1
|
Sheida A, Farshadi M, Mirzaei A, Najjar Khalilabad S, Zarepour F, Taghavi SP, Hosseini Khabr MS, Ravaei F, Rafiei S, Mosadeghi K, Yazdani MS, Fakhraie A, Ghattan A, Zamani Fard MM, Shahyan M, Rafiei M, Rahimian N, Talaei Zavareh SA, Mirzaei H. Potential of Natural Products in the Treatment of Glioma: Focus on Molecular Mechanisms. Cell Biochem Biophys 2024; 82:3157-3208. [PMID: 39150676 DOI: 10.1007/s12013-024-01447-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] [Accepted: 07/17/2024] [Indexed: 08/17/2024]
Abstract
Despite the waning of traditional treatments for glioma due to possible long-term issues, the healing possibilities of substances derived from nature have been reignited in the scientific community. These natural substances, commonly found in fruits and vegetables, are considered potential alternatives to pharmaceuticals, as they have been shown in prior research to impact pathways surrounding cancer progression, metastases, invasion, and resistance. This review will explore the supposed molecular mechanisms of different natural components, such as berberine, curcumin, coffee, resveratrol, epigallocatechin-3-gallate, quercetin, tanshinone, silymarin, coumarin, and lycopene, concerning glioma treatment. While the benefits of a balanced diet containing these compounds are widely recognized, there is considerable scope for investigating the efficacy of these natural products in treating glioma.
Collapse
Affiliation(s)
- Amirhossein Sheida
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | | | - Amirhossein Mirzaei
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Shakiba Najjar Khalilabad
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Zarepour
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Seyed Pouya Taghavi
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Maryam Sadat Hosseini Khabr
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Fatemeh Ravaei
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Sara Rafiei
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences (IUMS), Tehran, Iran
- Department of Internal Medicine, School of Medicine, Firoozgar Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Kimia Mosadeghi
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad Sepehr Yazdani
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Ali Fakhraie
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Alireza Ghattan
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad Masoud Zamani Fard
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Maryam Shahyan
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Moein Rafiei
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Neda Rahimian
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences (IUMS), Tehran, Iran.
- Department of Internal Medicine, School of Medicine, Firoozgar Hospital, Iran University of Medical Sciences, Tehran, Iran.
| | | | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
2
|
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: 1.5] [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
|
3
|
Wang W, Zhang A, Hao Y, Wang G, Jia Z. The emerging role of miR-19 in glioma. J Cell Mol Med 2018; 22:4611-4616. [PMID: 30073755 PMCID: PMC6156349 DOI: 10.1111/jcmm.13788] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Revised: 05/31/2018] [Accepted: 06/12/2018] [Indexed: 12/13/2022] Open
Abstract
Glioma has been regarded as the most common, highly proliferative and invasive brain tumour. Advances in research of miRNAs in glioma are toward further understanding of the pathogenesis of glioma. MiR‐19, a member of miR‐17~92 cluster, was reported to play an oncogenic role in tumourigenesis. Here we review the identified data about the effect of miR‐19 on proliferation, apoptosis, migration and invasion of glioma cells, the target genes regulated by miR‐19, and correlation of miR‐19 with the sensitivity of glioma cells to chemotherapy and radiotherapy. It is concluded that miR‐19 plays an important role in the pathogenesis of glioma and can be a potential target for gene therapy of glioma.
Collapse
Affiliation(s)
- Weihan Wang
- Department of Neurosurgery, Tianjin Medical University, General Hospital, Tianjin Neurological Institute, Laboratory of Neuro-Oncology, Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China
| | - Anling Zhang
- Department of Neurosurgery, Tianjin Medical University, General Hospital, Tianjin Neurological Institute, Laboratory of Neuro-Oncology, Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China
| | - Yubing Hao
- Department of Neurosurgery, Tianjin Medical University, General Hospital, Tianjin Neurological Institute, Laboratory of Neuro-Oncology, Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China
| | - Guangxiu Wang
- Department of Neurosurgery, Tianjin Medical University, General Hospital, Tianjin Neurological Institute, Laboratory of Neuro-Oncology, Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China
| | - Zhifan Jia
- Department of Neurosurgery, Tianjin Medical University, General Hospital, Tianjin Neurological Institute, Laboratory of Neuro-Oncology, Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China
| |
Collapse
|
4
|
Xiao Q, Dong M, Cheng F, Mao F, Zong W, Wu K, Wang H, Xie R, Wang B, Lei T, Guo D. LRIG2 promotes the proliferation and cell cycle progression of glioblastoma cells in vitro and in vivo through enhancing PDGFRβ signaling. Int J Oncol 2018; 53:1069-1082. [PMID: 30015847 PMCID: PMC6065455 DOI: 10.3892/ijo.2018.4482] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 06/29/2018] [Indexed: 01/12/2023] Open
Abstract
The leucine-rich repeats and immunoglobulin-like domains (LRIG) gene family, comprising LRIG1, 2 and 3, encodes integral membrane proteins. It has been well established that LRIG1 negatively regulates multiple growth factor signaling pathways and is considered to be a tumor suppressor; however, the biological functions of LRIG2 remain largely unexplored. It was previously demonstrated that LRIG2 positively regulates epidermal growth factor receptor (EGFR) signaling, the most common aberrant receptor tyrosine kinase (RTK) signaling in glioblastoma multiforme (GBM), which promotes GBM growth. In the present study, the effect of LRIG2 on the proliferation of GBM cells was further addressed, as well as the possible mechanisms underlying the regulatory effect of LRIG2 on platelet-derived growth factor receptor β (PDGFRβ) signaling, another common oncogenic RTK signaling pathway in GBM. First, the expression levels of endogenous LRIG2 and PDGFRβ were found to vary notably in human GBM, and the LRIG2 expression level was positively correlated with the expression level of PDGFRβ. Furthermore, to the best of our knowledge, this is the first study to demonstrate that LRIG2 promoted the PDGF-BB-induced proliferation of GBM cells in vitro and in vivo through regulating the PDGFRβ signaling-mediated cell cycle progression. Mechanistically, LRIG2 has the ability to physically interact with PDGFRβ, promoting the total expression and the activation of PDGFRβ, and enhancing its downstream signaling pathways of Akt and signal transducer and activator of transcription 3 and the effectors of key regulators of cell cycle progression, resulting in increased GBM cell proliferation. Collectively, these data indicated that LRIG2 may serve as a tumor promoter gene in gliomagenesis by positively regulating PDGFRβ signaling, another important oncogenic RTK signaling pathway, in addition to the previously reported EGFR signaling in GBM modulated by LRIG2, and validated LRIG2 as a promising therapeutic target for the treatment of GBM characterized by multiple aberrant RTK signaling.
Collapse
Affiliation(s)
- Qungen Xiao
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Minhai Dong
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Fangling Cheng
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Feng Mao
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Weifeng Zong
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Kang Wu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Heping Wang
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Ruifan Xie
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Baofeng Wang
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Ting Lei
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Dongsheng Guo
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| |
Collapse
|
5
|
Mao F, Wang B, Xiao Q, Cheng F, Lei T, Guo D. LRIG proteins in glioma: Functional roles, molecular mechanisms, and potential clinical implications. J Neurol Sci 2017; 383:56-60. [PMID: 29246624 DOI: 10.1016/j.jns.2017.10.025] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 09/26/2017] [Accepted: 10/17/2017] [Indexed: 12/26/2022]
Abstract
Gliomas are the most common intracranial tumors of the nervous system. These tumors are characterized by unlimited cell proliferation and excessive invasiveness. Despite the advances in diagnostic imaging, microneurosurgical techniques, radiation therapy, and chemotherapy, significant increases in the progression free survival of glioma patients have not been achieved. Improvements in our understanding of the molecular subtypes of gliomas and the underlying alterations in specific signaling pathways may impact both the diagnosis and the treatment strategies for patients with gliomas. Growth factors and their corresponding receptor tyrosine kinases are associated with oncogenesis and development of tumors in numerous human cancer types, including glioma. Leucine-rich repeats and immunoglobulin-like domains (LRIG) are integral membrane proteins which contain three vertebrate members including LRIG1, LRIG2 and LRIG3. They mainly function as regulators of growth factor signaling. Specifically, LRIG1 has been identified as a tumor suppressor in human cancers. In contrast, LRIG2 appears to function as a tumor promoter, while LRIG3 appears to have a function similar to that of LRIG1. In the present review, we summarize the functional roles, molecular mechanisms, and clinical perspectives of LRIG proteins in gliomas and propose that these proteins may be useful in the future as targets for treatment and prognostication in glioma patients.
Collapse
Affiliation(s)
- Feng Mao
- Department of Neurosurgery and Sino-German Neuro-Oncology Molecular Laboratory, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Baofeng Wang
- Department of Neurosurgery and Sino-German Neuro-Oncology Molecular Laboratory, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qungen Xiao
- Department of Neurosurgery and Sino-German Neuro-Oncology Molecular Laboratory, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fangling Cheng
- Department of Neurosurgery and Sino-German Neuro-Oncology Molecular Laboratory, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ting Lei
- Department of Neurosurgery and Sino-German Neuro-Oncology Molecular Laboratory, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dongsheng Guo
- Department of Neurosurgery and Sino-German Neuro-Oncology Molecular Laboratory, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
6
|
Chen L, Wang W, Zhu S, Jin X, Wang J, Zhu J, Zhou Y. MicroRNA-590-3p enhances the radioresistance in glioblastoma cells by targeting LRIG1. Exp Ther Med 2017; 14:1818-1824. [PMID: 28810655 DOI: 10.3892/etm.2017.4697] [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: 08/14/2015] [Accepted: 01/06/2017] [Indexed: 12/31/2022] Open
Abstract
microRNA (miR)-590 has been found to serve potential roles in cancer development; however, the expression and function of miR-590 in human gliomas remains to be elucidated. The present study aimed to investigate the expression of miR-590 in human glioma tissues and radioresistant human glioblastoma cells (U251R), and to determine the effect and related molecular mechanism of miR-590-3p on the radiosensitivity of U251R cells in vitro. The results from reverse transcription-quantitative polymerase chain reaction indicated that miR-590-3p was upregulated in human glioma tissues and radioresistant human glioblastoma cells, and that miR-590-3p expression was higher in high grade than in low grade gliomas. In vitro experiments revealed that the miR-590-3p inhibitor enhanced the radiosensitivity of U251R cells by suppressing cell viability, decreasing colony formation capacity and increasing cell apoptosis rate, as demonstrated by MTT, colony formation and flow cytometry analyses. A luciferase reporter assay demonstrated that leucine-rich repeats and immunoglobulin-like domains protein 1 (LRIG1) was a direct target of miR-590-3p. Furthermore, it was demonstrated that the effect of miR-590-3p suppression on cell viability, colony formation capacity and cell apoptosis rate was attenuated by the knockdown of LRIG1 in the U251R cells. In conclusion, the present study revealed that miR-590-3p was upregulated in human glioma tissues and radioresistant human glioblastoma cells, and miR-590-3p contributes to the radioresistance of human glioblastoma cells by directly targeting LRIG1. These findings may provide potential therapeutic strategies to prevent radioresistance in human gliomas.
Collapse
Affiliation(s)
- Long Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China.,Department of Neurosurgery, The Affiliated Hospital of Nanjing University of Chinese Medicine, Kunshan, Jiangsu 215300, P.R. China
| | - Wenhua Wang
- Department of Neurosurgery, The Affiliated Hospital of Nanjing University of Chinese Medicine, Kunshan, Jiangsu 215300, P.R. China
| | - Shengqiang Zhu
- Department of Neurosurgery, The Affiliated Hospital of Nanjing University of Chinese Medicine, Kunshan, Jiangsu 215300, P.R. China
| | - Xuegang Jin
- Department of Neurosurgery, The Affiliated Hospital of Nanjing University of Chinese Medicine, Kunshan, Jiangsu 215300, P.R. China
| | - Jian Wang
- Department of Neurosurgery, The Affiliated Hospital of Nanjing University of Chinese Medicine, Kunshan, Jiangsu 215300, P.R. China
| | - Jianfang Zhu
- Department of Neurosurgery, The Affiliated Hospital of Nanjing University of Chinese Medicine, Kunshan, Jiangsu 215300, P.R. China
| | - Youxin Zhou
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| |
Collapse
|
7
|
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: 1.3] [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
|
8
|
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
|
9
|
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: 1.7] [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
|
10
|
Guo D, Yang H, Guo Y, Xiao Q, Mao F, Tan Y, Wan X, Wang B, Lei T. LRIG3 modulates proliferation, apoptosis and invasion of glioblastoma cells as a potent tumor suppressor. J Neurol Sci 2015; 350:61-8. [DOI: 10.1016/j.jns.2015.02.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Revised: 01/13/2015] [Accepted: 02/08/2015] [Indexed: 10/24/2022]
|
11
|
Kondapalli KC, Llongueras JP, Capilla-González V, Prasad H, Hack A, Smith C, Guerrero-Cázares H, Quiñones-Hinojosa A, Rao R. A leak pathway for luminal protons in endosomes drives oncogenic signalling in glioblastoma. Nat Commun 2015; 6:6289. [PMID: 25662504 PMCID: PMC4354686 DOI: 10.1038/ncomms7289] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Accepted: 01/12/2015] [Indexed: 12/15/2022] Open
Abstract
Epidermal growth factor receptor (EGFR) signalling is a potent driver of glioblastoma, a malignant and lethal form of brain cancer. Disappointingly, inhibitors targeting receptor tyrosine kinase activity are not clinically effective and EGFR persists on the plasma membrane to maintain tumour growth and invasiveness. Here we show that endolysosomal pH is critical for receptor sorting and turnover. By functioning as a leak pathway for protons, the Na(+)/H(+) exchanger NHE9 limits luminal acidification to circumvent EGFR turnover and prolong downstream signalling pathways that drive tumour growth and migration. In glioblastoma, NHE9 expression is associated with stem/progenitor characteristics, radiochemoresistance, poor prognosis and invasive growth in vitro and in vivo. Silencing or inhibition of NHE9 in brain tumour-initiating cells attenuates tumoursphere formation and improves efficacy of EGFR inhibitor. Thus, NHE9 mediates inside-out control of oncogenic signalling and is a highly druggable target for pan-specific receptor clearance in cancer therapy.
Collapse
Affiliation(s)
- Kalyan C. Kondapalli
- Department of Physiology, The Johns Hopkins University School of Medicine, 725 North Wolfe Street, Baltimore, MD 21205, USA
| | - Jose P. Llongueras
- Department of Physiology, The Johns Hopkins University School of Medicine, 725 North Wolfe Street, Baltimore, MD 21205, USA
| | - Vivian Capilla-González
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, 725 North Wolfe Street, Baltimore, MD 21205, USA
| | - Hari Prasad
- Department of Physiology, The Johns Hopkins University School of Medicine, 725 North Wolfe Street, Baltimore, MD 21205, USA
| | - Anniesha Hack
- Department of Physiology, The Johns Hopkins University School of Medicine, 725 North Wolfe Street, Baltimore, MD 21205, USA
| | - Christopher Smith
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, 725 North Wolfe Street, Baltimore, MD 21205, USA
| | - Hugo Guerrero-Cázares
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, 725 North Wolfe Street, Baltimore, MD 21205, USA
| | - Alfredo Quiñones-Hinojosa
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, 725 North Wolfe Street, Baltimore, MD 21205, USA
| | - Rajini Rao
- Department of Physiology, The Johns Hopkins University School of Medicine, 725 North Wolfe Street, Baltimore, MD 21205, USA
| |
Collapse
|
12
|
Tian P, Peng C, Zhang L. Biodegradable polymeric gene delivering nanoscale hybrid micelles enhance the suppression effect of LRIG1 in breast cancer. RSC Adv 2015. [DOI: 10.1039/c5ra03740a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Biodegradable polymeric gene delivering nanoscale hybrid micelles enhance the suppression effect of LRIG1 in breast cancer.
Collapse
Affiliation(s)
- Peng Tian
- Chengdu Medical College – The First Affiliated Hospital of Chengdu Medical College
- Chengdu
- China
| | - ChaoMing Peng
- Chengdu Medical College – The First Affiliated Hospital of Chengdu Medical College
- Chengdu
- China
| | - Lei Zhang
- Chengdu Medical College – The First Affiliated Hospital of Chengdu Medical College
- Chengdu
- China
| |
Collapse
|
13
|
Yao Y, Ma J, Xue Y, Wang P, Li Z, Li Z, Hu Y, Shang X, Liu Y. MiR-449a exerts tumor-suppressive functions in human glioblastoma by targeting Myc-associated zinc-finger protein. Mol Oncol 2014; 9:640-56. [PMID: 25487955 DOI: 10.1016/j.molonc.2014.11.003] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Revised: 11/14/2014] [Accepted: 11/14/2014] [Indexed: 12/16/2022] Open
Abstract
Glioblastoma (GBM) is one of the most common and aggressive primary brain tumors in adults. Deregulated expression of microRNAs (miRNAs) has been associated with GBM progression through alterations in either oncogenic or tumor suppressor targets. Here, we elucidated the function and the possible molecular mechanisms of miR-449a in human GBM cell lines and tumor specimens-derived glioblastoma stem cells (GSCs). Quantitative real-time PCR demonstrated that miR-449a was down-regulated in human GBM cell lines and GSCs. Functionally, miR-449a acted as a tumor suppressor by reducing cell proliferation, migration and invasion as well as inducing apoptosis in human GBM cell lines and GSCs. Myc-associated zinc-finger protein (MAZ) was identified as a direct target of miR-449a, mediating these tumor-suppressive effects, demonstrated by Western blot assay and luciferase assays. Moreover, over-expression of miR-449a inhibited the expression of Podoplanin (PDPN) by down-regulating MAZ which could positively control the promoter activities via binding to the promoter of PDPN, demonstrated by luciferase assays and chromatin immunoprecipitation assays. Further, the PI3K/AKT pathway was blocked when MAZ was down-regulated by miR-449a. This process was coincided with the up-regulation of apoptotic proteins and the down-regulation of anti-apoptotic proteins, MMP2 and MMP9. Furthermore, nude mice carrying over-expressed miR-449a combined with knockdown MAZ tumors produced the smallest tumors and the highest survival. These results elucidated a novel molecular mechanism of GBM progression, and may thus suggest a promising application for GBM treatment.
Collapse
Affiliation(s)
- Yilong Yao
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China
| | - Jun Ma
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang 110001, People's Republic of China; Institute of Pathology and Pathophysiology, China Medical University, Shenyang 110001, People's Republic of China
| | - Yixue Xue
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang 110001, People's Republic of China; Institute of Pathology and Pathophysiology, China Medical University, Shenyang 110001, People's Republic of China
| | - Ping Wang
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang 110001, People's Republic of China; Institute of Pathology and Pathophysiology, China Medical University, Shenyang 110001, People's Republic of China
| | - Zhen Li
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China
| | - Zhiqing Li
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang 110001, People's Republic of China; Institute of Pathology and Pathophysiology, China Medical University, Shenyang 110001, People's Republic of China
| | - Yi Hu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China
| | - Xiuli Shang
- Department of Neurology, The First Affiliated Hospital, China Medical University, Shenyang 110001, People's Republic of China
| | - Yunhui Liu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China.
| |
Collapse
|
14
|
Soluble LRIG2 ectodomain is released from glioblastoma cells and promotes the proliferation and inhibits the apoptosis of glioblastoma cells in vitro and in vivo in a similar manner to the full-length LRIG2. PLoS One 2014; 9:e111419. [PMID: 25353163 PMCID: PMC4213030 DOI: 10.1371/journal.pone.0111419] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Accepted: 10/01/2014] [Indexed: 11/25/2022] Open
Abstract
The human leucine-rich repeats and immunoglobulin-like domains (LRIG) gene family contains LRIG1, 2 and 3, encoding integral membrane proteins with an ectodomain, a transmembrane domain and a cytoplasmic tail. LRIG1 negatively regulates multiple receptor tyrosine kinases signaling including the epidermal growth factor receptor (EGFR) and is a proposed tumor suppressor. The soluble LRIG1 ectodomain is demonstrated to be shed naturally and inhibit the progression of glioma. However, little is known regarding the functions of LRIG2. In oligodendroglioma, LRIG2 expression is associated with poor survival, suggesting that LRIG2 might have different functions compared with LRIG1. Since soluble LRIG1 ectodomain has a similar function to the full-length LRIG1, we hypothesize that the different roles exerted by LRIG2 and LRIG1 result from the difference of their ectodomains. Here, we addressed the functions of LRIG2 and LRIG2 ectodomain in the proliferation and apoptosis of glioma and the possible underlying mechanisms. Firstly, we found that LRIG2 expression levels positively correlated with the grade of glioma. Further, we demonstrated for the first time that soluble LRIG2 ectodomain was capable of being released from glioblastoma cells and exerted a pro-proliferative effect. Overexpression of LRIG2 ectodomain promoted the proliferation and inhibited the apoptosis of glioblastoma cells in vitro and in vivo in a similar manner to the full-length LRIG2. Both full-length LRIG2 and LRIG2 ectodomain were found to physically interact with EGFR, enhance the activation of EGFR and its downstream PI3 K/Akt pathway. To our knowledge, this is the first report demonstrating that soluble LRIG2 ectodomain is capable of being released from glioblastoma cells and exerts a similar role to the full-length LRIG2 in the regulation of EGFR signaling in the progression of glioblastoma. LRIG2 ectodomain, with potent pro-tumor effects, holds promise for providing a new therapeutic target for the treatment of glioblastoma.
Collapse
|
15
|
Fathima Hurmath K, Ramaswamy P, Nandakumar DN. IL-1β microenvironment promotes proliferation, migration, and invasion of human glioma cells. Cell Biol Int 2014; 38:1415-22. [DOI: 10.1002/cbin.10353] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Accepted: 06/09/2014] [Indexed: 12/11/2022]
Affiliation(s)
- K. Fathima Hurmath
- Department of Neurochemistry; National Institute of Mental Health and Neuro Sciences (NIMHANS); Bengaluru 560029 India
| | - Palaniswamy Ramaswamy
- Department of Neurochemistry; National Institute of Mental Health and Neuro Sciences (NIMHANS); Bengaluru 560029 India
| | | |
Collapse
|
16
|
Nagata M, Nakamura T, Sotozono C, Inatomi T, Yokoi N, Kinoshita S. LRIG1 as a potential novel marker for neoplastic transformation in ocular surface squamous neoplasia. PLoS One 2014; 9:e93164. [PMID: 24709893 PMCID: PMC3977825 DOI: 10.1371/journal.pone.0093164] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Accepted: 03/01/2014] [Indexed: 11/18/2022] Open
Abstract
The leucine rich repeats and immunoglobulin-like protein 1 (LRIG1) is a newly discovered negative regulator of epidermal growth factor receptor (EGFR) and a proposed tumor suppressor. It is not universally downregulated in human cancers, and its role in neoplastic transformation and tumorigenesis is not well-documented. In this study, we show the expression of LRIG1 as a novel potential marker for neoplastic transformation in ocular-surface squamous neoplasia (OSSN). The following two groups were included in this study: 1) benign group (3 cases; 1 with papilloma and 2 with dysplasia) and 2) malignant group (3 cases with squamous cell carcinoma (SCC)). In both groups, immunofluorescence analysis was firstly performed for keratins 4, 12, 13, and 15 to characterize the state of differentiation, and for Ki67 to evaluate the proliferation activity. Subsequently, LRIG1 and EGFR expression was analyzed. Either keratin 4 and/or 13, both non-keratinized epithelial cell markers, were generally expressed in both groups, except for 1 severe SCC case. Keratin 15, an undifferentiated basal cell marker, was more strongly expressed in the malignant cases than in the benign cases. The Ki67 index was significantly higher (P<0.002) in the malignant group (33.2%) than in the benign group (10.9%). LRIG1 expression was limited to basal epithelial cells in normal corneal epithelial tissue. Interestingly, LRIG1 was expressed throughout the epithelium in all the benign cases. In contrast, its expression was limited or totally disappeared in the malignant cases. Inversely, EGFR staining was faintly expressed in the benign cases, yet strongly expressed in the malignant cases. Malignant tissue with proliferative potential presented EGFR overexpression and inverse downregulation of LRIG1, consistent with LRIG1 being a suppressor of neoplastic transformation by counteracting the tumor growth property of EGFR. Our findings indicate that downregulation of LRIG1 is possibly a novel potential marker of transformation and tumorigenesis in OSSN cases.
Collapse
Affiliation(s)
- Maho Nagata
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Takahiro Nakamura
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, Kyoto, Japan
- Research Center for Inflammation and Regenerative Medicine, Doshisha University, Kyoto, Japan
- * E-mail:
| | - Chie Sotozono
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Tsutomu Inatomi
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Norihiko Yokoi
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Shigeru Kinoshita
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
17
|
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
|
18
|
Activation of NMDA receptor of glutamate influences MMP-2 activity and proliferation of glioma cells. Neurol Sci 2013; 35:823-9. [DOI: 10.1007/s10072-013-1604-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Accepted: 12/10/2013] [Indexed: 02/07/2023]
|
19
|
Effects of Rab27a on proliferation, invasion, and anti-apoptosis in human glioma cell. Tumour Biol 2013; 34:2195-203. [PMID: 23553027 DOI: 10.1007/s13277-013-0756-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2013] [Accepted: 03/18/2013] [Indexed: 12/22/2022] Open
Abstract
This study aims to investigate the relationship between Rab27a and the characteristics of glioma cell U251 such as proliferation, apoptosis, and invasion and to provide an experimental basis for future therapy in human glioma. Recombinant plasmid of pcDNA3.1-Rab27a was constructed and transfected into U251 cells with the help of Lipofectamine™2000. The expression of Rab27a was detected by Western blot. Cell viability, cell cycle, cell apoptosis, and cell migration were analyzed, respectively, by (3-(4,5)-dimethylthi-azol-2-yl)-2,5-diphenytetrazolium bromide (MTT) assay, flow cytometry, and Transwell invasion chamber methods. Meanwhile, the effect of Rab27a on secretion of cathepsin D in U251 cells was also examined. With the help of luciferase reporter assay system, the relationship between miR-124 and gene Rab27a expression was explored. Western blot showed that the expression of Rab27a was significantly increased in pcDNA3.1-Rab27a transfection group (p < 0.01) and that was significantly decreased in Rab27a-shRNA transfection group (p < 0.01) compared with control group. MTT assay, flow cytometry, and Transwell invasion chamber experiment indicated that cell viability (p < 0.01), proliferation index (p < 0.05), and invasion ability (p < 0.01) were improved significantly in pcDNA3.1-Rab27a transfection group compared with control group and that cell viability (p < 0.01), proliferation index (p < 0.05), and invasion ability (p < 0.01) were reduced markedly in Rab27a-shRNA transfection group compared with control group. The apoptosis analysis by flow cytometry demonstrated that the ratio of apoptosis in pcDNA3.1-Rab27a transfection group was significantly lower than that in control group (p < 0.05) and the ratio was notably higher in Rab27a-shRNAtransfection group than that in the control group. Cathepsin D activity assay indicated that the release of cathepsin D was enhanced in pcDNA3.1-Rab27a transfection group compared to that in the control group (p < 0.05). Rab27a could increase the glioma cell ability, promote proliferation and invasion, and suppress cell apoptosis. The above-stated effects of Rab27a possibly were exerted by increasing the secretion of cathepsin D and regulated by miR-124. In addition, the inhibition of expression of Rab27a perhaps benefited the therapy for glioma patients.
Collapse
|