1
|
Brada MD, Karakulak T, Schraml P, Haberecker M, Rutishauser D, Ross JS, Eberli D, Moch H. AXL and SRC in clear cell renal cell carcinoma: absence of mutations, rare alternative splicing events, but association of protein expression with poor prognosis. J Pathol Clin Res 2025; 11:e70028. [PMID: 40260977 DOI: 10.1002/2056-4538.70028] [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: 09/07/2024] [Revised: 03/05/2025] [Accepted: 03/20/2025] [Indexed: 04/24/2025]
Abstract
Novel treatment options for metastatic renal cell carcinomas (RCC) include specific MET inhibitors, GAS6/AXL inhibitors, and SRC inhibitors. The interplay between c-MET, SRC, AXL expression, and their gene mutation patterns in different renal carcinoma subtypes is unclear. To improve the understanding of these signaling pathways, we analyzed c-MET, AXL, and SRC expression in 590 clear cell RCC (ccRCC) and 127 papillary RCC (pRCC) by immunohistochemistry and integrated sequencing data to investigate the frequency of MET, AXL, and SRC gene mutations, their expression levels, and the presence of splice variants. In TCGA and in Foundation Medicine, Inc. (FMI) datasets, AXL and SRC gene alterations were extremely rare (<2%) or absent in ccRCC (n = 531 and 2,781, respectively) and pRCC (n = 290 and 566, respectively). On the other hand, MET mutations or amplifications were found in 9.7% (TCGA) and 10.2% (FMI) of pRCC. We show that strong SRC staining intensity by immunohistochemistry is associated with high tumor stage, high grade, and shorter survival in ccRCC (p < 0.001 each). AXL expression correlates with high stage and grade in ccRCC (p < 0.001 each). Both SRC and AXL expression were independent prognostic parameters in multivariate analysis (p < 0.05). MET expression is associated with longer survival in pRCC (p < 0.05). Our TCGA data analysis aligns with SRC immunohistochemistry findings on tumor stage and shorter survival in ccRCC. TCGA expression data showed a moderate positive correlation between AXL and c-MET in pRCC. In addition, we identified alternative splicing events reported for AXL in pRCC, and MET and SRC in ccRCC, across various alternative splicing databases. In conclusion, we identified high SRC expression as a biomarker for poor prognosis of ccRCC. Our data demonstrate c-MET, AXL, and SRC signaling pathway interactions independent of c-MET, AXL, and SRC mutations in ccRCC.
Collapse
Affiliation(s)
- Muriel D Brada
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Tülay Karakulak
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Peter Schraml
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Martina Haberecker
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Dorothea Rutishauser
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Jeffrey S Ross
- Foundation Medicine, Inc., Cambridge, MA, USA
- Department of Pathology, Upstate Medical University, Syracuse, NY, USA
- Department of Urology, Upstate Medical University, Syracuse, NY, USA
- Department of Medicine (Oncology), Upstate Medical University, Syracuse, NY, USA
| | - Daniel Eberli
- Department of Urology, University Hospital Zurich, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Holger Moch
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| |
Collapse
|
2
|
Imai T, Miyai M, Nemoto J, Tamai T, Ohta M, Yagi Y, Nakanishi O, Mochizuki H, Nakamori M. Pentatricopeptide repeat protein targeting CUG repeat RNA ameliorates RNA toxicity in a myotonic dystrophy type 1 mouse model. Sci Transl Med 2025; 17:eadq2005. [PMID: 40238915 DOI: 10.1126/scitranslmed.adq2005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 11/25/2024] [Accepted: 03/26/2025] [Indexed: 04/18/2025]
Abstract
Myotonic dystrophy type 1 (DM1) is an autosomal dominant multisystemic disorder caused by the expansion of a CTG-triplet repeat in the 3' untranslated region of the dystrophia myotonica protein kinase (DMPK) gene. It results in the transcription of toxic RNAs that contain expanded CUG repeats (CUGexp). Splicing factors, such as muscleblind-like 1 (MBNL1), are sequestered by CUGexp, thereby disrupting the normal splicing program that is essential for various cellular functions. Pentatricopeptide repeat (PPR) proteins, originally found in plants, regulate RNA in organelles by binding in a sequence-specific manner. Here, we designed PPR proteins that specifically bind to the hexamer of CUG repeat RNAs (CUG-PPRs) and showed that CUG-PPR1 could ameliorate RNA toxicity induced by CUGexp in cell models of DM1. A single systemic recombinant adeno-associated virus (AAV9) vector-mediated gene delivery of CUG-PPR1 demonstrated long-term therapeutic effects on myotonia and restored splicing activity in a mouse model of DM1. These results highlight the potential of PPR molecules to target pathogenic RNA sequences in DM1 and potentially other RNA-mediated disorders.
Collapse
Affiliation(s)
| | - Maiko Miyai
- Department of Neurology, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | - Joe Nemoto
- Department of Neurology, Yamaguchi University Graduate School of Medicine, Ube 755-8505, Japan
| | | | | | - Yusuke Yagi
- EditForce Inc., Fukuoka 819-0395, Japan
- Faculty of Agriculture, Kyushu University, Fukuoka 819-0395, Japan
| | | | - Hideki Mochizuki
- Department of Neurology, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | - Masayuki Nakamori
- Department of Neurology, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
- Department of Neurology, Yamaguchi University Graduate School of Medicine, Ube 755-8505, Japan
| |
Collapse
|
3
|
Cao L, Jia K, Van Tine BA, Yu Y, Peng Y, Chen X, Pan Q, Yang W, Zhang Z, Shao Z, Wu W. KPNA2 promotes osteosarcoma progression by regulating the alternative splicing of DDX3X mediated by YBX1. Oncogene 2025:10.1038/s41388-025-03375-3. [PMID: 40216969 DOI: 10.1038/s41388-025-03375-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 03/11/2025] [Accepted: 03/25/2025] [Indexed: 04/14/2025]
Abstract
Osteosarcoma (OS) is a rapidly progressive primary malignant bone tumor that occurs in children and adolescents aged between 15 and 19 years and adults aged over 60 years. As alternative splicing (AS) changes caused by abnormal splicing factors contribute to tumor progression, gene expression and AS analyses were performed on 44 osteosarcoma patients to create a genome-wide co-expression network of RNA-binding proteins (RBPs), AS events, and AS genes. A gain- or loss-of-function osteosarcoma cell model was established, and an interactive network analysis and enrichment analysis were performed. Karyopherin Subunit Alpha 2 (KPNA2) negatively correlated with patient survival. KPNA2 transports splicing factor Y-box Binding Protein 1 (YBX1) into the nucleus and YBX1 accelerates the degradation of the ATP-dependent RNA helicase DDX3X (DDX3X) through the nonsense-mediated decay (NMD) pathway to promote intron retention of the DDX3X gene, thus reducing DDX3X protein levels. KPNA2/YBX1 axis regulates the stability of DDX3X mRNA and cell cycle progression. KPNA2/YBX1/DDX3X axis might be potential targets for inhibiting disease progression and improving OS patient survival. It integrates AS control of DDX3X into the progression of OS and represents a potential prognostic biomarker and therapeutic target for OS therapy.
Collapse
Affiliation(s)
- Li Cao
- Department of Orthopaedic, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
- Division of Medical Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Ke Jia
- Department of Orthopaedic, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - B A Van Tine
- Division of Medical Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Yihan Yu
- Department of Orthopaedic, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - Yizhong Peng
- Department of Orthopaedic, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - Xuanzuo Chen
- Department of Orthopaedic, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - Qing Pan
- Department of Orthopaedic, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - Wenbo Yang
- Department of Orthopaedic, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - Zhicai Zhang
- Department of Orthopaedic, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China.
| | - Zengwu Shao
- Department of Orthopaedic, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China.
| | - Wei Wu
- Department of Orthopaedic, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China.
| |
Collapse
|
4
|
Zhang QQ, Miao YS, Hu JY, Liu RX, Hu YX, Wang F. The truncated AXIN1 isoform promotes hepatocellular carcinoma metastasis through SRSF9-mediated exon 9 skipping. Mol Cell Biochem 2025; 480:2247-2263. [PMID: 38748384 DOI: 10.1007/s11010-024-05012-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 04/14/2024] [Indexed: 04/02/2025]
Abstract
Axis inhibitor protein 1 (AXIN1) is a protein recognized for inhibiting tumor growth and is commonly involved in cancer development. In this study, we explored the potential molecular mechanisms that connect alternative splicing of AXIN1 to the metastasis of hepatocellular carcinoma (HCC). Transcriptome sequencing, RT‒PCR, qPCR and Western blotting were utilized to determine the expression levels of AXIN1 in human HCC tissues and HCC cells. The effects of the AXIN1 exon 9 alternative splice isoform and SRSF9 on the migration and invasion of HCC cells were assessed through wound healing and Transwell assays, respectively. The interaction between SRSF9 and AXIN1 was investigated using UV crosslink RNA immunoprecipitation, RNA pulldown, and RNA immunoprecipitation assays. Furthermore, the involvement of the AXIN1 isoform and SRSF9 in HCC metastasis was validated in a nude mouse model. AXIN1-L (exon 9 including) expression was downregulated, while AXIN1-S (exon 9 skipping) was upregulated in HCC. SRSF9 promotes the production of AXIN1-S by interacting with the sequence of exons 8 and 10 of AXIN1. AXIN1-S significantly promoted HCC cells migration and invasion by activating the Wnt pathway, while the opposite effects were observed for AXIN1-L. In vivo experiments demonstrated that AXIN1-L inhibited HCC metastasis, whereas SRSF9 promoted HCC metastasis in part by regulating the level of AXIN1-S. AXIN1, a tumor suppressor protein that targets the AXIN1/Wnt/β-catenin signaling axis, may be a promising prognostic factor and a valuable therapeutic target for HCC.
Collapse
Affiliation(s)
- Qian-Qian Zhang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Institute of Genomic Medicine, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou, 510632, China
| | - Ying-Shuang Miao
- International School, Jinan University, Guangzhou, 510632, China
| | - Jun-Yi Hu
- International School, Jinan University, Guangzhou, 510632, China
| | - Rui-Xuan Liu
- International School, Jinan University, Guangzhou, 510632, China
| | - Yue-Xiao Hu
- International School, Jinan University, Guangzhou, 510632, China
| | - Feng Wang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Institute of Genomic Medicine, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
5
|
Ni X, Wei Z, Peng Y, Zheng L, Shang J, Liu F, Li Y, Liu J. Triclosan exposure induces liver fibrosis in mice: The heterogeneous nuclear ribonucleoprotein A1/pyruvate kinase M2 axis drives hepatic stellate cell activation. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 294:118113. [PMID: 40157328 DOI: 10.1016/j.ecoenv.2025.118113] [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: 11/07/2024] [Revised: 03/23/2025] [Accepted: 03/25/2025] [Indexed: 04/01/2025]
Abstract
Triclosan (TCS) is an effective broad-spectrum antibacterial agent. TCS possesses a stable structure, can easily accumulate in the environment, and may have numerous negative impacts on human health. One organ particularly susceptible to TCS damage is the liver; however, the molecular mechanisms underlying TCS-induced liver damage remain unclear. A long-term TCS exposure model was established in C57BL/6 mice through maternal administration from gestation to postnatal 8-week-old. The offspring were randomly assigned to three groups (0, 50, and 100 mg/kg TCS) with six animals per group, ensuring an equal gender distribution (3 males and 3 females). The results showed that TCS-exposed mice exhibited serum aspartate aminotransferase, alanine aminotransferase, and alkaline phosphatase enzyme activities increased by 1.5-2 times when compared with vehicle-treated mice, along with features of liver fibrosis. In the LX-2 cell line, used as an in vitro model, TCS promoted proliferation and migration and induced the activation of hepatic stellate cells (HSCs). The level of pyruvate kinase M2 (PKM2) dimer increased by 200 % in LX-2 cells treated with TCS. PKM2 dimer overexpression stimulated HSC activation, whereas treatment with TEPP-46 (a PKM2 dimer inhibitor) significantly decreased the activation process. The expression of heterogeneous ribonucleoprotein particle A1 (hnRNPA1) was upregulated in the TCS treatment group and promoted the PKM2 expression. Moreover, disruption of the hnRNPA1/PKM2 axis reduced HSC proliferation and migration activated by TCS. Overall, our findings highlighted that TCS could cause liver fibrosis by stimulating the proliferation and migration of HSCs activated via the hnRNPA1/PKM2 axis, providing promising treatment options for TCS-related liver damage.
Collapse
Affiliation(s)
- Xiao Ni
- Department of Health Laboratory Technology, School of Public Health, China Medical University, Shenyang, Liaoning 110122, PR China
| | - Ziyun Wei
- Department of Health Laboratory Technology, School of Public Health, China Medical University, Shenyang, Liaoning 110122, PR China
| | - Yuxuan Peng
- Department of Health Laboratory Technology, School of Public Health, China Medical University, Shenyang, Liaoning 110122, PR China
| | - Linlin Zheng
- Department of Health Laboratory Technology, School of Public Health, China Medical University, Shenyang, Liaoning 110122, PR China
| | - Jianing Shang
- Department of Health Laboratory Technology, School of Public Health, China Medical University, Shenyang, Liaoning 110122, PR China
| | - Fu Liu
- Department of Health Laboratory Technology, School of Public Health, China Medical University, Shenyang, Liaoning 110122, PR China
| | - Yunwei Li
- Department of Anorectal Surgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, PR China.
| | - Jieyu Liu
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, PR China; Department of Health Laboratory Technology, School of Public Health, China Medical University, Shenyang, Liaoning 110122, PR China.
| |
Collapse
|
6
|
Zhou M, Tian M, Li Z, Wang C, Guo Z. Overview of splicing variation in ovarian cancer. Biochim Biophys Acta Rev Cancer 2025; 1880:189288. [PMID: 39993511 DOI: 10.1016/j.bbcan.2025.189288] [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: 10/14/2024] [Revised: 02/14/2025] [Accepted: 02/14/2025] [Indexed: 02/26/2025]
Abstract
Ovarian cancer remains one of the deadliest gynecological malignancies, with a persistently high mortality rate despite promising advancements in immunotherapy. Aberrant splicing events play a crucial role in cancer heterogeneity and treatment resistance. Many splicing variants, especially those involving key molecular markers such as BRCA1/2, are closely linked to disease progression and treatment outcomes. These variants and related splicing factors hold significant clinical value as diagnostic and prognostic biomarkers and therapeutic targets. This review provides a comprehensive overview of splicing variants in ovarian cancer, emphasizing their role in metastasis and resistance, and offers insights to advance biomarker development and treatment strategies.
Collapse
Affiliation(s)
- Min Zhou
- From the Department of Gynecology and Obstetrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Mengdie Tian
- From the Department of Gynecology and Obstetrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zhuoer Li
- From the Department of Gynecology and Obstetrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Chunli Wang
- From the Department of Gynecology and Obstetrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zhiqiang Guo
- From the Department of Gynecology and Obstetrics, Shengjing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
7
|
Sun S, Paniagua K, Ding L, Wang X, Huang Y, Flores MA, Gao SJ. KSHV Reprograms Host RNA Splicing via FAM50A to Activate STAT3 and Drive Oncogenic Cellular Transformation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.17.643747. [PMID: 40166334 PMCID: PMC11957025 DOI: 10.1101/2025.03.17.643747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
RNA alternative splicing is a fundamental cellular process implicated in cancer development. Kaposi's sarcoma-associated herpesvirus (KSHV), the etiological agent of multiple human malignancies, including Kaposi's sarcoma (KS), remains a significant concern, particularly in AIDS patients. A CRISPR-Cas9 screening of matched primary rat mesenchymal stem cells (MM) and KSHV-transformed MM cells (KMM) identified key splicing factors involved in KSHV-induced cellular transformation. To elucidate the mechanisms by which KSHV-driven splicing reprogramming mediates cellular transformation, we performed transcriptomic sequencing, identifying 131 differential alternative splicing transcripts, with exon skipping as the predominant event. Notably, these transcripts were enriched in vascular permeability, multiple metabolic pathways and ERK1/2 signaling cascades, which play key roles in KSHV-induced oncogenesis. Further analyses of cells infected with KSHV mutants lacking latent genes including vFLIP, vCyclin and viral miRNAs, as well as cells overexpressing LANA, revealed their involvement in alternative splicing regulation. Among the identified splicing factors, FAM50A, a component of the spliceosome complex C, was found to be crucial for KSHV-mediated transformation. FAM50A knockout resulted in distinct splicing profiles in both MM and KMM cells, and significantly inhibited KSHV-driven proliferation, cellular transformation and tumorigenesis. Mechanistically, FAM50A knockout altered SHP2 splicing, promoting an isoform with enhanced enzymatic activity that led to reduced STAT3 Y705 phosphorylation in KMM cells. These findings reveal a novel paradigm in which KSHV hijacks host splicing machinery, specifically FAM50A-mediated SHP2 splicing, to sustain STAT3 activation and drive oncogenic transformation. Importance Kaposi's sarcoma-associated herpesvirus (KSHV) causes cancers such as Kaposi's sarcoma, particularly in AIDS patients. This study uncovers how KSHV hijacks a fundamental cellular process called RNA splicing to promote cancer development. We identified key splicing events that alter critical pathways involved in vascular permeability, metabolism, and oncogenic signaling, particularly ERK1/2 and STAT3. A specific protein, FAM50A, was found to be essential for KSHV-driven cancerous transformation. Removing FAM50A disrupted splicing, weakening cancer-promoting signals. These findings provide new insights into how viruses manipulate host cells to drive cancer and highlight RNA splicing as a potential target for future therapies.
Collapse
Affiliation(s)
- Shenyu Sun
- Cancer Virology Program, University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Integrative System Biology Program, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Karla Paniagua
- Department of Electrical and Computer Engineering, University of Texas at San Antonio, San Antonio, TX
| | - Ling Ding
- Cancer Virology Program, University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Xian Wang
- Cancer Virology Program, University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Yufei Huang
- Cancer Virology Program, University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Department of Electrical and Computer Engineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mario A Flores
- Department of Electrical and Computer Engineering, University of Texas at San Antonio, San Antonio, TX
| | - Shou-Jiang Gao
- Cancer Virology Program, University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Integrative System Biology Program, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
8
|
Zhu X, Li W, Zhu T, Zheng W, Luo Q, Xu T, Sun Y. Identification and functional regulation of two alternative splicing isoforms of the Uhrf2 gene in Miichthysmiiuy. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2025; 166:105356. [PMID: 40074104 DOI: 10.1016/j.dci.2025.105356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 02/17/2025] [Accepted: 03/10/2025] [Indexed: 03/14/2025]
Abstract
Alternative splicing can produce a variety of splicing isoforms to increase protein diversity, participate in the regulation of gene expression and the occurrence and development of diseases, and thus play an important role in innate immunity. Ubiquitin like with PHD and ring finger domains 2 (Uhrf2) protein is associated with cell proliferation, inflammation, tumors, and cancer, and is currently the focus of medical immunology research, but there is little research on alternative splicing of the Uhrf2 gene. In this study, we identified two different splicing isoforms of Uhrf2 in Miichthys miiuy through Sanger sequencing, dual-luciferase reporter gene assay, qRT-PCR, subcellular localization experiments, and named them Uhrf2-α and Uhrf2-β. Subcellular localization experiments found that Uhrf2-α was mainly located in the nucleus, while Uhrf2-β was mainly located in the cytoplasm. Although their localization was different, both could significantly inhibit the activation of IRF3 and NF-κB signaling pathways, and effectively inhibit the levels of inflammatory cytokines. These results indicate that Uhrf2-α and Uhrf2-β play important negative regulatory roles in innate immune responses in fish.
Collapse
Affiliation(s)
- Xiangxiang Zhu
- Laboratory of Fish Molecular Immunology, College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, China
| | - Wenxin Li
- Laboratory of Fish Molecular Immunology, College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, China
| | - Tongtong Zhu
- Laboratory of Fish Molecular Immunology, College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, China
| | - Weiwei Zheng
- Laboratory of Fish Molecular Immunology, College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, China
| | - Qiang Luo
- Laboratory of Fish Molecular Immunology, College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, China
| | - Tianjun Xu
- Laboratory of Fish Molecular Immunology, College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, China; Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, China; Marine Biomedical Science and Technology Innovation Platform of Lin-gang Special Area, Shanghai, China.
| | - Yuena Sun
- Laboratory of Fish Molecular Immunology, College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, China; National Pathogen Collection Center for Aquatic Animals, Shanghai Ocean University, China; Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, China.
| |
Collapse
|
9
|
Denisko D, Kim J, Ku J, Zhao B, Lee EA. Inverted Alu repeats in loop-out exon skipping across hominoid evolution. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.07.642063. [PMID: 40161837 PMCID: PMC11952303 DOI: 10.1101/2025.03.07.642063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Background Changes in RNA splicing over the course of evolution have profoundly diversified the functional landscape of the human genome. While DNA sequences proximal to intron-exon junctions are known to be critical for RNA splicing, the impact of distal intronic sequences remains underexplored. Emerging evidence suggests that inverted pairs of intronic Alu elements can promote exon skipping by forming RNA stem-loop structures. However, their prevalence and influence throughout evolution remain unknown. Results Here, we present a systematic analysis of inverted Alu pairs across the human genome to assess their impact on exon skipping through predicted RNA stem-loop formation and their relevance to hominoid evolution. We found that inverted Alu pairs, particularly pairs of AluY-AluSx1 and AluSz-AluSx, are enriched in the flanking regions of skippable exons genome-wide and are predicted to form stable stem-loop structures. Exons defined by weak 3' acceptor and strong 5' donor splice sites appear especially prone to this skipping mechanism. Through comparative genome analysis across nine primate species, we identified 67,126 hominoid-specific Alu insertions, primarily from AluY and AluS subfamilies, which form inverted pairs enriched across skippable exons in genes of ubiquitination-related pathways. Experimental validation of exon skipping among several hominoid-specific inverted Alu pairs further reinforced their potential evolutionary significance. Conclusion This work extends our current knowledge of the roles of RNA secondary structure formed by inverted Alu pairs and details a newly emerging mechanism through which transposable elements have contributed to genomic innovation across hominoid evolution at the transcriptomic level.
Collapse
Affiliation(s)
- Danielle Denisko
- Division of Genetics and Genomics, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA
| | - Jeonghyeon Kim
- Division of Genetics and Genomics, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115, USA
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA 94305, USA
| | - Jayoung Ku
- Division of Genetics and Genomics, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02115, USA
- Manton Center for Orphan Disease Research, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Boxun Zhao
- Division of Genetics and Genomics, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02115, USA
- Manton Center for Orphan Disease Research, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Eunjung Alice Lee
- Division of Genetics and Genomics, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02115, USA
- Manton Center for Orphan Disease Research, Boston Children’s Hospital, Boston, MA 02115, USA
| |
Collapse
|
10
|
Zhang S, Zhang Y, Feng S, Han M, Wang Z, Qiao D, Tian J, Wang L, Du B, Zhang Z, Zhong J. Tumor-promoting effect and tumor immunity of SRSFs. Front Cell Dev Biol 2025; 13:1527309. [PMID: 40129567 PMCID: PMC11931056 DOI: 10.3389/fcell.2025.1527309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 02/17/2025] [Indexed: 03/26/2025] Open
Abstract
Serine/arginine-rich splicing factors (SRSFs) are a family of 12 RNA-binding proteins crucial for the precursor messenger RNA (pre-mRNA) splicing. SRSFs are involved in RNA metabolism events such as transcription, translation, and nonsense decay during the shuttle between the nucleus and cytoplasm, which are important components of genome diversity and cell viability. SRs recognize splicing elements on pre-mRNA and recruit the spliceosome to regulate splicing. In tumors, aberrant expression of SRSFs leads to aberrant splicing of RNA, affecting the proliferation, migration, and anti-apoptotic ability of tumor cells, highlighting the therapeutic potential of targeted SRSFs for the treatment of diseases. The body's immune system is closely related to the occurrence and development of tumor, and SRSFs can affect the function of immune cells in the tumor microenvironment by regulating the alternative splicing of tumor immune-related genes. We review the important role of SRSFs-induced aberrant gene expression in a variety of tumors and the immune system, and prospect the application of SRSFs in tumor. We hope that this review will inform future treatment of the disease.
Collapse
Affiliation(s)
- Shuai Zhang
- Department of Oncology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Department of Pathology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Department of Pathology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Yongxi Zhang
- Department of Oncology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Sijia Feng
- Department of Pathology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Miaomiao Han
- Department of Pathology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Zixi Wang
- Department of Pathology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Dan Qiao
- Department of Pathology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Jiaqi Tian
- Department of Pathology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Lan Wang
- Department of Pathology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Baoshun Du
- Second Department of Neurosurgery, Xinxiang Central Hospital, Xinxiang, China
| | - Zheying Zhang
- Department of Pathology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Jiateng Zhong
- Department of Oncology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Department of Pathology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Department of Pathology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
- Henan Province Engineering Technology Research Center of Tumor diagnostic biomarkers and RNA interference drugs, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
11
|
Luo S, Peng H, Shi Y, Cai J, Zhang S, Shao N, Li J. Integration of proteomics profiling data to facilitate discovery of cancer neoantigens: a survey. Brief Bioinform 2025; 26:bbaf087. [PMID: 40052441 PMCID: PMC11886573 DOI: 10.1093/bib/bbaf087] [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: 10/17/2024] [Revised: 12/29/2024] [Accepted: 02/19/2025] [Indexed: 03/10/2025] Open
Abstract
Cancer neoantigens are peptides that originate from alterations in the genome, transcriptome, or proteome. These peptides can elicit cancer-specific T-cell recognition, making them potential candidates for cancer vaccines. The rapid advancement of proteomics technology holds tremendous potential for identifying these neoantigens. Here, we provided an up-to-date survey about database-based search methods and de novo peptide sequencing approaches in proteomics, and we also compared these methods to recommend reliable analytical tools for neoantigen identification. Unlike previous surveys on mass spectrometry-based neoantigen discovery, this survey summarizes the key advancements in de novo peptide sequencing approaches that utilize artificial intelligence. From a comparative study on a dataset of the HepG2 cell line and nine mixed hepatocellular carcinoma proteomics samples, we demonstrated the potential of proteomics for the identification of cancer neoantigens and conducted comparisons of the existing methods to illustrate their limits. Understanding these limits, we suggested a novel workflow for neoantigen discovery as perspectives.
Collapse
Affiliation(s)
- Shifu Luo
- Faculty of Computer Science and Control Engineering, Shenzhen University of Advanced Technology, Shenzhen, 518107, Guangdong, China
- Faculty of Health Sciences, University of Macau, Taipa, Macao SAR 999078, China
| | - Hui Peng
- Faculty of Computer Science and Control Engineering, Shenzhen University of Advanced Technology, Shenzhen, 518107, Guangdong, China
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore
| | - Ying Shi
- Faculty of Computer Science and Control Engineering, Shenzhen University of Advanced Technology, Shenzhen, 518107, Guangdong, China
- School of Computer and Information Technology, Shanxi University, Taiyuan, 030006, Shanxi, China
| | - Jiaxin Cai
- Faculty of Computer Science and Control Engineering, Shenzhen University of Advanced Technology, Shenzhen, 518107, Guangdong, China
| | - Songming Zhang
- Faculty of Computer Science and Control Engineering, Shenzhen University of Advanced Technology, Shenzhen, 518107, Guangdong, China
| | - Ningyi Shao
- Faculty of Health Sciences, University of Macau, Taipa, Macao SAR 999078, China
| | - Jinyan Li
- Faculty of Computer Science and Control Engineering, Shenzhen University of Advanced Technology, Shenzhen, 518107, Guangdong, China
| |
Collapse
|
12
|
McGirr T, Onar O, Jafarnejad SM. Dysregulated ribosome quality control in human diseases. FEBS J 2025; 292:936-959. [PMID: 38949989 PMCID: PMC11880988 DOI: 10.1111/febs.17217] [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: 02/12/2024] [Revised: 05/31/2024] [Accepted: 06/20/2024] [Indexed: 07/03/2024]
Abstract
Precise regulation of mRNA translation is of fundamental importance for maintaining homeostasis. Conversely, dysregulated general or transcript-specific translation, as well as abnormal translation events, have been linked to a multitude of diseases. However, driven by the misconception that the transient nature of mRNAs renders their abnormalities inconsequential, the importance of mechanisms that monitor the quality and fidelity of the translation process has been largely overlooked. In recent years, there has been a dramatic shift in this paradigm, evidenced by several seminal discoveries on the role of a key mechanism in monitoring the quality of mRNA translation - namely, Ribosome Quality Control (RQC) - in the maintenance of homeostasis and the prevention of diseases. Here, we will review recent advances in the field and emphasize the biological significance of the RQC mechanism, particularly its implications in human diseases.
Collapse
Affiliation(s)
- Tom McGirr
- Patrick G. Johnston Centre for Cancer ResearchQueen's University BelfastUK
| | - Okan Onar
- Patrick G. Johnston Centre for Cancer ResearchQueen's University BelfastUK
- Department of Biology, Faculty of ScienceAnkara UniversityTurkey
| | | |
Collapse
|
13
|
Dho SE, Othman K, Zhang Y, McGlade CJ. NUMB alternative splicing and isoform-specific functions in development and disease. J Biol Chem 2025; 301:108215. [PMID: 39863103 PMCID: PMC11889595 DOI: 10.1016/j.jbc.2025.108215] [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: 10/04/2023] [Revised: 01/06/2025] [Accepted: 01/11/2025] [Indexed: 01/27/2025] Open
Abstract
The NUMB gene encodes a conserved adaptor protein with roles in asymmetric cell division and cell fate determination. First described as an inhibitor of Notch signaling, multifunctional NUMB proteins regulate multiple cellular pathways through protein complexes with ubiquitin ligases, polarity proteins and the endocytic machinery. The vertebrate NUMB protein isoforms were identified over 2 decades ago, yet the majority of functional studies exploring NUMB function in endocytosis, cell migration and adhesion, development and disease have largely neglected the potential for distinct isoform activity in design and interpretation. In this review we consolidate the literature that has directly addressed individual NUMB isoform functions, as well as interpret other functional studies through the lens of the specific isoforms that were utilized. We also summarize the emerging literature on the mechanisms that regulate alternative splicing of NUMB, and how this is subverted in disease. Finally, the importance of relative NUMB isoform expression as a determinant of activity and considerations for future studies of NUMB isoforms as unique proteins with distinct functions are discussed.
Collapse
Affiliation(s)
- Sascha E Dho
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Kamal Othman
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Yangjing Zhang
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - C Jane McGlade
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
14
|
Liang Y, Mi Z, Kuo PC. Differential MYC and PROM1 mRNA isoform expression in breast invasive carcinoma as biomarkers for subtyping and prognosis. Surgery 2025; 179:108798. [PMID: 39306567 DOI: 10.1016/j.surg.2024.07.056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 06/26/2024] [Accepted: 07/14/2024] [Indexed: 02/02/2025]
Abstract
BACKGROUND Cancer stem cells are a subpopulation of tumor cells with the ability to self-renew; evidence suggests that cancer stem cells are responsible for recurrence, metastasis, and resistance to therapy. MYC and CD133 (PROM1 gene) are clinical biomarkers for cancer stem cells, and their dysregulation is involved in the progression of many cancers. Alternative splicing of these genes may contribute to cancer stem cell differentiation. METHODS Transcriptional and clinical data of PROM1 and MYC mRNA isoforms in breast cancer samples were downloaded from the TCGA Splicing Variants Database site, a web-tool to explore mRNA alternative-splicing based on TCGA samples. Data include RSEM isoform expression, clinical sample types, survival data, and clinical receptor expression. Breast cancer subtypes (luminal A, luminal B, Her2 positive, triple negative) were assigned on the basis of estrogen, progesterone, and HER2 expression. RESULTS Expression of MYC isoforms uc003ysh.1 and uc003ysi.3 was significantly greater in triple-negative breast cancer compared with all other breast cancer subtypes (P < .001). Isoform uc003ysi.3 was associated with greater 5-year survival in luminal A breast cancer (hazard ratio, 0.79; 95% confidence interval, 0.65-0.96; P = .02). PROM1 isoforms uc003gop.2, uc003goq.3, uc003gos.2, and uc003gou.2 were expressed greatest in triple-negative breast cancer (P < .001). PROM1 isoform uc003gou.2 was associated with better 5-year survival in luminal A breast cancer (hazard ratio, 0.79; 95% confidence interval, 0.65-0.97; P = .02). CONCLUSIONS MYC and PROM1 isoforms are differentially expressed in breast cancer subtypes. Certain isoforms confer better survival prognosis. Further work should be done to study alternative splicing in cancer stem cells.
Collapse
Affiliation(s)
- Yifan Liang
- Department of Surgery, University of South Florida Morsani College of Medicine, Tampa, FL
| | - Zhiyong Mi
- Department of Surgery, University of South Florida Morsani College of Medicine, Tampa, FL
| | - Paul C Kuo
- Department of Surgery, University of South Florida Morsani College of Medicine, Tampa, FL; Bay Pines Veterans Affairs Health Care System, Bay Pines, FL.
| |
Collapse
|
15
|
Liu X, Song J, Zhou Z, He Y, Wu S, Yang J, Ren Z. Establishment of an alternative splicing prognostic risk model and identification of FN1 as a potential biomarker in glioblastoma multiforme. Sci Rep 2025; 15:6716. [PMID: 40000711 PMCID: PMC11862013 DOI: 10.1038/s41598-025-91038-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 02/18/2025] [Indexed: 02/27/2025] Open
Abstract
Aberrant alternative splicing and abnormal alternative splicing events (ASEs) in glioblastoma multiforme (GBM) remain largely elusive. The prognostic-associated ASEs in GBM were identified and summarized into 123 genes using GBM and LGG datasets from ASCancer Atlas and TCGA. The eleven genes (C2, COL3A1, CTSL, EIF3L, FKBP9, FN1, HPCAL1, HSPB1, IGFBP4, MANBA, PRKAR1B) were screened to develop an alternative splicing prognostic risk score (ASRS) model through machine learning algorithms. The model was trained on the TCGA-GBM cohort and validated with four external datasets from CGGA and GEO, achieving AUC values of 0.808, 0.814, 0.763, 0.859, and 0.836 for 3-year survival rates, respectively. ASRS could be an independent prognostic factor for GBM patients (HR > 1.8 across three datasets) through multivariate Cox regression analysis. The high-risk group demonstrated poorer prognosis, elevated immune scores, increased levels of immune cell infiltration, and greater differences in drug sensitivity. We found that FN1, used for model construction, contained 4 abnormal ASEs resulting in high expression of non-canonical transcripts and the presence of premature termination codon. These abnormal ASEs may be regulated by tumour-related splicing factors according to the PPI network. Furthermore, both mRNA and protein levels of FN1 were highly expressed in GBM compared to LGG, correlating with poor prognosis in GBM. In conclusion, our findings highlight the role of ASEs in affecting the progression of GBM, and the model showed a potential application for prognostic risk of patients. FN1 may serve as a promising splicing biomarker for GBM, and mechanisms of processes of aberrant splicing need to be revealed in the future.
Collapse
Affiliation(s)
- Xi Liu
- School of Medical Information and Engineering, Guangdong Pharmaceutical University, Guangzhou, 510006, China
- Guangdong Province Precise Medicine Big Data of Traditional Chinese Medicine Engineering Technology Research Center, Guangzhou, 51006, China
| | - Jinming Song
- School of Medical Information and Engineering, Guangdong Pharmaceutical University, Guangzhou, 510006, China
- Guangdong Province Precise Medicine Big Data of Traditional Chinese Medicine Engineering Technology Research Center, Guangzhou, 51006, China
| | - Zhiming Zhou
- School of Medical Information and Engineering, Guangdong Pharmaceutical University, Guangzhou, 510006, China
- Guangdong Province Precise Medicine Big Data of Traditional Chinese Medicine Engineering Technology Research Center, Guangzhou, 51006, China
| | - Yuting He
- School of Medical Information and Engineering, Guangdong Pharmaceutical University, Guangzhou, 510006, China
- Guangdong Province Precise Medicine Big Data of Traditional Chinese Medicine Engineering Technology Research Center, Guangzhou, 51006, China
| | - Shaochun Wu
- School of Medical Information and Engineering, Guangdong Pharmaceutical University, Guangzhou, 510006, China
- Guangdong Province Precise Medicine Big Data of Traditional Chinese Medicine Engineering Technology Research Center, Guangzhou, 51006, China
| | - Jin Yang
- School of Medical Information and Engineering, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
- Guangdong Province Precise Medicine Big Data of Traditional Chinese Medicine Engineering Technology Research Center, Guangzhou, 51006, China.
| | - Zhonglu Ren
- School of Medical Information and Engineering, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
- Guangdong Province Precise Medicine Big Data of Traditional Chinese Medicine Engineering Technology Research Center, Guangzhou, 51006, China.
| |
Collapse
|
16
|
Suri P, Badalov A, Ruggiu M. Alternative Splicing as a Modulator of the Interferon-Gamma Pathway. Cancers (Basel) 2025; 17:594. [PMID: 40002189 PMCID: PMC11853465 DOI: 10.3390/cancers17040594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 01/31/2025] [Accepted: 02/08/2025] [Indexed: 02/27/2025] Open
Abstract
Interferon-gamma (IFN-γ) is a critical cytokine that plays a pivotal role in immune system regulation. It is a key mediator of both cellular defense mechanisms and antitumor immunity. As the sole member of the type II interferon family, IFN-γ modulates immune responses by activating macrophages, enhancing natural killer cell function, and regulating gene expression across multiple cellular processes. Alternative splicing is a post-transcriptional gene expression regulatory mechanism that generates multiple mature messenger RNAs from a single gene, dramatically increasing proteome diversity without the need of a proportional genome expansion. This process occurs in 90-95% of human genes, with alternative splicing events allowing for the production of diverse protein isoforms that can have distinct-or even opposing-functional properties. Alternative splicing plays a crucial role in cancer immunology, potentially generating tumor neoepitopes and modulating immune responses. However, how alternative splicing affects IFN-γ's activity is still poorly understood. This review explores how alternative splicing regulates the expression and function of both upstream regulators and downstream effectors of IFN-γ, revealing complex mechanisms of gene expression and immune response modulation. Key transcription factors and signaling molecules of the IFN-γ pathway are alternatively spliced, and alternative splicing can dramatically alter IFN-γ signaling, immune cell function, and response to environmental cues. Specific splice variants can enhance or inhibit IFN-γ-mediated immune responses, potentially influencing cancer immunotherapy, autoimmune conditions, and infectious disease outcomes. The emerging understanding of these splicing events offers promising therapeutic strategies for manipulating immune responses through targeted molecular interventions.
Collapse
Affiliation(s)
- Parul Suri
- College of Pharmacy and Health Sciences, St. John’s University, 8000 Utopia Parkway Queens, New York, NY 11439, USA;
| | - Ariana Badalov
- Laboratory of RNA Biology and Molecular Neuroscience, Department of Biological Sciences, St. John’s University, 8000 Utopia Parkway Queens, New York, NY 11439, USA;
| | - Matteo Ruggiu
- Laboratory of RNA Biology and Molecular Neuroscience, Department of Biological Sciences, St. John’s University, 8000 Utopia Parkway Queens, New York, NY 11439, USA;
| |
Collapse
|
17
|
Ronemus M, Bradford D, Laster Z, Li S. Exploring genome-transcriptome correlations in cancer. Biochem Soc Trans 2025; 53:BST20240108. [PMID: 39910794 DOI: 10.1042/bst20240108] [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: 09/28/2024] [Revised: 12/16/2024] [Accepted: 12/23/2024] [Indexed: 02/07/2025]
Abstract
We examine the complex relationship between genomic copy number variation (CNV) and gene expression, highlighting the relevance to cancer biology and other biological contexts. By tracing the history of genometranscriptome correlations, we emphasize the complexity and challenges in understanding these interactions, particularly within the heterogeneous landscape of human cancers. Recent advances in computational algorithms and high-throughput single-cell multi-omic sequencing technologies are discussed, demonstrating their potential to refine our understanding of cancer biology and their limitations. The integration of genomic and transcriptomic analyses, which offers novel insights into tumor evolution and heterogeneity as well as therapeutic strategies, is presented as a crucial approach for advancing cancer research.
Collapse
Affiliation(s)
- Michael Ronemus
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, U.S.A
| | - Daniel Bradford
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, U.S.A
| | - Zachary Laster
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, U.S.A
| | - Siran Li
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, U.S.A
| |
Collapse
|
18
|
Wang H, Qian D, Wang J, Liu Y, Luo W, Zhang H, Cheng J, Li H, Wu Y, Li W, Wang J, Yang X, Zhang T, Han D, Wang Q, Zhang CZ, Liu L. HnRNPR-mediated UPF3B mRNA splicing drives hepatocellular carcinoma metastasis. J Adv Res 2025; 68:257-270. [PMID: 38402949 PMCID: PMC11785583 DOI: 10.1016/j.jare.2024.02.010] [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: 08/06/2023] [Revised: 02/06/2024] [Accepted: 02/15/2024] [Indexed: 02/27/2024] Open
Abstract
INTRODUCTION Abnormal alternative splicing (AS) contributes to aggressive intrahepatic invasion and metastatic spread, leading to the high lethality of hepatocellular carcinoma (HCC). OBJECTIVES This study aims to investigate the functional implications of UPF3B-S (a truncated oncogenic splice variant) in HCC metastasis. METHODS Basescope assay was performed to analyze the expression of UPF3B-S mRNA in tissues and cells. RNA immunoprecipitation, and in vitro and in vivo models were used to explore the role of UPF3B-S and the underlying mechanisms. RESULTS We show that splicing factor HnRNPR binds to the pre-mRNA of UPF3B via its RRM2 domain to generate an exon 8 exclusion truncated splice variant UPF3B-S. High expression of UPF3B-S is correlated with tumor metastasis and unfavorable overall survival in patients with HCC. The knockdown of UPF3B-S markedly suppresses the invasive and migratory capacities of HCC cells in vitro and in vivo. Mechanistically, UPF3B-S protein targets the 3'-UTR of CDH1 mRNA to enhance the degradation of CDH1 mRNA, which results in the downregulation of E-cadherin and the activation of epithelial-mesenchymal transition. Overexpression of UPF3B-S enhances the dephosphorylation of LATS1 and the nuclear accumulation of YAP1 to trigger the Hippo signaling pathway. CONCLUSION Our findings suggest that HnRNPR-induced UPF3B-S promotes HCC invasion and metastasis by exhausting CDH1 mRNA and modulating YAP1-Hippo signaling. UPF3B-S could potentially serve as a promising biomarker for the clinical management of invasive HCC.
Collapse
Affiliation(s)
- Hong Wang
- Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China; Department of Radiation Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Dong Qian
- Department of Radiation Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Jiabei Wang
- Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China; Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Yao Liu
- Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China; Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Wenguang Luo
- Department of Radiation Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Hongyan Zhang
- Department of Radiation Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Jingjing Cheng
- Department of Radiation Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Heng Li
- Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China; Department of Comprehensive Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC) West District/Anhui Provincial Cancer Hospital, Hefei, Anhui, China
| | - Yang Wu
- Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China; Department of General Surgery, Division of Life Science and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, China
| | - Wuhan Li
- Department of Emergency Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Jing Wang
- Department of Pathology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China; Intelligent Pathology Institute, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Xia Yang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Tianzhi Zhang
- Department of Pathology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Dong Han
- Tianjin Medical University Cancer Institute and Hospital, Department of Radiation Oncology, Tianjin, China
| | - Qinyao Wang
- Anhui Chest Hospital, Department of Radiation Oncology, Hefei, Anhui, China
| | - Chris Zhiyi Zhang
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China.
| | - Lianxin Liu
- Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China; Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.
| |
Collapse
|
19
|
Djamgoz MBA. Stemness of Cancer: A Study of Triple-negative Breast Cancer From a Neuroscience Perspective. Stem Cell Rev Rep 2025; 21:337-350. [PMID: 39531198 PMCID: PMC11872763 DOI: 10.1007/s12015-024-10809-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/21/2024] [Indexed: 11/16/2024]
Abstract
Stemness, giving cancer cells massive plasticity enabling them to survive in dynamic (e.g. hypoxic) environments and become resistant to treatment, especially chemotherapy, is an important property of aggressive tumours. Here, we review some essentials of cancer stemness focusing on triple-negative breast cancer (TNBC), the most aggressive form of all breast cancers. TNBC cells express a range of genes and mechanisms associated with stemness, including the fundamental four "Yamanaka factors". Most of the evidence concerns the transcription factor / oncogene c-Myc and an interesting case is the expression of the neonatal splice variant of voltage-gated sodium channel subtype Nav1.5. On the whole, measures that reduce the stemness make cancer cells less aggressive, reducing their invasive/metastatic potential and increasing/restoring their chemosensitivity. Such measures include gene silencing techniques, epigenetic therapies as well as novel approaches like optogenetics aiming to modulate the plasma membrane voltage. Indeed, simply hyperpolarizing their membrane potential can make stem cells differentiate. Finally, we give an overview of the clinical aspects and exploitation of cancer/TNBC stemness, including diagnostics and therapeutics. In particular, personalised mRNA-based therapies and mechanistically meaningful combinations are promising and the emerging discipline of 'cancer neuroscience' is providing novel insights to both fundamental issues and clinical applications.
Collapse
Affiliation(s)
- Mustafa B A Djamgoz
- Department of Life Sciences, Imperial College London, South Kensington Campus, London, SW7 2AZ, UK.
| |
Collapse
|
20
|
Lv X, Sun X, Gao Y, Song X, Hu X, Gong L, Han L, He M, Wei M. Targeting RNA splicing modulation: new perspectives for anticancer strategy? J Exp Clin Cancer Res 2025; 44:32. [PMID: 39885614 PMCID: PMC11781073 DOI: 10.1186/s13046-025-03279-w] [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: 11/18/2024] [Accepted: 01/07/2025] [Indexed: 02/01/2025] Open
Abstract
The excision of introns from pre-mRNA is a crucial process in the expression of the majority of genes. Alternative splicing allows a single gene to generate diverse mRNA and protein products. Aberrant RNA splicing is recognized as a molecular characteristic present in almost all types of tumors. Therefore, identifying cancer-specific subtypes from aberrant processing offers new opportunities for therapeutic development. Numerous splicing modulators, each utilizing different mechanisms, have been developed as promising anticancer therapies, some of which are in clinical trials. In this review, we summarize the splice-altered signatures of cancer cell transcriptomes and the contributions of splicing aberrations to tumorigenesis and progression. Especially, we discuss current and emerging RNA splicing-targeted strategies for cancer therapy, including pharmacological approaches and splice-switching antisense oligonucleotides (ASOs). Finally, we address the challenges and opportunities in translating these findings into clinical practice.
Collapse
Affiliation(s)
- Xuemei Lv
- Department of Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, 110122, P. R. China
- Central Laboratory, School of Pharmacy, China Medical University, Shenyang, Liaoning Province, China
| | - Xiaoyu Sun
- Department of Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, 110122, P. R. China
| | - Yang Gao
- Department of Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, 110122, P. R. China
| | - Xinyue Song
- Department of Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, 110122, P. R. China
| | - Xiaoyun Hu
- Scientific Experimental Center, School of Pharmacy, China Medical University, Shenyang, 110122, P. R. China
| | - Lang Gong
- Department of Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, 110122, P. R. China
| | - Li Han
- Department of Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, 110122, P. R. China.
- Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Shenyang, China.
| | - Miao He
- Department of Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, 110122, P. R. China.
- Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Shenyang, China.
| | - Minjie Wei
- Department of Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, 110122, P. R. China.
- Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Shenyang, China.
- Shenyang Kangwei Medical Laboratory Analysis Co. LTD, Shenyang, China.
| |
Collapse
|
21
|
Hou X, Dong Q, Hao J, Liu M, Ning J, Tao M, Wang Z, Guo F, Huang D, Shi X, Gao M, Li D, Zheng X. NSUN2-mediated m 5C modification drives alternative splicing reprogramming and promotes multidrug resistance in anaplastic thyroid cancer through the NSUN2/SRSF6/UAP1 signaling axis. Theranostics 2025; 15:2757-2777. [PMID: 40083919 PMCID: PMC11898302 DOI: 10.7150/thno.104713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 01/14/2025] [Indexed: 03/16/2025] Open
Abstract
Rationale: Anaplastic thyroid carcinoma (ATC) is an extraordinarily aggressive form of thyroid cancer, frequently presenting with locally advanced infiltration or distant metastases at the time of initial diagnosis, thus missing the optimal window for surgical intervention. Consequently, systemic chemotherapy and targeted therapies are vital for improving the prognosis of ATC. However, ATC exhibits significant resistance to conventional treatments, highlighting the need to elucidate the biological mechanisms underlying this drug resistance and identify novel therapeutic targets to overcome it. Methods: We conducted a comprehensive analysis of both bulk and single-cell RNA sequencing (scRNA-seq) data from ATC samples to screen for m5C modification-related genes associated with multidrug resistance (MDR). We then performed IC50 assays, flow cytometry, and employed a spontaneous tumorigenic ATC mouse model with Nsun2 knockout to demonstrate that NSUN2 promotes MDR in ATC. To investigate the mechanisms of NSUN2-mediated drug resistance, we generated NSUN2-knockout ATC cell lines and performed transcriptomic, proteomic, and MeRIP-seq analyses. Additionally, RNA sequencing and alternative splicing analyses were conducted to determine global changes upon NSUN2 knockout. We further explored the underlying mechanisms of the NSUN2/SRSF6/UAP1 axis through glycoprotein staining, denaturing IP ubiquitination, nuclear-cytoplasmic fractionation, and PCR. Lastly, we evaluated the synergistic effects of a small-molecule NSUN2 inhibitor with anticancer agents both in vitro and in vivo. Results: Our findings reveal that NSUN2 expression correlates significantly with MDR in ATC. NSUN2 operates as a "writer" and ALYREF as a "reader" of m5C on SRSF6 mRNA, inducing alternative splicing reprogramming and redirecting the splice form of the UAP1 gene from AGX1 to AGX2. As a result, AGX2 enhances the N-linked glycosylation of ABC transporters, stabilizing them by preventing ubiquitination-mediated degradation. Furthermore, an NSUN2 inhibitor reduces NSUN2 enzymatic activity and diminishes downstream target expression, presenting a novel, promising therapeutic approach to overcome MDR in ATC. Conclusions: These findings suggest that the NSUN2/SRSF6/UAP1 signaling axis plays a vital role in MDR of ATC and identify NSUN2 as a synergistic target for chemotherapy and targeted therapy in ATC.
Collapse
Affiliation(s)
- Xiukun Hou
- Department of Thyroid and Neck Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300040, China
| | - Qiman Dong
- Department of Thyroid and Neck Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300040, China
| | - Jie Hao
- Department of Thyroid and Neck Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300040, China
- Department of Thyroid and Breast Surgery, Tianjin Union Medical Center, Tianjin 300121, China
- Tianjin Key Laboratory of General Surgery in Construction, Tianjin Union Medical Center, Tianjin 300121, China
| | - Min Liu
- Department of Thyroid and Neck Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300040, China
| | - Junya Ning
- Department of Thyroid and Neck Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300040, China
| | - Mei Tao
- Department of Thyroid and Neck Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300040, China
| | - Zhongyu Wang
- Department of Thyroid and Neck Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300040, China
| | - Fengli Guo
- Department of Thyroid and Neck Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300040, China
| | - Dongmei Huang
- Department of Thyroid and Neck Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300040, China
| | - Xianle Shi
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Ming Gao
- Department of Thyroid and Neck Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300040, China
- Department of Thyroid and Breast Surgery, Tianjin Union Medical Center, Tianjin 300121, China
- Tianjin Key Laboratory of General Surgery in Construction, Tianjin Union Medical Center, Tianjin 300121, China
| | - Dapeng Li
- Department of Thyroid and Neck Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300040, China
| | - Xiangqian Zheng
- Department of Thyroid and Neck Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300040, China
| |
Collapse
|
22
|
Wang HC, Gao AC, Xia R, Wu CT, Hsu SW, Chen CH, Shih TC. Inhibition of Galectin-1 and Androgen Receptor Axis Enhances Enzalutamide Treatment in Enzalutamide Resistant Prostate Cancer. Cancers (Basel) 2025; 17:351. [PMID: 39941722 PMCID: PMC11816353 DOI: 10.3390/cancers17030351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 01/16/2025] [Accepted: 01/20/2025] [Indexed: 02/16/2025] Open
Abstract
BACKGROUND/OBJECTIVE Prostate cancer (PCa) remains a prevalent and deadly disease, particularly in its advanced stages. Despite various available treatments, resistance to drugs like enzalutamide continues to present significant challenges. This study aimed to investigate the role of Galectin-1 (Gal-1) in enzalutamide-resistant PCa and assess its potential as a therapeutic target to overcome resistance. METHODS The study utilized specific siRNA-mediated knockdown of Gal-1 in enzalutamide-resistant PCa cells to evaluate its effects on cell proliferation and response to enzalutamide treatment. An orthotopic mouse model was employed to examine the in vivo impact of Gal-1 knockdown. Pharmacological targeting of Gal-1 was conducted using LLS30, and its effects were assessed both in vitro and in vivo. RNA sequencing (RNA-seq) analysis was performed to explore the molecular mechanisms underlying the observed effects. RESULTS The findings demonstrated significant upregulation of Gal-1 in enzalutamide-resistant PCa cells. Gal-1 knockdown inhibited cell proliferation and resensitized resistant cells to enzalutamide treatment in the orthotopic mouse model. Elevated levels of androgen receptor full-length and AR-V7 are key mechanisms under-lying resistance to enzalutamide in PCa. Gal-1 knockdown suppressed AR and AR-V7 expression and their transcriptional activity. Treatment with LLS30 significantly suppressed the growth of enzalutamide-resistant PCa cells and exhibited synergistic effects when combined with enzalutamide. Notably, this combination therapy significantly inhibited the growth of enzalutamide-resistant xenografts in vivo. RNA-seq analysis revealed that LLS30 modulates AR and AR-V7 signaling through the inhibition of associated target genes. CONCLUSION These findings highlight Gal-1 as a promising therapeutic target for overcoming enzalutamide resistance in PCa. Targeting the Gal-1/AR/AR-V7 axis with LLS30 presents a novel strategy to enhance enzalutamide efficacy and address drug resistance in advanced PCa.
Collapse
Affiliation(s)
- Hsiao-Chi Wang
- Department of Research and Development, Kibio Inc., Houston, TX 77021, USA
| | - Allen C. Gao
- Department of Urologic Surgery, University of California at Davis, Davis, CA 95718, USA
| | - Roger Xia
- Department of Biomedical Data Science, Stanford University, Stanford, CA 94305, USA
| | - Chun-Te Wu
- Department of Urology, Chang Gung Memorial Hospital, Linko, Taoyuan 333423, Taiwan
| | - Ssu-Wei Hsu
- Divisions of Nephrology and Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of California at Davis, Davis, CA 95616, USA
- Comprehensive Cancer Center, University of California at Davis, Davis, CA 95616, USA
| | - Ching-Hsien Chen
- Divisions of Nephrology and Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of California at Davis, Davis, CA 95616, USA
- Comprehensive Cancer Center, University of California at Davis, Davis, CA 95616, USA
| | - Tsung-Chieh Shih
- Department of Research and Development, Kibio Inc., Houston, TX 77021, USA
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, 2450 Holcombe Boulevard, Houston, TX 77021, USA
| |
Collapse
|
23
|
Bao B, Tian M, Wang X, Yang C, Qu J, Zhou S, Cheng Y, Tong Q, Zheng L. SNORA37/CMTR1/ELAVL1 feedback loop drives gastric cancer progression via facilitating CD44 alternative splicing. J Exp Clin Cancer Res 2025; 44:15. [PMID: 39815331 PMCID: PMC11737211 DOI: 10.1186/s13046-025-03278-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: 10/26/2024] [Accepted: 01/03/2025] [Indexed: 01/18/2025] Open
Abstract
BACKGROUND Emerging evidence shows that small nucleolar RNA (snoRNA), a type of highly conserved non-coding RNA, is involved in tumorigenesis and aggressiveness. However, the roles of snoRNAs in regulating alternative splicing crucial for cancer progression remain elusive. METHODS High-throughput RNA sequencing and comprehensive analysis were performed to identify crucial snoRNAs and downstream alternative splicing events. Biotin-labeled RNA pull-down, mass spectrometry, cross-linking RNA immunoprecipitation, and in vitro binding assays were applied to explore interaction of snoRNAs with protein partners. Alternative splicing and gene expression was observed by real-time quantitative RT-PCR and western blot assays. In vitro and in vivo studies were performed to investigate biological effects of snoRNAs and their protein partners in gastric cancer. Survival analysis was undertaken by using Kaplan-Meier method and log-rank test. RESULTS SNORA37 was identified as an up-regulated snoRNA essential for tumorigenesis and aggressiveness of gastric cancer. Gain- and loss-of-function studies indicated that SNORA37 promoted the growth, invasion, and metastasis of gastric cancer cells in vitro and in vivo. Mechanistically, as an ELAV like RNA binding protein 1 (ELAVL1)-generated snoRNA, SNORA37 directly bound to cap methyltransferase 1 (CMTR1) to facilitate its interaction with ELAVL1, resulting in nuclear retention and activity of ELAVL1 in regulating alternative splicing of CD44. Rescue studies revealed that SNORA37 exerted oncogenic roles in gastric cancer progression via facilitating CMTR1-ELAVL1 interaction. In clinical gastric cancer cases, high levels of SNORA37, CMTR1, ELAVL1, or CD44 were associated with shorter survival and poor outcomes of patients. CONCLUSIONS These results indicated that SNORA37/CMTR1/ELAVL1 feedback loop drives gastric cancer progression via facilitating CD44 alternative splicing.
Collapse
Affiliation(s)
- Banghe Bao
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, Hubei Province, People's Republic of China
| | - Minxiu Tian
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, Hubei Province, People's Republic of China
| | - Xiaojing Wang
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, Hubei Province, People's Republic of China
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, Hubei Province, People's Republic of China
| | - Chunhui Yang
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, Hubei Province, People's Republic of China
| | - Jiaying Qu
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, Hubei Province, People's Republic of China
| | - Shunchen Zhou
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, Hubei Province, People's Republic of China
| | - Yang Cheng
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, Hubei Province, People's Republic of China
| | - Qiangsong Tong
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, Hubei Province, People's Republic of China.
| | - Liduan Zheng
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, Hubei Province, People's Republic of China.
| |
Collapse
|
24
|
Tian X, Fan R. A Novel Targeted Long-read Sequencing Approach Boosts Transcriptomic Profiling. GENOMICS, PROTEOMICS & BIOINFORMATICS 2025; 22:qzae090. [PMID: 39724297 PMCID: PMC11802469 DOI: 10.1093/gpbjnl/qzae090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 12/09/2024] [Accepted: 12/13/2024] [Indexed: 12/28/2024]
Affiliation(s)
- Xiaolong Tian
- Department of Genetics, Yale School of Medicine, New Haven, CT 06520, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA
| | - Rong Fan
- Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA
- Yale Stem Cell Center and Yale Cancer Center, Yale School of Medicine, New Haven, CT 06520, USA
- Department of Pathology, Yale School of Medicine, New Haven, CT 06520, USA
- Human and Translational Immunology, Yale School of Medicine, New Haven, CT 06520, USA
| |
Collapse
|
25
|
Kundu G, Elangovan S. Investigating the Role of Osteopontin (OPN) in the Progression of Breast, Prostate, Renal and Skin Cancers. Biomedicines 2025; 13:173. [PMID: 39857756 PMCID: PMC11762676 DOI: 10.3390/biomedicines13010173] [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: 10/22/2024] [Revised: 12/20/2024] [Accepted: 12/22/2024] [Indexed: 01/27/2025] Open
Abstract
Background/Objectives: Cancer is caused by disruptions in the homeostatic state of normal cells, which results in dysregulation of the cell cycle, and uncontrolled growth and proliferation in affected cells to form tumors. Successful development of tumorous cells proceeds through the activation of pathways promoting cell development and functionality, as well as the suppression of immune signaling pathways; thereby providing these cells with proliferative advantages, which subsequently metastasize into surrounding tissues. These effects are primarily caused by the upregulation of oncogenes, of which SPP1 (secreted phosphoprotein 1), a non-collagenous bone matrix protein, is one of the most well-known. Methods: In this study, we conducted a further examination of the transcriptomic expression profile of SPP1 (Osteopontin) during the progression of cancer in four human tissues, breast, prostate, renal and skin, in order to understand the circumstances conducive to its activation and dysregulation, the biological pathways and other mechanisms involved as well as differences in its splicing patterns influencing its expression and functionality. Results: A significant overexpression of SPP1, as well as a set of other highly correlated genes, was seen in most of these tissues, indicating their extensive implication in cancer. Increased expression was observed with higher tumor stages, especially in renal and skin cancer, while applying therapeutic modalities targeting these genes dampened this effect in breast, prostate and skin cancer. Pathway analyses showed gene signatures related to cell growth and development enriched in tumorigenic conditions and earlier cancer stages, while later stages of cancer showed pathways associated with weakened immune response, in all cancers studied. Moreover, the utilization of therapeutic methods showed the activation of immunogenic pathways in breast, prostate and skin cancer, thereby confirming their viability. Further analyses of differential transcript expression levels in these oncogenes showed their exonic regions to be selectively overexpressed similarly in tumorigenic samples in all cancers studied, while also displaying significant differences in exon selectivity between constituent transcripts, providing a basis for their high degree of multifunctionality in cancer. Conclusions: Overall, this study corroborates the entrenched role of SPP1 in the progression of these four types of cancer, as confirmed by its overexpression and activation of related oncogenes, their co-involvement in key cellular pathways, and predisposition to exhibit differential splicing between their transcripts, while the above effects were found to be highly inhibitable through treatment methods, thereby highlighting its promising role in therapeutic development.
Collapse
Affiliation(s)
| | - Selvakumar Elangovan
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT) Deemed to be University, Bhubaneswar 751024, Odisha, India;
| |
Collapse
|
26
|
Jin G, Song Y, Fang S, Yan M, Yang Z, Shao Y, Zhao K, Liu M, Wang Z, Guo Z, Dong Z. hnRNPU-mediated pathogenic alternative splicing drives gastric cancer progression. J Exp Clin Cancer Res 2025; 44:8. [PMID: 39773744 PMCID: PMC11705778 DOI: 10.1186/s13046-024-03264-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 12/20/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Alternative splicing (AS) is a process that facilitates the differential inclusion of exonic sequences from precursor messenger RNAs, significantly enhancing the diversity of the transcriptome and proteome. In cancer, pathogenic AS events are closely related to cancer progression. This study aims to investigate the role and regulatory mechanisms of AS in gastric cancer (GC). METHODS We analyzed AS events in various tumor samples and identified hnRNPU as a key splicing factor in GC. The effects of hnRNPU on cancer progression were assessed through in vitro and in vivo experiments. Gene knockout models and the FTO inhibitor (meclofenamic acid) were used to validate the interaction between hnRNPU and FTO and their impact on AS. RESULTS We found that hnRNPU serves as a key splicing factor in GC, and its high expression is associated with poor clinical prognosis. Genetic depletion of hnRNPU significantly reduced GC progression. Mechanistically, the m6A demethylase FTO interacts with hnRNPU transcripts, decreasing the m6A modification levels of hnRNPU, which leads to exon 14 skipping of the MET gene, thereby promoting GC progression. The FTO inhibitor meclofenamic acid effectively inhibited GC cell growth both in vitro and in vivo. CONCLUSION The FTO/hnRNPU axis induces aberrant exon skipping of MET, thereby promoting GC cell growth. Targeting the FTO/hnRNPU axis may interfere with abnormal AS events and provide a potential diagnostic and therapeutic strategy for GC.
Collapse
Affiliation(s)
- Guoguo Jin
- Henan Key Laboratory of Chronic Disease Management, Fuwai Central China Cardiovascular Hospital, Zhengzhou, 450000, China.
- China-US (Henan) Hormel Cancer Institute, No. 127, Dongming Road, Jinshui District, Zhengzhou, Henan, China.
- Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, 450046, China.
- Tianjian Laboratory of Advanced Biomedical Sciences, Institute of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, Henan, China.
| | - Yanming Song
- China-US (Henan) Hormel Cancer Institute, No. 127, Dongming Road, Jinshui District, Zhengzhou, Henan, China
| | - Shaobo Fang
- China-US (Henan) Hormel Cancer Institute, No. 127, Dongming Road, Jinshui District, Zhengzhou, Henan, China
- Department of Medical Imaging, Zhengzhou University People's Hospital& Henan Provincial People's Hospital, Zhengzhou, 450000, China
| | - Mingyang Yan
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- China-US (Henan) Hormel Cancer Institute, No. 127, Dongming Road, Jinshui District, Zhengzhou, Henan, China
| | - Zhaojie Yang
- Laboratory of Bone Tumor, Luoyang Orthopedic Hospital of Henan Province (Orthopedic Hospital of Henan Province), Zhengzhou, 450000, China
| | - Yang Shao
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- China-US (Henan) Hormel Cancer Institute, No. 127, Dongming Road, Jinshui District, Zhengzhou, Henan, China
| | - Kexin Zhao
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- China-US (Henan) Hormel Cancer Institute, No. 127, Dongming Road, Jinshui District, Zhengzhou, Henan, China
| | - Meng Liu
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- China-US (Henan) Hormel Cancer Institute, No. 127, Dongming Road, Jinshui District, Zhengzhou, Henan, China
| | - Zhenwei Wang
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- China-US (Henan) Hormel Cancer Institute, No. 127, Dongming Road, Jinshui District, Zhengzhou, Henan, China
| | - Zhiping Guo
- Henan Key Laboratory of Chronic Disease Management, Fuwai Central China Cardiovascular Hospital, Zhengzhou, 450000, China.
- Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, 450046, China.
| | - Zigang Dong
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China.
- China-US (Henan) Hormel Cancer Institute, No. 127, Dongming Road, Jinshui District, Zhengzhou, Henan, China.
- Tianjian Laboratory of Advanced Biomedical Sciences, Institute of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
27
|
Utkina M, Shcherbakova A, Deviatiiarov R, Ryabova A, Loguinova M, Trofimov V, Kuznetsova A, Petropavlovskiy M, Salimkhanov R, Maksimov D, Albert E, Golubeva A, Asaad W, Urusova L, Bondarenko E, Lapshina A, Shutova A, Beltsevich D, Gusev O, Dzeranova L, Melnichenko G, Minniakhmetov I, Dedov I, Mokrysheva N, Popov S. Comparative evaluation of ACetic - MEthanol high salt dissociation approach for single-cell transcriptomics of frozen human tissues. Front Cell Dev Biol 2025; 12:1469955. [PMID: 39839668 PMCID: PMC11748064 DOI: 10.3389/fcell.2024.1469955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 11/20/2024] [Indexed: 01/23/2025] Open
Abstract
Current dissociation methods for solid tissues in scRNA-seq studies do not guarantee intact single-cell isolation, especially for sensitive and complex human endocrine tissues. Most studies rely on enzymatic dissociation of fresh samples or nuclei isolation from frozen samples. Dissociating whole intact cells from fresh-frozen samples, commonly collected by biobanks, remains a challenge. Here, we utilized the acetic-methanol dissociation approach (ACME) to capture transcriptional profiles of individual cells from fresh-frozen tissue samples. This method combines acetic acid-based dissociation and methanol-based fixation. In our study, we optimized this approach for human endocrine tissue samples for the first time. We incorporated a high-salt washing buffer instead of the standard PBS to stabilize RNA and prevent RNases reactivation during rehydration. We have designated this optimized protocol as ACME HS (ACetic acid-MEthanol High Salt). This technique aims to preserve cell morphology and RNA integrity, minimizing transcriptome changes and providing a more accurate representation of mature mRNA. We compared the ability of enzymatic, ACME HS, and nuclei isolation methods to preserve major cell types, gene expression, and standard quality parameters across 41 tissue samples. Our results demonstrated that ACME HS effectively dissociates and fixes cells, preserving cell morphology and high RNA integrity. This makes ACME HS a valuable alternative for scRNA-seq protocols involving challenging tissues where obtaining a live cell suspension is difficult or disruptive.
Collapse
Affiliation(s)
- Marina Utkina
- Endocrinology Research Centre, Institute of Personalized Medicine, Moscow, Russia
| | | | - Ruslan Deviatiiarov
- Endocrinology Research Centre, Institute of Personalized Medicine, Moscow, Russia
- Graduate School of Medicine, Juntendo University, Bunkyo-ku, Japan
- Life Improvement by Future Technologies (LIFT) Center, Moscow, Russia
| | - Alina Ryabova
- Endocrinology Research Centre, Institute of Personalized Medicine, Moscow, Russia
| | - Marina Loguinova
- Endocrinology Research Centre, Institute of Personalized Medicine, Moscow, Russia
| | - Valentin Trofimov
- Endocrinology Research Centre, Institute of Personalized Medicine, Moscow, Russia
| | - Anna Kuznetsova
- Endocrinology Research Centre, Institute of Personalized Medicine, Moscow, Russia
| | | | - Rustam Salimkhanov
- Endocrinology Research Centre, Institute of Personalized Medicine, Moscow, Russia
| | - Denis Maksimov
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Eugene Albert
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Alexandra Golubeva
- Endocrinology Research Centre, Institute of Personalized Medicine, Moscow, Russia
| | - Walaa Asaad
- Endocrinology Research Centre, Institute of Personalized Medicine, Moscow, Russia
| | - Lilia Urusova
- Endocrinology Research Centre, Institute of Personalized Medicine, Moscow, Russia
| | - Ekaterina Bondarenko
- Endocrinology Research Centre, Institute of Personalized Medicine, Moscow, Russia
| | - Anastasia Lapshina
- Endocrinology Research Centre, Institute of Personalized Medicine, Moscow, Russia
| | - Alexandra Shutova
- Endocrinology Research Centre, Institute of Personalized Medicine, Moscow, Russia
| | - Dmitry Beltsevich
- Endocrinology Research Centre, Institute of Personalized Medicine, Moscow, Russia
| | - Oleg Gusev
- Endocrinology Research Centre, Institute of Personalized Medicine, Moscow, Russia
- Graduate School of Medicine, Juntendo University, Bunkyo-ku, Japan
- Life Improvement by Future Technologies (LIFT) Center, Moscow, Russia
- Regulatory Genomics Research Center, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Larisa Dzeranova
- Endocrinology Research Centre, Institute of Personalized Medicine, Moscow, Russia
| | - Galina Melnichenko
- Endocrinology Research Centre, Institute of Personalized Medicine, Moscow, Russia
| | - Ildar Minniakhmetov
- Endocrinology Research Centre, Institute of Personalized Medicine, Moscow, Russia
| | - Ivan Dedov
- Endocrinology Research Centre, Institute of Personalized Medicine, Moscow, Russia
| | - Natalya Mokrysheva
- Endocrinology Research Centre, Institute of Personalized Medicine, Moscow, Russia
| | - Sergey Popov
- Endocrinology Research Centre, Institute of Personalized Medicine, Moscow, Russia
| |
Collapse
|
28
|
Yang Y, Kumar H, Xie Y, Li Z, Li R, Chen W, Diala C, Ali MA, Xu Y, Wu A, Hosseini SR, Bi E, Zhao H, Kim P, Zheng W. ASpdb: an integrative knowledgebase of human protein isoforms from experimental and AI-predicted structures. Nucleic Acids Res 2025; 53:D331-D339. [PMID: 39530217 PMCID: PMC11701669 DOI: 10.1093/nar/gkae1018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 10/13/2024] [Accepted: 10/16/2024] [Indexed: 11/16/2024] Open
Abstract
Alternative splicing is a crucial cellular process in eukaryotes, enabling the generation of multiple protein isoforms with diverse functions from a single gene. To better understand the impact of alternative splicing on protein structures, protein-protein interaction and human diseases, we developed ASpdb (https://biodataai.uth.edu/ASpdb/), a comprehensive database integrating experimentally determined structures and AlphaFold 2-predicted models for human protein isoforms. ASpdb includes over 3400 canonical isoforms, each represented by both experimentally resolved and predicted structures, and >7200 alternative isoforms with AlphaFold 2 predictions. In addition to detailed splicing events, 3D structures, sequence variations and functional annotations, ASpdb uniquely offers comparative analyses and visualization of structural alterations among isoforms. This resource is invaluable for advancing research in alternative splicing, structural biology and disease mechanisms.
Collapse
Affiliation(s)
- Yuntao Yang
- McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, 7000 Fannin Street, Suite 600, Houston, TX 77030, USA
| | - Himansu Kumar
- McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, 7000 Fannin Street, Suite 600, Houston, TX 77030, USA
| | - Yuhan Xie
- Department of Biostatistics, Yale University School of Public Health, 300 George Street, Set 503, New Haven, CT 06511, USA
| | - Zhao Li
- McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, 7000 Fannin Street, Suite 600, Houston, TX 77030, USA
| | - Rongbin Li
- McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, 7000 Fannin Street, Suite 600, Houston, TX 77030, USA
| | - Wenbo Chen
- McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, 7000 Fannin Street, Suite 600, Houston, TX 77030, USA
| | - Chiamaka S Diala
- McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, 7000 Fannin Street, Suite 600, Houston, TX 77030, USA
| | - Meer A Ali
- McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, 7000 Fannin Street, Suite 600, Houston, TX 77030, USA
| | - Yi Xu
- McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, 7000 Fannin Street, Suite 600, Houston, TX 77030, USA
| | - Albon Wu
- Department of Computer Science and Engineering, University of Michigan, 2260 Hayward Street, Ann Arbor, MI 48109-2121, USA
| | - Sayed-Rzgar Hosseini
- McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, 7000 Fannin Street, Suite 600, Houston, TX 77030, USA
| | - Erfei Bi
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Room 1156, BRB II/III, 421 Curie Boulevard, Philadelphia, PA 19104-6058, USA
| | - Hongyu Zhao
- Department of Biostatistics, Yale University School of Public Health, 300 George Street, Set 503, New Haven, CT 06511, USA
| | - Pora Kim
- McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, 7000 Fannin Street, Suite 600, Houston, TX 77030, USA
| | - W Jim Zheng
- McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, 7000 Fannin Street, Suite 600, Houston, TX 77030, USA
| |
Collapse
|
29
|
Sun C, Zeng B, Zhou J, Li N, Li M, Zhu C, Xie S, Wang Y, Wang S, Wang X. Analysis of SLC genes alternative splicing identifies the SLC7A6 RI isoform as a therapeutic target for colorectal cancer. Cancer Sci 2025; 116:233-247. [PMID: 39403788 PMCID: PMC11711054 DOI: 10.1111/cas.16351] [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: 07/03/2024] [Revised: 08/30/2024] [Accepted: 09/07/2024] [Indexed: 01/11/2025] Open
Abstract
Alternative splicing (AS), a crucial mechanism in post-transcriptional regulation, has been implicated in diverse cancer processes. Several splicing variants of solute carrier (SLC) transporters reportedly play pivotal roles in tumorigenesis and tumor development. However, an in-depth analysis of AS landscapes of SLCs in colon adenocarcinoma (COAD) is lacking. Herein, we analyzed data from The Cancer Genome Atlas and identified 1215 AS events across 243 SLC genes, including 109 differentially expressed AS (DEAS) events involving 62 SLC genes in COAD. Differentially spliced SLCs were enriched in biological processes, including transmembrane transporter activity, transporter activity, ferroptosis, and choline metabolism. In patients with COAD, tumor tissues exhibited higher expression of longer mitochondrial carrier SLC25A16 isoforms than adjacent normal tissues, consistent with bioinformatics analysis. Protein-coding sequences and transmembrane helices of survival-related DEAS were predicted, revealing that shifts in splicing sites altered the number and structure of their transmembrane proteins. We developed a prognostic risk model based on the screened 6-SLC-AS (SLC7A6_RI_37208 (SLC7A6-RI), SLC11A2_AP_21724, SLC2A8_ES_87631, SLC35B1_AA_42317, SLC39A11_AD_43204, and SLC7A8_AP_26712). Knockdown of the intronic region of SLC7A6-RI isoform enhanced colon cancer cell proliferation. In vivo, knockdown of the intronic region of SLC7A6-RI isoform enhanced tumor growth in colon cancer. Mechanistically, si-SLC7A6-RI isoform exerted oncogenic effects by activating the PI3K-Akt-mTOR signaling pathway and promoting cell proliferation, evidenced by increased expression of key regulators Phosphorylated Mammalian Target of Rapamycin (p-mTOR) and a cell proliferation marker Proliferating Cell Nuclear Antigen (PCNA) using western blotting. Our study elucidated SLC-AS in COAD, highlighting its potential as a prognostic and therapeutic target and emphasizing the suppressive influence of SLC7A6-RI in colon cancer progression.
Collapse
Affiliation(s)
- Chao Sun
- Department of Pharmacy, Shenzhen People's HospitalThe Second Clinical Medical College, Jinan UniversityShenzhenChina
- Integrated Chinese and Western Medicine Postdoctoral Research StationJinan UniversityGuangzhouChina
| | - Boning Zeng
- Department of Pharmacy, Shenzhen People's HospitalThe Second Clinical Medical College, Jinan UniversityShenzhenChina
- Integrated Chinese and Western Medicine Postdoctoral Research StationJinan UniversityGuangzhouChina
| | - Jilong Zhou
- Department of Pharmacy, Shenzhen People's HospitalThe Second Clinical Medical College, Jinan UniversityShenzhenChina
| | - Nan Li
- School of Pharmaceutical SciencesShenzhen University Health Science CenterShenzhenChina
| | - Mingwei Li
- Department of Pharmacy, Shenzhen People's HospitalThe Second Clinical Medical College, Jinan UniversityShenzhenChina
| | - Chaowei Zhu
- Department of Pharmacy, Shenzhen People's HospitalThe Second Clinical Medical College, Jinan UniversityShenzhenChina
| | - Shouxia Xie
- Department of Pharmacy, Shenzhen People's HospitalThe Second Clinical Medical College, Jinan UniversityShenzhenChina
| | - Yifei Wang
- Institute of Biomedicine, College of Life Science and Technology, Guangdong Province Key Laboratory of Bioengineering Medicine, Key Laboratory of Innovative Technology Research on Natural Products and Cosmetics Raw MaterialsJinan UniversityGuangzhouChina
| | - Shaoxiang Wang
- School of Pharmaceutical SciencesShenzhen University Health Science CenterShenzhenChina
| | - Xiao Wang
- Department of Pharmacy, Shenzhen People's HospitalThe Second Clinical Medical College, Jinan UniversityShenzhenChina
| |
Collapse
|
30
|
Liu GL, Luo H, Liang DD, Zhong L, Dai N, Lan WH. Comprehensive Analysis of Prognostic Alternative Splicing Signatures in Tumor Immune Infiltration in Bladder Cancer. Recent Pat Anticancer Drug Discov 2025; 20:185-199. [PMID: 39473202 DOI: 10.2174/0115748928329276241020184935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 09/30/2024] [Accepted: 10/03/2024] [Indexed: 04/24/2025]
Abstract
BACKGROUND Bladder cancer exhibits substantial heterogeneity encompassing genetic expressions and histological features. This heterogeneity is predominantly attributed to alternative splicing (AS) and AS-regulated splicing factors (SFs), which, in turn, influence bladder cancer development, progression, and response to treatment. OBJECTIVE This study aimed to explore the immune landscape of aberrant AS in bladder cancer and establish the prognostic signatures for survival prediction. METHODS Bladder cancer-related RNA-Seq, transcriptome, and corresponding clinical information were downloaded from The Cancer Genome Atlas (TCGA). Gene set enrichment analysis (GSEA) was used to identify significantly enriched pathways of cancer-related AS events. The underlying interactions among differentially expressed genes (DEGs) and cancer-related AS events were assessed by a protein-protein interaction network. Univariate and multivariate Cox regression analyses were performed to identify crucial prognostic DEGs that co-occurred with cancer-related AS events (DEGAS) for overall survival. The area under the curve (AUC) of receiver operating characteristic (ROC) curves was used to assess the efficiency of the prognostic signatures. The CIBERSORT algorithm was used to explore the abundance of immune infiltrating cells. RESULTS A total of 3755 cancer-related AS events and 3110 DEGs in bladder cancer were identified. Among them, 379 DEGs co-occurred with cancer-related AS events (DEGAS), of which 102 DEGAS were associated with 14 dysregulated SFs. GSEA and KEGG analysis showed that cancer-related AS events were predominantly enriched in pathways related to immunity, tumorigenesis, and treatment difficulties of bladder cancer. Multivariate Cox regression analysis identified 8 DEGAS (CABP1, KCNN2, TNFRSF13B, PCDH7, SNRPA1, APOLD1, CX3CL1, and DENND5A) significantly associated with OS, and they were further integrated into the prediction model with good AUCs at 3-year, 5-year and 7-year ROC curves (all>0.7). Immune infiltration analysis revealed the significant enrichment of three immune cell types (B cells naïve, dendritic cells resting, and dendritic cell activated) in high-risk bladder cancer patients. CONCLUSION This study not only unveiled comprehensive prognostic signatures of AS events in bladder cancer but also established a robust prognostic model based on survival-related DEGAS. These aberrant AS events, dysregulated SFs, and the identified 8 DEGAS may have significant clinical potential as therapeutic targets for bladder cancer.
Collapse
Affiliation(s)
- Gao-Lei Liu
- Department of Urology, Army Medical Center, Chongqing, 400042, China
| | - Hao Luo
- Department of Oncology, Army Medical Center, Chongqing, 400042, China
| | - Dan-Dan Liang
- People's Hospital of Chong Qing Liang Jiang New Area, Chongqing, 400042, China
| | - Li Zhong
- Department of Oncology, Army Medical Center, Chongqing, 400042, China
| | - Nan Dai
- Department of Oncology, Army Medical Center, Chongqing, 400042, China
| | - Wei-Hua Lan
- Department of Urology, Army Medical Center, Chongqing, 400042, China
| |
Collapse
|
31
|
Rosenberg-Mogilevsky A, Siegfried Z, Karni R. Generation of tumor neoantigens by RNA splicing perturbation. Trends Cancer 2025; 11:12-24. [PMID: 39578174 DOI: 10.1016/j.trecan.2024.10.008] [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: 07/19/2024] [Revised: 10/16/2024] [Accepted: 10/23/2024] [Indexed: 11/24/2024]
Abstract
Immunotherapy has revolutionized cancer treatment, but the limited availability of tumor-specific neoantigens still remains a challenge. The potential of alternative mRNA splicing-derived neoantigens as a source of new immunotherapy targets has gained significant attention. Tumors exhibit unique splicing changes and splicing factor mutations which are prevalent in various cancers and play a crucial role in neoantigen production. We present advances in splicing modulation approaches, including small-molecule drugs, decoy and splice-switching antisense oligonucleotides (SSOs), CRISPR, small interfering RNAs (siRNAs), and nonsense-mediated RNA decay (NMD) inhibition, that can be adapted to enhance antitumor immune responses. Finally, we explore the clinical implications of these approaches, highlighting their potential to transform cancer immunotherapy and broaden its efficacy.
Collapse
Affiliation(s)
- Adi Rosenberg-Mogilevsky
- Department of Biochemistry and Molecular Biology, Institute for Medical Research Israel-Canada (IMRIC), Hebrew University and Hadassah Medical School, Jerusalem, Israel
| | - Zahava Siegfried
- Department of Biochemistry and Molecular Biology, Institute for Medical Research Israel-Canada (IMRIC), Hebrew University and Hadassah Medical School, Jerusalem, Israel
| | - Rotem Karni
- Department of Biochemistry and Molecular Biology, Institute for Medical Research Israel-Canada (IMRIC), Hebrew University and Hadassah Medical School, Jerusalem, Israel.
| |
Collapse
|
32
|
Panthong W, Pientong C, Nukpook T, Heawchaiyaphum C, Aromseree S, Ekalaksananan T. OSI-027 as a Potential Drug Candidate Targeting Upregulated Hub Protein TAF1 in Potential Mechanism of Sinonasal Squamous Cell Carcinoma: Insights from Proteomics and Molecular Docking. BIOLOGY 2024; 13:1089. [PMID: 39765756 PMCID: PMC11673211 DOI: 10.3390/biology13121089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 12/18/2024] [Accepted: 12/21/2024] [Indexed: 01/11/2025]
Abstract
Sinonasal squamous cell carcinoma (SNSCC) is a rare tumor with high mortality and recurrence rates. However, SNSCC carcinogenesis mechanisms and potential therapeutic drugs have not been fully elucidated. This study investigated the key molecular mechanisms and hub proteins involved in SNSCC carcinogenesis using proteomics and bioinformatic analysis. Dysregulated proteins were validated by RT-qPCR in SNSCC and nasal polyp (NP) tissues. Proteomic analysis revealed that differentially expressed proteins were clustered using MCODE scores ≥ 4 into three modules. The specific hub proteins in each module were analyzed in carcinogenesis pathways using STRING, highlighting potential mechanisms of histone modification and spliceosome dysregulation. Spliceosome components SNRNP200 and SF3A3 were significantly downregulated in SNSCC by RT-qPCR. Web-based applications L1000CDS2 and iLINCS were applied to identify 10 potential repurposable drugs that could reverse the gene expression pattern associated with SNSCC. Docking studies of TAF1, a protein in histone modification, with these 10 small molecule inhibitors indicated OSI-027 to be the most promising due to its strong binding interactions with key residues. These findings suggest that hub proteins involved in the underlying mechanism of SNSCC carcinogenesis may serve as valuable targets for drug development, with OSI-027 emerging as a novel candidate against TAF1 in SNSCC.
Collapse
Affiliation(s)
- Watcharapong Panthong
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; (W.P.); (T.N.); (C.H.); (S.A.)
- HPV&EBV and Carcinogenesis (HEC) Research Group, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Chamsai Pientong
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; (W.P.); (T.N.); (C.H.); (S.A.)
- HPV&EBV and Carcinogenesis (HEC) Research Group, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Thawaree Nukpook
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; (W.P.); (T.N.); (C.H.); (S.A.)
- HPV&EBV and Carcinogenesis (HEC) Research Group, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Chukkris Heawchaiyaphum
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; (W.P.); (T.N.); (C.H.); (S.A.)
- HPV&EBV and Carcinogenesis (HEC) Research Group, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Sirinart Aromseree
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; (W.P.); (T.N.); (C.H.); (S.A.)
- HPV&EBV and Carcinogenesis (HEC) Research Group, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Tipaya Ekalaksananan
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; (W.P.); (T.N.); (C.H.); (S.A.)
- HPV&EBV and Carcinogenesis (HEC) Research Group, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
| |
Collapse
|
33
|
Cao Y, Wang S, Liu J, Xu J, Liang Y, Ao F, Wei Z, Wang L. CARF regulates the alternative splicing and piwi/piRNA complexes during mouse spermatogenesis through PABPC1. Acta Biochim Biophys Sin (Shanghai) 2024; 57:656-666. [PMID: 39696987 DOI: 10.3724/abbs.2024224] [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] [Indexed: 12/20/2024] Open
Abstract
ADP-ribosylation factor collaborator (CARF), which is also known as CDKN2AIP, was first recognized as an ADP-ribosylation factor-interacting protein that participates in the activation of the ARF-p53-p21 (WAF1) signaling pathway under different conditions, such as oxidative and oncogenic stresses. The activation of this pathway often leads to cell growth arrest and apoptosis as well as senescence. Previous studies revealed that CARF, an RNA-binding protein, is critical for maintaining stem cell pluripotency and somatic differentiation. Nevertheless, its involvement in spermatogenesis has not been well examined. In this study, we show that male mice deficient in Carf expression present impaired spermatogenesis and fertility. IP-MS and RNA-seq analyses reveal that CARF/ Carf interacts with multiple key splicing factors, such as PABPC1, and directly targets 356 different types of mRNAs in spermatocytes. Carf-associated mRNAs display aberrant splicing patterns when Carf expression is deficient. In addition, our results demonstrate that PIWIL1 expression and localization are altered in the Carf -/ - mouse model through the downregulation of PABPC1, which further affects the ratio of pachytene-piRNA. Our study suggests that CARF is critical for regulating alternative splicing in mammalian spermatogenesis and determining infertility in male mice.
Collapse
|
34
|
Pallasaho S, Gondane A, Kutz J, Liang J, Yalala S, Duveau DY, Pospiech H, Thomas CJ, Loda M, Itkonen HM. Compromised CDK12 activity causes dependency on the high activity of O-GlcNAc transferase. Glycobiology 2024; 34:cwae081. [PMID: 39361894 PMCID: PMC11632362 DOI: 10.1093/glycob/cwae081] [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: 04/01/2024] [Revised: 08/19/2024] [Accepted: 10/01/2024] [Indexed: 10/05/2024] Open
Abstract
O-GlcNAc transferase (OGT) coordinates with regulators of transcription, including cyclin-dependent kinase 12 (CDK12), the major transcription elongation kinase. Here, we use inhibitor- and knockdown-based strategies to show that co-targeting of OGT and CDK12 is toxic to prostate cancer cells. OGT catalyzes all nucleocytoplasmic O-GlcNAcylation and due to its essentiality in higher eukaryotes, it is not an ideal drug target. Our glycoproteomics-data revealed that short-term CDK12 inhibition induces hyper-O-GlcNAcylation of the spliceosome-machinery in different models of prostate cancer. By integrating our glycoproteomics-, gene essentiality- and clinical-data from CDK12 mutant prostate cancer patients, we identify the non-essential serine-arginine protein kinase 1 (SRPK1) as a synthetic lethal partner with CDK12-inactivation. Both normal and cancer cells become highly sensitive against inhibitors of OGT and SRPK1 if they have lowered activity of CDK12. Inactivating mutations in CDK12 are enriched in aggressive prostate cancer, and we propose that these patients would benefit from therapy targeting the spliceosome.
Collapse
Affiliation(s)
- Satu Pallasaho
- Department of Biochemistry and Developmental Biology, Faculty of Medicine, University of Helsinki, Helsinki 00014, Finland
| | - Aishwarya Gondane
- Department of Biochemistry and Developmental Biology, Faculty of Medicine, University of Helsinki, Helsinki 00014, Finland
| | - Julia Kutz
- Project group Biochemistry, Leibniz Institute on Aging - Fritz Lipmann Institute, Jena D-07745, Germany
| | - Jing Liang
- Department of Biochemistry and Developmental Biology, Faculty of Medicine, University of Helsinki, Helsinki 00014, Finland
| | - Shivani Yalala
- Department of Biochemistry and Developmental Biology, Faculty of Medicine, University of Helsinki, Helsinki 00014, Finland
| | - Damien Y Duveau
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, Rockville, MD 20850, United States
| | - Helmut Pospiech
- Project group Biochemistry, Leibniz Institute on Aging - Fritz Lipmann Institute, Jena D-07745, Germany
- University Hospital and Medical Faculty of the Heinrich-Heine University Düsseldorf, Life Science Center, Düsseldorf D-40225, Germany
| | - Craig J Thomas
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, Rockville, MD 20850, United States
- Lymphoid Malignancies Branch, Center for Cancer Research, NCI, NIH, Bethesda, MD, United States
| | - Massimo Loda
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York-Presbyterian Hospital, New York, New York 10021, United States
- The Broad Institute of Harvard and MIT, Cambridge, MA 02142, United States
- The New York Genome Center, New York, New York 10013, United States
| | - Harri M Itkonen
- Department of Biochemistry and Developmental Biology, Faculty of Medicine, University of Helsinki, Helsinki 00014, Finland
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York-Presbyterian Hospital, New York, New York 10021, United States
| |
Collapse
|
35
|
Denti V, Greco A, Alviano AM, Capitoli G, Monza N, Smith A, Pilla D, Maggioni A, Ivanova M, Venetis K, Maffini F, Garancini M, Pincelli AI, Galimberti S, Magni F, Fusco N, L'Imperio V, Pagni F. Spatially Resolved Molecular Characterization of Noninvasive Follicular Thyroid Neoplasms with Papillary-like Nuclear Features (NIFTPs) Identifies a Distinct Proteomic Signature Associated with RAS-Mutant Lesions. Int J Mol Sci 2024; 25:13115. [PMID: 39684824 DOI: 10.3390/ijms252313115] [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: 10/24/2024] [Revised: 11/23/2024] [Accepted: 12/04/2024] [Indexed: 12/18/2024] Open
Abstract
Follicular-patterned thyroid neoplasms comprise a diverse group of lesions that pose significant challenges in terms of differential diagnosis based solely on morphologic and genetic features. Thus, the identification of easily testable biomarkers complementing microscopic and genetic analyses is a highly anticipated advancement that could improve diagnostic accuracy, particularly for noninvasive follicular thyroid neoplasms with papillary-like nuclear features (NIFTPs). These tumors exhibit considerable morphological and molecular heterogeneity, which may complicate their distinction from structurally similar neoplasms, especially when genetic analyses reveal shared genomic alterations (e.g., RAS mutations). Here, we integrated next-generation sequencing (NGS) with matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI-MSI) to perform a proteogenomic analysis on 85 NIFTPs (n = 30 RAS-mutant [RAS-mut] and n = 55 RAS-wild type [RAS-wt]), with the aim to detect putative biomarkers of RAS-mut lesions. Through this combined approach, we identified four proteins that were significantly underexpressed in RAS-mut as compared to RAS-wt NIFTPs. These proteins could serve as readily accessible markers in morphologically borderline cases showing RAS mutations. Additionally, our findings may provide insights into the distinct pathogenic pathways through which RAS-mut and RAS-wt NIFTPs arise, highlighting the pivotal role of constitutive RAS-mitogen-activated protein kinase (MAPK) pathway activation in the development and progression of RAS-mut tumors.
Collapse
Affiliation(s)
- Vanna Denti
- Proteomics and Metabolomics Unit, Department of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Angela Greco
- Department of Medicine and Surgery, Pathology, Center of Digital Medicine, University of Milano-Bicocca, Fondazione IRCCS San Gerardo dei Tintori, Via Cadore 48, 20900 Monza, Italy
| | - Antonio Maria Alviano
- Department of Medicine and Surgery, Pathology, Center of Digital Medicine, University of Milano-Bicocca, Fondazione IRCCS San Gerardo dei Tintori, Via Cadore 48, 20900 Monza, Italy
| | - Giulia Capitoli
- Bicocca Bioinformatics Biostatistics and Bioimaging Research Centre-B4, Department of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
- Biostatistics and Clinical Epidemiology, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy
| | - Nicole Monza
- Proteomics and Metabolomics Unit, Department of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Andrew Smith
- Proteomics and Metabolomics Unit, Department of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Daniela Pilla
- Department of Medicine and Surgery, Pathology, Center of Digital Medicine, University of Milano-Bicocca, Fondazione IRCCS San Gerardo dei Tintori, Via Cadore 48, 20900 Monza, Italy
| | - Alice Maggioni
- Department of Medicine and Surgery, Pathology, Center of Digital Medicine, University of Milano-Bicocca, Fondazione IRCCS San Gerardo dei Tintori, Via Cadore 48, 20900 Monza, Italy
| | - Mariia Ivanova
- Department of Pathology and Laboratory Medicine, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Konstantinos Venetis
- Department of Pathology and Laboratory Medicine, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Fausto Maffini
- Department of Pathology and Laboratory Medicine, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | | | | | - Stefania Galimberti
- Bicocca Bioinformatics Biostatistics and Bioimaging Research Centre-B4, Department of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
- Biostatistics and Clinical Epidemiology, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy
| | - Fulvio Magni
- Proteomics and Metabolomics Unit, Department of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Nicola Fusco
- Department of Pathology and Laboratory Medicine, European Institute of Oncology IRCCS, 20141 Milan, Italy
- Department of Oncology & Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Vincenzo L'Imperio
- Department of Medicine and Surgery, Pathology, Center of Digital Medicine, University of Milano-Bicocca, Fondazione IRCCS San Gerardo dei Tintori, Via Cadore 48, 20900 Monza, Italy
| | - Fabio Pagni
- Department of Medicine and Surgery, Pathology, Center of Digital Medicine, University of Milano-Bicocca, Fondazione IRCCS San Gerardo dei Tintori, Via Cadore 48, 20900 Monza, Italy
| |
Collapse
|
36
|
Ogawa T, Isik M, Wu Z, Kurmi K, Meng J, Cho S, Lee G, Fernandez-Cardenas LP, Mizunuma M, Blenis J, Haigis MC, Blackwell TK. Nutrient control of growth and metabolism through mTORC1 regulation of mRNA splicing. Mol Cell 2024; 84:4558-4575.e8. [PMID: 39571580 DOI: 10.1016/j.molcel.2024.10.037] [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: 04/11/2023] [Revised: 07/30/2024] [Accepted: 10/28/2024] [Indexed: 12/08/2024]
Abstract
Cellular growth and organismal development are remarkably complex processes that require the nutrient-responsive kinase mechanistic target of rapamycin complex 1 (mTORC1). Anticipating that important mTORC1 functions remained to be identified, we employed genetic and bioinformatic screening in C. elegans to uncover mechanisms of mTORC1 action. Here, we show that during larval growth, nutrients induce an extensive reprogramming of gene expression and alternative mRNA splicing by acting through mTORC1. mTORC1 regulates mRNA splicing and the production of protein-coding mRNA isoforms largely independently of its target p70 S6 kinase (S6K) by increasing the activity of the serine/arginine-rich (SR) protein RSP-6 (SRSF3/7) and other splicing factors. mTORC1-mediated mRNA splicing regulation is critical for growth; mediates nutrient control of mechanisms that include energy, nucleotide, amino acid, and other metabolic pathways; and may be conserved in humans. Although mTORC1 inhibition delays aging, mTORC1-induced mRNA splicing promotes longevity, suggesting that when mTORC1 is inhibited, enhancement of this splicing might provide additional anti-aging benefits.
Collapse
Affiliation(s)
- Takafumi Ogawa
- Research Division, Joslin Diabetes Center, Boston, MA 02215, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Unit of Biotechnology, Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Japan; Hiroshima Research Center for Healthy Aging (HiHA), Hiroshima University, Higashi-Hiroshima, Japan
| | - Meltem Isik
- Research Division, Joslin Diabetes Center, Boston, MA 02215, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Ziyun Wu
- Research Division, Joslin Diabetes Center, Boston, MA 02215, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Department of Food Science and Engineering, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Kiran Kurmi
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Ludwig Center, Harvard Medical School, Boston, MA 02115, USA
| | - Jin Meng
- Research Division, Joslin Diabetes Center, Boston, MA 02215, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Sungyun Cho
- Meyer Cancer Center and Department of Pharmacology, Weill Cornell Medicine, New York, NY 10021, USA
| | - Gina Lee
- Meyer Cancer Center and Department of Pharmacology, Weill Cornell Medicine, New York, NY 10021, USA; Department of Microbiology and Molecular Genetics, Chao Family Comprehensive Cancer Center, School of Medicine, University of California Irvine, Irvine, CA 92617, USA
| | - L Paulette Fernandez-Cardenas
- Research Division, Joslin Diabetes Center, Boston, MA 02215, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Masaki Mizunuma
- Unit of Biotechnology, Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Japan; Hiroshima Research Center for Healthy Aging (HiHA), Hiroshima University, Higashi-Hiroshima, Japan
| | - John Blenis
- Meyer Cancer Center and Department of Pharmacology, Weill Cornell Medicine, New York, NY 10021, USA
| | - Marcia C Haigis
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Ludwig Center, Harvard Medical School, Boston, MA 02115, USA
| | - T Keith Blackwell
- Research Division, Joslin Diabetes Center, Boston, MA 02215, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA.
| |
Collapse
|
37
|
Debeljak M, Cho S, Downs BM, Considine M, Avin-McKelvey B, Wang Y, Perez PN, Grizzle WE, Hoadley KA, Lynch CF, Hernandez BY, van Diest PJ, Cozen W, Hamilton AS, Hawes D, Gabrielson E, Cimino-Mathews A, Florea LD, Cope L, Umbricht CB. Multimodal genome-wide survey of progressing and non-progressing breast ductal carcinoma in-situ. Breast Cancer Res 2024; 26:178. [PMID: 39633428 PMCID: PMC11616160 DOI: 10.1186/s13058-024-01927-1] [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: 08/16/2024] [Accepted: 11/18/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND Ductal carcinoma in-situ (DCIS) is a pre-invasive form of invasive breast cancer (IBC). Due to improved breast cancer screening, it now accounts for ~ 25% of all breast cancers. While the treatment success rates are over 90%, this comes at the cost of considerable morbidity, considering that the majority of DCIS never become invasive and our understanding of the molecular changes occurring in DCIS that predispose to invasive disease is limited. The aim of this study is to characterize molecular changes that occur in DCIS, with the goal of improving DCIS risk stratification. METHODS We identified and obtained a total of 197 breast tissue samples from 5 institutions (93 DCIS progressors, 93 DCIS non-progressors, and 11 adjacent normal breast tissues) that had at least 10-year follow-up. We isolated DNA and RNA from archival tissue blocks and characterized genome-wide mRNA expression, DNA methylation, DNA copy number variation, and RNA splicing variation. RESULTS We obtained all four genomic data sets in 122 of the 197 samples. Our intrinsic expression subtype-stratified analyses identified multiple molecular differences both between DCIS subtypes and between DCIS and IBC. While there was heterogeneity in molecular signatures and outcomes within intrinsic subtypes, several gene sets that differed significantly between progressing and non-progressing DCIS were identified by Gene Set Enrichment Analysis. CONCLUSION DCIS is a molecularly highly heterogenous disease with variable outcomes, and the molecular events determining DCIS disease progression remain poorly defined. Our genome-wide multi-omic survey documents DCIS-associated alterations and reveals molecular heterogeneity within the intrinsic DCIS subtypes. Further studies investigating intrinsic subtype-stratified characteristics and molecular signatures are needed to determine if these may be exploitable for risk assessment and mitigation of DCIS progression. The highly significant associations of specific gene sets with IBC progression revealed by our Gene Set Enrichment Analysis may lend themselves to the development of a prognostic molecular score, to be validated on independent DCIS cohorts.
Collapse
Affiliation(s)
- Marija Debeljak
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Soonweng Cho
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Bradley M Downs
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA
| | - Michael Considine
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Yongchun Wang
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Phillip N Perez
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - William E Grizzle
- Department of Pathology, University of Alabama at Birmingham School of Medicine, Birmingham, AL, USA
| | - Katherine A Hoadley
- Department of Genetics, Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Charles F Lynch
- Department of Epidemiology, College of Public Health, University of Iowa, Iowa City, IA, USA
| | - Brenda Y Hernandez
- Population Sciences in the Pacific-Program, University of Hawaii Cancer Research Center, Honolulu, HI, USA
| | - Paul J van Diest
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Wendy Cozen
- Department of Medicine, School of Medicine, Susan and Henry Samueli College of Health Sciences, University of California at Irvine, Irvine, CA, USA
| | - Ann S Hamilton
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Debra Hawes
- Department of Pathology and Laboratory Medicine, Keck School of Medicine, Children's Hospital Los Angeles, University of Southern California, Los Angeles, CA, USA
| | - Edward Gabrielson
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ashley Cimino-Mathews
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Liliana D Florea
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Leslie Cope
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Biostatistics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Christopher B Umbricht
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- The Johns Hopkins University School of Medicine, Ross Building, Room 743, 720 Rutland Ave, Baltimore, MD, 21205, USA.
| |
Collapse
|
38
|
Li J, Qiu H, Dong Q, Yu H, Piao C, Li Z, Sun Y, Cui X. Androgen-targeted hsa_circ_0085121 encodes a novel protein and improves the development of prostate cancer through facilitating the activity of PI3K/Akt/mTOR pathway and enhancing AR-V7 alternative splicing. Cell Death Dis 2024; 15:848. [PMID: 39567496 PMCID: PMC11579034 DOI: 10.1038/s41419-024-07246-9] [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: 07/08/2024] [Revised: 11/10/2024] [Accepted: 11/12/2024] [Indexed: 11/22/2024]
Abstract
Prostate cancer (PCa) is the most prevalent type of cancer and the second leading cause of mortality in males, with a marked increase in incidence observed across the globe. In the present study, whole-transcriptome analysis was conducted to identify differentially expressed circular RNAs (DE-circRNAs). The coding abilities of the DE-circRNAs were analyses, and it was found that hsa_circ_0085121 (circRNF19A) not only exhibited overexpression in PCa cells and tumor samples, but also encoded a 490 amino acid polypeptide designated circRNF19A-490aa. The knockdown of circRNF19A was observed to notably inhibit the proliferation, invasion, migration and docetaxel resistance of PCa cells. In contrast, mutation of the IRES significantly impaired the tumor-promoting function of circRNF19A, indicating that circRNF19A-490aa is the primary form that regulates the malignant behaviors of PCa cells. Mechanistically, circRNF19A-490aa was demonstrated to interact with HSP90AA1, thereby enhancing AR activity and facilitating the activation of the Akt/mTOR and PLK1 pathways. Furthermore, circRNF19A-490aa was observed to interact with HNRNPF, facilitating the recruitment of HNRNPF to the splicing site of AR-V7 and enhancing its alternative splicing. Finally, the androgen receptor (AR) was observed to bind to the promoter region of the RNF19A gene, subsequently regulating the expression of circRNF19A and circRNF19A-490aa. These data indicate that circRNF19A plays a pivotal role in AR activation and AR-V7 generation by encoding a novel protein, circRNF19A-490aa, and targeting circRNF19A may prove an effective strategy for impeding the progression of CRPC.
Collapse
Affiliation(s)
- Jianfeng Li
- Department of Urology, First Hospital of China Medical University, #155 Nanjing North Road, 110001, Shenyang, China
| | - Hui Qiu
- Department of Gynecology and Obstetrics, Shengjing Hospital of China Medical University, #36 Sanhao Street, 110004, Shenyang, China
| | - Qingzhuo Dong
- Department of Urology, First Hospital of China Medical University, #155 Nanjing North Road, 110001, Shenyang, China
| | - Hongyuan Yu
- Department of Urology, First Hospital of China Medical University, #155 Nanjing North Road, 110001, Shenyang, China
| | - Chiyuan Piao
- Department of Urology, First Hospital of China Medical University, #155 Nanjing North Road, 110001, Shenyang, China
| | - Zhengxiu Li
- Department of Dermatology, First Hospital of China Medical University, #155 Nanjing North Road, 110001, Shenyang, China
| | - Yanbin Sun
- Department of Thoracic Surgery, First Hospital of China Medical University, #155 Nanjing North Road, 110001, Shenyang, China.
| | - Xiaolu Cui
- Department of Urology, First Hospital of China Medical University, #155 Nanjing North Road, 110001, Shenyang, China.
| |
Collapse
|
39
|
Ryzhkova A, Maltseva E, Battulin N, Kabirova E. Loop Extrusion Machinery Impairments in Models and Disease. Cells 2024; 13:1896. [PMID: 39594644 PMCID: PMC11592926 DOI: 10.3390/cells13221896] [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: 10/22/2024] [Revised: 11/15/2024] [Accepted: 11/15/2024] [Indexed: 11/28/2024] Open
Abstract
Structural maintenance of chromosomes (SMC) complexes play a crucial role in organizing the three-dimensional structure of chromatin, facilitating key processes such as gene regulation, DNA repair, and chromosome segregation. This review explores the molecular mechanisms and biological significance of SMC-mediated loop extrusion complexes, including cohesin, condensins, and SMC5/6, focusing on their structure, their dynamic function during the cell cycle, and their impact on chromatin architecture. We discuss the implications of impairments in loop extrusion machinery as observed in experimental models and human diseases. Mutations affecting these complexes are linked to various developmental disorders and cancer, highlighting their importance in genome stability and transcriptional regulation. Advances in model systems and genomic techniques have provided deeper insights into the pathological roles of SMC complex dysfunction, offering potential therapeutic avenues for associated diseases.
Collapse
Affiliation(s)
- Anastasiya Ryzhkova
- Institute of Cytology and Genetics, 630090 Novosibirsk, Russia; (A.R.); (N.B.)
| | - Ekaterina Maltseva
- Department of Genetics and Genetic Technologies, Sirius University of Science and Technology, 354340 Sirius, Russia;
| | - Nariman Battulin
- Institute of Cytology and Genetics, 630090 Novosibirsk, Russia; (A.R.); (N.B.)
- Department of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Evelyn Kabirova
- Institute of Cytology and Genetics, 630090 Novosibirsk, Russia; (A.R.); (N.B.)
- Department of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| |
Collapse
|
40
|
Zhang J, Ni Z, Zhang Y, Guo Y, Zhai R, Wang M, Gong Z, Wang M, Zeng F, Gu Z, Chen Q, Liu L, Wang Z, Zhu W. DAZAP1 Phase Separation Regulates Mitochondrial Metabolism to Facilitate Invasion and Metastasis of Oral Squamous Cell Carcinoma. Cancer Res 2024; 84:3818-3833. [PMID: 39120588 DOI: 10.1158/0008-5472.can-24-0067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 06/21/2024] [Accepted: 08/02/2024] [Indexed: 08/10/2024]
Abstract
Tumor invasion and metastasis are the underlying causes of high mortality rate due to oral squamous cell carcinoma (OSCC). Energy metabolism reprogramming has been identified as a crucial process mediating tumor metastasis, thus indicating an urgent need for an in-depth investigation of the specific mechanisms of tumor energy metabolism. Here, we identified an RNA-binding protein, DAZ-associated protein 1 (DAZAP1), as a tumor-promoting factor with an important role in OSCC progression. DAZAP1 was significantly upregulated in OSCC, which enhanced the migration and invasion of OSCC cells and induced the epithelial-mesenchymal transition (EMT). RNA sequencing analysis and experimental validation demonstrated that DAZAP1 regulates mitochondrial energy metabolism in OSCC. Mechanistically, DAZAP1 underwent liquid-liquid phase separation to accumulate in the nucleus where it enhanced cytochrome c oxidase 16 (COX16) expression by regulating pre-mRNA alternative splicing, thereby promoting OSCC invasion and mitochondrial respiration. In mouse OSCC models, loss of DAZAP1 suppressed EMT, downregulated COX16, and reduced tumor growth and metastasis. In samples from patients with OSCC, expression of DAZAP1 positively correlated with COX16 and a high expression of both proteins was associated with poor patient prognosis. Together, these findings revealed a mechanism by which DAZAP1 supports mitochondrial metabolism and tumor development of OSCC, suggesting the potential of therapeutic strategies targeting DAZAP1 to block OSCC invasion and metastasis. Significance: The RNA-binding protein DAZAP1 undergoes phase separation to enhance COX16 expression and mediate metabolic reprogramming that enables tumor metastasis, highlighting DAZAP1 as a potential metabolic target for cancer therapy.
Collapse
Affiliation(s)
- Jiayi Zhang
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, Nanjing, China
- Department of Basic Science of Stomatology, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Zihui Ni
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, Nanjing, China
- Department of Basic Science of Stomatology, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Yu Zhang
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, Nanjing, China
- Department of Basic Science of Stomatology, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Yan Guo
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, Nanjing, China
- Department of Basic Science of Stomatology, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Rundong Zhai
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, Nanjing, China
- Department of Basic Science of Stomatology, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Mengqi Wang
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, Nanjing, China
- Department of Basic Science of Stomatology, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Zizhen Gong
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, Nanjing, China
- Department of Basic Science of Stomatology, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Mengyao Wang
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, Nanjing, China
- Department of Basic Science of Stomatology, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Fanrui Zeng
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, Nanjing, China
- Department of Basic Science of Stomatology, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Ziyue Gu
- Department of Oral and Maxillofacial-Head Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qianming Chen
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - Laikui Liu
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, Nanjing, China
- Department of Basic Science of Stomatology, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Zhiyong Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - Weiwen Zhu
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, Nanjing, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, China
- Department of Oral Mucosal Diseases, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
41
|
Yang S, Ko M, Hur SC, Lee EK, Jeong SM. SF3B4 Regulates Cellular Senescence and Suppresses Therapy-induced Senescence of Cancer Cells. Cancer Genomics Proteomics 2024; 21:622-629. [PMID: 39467623 PMCID: PMC11534033 DOI: 10.21873/cgp.20478] [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: 07/29/2024] [Revised: 08/20/2024] [Accepted: 08/29/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND/AIM Cellular senescence is a state in which cells permanently exit the cell cycle, preventing tumor growth, but it can also contribute to aging and chronic inflammation. Senescence induced by cancer therapies, known as therapy-induced senescence (TIS), halts cancer cell proliferation and prevents metastasis. TIS has been investigated as an important therapeutic approach that could minimize cytotoxicity effects. This study aimed to elucidate the role of splicing factor 3B subunit 4 (SF3B4) in cellular senescence and TIS in cancer cells. MATERIALS AND METHODS β-galactosidase staining was used to examine senescence induction. SF3B4 and p21 expression were determined by RT-qPCR and western blot. Cell proliferation and cell death were evaluated. RESULTS SF3B4 expression decreases in replicative senescent human fibroblasts and its knockdown induces senescence via a p21-dependent pathway. In A549 non-small cell lung cancer (NSCLC) cells, SF3B4 knockdown also increased senescence markers. Notably, SF3B4 overexpression mitigated doxorubicin-induced senescence in A549 cells. CONCLUSION SF3B4 regulates senescence, and this study highlights its potential as a therapeutic target for developing better cancer treatment strategies by leveraging TIS to suppress tumor growth and enhance treatment efficacy.
Collapse
Affiliation(s)
- Seungyeon Yang
- Department of Biochemistry, Institute for Aging and Metabolic Diseases, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Department of Medical Sciences, Graduate School of The Catholic University of Korea, Seoul, Republic of Korea
| | - Minbeom Ko
- Department of Biochemistry, Institute for Aging and Metabolic Diseases, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Department of Medical Sciences, Graduate School of The Catholic University of Korea, Seoul, Republic of Korea
| | - Soojung Claire Hur
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, U.S.A
| | - Eun Kyung Lee
- Department of Biochemistry, Institute for Aging and Metabolic Diseases, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Department of Medical Sciences, Graduate School of The Catholic University of Korea, Seoul, Republic of Korea
| | - Seung Min Jeong
- Department of Biochemistry, Institute for Aging and Metabolic Diseases, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Department of Medical Sciences, Graduate School of The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
42
|
Gupta I, Gaykalova DA. Unveiling the role of PIK3R1 in cancer: A comprehensive review of regulatory signaling and therapeutic implications. Semin Cancer Biol 2024; 106-107:58-86. [PMID: 39197810 DOI: 10.1016/j.semcancer.2024.08.004] [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: 05/07/2024] [Revised: 07/11/2024] [Accepted: 08/20/2024] [Indexed: 09/01/2024]
Abstract
Phosphoinositide 3-kinase (PI3K) is responsible for phosphorylating phosphoinositides to generate secondary signaling molecules crucial for regulating various cellular processes, including cell growth, survival, and metabolism. The PI3K is a heterodimeric enzyme complex comprising of a catalytic subunit (p110α, p110β, or p110δ) and a regulatory subunit (p85). The binding of the regulatory subunit, p85, with the catalytic subunit, p110, forms an integral component of the PI3K enzyme. PIK3R1 (phosphoinositide-3-kinase regulatory subunit 1) belongs to class IA of the PI3K family. PIK3R1 exhibits structural complexity due to alternative splicing, giving rise to distinct isoforms, prominently p85α and p55α. While the primary p85α isoform comprises multiple domains, including Src homology 3 (SH3) domains, a Breakpoint Cluster Region Homology (BH) domain, and Src homology 2 (SH2) domains (iSH2 and nSH2), the shorter isoform, p55α, lacks certain domains present in p85α. In this review, we will highlight the intricate regulatory mechanisms governing PI3K signaling along with the impact of PIK3R1 alterations on cellular processes. We will further delve into the clinical significance of PIK3R1 mutations in various cancer types and their implications for prognosis and treatment outcomes. Additionally, we will discuss the evolving landscape of targeted therapies aimed at modulating PI3K-associated pathways. Overall, this review will provide insights into the dynamic interplay of PIK3R1 in cancer, fostering advancements in precision medicine and the development of targeted interventions.
Collapse
Affiliation(s)
- Ishita Gupta
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA; Department of Otorhinolaryngology-Head and Neck Surgery, Marlene & Stewart Greenebaum Comprehensive Cancer Center, University of Maryland Medical Center, Baltimore, MD, USA
| | - Daria A Gaykalova
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA; Department of Otorhinolaryngology-Head and Neck Surgery, Marlene & Stewart Greenebaum Comprehensive Cancer Center, University of Maryland Medical Center, Baltimore, MD, USA; Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
43
|
Chang YH, Head ST, Harrison T, Yu Y, Huff CD, Pasaniuc B, Lindström S, Bhattacharya A. Isoform-level analyses of 6 cancers uncover extensive genetic risk mechanisms undetected at the gene-level. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.10.29.24316388. [PMID: 39574839 PMCID: PMC11581093 DOI: 10.1101/2024.10.29.24316388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/01/2024]
Abstract
Integrating genome-wide association study (GWAS) and transcriptomic datasets can help identify potential mediators for germline genetic risk of cancer. However, traditional methods have been largely unsuccessful because of an overreliance on total gene expression. These approaches overlook alternative splicing, which can produce multiple isoforms from the same gene, each with potentially different effects on cancer risk. Here, we integrate genetic and multi-tissue isoform-level gene expression data from the Genotype Tissue-Expression Project (GTEx, N = 108-574) with publicly available European-ancestry GWAS summary statistics (all N > 20,000 cases) to identify both isoform- and gene-level risk associations with six cancers (breast, endometrial, colorectal, lung, ovarian, prostate) and six related cancer subtype classifications (N = 12 total). Compared to traditional methods leveraging total gene expression, directly modeling isoform expression through transcriptome-wide association studies (isoTWAS) substantially increases discovery of transcriptomic mechanisms underlying genetic associations. Using the same RNA-seq datasets, isoTWAS identified 164% more significant unique gene associations compared to TWAS (6,163 and 2,336, respectively), with isoTWAS-prioritized genes enriched 4-fold for evolutionarily-constrained genes (P = 6.1 × 10-13). isoTWAS tags transcriptomic associations at 52% more independent GWAS loci compared to TWAS across the six cancers. Additionally, isoform expression mediates an estimated 63% greater proportion of cancer risk SNP heritability compared to gene expression when evaluating cis-genetic influence on isoform expression. We highlight several notable isoTWAS associations that demonstrate GWAS colocalization at the isoform level but not at the gene level, including, CLPTM1L (lung cancer), LAMC1 (colorectal), and BABAM1 (breast). These results underscore the critical importance of modeling isoform-level expression to maximize discovery of genetic risk mechanisms for cancers.
Collapse
Affiliation(s)
- Yung-Han Chang
- Quantitative Sciences Program, The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX, USA
| | - S. Taylor Head
- Department of Epidemiology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Tabitha Harrison
- Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA, USA
| | - Yao Yu
- Department of Epidemiology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Chad D. Huff
- Department of Epidemiology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bogdan Pasaniuc
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sara Lindström
- Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA, USA
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Arjun Bhattacharya
- Department of Epidemiology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Institute for Data Science in Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
44
|
Temaj G, Chichiarelli S, Saha S, Telkoparan-Akillilar P, Nuhii N, Hadziselimovic R, Saso L. Alternative Splicing: A Potential Therapeutic Target in Hematological Malignancies. Hematol Rep 2024; 16:682-697. [PMID: 39584923 PMCID: PMC11587037 DOI: 10.3390/hematolrep16040066] [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: 08/23/2024] [Revised: 10/07/2024] [Accepted: 10/24/2024] [Indexed: 11/26/2024] Open
Abstract
Leukemia represents the most prevalent malignancy in children, constituting 30% of childhood cancer cases, with acute lymphoblastic leukemia (ALL) being particularly heterogeneous. This paper explores the role of alternative splicing in leukemia, highlighting its significance in cancer development and progression. Aberrant splicing is often driven by mutations in splicing-factor genes, which can lead to the production of variant proteins that contribute to oncogenesis. The spliceosome, a complex of small nuclear RNAs and proteins, facilitates RNA splicing, a process critical for generating diverse mRNA and protein products from single genes. Mutations in splicing factors, such as U2AF1, SF3B1, SRSF2, ZRSR2, and HNRNPH1, are frequently observed across various hematological malignancies and are associated with poor prognosis and treatment resistance. This research underscores the necessity of understanding the mechanisms of RNA splicing dysregulation in order to develop targeted therapies to correct these aberrant processes, thereby improving outcomes for patients with leukemia and related disorders.
Collapse
Affiliation(s)
- Gazmend Temaj
- Faculty of Pharmacy, College UBT, 10000 Prishtina, Kosovo;
| | - Silvia Chichiarelli
- Department of Biochemical Sciences “A. Rossi-Fanelli”, Sapienza University of Rome, 00185 Rome, Italy;
| | - Sarmistha Saha
- Department of Biotechnology, Institute of Applied Sciences & Humanities, GLA University, Mathura 00185, Uttar Pradesh, India
| | | | - Nexhibe Nuhii
- Department of Pharmacy, Faculty of Medical Sciences, State University of Tetovo, 1200 Tetovo, North Macedonia;
| | - Rifat Hadziselimovic
- Faculty of Science, University of Sarajevo, 71000 Sarajevo, Bosnia and Herzegovina;
| | - Luciano Saso
- Department of Physiology and Pharmacology “Vittorio Erspamer”, La Sapienza University, 00185 Rome, Italy;
| |
Collapse
|
45
|
Wang Y, Chen F, Qu W, Gong Y, Wang Y, Chen L, Zhou Q, Mo J, Zhang H, Lin L, Bi T, Wang X, Gu J, Li Y, Sui L. Alternative splicing in the genome of HPV and its regulation. Front Cell Infect Microbiol 2024; 14:1443868. [PMID: 39502170 PMCID: PMC11534716 DOI: 10.3389/fcimb.2024.1443868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 09/30/2024] [Indexed: 11/08/2024] Open
Abstract
Persistent infection with high-risk human papillomavirus (HR-HPV) is the main cause of cervical cancer. These chronic infections are characterized by high expression of the HPV E6 and E7 oncogenes and the absence of the L1 and L2 capsid proteins. The regulation of HPV gene expression plays a crucial role in both the viral life cycle and rare oncogenic events. Alternative splicing of HPV mRNA is a key mechanism in post-transcriptional regulation. Through alternative splicing, HPV mRNA is diversified into various splice isoforms with distinct coding potentials, encoding multiple proteins and influencing the expression of HPV genes. The spliced mRNAs derived from a donor splicing site within the E6 ORF and one of the different acceptor sites located in the early mRNA contain E6 truncated mRNAs, named E6*. E6* is one of the extensively studied splicing isoforms. However, the role of E6* proteins in cancer progression remains controversial. Here, we reviewed and compared the alternative splicing events occurring in the genomes of HR-HPV and LR-HPV. Recently, new HPV alternative splicing regulatory proteins have been continuously discovered, and we have updated the regulation of HPV alternative splicing. In addition, we summarized the functions of known splice isoforms from three aspects: anti-tumorigenic, tumorigenic, and other cancer-related functions, including not only E6*, but also E6^E7, E8^E2, and so on. Comprehending their contributions to cancer development enhances insights into the carcinogenic mechanisms of HPV and explores the potential utility of alternative splicing in the diagnosis and treatment of cervical cancer.
Collapse
Affiliation(s)
- Yaping Wang
- Department of Gynecology and Obstetrics, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Fang Chen
- Department of Gynecology and Obstetrics, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Wenjie Qu
- Department of Gynecology and Obstetrics, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Yingxin Gong
- Department of Gynecology and Obstetrics, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Yan Wang
- Department of Gynecology and Obstetrics, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Limei Chen
- Department of Gynecology and Obstetrics, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Qi Zhou
- Department of Gynecology and Obstetrics, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Jiayin Mo
- Department of Gynecology and Obstetrics, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Hongwei Zhang
- Department of Gynecology and Obstetrics, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Lin Lin
- Department of Gynecology and Obstetrics, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Tianyi Bi
- Department of Gynecology and Obstetrics, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Xujie Wang
- Department of Obstetrics and Gynecology, Shanghai Changning Maternity and Infant Health Hospital, Shanghai, China
| | - Jiashi Gu
- Department of Obstetrics and Gynecology, Shanghai Pudong Hospital of Fudan University, Shanghai, China
| | - Yanyun Li
- Department of Gynecology and Obstetrics, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Long Sui
- Department of Gynecology and Obstetrics, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| |
Collapse
|
46
|
Wu F, Li D. YB1 and its role in osteosarcoma: a review. Front Oncol 2024; 14:1452661. [PMID: 39497723 PMCID: PMC11532169 DOI: 10.3389/fonc.2024.1452661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 10/07/2024] [Indexed: 11/07/2024] Open
Abstract
YB1 (Y box binding protein 1), a multifunctional protein capable of binding to DNA/RNA, is present in most cells and acts as a splicing factor. It is involved in numerous cellular processes such as transcription, translation, and DNA repair, significantly affecting cell proliferation, differentiation, and apoptosis. Abnormal expression of this protein is closely linked to the formation of various malignancies (osteosarcoma, nasopharyngeal carcinoma, breast cancer, etc.). This review examines the multifaceted functions of YB1 and its critical role in osteosarcoma progression, providing new perspectives for potential therapeutic strategies.
Collapse
Affiliation(s)
| | - Dapeng Li
- Affiliated Hospital of Jiangsu University, Zhenjiang,
Jiangsu, China
| |
Collapse
|
47
|
Wei L, Li Y, Chen J, Wang Y, Wu J, Yang H, Zhang Y. Alternative splicing in ovarian cancer. Cell Commun Signal 2024; 22:507. [PMID: 39425166 PMCID: PMC11488268 DOI: 10.1186/s12964-024-01880-8] [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: 07/29/2024] [Accepted: 10/06/2024] [Indexed: 10/21/2024] Open
Abstract
Ovarian cancer is the second leading cause of gynecologic cancer death worldwide, with only 20% of cases detected early due to its elusive nature, limiting successful treatment. Most deaths occur from the disease progressing to advanced stages. Despite advances in chemo- and immunotherapy, the 5-year survival remains below 50% due to high recurrence and chemoresistance. Therefore, leveraging new research perspectives to understand molecular signatures and identify novel therapeutic targets is crucial for improving the clinical outcomes of ovarian cancer. Alternative splicing, a fundamental mechanism of post-transcriptional gene regulation, significantly contributes to heightened genomic complexity and protein diversity. Increased awareness has emerged about the multifaceted roles of alternative splicing in ovarian cancer, including cell proliferation, metastasis, apoptosis, immune evasion, and chemoresistance. We begin with an overview of altered splicing machinery, highlighting increased expression of spliceosome components and associated splicing factors like BUD31, SF3B4, and CTNNBL1, and their relationships to ovarian cancer. Next, we summarize the impact of specific variants of CD44, ECM1, and KAI1 on tumorigenesis and drug resistance through diverse mechanisms. Recent genomic and bioinformatics advances have enhanced our understanding. By incorporating data from The Cancer Genome Atlas RNA-seq, along with clinical information, a series of prognostic models have been developed, which provided deeper insights into how the splicing influences prognosis, overall survival, the immune microenvironment, and drug sensitivity and resistance in ovarian cancer patients. Notably, novel splicing events, such as PIGV|1299|AP and FLT3LG|50,941|AP, have been identified in multiple prognostic models and are associated with poorer and improved prognosis, respectively. These novel splicing variants warrant further functional characterization to unlock the underlying molecular mechanisms. Additionally, experimental evidence has underscored the potential therapeutic utility of targeting alternative splicing events, exemplified by the observation that knockdown of splicing factor BUD31 or antisense oligonucleotide-induced BCL2L12 exon skipping promotes apoptosis of ovarian cancer cells. In clinical settings, bevacizumab, a humanized monoclonal antibody that specifically targets the VEGF-A isoform, has demonstrated beneficial effects in the treatment of patients with advanced epithelial ovarian cancer. In conclusion, this review constitutes the first comprehensive and detailed exposition of the intricate interplay between alternative splicing and ovarian cancer, underscoring the significance of alternative splicing events as pivotal determinants in cancer biology and as promising avenues for future diagnostic and therapeutic intervention.
Collapse
Affiliation(s)
- Liwei Wei
- Medical School, Faculty of Medicine, Tianjin University, Tianjin, 300072, China
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310030, China
| | - Yisheng Li
- College of Pharmacy, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310030, China
| | - Jiawang Chen
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Wenzhou, Zhejiang, 325101, China
| | - Yuanmei Wang
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310030, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jianmin Wu
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310030, China
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Huanming Yang
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310030, China.
| | - Yi Zhang
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310030, China.
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China.
| |
Collapse
|
48
|
Kwon JH, Kim SH. YBX1 promotes epithelial-mesenchymal transition in hepatocellular carcinoma via transcriptional regulation of PLRG1. Med Oncol 2024; 41:280. [PMID: 39400789 DOI: 10.1007/s12032-024-02516-0] [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: 08/12/2024] [Accepted: 09/20/2024] [Indexed: 10/15/2024]
Abstract
Hepatocellular carcinoma (HCC) ranks as the sixth most prevalent cancer worldwide. The epithelial-mesenchymal transition (EMT) is a critical process in cancer progression, contributing to increased malignancy. While Pleiotropic Regulator 1 (PLRG1) is upregulated in HCC and is associated with enhanced cell proliferation, its oncogenic role in EMT remains unclear. In this study, we demonstrate that PLRG1 promotes EMT in HCC cells. Knockdown of PLRG1 in Huh7 cells resulted in decreased expression of the EMT markers N-cadherin and Snail, and impaired cell migration and invasion. Chromatin immunoprecipitation (ChIP) and luciferase assays identified Y-box binding protein 1 (YBX1) as a direct regulator of PLRG1 transcription, binding to its promoter region. Overexpression of YBX1 in SNU-449 cells led to increased PLRG1 expression and subsequent EMT activation, as well as enhanced migration, and invasion. These effects were attenuated by PLRG1 knockdown. Our findings indicate that YBX1 drives EMT in HCC by upregulating PLRG1, offering novel insights into the molecular mechanisms underlying HCC progression.
Collapse
Affiliation(s)
- Jae Hwan Kwon
- Department of Biology, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Sang Hoon Kim
- Department of Biology, Kyung Hee University, Seoul, 02447, Republic of Korea.
| |
Collapse
|
49
|
Cao AB, Devant P, Wang C, Sun M, Kennedy SN, Ruan J, Kagan JC. LPS binding caspase activation and recruitment domains (CARDs) are bipartite lipid binding modules. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.07.617105. [PMID: 39416091 PMCID: PMC11482759 DOI: 10.1101/2024.10.07.617105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Caspase-11 is an innate immune pattern recognition receptor (PRR) that detects cytosolic bacterial lipopolysaccharides (LPS) through its caspase activation and recruitment domain (CARD), triggering inflammatory cell death known as pyroptosis. Caspase-11 also detects eukaryotic (i.e. self) lipids. This observation raises the question of whether common or distinct mechanisms govern the interactions with self and nonself lipids. In this study, using biochemical, computational, and cell-based assays, we report that the caspase-11 CARD functions as a bipartite lipid-binding module. Distinct regions within the CARD bind to phosphate groups and long acyl chains of self and nonself lipids. Self-lipid binding capability is conserved across numerous caspase-11 homologs and orthologs. The symmetry in self and nonself lipid detection mechanisms enabled us to engineer an LPS-binding domain de novo, using an ancestral CARD-like domain present in the fish Amphilophus citrinellus. These findings offer critical insights into the molecular basis of LPS recognition by caspase-11 and highlight the fundamental and likely inseparable relationship between self and nonself discrimination.
Collapse
Affiliation(s)
- Anh B. Cao
- Division of Gastroenterology, Boston Children’s Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Pascal Devant
- Division of Gastroenterology, Boston Children’s Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Chengliang Wang
- Department of Immunology, UConn Health School of Medicine, 263 Farmington Ave., Farmington, CT 06030, USA
| | - Mengyu Sun
- Division of Gastroenterology, Boston Children’s Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Stephanie N. Kennedy
- Division of Gastroenterology, Boston Children’s Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Jianbin Ruan
- Department of Immunology, UConn Health School of Medicine, 263 Farmington Ave., Farmington, CT 06030, USA
| | - Jonathan C. Kagan
- Division of Gastroenterology, Boston Children’s Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| |
Collapse
|
50
|
Qu Z, Sakaguchi N, Kikutake C, Suyama M. Identification and analysis of short indels inducing exon extension/shrinkage events. FEBS Open Bio 2024; 14:1682-1690. [PMID: 39085971 PMCID: PMC11452298 DOI: 10.1002/2211-5463.13871] [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: 04/09/2024] [Revised: 06/24/2024] [Accepted: 07/19/2024] [Indexed: 08/02/2024] Open
Abstract
The search for genetic variants that act as causative factors in human diseases by disrupting the normal splicing process has primarily focused on single nucleotide variants (SNVs). It is worth noting that insertions or deletions (indels) have also been sporadically reported as causative disease variants through their potential impact on the splicing process. In this study, to perform identification of indels inducing exon extension/shrinkage events, we used individual-specific genomes and RNA sequencing (RNA-seq) data pertaining to the corresponding individuals and identified 12 exon extension/shrinkage events that were potentially induced by indels that disrupted authentic splice sites or created novel splice sites in 235 normal individuals. By evaluating the impact of these abnormal splicing events on the resulting transcripts, we found that five events led to the generation of premature termination codons (PTCs), including those occurring within genes associated with genetic disorders. Our analysis revealed that the potential functions of indels have been underexamined, and it is worth considering the possibility that indels may affect splice site usage, using RNA-seq data to discover novel potentially disease-associated mutations.
Collapse
Affiliation(s)
- Zhuo Qu
- Division of Bioinformatics, Medical Institute of BioregulationKyushu UniversityFukuokaJapan
| | - Narumi Sakaguchi
- Division of Bioinformatics, Medical Institute of BioregulationKyushu UniversityFukuokaJapan
| | - Chie Kikutake
- Division of Bioinformatics, Medical Institute of BioregulationKyushu UniversityFukuokaJapan
| | - Mikita Suyama
- Division of Bioinformatics, Medical Institute of BioregulationKyushu UniversityFukuokaJapan
| |
Collapse
|