1
|
Yu J, Deng X, Lin X, Xie L, Guo S, Lin X, Lin D. DST regulates cisplatin resistance in colorectal cancer via PI3K/Akt pathway. J Pharm Pharmacol 2024:rgae104. [PMID: 39419785 DOI: 10.1093/jpp/rgae104] [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: 02/06/2024] [Accepted: 07/26/2024] [Indexed: 10/19/2024]
Abstract
OBJECTIVES Dystonin (DST), a potential tumor suppressor gene, plays a crucial role in regulating cancer cell proliferation and resistance to chemotherapy. However, DST's specific role in colorectal cancer (CRC) has not been thoroughly investigated, and this study aims to elucidate its molecular role in modulating cisplatin (DDP) resistance in CRC. METHODS DST expression was analyzed in CRC tumors, DDP-resistant CRC tissues, paracancer tissues, and normal tissues. Lentiviral overexpression and shRNA knockdown were conducted in advanced CRC and DDP-resistant cell lines to assess cell viability, apoptosis, invasion, migration, proliferation, and angiogenesis. Xenograft mouse models studied DST's impact on CRC tumor growth and DDP resistance in vivo. RESULTS DST expression was significantly reduced in CRC tumor and DDP-resistant CRC tissues compared to paracancer and normal tissues (P < .001). Upregulating DST inhibited CRC and DDP-resistant cell viability, proliferation, invasion, and migration while promoting apoptosis. DST overexpression also reduced angiogenesis and attenuated DDP-induced cytotoxicity in CRC cells. Mechanistically, DST upregulation suppressed DDP resistance in CRC cells via the PI3K/Akt signaling pathway. DST upregulation reduced CRC tumor growth and mitigated DDP resistance, in vivo. CONCLUSION DST plays a crucial role in limiting CRC progression and overcoming DDP resistance, suggesting potential for targeted CRC therapies.
Collapse
Affiliation(s)
- Jianwei Yu
- Department of Gastroenterology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan 364000, Fujian Province, China
| | - Xueqiong Deng
- Department of Gastroenterology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan 364000, Fujian Province, China
| | - Xueqin Lin
- Department of Gastroenterology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan 364000, Fujian Province, China
| | - Li Xie
- Department of Gastroenterology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan 364000, Fujian Province, China
| | - Sisi Guo
- Department of Gastroenterology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan 364000, Fujian Province, China
| | - Xiaoliang Lin
- Department of Gastroenterology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan 364000, Fujian Province, China
| | - Dong Lin
- Department of Gastroenterology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan 364000, Fujian Province, China
| |
Collapse
|
2
|
Joseph SC, Eugin Simon S, Bohm MS, Kim M, Pye ME, Simmons BW, Graves DG, Thomas-Gooch SM, Tanveer UA, Holt JR, Ponnusamy S, Sipe LM, Hayes DN, Cook KL, Narayanan R, Pierre JF, Makowski L. FXR Agonism with Bile Acid Mimetic Reduces Pre-Clinical Triple-Negative Breast Cancer Burden. Cancers (Basel) 2024; 16:1368. [PMID: 38611046 PMCID: PMC11011133 DOI: 10.3390/cancers16071368] [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: 02/06/2024] [Revised: 03/20/2024] [Accepted: 03/26/2024] [Indexed: 04/14/2024] Open
Abstract
Bariatric surgery is associated with improved outcomes for several cancers, including breast cancer (BC), although the mechanisms mediating this protection are unknown. We hypothesized that elevated bile acid pools detected after bariatric surgery may be factors that contribute to improved BC outcomes. Patients with greater expression of the bile acid receptor FXR displayed improved survival in specific aggressive BC subtypes. FXR is a nuclear hormone receptor activated by primary bile acids. Therefore, we posited that activating FXR using an established FDA-approved agonist would induce anticancer effects. Using in vivo and in vitro approaches, we determined the anti-tumor potential of bile acid receptor agonism. Indeed, FXR agonism by the bile acid mimetic known commercially as Ocaliva ("OCA"), or Obeticholic acid (INT-747), significantly reduced BC progression and overall tumor burden in a pre-clinical model. The transcriptomic analysis of tumors in mice subjected to OCA treatment revealed differential gene expression patterns compared to vehicle controls. Notably, there was a significant down-regulation of the oncogenic transcription factor MAX (MYC-associated factor X), which interacts with the oncogene MYC. Gene set enrichment analysis (GSEA) further demonstrated a statistically significant downregulation of the Hallmark MYC-related gene set (MYC Target V1) following OCA treatment. In human and murine BC analyses in vitro, agonism of FXR significantly and dose-dependently inhibited proliferation, migration, and viability. In contrast, the synthetic agonism of another common bile acid receptor, the G protein-coupled bile acid receptor TGR5 (GPBAR1) which is mainly activated by secondary bile acids, failed to significantly alter cancer cell dynamics. In conclusion, agonism of FXR by primary bile acid memetic OCA yields potent anti-tumor effects potentially through inhibition of proliferation and migration and reduced cell viability. These findings suggest that FXR is a tumor suppressor gene with a high potential for use in personalized therapeutic strategies for individuals with BC.
Collapse
Affiliation(s)
- Sydney C. Joseph
- Department of Medicine, Division of Hematology and Oncology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Samson Eugin Simon
- Department of Medicine, Division of Hematology and Oncology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Margaret S. Bohm
- Department of Microbiology, Immunology and Biochemistry, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Minjeong Kim
- Department of Medicine, Division of Hematology and Oncology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Madeline E. Pye
- Department of Medicine, Division of Hematology and Oncology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Boston W. Simmons
- Department of Medicine, Division of Hematology and Oncology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Dillon G. Graves
- Department of Medicine, Division of Hematology and Oncology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Stacey M. Thomas-Gooch
- Department of Medicine, Division of Hematology and Oncology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Ubaid A. Tanveer
- Department of Medicine, Division of Hematology and Oncology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Jeremiah R. Holt
- Department of Medicine, Division of Hematology and Oncology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Suriyan Ponnusamy
- Department of Medicine, Division of Hematology and Oncology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Laura M. Sipe
- Department of Biological Sciences, University of Mary Washinton, Fredericksburg, VI 22401, USA
| | - D. Neil Hayes
- Department of Medicine, Division of Hematology and Oncology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN 38163, USA
- UTHSC Center for Cancer Research, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Katherine L. Cook
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, NC 27157, USA;
| | - Ramesh Narayanan
- Department of Medicine, Division of Hematology and Oncology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN 38163, USA
- UTHSC Center for Cancer Research, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Joseph F. Pierre
- Department of Nutritional Sciences, College of Agricultural and Life Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Liza Makowski
- Department of Medicine, Division of Hematology and Oncology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN 38163, USA
- Department of Microbiology, Immunology and Biochemistry, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN 38163, USA
- UTHSC Center for Cancer Research, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN 38163, USA
| |
Collapse
|
3
|
Cui Y, Wang Z, Wang X, Zhang Y, Zhang Y, Pan T, Zhang Z, Li S, Guo Y, Akutsu T, Song J. SMG: self-supervised masked graph learning for cancer gene identification. Brief Bioinform 2023; 24:bbad406. [PMID: 37950905 PMCID: PMC10639095 DOI: 10.1093/bib/bbad406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 09/26/2023] [Accepted: 10/24/2023] [Indexed: 11/13/2023] Open
Abstract
Cancer genomics is dedicated to elucidating the genes and pathways that contribute to cancer progression and development. Identifying cancer genes (CGs) associated with the initiation and progression of cancer is critical for characterization of molecular-level mechanism in cancer research. In recent years, the growing availability of high-throughput molecular data and advancements in deep learning technologies has enabled the modelling of complex interactions and topological information within genomic data. Nevertheless, because of the limited labelled data, pinpointing CGs from a multitude of potential mutations remains an exceptionally challenging task. To address this, we propose a novel deep learning framework, termed self-supervised masked graph learning (SMG), which comprises SMG reconstruction (pretext task) and task-specific fine-tuning (downstream task). In the pretext task, the nodes of multi-omic featured protein-protein interaction (PPI) networks are randomly substituted with a defined mask token. The PPI networks are then reconstructed using the graph neural network (GNN)-based autoencoder, which explores the node correlations in a self-prediction manner. In the downstream tasks, the pre-trained GNN encoder embeds the input networks into feature graphs, whereas a task-specific layer proceeds with the final prediction. To assess the performance of the proposed SMG method, benchmarking experiments are performed on three node-level tasks (identification of CGs, essential genes and healthy driver genes) and one graph-level task (identification of disease subnetwork) across eight PPI networks. Benchmarking experiments and performance comparison with existing state-of-the-art methods demonstrate the superiority of SMG on multi-omic feature engineering.
Collapse
Affiliation(s)
- Yan Cui
- Bioinformatics Center, Institute for Chemical Research, Kyoto University, Kyoto 611-0011, Japan
| | - Zhikang Wang
- Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC 3800, Australia
| | - Xiaoyu Wang
- Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC 3800, Australia
| | - Yiwen Zhang
- School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC 3004, Australia
| | - Ying Zhang
- School of Computer Science and Engineering, Nanjing University of Science and Technology, 200 Xiaolingwei, Nanjing, 210094, China
| | - Tong Pan
- Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC 3800, Australia
| | | | - Shanshan Li
- School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC 3004, Australia
| | - Yuming Guo
- School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC 3004, Australia
| | - Tatsuya Akutsu
- Bioinformatics Center, Institute for Chemical Research, Kyoto University, Kyoto 611-0011, Japan
| | - Jiangning Song
- Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC 3800, Australia
- Monash Data Futures Institute, Monash University, Melbourne, VIC 3800, Australia
| |
Collapse
|
4
|
Jayarathna DK, Rentería ME, Batra J, Gandhi NS. Integrative competing endogenous RNA network analyses identify novel lncRNA and genes implicated in metastatic breast cancer. Sci Rep 2023; 13:2423. [PMID: 36765262 PMCID: PMC9918521 DOI: 10.1038/s41598-023-29585-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 02/07/2023] [Indexed: 02/12/2023] Open
Abstract
Competing endogenous RNAs (ceRNAs) have gained attention in cancer research owing to their involvement in microRNA-mediated gene regulation. Previous studies have identified ceRNA networks of individual cancers. Nevertheless, none of these studies has investigated different cancer stages. We identify stage-specific ceRNAs in breast cancer using the cancer genome atlas data. Moreover, we investigate the molecular functions and prognostic ability of ceRNAs involved in stage I-IV networks. We identified differentially expressed candidate ceRNAs using edgeR and limma R packages. A three-step analysis was used to identify statistically significant ceRNAs of each stage. Survival analysis and functional enrichment analysis were conducted to identify molecular functions and prognostic ability. We found five genes and one long non-coding RNA unique to the stage IV ceRNA network. These genes have been described in previous breast cancer studies. Genes acted as ceRNAs are enriched in cancer-associated pathways. Two, three, and three microRNAs from stages I, II, and III were prognostic from the Kaplan-Meier survival analysis. Our results reveal a set of unique ceRNAs in metastatic breast cancer. Further experimental work is required to evaluate their role in metastasis. Moreover, identifying stage-specific ceRNAs will improve the understanding of personalised therapeutics in breast cancer.
Collapse
Affiliation(s)
- Dulari K Jayarathna
- Centre for Genomics and Personalised Health, School of Chemistry and Physics, Queensland University of Technology, 2 George Street, Brisbane, QLD, 4000, Australia
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia
| | - Miguel E Rentería
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Kelvin Grove, Brisbane, QLD, 4059, Australia
| | - Jyotsna Batra
- Centre for Genomics and Personalised Health, School of Chemistry and Physics, Queensland University of Technology, 2 George Street, Brisbane, QLD, 4000, Australia
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Kelvin Grove, Brisbane, QLD, 4059, Australia
- Translational Research Institute, 37 Kent Street, Woolloongabba, QLD, 4102, Australia
| | - Neha S Gandhi
- Centre for Genomics and Personalised Health, School of Chemistry and Physics, Queensland University of Technology, 2 George Street, Brisbane, QLD, 4000, Australia.
- Translational Research Institute, 37 Kent Street, Woolloongabba, QLD, 4102, Australia.
| |
Collapse
|
5
|
NUSAP1 and PCLAF (KIA0101) Downregulation by Neoadjuvant Therapy is Associated with Better Therapeutic Outcomes and Survival in Breast Cancer. JOURNAL OF ONCOLOGY 2022; 2022:6001947. [DOI: 10.1155/2022/6001947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 09/30/2022] [Accepted: 10/31/2022] [Indexed: 11/30/2022]
Abstract
Purpose. To evaluate whether changes in genomic expression that occur beginning with breast cancer (BC) diagnosis and through to tumor resection after neoadjuvant chemotherapy (NCT) reveal biomarkers that can help predict therapeutic response and survival. Materials and Methods. We determined gene expression profiles based on microarrays in tumor samples from 39 BC patients who showed pathologic complete response (pCR) or therapeutic failure (non-pCR) after NCT (cyclophosphamide-doxorubicin/epirubicin). Based on unsupervised clustering of gene expression, together with functional enrichment analyses of differentially expressed genes, we selected NUSAP1, PCLAF, MME, and DST. We evaluated the NCT response and the expression of these four genes in BC histologic subtypes. In addition, we study the presence of tumor-infiltrating lymphocytes. Finally, we analyze the correlation between NUSAP1 and PCLAF against disease-free survival (DFS) and overall survival (OS). Results. A signature of 43 differentially expressed genes discriminated pCR from non-pCR patients (|fold change >2|, false discovery rate <0.05) only in biopsies taken after surgery. Patients achieving pCR showed downregulation of NUSAP1 and PCLAF in tumor tissues and increased DFS and OS, while overexpression of these genes correlated with poor therapeutic response and OS. These genes are involved in the regulation of mitotic division. Conclusions. The downregulation of NUSAP1 and PCLAF after NCT is associated with the tumor response to chemotherapy and patient survival.
Collapse
|
6
|
Khalil K, Eon A, Janody F. Cell Architecture-Dependent Constraints: Critical Safeguards to Carcinogenesis. Int J Mol Sci 2022; 23:8622. [PMID: 35955754 PMCID: PMC9369145 DOI: 10.3390/ijms23158622] [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: 07/06/2022] [Revised: 07/29/2022] [Accepted: 07/30/2022] [Indexed: 02/04/2023] Open
Abstract
Animal cells display great diversity in their shape. These morphological characteristics result from crosstalk between the plasma membrane and the force-generating capacities of the cytoskeleton macromolecules. Changes in cell shape are not merely byproducts of cell fate determinants, they also actively drive cell fate decisions, including proliferation and differentiation. Global and local changes in cell shape alter the transcriptional program by a multitude of mechanisms, including the regulation of physical links between the plasma membrane and the nuclear envelope and the mechanical modulation of cation channels and signalling molecules. It is therefore not surprising that anomalies in cell shape contribute to several diseases, including cancer. In this review, we discuss the possibility that the constraints imposed by cell shape determine the behaviour of normal and pro-tumour cells by organizing the whole interconnected regulatory network. In turn, cell behaviour might stabilize cells into discrete shapes. However, to progress towards a fully transformed phenotype and to acquire plasticity properties, pro-tumour cells might need to escape these cell shape restrictions. Thus, robust controls of the cell shape machinery may represent a critical safeguard against carcinogenesis.
Collapse
Affiliation(s)
- Komal Khalil
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; (K.K.); (A.E.)
- IPATIMUP—Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Rua Dr. Roberto Frias s/n, 4200-465 Porto, Portugal
- Master Programme in Oncology, School of Medicine & Biomedical Sciences, University of Porto (ICBAS-UP), Rua Jorge Viterbo Ferreira 228, 4050-513 Porto, Portugal
| | - Alice Eon
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; (K.K.); (A.E.)
- IPATIMUP—Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Rua Dr. Roberto Frias s/n, 4200-465 Porto, Portugal
- Magistère Européen de Génétique, Université Paris Cité, 5 Rue Thomas Mann, 75013 Paris, France
| | - Florence Janody
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; (K.K.); (A.E.)
- IPATIMUP—Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Rua Dr. Roberto Frias s/n, 4200-465 Porto, Portugal
| |
Collapse
|
7
|
Ancient Origins of Cytoskeletal Crosstalk: Spectraplakin-like Proteins Precede the Emergence of Cortical Microtubule Stabilization Complexes as Crosslinkers. Int J Mol Sci 2022; 23:ijms23105594. [PMID: 35628404 PMCID: PMC9145010 DOI: 10.3390/ijms23105594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 05/12/2022] [Accepted: 05/12/2022] [Indexed: 11/17/2022] Open
Abstract
Adhesion between cells and the extracellular matrix (ECM) is one of the prerequisites for multicellularity, motility, and tissue specialization. Focal adhesions (FAs) are defined as protein complexes that mediate signals from the ECM to major components of the cytoskeleton (microtubules, actin, and intermediate filaments), and their mutual communication determines a variety of cellular processes. In this study, human cytoskeletal crosstalk proteins were identified by comparing datasets with experimentally determined cytoskeletal proteins. The spectraplakin dystonin was the only protein found in all datasets. Other proteins (FAK, RAC1, septin 9, MISP, and ezrin) were detected at the intersections of FAs, microtubules, and actin cytoskeleton. Homology searches for human crosstalk proteins as queries were performed against a predefined dataset of proteomes. This analysis highlighted the importance of FA communication with the actin and microtubule cytoskeleton, as these crosstalk proteins exhibit the highest degree of evolutionary conservation. Finally, phylogenetic analyses elucidated the early evolutionary history of spectraplakins and cortical microtubule stabilization complexes (CMSCs) as model representatives of the human cytoskeletal crosstalk. While spectraplakins probably arose at the onset of opisthokont evolution, the crosstalk between FAs and microtubules is associated with the emergence of metazoans. The multiprotein complexes contributing to cytoskeletal crosstalk in animals gradually gained in complexity from the onset of metazoan evolution.
Collapse
|
8
|
Zhang H, Lu X, Huang G, Hua M, Zhang W, Wang T, Huang L, Wang Z, Chen Q, Li J, Yang Q, Yang G. A genomic mutation spectrum of collecting duct carcinoma in the Chinese population. BMC Med Genomics 2022; 15:1. [PMID: 34980126 PMCID: PMC8722201 DOI: 10.1186/s12920-021-01143-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 12/01/2021] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Renal collecting duct carcinoma (CDC) is a rare and lethal subtype of renal cell carcinoma (RCC). The genomic profile of the Chinese population with CDC remains unclear. In addition, clinical treatments are contradictory. In this study, we aimed to identify the genomic mutation spectrum of CDC in the Chinese population. METHODS Whole-exome sequencing was performed using the Illumina Novaseq™ 6000 platform. MuTect2 detects single-nucleotide variants (SNVs) and small scale insertions/deletions (INDELs). The identified mutations were annotated with ANNOVAR and validated by Sanger sequencing. Control-FREEC was used to detect copy number variation (CNV), and GISTIC was applied to detect frequently mutated altered regions. These data were compared with associated The Cancer Genome Atlas cohorts. RESULTS Ten normal-matched CDC patients were included. The mean tumour mutation burden was 1.37 Mut/Mb. Six new recurrent somatic mutated genes were identified, including RBM14, MTUS1, GAK, DST, RNF213 and XIRP2 (20% and 2 of 10, respectively), and validated by Sanger sequencing. In terms of common mutated genes, SETD2 was altered in both CDC and other RCC subtypes but not in bladder urothelial carcinoma (BLCA); CDKN2A was a driver gene in both CDC (SNV: 10%, 1 of 10) and BLCA but not in other RCC subtypes. Next, 29 amplifications and 6 deletions of recurrent focal somatic CNVs were identified by GISTIC2.0, which displayed differences from kidney renal clear cell carcinoma (KIRC), kidney renal papillary cell carcinoma (KIRP) and BLCA cohorts. Of note, CDKN2A (CNV alteration: 30%, 3 of 10) and CDKN2A-AS1 were the only overlapping genes of these four cohorts. Importantly, the CDKN2A mutation in our cohort differed from previous studies in urinary carcinomas. Moreover, CDKN2A-altered cases had significantly worse overall survival than wild-type cases in both KIRC and KIRP cohorts. In addition, the most frequently altered genomic pathway of our CDC cohort was the CDKN2A-mediated p53/RB1 pathway. CONCLUSIONS Our study offers the first genomic spectrum of the Chinese population with CDC, which differs from that of the Western population. The altered CDKN2A-mediated p53/RB1 pathway might provide new insight into potential therapeutic targets for CDC patients.
Collapse
Affiliation(s)
- Huaru Zhang
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
- Department of Urology, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China
- Department of Urology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Xiaojun Lu
- Department of Urology, the First Affiliated Hospital, Naval Military Medical University, Shanghai, 200433, China
| | - Gang Huang
- Department of Urology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Meimian Hua
- Department of Urology, the First Affiliated Hospital, Naval Military Medical University, Shanghai, 200433, China
| | - Wenhui Zhang
- Department of Urology, the First Affiliated Hospital, Naval Military Medical University, Shanghai, 200433, China
| | - Tao Wang
- Department of Urology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Liqun Huang
- Department of Urology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Ziwei Wang
- Department of Urology, the First Affiliated Hospital, Naval Military Medical University, Shanghai, 200433, China
| | - Qing Chen
- Department of Urology, the First Affiliated Hospital, Naval Military Medical University, Shanghai, 200433, China
| | - Jing Li
- Department of Bioinformatics, Center for Translational Medicine, Second Military Medical University, Shanghai, 200433, China.
| | - Qing Yang
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200011, China.
| | - Guosheng Yang
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China.
- Department of Urology, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China.
- Department of Urology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.
| |
Collapse
|
9
|
Ayanlaja AA, Hong X, Cheng B, Zhou H, Kanwore K, Alphayo-Kambey P, Zhang L, Tang C, Adeyanju MM, Gao D. Susceptibility of cytoskeletal-associated proteins for tumor progression. Cell Mol Life Sci 2021; 79:13. [PMID: 34964908 PMCID: PMC11072373 DOI: 10.1007/s00018-021-04101-4] [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: 08/17/2021] [Revised: 12/11/2021] [Accepted: 12/16/2021] [Indexed: 10/19/2022]
Abstract
The traditional functions of cytoskeletal-associated proteins (CAPs) in line with polymerization and stabilization of the cytoskeleton have evolved and are currently underrated in oncology. Although therapeutic drugs have been developed to target the cytoskeletal components directly in cancer treatment, several recently established therapeutic agents designed for new targets block the proliferation of cancer cells and suppress resistance to existing target agents. It would seem like these targets only work toward inhibiting the polymerization of cytoskeletal components or hindering mitotic spindle formation in cancer cells, but a large body of literature points to CAPs and their culpability in cell signaling, molecular conformation, organelle trafficking, cellular metabolism, and genomic modifications. Here, we review those underappreciated functions of CAPs, and we delineate the implications of cellular signaling instigated by evasive properties induced by aberrant expression of CAPs in response to stress or failure to exert normal functions. We present an analogy establishing CAPs as vulnerable targets for cancer systems and credible oncotargets. This review establishes a paradigm in which the cancer machinery may commandeer the conventional functions of CAPs for survival, drug resistance, and energy generation; an interesting feature overdue for attention.
Collapse
Affiliation(s)
- Abiola Abdulrahman Ayanlaja
- Public Experimental Laboratory, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- Department of Neurology, Johns Hopkins University School of Medicine, 201 N Broadway, Baltimore, MD, 21287, USA
| | - Xiaoliang Hong
- Public Experimental Laboratory, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Bo Cheng
- The Affiliated Oriental Hospital of Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Han Zhou
- Public Experimental Laboratory, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Kouminin Kanwore
- Public Experimental Laboratory, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Piniel Alphayo-Kambey
- Public Experimental Laboratory, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Lin Zhang
- Public Experimental Laboratory, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Chuanxi Tang
- Public Experimental Laboratory, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | | | - Dianshuai Gao
- Public Experimental Laboratory, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
| |
Collapse
|
10
|
Zhang H, Cheng Z, Li W, Hu J, Zhao L, Chen D, Gao J, Chen J, Yan Y, Lin L, Shi A. WTS-1/LATS regulates endocytic recycling by restraining F-actin assembly in a synergistic manner. J Cell Sci 2021; 134:273738. [PMID: 34817059 DOI: 10.1242/jcs.259085] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 11/16/2021] [Indexed: 12/30/2022] Open
Abstract
The disruption of endosomal actin architecture negatively affects endocytic recycling. However, the underlying homeostatic mechanisms that regulate actin organization during recycling remain unclear. In this study, we identified a synergistic endosomal actin assembly restricting mechanism in C. elegans involving WTS-1, the homolog of LATS kinases, which is a core component of the Hippo pathway. WTS-1 resides on the sorting endosomes and colocalizes with the actin polymerization regulator PTRN-1 [the homolog of the calmodulin-regulated spectrin-associated proteins (CAMSAPs)]. We observed an increase in PTRN-1-labeled structures in WTS-1-deficient cells, indicating that WTS-1 can limit the endosomal localization of PTRN-1. Accordingly, the actin overaccumulation phenotype in WTS-1-depleted cells was mitigated by the associated PTRN-1 loss. We further demonstrated that recycling defects and actin overaccumulation in WTS-1-deficient cells were reduced by the overexpression of constitutively active UNC-60A(S3A) (a cofilin protein homolog), which aligns with the role of LATS as a positive regulator of cofilin activity. Altogether, our data confirmed previous findings, and we propose an additional model, that WTS-1 acts alongside the UNC-60A-mediated actin disassembly to restrict the assembly of endosomal F-actin by curbing PTRN-1 dwelling on endosomes, preserving recycling transport.
Collapse
Affiliation(s)
- Hanchong Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Zihang Cheng
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Wenbo Li
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Jie Hu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Linyue Zhao
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Dan Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Jinghu Gao
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Juan Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China.,Cell Architecture Research Center, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Yanling Yan
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China.,Cell Architecture Research Center, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Long Lin
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China.,Cell Architecture Research Center, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Anbing Shi
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China.,Cell Architecture Research Center, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| |
Collapse
|
11
|
Li P, Lin Z, Liu Q, Chen S, Gao X, Guo W, Gong F, Wei J, Lin H. Enhancer RNA SLIT2 Inhibits Bone Metastasis of Breast Cancer Through Regulating P38 MAPK/c-Fos Signaling Pathway. Front Oncol 2021; 11:743840. [PMID: 34722297 PMCID: PMC8554345 DOI: 10.3389/fonc.2021.743840] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 09/23/2021] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Breast cancer (BRCA) is the most common cancer in women, while the bones are one of the most common sites of metastasis. Although new diagnostic methods or radiation or chemotherapies and targeted therapies have made huge advances, the occurrence of bone metastasis is also linked with poorer survival. Enhancer RNAs (eRNAs) have been demonstrated to participate in the progression of tumorigenesis and metastasis. However, the role of eRNAs in BRCA bone metastasis remains largely unclear. METHOD Gene expression profiling of 1,211 primary BRCA and 17 bone metastases samples were retrieved from The Cancer Genome Atlas (TCGA) database, and the significant prognostic eRNAs were identified by Cox regression and least absolute shrinkage and selection operator (LASSO) regression. The acceptable accuracy and discrimination of the nomogram were indicated by the receiver operating characteristic (ROC) and the calibration curves. Then target genes of eRNA, immune cell percentage by CIBERSORT analysis, immune genes by single-sample gene set enrichment analysis (ssGSEA), hallmark of cancer signaling pathway by gene set variation analysis (GSVA), and reverse phase protein array (RPPA) protein chip were used to build a co-expression regulation network and identified the key eRNAs in bone metastasis of BRCA. Finally, Cell Counting Kit-8 (CCK8) assay, cell cycle assay, and transwell assay were used to study changes in cell proliferation, migration, and invasiveness. Immunoprecipitation assay and Western blotting were used to test the interaction and the regulation signaling pathways. RESULTS The 27 hub eRNAs were selected, and a survival-related linear risk assessment model with a relatively high accuracy (area under curve (AUC): 0.726) was constructed. In addition, seven immune-related eRNAs (SLIT2, CLEC3B, LBPL1, FRY, RASGEF1B, DST, and ITIH5) as prognostic signatures for bone metastasis of BRCA were further confirmed by LASSO and multivariate Cox regression and CIBERSORT analysis. Finally, in vitro assay demonstrated that overexpression of SLIT2 reduced proliferation and metastasis in BRCA cells. Using high-throughput co-expression regulation network, we identified that SLIT2 may regulating P38 MAPK/c-Fos signaling pathway to promote the effects of metastasis. CONCLUSION Based on the co-expression network for bone metastasis of BRCA, we screened key eRNAs to explore a prognostic model in predicting the bone metastasis by bioinformatics analysis. Besides, we identified the potential regulatory signaling pathway of SLIT2 in BRCA bone metastasis, which provides a promising therapeutic strategy for metastasis of BRCA.
Collapse
Affiliation(s)
- Peng Li
- Stem Cell Research and Cellular Therapy Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Zhiping Lin
- Orthopedic Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
- Orthopedic Center, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Qianzheng Liu
- Orthopedic Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Siyuan Chen
- Orthopedic Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Xiang Gao
- Stem Cell Research and Cellular Therapy Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Weixiong Guo
- Orthopedic Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Fan Gong
- Orthopedic Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Jinsong Wei
- Orthopedic Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Hao Lin
- Orthopedic Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
12
|
Talley EM, Watts CT, Aboyer S, Adamson MG, Akoto HA, Altemus H, Avella PJ, Bailey R, Bell ER, Bell KL, Breneman K, Burkhart JS, Chanley LJ, Cook SS, DesLaurier MT, Dorsey TR, Doyle CJ, Egloff ME, Fasawe AS, Garcia KK, Graves NP, Gray TK, Gustafson EM, Hall MJ, Hayes JD, Holic LJ, Jarvis BA, Klos PS, Kritzmire S, Kuzovko L, Lainez E, McCoy S, Mierendorf JC, Neri NA, Neville CR, Osborn K, Parker K, Parks ME, Peck K, Pitt R, Platta ME, Powell B, Rodriguez K, Ruiz C, Schaefer MN, Shields AB, Smiley JB, Stauffer B, Straub D, Sweeney JL, Termine KM, Thomas B, Toth SD, Veile TR, Walker KS, Webster PN, Woodard BJ, Yoder QL, Young MK, Zeedyk ML, Ziegler LN, Bieser KL, Puthoff DP, Stamm J, Vrailas-Mortimer AD, Kagey JD, Merkle JA. Genetic mapping and phenotypic analysis of shotH.3.2 in Drosophila melanogaster. MICROPUBLICATION BIOLOGY 2021; 2021. [PMID: 34278244 PMCID: PMC8278230 DOI: 10.17912/micropub.biology.000418] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/01/2021] [Accepted: 07/02/2021] [Indexed: 11/22/2022]
Abstract
Genetic screens are used to identify genes involved in specific biological processes. An EMS mutagenesis screen in Drosophila melanogaster identified growth control phenotypes in the developing eye. One mutant line from this screen, H.3.2, was phenotypically characterized using the FLP/FRT system and genetically mapped by complementation analysis and genomic sequencing by undergraduate students participating in the multi-institution Fly-CURE consortium. H.3.2 was found to have a nonsense mutation in short stop (shot), anortholog of the mammalian spectraplakin dystonin (DST). shot and DST are involved in cytoskeletal organization and play roles during cell growth and proliferation.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Kylee Peck
- University of Evansville, Evansville, IN USA
| | - Robyn Pitt
- Illinois State University, Normal, IL USA
| | | | | | | | - Clara Ruiz
- Illinois State University, Normal, IL USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Joyce Stamm
- University of Evansville, Evansville, IN USA
| | | | | | | |
Collapse
|
13
|
Sun X, Chen P, Chen X, Yang W, Chen X, Zhou W, Huang D, Cheng Y. KIF4A enhanced cell proliferation and migration via Hippo signaling and predicted a poor prognosis in esophageal squamous cell carcinoma. Thorac Cancer 2020; 12:512-524. [PMID: 33350074 PMCID: PMC7882383 DOI: 10.1111/1759-7714.13787] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/30/2020] [Accepted: 11/30/2020] [Indexed: 12/17/2022] Open
Abstract
Background In this study, we aimed to explore and clarify the function of KIF4A in esophageal squamous cell carcinoma (ESCC). Methods The microarray data were extracted from the Gene Expression Omnibus (GEO) database. We then used the database for Annotation, Visualization, and Integrated Discovery (DAVID) to perform the gene ontology function (GO) and KEGG Orthology‐Based Annotation System (KOBAS) to perform Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis of differentially expressed genes (DEGs). The six core candidate genes were identified using protein–protein interaction (PPI) network analysis and Cytoscape software. Among them, the expression of KIF4A were validated by UALCAN database from the Cancer Genome Atlas (TCGA) (P < 0.05). Western blotting, qRT‐PCR and IHC were used to detect the expression of KIF4A in tissues. Cell experiments (transwell migration assays, wound healing assay, CCK8 assay, and clone formation experiment) were utilized to verify the roles of KIF4A on the ESCC cells. Western blotting was used to explore the mechanism of KIF4A in ESCC. Results The expression level of KIF4A was upregulated in ESCC samples compared to those in paracancerous tissues. Transwell migration and wound healing assay showed overexpression of KIF4A significantly promoted the migration of ESCC cells. CCK8 assay and clone formation experiment analysis showed that overexpression of KIF4A promoted proliferation of ESCC cells. Western blot detection found that KIF4A could affect the phosphorylation level of Hippo signaling pathway related proteins. Conclusions In summary, KIF4A promotes ESCC cell proliferation and migration by regulating the biological function of ESCC cells through the Hippo signaling pathway. Key points Significant findings of the study We found that high KIF4A expression was associated with poor overall survival in esophageal squamous cell carcinoma. KIF4A expression also promoted the proliferation and migration of ESCC cells in vitro. What this study adds Our experimental results explain the role of KIF4A in ESCC, and provide a new biomolecular target for the treatment of ESCC.
Collapse
Affiliation(s)
- Xiaozheng Sun
- Department of Radiation Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Pengxiang Chen
- Department of Radiation Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xue Chen
- Department of Radiation Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Wenjing Yang
- Department of Radiation Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xuan Chen
- Department of Radiation Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Wei Zhou
- Department of Radiation Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Di Huang
- Department of Radiation Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yufeng Cheng
- Department of Radiation Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|