1
|
Chen Q, Chen Y, Zheng Q. The RNA-binding protein LSM family regulating reproductive development via different RNA metabolism. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167808. [PMID: 40139411 DOI: 10.1016/j.bbadis.2025.167808] [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: 11/25/2024] [Revised: 03/18/2025] [Accepted: 03/18/2025] [Indexed: 03/29/2025]
Abstract
The LSM (Like-Sm) protein family, characterized by highly conserved LSM domains, is integral to ribonucleic acid (RNA) metabolism. Ubiquitously present in both eukaryotes and select prokaryotes, these proteins bind to RNA molecules with high specificity through their LSM domains. They can also form ring-shaped complexes with other proteins, thereby facilitating various fundamental cellular processes such as mRNA degradation, splicing, and ribosome biogenesis. LSM proteins play crucial roles in gametogenesis, early embryonic development, sex determination, gonadal maturation, and reproductive system formation. In pathological conditions, the absence of LSM14B leads to arrest of oocytes at mid-meiosis, downregulation of LSM4 expression is associated with abnormal spermatogenesis, and aberrant expression of LSM1 protein is linked to the occurrence and progression of breast cancer. This review focuses on the recent advances in the functional research of LSM proteins in reproduction.
Collapse
Affiliation(s)
- Qin Chen
- Prenatal Diagnosis Center, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shennan Road, Shenzhen 518033, PR China
| | - Ying Chen
- Prenatal Diagnosis Center, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shennan Road, Shenzhen 518033, PR China
| | - Qingliang Zheng
- Prenatal Diagnosis Center, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shennan Road, Shenzhen 518033, PR China.
| |
Collapse
|
2
|
Yang C, Ge C, Zhang W, Xu J. LSM2 drives glioma progression through alternative splicing dysregulation: a multi-omics approach to identify a potential therapeutic target. Front Oncol 2025; 15:1521608. [PMID: 40365337 PMCID: PMC12069061 DOI: 10.3389/fonc.2025.1521608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Accepted: 04/07/2025] [Indexed: 05/15/2025] Open
Abstract
Background Glioma, particularly glioblastoma (GBM), remains a highly aggressive and challenging tumour, characterised by poor prognosis and limited therapeutic options. LSM2, an RNA-binding protein, has been implicated in tumour progression, yet its role in glioma remains underexplored. This study aims to investigate the expression, prognostic significance, and molecular mechanisms of LSM2 in glioma, focusing on its impact on RNA splicing regulation. Methods Clinical and transcriptomic data from 163 GBM and 518 lower-grade glioma (LGG) cases from The Cancer Genome Atlas (TCGA) were analysed to assess LSM2 expression and its prognostic value. RNA sequencing was performed on LSM2 knockdown in T98G glioblastoma cells to identify differentially expressed genes (DEGs) and alternative splicing events (ASEs). Bioinformatics tools were employed to perform functional enrichment analyses and construct protein-protein interaction (PPI) networks. Results LSM2 expression was significantly elevated in gliomas, particularly in GBM and in tumours with 1p/19q non-deletion or IDH1 mutation (p < 0.001). High LSM2 expression was correlated with shorter overall survival (HR = 1.7, p = 0.01). Knockdown of LSM2 in T98G cells identified 728 upregulated and 1,720 downregulated genes, alongside 1,949 splicing alterations, which primarily affected pathways related to RNA metabolism, DNA damage response, and cell cycle regulation. Key hub genes such as TLN1, FN1, and IRF7 were associated with glioma progression and poor prognosis. Conclusion Our findings demonstrate that LSM2 plays a critical role in glioma progression through the regulation of RNA splicing dynamics. Elevated LSM2 expression serves as a prognostic biomarker and offers promising potential as a therapeutic target in glioma.
Collapse
Affiliation(s)
| | | | | | - Jingxuan Xu
- Department of Neurosurgery, The Second Affiliated Hospital of Xinjiang Medical
University, Urumqi, China
| |
Collapse
|
3
|
Bronson K, Banik J, Lim J, Reddick MM, Hardy L, Childs GV, MacNicol MC, MacNicol AM. Musashi-dependent mRNA translational activation is mediated through association with the Scd6/Like-sm family member, LSM14B. Sci Rep 2025; 15:12363. [PMID: 40211036 PMCID: PMC11986153 DOI: 10.1038/s41598-025-97188-9] [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: 11/27/2024] [Accepted: 04/01/2025] [Indexed: 04/12/2025] Open
Abstract
The Musashi family of sequence-specific RNA binding proteins (Musashi1 and Musashi2) serve a critical role in mediating both physiological and pathological stem cell function in many tissue types by repressing the translation of target mRNAs that encode proteins that promote cell cycle inhibition and cell differentiation. In addition to repression of target mRNAs, we have also identified a role for Musashi proteins in activating the translation of target mRNAs in a context-dependent manner. However, the molecular mechanisms by which Musashi controls target mRNA translational activation have not been fully elucidated. Since Musashi lacks inherent enzymatic activity, its ability to modulate target mRNA translation likely involves recruitment of ancillary proteins to the target mRNA. We have previously identified a number of proteins that specifically associate with Musashi during Xenopus laevis oocyte maturation at a time when Musashi target mRNAs are translationally activated. Here, we demonstrate that one of these proteins, the Scd6/Like-sm family member LSM14B, is a mediator of the Musashi1-dependent mRNA translational activation that is required for oocyte maturation. Unlike previously characterized proteins which interact with the C-terminal domain of Musashi, LSM14B instead associates with the N-terminal RNA recognition motifs. Additionally, we demonstrate that the mammalian Prop1 mRNA, which encodes a key regulator of pituitary development, is translationally activated by Musashi1 in a LSM14B-dependent manner. Our studies support an evolutionarily conserved role for LSM14B in facilitating the ability of Musashi1 to promote target mRNA translation.
Collapse
Affiliation(s)
- Katherine Bronson
- Department of Neuroscience, University of Arkansas for Medical Sciences, 4301 W Markham, Slot 814, Little Rock, AR, 72205, USA
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, 4301 W Markham, Slot 814, Little Rock, AR, 72205, USA
| | - Jewel Banik
- Department of Neuroscience, University of Arkansas for Medical Sciences, 4301 W Markham, Slot 814, Little Rock, AR, 72205, USA
| | - Juchan Lim
- Department of Neuroscience, University of Arkansas for Medical Sciences, 4301 W Markham, Slot 814, Little Rock, AR, 72205, USA
| | - Milla M Reddick
- Department of Neuroscience, University of Arkansas for Medical Sciences, 4301 W Markham, Slot 814, Little Rock, AR, 72205, USA
| | - Linda Hardy
- Department of Neuroscience, University of Arkansas for Medical Sciences, 4301 W Markham, Slot 814, Little Rock, AR, 72205, USA
| | - Gwen V Childs
- Department of Neuroscience, University of Arkansas for Medical Sciences, 4301 W Markham, Slot 814, Little Rock, AR, 72205, USA
| | - Melanie C MacNicol
- Department of Neuroscience, University of Arkansas for Medical Sciences, 4301 W Markham, Slot 814, Little Rock, AR, 72205, USA
- Department of Biomedical Informatics, University of Arkansas for Medical Sciences, 4301 W Markham, Slot 814, Little Rock, AR, 72205, USA
| | - Angus M MacNicol
- Department of Neuroscience, University of Arkansas for Medical Sciences, 4301 W Markham, Slot 814, Little Rock, AR, 72205, USA.
| |
Collapse
|
4
|
Lin T, Deng L, Liu S, Chen Y, Tang Y, Zhou F, Gong G, He Y. Identification of RNA processing factor LSM5 as a new adverse biomarker in nasopharyngeal carcinoma. Sci Rep 2025; 15:9901. [PMID: 40121338 PMCID: PMC11929837 DOI: 10.1038/s41598-025-94968-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 03/18/2025] [Indexed: 03/25/2025] Open
Abstract
Dysregulated RNA processing is crucial in nasopharyngeal carcinoma (NPC) progression. Our research aimed to evaluate the prognostic values of RNA-processing genes (RPGs) in NPC through bioinformatic analysis of the GSE12452 and GSE102349 datasets, identifying differentially expressed RNA-processing genes (DE-RPGs). A prognostic model was developed using univariate and multivariate Cox analysis, with effectiveness assessed by ROC analysis. The correlation between risk scores, immune characteristics, and chemotherapy sensitivity was also analyzed. Model gene expression was validated by RT-qPCR, Western Blot, and immunohistochemistry, alongside functional assays. Bioinformatics indicated that RNA binding motif protein 20 (RBM20) and LSM5 are prognostic RPGs, with the ROC curve confirming their predictive ability for survival. Significant differences in drug sensitivity were noted between high- and low-risk groups. Experimental validation showed LSM5 is overexpressed in NPC tissues, correlating with poorer prognosis, and its down-regulation inhibits cell proliferation and migration. Thus, LSM5 is identified as a new adverse biomarker in NPC, with implications for targeted therapy and prognosis improvement.
Collapse
Affiliation(s)
- Ting Lin
- Hunan Provincial Engineering Research Center for Prevention and Treatment of Ophthalmology and Otolaryngology Diseases With Chinese Medicine and Visual Function Protection, Hunan University of Chinese Medicine, No. 300, Xuba Road, Hanpu Science and Education Park, Changsha, 410208, Hunan, People's Republic of China
- Hunan Provincial Key Laboratory for Prevention and Treatment of Ophthalmology and Otolaryngology Diseases With Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, People's Republic of China
| | - Linyu Deng
- College of Acupuncture, Massage and Rehabilitation of Hunan University of Chinese Medicine, Changsha, 410208, Hunan, People's Republic of China
| | - Siyu Liu
- Graduate School of Hunan University of Chinese Medicine, Changsha, 410208, Hunan, People's Republic of China
| | - Yan Chen
- Graduate School of Hunan University of Chinese Medicine, Changsha, 410208, Hunan, People's Republic of China
| | - Yinggang Tang
- Graduate School of Hunan University of Chinese Medicine, Changsha, 410208, Hunan, People's Republic of China
| | - Fangliang Zhou
- Hunan Provincial Engineering Research Center for Prevention and Treatment of Ophthalmology and Otolaryngology Diseases With Chinese Medicine and Visual Function Protection, Hunan University of Chinese Medicine, No. 300, Xuba Road, Hanpu Science and Education Park, Changsha, 410208, Hunan, People's Republic of China
- Hunan Provincial Key Laboratory for Prevention and Treatment of Ophthalmology and Otolaryngology Diseases With Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, People's Republic of China
| | - Guanghui Gong
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China.
- Department of Pathology, School of Basic Medical Science, Central South University, Changsha, 410013, Hunan, People's Republic of China.
| | - Yingchun He
- Hunan Provincial Engineering Research Center for Prevention and Treatment of Ophthalmology and Otolaryngology Diseases With Chinese Medicine and Visual Function Protection, Hunan University of Chinese Medicine, No. 300, Xuba Road, Hanpu Science and Education Park, Changsha, 410208, Hunan, People's Republic of China.
- Hunan Provincial Key Laboratory for Prevention and Treatment of Ophthalmology and Otolaryngology Diseases With Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, People's Republic of China.
| |
Collapse
|
5
|
Wang Y, Li C, Deng Q, Hu X, Zhao J, Yang H, Wen G. EDC3 protein of P-body suppresses PRRSV proliferation and functions by upregulating MyD88. Vet Microbiol 2025; 302:110414. [PMID: 39938412 DOI: 10.1016/j.vetmic.2025.110414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 01/17/2025] [Accepted: 01/27/2025] [Indexed: 02/14/2025]
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) causes significant economic losses to the swine industry worldwide. Despite numerous studies on this virus, the molecular mechanisms underlying PRRSV infection remain unclear. Enhancer of mRNA decapping 3 (EDC3) is a crucial scaffold protein for P-body assembly that stimulates mRNA decapping by alleviating self-inhibition of the DCP1/2 decapping complex. In this study, we investigated the impact of EDC3 on PRRSV proliferation, as well as its influence on three key adaptor proteins of the innate immune system: Toll-like receptor interleukin-1 receptor domain-containing adaptor inducing interferon-β (TRIF), mitochondrial antiviral signaling protein (MAVs), and myeloid differentiation factor 88 (MyD88). Western blotting and quantitative real-time PCR assays showed that EDC3 overexpression in both Marc-145 and PAM-CD163 cells significantly inhibited transcription of the PRRSV N gene and expression of the N protein. Conversely, knockdown of EDC3 expression in these cells significantly promoted the transcription of the PRRSV N gene and expression of the N protein. Furthermore, our results demonstrated that EDC3 overexpression significantly enhanced the expression of MyD88, whereas the inhibition of EDC3 expression led to a reduction in this process. In contrast, EDC3 did not effectively promote the increase in protein levels of TRIF and MAVs. In conclusion, the P-body EDC3 protein suppresses PRRSV proliferation and function by upregulating MyD88. This work is the first to report the mechanism of action of EDC3 against PRRSV, providing ideas for studying antiviral innate immunity.
Collapse
Affiliation(s)
- Yiran Wang
- College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Changhong Li
- College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Qiaomu Deng
- College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Xuan Hu
- College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Jing Zhao
- College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Hongxing Yang
- College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Guilan Wen
- College of Animal Science, Guizhou University, Guiyang 550025, China.
| |
Collapse
|
6
|
Hao W, Zhao D, Meng Y, Yang M, Ma M, Hu J, Liu J, Qin X. Screening of Cancer-Specific Biomarkers for Hepatitis B-Related Hepatocellular Carcinoma Based on a Proteome Microarray. Mol Cell Proteomics 2024; 23:100872. [PMID: 39489219 DOI: 10.1016/j.mcpro.2024.100872] [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: 06/11/2024] [Revised: 10/14/2024] [Accepted: 10/19/2024] [Indexed: 11/05/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is associated with one of the highest mortality rates among cancers, rendering its early diagnosis clinically invaluable. Serum biomarkers, specifically alpha-fetoprotein (AFP), represent the most promising and widely used diagnostic biomarkers for HCC. However, its detection rate is low in the early stages of HCC progression, and distinguishing specific false positives for other liver-related diseases, such as cirrhosis and acute hepatitis, remains challenging. Therefore, this study was conducted to identify biomarkers for hepatitis B (HBV)-related liver diseases by screening differentially expressed autoantibodies against tumor-associated antigens (TAAbs). We designed a large-scale multistage investigation, encompassing initial screening, HCC-focused, and ELISA validation cohorts to identify potential TAAbs in HBV-related liver diseases, spanning from healthy control (HC) individuals to patients with chronic hepatitis B (CHB), hepatitis B-related cirrhosis (HBC), and HCC, using protein microarray technology. The differential biological characteristics of TAAbs were analyzed using bioinformatics analysis. Validation of tumor-specific biomarkers for HCC was performed using ELISA. In the screening cohort, 547 candidate TAAbs were identified in the HCC group compared to those in the HC group. In the HCC-focused cohort, 64, 61, and 65 candidate TAAbs were identified in the CHB, HBC, and HCC groups, respectively, compared to those in the HC group. Thirty-four proteins exhibited continuously elevated expression from HCs to patients with CHB, HBC, and HCC. Among these, nine were identified as cancer-specific proteins. In the validation cohort, UBE2Z, CNOT3, and EID3 were correlated with liver function indicators in patients with hepatitis B-related HCC. Overall, UBE2Z, CNOT3, and EID3 emerged as cancer-specific biomarkers for HBV-related liver disease, providing a scientific basis for clinical application.
Collapse
Affiliation(s)
- Wudi Hao
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, China; Liaoning Clinical Research Center for Laboratory Medicine, Shenyang, China
| | - Danyang Zhao
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, China; Liaoning Clinical Research Center for Laboratory Medicine, Shenyang, China
| | - Yuan Meng
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, China; Liaoning Clinical Research Center for Laboratory Medicine, Shenyang, China
| | - Mei Yang
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, China; Liaoning Clinical Research Center for Laboratory Medicine, Shenyang, China
| | - Meichen Ma
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, China; Liaoning Clinical Research Center for Laboratory Medicine, Shenyang, China
| | - Jingwen Hu
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, China; Liaoning Clinical Research Center for Laboratory Medicine, Shenyang, China
| | - Jianhua Liu
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, China; Liaoning Clinical Research Center for Laboratory Medicine, Shenyang, China.
| | - Xiaosong Qin
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, China; Liaoning Clinical Research Center for Laboratory Medicine, Shenyang, China.
| |
Collapse
|
7
|
Wang C, He Y, Fang X, Zhang D, Huang J, Zhao S, Li L, Li G. METTL1-modulated LSM14A facilitates proliferation and migration in glioblastoma via the stabilization of DDX5. iScience 2024; 27:110225. [PMID: 39040050 PMCID: PMC11261005 DOI: 10.1016/j.isci.2024.110225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 02/06/2024] [Accepted: 06/06/2024] [Indexed: 07/24/2024] Open
Abstract
Glioblastoma (GBM) is characterized by aggressive growth, invasiveness, and poor prognosis. Elucidating the molecular mechanisms underlying GBM is crucial. This study explores the role of Sm-like protein 14 homolog A (LSM14A) in GBM. Bioinformatics and clinical tissue samples analysis demonstrated that overexpression of LSM14A in GBM correlates with poorer prognosis. CCK8, EdU, colony formation, and transwell assays revealed that LSM14A promotes proliferation, migration, and invasion in GBM in vitro. In vivo mouse xenograft models confirmed the results of the in vitro experiments. The mechanism of LSM14A modulating GBM cell proliferation was investigated using mass spectrometry, co-immunoprecipitation (coIP), protein half-life, and methylated RNA immunoprecipitation (MeRIP) analyses. The findings indicate that during the G1/S phase, LSM14A stabilizes DDX5 in the cytoplasm, regulating CDK4 and P21 levels. Furthermore, METTL1 modulates LSM14A expression via mRNA m7G methylation. Altogether, our work highlights the METTL1-LSM14A-DDX5 pathway as a potential therapeutic target in GBM.
Collapse
Affiliation(s)
- Changyu Wang
- Department of Neurosurgery, The First Hospital of China Medical University, NO. 155 Nanjing North Street, Heping District, Shenyang 110002, China
| | - Yan He
- Department of Laboratory Animal Science, China Medical University, 110122, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, P.R. China
| | - Xiang Fang
- Department of Neurosurgery, Central Hospital Affiliated to Shandong First Medical University, No. 105, Jiefang Road, Jinan, Shandong, People’s Republic of China
| | - Danyang Zhang
- Department of Immunology, College of Basic Medical Sciences of China Medical University, 110122, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, P.R. China
| | - Jinhai Huang
- Department of Neurosurgery, The First Hospital of China Medical University, NO. 155 Nanjing North Street, Heping District, Shenyang 110002, China
| | - Shuxin Zhao
- The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Lun Li
- Department of Neurosurgery, Anshan Hospital of the First Hospital of China Medical University, Anshan, China
| | - Guangyu Li
- Department of Neurosurgery, The First Hospital of China Medical University, NO. 155 Nanjing North Street, Heping District, Shenyang 110002, China
| |
Collapse
|
8
|
Mo CJ, Deng XY, Ma RL, Zhu K, Shi L, Li K. Sm-like 5 knockdown inhibits proliferation and promotes apoptosis of colon cancer cells by upregulating p53, CDKN1A and TNFRSF10B. World J Gastrointest Oncol 2024; 16:2704-2714. [DOI: 10.4251/wjgo.v16.i6.2704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 02/09/2024] [Accepted: 04/15/2024] [Indexed: 06/13/2024] Open
Abstract
BACKGROUND The role of Sm-like 5 (LSM5) in colon cancer has not been determined. In this study, we investigated the role of LSM5 in progression of colon cancer and the potential underlying mechanism involved.
AIM To determine the role of LSM5 in the progression of colon cancer and the potential underlying mechanism involved.
METHODS The Gene Expression Profiling Interactive Analysis database and the Human Protein Atlas website were used for LSM5 expression analysis and prognosis analysis. Real-time quantitative polymerase chain reaction and Western blotting were utilized to detect the expression of mRNAs and proteins. A lentivirus targeting LSM5 was constructed and transfected into colon cancer cells to silence LSM5 expression. Proliferation and apoptosis assays were also conducted to evaluate the growth of the colon cancer cells. Human GeneChip assay and bioinformatics analysis were performed to identify the potential underlying mechanism of LSM5 in colon cancer.
RESULTS LSM5 was highly expressed in tumor tissue and colon cancer cells. A high expression level of LSM5 was related to poor prognosis in patients with colon cancer. Knockdown of LSM5 suppressed proliferation and promoted apoptosis in colon cancer cells. Silencing of LSM5 also facilitates the expression of p53, cyclin-dependent kinase inhibitor 1A (CDKN1A) and tumor necrosis factor receptor superfamily 10B (TNFRSF10B). The inhibitory effect of LSM5 knockdown on the growth of colon cancer cells was associated with the upregulation of p53, CDKN1A and TNFRSF10B.
CONCLUSION LSM5 knockdown inhibited the proliferation and facilitated the apoptosis of colon cancer cells by upregulating p53, CDKN1A and TNFRSF10B.
Collapse
Affiliation(s)
- Cai-Jing Mo
- Department of Surgical Oncology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
| | - Xiao-Yuan Deng
- Department of Surgical Oncology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
| | - Ru-Lan Ma
- Department of Surgical Oncology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
| | - Kun Zhu
- Department of Surgical Oncology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
| | - Lei Shi
- Department of Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
| | - Kang Li
- Department of Surgical Oncology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
| |
Collapse
|
9
|
Mo CJ, Deng XY, Ma RL, Zhu K, Shi L, Li K. Sm-like 5 knockdown inhibits proliferation and promotes apoptosis of colon cancer cells by upregulating p53, CDKN1A and TNFRSF10B. World J Gastrointest Oncol 2024; 16:2716-2726. [PMID: 38994171 PMCID: PMC11236259 DOI: 10.4251/wjgo.v16.i6.2716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 03/18/2024] [Accepted: 04/15/2024] [Indexed: 06/14/2024] Open
Abstract
BACKGROUND The role of Sm-like 5 (LSM5) in colon cancer has not been determined. In this study, we investigated the role of LSM5 in progression of colon cancer and the potential underlying mechanism involved. AIM To determine the role of LSM5 in the progression of colon cancer and the potential underlying mechanism involved. METHODS The Gene Expression Profiling Interactive Analysis database and the Human Protein Atlas website were used for LSM5 expression analysis and prognosis analysis. Real-time quantitative polymerase chain reaction and Western blotting were utilized to detect the expression of mRNAs and proteins. A lentivirus targeting LSM5 was constructed and transfected into colon cancer cells to silence LSM5 expression. Proliferation and apoptosis assays were also conducted to evaluate the growth of the colon cancer cells. Human GeneChip assay and bioinformatics analysis were performed to identify the potential underlying mechanism of LSM5 in colon cancer. RESULTS LSM5 was highly expressed in tumor tissue and colon cancer cells. A high expression level of LSM5 was related to poor prognosis in patients with colon cancer. Knockdown of LSM5 suppressed proliferation and promoted apoptosis in colon cancer cells. Silencing of LSM5 also facilitates the expression of p53, cyclin-dependent kinase inhibitor 1A (CDKN1A) and tumor necrosis factor receptor superfamily 10B (TNFRSF10B). The inhibitory effect of LSM5 knockdown on the growth of colon cancer cells was associated with the upregulation of p53, CDKN1A and TNFRSF10B. CONCLUSION LSM5 knockdown inhibited the proliferation and facilitated the apoptosis of colon cancer cells by upregulating p53, CDKN1A and TNFRSF10B.
Collapse
Affiliation(s)
- Cai-Jing Mo
- Department of Surgical Oncology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
| | - Xiao-Yuan Deng
- Department of Surgical Oncology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
| | - Ru-Lan Ma
- Department of Surgical Oncology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
| | - Kun Zhu
- Department of Surgical Oncology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
| | - Lei Shi
- Department of Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
| | - Kang Li
- Department of Surgical Oncology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
| |
Collapse
|
10
|
Gao X, Ren X, Wang F, Ren X, Liu M, Cui G, Liu X. Immunotherapy and drug sensitivity predictive roles of a novel prognostic model in hepatocellular carcinoma. Sci Rep 2024; 14:9509. [PMID: 38664521 PMCID: PMC11045740 DOI: 10.1038/s41598-024-59877-9] [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: 02/20/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most significant causes of cancer-related deaths in the worldwide. Currently, predicting the survival of patients with HCC and developing treatment drugs still remain a significant challenge. In this study, we employed prognosis-related genes to develop and externally validate a predictive risk model. Furthermore, the correlation between signaling pathways, immune cell infiltration, immunotherapy response, drug sensitivity, and risk score was investigated using different algorithm platforms in HCC. Our results showed that 11 differentially expressed genes including UBE2C, PTTG1, TOP2A, SPP1, FCN3, SLC22A1, ADH4, CYP2C8, SLC10A1, F9, and FBP1 were identified as being related to prognosis, which were integrated to construct a prediction model. Our model could accurately predict patients' overall survival using both internal and external datasets. Moreover, a strong correlation was revealed between the signaling pathway, immune cell infiltration, immunotherapy response, and risk score. Importantly, a novel potential drug candidate for HCC treatment was discovered based on the risk score and also validated through ex vivo experiments. Our finds offer a novel perspective on prognosis prediction and drug exploration for cancer patients.
Collapse
Affiliation(s)
- Xiaoge Gao
- Cancer Institute, Xuzhou Medical University, Xuzhou, 221002, Jiangsu Province, People's Republic of China
| | - Xin Ren
- Cancer Institute, Xuzhou Medical University, Xuzhou, 221002, Jiangsu Province, People's Republic of China
- Department of Oncology, Jiangyin Clinical College, Xuzhou Medical University, Jiangyin, 214400, Jiangsu Province, People's Republic of China
| | - Feitong Wang
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, Jiangsu Province, People's Republic of China
| | - Xinxin Ren
- School of Information and Artificial Intelligence, Anhui Agricultural University, Hefei, 230036, People's Republic of China
| | - Mengchen Liu
- School of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519040, Guangdong Province, People's Republic of China
| | - Guozhen Cui
- School of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519040, Guangdong Province, People's Republic of China
| | - Xiangye Liu
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu Province, People's Republic of China.
- National Demonstration Center for Experimental Basic Medical Science Education (Xuzhou Medical University), Xuzhou, 221002, Jiangsu Province, People's Republic of China.
| |
Collapse
|
11
|
Jiang Y. Editorial: Multi-omics analysis of programmed cell death-mediated tumor microenvironment heterogeneity. Front Oncol 2024; 14:1265418. [PMID: 38595819 PMCID: PMC11002217 DOI: 10.3389/fonc.2024.1265418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Accepted: 02/15/2024] [Indexed: 04/11/2024] Open
Affiliation(s)
- Yanan Jiang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, China
| |
Collapse
|
12
|
Heßling LD, Troost-Kind B, Weiß M. NAADP-binding proteins - Linking NAADP signaling to cancer and immunity. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119531. [PMID: 37394011 DOI: 10.1016/j.bbamcr.2023.119531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 06/02/2023] [Accepted: 06/26/2023] [Indexed: 07/04/2023]
Abstract
NAADP is one of the most potent calcium mobilizing second messengers. Only recently, two NAADP-binding proteins have been identified: HN1L/JPT2 and LSM12. Further, ASPDH was suggested as a less selective binding partner. Apart from this newly uncovered link, little is known about the shared mechanisms between these proteins. The aim of this review is to assess potential functional connections between NAADP and its binding proteins. We here give a description of two major links. For one, HN1L/JPT2 and LSM12 both have potent oncogenic functions in several cancer types. Second, they are involved in similar cellular pathways in both cancer and immunity.
Collapse
Affiliation(s)
- Louisa D Heßling
- The Calcium Signaling Group, Dept. of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany.
| | - Berit Troost-Kind
- The Calcium Signaling Group, Dept. of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Mariella Weiß
- The Calcium Signaling Group, Dept. of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| |
Collapse
|
13
|
Zhou S, Van Bortle K. The Pol III transcriptome: Basic features, recurrent patterns, and emerging roles in cancer. WILEY INTERDISCIPLINARY REVIEWS. RNA 2023; 14:e1782. [PMID: 36754845 PMCID: PMC10498592 DOI: 10.1002/wrna.1782] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 01/13/2023] [Accepted: 01/18/2023] [Indexed: 02/10/2023]
Abstract
The RNA polymerase III (Pol III) transcriptome is universally comprised of short, highly structured noncoding RNA (ncRNA). Through RNA-protein interactions, the Pol III transcriptome actuates functional activities ranging from nuclear gene regulation (7SK), splicing (U6, U6atac), and RNA maturation and stability (RMRP, RPPH1, Y RNA), to cytoplasmic protein targeting (7SL) and translation (tRNA, 5S rRNA). In higher eukaryotes, the Pol III transcriptome has expanded to include additional, recently evolved ncRNA species that effectively broaden the footprint of Pol III transcription to additional cellular activities. Newly evolved ncRNAs function as riboregulators of autophagy (vault), immune signaling cascades (nc886), and translation (Alu, BC200, snaR). Notably, upregulation of Pol III transcription is frequently observed in cancer, and multiple ncRNA species are linked to both cancer progression and poor survival outcomes among cancer patients. In this review, we outline the basic features and functions of the Pol III transcriptome, and the evidence for dysregulation and dysfunction for each ncRNA in cancer. When taken together, recurrent patterns emerge, ranging from shared functional motifs that include molecular scaffolding and protein sequestration, overlapping protein interactions, and immunostimulatory activities, to the biogenesis of analogous small RNA fragments and noncanonical miRNAs, augmenting the function of the Pol III transcriptome and further broadening its role in cancer. This article is categorized under: RNA in Disease and Development > RNA in Disease RNA Processing > Processing of Small RNAs RNA Interactions with Proteins and Other Molecules > Protein-RNA Interactions: Functional Implications.
Collapse
Affiliation(s)
- Sihang Zhou
- Department of Cell and Developmental Biology, University of Illinois Urbana-Champaign, Urbana, Illinois, USA
| | - Kevin Van Bortle
- Department of Cell and Developmental Biology, University of Illinois Urbana-Champaign, Urbana, Illinois, USA
- Cancer Center at Illinois, University of Illinois Urbana-Champaign, Urbana, Illinois, USA
| |
Collapse
|
14
|
Qin P, Huang H, Wang J, Jiang T, Zeng N, Wang Q, He Y, Zhou Y. The mechanism of LSM2 in the progression of live hepatocellular carcinoma was analyzed based on bioinformatics. Med Oncol 2023; 40:276. [PMID: 37612479 DOI: 10.1007/s12032-023-02152-0] [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: 06/16/2023] [Accepted: 08/07/2023] [Indexed: 08/25/2023]
Abstract
Comprehensive analysis of the expression and probable function of LSM2 in Live hepatocellular carcinoma (LIHC), and validation via in vitro experiments. Integrated use of database resources to examine the differential expression, survival prognosis, clinicopathological characteristics, and functional enrichment of LSM2 in LIHC. The expression level of LSM2 in LIHC tissues and adjacent tissues was proven via immunohistochemical staining. The biological function of LSM2 in LIHC was detected by cell proliferation, cell cloning, cell scratch, cell migration, and invasion experiments in vitro. TIMER 2.0 and GEPIA indicated that LSM2 was highly expressed in cancers and was strongly associated with survival rates in LIHC, cholangiocarcinoma, breast cancer, and renal clear cell carcinoma. LSM2 was highly expressed in LIHC, which was closely associated to the clinicopathological characteristics of patients, and the overall survival rate and disease-free survival rate of patients with high expression of LSM2 were lower than those with low expression of LSM2. Functional enrichment results revealed that LSM2 was involved to ribosome formation, DNA replication, cell cycle, metabolic processes, JAK-STAT signaling pathways, and FoxO signaling pathways. Knockdown of LSM2 inhibited the proliferation, migration, and invasion of LIHC cells in vitro experiments. LSM2 was highly expressed in LIHC and was related to a poor prognosis. Knockdown of LSM2 could inhibit the proliferation, migration, and invasion of LIHC cells.
Collapse
Affiliation(s)
- Peifang Qin
- Institute of Pathogenic Biology, Guilin Medical University, Guilin, 541004, China
- Department of Clinical Laboratory, Guigang City People's Hospital, Guigang, Guangxi, 537100, China
- Guangxi Medical and Health Key Disciplines Infectious Diseases Key Disciplines, The Second Affiliated Hospital of Guilin Medical College, Guilin, 541004, China
| | - Haitao Huang
- Institute of Pathogenic Biology, Guilin Medical University, Guilin, 541004, China
| | - Jiahui Wang
- Institute of Pathogenic Biology, Guilin Medical University, Guilin, 541004, China
| | - Tingting Jiang
- Institute of Pathogenic Biology, Guilin Medical University, Guilin, 541004, China
| | - Nannan Zeng
- Department of Physiology, Guilin Medical University, Guilin, 541004, China
| | - Qi Wang
- Department of Physiology, Guilin Medical University, Guilin, 541004, China
| | - Yulin He
- Institute of Pathogenic Biology, Guilin Medical University, Guilin, 541004, China.
| | - Yali Zhou
- Institute of Pathogenic Biology, Guilin Medical University, Guilin, 541004, China.
- Guangxi Medical and Health Key Disciplines Infectious Diseases Key Disciplines, The Second Affiliated Hospital of Guilin Medical College, Guilin, 541004, China.
| |
Collapse
|
15
|
Sun X, Zhang J, Hu J, Han Q, Ge Z. LSM2 is associated with a poor prognosis and promotes cell proliferation, migration, and invasion in skin cutaneous melanoma. BMC Med Genomics 2023; 16:129. [PMID: 37312186 DOI: 10.1186/s12920-023-01564-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 05/31/2023] [Indexed: 06/15/2023] Open
Abstract
BACKGROUND Skin cutaneous melanoma (SKCM) is an extremely malignant tumor that is associated with a poor prognosis. LSM2 has been found to be related to different types of tumors; however, its role in SKCM is poorly defined. We aimed to determine the value of LSM2 as a prognostic biomarker for SKCM. METHODS The expression profile of LSM2 mRNA was compared between tumor and normal tissues in public databases, such as TCGA, GEO, and BioGPS. LSM2 protein expression was explored using immunohistochemistry (IHC) on a tissue microarray containing 44 SKCM tissues and 8 normal samples collected at our center. Kaplan-Meier analysis was performed to assess the prognostic value of LSM2 expression in patients with SKCM. SKCM cell lines with LSM2 knockdown were used to determine the effects of LSM2. Cell counting kit-8 (CCK8) and colony formation assays were conducted to assess cell proliferation, whereas wound healing and transwell assays were carried out to assess the migration and invasion abilities of SKCM cells. RESULTS LSM2 was more highly expressed at the mRNA and protein levels in SKCM than that in normal skin. Moreover, elevated expression of LSM2 was associated with shorter survival time and early recurrence in patients with SKCM. The in vitro results revealed that the silencing of LSM2 in SKCM cells significantly inhibited cell proliferation, migration, and invasion. CONCLUSION Overall, LSM2 contributes to malignant status and poor prognosis in patients with SKCM and may be identified as a novel prognostic biomarker and therapeutic target.
Collapse
Affiliation(s)
- Xiaofang Sun
- Department of Dermatology, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
- Department of Oral and Maxillofacial Surgery, the First Affiliated Hospital of Soochow University, Soochow University, Jiangsu, China
| | - Jianping Zhang
- Department of Dermatology, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Jiayuan Hu
- Department of Dermatology, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Qingdong Han
- Department of Dermatology, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Zili Ge
- Department of Oral and Maxillofacial Surgery, the First Affiliated Hospital of Soochow University, Soochow University, Jiangsu, China.
| |
Collapse
|
16
|
Liu Q, Lian Q, Song Y, Yang S, Jia C, Fang J. Identification of LSM family members as potential chemoresistance predictive and therapeutic biomarkers for gastric cancer. Front Oncol 2023; 13:1119945. [PMID: 37007092 PMCID: PMC10064066 DOI: 10.3389/fonc.2023.1119945] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 03/02/2023] [Indexed: 03/19/2023] Open
Abstract
IntroductionThe Like-Smith (LSM) family plays a critical role in the progression of several cancers. However, the function of LSMs in chemoresistance of gastric cancer (GC) is still elusive.MethodsThe Cancer Genome Atlas (TCGA) database, Gene Expression Omnibus (GEO) database and Tumor Immune Estimation Resource Analysis (TIMER) were utilized to analyze the expression, prognostic value and immune infiltration of LSMs in GC patients. Moreover, qPCR and immunohistochemistry (IHC) experiment were conducted with clinical samples.ResultsThe expression of LSMs was upregulated in GC tissues and most of LSMs were negatively correlated with overall survival of GC patients with 5-fluorouracil (5-FU) treatment. We further revealed that LSM5, 7 and 8 were hub genes of GEO (GSE14210). Besides, the qPCR results demonstrated that a higher level of LSM5 and LSM8 was associated with 5-FU chemoresistance in GC. Moreover, both TIMER and IHC revealed that a lower expression of LSM5 and LSM8 was correlated with high infiltration of T cells, regulatory T cells, B cells, macrophages, and neutrophils.DiscussionOur study systematically investigated the expression pattern and biological features of LSM family members in GC, and identified LSM5 and LSM8 as potential biomarkers in GC with 5-FU chemotherapy.
Collapse
Affiliation(s)
- Qianhui Liu
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Qinghai Lian
- Department of Cell-Gene Therapy Translational Medicine Research Center, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yingqiu Song
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Shangbin Yang
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Changchang Jia
- Department of Cell-Gene Therapy Translational Medicine Research Center, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Jiafeng Fang
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- *Correspondence: Jiafeng Fang,
| |
Collapse
|
17
|
ZHUANG YAN, NING CHUNLAN, LIU PENGFEI, ZHAO YANPENG, LI YUE, MA ZHENCHI, SHAN LULING, PIAO YINGZHE, ZHAO PENG, JIN XUN. LSM12 facilitates the progression of colorectal cancer by activating the WNT/CTNNB1 signaling pathway. Oncol Res 2023; 30:289-300. [PMID: 37303493 PMCID: PMC10207973 DOI: 10.32604/or.2022.028225] [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: 12/06/2022] [Accepted: 01/29/2023] [Indexed: 02/11/2023] Open
Abstract
Aberrant activation of the WNT signaling pathway is a joint event in colorectal cancer (CRC), but the molecular mechanism is still unclear. Recently, RNA-splicing factor LSM12 (like-Sm protein 12) is highly expressed in CRC tissues. This study aimed to verify whether LSM12 is involved in regulating CRC progression via regulating the WNT signaling pathway. Here, we found that LSM12 is highly expressed in CRC patient-derived tissues and cells. LSM12 is involved in the proliferation, invasion, and apoptosis of CRC cells, similar to the function of WNT signaling in CRC. Furthermore, protein interaction simulation and biochemical experiments proved that LSM12 directly binds to CTNNB1 (also known as β-Catenin) and regulates its protein stability to affect the CTTNB1-LEF1-TCF1 transcriptional complex formation and the associated WNT downstream signaling pathway. LSM12 depletion in CRC cells decreased the in vivo tumor growth through repression of cancer cell growth and acceleration of cancer cell apoptosis. Taken together, we suggest that the high expression of LSM12 is a novel factor leading to aberrant WNT signaling activation, and that strategies targeting this molecular mechanism may contribute to developing a new therapeutic method for CRC.
Collapse
Affiliation(s)
- YAN ZHUANG
- Department of Colorectal Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
| | - CHUNLAN NING
- Department of Biochemistry and Molecular Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
- Tianjin Medical University, Tianjin, 300070, China
| | - PENGFEI LIU
- Department of Oncology, Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin, 300120, China
| | - YANPENG ZHAO
- Tianjin Marvel Medical Laboratory, Tianjin Marvelbio Technology Co., Ltd., Tianjin, 300381, China
| | - YUE LI
- Department of Gastro Colorectal Oncology, Tianjin Cancer Hospital Airport Hospital, Tianjin, 300304, China
| | - ZHENCHI MA
- Department of Gastro Colorectal Oncology, Tianjin Cancer Hospital Airport Hospital, Tianjin, 300304, China
| | - LULING SHAN
- Department of Gastro Colorectal Oncology, Tianjin Cancer Hospital Airport Hospital, Tianjin, 300304, China
| | - YINGZHE PIAO
- Department of Neuro-Oncology and Neurosurgery, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy Tianjin, Tianjin’s Clinical Research Center for Cancer, Tianjin, 300060, China
| | - PENG ZHAO
- Department of Gastro Colorectal Oncology, Tianjin Cancer Hospital Airport Hospital, Tianjin, 300304, China
| | - XUN JIN
- Department of Biochemistry and Molecular Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
| |
Collapse
|
18
|
Liu T, Liu Y, Su X, Peng L, Chen J, Xing P, Qiao X, Wang Z, Di J, Zhao M, Jiang B, Qu H. Genome-wide transcriptomics and copy number profiling identify patient-specific CNV-lncRNA-mRNA regulatory triplets in colorectal cancer. Comput Biol Med 2023; 153:106545. [PMID: 36646024 DOI: 10.1016/j.compbiomed.2023.106545] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 12/19/2022] [Accepted: 01/10/2023] [Indexed: 01/13/2023]
Abstract
Screening cancer genomes has provided an in-depth characterization of genetic variants such as copy number variations (CNVs) and gene expression changes of non-coding transcripts. Single-dimensional experiments are often designed to differentiate a patient cohort into various sets with the aim of identifying molecular changes among groups; however, this may be inadequate to decipher the causal relationship between molecular signatures in individual patients. To overcome this challenge with respect to personalized medicine, we implemented a patient-specific multi-dimensional integrative approach to uncover coherent signals from multiple independent platforms. In particular, we focused on the consistent gene dosage effects of CNVs for both mRNA and long non-coding RNA (lncRNA) expression in nine colorectal cancer patients. We identified 511 CNV-lncRNA-mRNA regulatory triplets associated with CNVs and aberrant expression of both mRNAs and lncRNAs. By filtering out inconsistent changes among CNVs, mRNAs, and lncRNAs, we further characterized 165 coherent motifs associated with 56 genes. In total, 108 motifs were linked with 31 copy number gains, 44 upregulated lncRNAs, and 45 upregulated mRNAs. Another 57 coherent downregulated motifs were also collected. We discuss how for many of these CNV-lncRNA-mRNA regulatory triplets, their clinical impact remains to be explored, including survival time, microsatellite instability, tumor stage, and primary tumor sites. By validating two example CNV-lncRNA-mRNA triplets with up- and down-regulation, we confirmed that individual variations in multiple dimensions are a robust tool to identify reliable molecular signals for personalized medicine. In summary, we utilized a patient-specific computational pipeline to explore the consistent CNV-driven motifs consisting of lncRNAs and mRNAs. We also identified LSM14B as a potential promoter in colorectal cancer progression, suggesting that it may serve as a target for colorectal cancer treatment.
Collapse
Affiliation(s)
- Tianqi Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Yining Liu
- The School of Public Health, Institute for Chemical Carcinogenesis, Guangzhou Medical University, Guangzhou, China
| | - Xiangqian Su
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Lin Peng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Jiangbo Chen
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Pu Xing
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Xiaowen Qiao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Zaozao Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Jiabo Di
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Min Zhao
- School of Science, Technology and Engineering, University of the Sunshine Coast, Maroochydore DC, Queensland, 4558, Australia.
| | - Beihai Jiang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, Beijing, 100142, China.
| | - Hong Qu
- Center for Bioinformatics, State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, 100871, PR China.
| |
Collapse
|