1
|
Qu L, Wang F, Wang Y, Li Z. AJUBA promotes the proliferation, invasion and migration of NSCLC cells by activating the ERK/β-catenin pathway. Sci Rep 2025; 15:13123. [PMID: 40240814 PMCID: PMC12003803 DOI: 10.1038/s41598-025-98156-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 04/09/2025] [Indexed: 04/18/2025] Open
Abstract
Accumulating evidence indicates that AJUBA acts as a potential target for new therapeutics to treat cancers. Nevertheless, the role of AJUBA in non-small cell lung cancer (NSCLC) remains unclear. In the current study, immunohistochemistry (IHC) showed that expression of AJUBA was upregulated in 67.55% of NSCLC tumor samples and was associated with tumor size, lymph node metastasis, advanced tumor stage, poor differentiation and poor prognosis. Loss-of-function assays of AJUBA produced by silencing RNA (siAJUBA) significantly inhibited the proliferation, invasion and migration of H1299 and A549 cell lines. Mechanistically, inhibition of extracellular signal-regulated kinases (ERKs) blocked the AJUBA-induced proliferation, invasion and migration of NSCLC cells, and decreased the expression of proteins related to the endothelial-mesenchymal transition (EMT). Silencing of AJUBA repressed tumor growth and led to a decrease in p-ERK, β-catenin and N-cadherin in vivo. In conclusion,, overexpression of AJUBA facilitates the proliferation and motility of NSCLC cells via the ERK and Wnt/β-catenin pathways. AJUBA may be useful as a prognostic marker which may provide a promising approach for the treatment of NSCLC.
Collapse
Affiliation(s)
- Lianyue Qu
- Key Laboratory of Diagnostic Imaging and Interventional Radiology of Liaoning Province, Department of Radiology, The First Hospital of China Medical University, Shenyang, P. R. China
- Department of Pharmacy, The First Hospital of China Medical University, Shenyang, P. R. China
| | - Fan Wang
- Key Laboratory of Diagnostic Imaging and Interventional Radiology of Liaoning Province, Department of Radiology, The First Hospital of China Medical University, Shenyang, P. R. China
- Department of Interventional Radiology, The First Hospital of China Medical University, Shenyang, P. R. China
| | - Yuxiang Wang
- Key Laboratory of Diagnostic Imaging and Interventional Radiology of Liaoning Province, Department of Radiology, The First Hospital of China Medical University, Shenyang, P. R. China
- Department of Nuclear Medicine, The First Hospital of China Medical University, Shenyang, P. R. China
| | - Zixuan Li
- Key Laboratory of Diagnostic Imaging and Interventional Radiology of Liaoning Province, Department of Radiology, The First Hospital of China Medical University, Shenyang, P. R. China.
- Department of Radiology, The First Hospital of China Medical University, Shenyang, P. R. China.
| |
Collapse
|
2
|
Cuttano R, Mazzarelli F, Afanga KM, Bianchi F, Dama E. MicroRNAs and the Mediterranean diet: a nutri-omics perspective for lung cancer. J Transl Med 2024; 22:632. [PMID: 38972997 PMCID: PMC11227714 DOI: 10.1186/s12967-024-05454-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 06/30/2024] [Indexed: 07/09/2024] Open
Abstract
Lung cancer is the deadliest cancer type worldwide with ~ 1.8 million deaths per-year. Smoking accounts for ~ 85% of all cases, with a described joint effect with unhealthy diet in lung cancer risk increase. Public health policies to prevent carcinogens exposure, promote smoking cessation and advocacy for healthy nutrition, are therefore highly recommended. Here we have examined the benefits of the Mediterranean Diet (MedDiet) in protecting against some non-communicable diseases including lung cancer, highlighting the epidemiological and biomolecular aspects of MedDiet anti-inflammatory effect and its interaction with smoking habits closely linked to risk of lung cancer. Considering the high incidence and mortality rates of lung cancer, we discussed also about the global impact that a Planeterranean extension of the benefits of MedDiet could have on controlling lung cancer risk. We also debated the impact of personalized nutrition on lung cancer prevention, considering individual heterogeneity in response to diet plans as well as recent advancements on nutri-omics in lung cancer research, with a specific focus on the role of microRNAs (miRNAs) as a promising nutritional molecular hub for lung cancer prevention. We strongly believe that a deep understanding of the molecular link between food components and genetic/epigenetics factors can expand effective intervention strategies.
Collapse
Affiliation(s)
- Roberto Cuttano
- Unit of Cancer Biomarkers, Fondazione IRCCS Casa Sollievo della Sofferenza, Viale Cappuccini Snc, 71013, San Giovanni Rotondo, Italy
| | - Francesco Mazzarelli
- Unit of Cancer Biomarkers, Fondazione IRCCS Casa Sollievo della Sofferenza, Viale Cappuccini Snc, 71013, San Giovanni Rotondo, Italy
| | - Kuku Miriam Afanga
- Unit of Cancer Biomarkers, Fondazione IRCCS Casa Sollievo della Sofferenza, Viale Cappuccini Snc, 71013, San Giovanni Rotondo, Italy
| | - Fabrizio Bianchi
- Unit of Cancer Biomarkers, Fondazione IRCCS Casa Sollievo della Sofferenza, Viale Cappuccini Snc, 71013, San Giovanni Rotondo, Italy.
| | - Elisa Dama
- Unit of Cancer Biomarkers, Fondazione IRCCS Casa Sollievo della Sofferenza, Viale Cappuccini Snc, 71013, San Giovanni Rotondo, Italy
| |
Collapse
|
3
|
Sun L, Wan J, Sun B, Tian Q, Li M, Xu LX, Feng CX, Tong X, Feng X, Yang X, Ding X. LncRNA-mir3471-limd1 regulatory network plays critical roles in HIBD. Exp Brain Res 2024; 242:443-449. [PMID: 38147087 PMCID: PMC10806112 DOI: 10.1007/s00221-023-06755-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 11/11/2023] [Indexed: 12/27/2023]
Abstract
The purpose of this study was to identify the target genes of tcon_00044595, elucidate its activation site, and provide novel insights into the pathogenesis and treatment of neonatal hypoxic-ischemic brain damage (HIBD). Through homologous blast analysis, we identified predicted target sequences in the neighboring regions of the long non-coding RNA (lncRNA) tcon_00044595, suggesting that limd1 is its target gene. Starbase was utilized to identify potential candidate microRNAs associated with the lncRNA. The interaction between the candidate microRNAs and limd1 was investigated and validated using various experimental methods including in vitro cell culture, cell transfection, dual fluorescence reporter detection system, and real-time PCR. Homology alignment analysis revealed that the lncRNA tcon_00044595 exhibited a 246 bp homologous sequence at the 3' end of the adjacent limd1 gene, with a conservation rate of 68%. Analysis conducted on Starbase online identified three potential microRNA candidates: miR-3471, miR-883a-5p, and miR-214-3p. Intracellular expression of the limd1 gene was significantly down-regulated upon transfection with miR-3471, while the other two microRNAs did not produce noticeable effects. Luciferase reporter assays identified two interaction sites (UTR-1, UTR-2) between miR-3471 and the limd1 3'UTR, with UTR-1 exhibiting a strong influence. Further CCK8 assay indicated a protective role of miR-3471 during low oxygen stroke in HIBD. The potential regulatory relationship between lncRNA (tcon_00044595), miR-3471, and the target gene limd1 suggests their involvement in the occurrence and development of HIBD, providing new insights for investigating the underlying mechanisms and exploring targeted therapeutic approaches for HIBD.
Collapse
Affiliation(s)
- Li Sun
- Soochow Key Laboratory of Prevention and Treatment of Child Brain Injury;, Children's Hospital of Soochow University, Suzhou, 215025, China
| | - Jun Wan
- Soochow Key Laboratory of Prevention and Treatment of Child Brain Injury;, Children's Hospital of Soochow University, Suzhou, 215025, China
- Department of Pediatrics, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Bin Sun
- Department of Neonatology, Children's Hospital of Soochow University, No.92 Zhongnanjie Road, Suzhou, 215025, Jiangsu, China
| | - Qiuyan Tian
- Soochow Key Laboratory of Prevention and Treatment of Child Brain Injury;, Children's Hospital of Soochow University, Suzhou, 215025, China
| | - Mei Li
- Pediatrics Research Institute, Children's Hospital of Soochow University, Suzhou, 215025, China
| | - Li-Xiao Xu
- Pediatrics Research Institute, Children's Hospital of Soochow University, Suzhou, 215025, China
| | - Chen-Xi Feng
- Pediatrics Research Institute, Children's Hospital of Soochow University, Suzhou, 215025, China
| | - Xiao Tong
- Department of Pediatrics, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Xing Feng
- Soochow Key Laboratory of Prevention and Treatment of Child Brain Injury;, Children's Hospital of Soochow University, Suzhou, 215025, China
| | - Xiaofeng Yang
- Soochow Key Laboratory of Prevention and Treatment of Child Brain Injury;, Children's Hospital of Soochow University, Suzhou, 215025, China.
- Department of Neonatology, Children's Hospital of Soochow University, No.92 Zhongnanjie Road, Suzhou, 215025, Jiangsu, China.
| | - Xin Ding
- Soochow Key Laboratory of Prevention and Treatment of Child Brain Injury;, Children's Hospital of Soochow University, Suzhou, 215025, China.
- Department of Neonatology, Children's Hospital of Soochow University, No.92 Zhongnanjie Road, Suzhou, 215025, Jiangsu, China.
| |
Collapse
|
4
|
Tan R, You Q, Yu D, Xiao C, Adu-Amankwaah J, Cui J, Zhang T. Novel hub genes associated with pulmonary artery remodeling in pulmonary hypertension. Front Cardiovasc Med 2022; 9:945854. [PMID: 36531719 PMCID: PMC9748075 DOI: 10.3389/fcvm.2022.945854] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 11/15/2022] [Indexed: 12/02/2022] Open
Abstract
Pulmonary hypertension (PH) is a life-threatening disease with complex pathogenesis. According to etiology, PH is divided into five major groups in clinical classification. However, pulmonary artery (PA) remodeling is their common feature, in addition to bone morphogenetic protein receptor type 2; it is elusive whether there are other novel common genes and similar underlying mechanisms. To identify novel common hub genes involved in PA remodeling at different PH groups, we analyzed mRNA-Seq data located in the general gene expression profile GSE130391 utilizing bioinformatics technology. This database contains PA samples from different PH groups of hospitalized patients with chronic thromboembolic pulmonary hypertension (CTEPH), idiopathic pulmonary artery hypertension (IPAH), and PA samples from organ donors without known pulmonary vascular diseases as control. We screened 22 hub genes that affect PA remodeling, most of which have not been reported in PH. We verified the top 10 common hub genes in hypoxia with Sugen-induced PAH rat models by qRT-PCR. The three upregulated candidate genes are WASF1, ARHGEF1 and RB1 and the seven downregulated candidate genes are IL1R1, RHOB, DAPK1, TNFAIP6, PKN1, PLOD2, and MYOF. WASF1, ARHGEF1, and RB1 were upregulated significantly in hypoxia with Sugen-induced PAH, while IL1R1, DAPK1, and TNFA1P6 were upregulated significantly in hypoxia with Sugen-induced PAH. The DEGs detected by mRNA-Seq in hospitalized patients with PH are different from those in animal models. This study will provide some novel target genes to further study PH mechanisms and treatment.
Collapse
Affiliation(s)
- Rubin Tan
- Department of Physiology, Basic Medical School, Xuzhou Medical University, Xuzhou, China
- *Correspondence: Rubin Tan
| | - Qiang You
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Dongdong Yu
- Department of Tumor Radiotherapy, Renmin Hospital of Wuhan University, Wuhan, China
- Department of Pathophysiology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Pulmonary Diseases of Ministry of Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chushu Xiao
- Department of Pathophysiology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Pulmonary Diseases of Ministry of Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Joseph Adu-Amankwaah
- Department of Physiology, Basic Medical School, Xuzhou Medical University, Xuzhou, China
| | - Jie Cui
- Department of Physiology, Basic Medical School, Xuzhou Medical University, Xuzhou, China
| | - Ting Zhang
- Department of Pathophysiology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Pulmonary Diseases of Ministry of Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
5
|
Elitist random swapped particle swarm optimization embedded with variable k-nearest neighbour classification: a new PSO variant applied to gene identification. Soft comput 2022. [DOI: 10.1007/s00500-022-07515-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/10/2022]
|
6
|
PLOD3 regulates the expression of YAP1 to affect the progression of non-small cell lung cancer via the PKCδ/CDK1/LIMD1 signaling pathway. J Transl Med 2022; 102:440-451. [PMID: 35039611 DOI: 10.1038/s41374-021-00674-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 08/19/2021] [Accepted: 08/30/2021] [Indexed: 12/25/2022] Open
Abstract
Procollagen-lysine, 2-oxoglutarate 5-dioxygenase (PLOD3) is a crucial oncogene in human lung cancer, whereas protein kinase C δ (PKCδ) acts as a tumor suppressor. In this study, we aimed to explore the regulation by PLOD3 on the expression of YAP1 to affect the progression of non-small cell lung cancer (NSCLC) via the PKCδ/CDK1/LIMD1 signaling pathway. We found that PLOD3, CDK1, and YAP1 were highly expressed, while LIMD1 was poorly expressed in NSCLC tissues. Mechanistic investigation demonstrated that silencing PLOD3 promoted the cleavage of PKCδ in a caspase-dependent manner to generate a catalytically active fragment cleaved PKCδ, enhanced phosphorylation levels of CDK1, and LIMD1 but suppressed nuclear translocation of YAP1. Furthermore, functional experimental results suggested that loss of PLOD3 led to increased phosphorylation levels of CDK1 and LIMD1 and downregulated YAP1, thereby suppressing the proliferation, colony formation, cell cycle entry, and resistance to apoptosis of NSCLC cells in vitro and inhibiting tumor growth in vivo. Taken together, these results show that PLOD3 silencing activates the PKCδ/CDK1/LIMD1 signaling pathway to prevent the progression of NSCLC, thus providing novel insight into molecular targets for treating NSCLC.
Collapse
|
7
|
Davidson K, Grevitt P, Contreras-Gerenas MF, Bridge KS, Hermida M, Shah KM, Mardakheh FK, Stubbs M, Burke R, Casado P, Cutillas PR, Martin SA, Sharp TV. Targeted therapy for LIMD1-deficient non-small cell lung cancer subtypes. Cell Death Dis 2021; 12:1075. [PMID: 34764236 PMCID: PMC8586256 DOI: 10.1038/s41419-021-04355-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 10/13/2021] [Accepted: 10/25/2021] [Indexed: 12/15/2022]
Abstract
An early event in lung oncogenesis is loss of the tumour suppressor gene LIMD1 (LIM domains containing 1); this encodes a scaffold protein, which suppresses tumorigenesis via a number of different mechanisms. Approximately 45% of non-small cell lung cancers (NSCLC) are deficient in LIMD1, yet this subtype of NSCLC has been overlooked in preclinical and clinical investigations. Defining therapeutic targets in these LIMD1 loss-of-function patients is difficult due to a lack of 'druggable' targets, thus alternative approaches are required. To this end, we performed the first drug repurposing screen to identify compounds that confer synthetic lethality with LIMD1 loss in NSCLC cells. PF-477736 was shown to selectively target LIMD1-deficient cells in vitro through inhibition of multiple kinases, inducing cell death via apoptosis. Furthermore, PF-477736 was effective in treating LIMD1-/- tumours in subcutaneous xenograft models, with no significant effect in LIMD1+/+ cells. We have identified a novel drug tool with significant preclinical characterisation that serves as an excellent candidate to explore and define LIMD1-deficient cancers as a new therapeutic subgroup of critical unmet need.
Collapse
Affiliation(s)
- Kathryn Davidson
- Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M6 BQ, UK
| | - Paul Grevitt
- Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M6 BQ, UK
| | - Maria F Contreras-Gerenas
- Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M6 BQ, UK
| | - Katherine S Bridge
- York Biomedical Research Institute, University of York, Wentworth Way, York, YO10 5DD, UK
| | - Miguel Hermida
- Department of Bioengineering, Imperial College, London, UK
| | - Kunal M Shah
- Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M6 BQ, UK
| | - Faraz K Mardakheh
- Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M6 BQ, UK
| | - Mark Stubbs
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, 15 Cotswold Road, Sutton, SM2 5NG, UK
| | - Rosemary Burke
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, 15 Cotswold Road, Sutton, SM2 5NG, UK
| | - Pedro Casado
- Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M6 BQ, UK
| | - Pedro R Cutillas
- Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M6 BQ, UK
| | - Sarah A Martin
- Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M6 BQ, UK.
| | - Tyson V Sharp
- Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M6 BQ, UK.
| |
Collapse
|
8
|
Human chromosome 3p21.3 carries TERT transcriptional regulators in pancreatic cancer. Sci Rep 2021; 11:15355. [PMID: 34321527 PMCID: PMC8319171 DOI: 10.1038/s41598-021-94711-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 07/15/2021] [Indexed: 11/23/2022] Open
Abstract
Frequent loss of heterozygosity (LOH) on the short arm of human chromosome 3 (3p) region has been found in pancreatic cancer (PC), which suggests the likely presence of tumor suppressor genes in this region. However, the functional significance of LOH in this region in the development of PC has not been clearly defined. The human telomerase reverse transcriptase gene (hTERT) contributes to unlimited proliferative and tumorigenicity of malignant tumors. We previously demonstrated that hTERT expression was suppressed by the introduction of human chromosome 3 in several cancer cell lines. To examine the functional role of putative TERT suppressor genes on chromosome 3 in PC, we introduced an intact human chromosome 3 into the human PK9 and murine LTPA PC cell lines using microcell-mediated chromosome transfer. PK9 microcell hybrids with an introduced human chromosome 3 showed significant morphological changes and rapid growth arrest. Intriguingly, microcell hybrid clones of LTPA cells with an introduced human chromosome 3 (LTPA#3) showed suppression of mTert transcription, cell proliferation, and invasion compared with LTPA#4 cells containing human chromosome 4 and parental LTPA cells. Additionally, the promoter activity of mTert was downregulated in LTPA#3. Furthermore, we confirmed that TERT regulatory gene(s) are present in the 3p21.3 region by transfer of truncated chromosomes at arbitrary regions. These results provide important information on the functional significance of the LOH at 3p for development and progression of PC.
Collapse
|
9
|
Hypomethylation of LIMD1 and P16 by downregulation of DNMT1 results in restriction of liver carcinogenesis by amarogentin treatment. J Biosci 2021. [DOI: 10.1007/s12038-021-00176-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
10
|
Basu M, Chatterjee A, Chakraborty B, Chatterjee E, Ghosh S, Samadder S, Pal DK, Roy A, Chakrabarti J, Ghosh A, Panda CK. High nuclear expression of HIF1α, synergizing with inactivation of LIMD1 and VHL, portray worst prognosis among the bladder cancer patients: association with arsenic prevalence. J Cancer Res Clin Oncol 2021; 147:2309-2322. [PMID: 34080067 DOI: 10.1007/s00432-021-03661-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 05/05/2021] [Indexed: 11/25/2022]
Abstract
PURPOSE Our study was aimed to understand the importance of LIMD1-VHL-HIF1α pathway in development of bladder carcinoma (BlCa) in association with arsenic prevalence. METHODS At first, the mRNA expression pattern of the genes of this pathway (LIMD1, VHL and HIF1α) was checked in GEO datasets and in our samples. Next, genetic and epigenetic profiling of LIMD1 and VHL was done in our sample pool, validated in T24 BlCa cell line. The results were next correlated with various clinico-pathological parameters. RESULTS Differential under-expression of LIMD1 and VHL genes was found in muscle-invasive BlCa (MIBC) in comparison to non-muscle-invasive BlCa (NMIBC). However, HIF1α protein, but mRNA, was found to be overexpressed among the MIBC samples; depicting the probability of HIF1α protein stabilization. Analysis of genetic and epigenetic profiles of LIMD1 and VHL exposed a frequent promoter methylation of LIMD1 gene in MIBC samples. Further, in-depth look into the results unveiled that the high nuclear expression of HIF1α was significantly correlated with genetic alterations of LIMD1, alone or in combination with VHL. Moreover, treating the T24 cells with a de-methylating agent (5-aza-2'-deoxycytidine) re-expressed the methylated LIMD1 and VHL genes, which in turn, reduced the HIF1α protein level significantly. Additionally, patients with high arsenic content (> 112 ng/g, AsH) seemed to have recurrent promoter methylation in LIMD1, as well as co-methylation/alteration of LIMD1 and VHL gene. Lastly, high nuclear expression of HIF1α in association with co-alteration of VHL and LIMD1 showed the worst overall survival (OS) among the patients. CONCLUSION To conclude, MIBC samples portrayed higher alterations in VHL and LIMD1, thereby, stabilizing HIF1α protein and lowering the OS of patients.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Arsenic Poisoning/diagnosis
- Arsenic Poisoning/epidemiology
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Carcinoma, Transitional Cell/diagnosis
- Carcinoma, Transitional Cell/epidemiology
- Carcinoma, Transitional Cell/genetics
- Carcinoma, Transitional Cell/pathology
- Cell Line, Tumor
- Cell Nucleus/genetics
- Cell Nucleus/metabolism
- Cell Nucleus/pathology
- Comorbidity
- DNA Methylation
- Datasets as Topic
- Female
- Gene Expression Regulation, Neoplastic
- Gene Silencing
- Humans
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Intracellular Signaling Peptides and Proteins/genetics
- Intracellular Signaling Peptides and Proteins/metabolism
- LIM Domain Proteins/genetics
- LIM Domain Proteins/metabolism
- Male
- Middle Aged
- Neoplasm Invasiveness
- Prevalence
- Prognosis
- Survival Analysis
- Urinary Bladder Neoplasms/diagnosis
- Urinary Bladder Neoplasms/epidemiology
- Urinary Bladder Neoplasms/genetics
- Urinary Bladder Neoplasms/pathology
- Von Hippel-Lindau Tumor Suppressor Protein/genetics
- Von Hippel-Lindau Tumor Suppressor Protein/metabolism
Collapse
Affiliation(s)
- Mukta Basu
- Department of Oncogene Regulation, Chittaranjan National Cancer Institute, 37 S. P. Mukherjee Road, Kolkata, West Bengal, 700026, India
| | - Amvrin Chatterjee
- Department of Urology, IPGMER, SSKM, 244 A.J.C. Bose Road, Kolkata, 700020, India
| | - Balarko Chakraborty
- Department of Oncogene Regulation, Chittaranjan National Cancer Institute, 37 S. P. Mukherjee Road, Kolkata, West Bengal, 700026, India
| | - Essha Chatterjee
- National Institute of Pharmaceutical Education and Research, Kukatpally Industrial Estate, Balanagar, Hyderabad, Telangana, 500037, India
| | - Sabnam Ghosh
- Department of Life Science, Presidency University, 86/1, College Street, Kolkata, 700073, India
| | - Sudip Samadder
- Department of Oncogene Regulation, Chittaranjan National Cancer Institute, 37 S. P. Mukherjee Road, Kolkata, West Bengal, 700026, India
| | - Dilip Kumar Pal
- Department of Urology, IPGMER, SSKM, 244 A.J.C. Bose Road, Kolkata, 700020, India
| | - Anup Roy
- Nil Ratan Sarkar Medical College and Hospital, Kolkata, West Bengal, India
| | - Jayanta Chakrabarti
- Department of Oncogene Regulation, Chittaranjan National Cancer Institute, 37 S. P. Mukherjee Road, Kolkata, West Bengal, 700026, India
| | - Amlan Ghosh
- Department of Life Science, Presidency University, 86/1, College Street, Kolkata, 700073, India
| | - Chinmay Kumar Panda
- Department of Oncogene Regulation, Chittaranjan National Cancer Institute, 37 S. P. Mukherjee Road, Kolkata, West Bengal, 700026, India.
| |
Collapse
|
11
|
Wang Y, Zhang C, Yang W, Shao S, Xu X, Sun Y, Li P, Liang L, Wu C. LIMD1 phase separation contributes to cellular mechanics and durotaxis by regulating focal adhesion dynamics in response to force. Dev Cell 2021; 56:1313-1325.e7. [PMID: 33891898 DOI: 10.1016/j.devcel.2021.04.002] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 12/25/2020] [Accepted: 03/30/2021] [Indexed: 01/09/2023]
Abstract
Cells sense and respond to extracellular mechanical cues through cell-matrix adhesions. Interestingly, the maturation of focal adhesions (FAs) is reciprocally force dependent. How biomechanical cues dictate the status of cell motility and how FAs spatial temporally coordinate force sensing and self-organization remain enigmatic. Here, we identify that LIMD1, a member of the LIM domain scaffolding proteins, undergoes force-sensitive condensation at the FAs. We also unveil that the multivalent interactions of LIMD1 intrinsically disordered region (IDR) and the LIM domains concertedly drive this phase transition under the regulation of phosphorylation. Intriguingly, formation of condensed LIMD1 protein compartments is sufficient to specifically enrich and localize late FA proteins. We further discover that LIMD1 regulates cell spreading, maintains FA dynamics and cellular contractility, and is critical for durotaxis-the ability of cells to crawl along gradients of substrate stiffness. Our results suggest a model that recruitment of LIMD1 to the FAs, via mechanical force triggered inter-molecular interaction, serves as a phase separation hub to assemble and organize matured FAs, thus allowing for efficient mechano-transduction and cell migration.
Collapse
Affiliation(s)
- Yuan Wang
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing Key Laboratory of Tumor Systems Biology, Beijing 100191, China
| | - Chunlei Zhang
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing Key Laboratory of Tumor Systems Biology, Beijing 100191, China
| | - Wenzhong Yang
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing Key Laboratory of Tumor Systems Biology, Beijing 100191, China
| | - ShiPeng Shao
- State Key Laboratory of Membrane Biology, Biodynamic Optical Imaging Center (BIOPIC), School of Life Sciences, Peking University, Beijing 100871, China; Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China
| | - Xinmin Xu
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing Key Laboratory of Tumor Systems Biology, Beijing 100191, China
| | - Yujie Sun
- State Key Laboratory of Membrane Biology, Biodynamic Optical Imaging Center (BIOPIC), School of Life Sciences, Peking University, Beijing 100871, China
| | - Pilong Li
- Beijing Advanced Innovation Center for Structural Biology, Beijing Frontier Research Center for Biological Structure, Tsinghua University, Peking University Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Ling Liang
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China.
| | - Congying Wu
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing Key Laboratory of Tumor Systems Biology, Beijing 100191, China.
| |
Collapse
|
12
|
Wang L, Sparks-Wallace A, Casteel JL, Howell MEA, Ning S. Algorithm-Based Meta-Analysis Reveals the Mechanistic Interaction of the Tumor Suppressor LIMD1 With Non-Small-Cell Lung Carcinoma. Front Oncol 2021; 11:632638. [PMID: 33869018 PMCID: PMC8044451 DOI: 10.3389/fonc.2021.632638] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 03/15/2021] [Indexed: 12/25/2022] Open
Abstract
Non-small-cell lung carcinoma (NSCLC) is the major type of lung cancer, which is among the leading causes of cancer-related deaths worldwide. LIMD1 was previously identified as a tumor suppressor in lung cancer, but their detailed interaction in this setting remains unclear. In this study, we have carried out multiple genome-wide bioinformatic analyses for a comprehensive understanding of LIMD1 in NSCLC, using various online algorithm platforms that have been built for mega databases derived from both clinical and cell line samples. Our results indicate that LIMD1 expression level is significantly downregulated at both mRNA and protein levels in both lung adenocarcinoma (LUAD) and lung squamous cell carcinoma (LUSC), with a considerable contribution from its promoter methylation rather than its gene mutations. The Limd1 gene undergoes mutation only at a low rate in NSCLC (0.712%). We have further identified LIMD1-associated molecular signatures in NSCLC, including its natural antisense long non-coding RNA LIMD1-AS1 and a pool of membrane trafficking regulators. We have also identified a subgroup of tumor-infiltrating lymphocytes, especially neutrophils, whose tumor infiltration levels significantly correlate with LIMD1 level in both LUAD and LUSC. However, a significant correlation of LIMD1 with a subset of immune regulatory molecules, such as IL6R and TAP1, was only found in LUAD. Regarding the clinical outcomes, LIMD1 expression level only significantly correlates with the survival of LUAD (p<0.01) but not with that of LUSC (p>0.1) patients. These findings indicate that LIMD1 plays a survival role in LUAD patients at least by acting as an immune regulatory protein. To further understand the mechanisms underlying the tumor-suppressing function of LIMD1 in NSCLC, we show that LIMD1 downregulation remarkably correlates with the deregulation of multiple pathways that play decisive roles in the oncogenesis of NSCLC, especially those mediated by EGFR, KRAS, PIK3CA, Keap1, and p63, in both LUAD and LUSC, and those mediated by p53 and CDKN2A only in LUAD. This study has disclosed that LIMD1 can serve as a survival prognostic marker for LUAD patients and provides mechanistic insights into the interaction of LIMD1 with NSCLC, which provide valuable information for clinical applications.
Collapse
Affiliation(s)
- Ling Wang
- Department of Internal Medicine, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States.,Center of Excellence for Inflammation, Infectious Diseases and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Ayrianna Sparks-Wallace
- Department of Internal Medicine, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Jared L Casteel
- Department of Internal Medicine, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Mary E A Howell
- Department of Internal Medicine, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Shunbin Ning
- Department of Internal Medicine, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States.,Center of Excellence for Inflammation, Infectious Diseases and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| |
Collapse
|
13
|
Guo ZZ, Ma ZJ, He YZ, Jiang W, Xia Y, Pan CF, Wei K, Shi YJ, Chen L, Chen YJ. miR-550a-5p Functions as a Tumor Promoter by Targeting LIMD1 in Lung Adenocarcinoma. Front Oncol 2020; 10:570733. [PMID: 33194664 PMCID: PMC7655921 DOI: 10.3389/fonc.2020.570733] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 09/14/2020] [Indexed: 12/21/2022] Open
Abstract
Lung adenocarcinoma accounts for half of all lung cancer cases in most countries. Mounting evidence has demonstrated that microRNAs play important roles in cancer progression, and some of them can be identified as potential biomarkers. This study aimed to explore the role of miR-550a-5p, a lung adenocarcinoma-associated mature microRNA screened out from the TCGA database via R-studio and Perl, with abundant expression in samples and with 5-year survival prognosis difference, as well as having not been studied in lung cancer yet. Potential target genes were predicted by the online database. Gene ontology enrichment, pathway enrichment, protein–protein interaction network, and hub genes–microRNA network were constructed by FunRich, STRING database, and Cytoscape. Then, LIMD1, a known tumor suppressor gene reported by multiple articles, was found to have a negative correlation with miR-550a-5p. The expression of miR-550a-5p was up-regulated in tumor samples and tumor-associated cell lines. Its high expression was also correlated with tumor size. Cell line A549 treated with miR-550a-5p overexpression promoted tumor proliferation, while H1299 treated with miR-550a-5p knockdown showed the opposite result. Mechanically, miR-550a-5p negatively regulated LIMD1 by directly binding to its 3′-UTR validated by dual luciferase assay. In summary, a new potential prognostic and therapeutic biomarker, miR-550a-5p, has been identified by bioinformatics analysis and experimental validation in vitro and in vivo, which promotes lung adenocarcinoma by silencing a known suppressor oncogene LIMD1.
Collapse
Affiliation(s)
- Zi-Zhang Guo
- Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zi-Jian Ma
- Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yao-Zhou He
- Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wei Jiang
- Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yang Xia
- Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Chun-Feng Pan
- Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ke Wei
- Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yi-Jun Shi
- Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Liang Chen
- Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yi-Jiang Chen
- Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
14
|
Landry NM, Dixon IMC. Fibroblast mechanosensing, SKI and Hippo signaling and the cardiac fibroblast phenotype: Looking beyond TGF-β. Cell Signal 2020; 76:109802. [PMID: 33017619 DOI: 10.1016/j.cellsig.2020.109802] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 09/29/2020] [Accepted: 09/30/2020] [Indexed: 12/19/2022]
Abstract
Cardiac fibroblast activation to hyper-synthetic myofibroblasts following a pathological stimulus or in response to a substrate with increased stiffness may be a key tipping point for the evolution of cardiac fibrosis. Cardiac fibrosis per se is associated with progressive loss of heart pump function and is a primary contributor to heart failure. While TGF-β is a common cytokine stimulus associated with fibroblast activation, a druggable target to quell this driver of fibrosis has remained an elusive therapeutic goal due to its ubiquitous use by different cell types and also in the signaling complexity associated with SMADs and other effector pathways. More recently, mechanical stimulus of fibroblastic cells has been revealed as a major point of activation; this includes cardiac fibroblasts. Further, the complexity of TGF-β signaling has been offset by the discovery of members of the SKI family of proteins and their inherent anti-fibrotic properties. In this respect, SKI is a protein that may bind a number of TGF-β associated proteins including SMADs, as well as signaling proteins from other pathways, including Hippo. As SKI is also known to directly deactivate cardiac myofibroblasts to fibroblasts, this mode of action is a putative candidate for further study into the amelioration of cardiac fibrosis. Herein we provide a synthesis of this topic and highlight novel candidate pathways to explore in the treatment of cardiac fibrosis.
Collapse
Affiliation(s)
- Natalie M Landry
- Department of Physiology and Pathophysiology, Institute of Cardiovascular Sciences, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Canada
| | - Ian M C Dixon
- Department of Physiology and Pathophysiology, Institute of Cardiovascular Sciences, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Canada.
| |
Collapse
|
15
|
Zeng X, Wang H, He D, Jia W, Ma R. LIMD1 Increases the Sensitivity of Lung Adenocarcinoma Cells to Cisplatin via the GADD45α/p38 MAPK Signaling Pathway. Front Oncol 2020; 10:969. [PMID: 32754438 PMCID: PMC7365921 DOI: 10.3389/fonc.2020.00969] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 05/18/2020] [Indexed: 11/30/2022] Open
Abstract
Objective: To investigate the effect of LIM domain-containing protein 1 (LIMD1) on the sensitivity of lung adenocarcinoma cells to cisplatin and explore the mechanism. Methods: A549 and H1299 cells were transfected with lentivirus to establish LIMD1-overexpressing cell lines and their respective controls. The protein expression of DNA damage-inducible 45 alpha (GADD45α) and p38 mitogen-activated protein kinase (MAPK) was detected by Western blot. The survival of A549-vec, A549-LIMD1, H1299-vec, and H1299-LIMD1 cells after cisplatin treatment was observed by CCK-8, and the viability was calculated accordingly. Then, SB203580 was used to inhibit the activity of the p38 MAPK signaling pathway, after which the survival of A549-vec, A549-LIMD1, H1299-vec, and H1299-LIMD1 cells in response to cisplatin was observed again by CCK-8, and the viability was calculated accordingly. Results: When LIMD1 was overexpressed in A549 and H1299 cells, the levels of GADD45α and p-p38 MAPK were increased, but total p38 MAPK expression showed no significant change. After adding 30 μM cisplatin, the optical density (OD) values of A549-LIMD1 and H1299-LIMD1 cells were significantly lower than those of their respective controls at 24, 48, and 72 h. The viability of A549-LIMD1 and H1299-LIMD1 cells was significantly lower than that of their respective controls at all the times tested (p < 0.05). The Western blot results showed that the expression of apoptotic proteins cleaved caspase 3 and cleaved PARP in cisplatin-treated A549-LIDM1 and H1299-LIMD1 cells was significantly higher than that in their respective control cells. Flow cytometry showed that the apoptosis rates of A549-LIMD1 and H1299-LIMD1 cells were significantly higher than those of their respective controls (p < 0.05). SB203580 significantly inhibited the activation of the p38 MAPK signaling pathway in lung adenocarcinoma cells; however, neither the OD values nor the viability of A549-LIMD1 cells and H1299-LIMD1 cells showed no significant difference from those of their controls at 24, 48, and 72 h after cisplatin and SB203580 treatment (p > 0.05 for both). Western blot analysis showed that after SB203580 was added, the expression of cleaved caspase 3 and cleaved PARP in A549-LIMD1 and H1299-LIMD1 cells presented no significant difference compared with that in their respective controls. Conclusion: LIMD1 increases the sensitivity of lung adenocarcinoma cells to cisplatin by activating the GADD45α/p38 MAPK signaling pathway.
Collapse
Affiliation(s)
- Xiaofei Zeng
- Department of Cardiothoracic Surgery, First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Hong Wang
- Department of Cardiothoracic Surgery, First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Dongsheng He
- Department of Cardiothoracic Surgery, First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Weikun Jia
- Department of Cardiothoracic Surgery, First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Ruidong Ma
- Department of Cardiothoracic Surgery, First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| |
Collapse
|
16
|
Pan J, Tang Y, Liu S, Li L, Yu B, Lu Y, Wang Y. LIMD1-AS1 suppressed non-small cell lung cancer progression through stabilizing LIMD1 mRNA via hnRNP U. Cancer Med 2020; 9:3829-3839. [PMID: 32239804 PMCID: PMC7286462 DOI: 10.1002/cam4.2898] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 01/20/2020] [Accepted: 01/21/2020] [Indexed: 12/30/2022] Open
Abstract
Background Non‐small cell lung cancer (NSCLC) occupies the majority of lung cancer cases and is notorious for the awful prognosis. LIM domains‐containing 1 (LIMD1) is suggested as a tumor suppressor in lung cancer, but its mechanism in NSCLC remains elusive. Present study aimed to uncover the mechanism of LIMD1 in NSCLC. Methods qRT‐PCR was performed to analyze the level of LIMD1. The functions of LIMD1 in NSCLC cells were evaluated by CCK‐8, EdU, and caspase‐3 activity assays. RIP and pull‐down assays were applied to determine the interaction of LIMD1 with heterogeneous nuclear ribonucleoprotein U (hnRNP U) and LIMD1‐AS1. Results LIMD1 was downregulated in NSCLC samples and cells. Functionally, LIMD1 hindered proliferation and drove apoptosis in NSCLC cells. Moreover, long noncoding RNA (lncRNA) LIMD1 antisense RNA 1 (LIMD1‐AS1) was downregulated in NSCLC samples and cell lines. LIMD1‐AS1 knockdown abrogated NSCLC cell growth in vitro and in vivo. Mechanistically, LIMD1‐AS1 stabilized LIMD1 mRNA through interacting with hnRNP U. Rescue experiments suggested that LIMD1‐AS1 repressed NSCLC progression through LIMD1. Conclusions LIMD1‐AS1 suppressed NSCLC progression through stabilizing LIMD1 mRNA via hnRNP U, providing new thoughts for the improvement of molecular‐targeted therapy for NSCLC.
Collapse
Affiliation(s)
- Jianyuan Pan
- Cardiovascular Department, The Second People's Hospital of Hefei, Hefei, China
| | - Yongqin Tang
- General Surgery, The First People's Hospital of Chuzhou, Chuzhou, China
| | - Shumei Liu
- Respiratory Medicine Department, Liaocheng People's Hospital, Liaocheng, China
| | - Lily Li
- Respiratory Medicine Department, Liaocheng People's Hospital, Liaocheng, China
| | - Bo Yu
- Respiratory Medicine Department, Liaocheng People's Hospital, Liaocheng, China
| | - Yuanyuan Lu
- Respiratory Medicine Department, Liaocheng People's Hospital, Liaocheng, China
| | - Yu Wang
- Respiratory Medicine Department, Liaocheng People's Hospital, Liaocheng, China
| |
Collapse
|
17
|
Malyla V, Paudel KR, Shukla SD, Donovan C, Wadhwa R, Pickles S, Chimankar V, Sahu P, Bielefeldt-Ohmann H, Bebawy M, Hansbro PM, Dua K. Recent advances in experimental animal models of lung cancer. Future Med Chem 2020; 12:567-570. [PMID: 32175774 DOI: 10.4155/fmc-2019-0338] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 01/16/2020] [Indexed: 02/02/2023] Open
Affiliation(s)
- Vamshikrishna Malyla
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Chippendale, NSW 2007, Australia
- Centre for Inflammation, Centenary Institute, Sydney, NSW 2050, Australia
| | - Keshav Raj Paudel
- Centre for Inflammation, Centenary Institute, Sydney, NSW 2050, Australia
- School of Life Sciences, University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Shakti D Shukla
- Priority Research Centre for Healthy Lungs & Hunter Medical Research Institute, The University of Newcastle, Newcastle, NSW 2305, Australia
| | - Chantal Donovan
- Centre for Inflammation, Centenary Institute, Sydney, NSW 2050, Australia
- School of Life Sciences, University of Technology Sydney, Sydney, NSW 2007, Australia
- Priority Research Centre for Healthy Lungs & Hunter Medical Research Institute, The University of Newcastle, Newcastle, NSW 2305, Australia
| | - Ridhima Wadhwa
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Chippendale, NSW 2007, Australia
- Centre for Inflammation, Centenary Institute, Sydney, NSW 2050, Australia
| | - Sophie Pickles
- Centre for Inflammation, Centenary Institute, Sydney, NSW 2050, Australia
- Priority Research Centre for Healthy Lungs & Hunter Medical Research Institute, The University of Newcastle, Newcastle, NSW 2305, Australia
| | - Vrushali Chimankar
- Centre for Inflammation, Centenary Institute, Sydney, NSW 2050, Australia
- Priority Research Centre for Healthy Lungs & Hunter Medical Research Institute, The University of Newcastle, Newcastle, NSW 2305, Australia
| | - Priyanka Sahu
- Centre for Inflammation, Centenary Institute, Sydney, NSW 2050, Australia
- Priority Research Centre for Healthy Lungs & Hunter Medical Research Institute, The University of Newcastle, Newcastle, NSW 2305, Australia
| | - Helle Bielefeldt-Ohmann
- Australian Infectious Diseases Research Centre, School of Chemistry & Molecular Biosciences, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Mary Bebawy
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Chippendale, NSW 2007, Australia
| | - Philip M Hansbro
- Centre for Inflammation, Centenary Institute, Sydney, NSW 2050, Australia
- School of Life Sciences, University of Technology Sydney, Sydney, NSW 2007, Australia
- Priority Research Centre for Healthy Lungs & Hunter Medical Research Institute, The University of Newcastle, Newcastle, NSW 2305, Australia
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Chippendale, NSW 2007, Australia
- Centre for Inflammation, Centenary Institute, Sydney, NSW 2050, Australia
- Priority Research Centre for Healthy Lungs & Hunter Medical Research Institute, The University of Newcastle, Newcastle, NSW 2305, Australia
| |
Collapse
|
18
|
Zhou J, Zhang L, Zhou W, Chen Y, Cheng Y, Dong J. LIMD1 phosphorylation in mitosis is required for mitotic progression and its tumor-suppressing activity. FEBS J 2019; 286:963-974. [PMID: 30600590 DOI: 10.1111/febs.14743] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 10/23/2018] [Accepted: 01/01/2019] [Indexed: 12/13/2022]
Abstract
LIM domains containing 1 (LIMD1) is a member of the Zyxin family proteins and functions as a tumor suppressor in lung cancer. LIMD1 has been shown to regulate Hippo-YAP signaling activity. Here, we report a novel regulatory mechanism for LIMD1. We found that cyclin-dependent kinase 1 (CDK1) and c-Jun NH2-terminal kinases 1/2 (JNK1/2) phosphorylate LIMD1 in vitro and in cells during anti-tubulin drug-induced mitotic arrest. Phosphorylation also occurs during normal mitosis. S272, S277, S421, and S424 were identified as the main phosphorylation sites in LIMD1. Deletion of LIMD1 resulted in a shortened mitotic cell cycle and phosphorylation of LIMD1 is required for proper mitotic progression. We further showed that the phosphorylation-deficient mutant LIMD1-4A is less active in suppressing cell proliferation, anchorage-independent growth, cell migration, and invasion in lung cancer cells. Together, our findings suggest that LIMD1 is a key regulator of mitotic progression, and that dysregulation of LIMD1 contributes to tumorigenesis.
Collapse
Affiliation(s)
- Jiuli Zhou
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA.,Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Lin Zhang
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, China
| | - Wei Zhou
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, China
| | - Yuanhong Chen
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Yufeng Cheng
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, China
| | - Jixin Dong
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
19
|
Tang J, Zhu L, Huang Y, Shi B, Zhang S, Gu L, Zhao J, Deng M, Zhu J, Xun H, Wang Y, Wang C. Silencing of LIMD1 promotes proliferation and reverses cell adhesion-mediated drug resistance in non-Hodgkin's lymphoma. Oncol Lett 2019; 17:2993-3000. [PMID: 30854077 DOI: 10.3892/ol.2019.9921] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 11/08/2018] [Indexed: 01/05/2023] Open
Abstract
LIM domains-containing protein 1 (LIMD1) is a tumor suppressor protein downregulated in numerous solid malignancies. However, the functional role of LIMD1 in non-Hodgkin's lymphoma (NHL) remains unclear. In the present study, it was demonstrated that LIMD1 is associated with the proliferation of NHL and cell adhesion mediated-drug resistance (CAM-DR). It was indicated by western blot analysis that LIMD1expression is lower in progressive lymphoma compared with indolent lymphoma. Furthermore, it was indicated that the role of LIMD1 in cell viability and proliferation remains unclear. Finally, the present study demonstrated that LIMD1 serves an important role in CAM-DR by regulating cell cycle progression. Silencing of LIMD1 may reverse CAM-DR in NHL. Therefore, the findings of the present study suggested that LIMD1 may be a potential therapeutic target for patients with NHL.
Collapse
Affiliation(s)
- Jie Tang
- Department of Oncology, Liyang People's Hospital, Liyang, Jiangsu 213300, P.R. China
| | - Liqun Zhu
- Department of Oncology, Liyang People's Hospital, Liyang, Jiangsu 213300, P.R. China
| | - Yuejiao Huang
- Department of Oncology, Affiliated Cancer Hospital of Nantong University, Nantong, Jiangsu 226002, P.R. China
| | - Bing Shi
- Department of Oncology, The Second People's Hospital of Nantong, Nantong, Jiangsu 226002, P.R. China
| | - Shuqing Zhang
- Department of Oncology, The Second People's Hospital of Nantong, Nantong, Jiangsu 226002, P.R. China
| | - Lingli Gu
- Department of Oncology, The Second People's Hospital of Nantong, Nantong, Jiangsu 226002, P.R. China
| | - Jie Zhao
- Department of Pathogenic Biology, Medical College, Nantong University, Nantong, Jiangsu 226002, P.R. China
| | - Minghao Deng
- Department of Pathogenic Biology, Medical College, Nantong University, Nantong, Jiangsu 226002, P.R. China
| | - Jiahao Zhu
- Department of Pathogenic Biology, Medical College, Nantong University, Nantong, Jiangsu 226002, P.R. China
| | - He Xun
- Department of Pathogenic Biology, Medical College, Nantong University, Nantong, Jiangsu 226002, P.R. China
| | - Yuchan Wang
- Department of Pathogenic Biology, Medical College, Nantong University, Nantong, Jiangsu 226002, P.R. China
| | - Chun Wang
- Department of Oncology, Liyang People's Hospital, Liyang, Jiangsu 213300, P.R. China
| |
Collapse
|
20
|
Zhang D, Li S, Yu W, Chen C, Liu T, Sun Y, Zhao Z, Liu L. LIMD1 is a survival prognostic marker of gastric cancer and hinders tumor progression by suppressing activation of YAP1. Cancer Manag Res 2018; 10:4349-4361. [PMID: 30349368 PMCID: PMC6188213 DOI: 10.2147/cmar.s174856] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
PURPOSE The purpose of this study was to investigate the clinical significance of LIMD1 and its biological roles in gastric cancer (GC). MATERIALS AND METHODS The prognostic value of LIMD1 in GC patients was determined by the online tool Kaplan-Meier Plotter. The biological functions of LIMD1 in GC were examined by in vitro assays, including proliferation, anchorage-independent growth, migration, invasion, and epithelial to mesenchymal transition (EMT) assays. The levels of downstream YAP1 regulated by LIMD1 were measured by Western blot analysis, and the sub-localization of YAP1 in GC cells was visualized by immunofluorescence staining. Differential expression levels and copy number levels of LIMD1 between GC and normal tissues were compared using the Oncomine database. A correlation of LIMD1 mRNA level and the copy number level was depicted by cBioPortal. We also evaluated the methylation status around the LIMD1 genes by Wanderer. RESULTS The expression level of LIMD1 positively correlated with the prognosis of GC patients regardless of tumor stage, size, lymph node, metastasis, Lauren's classification, differentiation, gender, treatment, and ERBB2 amplification status. Overexpression of LIMD1 impeded the tumor growth, cell motility, invasiveness, and metastasis, and knockdown of LIMD1 promoted these phenotypes in GC cells. Mechanistically, YAP1 was one of the downstream effectors of LIMD1; LIMD1 suppressed the expression of YAP1 as well as its intracellular translocation. Furthermore, we found that LIMD1 expression was reduced in some of the GC profiling datasets. Gene deletion, instead of DNA methylation, contributed to the reduced expression of LIMD1 in GC. CONCLUSION Our results identified LIMD1 as a convincing prognostic marker as well as a potentially therapeutic target for GC.
Collapse
Affiliation(s)
- Di Zhang
- Department of Chemotherapy, Cancer Center, Qilu Hospital of Shandong University, Jinan, Shandong, China,
| | - Song Li
- Department of Chemotherapy, Cancer Center, Qilu Hospital of Shandong University, Jinan, Shandong, China,
| | - Wenbin Yu
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Cheng Chen
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Teng Liu
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Yiting Sun
- Department of Chemotherapy, Cancer Center, Qilu Hospital of Shandong University, Jinan, Shandong, China,
| | - Zeyi Zhao
- Department of Chemotherapy, Cancer Center, Qilu Hospital of Shandong University, Jinan, Shandong, China,
| | - Lian Liu
- Department of Chemotherapy, Cancer Center, Qilu Hospital of Shandong University, Jinan, Shandong, China,
| |
Collapse
|
21
|
Foxler DE, Bridge KS, Foster JG, Grevitt P, Curry S, Shah KM, Davidson KM, Nagano A, Gadaleta E, Rhys HI, Kennedy PT, Hermida MA, Chang TY, Shaw PE, Reynolds LE, McKay TR, Wang HW, Ribeiro PS, Plevin MJ, Lagos D, Lemoine NR, Rajan P, Graham TA, Chelala C, Hodivala-Dilke KM, Spendlove I, Sharp TV. A HIF-LIMD1 negative feedback mechanism mitigates the pro-tumorigenic effects of hypoxia. EMBO Mol Med 2018; 10:e8304. [PMID: 29930174 PMCID: PMC6079541 DOI: 10.15252/emmm.201708304] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 05/23/2018] [Accepted: 05/28/2018] [Indexed: 12/23/2022] Open
Abstract
The adaptive cellular response to low oxygen tensions is mediated by the hypoxia-inducible factors (HIFs), a family of heterodimeric transcription factors composed of HIF-α and HIF-β subunits. Prolonged HIF expression is a key contributor to cellular transformation, tumorigenesis and metastasis. As such, HIF degradation under hypoxic conditions is an essential homeostatic and tumour-suppressive mechanism. LIMD1 complexes with PHD2 and VHL in physiological oxygen levels (normoxia) to facilitate proteasomal degradation of the HIF-α subunit. Here, we identify LIMD1 as a HIF-1 target gene, which mediates a previously uncharacterised, negative regulatory feedback mechanism for hypoxic HIF-α degradation by modulating PHD2-LIMD1-VHL complex formation. Hypoxic induction of LIMD1 expression results in increased HIF-α protein degradation, inhibiting HIF-1 target gene expression, tumour growth and vascularisation. Furthermore, we report that copy number variation at the LIMD1 locus occurs in 47.1% of lung adenocarcinoma patients, correlates with enhanced expression of a HIF target gene signature and is a negative prognostic indicator. Taken together, our data open a new field of research into the aetiology, diagnosis and prognosis of LIMD1-negative lung cancers.
Collapse
Affiliation(s)
- Daniel E Foxler
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Katherine S Bridge
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - John G Foster
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Paul Grevitt
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Sean Curry
- Faculty of Medicine and Life Sciences, University of Nottingham, Nottingham, UK
| | - Kunal M Shah
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Kathryn M Davidson
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Ai Nagano
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Emanuela Gadaleta
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | | | - Paul T Kennedy
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Miguel A Hermida
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Ting-Yu Chang
- Institute of Microbiology and Immunology, National Yang Ming University, Taipei City, Taiwan
| | - Peter E Shaw
- Faculty of Medicine and Life Sciences, University of Nottingham, Nottingham, UK
| | - Louise E Reynolds
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Tristan R McKay
- School of Healthcare Science, Manchester Metropolitan University, Manchester, UK
| | - Hsei-Wei Wang
- Institute of Microbiology and Immunology, National Yang Ming University, Taipei City, Taiwan
| | - Paulo S Ribeiro
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | | | - Dimitris Lagos
- Centre for Immunology and Infection, Hull York Medical School and Department of Biology, University of York, York, UK
| | - Nicholas R Lemoine
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Prabhakar Rajan
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Trevor A Graham
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Claude Chelala
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | | | - Ian Spendlove
- Faculty of Medicine and Life Sciences, University of Nottingham, Nottingham, UK
| | - Tyson V Sharp
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London, UK
| |
Collapse
|
22
|
Chakraborty C, Mitra S, Roychowdhury A, Samadder S, Dutta S, Roy A, Das P, Mandal RK, Sharp TV, Roychoudhury S, Panda CK. Deregulation of LIMD1-VHL-HIF-1α-VEGF pathway is associated with different stages of cervical cancer. Biochem J 2018; 475:1793-1806. [PMID: 29654110 DOI: 10.1042/bcj20170649] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 04/05/2018] [Accepted: 04/12/2018] [Indexed: 03/25/2024]
Abstract
To understand the mechanism of cellular stress in basal-parabasal layers of normal cervical epithelium and during different stages of cervical carcinoma, we analyzed the alterations (expression/methylation/copy number variation/mutation) of HIF-1α and its associated genes LIMD1, VHL and VEGF in disease-free normal cervix (n = 9), adjacent normal cervix of tumors (n = 70), cervical intraepithelial neoplasia (CIN; n = 32), cancer of uterine cervix (CACX; n = 174) samples and two CACX cell lines. In basal-parabasal layers of normal cervical epithelium, LIMD1 showed high protein expression, while low protein expression of VHL was concordant with high expression of HIF-1α and VEGF irrespective of HPV-16 (human papillomavirus 16) infection. This was in concordance with the low promoter methylation of LIMD1 and high in VHL in the basal-parabasal layers of normal cervix. LIMD1 expression was significantly reduced while VHL expression was unchanged during different stages of cervical carcinoma. This was in concordance with their frequent methylation during different stages of this tumor. In different stages of cervical carcinoma, the expression pattern of HIF-1α and VEGF was high as seen in basal-parabasal layers and inversely correlated with the expression of LIMD1 and VHL. This was validated by demethylation experiments using 5-aza-2'-deoxycytidine in CACX cell lines. Additional deletion of LIMD1 and VHL in CIN/CACX provided an additional growth advantage during cervical carcinogenesis through reduced expression of genes and associated with poor prognosis of patients. Our data showed that overexpression of HIF-1α and its target gene VEGF in the basal-parabasal layers of normal cervix was due to frequent inactivation of VHL by its promoter methylation. This profile was maintained during different stages of cervical carcinoma with additional methylation/deletion of VHL and LIMD1.
Collapse
Affiliation(s)
| | - Sraboni Mitra
- Department of Systems Biology, MD Anderson Cancer Center, Houston, TX, U.S.A
| | - Anirban Roychowdhury
- Department of Oncogene Regulation, Chittaranjan National Cancer Institute, Kolkata, India
| | - Sudip Samadder
- Department of Oncogene Regulation, Chittaranjan National Cancer Institute, Kolkata, India
| | - Sankhadeep Dutta
- Infections and Cancer Biology Division, International Agency for Research on Cancer, Lyon, France
| | - Anup Roy
- Department of Pathology, Nil Ratan Sircar Medical College and Hospital, Kolkata, India
| | - Pradip Das
- Department of Gynaecologic Oncology, Chittaranjan National Cancer Institute, Kolkata, India
| | - Ranajit Kumar Mandal
- Department of Gynaecologic Oncology, Chittaranjan National Cancer Institute, Kolkata, India
| | - Tyson V Sharp
- Barts Cancer Institute, Queen Mary University of London, London, U.K
| | - Susanta Roychoudhury
- Saroj Gupta Cancer Centre and Research Institute, MG Road, Thakurpukur, Kolkata, India
| | - Chinmay Kumar Panda
- Department of Oncogene Regulation, Chittaranjan National Cancer Institute, Kolkata, India
| |
Collapse
|
23
|
Sarkar S, Alam N, Mandal SS, Chatterjee K, Ghosh S, Roychoudhury S, Panda CK. Differential transmission of the molecular signature of RBSP3, LIMD1 and CDC25A in basal/ parabasal versus spinous of normal epithelium during head and neck tumorigenesis: A mechanistic study. PLoS One 2018; 13:e0195937. [PMID: 29672635 PMCID: PMC5909606 DOI: 10.1371/journal.pone.0195937] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 04/03/2018] [Indexed: 12/16/2022] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is a global disease and mortality burden, necessitating the elucidation of its molecular progression for effective disease management. The study aims to understand the molecular profile of three candidate cell cycle regulatory genes, RBSP3, LIMD1 and CDC25A in the basal/ parabasal versus spinous layer of normal oral epithelium and during head and neck tumorigenesis. Immunohistochemical expression and promoter methylation was used to determine the molecular signature in normal oral epithelium. The mechanism of alteration transmission of this profile during tumorigenesis was then explored through additional deletion and mutation in HPV/ tobacco etiological groups, followed byclinico-pathological correlation. In basal/parabasal layer, the molecular signature of the genes was low protein expression/ high promoter methylation of RBSP3, high expression/ low methylation of LIMD1 and high expression of CDC25A. Dysplastic epithelium maintained the signature of RBSP3 through high methylation/ additional deletion with loss of the signatures of LIMD1 and CDC25A via deletion/ additional methylation. Similarly, maintenance and / or loss of signature in invasive tumors was by recurrent deletion/ methylation. Thus, differential patterns of alteration of the genes might be pre-requisite for the development of dysplastic and invasive lesions. Etiological factors played a key role in promoting genetic alterations and determining prognosis. Tobacco negative HNSCC patients had significantly lower alterations of LIMD1 and CDC25A, along with better survival among tobacco negative/ HPV positive patients. Our data suggests the necessity for perturbation of normal molecular profile of RBSP3, LIMD1 and CDC25A in conjunction with etiological factors for head and neck tumorigenesis, implying their diagnostic and prognostic significance.
Collapse
Affiliation(s)
- Shreya Sarkar
- Department of Oncogene Regulation, Chittaranjan National Cancer Institute, Kolkata, India
| | - Neyaz Alam
- Department of Surgical Oncology, Chittaranjan National Cancer Institute, Kolkata, India
| | - Syam Sundar Mandal
- Department of Epidemiology and Biostatistics, Chittaranjan National Cancer Institute, Kolkata, India
| | - Kabita Chatterjee
- Department of Oral and Maxillofacial Pathology, Buddha Institute of Dental Sciences and Hospital, Patna, India
| | - Supratim Ghosh
- Department of Oral and Maxillofacial Pathology, Burdwan Dental College and Hospital, Burdwan, India
| | - Susanta Roychoudhury
- Basic Research, Saroj Gupta Cancer Centre and Research Institute, Kolkata, India
| | - Chinmay Kumar Panda
- Department of Oncogene Regulation, Chittaranjan National Cancer Institute, Kolkata, India
- * E-mail:
| |
Collapse
|
24
|
Over expression of HIF1α is associated with inactivation of both LimD1 and VHL in renal cell carcinoma: Clinical importance. Pathol Res Pract 2017; 213:1477-1481. [DOI: 10.1016/j.prp.2017.10.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 10/03/2017] [Accepted: 10/07/2017] [Indexed: 01/07/2023]
|
25
|
Chen X, Stauffer S, Chen Y, Dong J. Ajuba Phosphorylation by CDK1 Promotes Cell Proliferation and Tumorigenesis. J Biol Chem 2016; 291:14761-72. [PMID: 27226586 DOI: 10.1074/jbc.m116.722751] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Indexed: 12/21/2022] Open
Abstract
Recent studies identified the adaptor protein Ajuba as a positive regulator of Yes-associated protein (YAP) oncogenic activity through inhibiting large tumor suppressor (Lats1/2) core kinases of the Hippo pathway, a signaling pathway that plays important roles in cancer. In this study, we define a novel mechanism for phospho-regulation of Ajuba in mitosis and its biological significance in cancer. We found that Ajuba is phosphorylated in vitro and in vivo by cyclin-dependent kinase 1 (CDK1) at Ser(119) and Ser(175) during the G2/M phase of the cell cycle. Mitotic phosphorylation of Ajuba controls the expression of multiple cell cycle regulators; however, it does not affect Hippo signaling activity, nor does it induce epithelial-mesenchymal transition. We further showed that mitotic phosphorylation of Ajuba is sufficient to promote cell proliferation and anchorage-independent growth in vitro and tumorigenesis in vivo Collectively, our discoveries reveal a previously unrecognized mechanism for Ajuba regulation in mitosis and its role in tumorigenesis.
Collapse
Affiliation(s)
- Xingcheng Chen
- From the Eppley Institute for Research in Cancer, Fred and Pamela Buffett Cancer Center and the Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska 68198
| | - Seth Stauffer
- From the Eppley Institute for Research in Cancer, Fred and Pamela Buffett Cancer Center and the Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska 68198
| | - Yuanhong Chen
- From the Eppley Institute for Research in Cancer, Fred and Pamela Buffett Cancer Center and
| | - Jixin Dong
- From the Eppley Institute for Research in Cancer, Fred and Pamela Buffett Cancer Center and
| |
Collapse
|
26
|
Abstract
Schimizzi and Longmore summarise what we know about Ajuba LIM-domain proteins and their various subcellular roles.
Collapse
Affiliation(s)
- Gregory V Schimizzi
- ICCE Institute, Department of Medicine and Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Gregory D Longmore
- ICCE Institute, Department of Medicine and Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
27
|
Foster JG, Wong SCK, Sharp TV. The hypoxic tumor microenvironment: driving the tumorigenesis of non-small-cell lung cancer. Future Oncol 2015; 10:2659-74. [PMID: 25531051 DOI: 10.2217/fon.14.201] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Since the application of molecular biology in cancer biology, lung cancer research has classically focused on molecular drivers of disease. One such pathway, the hypoxic response pathway, is activated by reduced local oxygen concentrations at the tumor site. Hypoxia-driven gene and protein changes enhance epithelial-to-mesenchymal transition, remodel the extracellular matrix, drive drug resistance, support cancer stem cells and aid evasion from immune cells. However, it is not the tumor cells alone which drive this response to hypoxia, but rather their interaction with a complex milieu of supporting cells. This review will focus on recent advances in our understanding of how these cells contribute to the tumor response to hypoxia in non-small-cell lung cancer.
Collapse
Affiliation(s)
- John G Foster
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | | | | |
Collapse
|
28
|
RHOBTB3 promotes proteasomal degradation of HIFα through facilitating hydroxylation and suppresses the Warburg effect. Cell Res 2015. [PMID: 26215701 PMCID: PMC4559813 DOI: 10.1038/cr.2015.90] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Hypoxia-inducible factors (HIFs) are master regulators of adaptive responses to low oxygen, and their α-subunits are rapidly degraded through the ubiquitination-dependent proteasomal pathway after hydroxylation. Aberrant accumulation or activation of HIFs is closely linked to many types of cancer. However, how hydroxylation of HIFα and its delivery to the ubiquitination machinery are regulated remains unclear. Here we show that Rho-related BTB domain-containing protein 3 (RHOBTB3) directly interacts with the hydroxylase PHD2 to promote HIFα hydroxylation. RHOBTB3 also directly interacts with the von Hippel-Lindau (VHL) protein, a component of the E3 ubiquitin ligase complex, facilitating ubiquitination of HIFα. Remarkably, RHOBTB3 dimerizes with LIMD1, and constructs a RHOBTB3/LIMD1-PHD2-VHL-HIFα complex to effect the maximal degradation of HIFα. Hypoxia reduces the RHOBTB3-centered complex formation, resulting in an accumulation of HIFα. Importantly, the expression level of RHOBTB3 is greatly reduced in human renal carcinomas, and RHOBTB3 deficiency significantly elevates the Warburg effect and accelerates xenograft growth. Our work thus reveals that RHOBTB3 serves as a scaffold to organize a multi-subunit complex that promotes the hydroxylation, ubiquitination and degradation of HIFα.
Collapse
|
29
|
Haubner BJ, Moik D, Schuetz T, Reiner MF, Voelkl JG, Streil K, Bader K, Zhao L, Scheu C, Mair J, Pachinger O, Metzler B. In vivo cardiac role of migfilin during experimental pressure overload. Cardiovasc Res 2015; 106:398-407. [DOI: 10.1093/cvr/cvv125] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 03/14/2015] [Indexed: 11/14/2022] Open
|
30
|
Wang L, Yao ZQ, Moorman JP, Xu Y, Ning S. Gene expression profiling identifies IRF4-associated molecular signatures in hematological malignancies. PLoS One 2014; 9:e106788. [PMID: 25207815 PMCID: PMC4160201 DOI: 10.1371/journal.pone.0106788] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 07/31/2014] [Indexed: 12/29/2022] Open
Abstract
The lymphocyte-specific transcription factor Interferon (IFN) Regulatory Factor 4 (IRF4) is implicated in certain types of lymphoid and myeloid malignancies. However, the molecular mechanisms underlying its interactions with these malignancies are largely unknown. In this study, we have first profiled molecular signatures associated with IRF4 expression in associated cancers, by analyzing existing gene expression profiling datasets. Our results show that IRF4 is overexpressed in melanoma, in addition to previously reported contexts including leukemia, myeloma, and lymphoma, and that IRF4 is associated with a unique gene expression pattern in each context. A pool of important genes involved in B-cell development, oncogenesis, cell cycle regulation, and cell death including BATF, LIMD1, CFLAR, PIM2, and CCND2 are common signatures associated with IRF4 in non-Hodgkin B cell lymphomas. We confirmed the correlation of IRF4 with LIMD1 and CFLAR in a panel of cell lines derived from lymphomas. Moreover, we profiled the IRF4 transcriptome in the context of EBV latent infection, and confirmed several genes including IFI27, IFI44, GBP1, and ARHGAP18, as well as CFLAR as novel targets for IRF4. These results provide valuable information for understanding the IRF4 regulatory network, and improve our knowledge of the unique roles of IRF4 in different hematological malignancies.
Collapse
Affiliation(s)
- Ling Wang
- Center for Inflammation, Infectious Diseases and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, United States of America
| | - Zhi Q. Yao
- Center for Inflammation, Infectious Diseases and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, United States of America
- HIV/HCV Program, James H. Quillen VA Medical Center, Johnson City, Tennessee, United States of America
| | - Jonathan P. Moorman
- Center for Inflammation, Infectious Diseases and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, United States of America
- HIV/HCV Program, James H. Quillen VA Medical Center, Johnson City, Tennessee, United States of America
| | - Yanji Xu
- Shaun and Lilly International, LLC, Collierville, Tennessee, United States of America
| | - Shunbin Ning
- Center for Inflammation, Infectious Diseases and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, United States of America
- * E-mail:
| |
Collapse
|
31
|
Zhang L, Ramchandren R, Papenhausen P, Loughran TP, Sokol L. Transformed aggressive γδ-variant T-cell large granular lymphocytic leukemia with acquired copy neutral loss of heterozygosity at 17q11.2q25.3 and additional aberrations. Eur J Haematol 2014; 93:260-4. [PMID: 24635703 DOI: 10.1111/ejh.12313] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/11/2014] [Indexed: 11/28/2022]
Abstract
T-cell large granular lymphocytic leukemia (T-LGLL) is a rare indolent lymphoproliferative disorder characterized by cytopenias, splenomegaly, and various degrees of T-cell lymphocytosis, due to a clonal expansion of CD8-positive cytotoxic T-cells. Phenotypic variants of T-LGLL include CD4(+) /CD8(-) T-cells, with dual CD4(-) /CD8(-) /γδ(+) T-cells being even rarer. Cytogenetic abnormalities in T-LGLL have rarely been reported, and there is scientific debate regarding the existence of aggressive or transformed variants of T-LGLL. We report a patient with T-LGLL, γδ variant, with nearly 20-year-long duration of cytopenias before transformation to an unusual clinical scenario, manifesting with marked lymphocytosis >100 × 10(9) /L and infiltration of lymph nodes, tonsils, and subcutaneous tissue. Single-nucleotide polymorphism assays revealed acquired copy neutral loss of heterozygosity at 17q and deletion of 3p21.31, in addition to trisomy 5, monosomy X, and monosomy 21. These genetic abnormalities provided a better understanding of the molecular nature and the potentiality of disease transformation.
Collapse
Affiliation(s)
- Ling Zhang
- Department of Hematopathology and Laboratory Medicine, H Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | | | | | | | | |
Collapse
|
32
|
Kooistra SM, Nørgaard LCR, Lees MJ, Steinhauer C, Johansen JV, Helin K. A screen identifies the oncogenic micro-RNA miR-378a-5p as a negative regulator of oncogene-induced senescence. PLoS One 2014; 9:e91034. [PMID: 24651706 PMCID: PMC3961217 DOI: 10.1371/journal.pone.0091034] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Accepted: 02/06/2014] [Indexed: 12/15/2022] Open
Abstract
Oncogene-induced senescence (OIS) can occur in response to hyperactive oncogenic signals and is believed to be a fail-safe mechanism protecting against tumorigenesis. To identify new factors involved in OIS, we performed a screen for microRNAs that can overcome or inhibit OIS in human diploid fibroblasts. This screen led to the identification of miR-378a-5p and in addition several other miRNAs that have previously been shown to play a role in senescence. We show that ectopic expression of miR-378a-5p reduces the expression of several senescence markers, including p16INK4A and senescence-associated β-galactosidase. Moreover, cells with ectopic expression of miR-378a-5p retain proliferative capacity even in the presence of an activated Braf oncogene. Finally, we identified several miR-378a-5p targets in diploid fibroblasts that might explain the mechanism by which the microRNA can delay OIS. We speculate that miR-378a-5p might positively influence tumor formation by delaying OIS, which is consistent with a known pro-oncogenic function of this microRNA.
Collapse
Affiliation(s)
- Susanne Marije Kooistra
- Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark; Centre for Epigenetics, University of Copenhagen, Copenhagen, Denmark
| | - Lise Christine Rudkjær Nørgaard
- Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark; Centre for Epigenetics, University of Copenhagen, Copenhagen, Denmark
| | - Michael James Lees
- Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark; Centre for Epigenetics, University of Copenhagen, Copenhagen, Denmark
| | - Cornelia Steinhauer
- Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark; Centre for Epigenetics, University of Copenhagen, Copenhagen, Denmark
| | - Jens Vilstrup Johansen
- Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark; The Bioinformatics Centre, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Kristian Helin
- Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark; Centre for Epigenetics, University of Copenhagen, Copenhagen, Denmark; The Danish Stem Cell Center (DanStem), University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
33
|
Mayank AK, Sharma S, Deshwal RK, Lal SK. LIMD1 antagonizes E2F1 activity and cell cycle progression by enhancing Rb function in cancer cells. Cell Biol Int 2014; 38:809-17. [DOI: 10.1002/cbin.10266] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2013] [Accepted: 01/30/2014] [Indexed: 12/12/2022]
Affiliation(s)
- Adarsh K. Mayank
- School of Life Sciences; Singhania University; Pacheri Beri Rajasthan India
| | - Shipra Sharma
- Virology Group; International Centre for Genetic Engineering and Biotechnology; Aruna Asaf Ali Marg New Delhi India
| | - Ravi K Deshwal
- Apex Institute of Management and Science; Jaipur Rajasthan
| | - Sunil K. Lal
- Virology Group; International Centre for Genetic Engineering and Biotechnology; Aruna Asaf Ali Marg New Delhi India
| |
Collapse
|
34
|
A novel 3p22.3 gene CMTM7 represses oncogenic EGFR signaling and inhibits cancer cell growth. Oncogene 2013; 33:3109-18. [PMID: 23893243 DOI: 10.1038/onc.2013.282] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2012] [Revised: 04/11/2013] [Accepted: 04/29/2013] [Indexed: 12/15/2022]
Abstract
Deletion of 3p12-22 is frequent in multiple cancer types, indicating the presence of critical tumor-suppressor genes (TSGs) at this region. We studied a novel candidate TSG, CMTM7, located at the 3p22.3 CMTM-gene cluster, for its tumor-suppressive functions and related mechanisms. The three CMTM genes, CMTM6, 7 and 8, are broadly expressed in human normal adult tissues and normal epithelial cell lines. Only CMTM7 is frequently silenced or downregulated in esophageal and nasopharyngeal cell lines, but uncommon in other carcinoma cell lines. Immunostaining of tissue microarrays for CMTM7 protein showed its downregulation or absence in esophageal, gastric, pancreatic, liver, lung and cervix tumor tissues. Promoter CpG methylation and loss of heterozygosity were both found contributing to CMTM7 downregulation. Ectopic expression of CMTM7 in carcinoma cells inhibits cell proliferation, motility and tumor formation in nude mice, but not in immortalized normal cells, suggesting a tumor inhibitory role of CMTM7. The tumor-suppressive function of CMTM7 is associated with its role in G1/S cell cycle arrest, through upregulating p27 and downregulating cyclin-dependent kinase 2 (CDK2) and 6 (CDK6). Moreover, CMTM7 could promote epidermal growth factor receptor (EGFR) internalization, and further suppress AKT signaling pathway. Thus, our findings suggest that CMTM7 is a novel 3p22 tumor suppressor regulating G1/S transition and EGFR/AKT signaling during tumor pathogenesis.
Collapse
|
35
|
Schiller HB, Fässler R. Mechanosensitivity and compositional dynamics of cell-matrix adhesions. EMBO Rep 2013; 14:509-19. [PMID: 23681438 DOI: 10.1038/embor.2013.49] [Citation(s) in RCA: 212] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2012] [Accepted: 03/21/2013] [Indexed: 12/27/2022] Open
Abstract
Cells perceive information about the biochemical and biophysical properties of their tissue microenvironment through integrin-mediated cell-matrix adhesions, which connect the cytoskeleton with the extracellular matrix and thereby allow cohesion and long-range mechanical connections within tissues. The formation of cell-matrix adhesions and integrin signalling involves the dynamic recruitment and assembly of an inventory of proteins, collectively termed the 'adhesome', at the adhesive site. The recruitment of some adhesome proteins, most notably the Lin11-, Isl1- and Mec3-domain-containing proteins, depends on mechanical tension generated by myosin II-mediated contractile forces exerted on cell-matrix adhesions. When exposed to force, mechanosensitive adhesome proteins can change their conformation or expose cryptic-binding sites leading to the recruitment of proteins, rearrangement of the cytoskeleton, reinforcement of the adhesive site and signal transduction. Biophysical methods and proteomics revealed force ranges within the adhesome and cytoskeleton, and also force-dependent changes in adhesome composition. In this review, we provide an overview of the compositional dynamics of cell-matrix adhesions, discuss the most prevalent functional domains in adhesome proteins and review literature and concepts about mechanosensing mechanisms that operate at the adhesion site.
Collapse
Affiliation(s)
- Herbert B Schiller
- Department of Molecular Medicine, Max Planck Institute of Biochemistry, Am Klopferspitz 18, D-82152 Martinsried, Germany
| | | |
Collapse
|
36
|
Zhang K, Corsa CA, Ponik SM, Prior JL, Piwnica-Worms D, Eliceiri KW, Keely PJ, Longmore GD. The collagen receptor discoidin domain receptor 2 stabilizes SNAIL1 to facilitate breast cancer metastasis. Nat Cell Biol 2013; 15:677-87. [PMID: 23644467 DOI: 10.1038/ncb2743] [Citation(s) in RCA: 316] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2013] [Accepted: 03/28/2013] [Indexed: 12/16/2022]
Abstract
Increased stromal collagen deposition in human breast tumours correlates with metastases. We show that activation of the collagen I receptor DDR2 (discoidin domain receptor 2) regulates SNAIL1 stability by stimulating ERK2 activity, in a Src-dependent manner. Activated ERK2 directly phosphorylates SNAIL1, leading to SNAIL1 nuclear accumulation, reduced ubiquitylation and increased protein half-life. DDR2-mediated stabilization of SNAIL1 promotes breast cancer cell invasion and migration in vitro, and metastasis in vivo. DDR2 expression was observed in most human invasive ductal breast carcinomas studied, and was associated with nuclear SNAIL1 and absence of E-cadherin expression. We propose that DDR2 maintains SNAIL1 level and activity in tumour cells that have undergone epithelial-mesenchymal transition (EMT), thereby facilitating continued tumour cell invasion through collagen-I-rich extracellular matrices by sustaining the EMT phenotype. As such, DDR2 could be an RTK (receptor tyrosine kinase) target for the treatment of breast cancer metastasis.
Collapse
Affiliation(s)
- Kun Zhang
- BRIGHT Institute, Washington University, St Louis, Missouri 63110, USA
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Zhang X, Li HM, Liu Z, Zhou G, Zhang Q, Zhang T, Zhang J, Zhang C. Loss of heterozygosity and methylation of multiple tumor suppressor genes on chromosome 3 in hepatocellular carcinoma. J Gastroenterol 2013; 48:132-43. [PMID: 22766745 DOI: 10.1007/s00535-012-0621-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2011] [Accepted: 05/21/2012] [Indexed: 02/04/2023]
Abstract
BACKGROUND Genetic and epigenetic alterations are the two key mechanisms in the development of hepatocellular carcinoma (HCC). However, how they contribute to hepatocarcinogenesis and the correlation between them has not been fully elucidated. METHODS A total of 48 paired HCCs and noncancerous tissues were used to detect loss of heterozygosity (LOH) and the methylation profiles of five tumor suppressor genes (RASSF1A, BLU, FHIT, CRBP1, and HLTF) on chromosome 3 by using polymerase chain reaction (PCR) and methylation-specific PCR. Gene expression was analyzed by immunohistochemistry and reverse transcription (RT)-PCR. RESULTS Sixteen of 48 (33.3 %) HCCs had LOH on at least one locus on chromosome 3, and two smallest common deleted regions (3p22.3-24.3 and 3p12.3-14.2) were identified. RASSF1A, BLU, and FHIT showed very high frequencies of methylation in HCCs (100, 81.3, and 64.6 %, respectively) and noncancerous tissues, but not in liver tissues from control patients. Well-differentiated HCCs showed high methylation frequencies of these genes but very low frequencies of LOH. Furthermore, BLU methylation was associated with an increased level of alpha-fetoprotein, and FHIT methylation was inversely correlated with HCC recurrence. In comparison, CRBP1 showed moderate frequencies of methylation, while HLTF showed low frequencies of methylation, and CRBP1 methylation occurred mainly in elderly patients. Treatment with 5-aza-2'-deoxycytidine demethylated at least one of these genes and restored their expression in a DNA methylation-dependent or -independent manner. CONCLUSIONS Hypermethylation of RASSF1A, BLU, and FHIT is a common and very early event in hepatocarcinogenesis; CRBP1 methylation may also be involved in the later stage. Although LOH was not too frequent on chromosome 3, it may play a role as another mechanism in hepatocarcinogenesis.
Collapse
Affiliation(s)
- Xiaoying Zhang
- Institute of Pathology and Pathophysiology, Shandong University School of Medicine, Jinan, 250012, China
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Maiti GP, Ghosh A, Chatterjee R, Roy A, Sharp TV, Roychoudhury S, Panda CK. Reduced Expression of Limd1 in Ulcerative Oral Epithelium Associated with Tobacco and Areca Nut. Asian Pac J Cancer Prev 2012; 13:4341-6. [DOI: 10.7314/apjcp.2012.13.9.4341] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
39
|
Yang M, Li W, Liu YY, Fu S, Qiu GB, Sun KL, Fu WN. Promoter hypermethylation-induced transcriptional down-regulation of the gene MYCT1 in laryngeal squamous cell carcinoma. BMC Cancer 2012; 12:219. [PMID: 22672838 PMCID: PMC3472177 DOI: 10.1186/1471-2407-12-219] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2012] [Accepted: 06/06/2012] [Indexed: 12/21/2022] Open
Abstract
Background MYCT1, previously named MTLC, is a novel candidate tumor suppressor gene. MYCT1 was cloned from laryngeal squamous cell cancer (LSCC) and has been found to be down-regulated in LSCC; however, the regulatory details have not been fully elucidated. Methods Here, we sought to investigate the methylation status of the CpG islands of MYCT1 and mRNA levels by bisulfite-specific PCR (BSP) based on sequencing restriction enzyme digestion, reverse transcription and real-time quantitative polymerase chain reaction (RQ-PCR). The function of specific sites in the proximal promoter of MYCT1 in LSCC was measured by transient transfection, luciferase assays, electrophoretic mobility shift assay (EMSA) and chromatin immunoprecipitation assay (ChIP). Results The results suggested hypermethylation of 12 CpG sites of the promoter in both laryngeal cancer tissues and the laryngeal cancer line Hep-2 cell. The hypermethylation of the site CGCG (−695 to −692), which has been identified as the c-Myc binding site, was identified in laryngeal cancer tissues (59/73) compared to paired mucosa (13/73); in addition, statistical analysis revealed that the methylation status of this site significantly correlated with cancer cell differentiation(p < 0.01). The mRNA level of MYCT1 increased in Hep-2 cells treated with 5-aza-C (p < 0.01). The luciferase activity from mutant transfectants pGL3-MYCT1m (−852/+12, mut-695-C > A, mut-693-C > G) was significantly reduced compared with the wild type pGL3-MYCT1 (−852/+12), while the luciferase activity from wild transfectants pGL3-MYCT1 (−852/+12) rose after 5-aza treatment in Hep-2 cells. Finally, EMSA and ChIP confirmed that the methylation of the CGCG (−695 to −692) site prevented c-Myc from binding of the site and demethylation treatment of the 5′ flanking region of MYCT1 by 5-aza induced the increased occupation of the core promoter by c-Myc (p < 0.01). Conclusion In summary, this study concluded that hypermethylation contributed to the transcriptional down-regulation of MYCT1 and could inhibit cancer cell differentiation in LSCC. DNA methylation of the CGCG site (−695 to −692) of MYCT1 altered the promoter activity by interfering with its binding to c-Myc in LSCC. Epigenetic therapy of reactivating MYCT1 by 5-aza should be further evaluated in clinical trails of LSCC.
Collapse
Affiliation(s)
- Min Yang
- Department of Medical Genetics, China Medical University, Shenyang, P.R. China
| | | | | | | | | | | | | |
Collapse
|
40
|
LIM-domain proteins in transforming growth factor β-induced epithelial-to-mesenchymal transition and myofibroblast differentiation. Cell Signal 2012; 24:819-25. [DOI: 10.1016/j.cellsig.2011.12.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2011] [Revised: 11/15/2011] [Accepted: 12/04/2011] [Indexed: 12/16/2022]
|
41
|
Foxler DE, Bridge KS, James V, Webb TM, Mee M, Wong SCK, Feng Y, Constantin-Teodosiu D, Petursdottir TE, Bjornsson J, Ingvarsson S, Ratcliffe PJ, Longmore GD, Sharp TV. The LIMD1 protein bridges an association between the prolyl hydroxylases and VHL to repress HIF-1 activity. Nat Cell Biol 2012; 14:201-8. [PMID: 22286099 DOI: 10.1038/ncb2424] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2011] [Accepted: 12/16/2011] [Indexed: 12/15/2022]
Abstract
There are three prolyl hydroxylases (PHD1, 2 and 3) that regulate the hypoxia-inducible factors (HIFs), the master transcriptional regulators that respond to changes in intracellular O(2) tension. In high O(2) tension (normoxia) the PHDs hydroxylate two conserved proline residues on HIF-1α, which leads to binding of the von Hippel-Lindau (VHL) tumour suppressor, the recognition component of a ubiquitin-ligase complex, initiating HIF-1α ubiquitylation and degradation. However, it is not known whether PHDs and VHL act separately to exert their enzymatic activities on HIF-1α or as a multiprotein complex. Here we show that the tumour suppressor protein LIMD1 (LIM domain-containing protein) acts as a molecular scaffold, simultaneously binding the PHDs and VHL, thereby assembling a PHD-LIMD1-VHL protein complex and creating an enzymatic niche that enables efficient degradation of HIF-1α. Depletion of endogenous LIMD1 increases HIF-1α levels and transcriptional activity in both normoxia and hypoxia. Conversely, LIMD1 expression downregulates HIF-1 transcriptional activity in a manner depending on PHD and 26S proteasome activities. LIMD1 family member proteins Ajuba and WTIP also bind to VHL and PHDs 1 and 3, indicating that these LIM domain-containing proteins represent a previously unrecognized group of hypoxic regulators.
Collapse
Affiliation(s)
- Daniel E Foxler
- School of Biomedical Sciences, University of Nottingham, Queen's Medical Centre, NG7 2UH, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Das M, Ithychanda SS, Qin J, Plow EF. Migfilin and filamin as regulators of integrin activation in endothelial cells and neutrophils. PLoS One 2011; 6:e26355. [PMID: 22043318 PMCID: PMC3197140 DOI: 10.1371/journal.pone.0026355] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2011] [Accepted: 09/25/2011] [Indexed: 12/18/2022] Open
Abstract
Cell adhesion and migration depend on engagement of extracellular matrix ligands by integrins. Integrin activation is dynamically regulated by interactions of various cytoplasmic proteins, such as filamin and integrin activators, talin and kindlin, with the cytoplasmic tail of the integrin β subunit. Although filamin has been suggested to be an inhibitor of integrin activation, direct functional evidence for the inhibitory role of filamin is limited. Migfilin, a filamin-binding protein enriched at cell-cell and cell-extracellular matrix contact sites, can displace filamin from β1 and β3 integrins and promote integrin activation. However, its role in activation and functions of different β integrins in human vascular cells is unknown. In this study, using flow cytometry, we demonstrate that filamin inhibits β1 and αIIbβ3 integrin activation, and migfilin can overcome its inhibitory effect. Migfilin protein is widely expressed in different adherent and circulating blood cells and can regulate integrin activation in naturally-occurring vascular cells, endothelial cells and neutrophils. Migfilin can activate β1, β2 and β3 integrins and promote integrin mediated responses while migfilin depletion impairs the spreading and migration of endothelial cells. Thus, filamin can act broadly as an inhibitor and migfilin is a promoter of integrin activation.
Collapse
Affiliation(s)
- Mitali Das
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Sujay Subbayya Ithychanda
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Jun Qin
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Edward F. Plow
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
- * E-mail:
| |
Collapse
|
43
|
Wen J, Fu J, Zhang W, Guo M. Genetic and epigenetic changes in lung carcinoma and their clinical implications. Mod Pathol 2011; 24:932-43. [PMID: 21423157 DOI: 10.1038/modpathol.2011.46] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Lung cancer is the leading cause of cancer deaths worldwide. Recent advance in targeted therapy for lung cancer patients with epidermal growth factor receptor (EGFR) mutations has demonstrated a promising development toward personalized therapy for lung cancer patients. The development of lung cancer is a complex process, involving a series of genetic and epigenetic changes. Tobacco smoke is the predominant etiologic risk factor for lung cancer. However, some lung cancers, especially adenocarcinomas, arise in patients who have never smoked, suggesting the importance of host genetic/epigenetic susceptibility in the occurrence and development of lung cancer. Understanding of these genetic and epigenetic changes will further aid in the biomarker-driven personalized therapy for lung cancer patients. In this review, we summarize the genetic and epigenetic alterations observed in lung cancers, including chromosomal loss of heterozygosity, tumor-suppressor gene mutation, gene methylation, histone modification, and microRNA expression changes. Clinical and preclinical studies have implied specific genetic/epigenetic changes for clinical application in lung cancer patients. However, more efforts are required in validation of the identified molecular markers in lung cancer patients for early detections, assessment for treatment response, and survival predictions.
Collapse
Affiliation(s)
- Jing Wen
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | | | | | | |
Collapse
|
44
|
Foxler DE, James V, Shelton SJ, Vallim TQDA, Shaw PE, Sharp TV. PU.1 is a major transcriptional activator of the tumour suppressor gene LIMD1. FEBS Lett 2011; 585:1089-96. [PMID: 21402070 PMCID: PMC3078326 DOI: 10.1016/j.febslet.2011.03.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2010] [Revised: 02/24/2011] [Accepted: 03/07/2011] [Indexed: 12/30/2022]
Abstract
LIMD1 is a tumour suppressor gene (TSG) down regulated in ∼80% of lung cancers with loss also demonstrated in breast and head and neck squamous cell carcinomas. LIMD1 is also a candidate TSG in childhood acute lymphoblastic leukaemia. Mechanistically, LIMD1 interacts with pRB, repressing E2F-driven transcription as well as being a critical component of microRNA-mediated gene silencing. In this study we show a CpG island within the LIMD1 promoter contains a conserved binding motif for the transcription factor PU.1. Mutation of the PU.1 consensus reduced promoter driven transcription by 90%. ChIP and EMSA analysis demonstrated that PU.1 specifically binds to the LIMD1 promoter. siRNA depletion of PU.1 significantly reduced endogenous LIMD1 expression, demonstrating that PU.1 is a major transcriptional activator of LIMD1.
Collapse
Affiliation(s)
- Daniel E. Foxler
- School of Biomedical Sciences, University of Nottingham Medical School, Nottingham NG7 1UH, UK
| | - Victoria James
- School of Biomedical Sciences, University of Nottingham Medical School, Nottingham NG7 1UH, UK
| | - Samuel J. Shelton
- School of Biomedical Sciences, University of Nottingham Medical School, Nottingham NG7 1UH, UK
| | - Thomas Q. de A. Vallim
- Department of Biological Chemistry, University of California Los Angeles, David Geffen School of Medicine, Los Angeles, CA 90095, United States
- Department of Cardiology, University of California Los Angeles, David Geffen School of Medicine, Los Angeles, CA 90095, United States
| | - Peter E. Shaw
- School of Biomedical Sciences, University of Nottingham Medical School, Nottingham NG7 1UH, UK
| | - Tyson V. Sharp
- School of Biomedical Sciences, University of Nottingham Medical School, Nottingham NG7 1UH, UK
| |
Collapse
|
45
|
Moik DV, Janbandhu VC, Fässler R. Loss of migfilin expression has no overt consequences on murine development and homeostasis. J Cell Sci 2011; 124:414-21. [PMID: 21224394 DOI: 10.1242/jcs.075960] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Migfilin is a LIM-domain-containing protein of the zyxin family of adaptor proteins and is found at cell-matrix and cell-cell adhesion sites and in the nucleus. In vitro studies have suggested that migfilin promotes β1 integrin activity, regulates cell spreading and migration and induces cardiomyocyte differentiation. To test directly the function of migfilin in vivo, we generated a migfilin-null mouse strain. Here, we report that loss of migfilin expression permits normal development and normal postnatal aging. Fibroblasts and keratinocytes from migfilin-null mice display normal spreading and adhesion, and normal integrin expression and activation. The migration velocity and directionality of migfilin-null embryonic fibroblasts were normal, whereas the velocity of migfilin-null keratinocytes in wound scratch assays was slightly but significantly reduced. Our findings indicate that the roles of migfilin are functionally redundant during mouse development and tissue homeostasis.
Collapse
Affiliation(s)
- Daniel V Moik
- Max-Planck-Institute of Biochemistry, Department of Molecular Medicine, 82152 Martinsried, Germany
| | | | | |
Collapse
|
46
|
Abstract
While no real improvement in the long term survival has been obtained in lung cancer, during this decade a significant improvement in cancer control has been obtained by biology driven targeted therapy as with anti EGFR tyrosine kinase. Two phases can be described in the knowledge of lung cancer biology: a first phase open in the 1980s describing the main molecular anomalies and impaired cell control mechanisms, and a second phase starting in the 2004-2005 giving rise to the therapeutic applications of this knowledge. A new molecular classification of lung cancer, particularly adenocarcinomas will soon be proposed for therapeutic application.
Collapse
|
47
|
Hou D, Guan Y, Liu J, Xiao Z, Ouyang Y, Yu Y, Chen Z. Cloning and characterization of the NPCEDRG gene promoter. Mol Cell Biochem 2010; 346:1-10. [PMID: 20821255 DOI: 10.1007/s11010-010-0584-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2010] [Accepted: 08/27/2010] [Indexed: 11/25/2022]
Abstract
NPCEDRG is a novel tumor suppressive gene that localizes to 3p21.3, a chromosomal region frequently associated with loss of heterozygosity (LOH) in a number of malignancies including nasopharyngeal carcinoma (NPC). Its transcriptional down-expression has been shown in the cell lines and primary tumor tissues of NPC. Reintroduction of NPCEDRG into CNE2, a cell line derived from NPC, was effective to induce cell differentiation, control cell growth, and regulate the cell cycle. Little is known about the transcriptional mechanisms controlling NPCEDRG gene expression. In this article, we describe the NPCEDRG gene structure and the transcriptional expression of NPCEDRG; we found that NPCEDRG was expressed weakly in most of NPC cell lines. Using 5' rapid amplification of complementary DNA ends (5'-RACEs), we found that the NPCEDRG gene has several transcription start sites (TSSs) due to the existence of alternatively spliced variants, and the specific TSS of NPCEDRG was located -25 nucleotides upstream of the translation start site. We amend that Human NPCEDRG CDS containing 516 bp but not the 510 bp reported previously. To characterize the NPCEDRG promoter, transient luciferase and/or EGFP reporter assay were carried out with the constructs including various lengths of the 5' flanking region of the NPCEDRG gene. The results demonstrated that the basal promoter is located at the region from -215 to -8 nucleotides, and the optimal promoter is located at the region from -625 to -8 nucleotides upstream of the translation start site. In silico analysis suggested that the promoter region contained potential binding sites for SP1, c-Myb, AREB6, Nkx2-5, and so on. These results provide important clues to elucidate the regulation of NPCEDRG gene expression and function. Further studies are apparently required for the identification of the transcription factors, essential for NPCEDRG expression, which would lead to better understanding of the molecular mechanism of NPCEDRG expression in nasopharyngeal epithelial cells.
Collapse
Affiliation(s)
- Defu Hou
- Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, 87# Xiangya Road, Changsha, 410008, Hunan, China
| | | | | | | | | | | | | |
Collapse
|
48
|
James V, Zhang Y, Foxler DE, de Moor CH, Kong YW, Webb TM, Self TJ, Feng Y, Lagos D, Chu CY, Rana TM, Morley SJ, Longmore GD, Bushell M, Sharp TV. LIM-domain proteins, LIMD1, Ajuba, and WTIP are required for microRNA-mediated gene silencing. Proc Natl Acad Sci U S A 2010; 107:12499-504. [PMID: 20616046 PMCID: PMC2906597 DOI: 10.1073/pnas.0914987107] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
In recent years there have been major advances with respect to the identification of the protein components and mechanisms of microRNA (miRNA) mediated silencing. However, the complete and precise repertoire of components and mechanism(s) of action remain to be fully elucidated. Herein we reveal the identification of a family of three LIM domain-containing proteins, LIMD1, Ajuba and WTIP (Ajuba LIM proteins) as novel mammalian processing body (P-body) components, which highlight a novel mechanism of miRNA-mediated gene silencing. Furthermore, we reveal that LIMD1, Ajuba, and WTIP bind to Ago1/2, RCK, Dcp2, and eIF4E in vivo, that they are required for miRNA-mediated, but not siRNA-mediated gene silencing and that all three proteins bind to the mRNA 5' m(7)GTP cap-protein complex. Mechanistically, we propose the Ajuba LIM proteins interact with the m(7)GTP cap structure via a specific interaction with eIF4E that prevents 4EBP1 and eIF4G interaction. In addition, these LIM-domain proteins facilitate miRNA-mediated gene silencing by acting as an essential molecular link between the translationally inhibited eIF4E-m(7)GTP-5(')cap and Ago1/2 within the miRISC complex attached to the 3'-UTR of mRNA, creating an inhibitory closed-loop complex.
Collapse
Affiliation(s)
- Victoria James
- School of Biomedical Sciences, University of Nottingham Medical School, Queen’s Medical Centre, Nottingham NG7 2UH, United Kingdom
| | - Yining Zhang
- School of Biomedical Sciences, University of Nottingham Medical School, Queen’s Medical Centre, Nottingham NG7 2UH, United Kingdom
| | - Daniel E. Foxler
- School of Biomedical Sciences, University of Nottingham Medical School, Queen’s Medical Centre, Nottingham NG7 2UH, United Kingdom
| | - Cornelia H. de Moor
- School of Pharmacy, University of Nottingham, University Park, Nottingham NG7 2RD, United Kingdom
| | - Yi Wen Kong
- School of Pharmacy, University of Nottingham, University Park, Nottingham NG7 2RD, United Kingdom
| | - Thomas M. Webb
- School of Biomedical Sciences, University of Nottingham Medical School, Queen’s Medical Centre, Nottingham NG7 2UH, United Kingdom
| | - Tim J. Self
- Institute of Cell Signalling, School of Biomedical Sciences, University of Nottingham Medical School, Queen’s Medical Centre, Nottingham NG7 2UH, United Kingdom
| | - Yungfeng Feng
- Washington University School of Medicine, Campus Box 8086, Room 770 McDonnell Sciences Building, 660 South Euclid Avenue, St. Louis, MO 63110
| | - Dimitrios Lagos
- Cancer Research UK Viral Oncology Group, University College London Cancer Institute, Paul O’Gorman Building, Huntley Street, University College London, London WC1E 6BT, United Kingdom
| | - Chia-Ying Chu
- Institute of Zoology, School of Life Sciences, National Taiwan University, Taipei 10617, Taiwan
| | - Tariq M. Rana
- Program for RNA Biology, Sandford–Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037; and
| | - Simon J. Morley
- Department of Biochemistry, School of Life Sciences, University of Sussex, Brighton BN1 9QG, United Kingdom
| | - Gregory D. Longmore
- Washington University School of Medicine, Campus Box 8086, Room 770 McDonnell Sciences Building, 660 South Euclid Avenue, St. Louis, MO 63110
| | - Martin Bushell
- School of Pharmacy, University of Nottingham, University Park, Nottingham NG7 2RD, United Kingdom
| | - Tyson V. Sharp
- School of Biomedical Sciences, University of Nottingham Medical School, Queen’s Medical Centre, Nottingham NG7 2UH, United Kingdom
| |
Collapse
|
49
|
Cheng IKC, Ching AKK, Chan TC, Chan AWH, Wong CK, Choy KW, Kwan M, Lai PBS, Wong N. Reduced CRYL1 expression in hepatocellular carcinoma confers cell growth advantages and correlates with adverse patient prognosis. J Pathol 2010; 220:348-60. [PMID: 19927314 DOI: 10.1002/path.2644] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Homozygous deletion screening has been widely utilized to define tumour suppressor genes (TSGs) in cancers. Although these biallelic deletions are infrequent, their identification has facilitated the discovery of many important TSGs. We have systematically examined the genome of hepatocellular carcinoma (HCC), a highly malignant tumour that is rapidly fatal, for the presence of homozygous deletions. Array-CGH analysis on early passage of HCC cultures and cell lines led us to identify six homozygous deleted (HD) regions. A high concordance between array-CGH and expression of HD genes was demonstrated, where crystallin Lambda1 (CRYL1; located on chromosome 13q12.11) displayed the most frequent down-regulation. We found that reduced mRNA expression of CRYL1 was common in HCC tumours when compared with their adjacent non-tumoural liver (p = 0.0097). Significant associations could also be drawn between repressed CRYL1 and advanced tumour staging, increased tumour size, and shorter disease-free survival of patients (p < 0.037). Moreover, homozygous deletions on CRYL1 could be detected in 36% of HCC cases, where recurrent HDs were identified on exons 1, 5, and 8. Examination of other causal events suggested histone deacetylation and promoter hypermethylation to be likely inactivating mechanisms as well. Re-expression of CRYL1 in the SK-Hep1 cell line, where biallelic loss of CRYL1 was found, induced profound inhibition of cellular proliferation and cell growth (p < 0.0015). By Annexin V staining, CRYL1 restoration readily increased pro-apoptotic cells with an induction of PARP cleavage. Flow cytometry further revealed that CRYL1 could prolong the G(2)-M phase, possibly through interruption of the Cdc2/cyclin B pathway. Given that regional chromosome 13q12-q14 loss is a causal genomic event in HCC tumourigenesis, our finding may have implications for identifying a novel TSG CRYL1 within this important locus.
Collapse
Affiliation(s)
- Ibis K-C Cheng
- Department of Anatomical and Cellular Pathology at the Li Ka-Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Ghosh S, Ghosh A, Maiti GP, Mukherjee N, Dutta S, Roy A, Roychoudhury S, Panda CK. LIMD1 is more frequently altered than RB1 in head and neck squamous cell carcinoma: clinical and prognostic implications. Mol Cancer 2010; 9:58. [PMID: 20226061 PMCID: PMC2848626 DOI: 10.1186/1476-4598-9-58] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2009] [Accepted: 03/12/2010] [Indexed: 12/04/2022] Open
Abstract
Introduction To understand the role of two interacting proteins LIMD1 and pRB in development of head and neck squamous cell carcinoma (HNSCC), alterations of these genes were analyzed in 25 dysplastic head and neck lesions, 58 primary HNSCC samples and two HNSCC cell lines. Methods Deletions of LIMD1 and RB1 were analyzed along with mutation and promoter methylation analysis of LIMD1. The genotyping of LIMD1 linked microsatellite marker, hmlimD1, was done to find out any risk allele. The mRNA expression of LIMD1 and RB1 were analyzed by Q-PCR. Immunohistochemical analysis of RB1 was performed. Alterations of these genes were correlated with different clinicopathological parameters. Results High frequency [94% (78/83)] of LIMD1 alterations was observed in the samples studied. Compare to frequent deletion and methylation, mutation of LIMD1 was increased during tumor progression (P = 0.007). Six novel mutations in exon1 and one novel intron4/exon5 splice-junction mutation were detected in LIMD1 along with a susceptible hmlimD1 (CA)20 allele. Some of these mutations [42% (14/33)] produced non-functional proteins. RB1 deletion was infrequent (27%). Highly reduced mRNA expression of LIMD1 (25.1 ± 19.04) was seen than RB1 (3.8 ± 8.09), concordant to their molecular alterations. The pRB expression supported this data. Tumors with LIMD1 alterations in tobacco addicted patients without HPV infection showed poor prognosis. Co-alterations of these genes led the worse patients' outcome. Conclusions Our study suggests LIMD1 inactivation as primary event than inactivation of RB1 in HNSCC development.
Collapse
Affiliation(s)
- Susmita Ghosh
- Department of Oncogene Regulation, Chittaranjan National Cancer Institute, 37, SP Mukherjee Road, Kolkata 700026, India
| | | | | | | | | | | | | | | |
Collapse
|