1
|
Downstream Targets of VHL/HIF-α Signaling in Renal Clear Cell Carcinoma Progression: Mechanisms and Therapeutic Relevance. Cancers (Basel) 2023; 15:cancers15041316. [PMID: 36831657 PMCID: PMC9953937 DOI: 10.3390/cancers15041316] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/09/2023] [Accepted: 02/11/2023] [Indexed: 02/22/2023] Open
Abstract
The clear cell variant of renal cell carcinoma (ccRCC) is the most common renal epithelial malignancy and responsible for most of the deaths from kidney cancer. Patients carrying inactivating mutations in the Von Hippel-Lindau (VHL) gene have an increased proclivity to develop several types of tumors including ccRCC. Normally, the Hypoxia Inducible Factor alpha (HIF-α) subunits of the HIF heterodimeric transcription factor complex are regulated by oxygen-dependent prolyl-hydroxylation, VHL-mediated ubiquitination and proteasomal degradation. Loss of pVHL function results in elevated levels of HIF-α due to increased stability, leading to RCC progression. While HIF-1α acts as a tumor suppressor, HIF-2α promotes oncogenic potential by driving tumor progression and metastasis through activation of hypoxia-sensitive signaling pathways and overexpression of HIF-2α target genes. One strategy to suppress ccRCC aggressiveness is directed at inhibition of HIF-2α and the associated molecular pathways leading to cell proliferation, angiogenesis, and metastasis. Indeed, clinical and pre-clinical data demonstrated the effectiveness of HIF-2α targeted therapy in attenuating ccRCC progression. This review focuses on the signaling pathways and the involved genes (cyclin D, c-Myc, VEGF-a, EGFR, TGF-α, GLUT-1) that confer oncogenic potential downstream of the VHL-HIF-2α signaling axis in ccRCC. Discussed as well are current treatment options (including receptor tyrosine kinase inhibitors such as sunitinib), the medical challenges (high prevalence of metastasis at the time of diagnosis, refractory nature of advanced disease to current treatment options), scientific challenges and future directions.
Collapse
|
2
|
Taub M, Mahmoudzadeh NH, Tennessen J, Sudarshan S. Renal oncometabolite L-2-hydroxyglutarate imposes a block in kidney tubulogenesis: Evidence for an epigenetic basis for the L-2HG-induced impairment of differentiation. Front Endocrinol (Lausanne) 2022; 13:932286. [PMID: 36133305 PMCID: PMC9483015 DOI: 10.3389/fendo.2022.932286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 08/12/2022] [Indexed: 12/03/2022] Open
Abstract
2-Hydroxyglutarate (2HG) overproducing tumors arise in a number of tissues, including the kidney. The tumorigenesis resulting from overproduced 2HG has been attributed to the ability of 2HG alter gene expression by inhibiting α-ketoglutarate (αKG)-dependent dioxygenases, including Ten-eleven-Translocation (TET) enzymes. Genes that regulate cellular differentiation are reportedly repressed, blocking differentiation of mesenchymal cells into myocytes, and adipocytes. In this report, the expression of the enzyme responsible for L2HG degradation, L-2HG dehydrogenase (L2HGDH), is knocked down, using lentiviral shRNA, as well as siRNA, in primary cultures of normal Renal Proximal Tubule (RPT) cells. The knockdown (KD) results in increased L-2HG levels, decreased demethylation of 5mC in genomic DNA, and increased methylation of H3 Histones. Consequences include reduced tubulogenesis by RPT cells in matrigel, and reduced expression of molecular markers of differentiation, including membrane transporters as well as HNF1α and HNF1β, which regulate their transcription. These results are consistent with the hypothesis that oncometabolite 2HG blocks RPT differentiation by altering the methylation status of chromatin in a manner that impedes the transcriptional events required for normal differentiation. Presumably, similar alterations are responsible for promoting the expansion of renal cancer stem-cells, increasing their propensity for malignant transformation.
Collapse
Affiliation(s)
- Mary Taub
- Biochemistry Department, School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, United States
| | | | - Jason M. Tennessen
- Department of Biology, Indiana University, Bloomington, IN, United States
| | - Sunil Sudarshan
- Department of Urology, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
3
|
Ji Y, Kumar R, Gokhale A, Chao HP, Rycaj K, Chen X, Li Q, Tang DG. LRIG1, a regulator of stem cell quiescence and a pleiotropic feedback tumor suppressor. Semin Cancer Biol 2021; 82:120-133. [PMID: 33476721 PMCID: PMC8286266 DOI: 10.1016/j.semcancer.2020.12.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 12/04/2020] [Accepted: 12/16/2020] [Indexed: 12/14/2022]
Abstract
LRIG1, leucine-rich repeats and immunoglobulin-like domains protein 1, was discovered more than 20 years ago and has been shown to be downregulated or lost, and to function as a tumor suppressor in several cancers. Another well-reported biological function of LRIG1 is to regulate and help enforce the quiescence of adult stem cells (SCs). In both contexts, LRIG1 regulates SC quiescence and represses tumor growth via, primarily, antagonizing the expression and activities of ERBB and other receptor tyrosine kinases (RTKs). We have recently reported that in treatment-naïve human prostate cancer (PCa), LRIG1 is primarily regulated by androgen receptor (AR) and is prominently overexpressed. In castration-resistant PCa (CRPC), both LRIG1 and AR expression becomes heterogeneous and, frequently, discordant. Importantly, in both androgen-dependent PCa and CRPC models, LRIG1 exhibits tumor-suppressive functions. Moreover, LRIG1 induction inhibits the growth of pre-established AR+ and AR− PCa. Here, upon a brief introduction of the LRIG1 and the LRIG family, we provide an updated overview on LRIG1 functions in regulating SC quiescence and repressing tumor development. We further highlight the expression, regulation and functions of LRIG1 in treatment-naïve PCa and CRPC. We conclude by offering the perspectives of identifying novel cancer-specific LRIG1-interacting signaling partners and developing LRIG1-based anti-cancer therapeutics and diagnostic/prognostic biomarkers.
Collapse
Affiliation(s)
- Yibing Ji
- Department of Pharmacology & Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA.
| | - Rahul Kumar
- Department of Pharmacology & Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Abhiram Gokhale
- Department of Pharmacology & Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Hseu-Ping Chao
- Department of Epigenetics & Mol. Carcinogenesis, the University of Texas M.D Anderson Cancer Center, Smithville, TX 78957, USA
| | - Kiera Rycaj
- Department of Pharmacology & Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA; Department of Epigenetics & Mol. Carcinogenesis, the University of Texas M.D Anderson Cancer Center, Smithville, TX 78957, USA
| | - Xin Chen
- Department of Pharmacology & Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Qiuhui Li
- Department of Pharmacology & Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA.
| | - Dean G Tang
- Department of Pharmacology & Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA; Department of Epigenetics & Mol. Carcinogenesis, the University of Texas M.D Anderson Cancer Center, Smithville, TX 78957, USA.
| |
Collapse
|
4
|
Liu C, Liu L, Wang K, Li XF, Ge LY, Ma RZ, Fan YD, Li LC, Liu ZF, Qiu M, Hao YC, Shi ZF, Xia CY, Strååt K, Huang Y, Ma LL, Xu D. VHL-HIF-2α axis-induced SMYD3 upregulation drives renal cell carcinoma progression via direct trans-activation of EGFR. Oncogene 2020; 39:4286-4298. [PMID: 32291411 DOI: 10.1038/s41388-020-1291-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Revised: 03/30/2020] [Accepted: 03/30/2020] [Indexed: 12/17/2022]
Abstract
It has been well established that the von Hippel-Lindau/hypoxia-inducible factor α (VHL-HIFα) axis and epidermal growth factor receptor (EGFR) signaling pathway play a critical role in the pathogenesis and progression of renal cell carcinoma (RCC). However, few studies have addressed the relationship between the two oncogenic drivers in RCC. SET and MYND domain-containing protein 3 (SMYD3) is a histone methyltransferase involved in gene transcription and oncogenesis, but its expression and function in RCC remain unclear. In the present study, we found that SMYD3 expression was significantly elevated in RCC tumors and correlated with advanced tumor stage, histological and nuclear grade, and shorter survival. Depletion of SMYD3 inhibited RCC cell proliferation, colony numbers, and xenograft tumor formation, while promoted apoptosis. Mechanistically, SMYD3 cooperates with SP1 to transcriptionally promote EGFR expression, amplifying its downstream signaling activity. TCGA data analyses revealed a significantly increased SMYD3 expression in primary RCC tumors carrying the loss-of-function VHL mutations. We further showed that HIF-2α can directly bind to the SMYD3 promoter and subsequently induced SMYD3 transcription and expression. Taken together, we identify the VHL/HIF-2α/SMYD3 signaling cascade-mediated EGFR hyperactivity through which SMYD3 promotes RCC progression. Our study suggests that SMYD3 is a potential therapeutic target and prognostic factor in RCC.
Collapse
Affiliation(s)
- Cheng Liu
- Department of Urology, Peking University Third Hospital, Beijing, China.
| | - Li Liu
- School of Nursing, Beijing University of Chinese Medicine, Beijing, China
| | - Kun Wang
- Key Lab for Cancer Prevention and Therapy, National Clinical Research Centre for Cancer, Department of Urology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Xiao-Feng Li
- Department of Urology, Shandong University Qilu Hospital, Jinan, China
| | - Li-Yuan Ge
- Department of Urology, Peking University Third Hospital, Beijing, China.,Department of Urology, The People's Hospital of Xinjiang Uyghur Autonomous Region, Xinjiang, China
| | - Run-Zhuo Ma
- Department of Urology, Peking University Third Hospital, Beijing, China
| | - Yi-Dong Fan
- Department of Urology, Shandong University Qilu Hospital, Jinan, China
| | - Lu-Chao Li
- Department of Urology, Shandong University Qilu Hospital, Jinan, China
| | - Zheng-Fang Liu
- Department of Urology, Shandong University Qilu Hospital, Jinan, China
| | - Min Qiu
- Department of Urology, Peking University Third Hospital, Beijing, China
| | - Yi-Chang Hao
- Department of Urology, Peking University Third Hospital, Beijing, China
| | - Zhen-Feng Shi
- Department of Urology, The People's Hospital of Xinjiang Uyghur Autonomous Region, Xinjiang, China
| | - Chuan-You Xia
- Department of Medicine, Division of Hematology, Bioclinicum and Centre for Molecular Medicine, Karolinska University Hospital Solna, Karolinska Institutet, Stockholm, Sweden
| | - Klas Strååt
- Department of Medicine, Division of Hematology, Bioclinicum and Centre for Molecular Medicine, Karolinska University Hospital Solna, Karolinska Institutet, Stockholm, Sweden
| | - Yi Huang
- Department of Urology, Peking University Third Hospital, Beijing, China
| | - Lu-Lin Ma
- Department of Urology, Peking University Third Hospital, Beijing, China.
| | - Dawei Xu
- Department of Medicine, Division of Hematology, Bioclinicum and Centre for Molecular Medicine, Karolinska University Hospital Solna, Karolinska Institutet, Stockholm, Sweden.,Karolinska Institute-Shandong University Collaborative Laboratory for Cancer and Stem Cell Research, Jinan, China
| |
Collapse
|
5
|
Zhang Q, Liu JH, Liu JL, Qi CT, Yan L, Chen Y, Yu Q. Activation and function of receptor tyrosine kinases in human clear cell renal cell carcinomas. BMC Cancer 2019; 19:1044. [PMID: 31690270 PMCID: PMC6833303 DOI: 10.1186/s12885-019-6159-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 09/13/2019] [Indexed: 02/07/2023] Open
Abstract
Background The receptor tyrosine kinases (RTKs) play critical roles in the development of cancers. Clear cell renal cell carcinoma (ccRCC) accounts for 75% of the RCC. The previous studies on the RTKs in ccRCCs mainly focused on their gene expressions. The activation and function of the RTKs in ccRCC have not been fully investigated. Methods In the present study, we analyzed the phosphorylation patterns of RTKs in human ccRCC patient samples, human ccRCC and papillary RCC cell lines, and other kidney tumor samples using human phospho-RTK arrays. We further established ccRCC patient-derived xenograft models in nude mice and assessed the effects of RTKIs (RTK Inhibitors) on the growth of these cancer cells. Immunofluorescence staining was used to detect the localization of keratin, vimentin and PDGFRβ in ccRCCs. Results We found that the RTK phosphorylation patterns of the ccRCC samples were all very similar, but different from that of the cell lines, other kidney tumor samples, as well as the adjacent normal tissues. 9 RTKs, EGFR1–3, Insulin R, PDGFRβ, VEGFR1, VEGFR2, HGFR and M-CSFR were found to be phosphorylated in the ccRCC samples. The adjacent normal tissues, on the other hand, had predominantly only two of the 4 EGFR family members, EGFR and ErbB4, phosphorylated. What’s more, the RTK phosphorylation pattern of the xenograft, however, was different from that of the primary tissue samples. Treatment of the xenograft nude mice with corresponding RTK inhibitors effectively inhibited the Erk1/2 signaling pathway as well as the growth of the tumors. In addition, histological staining of the cancer samples revealed that most of the PDGFRβ expressing cells were localized in the vimentin-positive periepithelial stroma. Conclusions Overall, we have identified a set of RTKs that are characteristically phosphorylated in ccRCCs. The phosphorylation of RTKs in ccRCCs were determined by the growing environments. These phosphorylated/activated RTKs will guide targeting drugs development of more effective therapies in ccRCCs. The synergistical inhibition of RTKIs combination on the ccRCC suggest a novel strategy to use a combination of RTKIs to treat ccRCCs.
Collapse
Affiliation(s)
- Qing Zhang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Room 2-224, Shanghai, 201203, China
| | - Jian-He Liu
- The Department of Urology, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, China
| | - Jing-Li Liu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Room 2-224, Shanghai, 201203, China
| | - Chun-Ting Qi
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Room 2-224, Shanghai, 201203, China
| | - Lei Yan
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Room 2-224, Shanghai, 201203, China
| | - Yu Chen
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Room 2-224, Shanghai, 201203, China
| | - Qiang Yu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Room 2-224, Shanghai, 201203, China.
| |
Collapse
|
6
|
Gbenedio OM, Bonnans C, Grun D, Wang CY, Hatch AJ, Mahoney MR, Barras D, Matli M, Miao Y, Garcia KC, Tejpar S, Delorenzi M, Venook AP, Nixon AB, Warren RS, Roose JP, Depeille P. RasGRP1 is a potential biomarker to stratify anti-EGFR therapy response in colorectal cancer. JCI Insight 2019; 5:127552. [PMID: 31237864 DOI: 10.1172/jci.insight.127552] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Colorectal cancer (CRC) is the third most frequent neoplastic disorder and is a main cause of tumor-related mortality as many patients progress to stage IV metastatic CRC. Standard care consists of combination chemotherapy (FOLFIRI or FOLFOX). Patients with WT KRAS typing are eligible to receive anti-EGFR therapy combined with chemotherapy. Unfortunately, predicting efficacy of CRC anti-EGFR therapy has remained challenging. Here we uncover that the EGFR-pathway component RasGRP1 acts as CRC tumor suppressor in the context of aberrant Wnt signaling. We find that RasGRP1 suppresses EGF-driven proliferation of colonic epithelial organoids. Having established that RasGRP1 dosage levels impacts biology, we focused on CRC patients next. Mining five different data platforms, we establish that RasGRP1 expression levels decrease with CRC progression and predict poor clinical outcome of patients. Lastly, deletion of one or two Rasgrp1 alleles makes CRC spheroids more susceptible to EGFR inhibition. Retrospective analysis of the CALGB80203 clinical trial shows that addition of anti-EGFR therapy to chemotherapy significantly improves outcome for CRC patients when tumors express low RasGRP1 suppressor levels. In sum, RasGRP1 is a unique biomarker positioned in the EGFR pathway and of potential relevance to anti-EGFR therapy for CRC patients.
Collapse
Affiliation(s)
| | - Caroline Bonnans
- Department of Anatomy, UCSF, San Francisco, California, USA.,Institut National de la Santé et de la Recherche Médicale, Montpellier, France
| | - Delphine Grun
- Bioinformatics Core Facility (BCF) at SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Chih-Yang Wang
- Department of Anatomy, UCSF, San Francisco, California, USA
| | - Ace J Hatch
- Department of Medicine, Duke University, Durham, North Carolina, USA
| | - Michelle R Mahoney
- Alliance Statistics and Data Center, Mayo Clinic, Rochester, Minnesota, USA
| | - David Barras
- Bioinformatics Core Facility (BCF) at SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Mary Matli
- Department of Surgery, UCSF, San Francisco, California, USA
| | - Yi Miao
- Department of Molecular and Cellular Physiology, Department of Structural Biology and Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, California, USA
| | - K Christopher Garcia
- Department of Molecular and Cellular Physiology, Department of Structural Biology and Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, California, USA
| | - Sabine Tejpar
- Institut National de la Santé et de la Recherche Médicale, Montpellier, France
| | - Mauro Delorenzi
- Bioinformatics Core Facility (BCF) at SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland.,Department of Oncology, University of Lausanne, Lausanne, Switzerland
| | - Alan P Venook
- Hematology/Oncology, Department of Medicine, UCSF, San Francisco, California, USA
| | - Andrew B Nixon
- Alliance Statistics and Data Center, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Jeroen P Roose
- Department of Anatomy, UCSF, San Francisco, California, USA
| | | |
Collapse
|
7
|
Karlsson T, Kvarnbrink S, Holmlund C, Botling J, Micke P, Henriksson R, Johansson M, Hedman H. LMO7 and LIMCH1 interact with LRIG proteins in lung cancer, with prognostic implications for early-stage disease. Lung Cancer 2018; 125:174-184. [PMID: 30429017 DOI: 10.1016/j.lungcan.2018.09.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 09/20/2018] [Accepted: 09/21/2018] [Indexed: 10/28/2022]
Abstract
OBJECTIVES The human leucine-rich repeats and immunoglobulin-like domains (LRIG) protein family comprises the integral membrane proteins LRIG1, LRIG2 and LRIG3. LRIG1 is frequently down-regulated in human cancer, and high levels of LRIG1 in tumor tissue are associated with favorable clinical outcomes in several tumor types including non-small cell lung cancer (NSCLC). Mechanistically, LRIG1 negatively regulates receptor tyrosine kinases and functions as a tumor suppressor. However, the details of the molecular mechanisms involved are poorly understood, and even less is known about the functions of LRIG2 and LRIG3. The aim of this study was to further elucidate the functions and molecular interactions of the LRIG proteins. MATERIALS AND METHODS A yeast two-hybrid screen was performed using a cytosolic LRIG3 peptide as bait. In transfected human cells, co-immunoprecipitation and co-localization experiments were performed. Proximity ligation assay was performed to investigate interactions between endogenously expressed proteins. Expression levels of LMO7 and LIMCH1 in normal and malignant lung tissue were investigated using qRT-PCR and through in silico analyses of public data sets. Finally, a clinical cohort comprising 355 surgically treated NSCLC cases was immunostained for LMO7. RESULTS In the yeast two-hybrid screen, the two paralogous proteins LMO7 and LIMCH1 were identified as interaction partners to LRIG3. LMO7 and LIMCH1 co-localized and co-immunoprecipitated with both LRIG1 and LRIG3. Endogenously expressed LMO7 was in close proximity of both LRIG1 and LRIG3. LMO7 and LIMCH1 were highly expressed in normal lung tissue and down-regulated in malignant lung tissue. LMO7 immunoreactivity was shown to be a negative prognostic factor in LRIG1 positive tumors, predicting poor patient survival. CONCLUSION These findings suggest that LMO7 and LIMCH1 physically interact with LRIG proteins and that expression of LMO7 is of clinical importance in NSCLC.
Collapse
Affiliation(s)
- Terese Karlsson
- Department of Radiation Sciences, Oncology, Umeå University, SE-901 87, Umeå, Sweden
| | - Samuel Kvarnbrink
- Department of Radiation Sciences, Oncology, Umeå University, SE-901 87, Umeå, Sweden.
| | - Camilla Holmlund
- Department of Radiation Sciences, Oncology, Umeå University, SE-901 87, Umeå, Sweden
| | - Johan Botling
- Department of Immunology, Genetics and Pathology, Molecular and Morphological Pathology, Uppsala University, SE-751 85, Uppsala, Sweden
| | - Patrick Micke
- Department of Immunology, Genetics and Pathology, Molecular and Morphological Pathology, Uppsala University, SE-751 85, Uppsala, Sweden
| | - Roger Henriksson
- Department of Radiation Sciences, Oncology, Umeå University, SE-901 87, Umeå, Sweden
| | - Mikael Johansson
- Department of Radiation Sciences, Oncology, Umeå University, SE-901 87, Umeå, Sweden
| | - Håkan Hedman
- Department of Radiation Sciences, Oncology, Umeå University, SE-901 87, Umeå, Sweden
| |
Collapse
|
8
|
Meléndez-Rodríguez F, Roche O, Sanchez-Prieto R, Aragones J. Hypoxia-Inducible Factor 2-Dependent Pathways Driving Von Hippel-Lindau-Deficient Renal Cancer. Front Oncol 2018; 8:214. [PMID: 29938199 PMCID: PMC6002531 DOI: 10.3389/fonc.2018.00214] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 05/24/2018] [Indexed: 12/20/2022] Open
Abstract
The most common type of the renal cancers detected in humans is clear cell renal cell carcinomas (ccRCCs). These tumors are usually initiated by biallelic gene inactivation of the Von Hippel-Lindau (VHL) factor in the renal epithelium, which deregulates the hypoxia-inducible factors (HIFs) HIF1α and HIF2α, and provokes their constitutive activation irrespective of the cellular oxygen availability. While HIF1α can act as a ccRCC tumor suppressor, HIF2α has emerged as the key HIF isoform that is essential for ccRCC tumor progression. Indeed, preclinical and clinical data have shown that pharmacological inhibitors of HIF2α can efficiently combat ccRCC growth. In this review, we discuss the molecular basis underlying the oncogenic potential of HIF2α in ccRCC by focusing on those pathways primarily controlled by HIF2α that are thought to influence the progression of these tumors.
Collapse
Affiliation(s)
- Florinda Meléndez-Rodríguez
- Research Unit, Hospital of Santa Cristina, Research Institute Princesa (IP), Autonomous University of Madrid, Madrid, Spain
- CIBER de Enfermedades Cardiovasculares, Carlos III Health Institute, Madrid, Spain
| | - Olga Roche
- Unidad Asociada de Biomedicina, Universidad de Castilla-La Mancha, Consejo Superior de Investigaciones Científicas (CSIC), Albacete, Spain
- Departamento de Ciencias Médicas, Facultad de Medicina, Universidad Castilla-La Mancha, Albacete, Spain
- Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas, Universidad de Castilla-La Mancha, Albacete, Spain
| | - Ricardo Sanchez-Prieto
- Unidad Asociada de Biomedicina, Universidad de Castilla-La Mancha, Consejo Superior de Investigaciones Científicas (CSIC), Albacete, Spain
- Departamento de Ciencias Médicas, Facultad de Medicina, Universidad Castilla-La Mancha, Albacete, Spain
- Departamento de Biología del Cáncer, Instituto de investigaciones Biomedicas Alberto Sols, Universidad Autónoma de Madrid, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Julian Aragones
- Research Unit, Hospital of Santa Cristina, Research Institute Princesa (IP), Autonomous University of Madrid, Madrid, Spain
- CIBER de Enfermedades Cardiovasculares, Carlos III Health Institute, Madrid, Spain
| |
Collapse
|
9
|
Liu F, Jiao Y, Jiao Y, Garcia-Godoy F, Gu W, Liu Q. Sex difference in EGFR pathways in mouse kidney-potential impact on the immune system. BMC Genet 2016; 17:146. [PMID: 27881077 PMCID: PMC5122204 DOI: 10.1186/s12863-016-0449-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 10/15/2016] [Indexed: 12/23/2022] Open
Abstract
Background Epidermal growth factor receptor (Egfr) has been the target of several drugs for cancers. The potential gender differences in genes in the Egfr axis have been suggested in humans and in animal models. Female and male mice from the same recombinant inbred (RI) strain have the same genomic components except the sex difference. A population of different RI mouse strains allows to conduct precise analysis of molecular pathways and regulation of Egfr between female and male mice. Methods The whole genome expression profiles of 70 genetically diverse RI strains of mice were used to compare three major molecular aspects of Egfr gene: the relative expression levels, gene network and expression quantitative trait loci (eQTL) that regulate the expression of Egfr between female and male mice. Results Our data showed that there is a significant sex difference in the expression levels in kidney. A considerable number of genes in the gene network of Egfr are sex differentially expressed. The expression levels of Egfr in mice are statistical significant different between C57BL/6 J (B6) and DBA/2 J (D2) genotypes in male while no difference in female mice. The eQTLs that regulate the expression levels of Egfr between female and male mice are also different. Furthermore, the differential expression levels of Egfr showed significantly different correlations with two known biological traits between male and female mice. Conclusion Overall there is a substantial sex difference in the Egfr pathways in mice. These data may have significant impact on drug target design, development, formulation, and dosage determinant for women and men in clinical trials. Electronic supplementary material The online version of this article (doi:10.1186/s12863-016-0449-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Fengxia Liu
- The Fourth Hospital, Hebei Medical University, Shijiazhuang, Hebei, 050011, China.,Department of Orthopaedic Surgery and BME-Campbell Clinic, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Yan Jiao
- Department of Orthopaedic Surgery and BME-Campbell Clinic, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Yun Jiao
- Department of Neuroscience, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Franklin Garcia-Godoy
- Bioscience Research Center, College of Dentistry, University of Tennessee Health Science Center, 875 Union Avenue, Memphis, TN, USA
| | - Weikuan Gu
- Department of Orthopaedic Surgery and BME-Campbell Clinic, University of Tennessee Health Science Center, Memphis, TN, USA. .,Research Service, Veterans Affairs Medical Center, 1030 Jefferson Avenue, Memphis, TN, USA. .,, 956 Court Ave, Memphis, TN, 38163, USA.
| | - Qingyi Liu
- The Fourth Hospital, Hebei Medical University, Shijiazhuang, Hebei, 050011, China.
| |
Collapse
|
10
|
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
|
11
|
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: 0.9] [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
|
12
|
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: 0.3] [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
|
13
|
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
|
14
|
Gudas LJ, Fu L, Minton DR, Mongan NP, Nanus DM. The role of HIF1α in renal cell carcinoma tumorigenesis. J Mol Med (Berl) 2014; 92:825-36. [PMID: 24916472 DOI: 10.1007/s00109-014-1180-z] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Revised: 05/22/2014] [Accepted: 05/28/2014] [Indexed: 01/26/2023]
Abstract
UNLABELLED The transcription factor HIF1α is implicated in the development of clear cell renal cell carcinoma (ccRCC). Although HIF1α was initially believed to be essential for ccRCC development, recent studies hypothesize an oncogenic role for HIF2α in ccRCC, but a tumor suppressor role for HIF1α, leading to uncertainty as to the precise roles of the different HIF transcription factors in this disease. Using evidence available from studies with human ccRCC cell lines, mouse xenografts, murine models of ccRCC, and human ccRCC specimens, we evaluate the roles of HIF1α and HIF2α in the pathogenesis of ccRCC. We present a convergence of clinical and mechanistic data supporting an important role for HIF1α in promoting tumorigenesis in a clinically important and large subset of ccRCC. This indicates that current understanding of the exact roles of HIF1α and HIF2α is incomplete and that further research is required to determine the diverse roles of HIF1α and HIF2α in ccRCC. KEY MESSAGES The TRACK mouse ccRCC model with constitutively active HIF1α but not HIF2α expressed in proximal tubules develops RCC. HIF1α protein is expressed in the majority of human ccRCC specimens. Elevated HIF1α in ccRCC correlates with a worse prognosis. Many publications do not support a tumor suppressor role for HIF1α in ccRCC. HIF1α, but not HIF2α, is expressed in some types of cancer stem cells.
Collapse
Affiliation(s)
- Lorraine J Gudas
- Department of Pharmacology, Weill Cornell Medical College (WCMC) of Cornell University, New York, NY, 10065, USA,
| | | | | | | | | |
Collapse
|
15
|
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
|
16
|
Staruschenko A, Palygin O, Ilatovskaya DV, Pavlov TS. Epidermal growth factors in the kidney and relationship to hypertension. Am J Physiol Renal Physiol 2013; 305:F12-20. [PMID: 23637204 DOI: 10.1152/ajprenal.00112.2013] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Members of the epidermal growth factor (EGF)-family bind to ErbB (EGFR)-family receptors that play an important role in the regulation of various fundamental cell processes in many organs including the kidney. In this field, most of the research efforts are focused on the role of EGF-ErbB axis in cancer biology. However, many studies indicate that abnormal ErbB-mediated signaling pathways are critical in the development of renal and cardiovascular pathologies. The kidney is a major site of the EGF-family ligands synthesis, and it has been shown to express all four members of the ErbB receptor family. The study of kidney disease regulation by ErbB receptor ligands has expanded considerably in recent years. In vitro and in vivo studies have provided direct evidence of the role of ErbB signaling in the kidney. Recent advances in the understanding of how the proteins in the EGF-family regulate sodium transport and development of hypertension are specifically discussed here. Collectively, these results suggest that EGF-ErbB signaling pathways could be major determinants in the progress of renal lesions, including its effects on the regulation of sodium reabsorption in collecting ducts.
Collapse
Affiliation(s)
- Alexander Staruschenko
- Department of Physiology, Medical College of Wisconsin, 8701 Watertown Plank Rd., Milwaukee, WI 53226, USA.
| | | | | | | |
Collapse
|
17
|
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: 67] [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
|