819.2021.2005623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Bladder carcinoma is the second most frequent cancer in Egyptian males. Leucine-rich and immunoglobulin-like domains (LRIGs) are usually dysregulated in various human tumors. The aim of this study is to explore the immunohistochemical expression of LRIG2 and LRIG3 in urothelial bladder carcinoma (UBC) and their relationship to patients clinicopathological data including survival. The study cohort included 79 UBC cases (14 non muscle invasive (NMI) and 65 muscle invasive (MI)). We assessed the associations of LRIG2 and LRIG3 expression with clinicopathological data, as well as progression-free and overall survival. Most of studied cases (>50%) express LRIG2 and LRIG3. Statistically significant association was observed between positivity for LRIG3 and muscle invasion (P = 0.001), high grade (P = 0.03), and female gender (P = 0.02). Moreover, positive LRIG2 staining was associated with early stage (T2) (P = 0.03), lymphovascular invasion (P = 0.004), and tendency to non-muscle invasive stage (P = 0.07). Grouping of cases according to positivity/negativity of both markers showed that cases with dual positivity for both proteins are associated with muscle invasion (P = 0.001) and paradoxically with prolonged overall survival (P = 0.037). We conclude that although the association of LRIG3 with MI and high-grade tumors, its expression is related to better survival. LRIG3 has the dominant role even if it coexists with LRIG2. The role of LRIG2 remains to be further investigated.
Collapse
|
7
|
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: 1] [Impact Index Per Article: 0.3] [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
|
8
|
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: 6] [Impact Index Per Article: 2.0] [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
|
9
|
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: 11] [Impact Index Per Article: 2.2] [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
|
10
|
Mao F, Holmlund C, Faraz M, Wang W, Bergenheim T, Kvarnbrink S, Johansson M, Henriksson R, Hedman H. Lrig1 is a haploinsufficient tumor suppressor gene in malignant glioma. Oncogenesis 2018; 7:13. [PMID: 29391393 PMCID: PMC5833707 DOI: 10.1038/s41389-017-0012-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 11/09/2017] [Indexed: 11/19/2022] Open
Abstract
Recently, a genome-wide association study showed that a single nucleotide polymorphism (SNP) —rs11706832—in intron 2 of the human LRIG1 (Leucine-rich repeats and immunoglobulin-like domains 1) gene is associated with susceptibility to glioma. However, the mechanism by which rs11706832 affects glioma risk remains unknown; additionally, it is unknown whether the expression levels of LRIG1 are a relevant determinant of gliomagenesis. Here, we investigated the role of Lrig1 in platelet-derived growth factor (PDGF)-induced experimental glioma in mice by introducing mono-allelic and bi-allelic deletions of Lrig1 followed by inducing gliomagenesis via intracranial retroviral transduction of PDGFB in neural progenitor cells. Lrig1 was expressed in PDGFB-induced gliomas in wild-type mice as assessed using in situ hybridization. Intriguingly, Lrig1-heterozygous mice developed higher grade gliomas than did wild-type mice (grade IV vs. grade II/III, p = 0.002). Reciprocally, the ectopic expression of LRIG1 in the TB107 high-grade human glioma (glioblastoma, grade IV) cell line decreased the invasion of orthotopic tumors in immunocompromised mice in vivo and reduced cell migration in vitro. Concomitantly, the activity of the receptor tyrosine kinase MET was downregulated, which partially explained the reduction in cell migration. In summary, Lrig1 is a haploinsufficient suppressor of PDGFB-driven glioma, possibly in part via negative regulation of MET-driven cell migration and invasion. Thus, for the first time, changes in physiological Lrig1 expression have been linked to gliomagenesis, whereby the SNP rs11706832 may affect glioma risk by regulating LRIG1 expression.
Collapse
Affiliation(s)
- Feng Mao
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, Sweden.,Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Camilla Holmlund
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, Sweden
| | - Mahmood Faraz
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, Sweden
| | - Wanzhong Wang
- Department of Pathology/Cytology, Karolinska University Hospital, Huddinge, Sweden
| | - Tommy Bergenheim
- Department of Pharmacology and Clinical Neuroscience, Section of Neurosurgery, Umeå University, Umeå, Sweden
| | - Samuel Kvarnbrink
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, Sweden
| | - Mikael Johansson
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, Sweden
| | - Roger Henriksson
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, Sweden.,Regionalt Cancercentrum Stockholm Gotland, Karolinska, Stockholm, Sweden
| | - Håkan Hedman
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, Sweden.
| |
Collapse
|
11
|
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: 0.7] [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
|
12
|
Neirinckx V, Hedman H, Niclou SP. Harnessing LRIG1-mediated inhibition of receptor tyrosine kinases for cancer therapy. Biochim Biophys Acta Rev Cancer 2017; 1868:109-116. [PMID: 28259645 DOI: 10.1016/j.bbcan.2017.02.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 02/27/2017] [Accepted: 02/28/2017] [Indexed: 02/07/2023]
Abstract
Leucine-rich repeats and immunoglobulin-like domains containing protein 1 (LRIG1) is an endogenous feedback regulator of receptor tyrosine kinases (RTKs) and was recently shown to inhibit growth of different types of malignancies. Additionally, this multifaceted RTK inhibitor was reported to be a tumor suppressor, a stem cell regulator, and a modulator of different cellular phenotypes. This mini-review provides a concise and up-to-date summary about the known functions of LRIG1 and its related family members, with a special emphasis on underlying molecular mechanisms and the opportunities for harnessing its therapeutic potential against cancer.
Collapse
Affiliation(s)
- Virginie Neirinckx
- NorLux Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health, 1526, Luxembourg
| | - Hakan Hedman
- Oncology Research Laboratory, Department of Radiation Sciences, Umeå University, 90187 Umeå, Sweden
| | - Simone P Niclou
- NorLux Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health, 1526, Luxembourg; K.G. Jebsen Brain Tumour Research Centre, Department of Biomedicine, University of Bergen, 5020 Bergen, Norway.
| |
Collapse
|
13
|
Jiang X, Li H. Overexpression of LRIG1 regulates PTEN via MAPK/MEK signaling pathway in esophageal squamous cell carcinoma. Exp Ther Med 2016; 12:2045-2052. [PMID: 27698691 PMCID: PMC5038857 DOI: 10.3892/etm.2016.3606] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 06/20/2016] [Indexed: 01/05/2023] Open
Abstract
The present study aimed to evaluate the role of leucine-rich repeats and immunoglobulin-like domain protein 1 (LRIG1) in the regulation of phosphatase and tensin homolog (PTEN) expression in esophageal carcinogenesis. LRIG1 was overexpressed in esophageal squamous cell carcinoma (ESCC) cell lines, and the effect of LRIG1 overexpression on the mRNA and protein expression levels of PTEN was evaluated by reverse transcription-quantitative polymerase chain reaction and western blotting. Furthermore, the effects of LRIG1 overexpression on the cell cycle distribution and apoptosis of ESCC cells were examined by flow cytometry. Various cell signaling pathway inhibitors were used to assess the effects of LRIG1 on downstream signaling in ESCC cell lines. In addition, the association between LRIG1 and PTEN expression was examined in 48 samples from patients with ESCC. LRIG1 overexpression was demonstrated to downregulate PTEN expression in ESCC cell lines, and promote their proliferation and inhibit apoptosis. In addition, LRIG1-mediated suppression of PTEN expression was inhibited by the U0126 inhibitor, which suggests that LRIG1 may inhibit the activation of PTEN signaling molecules by triggering the mitogen-activated protein kinase (MAPK)/MAPK kinase 1 (MEK) signaling pathway. In conclusion, the present study demonstrated that overexpression of LRIG1 significantly and adversely affected the survival of ESCC cells, and that the MAPK/MEK signaling pathway may be responsible for the repression of PTEN expression and function.
Collapse
Affiliation(s)
- Xiaofang Jiang
- Central Laboratory, Xinjiang Medical University, Urumqi, Xinjiang 830011, P.R. China
| | - Huiwu Li
- Department of Biochemistry, School of Basic Medicine, Xinjiang Medical University, Urumqi, Xinjiang 830011, P.R. China; Tumor Institute, Affiliated Tumor Hospital, Xinjiang Medical University, Urumqi, Xinjiang 830011, P.R. China
| |
Collapse
|
14
|
Yang H, Yao J, Yin J, Wei X. Decreased LRIG1 in Human Ovarian Cancer Cell SKOV3 Upregulates MRP-1 and Contributes to the Chemoresistance of VP16. Cancer Biother Radiopharm 2016; 31:125-32. [PMID: 27183435 DOI: 10.1089/cbr.2015.1970] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Affiliation(s)
- Hua Yang
- Department of Gynaecology, The Second Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Jun Yao
- Department of Gynaecology, The Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Jiangpin Yin
- Department of Gynaecology, The Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Xuan Wei
- Department of Gynaecology, The Second Affiliated Hospital of Guilin Medical University, Guilin, China
| |
Collapse
|
15
|
Hellström M, Ericsson M, Johansson B, Faraz M, Anderson F, Henriksson R, Nilsson SK, Hedman H. Cardiac hypertrophy and decreased high-density lipoprotein cholesterol in Lrig3-deficient mice. Am J Physiol Regul Integr Comp Physiol 2016; 310:R1045-52. [PMID: 27009049 DOI: 10.1152/ajpregu.00309.2015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 03/21/2016] [Indexed: 11/22/2022]
Abstract
Genetic factors confer risk for cardiovascular disease. Recently, large genome-wide population studies have shown associations between genomic loci close to LRIG3 and heart failure and plasma high-density lipoprotein (HDL) cholesterol level. Here, we ablated Lrig3 in mice and investigated the importance of Lrig3 for heart function and plasma lipid levels. Quantitative reverse transcription-polymerase chain reaction (RT-PCR) was used to analyze Lrig3 expression in the hearts of wild-type and Lrig3-deficient mice. In addition, molecular, physiological, and functional parameters such as organ weights, heart rate, blood pressure, heart structure and function, gene expression in the heart, and plasma insulin, glucose, and lipid levels were evaluated. The Lrig3-deficient mice were smaller than the wild-type mice but otherwise appeared grossly normal. Lrig3 was expressed at detectable but relatively low levels in adult mouse hearts. At 9 mo of age, ad libitum-fed Lrig3-deficient mice had lower insulin levels than wild-type mice. At 12 mo of age, Lrig3-deficient mice exhibited increased blood pressure, and the Lrig3-deficient female mice displayed signs of cardiac hypertrophy as assessed by echocardiography, heart-to-body weight ratio, and expression of the cardiac hypertrophy marker gene Nppa. Additionally, Lrig3-deficient mice had reduced plasma HDL cholesterol and free glycerol. These findings in mice complement the human epidemiological results and suggest that Lrig3 may influence heart function and plasma lipid levels in mice and humans.
Collapse
Affiliation(s)
- Martin Hellström
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, Sweden;
| | - Madelene Ericsson
- Department of Medical Biosciences, Physiological Chemistry, Umeå University, Umeå, Sweden
| | - Bengt Johansson
- Department of Public Health and Clinical Medicine, Medicine, Umeå University, Umeå, Sweden; and
| | - Mahmood Faraz
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, Sweden
| | - Fredrick Anderson
- Department of Medical Biosciences, Physiological Chemistry, Umeå University, Umeå, Sweden
| | - Roger Henriksson
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, Sweden; Regional Cancer Center Stockholm/Gotland, Stockholm, Sweden
| | - Stefan K Nilsson
- Department of Medical Biosciences, Physiological Chemistry, Umeå University, Umeå, Sweden
| | - Håkan Hedman
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, Sweden
| |
Collapse
|
16
|
D'Uva G, Lauriola M. Towards the emerging crosstalk: ERBB family and steroid hormones. Semin Cell Dev Biol 2015; 50:143-52. [PMID: 26582250 DOI: 10.1016/j.semcdb.2015.11.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Revised: 10/28/2015] [Accepted: 11/09/2015] [Indexed: 01/05/2023]
Abstract
Growth factors acting through receptor tyrosine kinases (RTKs) of ERBB family, along with steroid hormones (SH) acting through nuclear receptors (NRs), are critical signalling mediators of cellular processes. Deregulations of ERBB and steroid hormone receptors are responsible for several diseases, including cancer, thus demonstrating the central role played by both systems. This review will summarize and shed light on an emerging crosstalk between these two important receptor families. How this mutual crosstalk is attained, such as through extensive genomic and non-genomic interactions, will be addressed. In light of recent studies, we will describe how steroid hormones are able to fine-tune ERBB feedback loops, thus impacting on cellular output and providing a new key for understanding the complexity of biological processes in physiological or pathological conditions. In our understanding, the interactions between steroid hormones and RTKs deserve further attention. A system biology approach and advanced technologies for the analysis of RTK-SH crosstalk could lead to major advancements in molecular medicine, providing the basis for new routes of pharmacological intervention in several diseases, including cancer.
Collapse
Affiliation(s)
- Gabriele D'Uva
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel.
| | - Mattia Lauriola
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel; Department of Experimental, Diagnostic and Specialty Medicine - DIMES, University of Bologna, Bologna 40138, Italy.
| |
Collapse
|
17
|
Adamo HH, Strömvall K, Nilsson M, Halin Bergström S, Bergh A. Adaptive (TINT) Changes in the Tumor Bearing Organ Are Related to Prostate Tumor Size and Aggressiveness. PLoS One 2015; 10:e0141601. [PMID: 26536349 PMCID: PMC4633147 DOI: 10.1371/journal.pone.0141601] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 10/09/2015] [Indexed: 11/18/2022] Open
Abstract
In order to grow, tumors need to induce supportive alterations in the tumor-bearing organ, by us named tumor instructed normal tissue (TINT) changes. We now examined if the nature and magnitude of these responses were related to tumor size and aggressiveness. Three different Dunning rat prostate tumor cells were implanted into the prostate of immune-competent rats; 1) fast growing and metastatic MatLyLu tumor cells 2) fast growing and poorly metastatic AT-1 tumor cells, and 3) slow growing and non-metastatic G tumor cells. All tumor types induced increases in macrophage, mast cell and vascular densities and in vascular cell-proliferation in the tumor-bearing prostate lobe compared to controls. These increases occurred in parallel with tumor growth. The most pronounced and rapid responses were seen in the prostate tissue surrounding MatLyLu tumors. They were, also when small, particularly effective in attracting macrophages and stimulating growth of not only micro-vessels but also small arteries and veins compared to the less aggressive AT-1 and G tumors. The nature and magnitude of tumor-induced changes in the tumor-bearing organ are related to tumor size but also to tumor aggressiveness. These findings, supported by previous observation in patient samples, suggest that one additional way to evaluate prostate tumor aggressiveness could be to monitor its effect on adjacent tissues.
Collapse
Affiliation(s)
- Hanibal Hani Adamo
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| | - Kerstin Strömvall
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| | - Maria Nilsson
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| | | | - Anders Bergh
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
- * E-mail:
| |
Collapse
|
18
|
Kou C, Zhou T, Han X, Zhuang H, Qian H. LRIG1, a 3p tumor suppressor, represses EGFR signaling and is a novel epigenetic silenced gene in colorectal cancer. Biochem Biophys Res Commun 2015; 464:519-25. [PMID: 26159916 DOI: 10.1016/j.bbrc.2015.06.173] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 06/30/2015] [Indexed: 10/23/2022]
Abstract
Downregulation of LRIG1 was found in many types of cancer. However, data concerning the possible mechanism of LRIG1 reduction in cancers were not reported yet. To analyze the regulation and function of LRIG1 in colorectal cancer (CRC), 6 cell lines, 46 paired tissues from primary CRC cases were employed in this study. In CRC cell lines, under-expression of LRIG1 was correlated with promoter region hypermethylation, and restoration of LRIG1 was induced by 5-Aza-2'-deoxyazacytidine treatment. Subsequently, we ectopically expressed LRIG1 in LRIG1 low-expressing HCT-116 cells and suppressed LRIG1 in LRIG1 high-expressing LoVo cells. We found that over-expression of LRIG1 inhibits cell proliferation and colony formation and tumor growth, while knockdown of LRIG1 promotes cell proliferation and colony formation. Decreased and increased EGFR/AKT signaling pathway may partially explain the lower and higher rates of proliferation in CRC cells transfected with LRIG1 cDNA or shRNA. In clinical samples, we compared the methylation, mRNA and protein expression of LRIG1 in samples of CRC tissues. A significant increase in LRIG1 methylation was identified in CRC specimens compared to adjacent normal tissues and that it was negatively correlated with its mRNA and protein expression. In conclusion, LRIG1 is frequently methylated in human CRC and consequent mRNA and protein downregulation may contribute to tumor growth by activating EGFR/AKT signaling.
Collapse
Affiliation(s)
- Changhua Kou
- Department of Oncological Surgery, The Central Hospital of Xuzhou City, Xuzhou, Jiangsu 221000, China.
| | - Tian Zhou
- Department of Gastroenterology, The Central Hospital of Xuzhou City, Xuzhou, Jiangsu 221000, China
| | - Xilin Han
- Department of Oncological Surgery, The Central Hospital of Xuzhou City, Xuzhou, Jiangsu 221000, China
| | - Huijie Zhuang
- Department of Oncological Surgery, The Central Hospital of Xuzhou City, Xuzhou, Jiangsu 221000, China
| | - Haixin Qian
- The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, China.
| |
Collapse
|
19
|
Construction of human LRIG1-TAT fusions and TAT-mediated LRIG1 protein delivery. Biomed Pharmacother 2014; 69:396-401. [PMID: 25661388 DOI: 10.1016/j.biopha.2014.12.034] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 12/10/2014] [Indexed: 11/23/2022] Open
Abstract
Human leucine-rich repeats and immunoglobulin-like domains (LRIG1) is a tumor suppressor in animals and also functions as an endogenous suppressor in human tumor. The level of LRIG1 expression is highly associated with patient survival in clinic. The exploration of LRIG1 as a protein drug is an important task. HIV-1 transactivator of transcription peptide (TAT) is an excellent candidate for protein transduction. In this study, human LRIG1 was cloned and LRIG1-TAT fusion gene was constructed. The fusion proteins were produced by an Escherichia coli strain and purified by Ni(2+)-resin. Western blot assay and immunofluorescence microscopy were employed for monitoring LRIG1-TAT protein transduction into human neuroblastoma cells. Cell proliferation and invasion were measured for evaluating the effect of LRIG1-TAT on neuroblastoma cell. Our data showed that LRIG1 protein can be delivered into cells or organs in living animals by TAT. One-time transduction of LRIG1 proteins into human neuroblastoma cells enhanced cell proliferation and increased cell invasion. In vivo transduction showed that LRIG1-TAT protein can be presented in living animal organs. Our experiments provide a new vision on LRIG1 applications and also offer a therapy window for revealing the intrinsic function of LRIG1 on cells.
Collapse
|
20
|
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
|
21
|
Guo Z, Chen Q, Liu B, Tian D, Zhang S, Li M. LRIG1 enhances chemosensitivity by modulating BCL-2 expression and receptor tyrosine kinase signaling in glioma cells. Yonsei Med J 2014; 55:1196-205. [PMID: 25048475 PMCID: PMC4108802 DOI: 10.3349/ymj.2014.55.5.1196] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
PURPOSE Leucine-rich repeats and immunoglobulin-like domains 1 (LRIG1) are an inhibitor of receptor tyrosine kinases (RTKs) that was discovered in recent years, and many studies showed that LRIG1 is a tumor suppressor gene and may be related to tumor drug resistance. In this study, we explored whether LRIG1 protein expression can improve the chemosensitivity of glioma cells and what was its mechanism. MATERIALS AND METHODS We collected 93 cases of glioma tissues and detected the expression of LRIG1 and BCL-2. We constructed a multidrug resistance cell line U251/multidrug resistance (MDR) and examined the change of LRIG1 and BCL-2 at mRNA and protein expression levels. LRIG1 expression was upregulated in U251/MDR cells and we detected the change of multidrug resistance. Meanwhile, we changed the expression of LRIG1 and BCL-2 and explored the relationship between LRIG1 and BCL-2. Finally, we also explored the relationship between LRIG1 and RTKs. RESULTS LRIG1 was negatively correlated with BCL-2 expression in glioma tissue and U251/MDR cells, and upregulation of LRIG1 can enhance chemosensitivity and inhibit BCL-2 expression. Furthermore, LRIG1 was negatively correlated with RTKs in U251/MDR cells. CONCLUSION These results demonstrated that LRIG1 can improve chemosensitivity by modulating BCL-2 expression and RTK signaling in glioma cells.
Collapse
Affiliation(s)
- Zhentao Guo
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Qianxue Chen
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.
| | - Baohui Liu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Daofeng Tian
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Shenqi Zhang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Mingchang Li
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
22
|
Häggström J, Cipriano M, Forshell LP, Persson E, Hammarsten P, Stella N, Fowler CJ. Potential upstream regulators of cannabinoid receptor 1 signaling in prostate cancer: a Bayesian network analysis of data from a tissue microarray. Prostate 2014; 74:1107-17. [PMID: 24913716 PMCID: PMC4145668 DOI: 10.1002/pros.22827] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Accepted: 04/30/2014] [Indexed: 01/12/2023]
Abstract
BACKGROUND The endocannabinoid system regulates cancer cell proliferation, and in prostate cancer a high cannabinoid CB1 receptor expression is associated with a poor prognosis. Down-stream mediators of CB1 receptor signaling in prostate cancer are known, but information on potential upstream regulators is lacking. RESULTS Data from a well-characterized tumor tissue microarray were used for a Bayesian network analysis using the max-min hill-climbing method. In non-malignant tissue samples, a directionality of pEGFR (the phosphorylated form of the epidermal growth factor receptor) → CB1 receptors were found regardless as to whether the endocannabinoid metabolizing enzyme fatty acid amide hydrolase (FAAH) was included as a parameter. A similar result was found in the tumor tissue, but only when FAAH was included in the analysis. A second regulatory pathway, from the growth factor receptor ErbB2 → FAAH was also identified in the tumor samples. Transfection of AT1 prostate cancer cells with CB1 receptors induced a sensitivity to the growth-inhibiting effects of the CB receptor agonist CP55,940. The sensitivity was not dependent upon the level of receptor expression. Thus a high CB1 receptor expression alone does not drive the cells towards a survival phenotype in the presence of a CB receptor agonist. CONCLUSIONS The data identify two potential regulators of the endocannabinoid system in prostate cancer and allow the construction of a model of a dysregulated endocannabinoid signaling network in this tumor. Further studies should be designed to test the veracity of the predictions of the network analysis in prostate cancer and other solid tumors.
Collapse
Affiliation(s)
- Jenny Häggström
- Department of Statistics, Umeå School of Business and Economics, Umeå UniversityUmeå, Sweden
| | - Mariateresa Cipriano
- Department of Pharmacology and Clinical Neuroscience, Umeå UniversityUmeå, Sweden
| | - Linus Plym Forshell
- Department of Pharmacology and Clinical Neuroscience, Umeå UniversityUmeå, Sweden
| | - Emma Persson
- Department of Radiation Sciences, Oncology, Umeå UniversityUmeå, Sweden
| | - Peter Hammarsten
- Department of Medical Biosciences, Pathology, Umeå UniversityUmeå, Sweden
| | - Nephi Stella
- Department of Pharmacology, Psychiatry and Behavioral Sciences, University of WashingtonSeattle, Washington
| | - Christopher J Fowler
- Department of Pharmacology and Clinical Neuroscience, Umeå UniversityUmeå, Sweden
- *Correspondence to: Professor Christopher J. Fowler, Department of Pharmacology and Clinical Neuroscience, Umeå University, SE-901 87, Umeå, Sweden. E-mail:
| |
Collapse
|
23
|
Sutinen P, Malinen M, Heikkinen S, Palvimo JJ. SUMOylation modulates the transcriptional activity of androgen receptor in a target gene and pathway selective manner. Nucleic Acids Res 2014; 42:8310-9. [PMID: 24981513 PMCID: PMC4117771 DOI: 10.1093/nar/gku543] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Androgen receptor (AR) plays an important regulatory role in prostate cancer. AR's transcriptional activity is regulated by androgenic ligands, but also by post-translational modifications, such as SUMOylation. To study the role of AR SUMOylation in genuine chromatin environment, we compared androgen-regulated gene expression and AR chromatin occupancy in PC-3 prostate cancer cell lines stably expressing wild-type (wt) or doubly SUMOylation site-mutated AR (AR-K386R,K520R). Our genome-wide gene expression analyses reveal that the SUMOylation modulates the AR function in a target gene and pathway selective manner. The transcripts that are differentially regulated by androgen and SUMOylation are linked to cellular movement, cell death, cellular proliferation, cellular development and cell cycle. Fittingly, SUMOylation mutant AR cells proliferate faster and are more sensitive to apoptosis. Moreover, ChIP-seq analyses show that the SUMOylation can modulate the chromatin occupancy of AR on many loci in a fashion that parallels their differential androgen-regulated expression. De novo motif analyses reveal that FOXA1, C/EBP and AP-1 motifs are differentially enriched at the wtAR- and the AR-K386R,K520R-preferred genomic binding positions. Taken together, our data indicate that SUMOylation does not simply repress the AR activity, but it regulates AR's interaction with the chromatin and the receptor's target gene selection.
Collapse
Affiliation(s)
- Päivi Sutinen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, PO Box 1627, FI-70211 Kuopio, Finland
| | - Marjo Malinen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, PO Box 1627, FI-70211 Kuopio, Finland
| | - Sami Heikkinen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, PO Box 1627, FI-70211 Kuopio, Finland
| | - Jorma J Palvimo
- Institute of Biomedicine, University of Eastern Finland, Kuopio, PO Box 1627, FI-70211 Kuopio, Finland Department of Pathology, Kuopio University Hospital, Kuopio, Finland
| |
Collapse
|
24
|
Suraneni MV, Moore JR, Zhang D, Badeaux M, Macaluso MD, DiGiovanni J, Kusewitt D, Tang DG. Tumor-suppressive functions of 15-Lipoxygenase-2 and RB1CC1 in prostate cancer. Cell Cycle 2014; 13:1798-810. [PMID: 24732589 PMCID: PMC4111726 DOI: 10.4161/cc.28757] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
15-Lipoxygenase-2 (15-LOX2) is a human-specific lipid-peroxidizing enzyme most prominently expressed in epithelial cells of normal human prostate but downregulated or completely lost in>70% of prostate cancer (PCa) cases. Transgenic expression of 15-LOX2 in the mouse prostate surprisingly causes hyperplasia. Here we first provide evidence that 15-LOX2-induced prostatic hyperplasia does not progress to PCa even in p53(+/-) or p53(-/-) background. More important, by generating 15-LOX2; Hi-Myc double transgenic (dTg) mice, we show that 15-LOX2 expression inhibits Myc-induced PCa development, such that in the 3-month- and 6-month-old dTg mice, there is a significant reduction in prostate intraneoplasia (PIN) and PCa prevalent in age-matched Hi-Myc prostates. The dTg prostates show increased cell senescence and expression of several senescence-associated molecules, including p27, phosphorylated Rb, and Rb1cc1. We further show that in HPCa, 15-LOX2 and c-Myc manifest reciprocal protein expression patterns. Moreover, RB1CC1 accumulates in senescing normal human prostate (NHP) cells, and in both NHP and RWPE-1 cells, the 15-LOX2 metabolic products 15(S)-HPETE and 15(S)-HETE induce RB1CC1. We finally show that unlike 15-LOX2, RB1CC1 is not lost but rather frequently overexpressed in PCa samples. RB1CC1 knockdown in PC3 cells enhances clonal growth in vitro and tumor growth in vivo. Together, our present studies provide evidence for tumor-suppressive functions for both 15-LOX2 and RB1CC1.
Collapse
Affiliation(s)
- Mahipal V Suraneni
- Department of Molecular Carcinogenesis; The University of Texas MD Anderson Cancer Center; Smithville, TX USA
| | - John R Moore
- Department of Molecular Carcinogenesis; The University of Texas MD Anderson Cancer Center; Smithville, TX USA
| | - Dingxiao Zhang
- Department of Molecular Carcinogenesis; The University of Texas MD Anderson Cancer Center; Smithville, TX USA
| | - Mark Badeaux
- Department of Molecular Carcinogenesis; The University of Texas MD Anderson Cancer Center; Smithville, TX USA
| | - Marc D Macaluso
- Department of Molecular Carcinogenesis; The University of Texas MD Anderson Cancer Center; Smithville, TX USA
| | - John DiGiovanni
- Division of Pharmacology and Toxicology; College of Pharmacy; The University of Texas at Austin; Austin, TX USA
| | - Donna Kusewitt
- Department of Molecular Carcinogenesis; The University of Texas MD Anderson Cancer Center; Smithville, TX USA
| | - Dean G Tang
- Department of Molecular Carcinogenesis; The University of Texas MD Anderson Cancer Center; Smithville, TX USA; Cancer Stem Cell Institute; Research Center for Translational Medicine; Shanghai East Hospital; Tongji University School of Medicine; Shanghai, China
| |
Collapse
|
25
|
Lindquist D, Näsman A, Tarján M, Henriksson R, Tot T, Dalianis T, Hedman H. Expression of LRIG1 is associated with good prognosis and human papillomavirus status in oropharyngeal cancer. Br J Cancer 2014; 110:1793-800. [PMID: 24548859 PMCID: PMC3974094 DOI: 10.1038/bjc.2014.87] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Revised: 12/02/2013] [Accepted: 01/22/2014] [Indexed: 01/08/2023] Open
Abstract
Background: The incidence of human papillomavirus (HPV)-associated oropharyngeal cancer has increased rapidly during the past decades. HPV is typically associated with a favourable outcome; however, a need exists for new and more effective prognostic and predictive markers for this disease. Leucine-rich repeats and immunoglobulin-like domains (LRIG)-1 is a tumour suppressor protein that belongs to the LRIG family. LRIG1 expression has prognostic significance in various human cancers, including cervical cancer, where HPV is a key aetiological agent. Methods: The prognostic value of LRIG1 and LRIG2 immunoreactivity was investigated in tumour specimens from a Swedish cohort of patients with tonsillar and base of tongue oropharyngeal cancers, including 278 patients. Results: LRIG1 immunoreactivity correlated with disease-free survival and overall survival in univariate and multivariate analyses. Notably, patients with HPV-positive tumours with high LRIG1 staining intensity or a high percentage of LRIG1-positive cells showed a very good prognosis. Furthermore, LRIG1 expression correlated with HPV status, whereas LRIG2 expression inversely correlated with HPV status. Conclusions: Taken together, the results suggest that LRIG1 immunoreactivity could be a clinically important prognostic marker in HPV-associated oropharyngeal cancer.
Collapse
Affiliation(s)
- D Lindquist
- Department of Radiation Sciences, Umeå University, SE-901 87, Umeå, Sweden
| | - A Näsman
- Department of Oncology-Pathology, Karolinska Institute, SE-171 76, Stockholm, Sweden
| | - M Tarján
- Department of Pathology and Clinical Cytology, Central Hospital Falun, SE-791 29, Falun, Sweden
| | - R Henriksson
- Department of Radiation Sciences, Umeå University, SE-901 87, Umeå, Sweden
| | - T Tot
- Department of Pathology and Clinical Cytology, Central Hospital Falun, SE-791 29, Falun, Sweden
| | - T Dalianis
- Department of Oncology-Pathology, Karolinska Institute, SE-171 76, Stockholm, Sweden
| | - H Hedman
- Department of Radiation Sciences, Umeå University, SE-901 87, Umeå, Sweden
| |
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: 3.7] [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
|
27
|
Winther M, Walmod PS. Neural cell adhesion molecules belonging to the family of leucine-rich repeat proteins. ADVANCES IN NEUROBIOLOGY 2014; 8:315-95. [PMID: 25300143 DOI: 10.1007/978-1-4614-8090-7_14] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Leucine-rich repeats (LRRs) are motifs that form protein-ligand interaction domains. There are approximately 140 human genes encoding proteins with extracellular LRRs. These encode cell adhesion molecules (CAMs), proteoglycans, G-protein-coupled receptors, and other types of receptors. Here we give a brief description of 36 proteins with extracellular LRRs that all can be characterized as CAMs or putative CAMs expressed in the nervous system. The proteins are involved in multiple biological processes in the nervous system including the proliferation and survival of cells, neuritogenesis, axon guidance, fasciculation, myelination, and the formation and maintenance of synapses. Moreover, the proteins are functionally implicated in multiple diseases including cancer, hearing impairment, glaucoma, Alzheimer's disease, multiple sclerosis, Parkinson's disease, autism spectrum disorders, schizophrenia, and obsessive-compulsive disorders. Thus, LRR-containing CAMs constitute a large group of proteins of pivotal importance for the development, maintenance, and regeneration of the nervous system.
Collapse
|
28
|
Chang L, Shi R, Yang T, Li F, Li G, Guo Y, Lang B, Yang W, Zhuang Q, Xu H. Restoration of LRIG1 suppresses bladder cancer cell growth by directly targeting EGFR activity. J Exp Clin Cancer Res 2013; 32:101. [PMID: 24314030 PMCID: PMC3880093 DOI: 10.1186/1756-9966-32-101] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Accepted: 12/04/2013] [Indexed: 11/10/2022] Open
Abstract
Background Recently, leucine-rich repeats and immunoglobulin-like domains 1 (LRIG1), a negative regulator of EGFR, was discovered is a novel agent for suppressing bladder cancer. The aim of this study was to investigate the impact of LRIG1 on the biological features of aggressive bladder cancer cells and the possible mechanisms of enhanced apoptosis induced by upregulation of LRIG1. Methods In this study, we examined the mRNA and protein expression of LRIG1 and EGFR in bladder cancers and normal bladder. Meanwhile, we overexpressed LRIG1 with adenovirus vector in T24/5637 bladder cancer cell lines, and we used real time-PCR, western blot, and co-immunoprecipitation analysis in order to examine the effects of LRIG1 gene on EGFR. Furthermore, we evaluate the impact of LRIG1 gene on the function of human bladder cancer cells and EGFR signaling. Results The expression of LRIG1 was decreased, while the expression of EGFR was increased in the majority of bladder cancer, and the ratio of EGFR/LRIG1 was increased in tumors versus normal tissue. We found that upregulation of LRIG1 induced cell apoptosis and cell growth inhibition, and further reversed invasion in bladder cancer cell lines in vitro by inhibiting phosphorylation of downstream MAPK and AKT signaling pathway. Conclusion Taken together, our findings provide us with an insight into LRIG1 function, and we conclude that LRIG1 evolved in bladder cancer as a rare feedback negative attenuator of EGFR, thus could offer a novel therapeutic target to treat patients with bladder cancer.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Hua Xu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
29
|
Rafidi H, Mercado F, Astudillo M, Fry WHD, Saldana M, Carraway KL, Sweeney C. Leucine-rich repeat and immunoglobulin domain-containing protein-1 (Lrig1) negative regulatory action toward ErbB receptor tyrosine kinases is opposed by leucine-rich repeat and immunoglobulin domain-containing protein 3 (Lrig3). J Biol Chem 2013; 288:21593-605. [PMID: 23723069 DOI: 10.1074/jbc.m113.486050] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Lrig1 is the founding member of the Lrig family of transmembrane leucine-rich repeat proteins, which also includes Lrig2 and Lrig3. Lrig1 is a negative regulator of oncogenic receptor tyrosine kinases, including ErbB and Met receptors, and promotes receptor degradation. Lrig1 has recently emerged as both a tumor suppressor and a key regulator of epidermal and epithelial stem cell quiescence. Despite this, little is known of the mechanisms by which Lrig1 is regulated. Lrig3 was recently reported to increase ErbB receptor expression suggesting that it may function in a manner opposite to Lrig1. In this study, we explore the interaction between Lrig1 and Lrig3 and demonstrate that Lrig1 and Lrig3 functionally oppose one another. Lrig3 opposes Lrig1 negative regulatory activity and stabilizes ErbB receptors. Conversely, Lrig1 destabilizes Lrig3, limiting Lrig3's positive effects on receptors and identifying Lrig3 as a new target of Lrig1. These studies provide new insight into the regulation of Lrig1 and uncover a complex cross-talk between Lrig1 and Lrig3.
Collapse
Affiliation(s)
- Hanine Rafidi
- Department of Biochemistry and Molecular Medicine, University of California at Davis Comprehensive Cancer Center, Sacramento, California 95817, USA
| | | | | | | | | | | | | |
Collapse
|
30
|
Johansson M, Oudin A, Tiemann K, Bernard A, Golebiewska A, Keunen O, Fack F, Stieber D, Wang B, Hedman H, Niclou SP. The soluble form of the tumor suppressor Lrig1 potently inhibits in vivo glioma growth irrespective of EGF receptor status. Neuro Oncol 2013; 15:1200-11. [PMID: 23723255 PMCID: PMC3748912 DOI: 10.1093/neuonc/not054] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background Deregulated growth factor signaling is a major driving force in the initiation and progression of glioblastoma. The tumor suppressor and stem cell marker Lrig1 is a negative regulator of the epidermal growth factor receptor (EGFR) family. Here, we addressed the therapeutic potential of the soluble form of Lrig1 (sLrig1) in glioblastoma treatment and the mechanism of sLrig1-induced growth inhibition. Methods With use of encapsulated cells, recombinant sLrig1 was locally delivered in orthotopic glioblastoma xenografts generated from freshly isolated patient tumors. Tumor growth and mouse survival were evaluated. The efficacy of sLrig1 and the affected downstream signaling was studied in vitro and in vivo in glioma cells displaying variable expression of wild-type and/or a constitutively active EGFR mutant (EGFRvIII). Results Continuous interstitial delivery of sLrig1 in genetically diverse patient-derived glioma xenografts led to strong tumor growth inhibition. Glioma cell proliferation in vitro and tumor growth in vivo were potently inhibited by sLrig1, irrespective of EGFR expression levels. Of importance, tumor growth was also suppressed in EGFRvIII-driven glioma. sLrig1 induced cell cycle arrest without changing total receptor level or phosphorylation. Affected downstream effectors included MAP kinase but not AKT signaling. Of importance, local delivery of sLrig1 into established tumors led to a 32% survival advantage in treated mice. Conclusions To our knowledge, this is the first report demonstrating that sLrig1 is a potent inhibitor of glioblastoma growth in clinically relevant experimental glioma models and that this effect is largely independent of EGFR status. The potent anti-tumor effect of sLrig1, in combination with cell encapsulation technology for in situ delivery, holds promise for future treatment of glioblastoma.
Collapse
Affiliation(s)
- Mikael Johansson
- NorLux Neuro-Oncology Laboratory, Department of Oncology, Centre de Recherche Public de la Santé, Luxembourg, Luxembourg, Sweden
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
LRIG1 is a triple threat: ERBB negative regulator, intestinal stem cell marker and tumour suppressor. Br J Cancer 2013; 108:1765-70. [PMID: 23558895 PMCID: PMC3658528 DOI: 10.1038/bjc.2013.138] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
In baseball parlance, a triple threat is a person who can run, hit and throw with aplomb. Leucine-rich repeats and immunoglobulin-like domains 1 (LRIG1) is a cell surface protein that antagonises ERBB receptor signalling by downregulating receptor levels. Over 10 years ago, Hedman et al postulated that LRIG1 might be a tumour suppressor. Recently, Powell et al provided in vivo evidence substantiating that claim by demonstrating that Lrig1 loss in mice leads to spontaneously arising, highly penetrant intestinal adenomas. Interestingly, Lrig1 also marks stem cells in the gut, suggesting a potential role for Lrig1 in maintaining intestinal epithelial homeostasis. In this review, we will discuss the ability of LRIG1 to act as a triple threat: pan-ERBB negative regulator, intestinal stem cell marker and tumour suppressor. We will summarise studies of LRIG1 expression in human cancers and discuss possible related roles for LRIG2 and LRIG3.
Collapse
|
32
|
Ghasimi S, Haapasalo H, Eray M, Korhonen K, Brännström T, Hedman H, Andersson U. Immunohistochemical analysis of LRIG proteins in meningiomas: correlation between estrogen receptor status and LRIG expression. J Neurooncol 2012; 108:435-41. [PMID: 22484910 DOI: 10.1007/s11060-012-0856-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2011] [Accepted: 03/22/2012] [Indexed: 01/16/2023]
Abstract
The leucine-rich repeats and immunoglobulin-like domains (LRIG) protein family is comprised of three integral membrane proteins: LRIG1, LRIG2, and LRIG3. LRIG1 is a negative regulator of growth factor signaling. The expression and subcellular localization of LRIG proteins have prognostic implications in primary brain tumors, such as oligodendrogliomas and astrocytomas. The expression of LRIG proteins has not previously been studied in meningiomas. In this study, the expression of LRIG1, LRIG2, and LRIG3 was analyzed in 409 meningiomas by immunohistochemistry, and potential associations between LRIG protein expression and tumor grade, gender, progesterone receptor status, and estrogen receptor (ER) status were investigated. The LRIG proteins were most often expressed in the cytoplasm, though LRIG1 also showed prominent nuclear expression. Cytoplasmic expression of LRIG1 and LRIG2 correlated with histological subtypes of meningiomas (p = 0.038 and 0.013, respectively). Nuclear and cytoplasmic expression of LRIG1 was correlated with ER status (p = 0.003 and 0.004, respectively), as was cytoplasmic expression of LRIG2 (p = 0.006). This study is the first to examine the expression of LRIG proteins in meningiomas, and it shows a correlation between ER status and the expression of LRIG1 and LRIG2, which suggests a possible role for LRIG proteins in meningioma pathogenesis.
Collapse
Affiliation(s)
- Soma Ghasimi
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, Sweden.
| | | | | | | | | | | | | |
Collapse
|
33
|
Krig SR, Frietze S, Simion C, Miller JK, Fry WHD, Rafidi H, Kotelawala L, Qi L, Griffith OL, Gray JW, Carraway KL, Sweeney C. Lrig1 is an estrogen-regulated growth suppressor and correlates with longer relapse-free survival in ERα-positive breast cancer. Mol Cancer Res 2011; 9:1406-17. [PMID: 21821674 DOI: 10.1158/1541-7786.mcr-11-0227] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Lrig1 is the founding member of the Lrig family and has been implicated in the negative regulation of several oncogenic receptor tyrosine kinases including ErbB2. Lrig1 is expressed at low levels in several cancer types but is overexpressed in some prostate and colorectal tumors. Given this heterogeneity, whether Lrig1 functions to suppress or promote tumor growth remains a critical question. Previously, we found that Lrig1 was poorly expressed in ErbB2-positive breast cancer, suggesting that Lrig1 has a growth-inhibitory role in this tumor type. However, breast cancer is a complex disease, with ErbB2-positive tumors accounting for just 25% of all breast cancers. To gain a better understanding of the role of Lrig1 in breast cancer, we examined its expression in estrogen receptor α (ERα)-positive disease which accounts for the majority of breast cancers. We find that Lrig1 is expressed at significantly higher levels in ERα-positive disease than in ERα-negative disease. Our study provides a molecular rationale for Lrig1 enrichment in ERα-positive disease by showing that Lrig1 is a target of ERα. Estrogen stimulates Lrig1 accumulation and disruption of this induction enhances estrogen-dependent tumor cell growth, suggesting that Lrig1 functions as an estrogen-regulated growth suppressor. In addition, we find that Lrig1 expression correlates with prolonged relapse-free survival in ERα-positive breast cancer, identifying Lrig1 as a new prognostic marker in this setting. Finally, we show that ErbB2 activation antagonizes ERα-driven Lrig1 expression, providing a mechanistic explanation for Lrig1 loss in ErbB2-positive breast cancer. This work provides strong evidence for a growth-inhibitory role for Lrig1 in breast cancer.
Collapse
Affiliation(s)
- Sheryl R Krig
- Division of Basic Sciences, University of California Davis Cancer Center, Sacramento, CA 95817, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|