1
|
Saroj N, Dholaniya PS, Alvi SB, Sridharan D, Soni N, Ashraf SA, Choudhry A, Ashraf YA, Mikula SK, Singla DK, Khan M. SiRNA-mediated knockdown of TOP2B protects hiPSC-derived cardiomyocytes from doxorubicin-induced toxicity. Life Sci 2025; 371:123595. [PMID: 40158615 DOI: 10.1016/j.lfs.2025.123595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 03/22/2025] [Accepted: 03/27/2025] [Indexed: 04/02/2025]
Abstract
AIMS Doxorubicin (Dox) is a potent chemotherapeutic agent, but its use is limited by cardiotoxicity, primarily due to the disruption of Topoisomerase-2 beta (TOP2B) activity. Dexrazoxane (Dex), an FDA-approved cardioprotective drug, alleviates Dox-induced toxicity but lacks heart-specific targeting. This study investigates siRNA-mediated TOP2B knockdown as a more targeted strategy to protect cardiomyocytes from Dox-induced damage. MATERIALS AND METHODS Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) were treated with siRNA to knock down TOP2B and were then exposed to Dox. We compared the cardioprotective effects of siRNA-mediated knockdown to Dex treatment using cell viability, cell toxicity assay and electrophysiological evaluation was performed using a multielectrode array (MEA). KEY FINDINGS Our results demonstrate that TOP2B silencing significantly decreases apoptosis and improved cell viability, as compared to the Dex treatment. Additionally, electrophysiological assays using a Multielectrode Array (MEA) demonstrated enhanced contractility and conductivity in siRNA-treated hiPSC-CMs. Furthermore, transmission electron microscopy (TEM) data revealed that TOP2B knockdown preserves mitochondrial morphology and sarcomere structure, compared to Dox and Dex-treated groups. SIGNIFICANCE These findings suggest that siRNA-mediated TOP2B inhibition could provide a safer, more specific approach to mitigate Dox-induced cardiotoxicity.
Collapse
Affiliation(s)
- Neha Saroj
- Division of Basic and Translational Research, Department of Emergency Medicine, The Ohio State University, Columbus, OH, USA; Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA; Department of Biotechnology & Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad, India
| | - Pankaj Singh Dholaniya
- Division of Basic and Translational Research, Department of Emergency Medicine, The Ohio State University, Columbus, OH, USA; Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA; Department of Biotechnology & Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad, India.
| | - Syed Baseeruddin Alvi
- Division of Basic and Translational Research, Department of Emergency Medicine, The Ohio State University, Columbus, OH, USA; Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Divya Sridharan
- Division of Basic and Translational Research, Department of Emergency Medicine, The Ohio State University, Columbus, OH, USA; Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Navisha Soni
- Division of Basic and Translational Research, Department of Emergency Medicine, The Ohio State University, Columbus, OH, USA; Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Syed Abdullah Ashraf
- Division of Basic and Translational Research, Department of Emergency Medicine, The Ohio State University, Columbus, OH, USA; Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Ayza Choudhry
- Division of Basic and Translational Research, Department of Emergency Medicine, The Ohio State University, Columbus, OH, USA; Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Yusuf Ali Ashraf
- Division of Basic and Translational Research, Department of Emergency Medicine, The Ohio State University, Columbus, OH, USA; Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Sarah Kathleen Mikula
- Center for Electron Microscopy and Analysis, The Ohio State University, Columbus, OH, USA
| | - Dinender Kumar Singla
- Biomolecular Science Center, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Mahmood Khan
- Division of Basic and Translational Research, Department of Emergency Medicine, The Ohio State University, Columbus, OH, USA; Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, USA; Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
2
|
He Z, Cao J, Wang X, Yang S, Gao H, Yu Y, Di Z, Peng C. Single-cell analyses unravel ecosystem dynamics and intercellular crosstalk during gallbladder cancer malignant transformation. Hepatol Commun 2025; 9:e0697. [PMID: 40377484 PMCID: PMC12088638 DOI: 10.1097/hc9.0000000000000697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Accepted: 03/04/2025] [Indexed: 05/18/2025] Open
Abstract
BACKGROUND Gallbladder cancer (GBC) is a rare but aggressive malignancy, often detected late due to early asymptomatic stages. Understanding cellular and molecular changes from normal tissue to high-grade intraepithelial neoplasia (HGIN) and invasive GBC is vital for identifying early biomarkers and therapeutic targets. METHODS We performed single-cell RNA sequencing on 98,113 cells derived from 2 normal adjacent tissues (NAT), 2 HGIN, and 6 GBC samples. The cellular diversity and heterogeneity, particularly within epithelial and immune cell populations in NAT-HGIN-GBC, were investigated utilizing single-cell RNA sequencing, bulk RNA sequencing (bulk RNA-seq), and 10 machine learning methodologies. Furthermore, the intercellular crosstalk between epithelial cells and tumor immune microenvironment cells was examined and validated through multiplex immunofluorescence staining. RESULTS The constructed cell atlas elucidated alterations in the immune landscape across various states of NAT-HGIN-GBC, highlighting a more pronounced inhibitory immune microenvironment in GBC. The epithelial subtype TOP2A+ Epi is markedly elevated in GBC and is correlated with a poor prognosis. Key genes associated with this subtype may include GMNN, CYTOR, KLK6, and BIRC5. Similarly, immunosuppressive macrophages, identified as TOP2A+ Macro, also increase along the NAT-HGIN-GBC sequence and are linked to reduced patient survival. Furthermore, TOP2A+ Macro and CD8+ exhausted T cells (CD8+ Tex) engage in intercellular communication with epithelial TOP2A+Epi cells via the TWEAK/FN14 signaling pathway, thereby promoting tumor progression and immune evasion in GBC. The findings were further corroborated through multiplex immunofluorescence staining conducted on specimens from patients. CONCLUSIONS This study elucidates significant alteration in the cellular ecosystems and intercellular signaling within the tumor immune microenvironment across the NAT-HGIN-GBC sequence. It identifies TOP2A, TWEAK, and FN14 as potential biomarkers and therapeutic targets for GBC.
Collapse
Affiliation(s)
- Zhaobin He
- Department of Hepatobiliary Surgery, Qilu Hospital, Shandong University, Jinan, Shandong Province, China
- Department of Hepatobiliary Minimally Invasive Surgery, Shandong University Institute of Endoscopic Minimally Invasive Surgery, Jinan, Shandong Province, China
| | - Jianqiang Cao
- Department of Hepatobiliary Surgery, Qilu Hospital, Shandong University, Jinan, Shandong Province, China
- Department of Hepatobiliary Minimally Invasive Surgery, Shandong University Institute of Endoscopic Minimally Invasive Surgery, Jinan, Shandong Province, China
| | - Xiqiang Wang
- Department of Hepatobiliary Surgery, Qilu Hospital, Shandong University, Jinan, Shandong Province, China
- Department of Hepatobiliary Minimally Invasive Surgery, Shandong University Institute of Endoscopic Minimally Invasive Surgery, Jinan, Shandong Province, China
| | - Shengbiao Yang
- Department of Hepatobiliary Surgery, Qilu Hospital, Shandong University, Jinan, Shandong Province, China
- Department of Hepatobiliary Minimally Invasive Surgery, Shandong University Institute of Endoscopic Minimally Invasive Surgery, Jinan, Shandong Province, China
| | - Huijie Gao
- Department of Hepatobiliary Surgery, Qilu Hospital, Shandong University, Jinan, Shandong Province, China
- Department of Hepatobiliary Minimally Invasive Surgery, Shandong University Institute of Endoscopic Minimally Invasive Surgery, Jinan, Shandong Province, China
| | - Yongzhe Yu
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| | - Zequn Di
- Department of Hepatobiliary Minimally Invasive Surgery, Shandong University Institute of Endoscopic Minimally Invasive Surgery, Jinan, Shandong Province, China
- Department of Clinical Medicine, School of Basic Medical Sciences Nanchang University, Nanchang, Jiangxi Province, China
| | - Cheng Peng
- Department of Hepatobiliary Surgery, Qilu Hospital, Shandong University, Jinan, Shandong Province, China
- Department of Hepatobiliary Minimally Invasive Surgery, Shandong University Institute of Endoscopic Minimally Invasive Surgery, Jinan, Shandong Province, China
| |
Collapse
|
3
|
Chen Z, Wang J, He T, Rao D, Wang Z, Zhu J. Vincristine exerts antiglioma effects by inhibiting the PI3K/AKT signaling pathway: A mechanistic study based on network pharmacology, bioinformatics analysis, and experimental validation. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:7107-7120. [PMID: 39718608 DOI: 10.1007/s00210-024-03693-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 11/27/2024] [Indexed: 12/25/2024]
Abstract
In clinical settings, glioma patients often develop secondary resistance to first-line chemotherapy drugs. Vincristine has been reported for its application in cancer chemotherapy, but its molecular mechanism of action remains unclear. This study aimed to identify potential targets of vincristine in glioma using network pharmacology and to experimentally validate the possible molecular mechanisms against glioma. First, the potential targets of vincristine were predicted using CTD, SwissTargetPrediction, and TargetNet databases. Differential expression analysis and WGCNA algorithm were employed on glioma data from the GEO database to obtain important glioma-related target genes, which were then used to identify the anti-glioma targets of vincristine. The intersecting targets were input into the String database to construct a PPI network, and core targets were identified using the cytohubba plugin in Cytoscape. GO and KEGG analyses were conducted to investigate the functional and pathway enrichment of the intersecting targets. The expression and prognostic significance of the core targets were validated using data from the TCGA and HPA databases. Finally, the anti-glioma proliferation effect of vincristine was validated through CCK-8 assay, flow cytometry for cell cycle analysis, RT-qPCR, and Western blotting. A total of 175 vincristine targets and 1673 glioma targets were identified, with 11 shared targets between vincristine and glioma tissues. Network pharmacology studies suggested that CDC25B, CDK4, CDK6, TOP2A, and the PI3K/AKT signaling pathway might be important core targets and pathways through which vincristine exerts its anti-glioma effects. In vitro experiments confirmed that vincristine successfully inhibited U87 cell proliferation and induced G1 phase arrest via the PI3K/AKT signaling pathway, thereby reducing cell growth. The study results indicate that the PI3K/AKT signaling pathway may be involved in the mechanism by which vincristine inhibits the proliferation of glioma cells.
Collapse
Affiliation(s)
- Zhihua Chen
- Department of Neurosurgery, the Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Donghu District, Nanchang City, Jiangxi, 330006, China
| | - Jiahong Wang
- Department of Neurosurgery, the Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Donghu District, Nanchang City, Jiangxi, 330006, China
| | - Ting He
- Department of Neurosurgery, the Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Donghu District, Nanchang City, Jiangxi, 330006, China
| | - Donggen Rao
- Department of Neurosurgery, the Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Donghu District, Nanchang City, Jiangxi, 330006, China
| | - Ziyang Wang
- Department of Neurosurgery, the Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Donghu District, Nanchang City, Jiangxi, 330006, China
| | - Jianming Zhu
- Department of Neurosurgery, the Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Donghu District, Nanchang City, Jiangxi, 330006, China.
| |
Collapse
|
4
|
Yang Y, Liu S, Xiao X. TOP2A Promotes Proliferation, Migration, and Inflammatory Response in M5-Treated Keratinocytes by Binding CTBP1 to Activate Wnt/β-Catenin Signaling. Cell Biochem Biophys 2025; 83:2101-2113. [PMID: 39565516 DOI: 10.1007/s12013-024-01620-2] [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: 11/08/2024] [Indexed: 11/21/2024]
Abstract
Psoriasis is a chronic cutaneous disease, affecting a significant portion of the global population. Topoisomerase II alpha (TOP2A) is upregulated in psoriasis samples, but the precise molecular mechanism remains unclear. We aimed to clarify the biological contribution of TOP2A in psoriasis progression. An in vitro psoriasis model was established on M5-induced keratinocytes (HaCaT cells) to simulate the psoriasis-like alterations. Following TOP2A knockdown without or with c terminal binding protein 1 (CTBP1) overexpression, CCK-8 and EDU staining were employed to analyze the viability and proliferation of HaCaT cells under M5 conditions. The capacities of HaCaT cell migration and invasion were examined with wound healing- and transwell assays. RT-qPCR and immunoblotting were adopted for evaluation of the inflammation and differentiation of M5-stimualted HaCaT cells. Additionally, the binding between TOP2A and CTBP1 was predicated using bioinformatics tools and detected by Co-IP. Finally, the expression of proteins in Wnt/β-catenin signaling was analyzed with the application of immunoblotting. Results suggested that TOP2A was upregulated in psoriasis skin lesions and M5-induced HaCaT cells. Interference with TOP2A attenuated the proliferation, migration, invasion, and inflammatory response in M5-treated HaCaT cells. In particular, TOP2A bound to CTBP1 and upregulated CTBP1 expression in HaCaT cells. Remarkably, CTBP1 upregulation blocked the impacts of TOP2A depletion on the biological behaviors of M5-treated HaCaT cells. Besides, TOP2A deficiency upregulated DKK1 expression as well as downregulated Wnt1, β-catenin, and c-Myc expression in HaCaT cells exposed to M5, which was restored by further CTBP1 overexpression. In summary, TOP2A binds CTBP1 to activate Wnt/β-catenin signaling, thereby promoting the progression of psoriasis.
Collapse
Affiliation(s)
- Yuanwen Yang
- Department of Dermatology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan, Shanxi, China
| | - Shumei Liu
- Medical Cosmetology Department, Shenzhen Jiarong Comprehensive Outpatient Department, Shenzhen, Guangdong, China
| | - Xia Xiao
- Medical Record Room of Shanxi Traditional Chinese Medicine Hospital, Taiyuan, Shanxi, China.
| |
Collapse
|
5
|
Kubeš J, Karabanovich G, Cong ATQ, Melnikova I, Lenčová O, Kollárová P, Bavlovič Piskáčková H, Keresteš V, Applová L, Arrouye LCM, Alvey JR, Paluncic J, Witter TL, Jirkovská A, Kuneš J, Štěrbová-Kovaříková P, Austin CA, Štěrba M, Šimůnek T, Roh J, Schellenberg MJ. Topobexin targets the Topoisomerase II ATPase domain for beta isoform-selective inhibition and anthracycline cardioprotection. Nat Commun 2025; 16:4928. [PMID: 40425539 PMCID: PMC12116762 DOI: 10.1038/s41467-025-60167-9] [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: 10/28/2024] [Accepted: 05/14/2025] [Indexed: 05/29/2025] Open
Abstract
Topoisomerase II alpha and beta (TOP2A and TOP2B) isoenzymes perform essential and non-redundant cellular functions. Anthracyclines induce their potent anti-cancer effects primarily via TOP2A, but at the same time they induce a dose limiting cardiotoxicity through TOP2B. Here we describe the development of the obex class of TOP2 inhibitors that bind to a previously unidentified druggable pocket in the TOP2 ATPase domain to act as allosteric catalytic inhibitors by locking the ATPase domain conformation with the capability of isoform-selective inhibition. Through rational drug design we have developed topobexin, which interacts with residues that differ between TOP2A and TOP2B to provide inhibition that is both selective for TOP2B and superior to dexrazoxane. Topobexin is a potent protectant against chronic anthracycline cardiotoxicity in an animal model. This demonstration of TOP2 isoform-specific inhibition underscores the broader potential to improve drug specificity and minimize adverse effects in various medical treatments.
Collapse
Affiliation(s)
- Jan Kubeš
- Department of Biochemical Sciences, Faculty of Pharmacy in Hradec Králové, Charles University; Hradec, Králové, 500 03, Czech Republic
| | - Galina Karabanovich
- Department of Organic and Bioorganic Chemistry, Faculty of Pharmacy in Hradec Králové, Charles University; Hradec, Králové, 500 03, Czech Republic
| | - Anh T Q Cong
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, 55905, MN, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, 55905, MN, USA
| | - Iuliia Melnikova
- Department of Organic and Bioorganic Chemistry, Faculty of Pharmacy in Hradec Králové, Charles University; Hradec, Králové, 500 03, Czech Republic
| | - Olga Lenčová
- Department of Pharmacology, Faculty of Medicine in Hradec Králové, Charles University; Hradec, Králové, 500 03, Czech Republic
| | - Petra Kollárová
- Department of Pharmacology, Faculty of Medicine in Hradec Králové, Charles University; Hradec, Králové, 500 03, Czech Republic
| | - Hana Bavlovič Piskáčková
- Department of Pharmaceutical Chemistry and Pharmaceutical Analysis, Faculty of Pharmacy in Hradec Králové, Charles University; Hradec, Králové, 500 03, Czech Republic
| | - Veronika Keresteš
- Department of Biochemical Sciences, Faculty of Pharmacy in Hradec Králové, Charles University; Hradec, Králové, 500 03, Czech Republic
| | - Lenka Applová
- Department of Biochemical Sciences, Faculty of Pharmacy in Hradec Králové, Charles University; Hradec, Králové, 500 03, Czech Republic
| | - Lise C M Arrouye
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, 55905, MN, USA
| | - Julia R Alvey
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, 55905, MN, USA
| | - Jasmina Paluncic
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, 55905, MN, USA
| | - Taylor L Witter
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, 55905, MN, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, 55905, MN, USA
| | - Anna Jirkovská
- Department of Biochemical Sciences, Faculty of Pharmacy in Hradec Králové, Charles University; Hradec, Králové, 500 03, Czech Republic
| | - Jiří Kuneš
- Department of Organic and Bioorganic Chemistry, Faculty of Pharmacy in Hradec Králové, Charles University; Hradec, Králové, 500 03, Czech Republic
| | - Petra Štěrbová-Kovaříková
- Department of Pharmaceutical Chemistry and Pharmaceutical Analysis, Faculty of Pharmacy in Hradec Králové, Charles University; Hradec, Králové, 500 03, Czech Republic
| | - Caroline A Austin
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, NE1 7RU, UK.
| | - Martin Štěrba
- Department of Pharmacology, Faculty of Medicine in Hradec Králové, Charles University; Hradec, Králové, 500 03, Czech Republic.
| | - Tomáš Šimůnek
- Department of Biochemical Sciences, Faculty of Pharmacy in Hradec Králové, Charles University; Hradec, Králové, 500 03, Czech Republic.
| | - Jaroslav Roh
- Department of Organic and Bioorganic Chemistry, Faculty of Pharmacy in Hradec Králové, Charles University; Hradec, Králové, 500 03, Czech Republic.
| | - Matthew J Schellenberg
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, 55905, MN, USA.
| |
Collapse
|
6
|
Goli M, Sandilya V, Ghandour B, Hajj HE, Kobeissy F, Darwiche N, Mechref Y. Exploring the Anti-Leukemic Effect of the Synthetic Retinoid ST1926 on Malignant T Cells: A Comprehensive Proteomics Approach. Int J Mol Sci 2025; 26:4651. [PMID: 40429796 PMCID: PMC12111145 DOI: 10.3390/ijms26104651] [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: 02/10/2025] [Revised: 05/01/2025] [Accepted: 05/10/2025] [Indexed: 05/29/2025] Open
Abstract
T-cell malignancies represent a group of complex cancers arising from T cells and include aggressive subtypes such as Adult T-cell Leukemia/Lymphoma (ATL) and T-cell Acute Lymphoblastic Leukemia (T-ALL). Patients with these aggressive subtypes still represent an unmet medical condition. The synthetic adamantyl retinoid ST1926, a potent DNA polymerase-α inhibitor, proved a promising potency in preclinical models of ATL and peripheral T-cell lymphoma. Using advanced liquid chromatography-mass spectrometry (LC-MS/MS) techniques, we explored the effects of ST1926 on global protein expression in ATL (HuT-102) and T-ALL (MOLT-4) cells. We demonstrate that ST1926 triggers differentiation and apoptosis in malignant T-cells while halting tumor progression. Evidence at the proteomics level reveals the impact of ST1926 on crucial DNA replication enzymes and cell cycle regulation, highlighting its potential to reduce leukemogenesis and promote apoptosis. Our findings underscore the potential of ST1926 as an innovative therapeutic approach to address these aggressive T-cell malignancies, providing valuable insights into developing new targeted therapies and improving the outcomes and prognosis of patients with these challenging diseases.
Collapse
Affiliation(s)
- Mona Goli
- Chemistry and Biochemistry Department, Texas Tech University, Lubbock, TX 79409, USA; (M.G.); (V.S.)
| | - Vishal Sandilya
- Chemistry and Biochemistry Department, Texas Tech University, Lubbock, TX 79409, USA; (M.G.); (V.S.)
| | - Botheina Ghandour
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut 1107 2020, Lebanon; (B.G.); (F.K.); (N.D.)
| | - Hiba El Hajj
- Department of Experimental Pathology, Immunology and Microbiology, American University of Beirut, Beirut 1107 2020, Lebanon;
| | - Firas Kobeissy
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut 1107 2020, Lebanon; (B.G.); (F.K.); (N.D.)
- Center for Neurotrauma, Multiomics & Biomarkers, Department of Neurobiology, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Nadine Darwiche
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut 1107 2020, Lebanon; (B.G.); (F.K.); (N.D.)
| | - Yehia Mechref
- Chemistry and Biochemistry Department, Texas Tech University, Lubbock, TX 79409, USA; (M.G.); (V.S.)
| |
Collapse
|
7
|
Wang YF, Zheng D, Zhang Y, Li XF, Xia M, Tang HM, Huang CH, Li MJ, Lou DD. Renal tubular epithelial cell related partial epithelial-mesenchymal transition in AAⅠ induced renal fibrosis via Wnt7b/β-catenin signaling. Front Pharmacol 2025; 16:1571960. [PMID: 40432885 PMCID: PMC12106489 DOI: 10.3389/fphar.2025.1571960] [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: 02/06/2025] [Accepted: 04/17/2025] [Indexed: 05/29/2025] Open
Abstract
Introduction This study investigates the pathological progressions in kidneys affected by aristolochic acid nephropathy (AAN) and explores the molecular mechanisms underlying the fibrotic process, specifically focusing on the Wnt7b/β-catenin signaling pathway. Methods Both mice and human kidney-2 (HK-2) cells were treated with aristolochic acid I (AAI). In mice, we monitored blood urea nitrogen (BUN), serum creatinine (Scr), kidney injury molecule-1 (KIM-1), pathological modifications of renal tubular epithelial cells (RTECs), and fibrosis degrees during acute/chronic disease phases. Wnt7b/β-catenin expression was evaluated through transcriptome analysis and laboratory assays (immunohistochemistry, Western blotting, immunoelectron microscopy) in acute AAN and cultured cells. Concurrent assays measured representative proteins: Aquaporin 1 (AQP1), Topoisomerase IIα (TOP2A), Vascular Cell Adhesion Molecule-1 (VCAM-1), and α-smooth muscle actin (α-SMA) in chronic AAN RTECs. Results AAI increased Scr, BUN, and KIM-1 levels by causing RTEC necrotic shedding in acute stages and promoted renal interstitial fibrosis chronically. Elevated Wnt7b pathway proteins enhanced damaged RTEC regeneration, with regenerated cells expressing mesenchymal proteins VCAM-1 and α-SMA. Discussion The Wnt7b/β-catenin signaling pathway connects acute tubule damage to fibrosis, explaining AAN's pathological continuum. These findings clarify how acute injury progresses to chronic fibrosis in AAN.
Collapse
Affiliation(s)
- Yi-Fan Wang
- Judicial Appraisal Center, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
- Key Laboratory of Forensic Toxicology of Herbal Medicines, Guizhou Education Department, Guiyang, Guizhou, China
| | - Dan Zheng
- Guiyang Maternal and Child Healthcare Hospital, Guiyang, Guizhou, China
| | - Ying Zhang
- Judicial Appraisal Center, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
- Key Laboratory of Forensic Toxicology of Herbal Medicines, Guizhou Education Department, Guiyang, Guizhou, China
| | - Xiao-Fen Li
- Judicial Appraisal Center, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
- Key Laboratory of Forensic Toxicology of Herbal Medicines, Guizhou Education Department, Guiyang, Guizhou, China
| | - Ming Xia
- Judicial Appraisal Center, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
- Key Laboratory of Forensic Toxicology of Herbal Medicines, Guizhou Education Department, Guiyang, Guizhou, China
| | - Hai-Ming Tang
- Judicial Appraisal Center, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
- Key Laboratory of Forensic Toxicology of Herbal Medicines, Guizhou Education Department, Guiyang, Guizhou, China
| | - Chun-Hua Huang
- Judicial Appraisal Center, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
- Key Laboratory of Forensic Toxicology of Herbal Medicines, Guizhou Education Department, Guiyang, Guizhou, China
| | - Mao-Juan Li
- Judicial Appraisal Center, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
- Key Laboratory of Forensic Toxicology of Herbal Medicines, Guizhou Education Department, Guiyang, Guizhou, China
| | - Di-Dong Lou
- Judicial Appraisal Center, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
- Key Laboratory of Forensic Toxicology of Herbal Medicines, Guizhou Education Department, Guiyang, Guizhou, China
| |
Collapse
|
8
|
Qin J, Huang D, Li C, Liu Y, Feng Q, Lan Y, Huang X. Integrative PPI network and random forest analysis identifies KIF4A, TOP2A, and ASPM protein macromolecules as novel protein biomarkers in renal carcinoma pathogenesis. Int J Biol Macromol 2025; 311:144058. [PMID: 40345279 DOI: 10.1016/j.ijbiomac.2025.144058] [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: 03/22/2025] [Revised: 05/02/2025] [Accepted: 05/07/2025] [Indexed: 05/11/2025]
Abstract
The pathogenesis of kidney cancer is not fully understood, so there is an urgent need to identify new biomarkers to improve diagnosis and treatment. The study identified KIF4A, TOP2A and ASPM as novel protein biomarkers for renal cancer and explored their biological functions in the development and progression of renal cancer. A variety of bioinformatics methods were used to process and batch calibrate the transcriptome data of renal cancer. Differential gene expression analysis was performed using Limma packages, followed by functional enrichment analysis to identify biological pathways associated with kidney cancer. Construct a protein-protein interaction (PPI) network to prioritize core genes and use machine learning methods to further screen key signaling pathways. Epithelial-mesenchymal transition (EMT) marker score was used to evaluate the prognosis of renal carcinoma, and the results were validated by cell culture experiment. Integrated principal component analysis (PCA) showed significant effect of batch correction in the renal cancer cohort. Transcriptome analysis revealed dysregulation of conserved gene expression in renal carcinoma. Functional enrichment analysis showed that metabolic and immune signaling pathways play an important role in the pathogenesis of renal cancer. The integrated PPI network prioritising KIF4A, TOP2A and ASPM as key mitotic regulators and further confirming these three as core carcinogenic drivers through machine learning. Finally, integrated prognostic analyses identified genetic features associated with EMT that are clinically significant.
Collapse
Affiliation(s)
- Junkai Qin
- Department of Urology, Minzu Hospital of Guangxi Zhuang Autonomous Region, Nanning 530001, China
| | - Dongjun Huang
- Department of Urology, Minzu Hospital of Guangxi Zhuang Autonomous Region, Nanning 530001, China
| | - Chaobin Li
- Department of Urology, Minzu Hospital of Guangxi Zhuang Autonomous Region, Nanning 530001, China
| | - Yu Liu
- Department of Urology, Minzu Hospital of Guangxi Zhuang Autonomous Region, Nanning 530001, China
| | - Qincong Feng
- Pathology Department, The 923rd Hospital of the Chinese People's Liberation Army Joint Logistics Support Force, Guangxi, 530000, China
| | - Yibi Lan
- Department of Urology, Minzu Hospital of Guangxi Zhuang Autonomous Region, Nanning 530001, China.
| | - Xijian Huang
- Department of Urology, Minzu Hospital of Guangxi Zhuang Autonomous Region, Nanning 530001, China.
| |
Collapse
|
9
|
Qu S, Guan X, Li H, Yang J, Sun P, Song C, Surzenko N, Wang Y. Dietary Intake of Octanoic Acid Restores UBE3A Expression and Improves the Behavioral Phenotypes in a Mouse Model of Angelman Syndrome. FASEB J 2025; 39:e70559. [PMID: 40277210 PMCID: PMC12023718 DOI: 10.1096/fj.202403130rr] [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: 12/07/2024] [Revised: 04/09/2025] [Accepted: 04/14/2025] [Indexed: 04/26/2025]
Abstract
Angelman syndrome (AS) is a severe neurodevelopmental disorder with no effective therapies. Most of the behavioral deficits observed in AS patients arise from the absence of ubiquitin protein ligase E3A (UBE3A) in the brain during development, driven by the loss of maternally expressed UBE3A and silencing of the paternal copy of this gene through imprinting. Safe and effective therapies aiming at restoring the expression of the paternal UBE3A gene early in human life are currently lacking. In this study, we investigated whether octanoic acid (OA), a medium-chain fatty acid, could unsilence the paternal Ube3a allele in neurons and ameliorate the behavioral defects in a murine model of AS. To this end, Ube3am-/p+ and Ube3am+/pYFP mice, as well as their wild-type littermates, were fed either a control or OA-supplemented diet from postnatal day 0 through adulthood, and the improvements in AS-related cellular and behavioral deficits were characterized. We demonstrate that dietary intake of OA activates the expression of the silenced, paternal Ube3a in neurons and improves select AS behavioral phenotypes in mice. We further show that downregulation of topoisomerase II beta and restoration of dendritic spine development may underlie the unsilencing of Ube3a and the behavioral improvements in OA-supplemented animals, respectively. Together, our findings suggest that dietary supplementation with OA could serve as an early, safe, and clinically feasible therapeutic strategy for reactivation of the paternal UBE3A allele in patients with AS.
Collapse
Affiliation(s)
- Song Qu
- Department of Medical Genetics, College of Basic Medical ScienceArmy Medical University (Third Military Medical University)ChongqingChina
| | - Xingying Guan
- Department of Medical Genetics, College of Basic Medical ScienceArmy Medical University (Third Military Medical University)ChongqingChina
| | - Hongyan Li
- Department of Medical Genetics, College of Basic Medical ScienceArmy Medical University (Third Military Medical University)ChongqingChina
| | - Jian Yang
- Undergraduate Student Brigade, College of Basic Medical ScienceArmy Medical University (Third Military Medical University)ChongqingChina
| | - Pu Sun
- Maternal and Child Health Hospital of Yongchuan DistrictChongqingChina
| | - Cui Song
- Department of Endocrinology and Genetic Metabolism Disease, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of PediatricsChildren's Hospital of Chongqing Medical UniversityChongqingChina
| | | | - Yanyan Wang
- Department of Medical Genetics, College of Basic Medical ScienceArmy Medical University (Third Military Medical University)ChongqingChina
| |
Collapse
|
10
|
Saha S, Zhang Y, Gibert MK, Dube C, Hanif F, Mulcahy E, Bednarek S, Marcinkiewicz P, Wang X, Kwak G, Hudson K, Sun Y, Dinda M, Saha T, Guessous F, Cruickshanks N, Colon RR, Dell'Olio LG, Anbu R, Kefas B, Kumar P, Klibanov AL, Schiff D, Suk JS, Hanes J, Mata J, Hafner M, Abounader R. Discovery and therapeutic exploitation of Master Regulatory miRNAs in Glioblastoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.01.646663. [PMID: 40236125 PMCID: PMC11996502 DOI: 10.1101/2025.04.01.646663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Glioblastoma is a fatal primary malignant brain tumor. Despite therapies involving surgical resection, chemotherapy, and radiation therapy, the average survival for glioblastoma patients remains at approximately 15 months. MicroRNAs (miRNAs) are short noncoding RNA molecules that regulate the expression of the majority of human genes. Numerous genes are concurrently deregulated in glioblastoma. Consequently, molecular monotherapies have failed to achieve improvements in clinical outcomes. Several lines of evidence suggest that simultaneous targeting of several deregulated molecules is required to achieve better therapies. However, the simultaneous targeting of several deregulated oncogenic drivers is severely limited by the fact that the drugs needed to target many deregulated molecules do not currently exist, and because combining several drugs in a clinical setting leads to an exponential increase in toxicity. We hypothesized that we can develop and use miRNA to simultaneously inhibit multiple deregulated genes for more efficacious glioblastoma therapies. The goal of this study was therefore to identify master regulatory microRNAs (miRNAs) and use them to simultaneously target multiple deregulated molecules for GBM therapy. We defined master regulatory miRNAs as those that target several deregulated genes in glioblastoma. To find master regulatory miRNAs, we first used PAR-CLIP screenings to identify all targets of all miRNAs in glioblastoma cells. We then analyzed TCGA tumor data to determine which of these targets are deregulated in human tumors. We developed and used an algorithm to rank these targets for significance in glioblastoma malignancy based on their magnitude of deregulation, frequency of deregulation, and correlation with patient survival. We then ranked the miRNAs for their capacity of targeting multiple glioblastoma-deregulated genes and therefore the potential to exhibit strong anti-tumor effects when delivered as therapy. Using this strategy, we selected two tumor suppressor master regulatory miRNAs, miR-340, miR-382 and an oncogenic master regulatory miRNA, miR-17. We validated the target genes of the miRNAs and showed that they form part of important glioblastoma regulatory pathways. We then showed that the miRNAs (miR-340 and miR-582) or the miR-17 inhibitor have strong inhibitory effects on glioblastoma cell growth, survival, invasion, stemness and in vivo tumor growth. Ultimately, we developed and successfully tested a new therapeutic approach to delivery miR-340 using MRI guided focused ultrasound and microbubbles (FUS-MB) and special brain penetrating nanoparticles (BPN). This approach resulted in a substantial reduction in tumor volume and prolongation of the survival of glioblastoma-bearing mice and can be translated into clinical trials. We therefore developed and successfully tested a novel strategy to discover and deliver miRNAs for glioblastoma and cancer therapy.
Collapse
|
11
|
Quan H, Yin H, Wang Z, Lv Y, Sun Q, Yin T. Identification of key hub genes and potential therapeutic drugs for nasopharyngeal carcinoma: Insights into molecular mechanisms and treatment strategies. Braz J Otorhinolaryngol 2025; 91:101618. [PMID: 40286596 PMCID: PMC12056400 DOI: 10.1016/j.bjorl.2025.101618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 03/03/2025] [Accepted: 03/29/2025] [Indexed: 04/29/2025] Open
Abstract
OBJECTIVE Nasopharyngeal Carcinoma (NPC) is a highly malignant cancer with a high incidence in East and Southeast Asia, including southern China. Despite advances in treatment, the prognosis for advanced NPC remains poor due to high recurrence and metastasis rates. The molecular mechanisms driving NPC progression are not fully understood, and identifying key genes and potential therapeutic agents is critical. This study aims to uncover critical genes and screen therapeutic drugs, providing insights into NPC pathogenesis and novel treatment strategies. METHODS Three GEO datasets (GSE12452, GSE53819, and GSE61218) were analyzed to identify overlapping Differentially Expressed Genes (DEGs) in NPC. Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and Gene Set Enrichment Analysis (GSEA) were used to explore the biological roles of DEGs. Protein-Protein Interaction (PPI) and mRNA-miRNA-lncRNA interaction networks were constructed to identify key hub genes. Potential therapeutic drugs were predicted via a Drug-Gene Interaction network. The overexpression of hub genes was validated in NPC cells using CCK-8 assays, and the anti-proliferative effects of three drugs ‒ valproic acid, cyclosporine, and calcitriol ‒ were tested. RESULTS Eight hub genes (ASPM, BIRC5, BUB1B, CDK1, KIF23, PBK, TOP2A, and TTK) were identified, with ASPM reported for the first time in the context of NPC. Overexpression of these genes significantly promoted NPC cell proliferation. Among the tested drugs, calcitriol exhibited the most potent anti-proliferative effect, with IC50 values of 0.90 μM, 0.47 μM, and 0.31 μM at 24-, 48-, and 72-hs, respectively. CONCLUSION This study identified eight key genes as potential biomarkers for NPC and validated calcitriol as a promising therapeutic agent, providing a foundation for further research into NPC treatment. LEVEL OF EVIDENCE Level 2 (Individual cross-sectional studies or systematic review of surveys that allow matching to local circumstances).
Collapse
Affiliation(s)
- Haiyan Quan
- Hunan Polytechnic of Environment and Biology, Hengyang, Hunan, China
| | - Hongguo Yin
- Hunan Polytechnic of Environment and Biology, Hengyang, Hunan, China
| | - Zhen Wang
- Hunan Polytechnic of Environment and Biology, Hengyang, Hunan, China
| | - Yuan Lv
- Hunan Polytechnic of Environment and Biology, Hengyang, Hunan, China
| | - Qiong Sun
- Hunan Polytechnic of Environment and Biology, Hengyang, Hunan, China
| | - Ting Yin
- Department of Ophthalmology, The Second Affiliated Hospital, University of South China, Hengyang, Hunan, China.
| |
Collapse
|
12
|
Kang X, Li X, Zhou J, Zhang Y, Qiu L, Tian C, Deng Z, Liang X, Zhang Z, Du S, Hu S, Wang N, Yue Z, Xu Y, Gao Y, Dai J, Wang Z, Yu C, Chen J, Wu Y, Chen L, Yao Y, Yao S, Yang X, Yan L, Wen Q, Depies OM, Chan K, Liang X, Li G, Zi Z, Liu X, Gan H. Extrachromosomal DNA replication and maintenance couple with DNA damage pathway in tumors. Cell 2025:S0092-8674(25)00414-3. [PMID: 40300601 DOI: 10.1016/j.cell.2025.04.012] [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: 10/18/2024] [Revised: 02/19/2025] [Accepted: 04/06/2025] [Indexed: 05/01/2025]
Abstract
Extrachromosomal DNA (ecDNA) drives the evolution of cancer cells. However, the functional significance of ecDNA and the molecular components involved in its replication and maintenance remain largely unknown. Here, using CRISPR-C technology, we generated ecDNA-carrying (ecDNA+) cell models. By leveraging these models alongside other well-established systems, we demonstrated that ecDNA can replicate and be maintained in ecDNA+ cells. The replication of ecDNA activates the ataxia telangiectasia mutated (ATM)-mediated DNA damage response (DDR) pathway. Topoisomerases, such as TOP1 and TOP2B, play a role in ecDNA replication-induced DNA double-strand breaks (DSBs). A subset of these elevated DSBs persists into the mitotic phase and is primarily repaired by the alternative non-homologous end joining (alt-NHEJ) pathway, which involves POLθ and LIG3. Correspondingly, ecDNA maintenance requires DDR, and inhibiting DDR impairs the circularization of ecDNA. In summary, we demonstrate reciprocal interactions between ecDNA maintenance and DDR, providing new insights into the detection and treatment of ecDNA+ tumors.
Collapse
Affiliation(s)
- Xing Kang
- Guangdong Provincial Key Laboratory of Synthetic Genomics, Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Xinran Li
- Guangdong Provincial Key Laboratory of Synthetic Genomics, Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jiaqi Zhou
- Guangdong Provincial Key Laboratory of Synthetic Genomics, Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yang Zhang
- Guangdong Provincial Key Laboratory of Synthetic Genomics, Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Lingyu Qiu
- Guangdong Provincial Key Laboratory of Synthetic Genomics, Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Congcong Tian
- Guangdong Provincial Key Laboratory of Synthetic Genomics, Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Zhiwen Deng
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Marshall Laboratory of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen, China
| | - Xiaoyan Liang
- Guangdong Provincial Key Laboratory of Synthetic Genomics, Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Ziwei Zhang
- Guangdong Provincial Key Laboratory of Synthetic Genomics, Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Songlin Du
- College of Animal Science and Technology, Northwest A&F University, Yangling, Xianyang 712100, Shaanxi, China
| | - Suili Hu
- Guangdong Provincial Key Laboratory of Synthetic Genomics, Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Nan Wang
- Guangdong Provincial Key Laboratory of Synthetic Genomics, Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Zhen Yue
- Guangdong Provincial Key Laboratory of Synthetic Genomics, Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yajing Xu
- Guangdong Provincial Key Laboratory of Synthetic Genomics, Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Yuan Gao
- Department of Pharmacology, Case Western Reserve University, School of Medicine, Cleveland, OH, USA
| | - Junbiao Dai
- CAS Key Laboratory of Quantitative Engineering Biology, Guangdong Provincial Key Laboratory of Synthetic Genomics and Shenzhen Key Laboratory of Synthetic Genomics, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Key Laboratory of Synthetic Biology, Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Zhiquan Wang
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Chuanhe Yu
- Hormel Institute, University of Minnesota, Austin, MN, USA
| | - Jinyi Chen
- Guangdong Provincial Key Laboratory of Synthetic Genomics, Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yuchun Wu
- Guangdong Provincial Key Laboratory of Synthetic Genomics, Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; School of Basic Medicine, Qingdao University, Qingdao, China
| | - Liangming Chen
- Guangdong Provincial Key Laboratory of Synthetic Genomics, Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yuan Yao
- Guangdong Provincial Key Laboratory of Synthetic Genomics, Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Sitong Yao
- Guangdong Provincial Key Laboratory of Synthetic Genomics, Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Xinran Yang
- Guangdong Provincial Key Laboratory of Synthetic Genomics, Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Lixia Yan
- Guangdong Provincial Key Laboratory of Synthetic Genomics, Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qing Wen
- Guangdong Provincial Key Laboratory of Synthetic Genomics, Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Olivia M Depies
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Kuiming Chan
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Xiaohuan Liang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Gang Li
- Centre of Reproduction, Development and Aging, Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau, China
| | - Zhike Zi
- Guangdong Provincial Key Laboratory of Synthetic Genomics, Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Xiangyu Liu
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Marshall Laboratory of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen, China
| | - Haiyun Gan
- Guangdong Provincial Key Laboratory of Synthetic Genomics, Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.
| |
Collapse
|
13
|
Ejaz H, Mushtaq M, Khan S, Azim N, Hussain A, Kakar K, Khan MZ, Hafeez A, Moeezullah S. Investigation of multi-drug resistant Candida auris using species-specific molecular markers in immunocompromised patients from a tertiary care hospital in Quetta, Pakistan. PLoS One 2025; 20:e0319485. [PMID: 40273190 PMCID: PMC12021172 DOI: 10.1371/journal.pone.0319485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 02/03/2025] [Indexed: 04/26/2025] Open
Abstract
INTRODUCTION Candida auris is an emerging multidrug-resistant pathogen responsible for nosocomial infections worldwide, characterized by high mortality rates and significant challenges in detection due to frequent misidentification. Classified by the WHO as a pathogen of critical importance since it exhibits resistance to multiple antifungal agents, particularly fluconazole, and is highly transmissible in healthcare settings. Conventional detection methods often lack the accuracy required for effective infection control. This study aimed to conduct inferential and molecular analyses of C. auris and other yeast species infecting immunocompromised patients in the Special and Intensive Care Units (SCU and ICU) of a tertiary care hospital in Quetta, Pakistan. In this region, C. auris remains rarely studied and is frequently misdiagnosed by clinical staff due to limited awareness and diagnostic challenges. Notably, no prior research has been conducted on C. auris in Quetta. The study also sought to develop reliable diagnostic methods suitable for resource-limited settings, addressing a critical gap in healthcare infrastructure. MATERIALS AND METHODS Samples (150 each) from the ear, axilla, groin, and saliva of SCU/ICU patients were collected and processed on yeast malt agar, with preliminary identification using Brilliance Candida Agar (BCA) and CHROMagar Candida Plus (CCP). Advanced techniques, including PCR amplification of ITS regions, DNA sequencing, RFLP with Msp1, MALDI-TOF, Vitek 2, and species-specific primers, were used for identification. Antifungal susceptibility to fluconazole, amphotericin B, and voriconazole were also assessed. RESULTS The culture test revealed that 42.6% samples were positive for yeast infections. In addition to detecting Candida auris in 4 cultures, chromogenic media identified 6 other Candida species: C. albicans, C. dubliniensis, C. glabrata, C. krusei, C. parapsilosis, and C. tropicalis. Further validation through advanced techniques, including molecular diagnostics and MALDI-TOF, enabled the identification of additional species: C. famata, C. kefyr, C. lusitaniae, and Meyerozyma (Candida) guilliermondii. Out of all identified yeast species C. dubliniensis was the most common, followed by C. albicans and C. tropicalis, with the highest infection rates observed in saliva samples. Antifungal Susceptibility Tests (AST) revealed that C. auris isolates were resistant to Fluconazole, Amphotericin B, and Voriconazole, highlighting multidrug resistance. This study represents the first report of novel multidrug-resistant C. auris from Quetta, Pakistan, indicating that C. auris is prevalent among ICU and SCU patients. Novel species specific primers targeting phospholipase B, topoisomerase II, CDR and 18s genes were designed in our laboratory and not previously reported in earlier studies, proved highly effective for the rapid identification of Candida species. The established protocol using these primers is recommended for implementation in resource-limited laboratory settings. The statistical analysis demonstrated significant correlations between Candida species infection (dependent variable) and several independent factors (variables) emphasizing the importance of targeted diagnostics and intervention strategies.
Collapse
Affiliation(s)
- Hira Ejaz
- Department of Biotechnology, Faculty of Life Sciences & Informatics (FLS&I), Balochistan University of Information Technology, Engineering and Management Sciences (BUITEMS), Balili, Quetta, Balochistan, Pakistan
| | - Muhammad Mushtaq
- Department of Biotechnology, Faculty of Life Sciences & Informatics (FLS&I), Balochistan University of Information Technology, Engineering and Management Sciences (BUITEMS), Balili, Quetta, Balochistan, Pakistan
| | | | - Nasir Azim
- Fatima Jinnah Chest Hospital, Quetta, Pakistan
| | - Abrar Hussain
- Department of Biotechnology, Faculty of Life Sciences & Informatics (FLS&I), Balochistan University of Information Technology, Engineering and Management Sciences (BUITEMS), Balili, Quetta, Balochistan, Pakistan
| | - Kaleemullah Kakar
- Department of Biotechnology, Faculty of Life Sciences & Informatics (FLS&I), Balochistan University of Information Technology, Engineering and Management Sciences (BUITEMS), Balili, Quetta, Balochistan, Pakistan
| | - Muhammad Zubair Khan
- Department of Mathematics, Faculty of Basic Sciences, BUITEMS, Takatu Campus, Quetta, Balochistan, Pakistan
| | - Ayisha Hafeez
- Department of Biotechnology, Faculty of Life Sciences & Informatics (FLS&I), Balochistan University of Information Technology, Engineering and Management Sciences (BUITEMS), Balili, Quetta, Balochistan, Pakistan
| | - Syed Moeezullah
- Department of Biotechnology, Faculty of Life Sciences & Informatics (FLS&I), Balochistan University of Information Technology, Engineering and Management Sciences (BUITEMS), Balili, Quetta, Balochistan, Pakistan
| |
Collapse
|
14
|
Li S, Cui Z, Gao M, Shan Y, Ren Y, Zhao Y, Wang D, Meng T, Liu H, Yin Z. Hsa_circ_0072088 promotes non-small cell lung cancer progression through modulating miR-1270/TOP2A axis. Cancer Cell Int 2025; 25:156. [PMID: 40259294 PMCID: PMC12010575 DOI: 10.1186/s12935-025-03749-3] [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: 05/10/2024] [Accepted: 03/12/2025] [Indexed: 04/23/2025] Open
Abstract
According to the data released by the International Agency for Research on Cancer (IARC) in 2020, lung cancer ranks second among newly diagnosed malignant tumors globally. As a special class of non-coding RNA, circRNA has become a new hotspot in the field of biomarker research. With the continuous deepening of molecular-level investigations, the underlying mechanisms of circRNA are being gradually unveiled. The more widely studied mechanism is the competitive endogenous RNA mechanism of circRNA. Studies related to circRNA expression were searched in GEO database and statistically analyzed using the "limma" package and weighted gene co-expression network analysis. The expression of circRNA, microRNA and mRNA in cells and tissues were examined via qRT-PCR. MTS assay was used to measure cell proliferation, Transwell assay was used to measure cell migration, and apoptosis assay was carried out to detect cell apoptosis. Additionally, a dual-luciferase reporter assay was further executed to explore the targeted binding relationships between circRNA-microRNA and microRNA-mRNA. It was discovered that hsa_circRNA_103809 was differentially highly expressed in non-small cell lung cancer cells, whereas miR-1270 was differentially lowly expressed. The knockdown of circ_0072088 inhibited the cell proliferation and migration, while promoting cell apoptosis. The same biological function was found with the overexpression of miR-1270. The rescue experiment further validated that circ_0072088 could regulate the biological function of cells by influencing miR-1270. Finally, the targeted binding relationship was verified by dual luciferase reporting experiment. In conclusion, circ_0072088 is differentially highly expressed in non-small cell lung cancer and can affect the progression of non-small cell lung cancer through the circ_0072088/miR-1270/TOP2A axis.
Collapse
Affiliation(s)
- Sixuan Li
- Postdoctoral Research Station, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, 110042, China
- Department of Epidemiology, School of Public Health, China Medical University, Shenyang, 110122, Liaoning, China
| | - Zhigang Cui
- School of Nursing, China Medical University, Shenyang, 110122, Liaoning, China
| | - Min Gao
- Department of Epidemiology, School of Public Health, China Medical University, Shenyang, 110122, Liaoning, China
| | - Yanan Shan
- Department of Epidemiology, School of Public Health, China Medical University, Shenyang, 110122, Liaoning, China
| | - Yihong Ren
- Department of Epidemiology, School of Public Health, China Medical University, Shenyang, 110122, Liaoning, China
| | - Yuxin Zhao
- Department of Epidemiology, School of Public Health, China Medical University, Shenyang, 110122, Liaoning, China
| | - Di Wang
- Department of Epidemiology, School of Public Health, China Medical University, Shenyang, 110122, Liaoning, China
| | - Tingyu Meng
- Department of Epidemiology, School of Public Health, China Medical University, Shenyang, 110122, Liaoning, China
| | - Hongxu Liu
- Department of Thoracic Surgery, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, 110042, China.
| | - Zhihua Yin
- Department of Epidemiology, School of Public Health, China Medical University, Shenyang, 110122, Liaoning, China.
| |
Collapse
|
15
|
Liu J, Li H, Lin X, Xiong J, Wu G, Ding L, Lin B. Deciphering the heterogeneity of epithelial cells in pancreatic ductal adenocarcinoma: implications for metastasis and immune evasion. World J Surg Oncol 2025; 23:144. [PMID: 40240899 PMCID: PMC12004766 DOI: 10.1186/s12957-025-03793-3] [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: 02/05/2025] [Accepted: 03/29/2025] [Indexed: 04/18/2025] Open
Abstract
OBJECTIVE This study examines the cellular heterogeneity of epithelial cells within pancreatic ductal adenocarcinoma (PDAC) and their contributions to tumor progression, metastasis, and immunosuppressive interactions using single-cell RNA sequencing. METHODS Single-cell RNA-sequencing data from two datasets (GSE154778 and GSE158356) were integrated using the Harmony algorithm, followed by quality control, clustering, and differential gene expression analysis. Distinct subpopulations of epithelial cells were identified, and their gene expression profiles were analyzed. To assess the malignancy of these subpopulations, single-cell copy number variation (CNV) analysis and trajectory analysis were conducted. Additionally, intercellular communication was examined using the CellChat platform. RESULTS The analysis revealed pronounced heterogeneity among PDAC epithelial cells, with specific subpopulations exhibiting distinct roles in tumor proliferation, extracellular matrix remodeling, and metastatic dissemination. Subpopulations 4 and 6 were characterized by increased CNV levels and a more malignant phenotype, suggesting an enhanced capacity for metastasis. Single-cell trajectory analysis, along with CellChat, mapped the temporal evolution of epithelial cells, identifying key regulatory genes such as DCBLD2 and JUN. A prognostic model incorporating five key genes, including KLF6, was developed and demonstrated strong predictive accuracy for patient outcomes. Notably, KLF6 emerged as a critical prognostic marker associated with immune modulation, particularly through interactions with M2 macrophages. CONCLUSION The study highlights the pronounced heterogeneity of epithelial cells in PDAC and their distinct contributions to tumor progression, metastasis, and immune modulation. Through single-cell transcriptomic and CNV analyses, we identified epithelial subpopulations with varying malignant potentials and distinct interactions with the tumor microenvironment. Among these, KLF6 emerged as a key regulator associated with immune modulation and metastasis. Our findings emphasize the significance of epithelial cell heterogeneity in shaping pancreatic cancer progression. These insights provide a foundation for future investigations into novel prognostic markers and therapeutic strategies.
Collapse
Affiliation(s)
- Jie Liu
- Department of Hepatopancreatobiliary Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, China
| | - Hui Li
- Department of Hepatopancreatobiliary Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, China
| | - Xiuyun Lin
- Department of Hepatopancreatobiliary Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, China
| | - Jiani Xiong
- Department of Hepatopancreatobiliary Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, China
| | - Guangfeng Wu
- Department of Hepatopancreatobiliary Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, China
| | - Lingyan Ding
- Department of Hepatopancreatobiliary Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, China
| | - Bin Lin
- Department of Orthodontics, Fujian Medical University Union Hospital, No. 29 of Xinquan Road, Gulou District, Fuzhou, 350001, China.
| |
Collapse
|
16
|
Li X, Li X, Ren Y, Wang L, Mao Z, Gao S, Ma P, Chen J. HJURP modulates cell proliferation and chemoresistance via the MYC/TOP2A transcriptional axis in gastric cancer. Front Mol Biosci 2025; 12:1566293. [PMID: 40290723 PMCID: PMC12021643 DOI: 10.3389/fmolb.2025.1566293] [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: 01/24/2025] [Accepted: 03/31/2025] [Indexed: 04/30/2025] Open
Abstract
Background The histone chaperone Holliday Junction Recognition Protein (HJURP) has been associated with multiple types of cancers, but its role in GC is not yet fully understood. Considering its functions in centromere stability and DNA repair, investigating HJURP's role in GC may offer novel therapeutic perspectives. Methods HJURP expression was examined in a dataset comprising TCGA-STAD samples and an internal group of GC patients, utilizing RNA sequencing and Western blot techniques. Functional experiments were carried out on the AGS and HGC-27 GC cell lines. The expression levels of HJURP, MYC, and Topoisomerase II alpha (TOP2A) were assessed via quantitative real-time PCR and Western blot. Proliferation rates of the cells were determined through EdU, CCK-8, and colony formation assays. Results Compared to adjacent normal tissues, HJURP expression was notably increased in GC tissues, a finding consistent across both the TCGA-STAD database and our internal patient group. Silencing HJURP markedly reduced GC cell growth and chemoresistance. Mechanistically, HJURP enhanced MYC stability, which in turn promoted TOP2A transcription. Rescue experiments confirmed that overexpression of TOP2A alters proliferation and chemoresistance of GC cells with HJURP knockdown, indicating the dependency of this axis on MYC activity. Conclusion Our study demonstrates that HJURP is critical for promoting GC proliferation and chemoresistance through the regulation of the MYC/TOP2A transcriptional network. Targeting HJURP might offer a novel therapeutic avenue for GC, necessitating further exploration of its clinical potential. This work underscores the value of investigating histone chaperones as potential targets in cancer treatment.
Collapse
Affiliation(s)
- Xu Li
- Department of Gastrointestinal Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
- Clinical Medical Research Center, Affiliated Hospital of Nantong University, Nantong, China
| | - Xiwen Li
- Department of Central Laboratory, Kunshan Hospital of Chinese Medicine, Affiliated Hospital of Yangzhou University, Kunshan, China
| | - Yanlin Ren
- Department of Labor Hygiene and Occupational Disease Prevention and Control, Nantong Center for Disease Control and Prevention, Nantong, China
| | - Ling Wang
- Department of Hematology, Affiliated Hospital 2 of Nantong University, Nantong, China
| | - Zehao Mao
- Clinical Medical Research Center, Affiliated Hospital of Nantong University, Nantong, China
| | - Shikun Gao
- Department of Gastrointestinal Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
- Clinical Medical Research Center, Affiliated Hospital of Nantong University, Nantong, China
| | - Peng Ma
- Department of Gastrointestinal Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Junjie Chen
- Clinical Medical Research Center, Affiliated Hospital of Nantong University, Nantong, China
- Nantong Key Laboratory of Gastrointestinal Oncology, Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
17
|
Marian AJ. Causes and consequences of DNA double-stranded breaks in cardiovascular disease. Mol Cell Biochem 2025; 480:2043-2064. [PMID: 39404936 DOI: 10.1007/s11010-024-05131-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 09/29/2024] [Indexed: 04/02/2025]
Abstract
The genome, whose stability is essential for survival, is incessantly exposed to internal and external stressors, which introduce an estimated 104 to 105 lesions, such as oxidation, in the nuclear genome of each mammalian cell each day. A delicate homeostatic balance between the generation and repair of DNA lesions maintains genomic stability. To initiate transcription, DNA strands unwind to form a transcription bubble and provide a template for the RNA polymerase II (RNAPII) complex to synthesize nascent RNA. The process generates DNA supercoils and introduces torsional stress. To enable RNAPII processing, the supercoils are released by topoisomerases by introducing strand breaks, including double-stranded breaks (DSBs). Thus, DSBs are intrinsic genomic features of gene expression. The breaks are quickly repaired upon processing of the transcription. DNA lesions and damaged proteins involved in transcription could impede the integrity and efficiency of RNAPII processing. The impediment, which is referred to as transcription stress, not only could lead to the generation of aberrant RNA species but also the accumulation of DSBs. The latter is particularly the case when topoisomerase processing and/or the repair mechanisms are compromised. The DSBs activate the DNA damage response (DDR) pathways to repair the damaged DNA and/or impose cell cycle arrest and cell death. In addition, the release of DSBs into the cytosol activates the cytosolic DNA-sensing proteins (CDSPs), which along with the nuclear DDR pathways induce the expression of senescence-associated secretory phenotype (SASP), cell cycle arrest, senescence, cell death, inflammation, and aging. The primary stimulus in hereditary cardiomyopathies is a mutation(s) in genes encoding the protein constituents of cardiac myocytes; however, the phenotype is the consequence of intertwined complex interactions among numerous stressors and the causal mutation(s). Increased internal DNA stressors, such as oxidation, alkylation, and cross-linking, are expected to be common in pathological conditions, including in hereditary cardiomyopathies. In addition, dysregulation of gene expression also imposes transcriptional stress and collectively with other stressors provokes the generation of DSBs. In addition, the depletion of nicotinamide adenine dinucleotide (NAD), which occurs in pathological conditions, impairs the repair mechanism and further facilitates the accumulation of DSBs. Because DSBs activate the DDR pathways, they are expected to contribute to the pathogenesis of cardiomyopathies. Thus, interventions to reduce the generation of DSBs, enhance their repair, and block the deleterious DDR pathways would be expected to impart salubrious effects not only in pathological states, as in hereditary cardiomyopathies but also aging.
Collapse
Affiliation(s)
- A J Marian
- Center for Cardiovascular Genetic Studies, Institute of Molecular Medicine, The University of Texas Health Science Center, 6770 Bertner Street, Suite C900A, Houston, TX, 77030, USA.
| |
Collapse
|
18
|
Safiullina AA, Potievskaya VI, Vitsenya MV, Cheremisina IA. A Multicenter Prospective Observational Study to Examine the Experience of Using Phosphocreatine in Combination Therapy for Heart Failure Caused by Cancer Treatment. Rationale and Design of the Study. KARDIOLOGIIA 2025; 65:21-25. [PMID: 40195775 DOI: 10.18087/cardio.2025.3.n2870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 02/04/2025] [Indexed: 04/09/2025]
Abstract
Enhanced cancer treatment efficacy has resulted in a significant increase in the number of cancer survivors after the cure of malignant tumors. However, cardiovascular morbidity, including chronic heart failure, has become the leading cause of death and decreased life expectancy among cancer survivors. This is due, in particular, to the cardiotoxic effects of anticancer drugs and associated factors. Cardioprotective approaches aim to reduce the incidence and severity of cardiotoxicity through the use of cardioprotective agents (e.g., dexrazoxane), liposomal drug delivery systems (e.g., liposomal doxorubicin), and optimization of drug administration schedules. Reducing the cardiotoxicity of cancer treatments is a clinically important goal. Phosphocreatine-based therapy represents a potentially valuable new strategy in this area. In this regard, the study "Multicenter prospective observational study to investigate the experience of using phosphocreatine in combination therapy for heart failure caused by cancer treatment" was initiated. This publication presents the protocol of the observational non-interventional NEOCARD study.
Collapse
Affiliation(s)
- A A Safiullina
- Chazov National Medical Research Center of Cardiology, Moscow
| | | | - M V Vitsenya
- Chazov National Medical Research Center of Cardiology, Moscow
| | | |
Collapse
|
19
|
Ali MS, Hadda V, Verma S, Chopra A, Mittal S, Madan K, Tiwari P, Suri TM, Mohan A. Unravelling the transcriptomic characteristics of bronchoalveolar lavage in post-covid pulmonary fibrosis. BMC Med Genomics 2025; 18:54. [PMID: 40098116 PMCID: PMC11917041 DOI: 10.1186/s12920-025-02110-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Accepted: 02/20/2025] [Indexed: 03/19/2025] Open
Abstract
BACKGROUND Post-Covid Pulmonary Fibrosis (PCPF) has emerged as a significant global issue associated with a poor quality of life and significant morbidity. Currently, our understanding of the molecular pathways of PCPF is limited. Hence, in this study, we performed whole transcriptome sequencing of the RNA isolated from the bronchoalveolar lavage (BAL) samples of PCPF and compared it with idiopathic pulmonary fibrosis (IPF) and non-ILD (Interstitial Lung Disease) control to understand the gene expression profile and associated pathways. METHODS BAL samples from PCPF (n = 3), IPF (n = 3), and non-ILD Control (n = 3) (individuals with apparent healthy lung without interstitial lung disease) groups were obtained and RNA were isolated for whole transcriptomic sequencing. Differentially Expressed Genes (DEGs) were determined followed by functional enrichment analysis and qPCR validation. RESULTS A panel of differentially expressed genes were identified in bronchoalveolar lavage fluid cells (BALF) of PCPF as compare to control and IPF. Our analysis revealed dysregulated pathways associated with cell cycle regulation, immune responses, and neuroinflammatory processes. Real-time validation further supported these findings. The PPI network and module analysis shed light on potential biomarkers and underscore the complex interplay of molecular mechanisms in PCPF. The comparison of PCPF and IPF identified a significant downregulation of pathways that were more prominent in IPF. CONCLUSION This investigation provides crucial insights into the molecular mechanism of PCPF and also outlines avenues for prospective research and the development of therapeutic approaches.
Collapse
Affiliation(s)
- Mohammad Shadab Ali
- Department of Pulmonary, Critical Care, & Sleep Medicine, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Vijay Hadda
- Department of Pulmonary, Critical Care, & Sleep Medicine, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India.
| | - Sonia Verma
- Division of Neuroscience and Ageing Biology, CSIR-Central Drug Research Institute, Lucknow, UP, 226031, India
| | - Anita Chopra
- Lab Oncology, Dr. BRAIRCH All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Saurabh Mittal
- Department of Pulmonary, Critical Care, & Sleep Medicine, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Karan Madan
- Department of Pulmonary, Critical Care, & Sleep Medicine, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Pawan Tiwari
- Department of Pulmonary, Critical Care, & Sleep Medicine, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Tejas Menon Suri
- Department of Pulmonary, Critical Care, & Sleep Medicine, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Anant Mohan
- Department of Pulmonary, Critical Care, & Sleep Medicine, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| |
Collapse
|
20
|
Khalili S, Mohseninia A, Liu C, Banister CE, Heine P, Khazan M, Morrison SE, Gokare P, Cowley GS, Weir BA, Pocalyko D, Bachman KE, Buckhaults PJ. Comprehensive genomic dependency landscape of a human colon cancer organoid. Commun Biol 2025; 8:436. [PMID: 40082551 PMCID: PMC11906589 DOI: 10.1038/s42003-025-07822-5] [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: 08/14/2024] [Accepted: 02/26/2025] [Indexed: 03/16/2025] Open
Abstract
Identifying genetic dependencies in human colon cancer could help identify effective treatment strategies. Genome-wide CRISPR-Cas9 dropout screens have the potential to reveal genetic dependencies, some of which could be exploited as therapeutic targets using existing drugs. In this study, we comprehensively characterized genetic dependencies present in a colon cancer organoid avatar, and validated tumor-specific selectivity of select pharmacologic agents. We conducted a genome-wide CRISPR dropout screen to elucidate the genetic dependencies that interacted with select driver somatic mutations. We found distinct genetic dependencies that interacted with WNT, MAPK, PI3K, TP53, and mismatch repair pathways and validated targets that could be exploited as treatments for this specific subtype of colon cancer. These findings demonstrate the utility of functional genomic screening in the context of personalized medicine.
Collapse
Affiliation(s)
| | | | | | | | - Paige Heine
- University of South Carolina, Columbia, SC, US
| | | | | | - Prashanth Gokare
- Janssen Research and Development, LLC Spring House, Spring House, PA, US
| | - Glenn S Cowley
- Janssen Research and Development, LLC Cambridge, Cambridge, MA, US
| | - Barbara A Weir
- Janssen Research and Development, LLC Cambridge, Cambridge, MA, US
| | - David Pocalyko
- Janssen Research and Development, LLC Spring House, Spring House, PA, US
| | - Kurtis E Bachman
- Janssen Research and Development, LLC Spring House, Spring House, PA, US
| | | |
Collapse
|
21
|
Lyu J, Xu X, Chen C. A convenient single-cell newly synthesized transcriptome assay reveals FLI1 downregulation during T-cell activation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.08.22.609222. [PMID: 39372732 PMCID: PMC11451745 DOI: 10.1101/2024.08.22.609222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
Sequencing newly synthesized transcriptome alongside regular transcriptome in single cells enables the study of gene expression temporal dynamics during rapid chromatin and gene regulation processes. Existing assays for profiling single-cell newly synthesized transcriptome often require specialized technical expertise to achieve high cellular throughput, limiting their accessibility. Here, we developed NOTE-seq, a method for simultaneous profiling of regular and newly synthesized transcriptomes in single cells with high cellular throughput. NOTE-seq integrates 4-thiouridine labeling of newly synthesized RNA, thiol-alkylation-based chemical conversion, and a streamlined 10X Genomics workflow, making it accessible and convenient for biologists without extensive single-cell expertise. Using NOTE-seq, we investigated the temporal dynamics of gene expression during early-stage T-cell activation, identified transcription factors and regulons in Jurkat and naïve T cells, and uncovered the down-regulation of FLI1 as a master transcription factor upon T-cell stimulation. Notably, topoisomerase inhibition led to the depletion of both topoisomerases and FLI1 in T cells through a proteasome-dependent mechanism driven by topoisomerase cleavage complexes, highlighting potential complications topoisomerase-targeting cancer chemotherapies could pose to the immune system.
Collapse
|
22
|
Xiong Y, Lei J, Wen M, Ma Y, Zhao J, Tian Y, Wan Z, Li X, Zhu J, Wang W, Ji X, Sun Y, Yang J, Zhang J, Xin S, Liu Y, Jia L, Han Y, Jiang T. CENPF (+) cancer cells promote malignant progression of early-stage TP53 mutant lung adenocarcinoma. Oncogenesis 2025; 14:5. [PMID: 40044674 PMCID: PMC11882812 DOI: 10.1038/s41389-025-00546-5] [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: 07/01/2024] [Revised: 12/18/2024] [Accepted: 01/23/2025] [Indexed: 03/09/2025] Open
Abstract
The prevention and precise treatment of early-stage lung adenocarcinoma (LUAD) characterized by small nodules (stage IA) remains a significant challenge for clinicians, which is due largely to the limited understanding of the oncogenic mechanisms spanning from preneoplasia to invasive adenocarcinoma. Our study highlights the pivotal role of cancer cells exhibiting high expression of centromere protein F (CENPF), driven by TP53 mutations, which become increasingly prevalent during the transition from preneoplasia to invasive LUAD. Biologically, cancer cells (CENPF+) exhibited robust proliferative and stem-like capabilities, thereby propelling the malignant progression of early-stage LUAD. Clinically, autoantibodies against CENPF in the serum and elevated cancer cells (CENPF+) in tissue correlated positively with the progression of early-stage LUAD, especially those in stage IA. Our findings suggest that cancer cells (CENPF+) play a central role in orchestrating the malignant evolution of LUAD and hold potential as a novel biomarker for early-stage detection and management of the disease.
Collapse
Affiliation(s)
- Yanlu Xiong
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
- Innovation Center for Advanced Medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
- Department of Thoracic Surgery, First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Jie Lei
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Miaomiao Wen
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Yongfu Ma
- Department of Thoracic Surgery, First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Jinbo Zhao
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Yahui Tian
- Department of Thoracic Surgery, Air Force Medical Center, PLA, Beijing, China
| | - Zitong Wan
- College of Life Sciences, Northwestern University, Xi'an, China
| | - Xiaoyan Li
- Department of Blood Transfusion, Shanxi Provincial People's Hospital, Taiyuan, China
| | - Jianfei Zhu
- Department of Thoracic Surgery, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Wenchen Wang
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Xiaohong Ji
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Ying Sun
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Jie Yang
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Jiao Zhang
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Shaowei Xin
- Department of Thoracic Surgery, Air Force Medical Center, PLA, Beijing, China
| | - Yang Liu
- Department of Thoracic Surgery, First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Lintao Jia
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, China
| | - Yong Han
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China.
- Department of Thoracic Surgery, Air Force Medical Center, PLA, Beijing, China.
| | - Tao Jiang
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
23
|
Li S, Li Z, Kuo HCD, Kong AN. Ursolic Acid as a Protective Agent against UVB-Induced Metabolic and Epigenetic Alterations in Human Skin Keratinocytes: An Omics-Based Study. Cancer Prev Res (Phila) 2025; 18:135-144. [PMID: 39718470 PMCID: PMC11875927 DOI: 10.1158/1940-6207.capr-24-0441] [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: 09/26/2024] [Revised: 11/26/2024] [Accepted: 12/20/2024] [Indexed: 12/25/2024]
Abstract
This study aimed to assess how ursolic acid (UA) can protect human skin keratinocytes from damage caused by UVB radiation. Utilizing an omics-based approach, we characterized the features of photodamage and investigated the potential of UA to reverse HaCaT cell subpopulation injury caused by UVB radiation. The most significant changes in metabolite levels after UA treatment were in pathways associated with phosphatidylcholine biosynthesis and arginine and proline metabolism. Treatment with UA can reverse the levels of certain metabolites, including creatinine, creatine phosphate, and succinic acid. Pathways activated by UA treatment in UVB-irradiated HaCaT cells were associated with several biological processes, including the positive regulation of protein modification process, cell division, and enzyme-linked receptor protein signaling pathway. Treatment with UA demonstrates the capability to mitigate the effects of UVB radiation on specific genes, including S100 calcium-binding protein A9 and IL6 receptor. DNA/CpG methylation indicates that UA can partially reverse some of the alterations in the UVB-induced CpG methylome. Utilizing integrated RNA sequencing and methylation sequencing data, starburst plots illustrate the correlation between mRNA expression and CpG methylation status. UA potentially influences the metabolic pathway of glycerophospholipid metabolism by modulating the expression of several key enzymes, including phospholipase A2 group IIA and lipin 2. Altogether, these results indicate that UVB radiation induces metabolic reprogramming, epigenetic changes, and transcriptomic shifts. Meanwhile, UA demonstrates the capacity to inhibit or reduce the severity of these alterations, which may underlie its potential protective role against skin damage caused by UVB exposure. Prevention Relevance: Our research indicates that UA has the potential to mitigate or lessen the impact of UVB radiation, which is known to cause metabolic reprogramming, epigenetic alterations, and transcriptomic changes. These effects could be responsible for UA's possible protective function against skin damage induced by UVB exposure.
Collapse
Affiliation(s)
- Shanyi Li
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, PR China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, 225001, PR China
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Zixin Li
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, PR China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, 225001, PR China
| | - Hsiao-Chen Dina Kuo
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Ah-Ng Kong
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| |
Collapse
|
24
|
Zhang J, Yang T, Wang K, Pan J, Qiu J, Zheng S, Li X, Li G. Multi-omics analysis reveals the panoramic picture of TOP2A in pan-cancer. Sci Rep 2025; 15:6046. [PMID: 39972040 PMCID: PMC11840046 DOI: 10.1038/s41598-025-85929-9] [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: 10/07/2024] [Accepted: 01/07/2025] [Indexed: 02/21/2025] Open
Abstract
Topoisomerases are critical nuclear enzymes that resolve DNA topological challenges during genetic processes. However, there is currently a lack of comprehensive multi-omics analysis of TOP2A from a pan-cancer perspective, despite its significance. A multiomics analysis was conducted to investigate TOP2A across various cancer types. This study involved the integration of over 10,000 multidimensional genomic datasets from 33 distinct cancer types, obtained from The Cancer Genome Atlas (TCGA). The analysis focused on evaluating the overall activity levels of TOP2A in pan-cancers, which encompassed differential expression, clinical significance, immune cell infiltration, and the regulation of pathways related to cancer. Aberrant epigenetic modifications and genomic alterations have been identified as being associated with the dysregulation of TOP2A expression levels. These molecular changes have substantial impacts on cancer progression, intratumoral heterogeneity, immunological status, and the regulation of pathways related to cancer biomarkers. Consequently, patient prognosis varies significantly based on the presence and specific nature of these alterations. The potential of TOP2A to serve as a novel biomarker for prognosis may offer valuable insights into the diagnosis and treatment of cancer.
Collapse
Affiliation(s)
- Jin Zhang
- Department of Orthopedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Tianxiao Yang
- Department of Orthopedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Kenie Wang
- The First Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Jie Pan
- Department of Spine, Shanghai East Hospital, Shanghai, China
| | - Junjun Qiu
- Department of Orthopedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Shengnai Zheng
- Department of Orthopedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China.
| | - Xuesong Li
- Department of Pediatric Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China.
| | - Guanghao Li
- Department of Urology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| |
Collapse
|
25
|
Li S, Ye J, Yang K, Xu C, Qin Z, Xue Y, Yu L, Zhou T, Yin Z, Sun B, Xu J. Targeting the AURKB- MAD2L2 Axis Disrupts the DNA Damage Response and Glycolysis to Inhibit Colorectal Cancer Progression. FRONT BIOSCI-LANDMRK 2025; 30:26532. [PMID: 40018943 DOI: 10.31083/fbl26532] [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: 09/30/2024] [Revised: 11/14/2024] [Accepted: 12/31/2024] [Indexed: 03/01/2025]
Abstract
BACKGROUND Dysregulated metabolic pathways, including glycolysis and a compromised DNA damage response (DDR), are linked to the progression of colorectal cancer (CRC). The mitotic arrest deficient-like 2 (MAD2L2) and aurora kinase B (AURKB) genes play roles in cell cycle regulation and the DDR, making them potential targets for CRC therapy. METHODS Differential expression analysis was performed using The Cancer Genome Atlas-Colon Adenocarcinoma (TCGA-COAD) and GSE47074 datasets. A predictive model was established, and gene expression levels were further analyzed. The Gene Expression Profiling Interaction Analysis database and co-immunoprecipitation experiments assessed the correlation between AURKB and MAD2L2. Knockdown experiments in CRC cell lines further investigated the role of AURKB, followed by analyses of cell behavior, oxidative stress, glycolysis, DDR, and interaction with MAD2L2. RESULTS The risk model identified six prognostic genes (BUB1 mitotic checkpoint serine/threonine kinase B (BUB1B), AURKB, aurora kinase A (AURKA), exonuclease 1 (EXO1), topoisomerase II alpha (TOP2A), cyclin A2 (CCNA2)) associated with CRC, which were significantly expressed in tumor samples from the TCGA-COAD and GSE47074 datasets. In vitro assays confirmed that AURKB knockdown inhibited CRC cell behavior, induced G1 cell cycle arrest, and increased oxidative stress and apoptosis. AURKB knockdown also impaired glycolysis, reducing lactate production, glucose uptake, and ATP levels. Overexpression of MAD2L2 partially reversed these effects, restored glycolytic activity, and mitigated the cell cycle arrest and DDR caused by AURKB knockdown. CONCLUSION AURKB regulates CRC progression by modulating glycolysis and DDR pathways. Targeting the AURKB-MAD2L2 axis offers a promising therapeutic strategy for disrupting fundamental metabolic and DNA repair mechanisms in CRC.
Collapse
Affiliation(s)
- Shengjie Li
- Department of Gastroenterology Surgery, Yichang Central People's Hospital, The First College of Clinical Medical Science, China Three Gorges University, 443000 Yichang, Hubei, China
| | - Jiayou Ye
- Department of Gastroenterology Surgery, Yichang Central People's Hospital, The First College of Clinical Medical Science, China Three Gorges University, 443000 Yichang, Hubei, China
| | - Kaifeng Yang
- Department of Gastroenterology Surgery, Yichang Central People's Hospital, The First College of Clinical Medical Science, China Three Gorges University, 443000 Yichang, Hubei, China
| | - Chengfan Xu
- Department of Gastroenterology Surgery, Yichang Central People's Hospital, The First College of Clinical Medical Science, China Three Gorges University, 443000 Yichang, Hubei, China
| | - Zhixiang Qin
- Department of Gastroenterology Surgery, Yichang Central People's Hospital, The First College of Clinical Medical Science, China Three Gorges University, 443000 Yichang, Hubei, China
| | - Yiyang Xue
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, CA 93106, USA
| | - Lanjian Yu
- Department of Gastroenterology Surgery, Yichang Central People's Hospital, The First College of Clinical Medical Science, China Three Gorges University, 443000 Yichang, Hubei, China
| | - Tianyu Zhou
- Department of Gastroenterology Surgery, Yichang Central People's Hospital, The First College of Clinical Medical Science, China Three Gorges University, 443000 Yichang, Hubei, China
| | - Ziming Yin
- Department of Chemistry, University of Warwick Coventry, CV4 7AL Coventry, UK
| | - Bin Sun
- Center for Clinical Research and Translational Medicine, Yangpu Hospital, School of Medicine, Tongji University, 200438 Shanghai, China
| | - Jun Xu
- Department of Gastroenterology Surgery, Yichang Central People's Hospital, The First College of Clinical Medical Science, China Three Gorges University, 443000 Yichang, Hubei, China
| |
Collapse
|
26
|
Zhou J, Zhang Z, Huang R, Zhuang X, Ni S. Mechanisms of Picrasma quassioides against hepatocellular carcinoma elucidated by network pharmacology and experimental validation. Am J Transl Res 2025; 17:1402-1415. [PMID: 40092104 PMCID: PMC11909513 DOI: 10.62347/vlgd3371] [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: 09/26/2024] [Accepted: 01/18/2025] [Indexed: 03/19/2025]
Abstract
OBJECTIVE The medicinal plant Picrasma quassioides (P. quassioides) Benn exerts an inhibitory effect on the growth of hepatocellular carcinoma (HCC) cells via an unknown mechanism. This study explored the targets and signaling pathways underlying the mechanism of P. quassioides against HCC. METHODS Targets of P. quassioides active compounds were identified using the HERB database, and the HCC targets were found with the GeneCards database. The optimal serum concentration and intervention time were determined using the CCK-8 assay. Apoptosis, cell cycle, invasion, cloning, and wound-healing abilities were assessed using flow cytometry. Core protein targets and signaling pathway-related metabolic enzymes were evaluated with Western blotting. The anti-HCC effect of P. quassioides medicated serum was verified using arachidonic acid (AA)-related enzyme agonists. RESULTS Network pharmacology identified 19 effective compounds of P. quassioides and 105 HCC-associated targets. It also revealed the AA pathway was the central pathway of P. quassioides against HCC, with AURKA, AURKB, KIF11, and TOP2A identified as core targets that inhibit excessive HCC cell proliferation and promote apoptosis. Flow cytometry findings supported that P. quassioides medicated serum significantly inhibited HCC cell proliferation and promoted apoptosis. By contrast, enzyme agonists related to the AA pathway markedly counteracted the anti-HCC effect of P. quassioides, promoting HCC growth. CONCLUSION P. quassioides medicated serum exerts a prominent anti-HCC effect in vitro. The AA pathway constitutes the mechanism by which P. quassioides medicated serum inhibits excessive proliferation and promotes apoptosis of HCC cells.
Collapse
Affiliation(s)
- Jie Zhou
- School of Pharmacy, Anhui Medical University Hefei 230012, Anhui, China
| | - Zhilan Zhang
- School of Pharmacy, Anhui Medical University Hefei 230012, Anhui, China
| | - Ruiru Huang
- School of Pharmacy, Anhui Medical University Hefei 230012, Anhui, China
| | - Xingxing Zhuang
- Department of Pharmacy, Chaohu Hospital of Anhui Medical University Chaohu 238000, Anhui, China
| | - Shoudong Ni
- School of Pharmacy, Anhui Medical University Hefei 230012, Anhui, China
- Department of Pharmacy, Chaohu Hospital of Anhui Medical University Chaohu 238000, Anhui, China
| |
Collapse
|
27
|
Zhu M, Ma M, Luo L, Li F, Zheng J, Pan Y, Yang L, Xiao Y, Wang Z, Xian B, Zheng Y, Li H, Yang J. Reduction of DNA Topoisomerase Top2 Reprograms the Epigenetic Landscape and Extends Health and Life Span Across Species. Aging Cell 2025:e70010. [PMID: 39935344 DOI: 10.1111/acel.70010] [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: 11/15/2024] [Revised: 12/25/2024] [Accepted: 01/22/2025] [Indexed: 02/13/2025] Open
Abstract
DNA topoisomerases are essential molecular machines that manage DNA topology in the cell and play important roles in DNA replication and transcription. We found that knocking down the enzyme topoisomerase Top2 or its mammalian homolog TOP2B increases the lifespan of S. cerevisiae, C. elegans, and mice. TOP2B reduction also extends the health span of mice and alleviates the pathologies of aging in multiple tissues. At the cellular/molecular level, TOP2B reduction alleviates the major hallmarks of aging, including senescence, DNA damage, and deregulated nutrient sensing. We observed that TOP2B reduction changes the epigenetic landscape of various tissues in old mice toward that of the young animals, and differentially downregulates genes with active promoter and high expression. Our observations suggest that Top2 reduction confers pro-longevity effect across species possibly through a conserved mechanism and may be a promising strategy for longevity intervention.
Collapse
Affiliation(s)
- Man Zhu
- Department of Health Management & Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Laboratory of Aging Research, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Meng Ma
- Laboratory of Aging Research, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Lunan Luo
- Laboratory of Aging Research, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Feiyang Li
- Laboratory of Aging Research, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Jiashun Zheng
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, California, USA
| | - Yan Pan
- Laboratory of Aging Research, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Lu Yang
- Laboratory of Aging Research, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Ying Xiao
- Laboratory of Aging Research, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Ziyan Wang
- Laboratory of Aging Research, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Bo Xian
- Laboratory of Aging Research, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yi Zheng
- Department of Health Management & Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Laboratory of Aging Research, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Hao Li
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, California, USA
| | - Jing Yang
- Department of Health Management & Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Laboratory of Aging Research, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
28
|
Li H, Jia Y, Chen Z, Yang L, Ni L, Cao Y, Fan R, Yuan Z, Zhu K, Zhang Z, Zuo L, Wu P, Gao Y, Lin Y. Bioinformatics analysis of coronary microvascular dysfunction in rats based on single-cell RNA sequencing. Sci Rep 2025; 15:5050. [PMID: 39934189 PMCID: PMC11814319 DOI: 10.1038/s41598-025-85318-2] [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: 11/04/2024] [Accepted: 01/01/2025] [Indexed: 02/13/2025] Open
Abstract
Coronary microvascular dysfunction serves as one of the etiological factors for ischemic heart disease and represents a novel therapeutic direction for coronary artery diseases; however, the research on its pathogenesis remains inconsistent. This study aims to explore the single-cell gene expression profiles in rats with coronary microvascular dysfunction using single-cell RNA sequencing, with a particular focus on the in-depth analysis of endothelial cell gene expression characteristics. By establishing a rat model of coronary microvascular dysfunction, we collected cardiac apical tissue to prepare single-cell suspensions and further analyzed them using bioinformatics methods. From a total of 55,419 cells, we identified 28 distinct cell clusters, with endothelial cells and fibroblasts being the predominant cell types. Compared to the NC group, the proportion of endothelial cells in the CMD group was significantly reduced, while the number of fibroblasts was significantly increased. Through further analysis of the endothelial cells, we classified them into normal phenotype endothelial cells, mesenchymal phenotype endothelial cells, proliferative phenotype endothelial cells, and lymphatic endothelial cells, with mesenchymal and proliferative endothelial cells originating from normal phenotype endothelial cells. Additionally, the CMD group exhibited an increase in immune cells, enhanced inflammatory response, and increased oxidative stress. These findings may provide novel potential therapeutic targets for the treatment of Coronary microvascular dysfunction.
Collapse
Affiliation(s)
- Hao Li
- LinFen Central Hospital, Department of Cardiology, LinFen, 041000, China
| | - Yiding Jia
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Department of Cardiology, 99 Longcheng Street, Xiaodian District, Taiyuan, 030032, Shanxi, China
| | - Zelin Chen
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Department of Cardiology, 99 Longcheng Street, Xiaodian District, Taiyuan, 030032, Shanxi, China
| | - Luqun Yang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Department of Cardiology, 99 Longcheng Street, Xiaodian District, Taiyuan, 030032, Shanxi, China
| | - Lin Ni
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Department of Cardiology, 99 Longcheng Street, Xiaodian District, Taiyuan, 030032, Shanxi, China
| | - Yuchen Cao
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Department of Cardiology, 99 Longcheng Street, Xiaodian District, Taiyuan, 030032, Shanxi, China
| | - Rong Fan
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Department of Cardiology, 99 Longcheng Street, Xiaodian District, Taiyuan, 030032, Shanxi, China
| | - Zitong Yuan
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Department of Cardiology, 99 Longcheng Street, Xiaodian District, Taiyuan, 030032, Shanxi, China
| | - Kaiyi Zhu
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Department of Cardiology, 99 Longcheng Street, Xiaodian District, Taiyuan, 030032, Shanxi, China
| | - Zhijun Zhang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Department of Cardiology, 99 Longcheng Street, Xiaodian District, Taiyuan, 030032, Shanxi, China
| | - Lin Zuo
- Shanxi Medical UniversityLaboratory of Cell Physiology, Jinzhong, 030600, China
| | - Ping Wu
- First Hospital of Shanxi Medical University, Department of Cardiology, Taiyuan, 030012, China
| | - Yuping Gao
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Department of Cardiology, 99 Longcheng Street, Xiaodian District, Taiyuan, 030032, Shanxi, China.
| | - Yuanyuan Lin
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Department of Cardiology, 99 Longcheng Street, Xiaodian District, Taiyuan, 030032, Shanxi, China.
| |
Collapse
|
29
|
de Klein B, Eickhoff N, Zwart W. The emerging regulatory interface between DNA repair and steroid hormone receptors in cancer. Trends Mol Med 2025:S1471-4914(25)00006-1. [PMID: 39934021 DOI: 10.1016/j.molmed.2025.01.006] [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: 11/15/2024] [Revised: 01/10/2025] [Accepted: 01/15/2025] [Indexed: 02/13/2025]
Abstract
Human cells potentiate highly diverse functions through tight transcriptional regulation and maintenance of genome integrity. While the DNA damage response (DDR) safeguards the genome, ligand-activated transcription factors, such as steroid hormone receptors (SHRs), provide complex transcriptional outputs. Interestingly, an increasing body of evidence reveals a direct biological and functional interplay between DDR factors and SHR cascades in cancer. SHRs can directly affect DDR gene expression, but DDR factors in turn act as transcriptional coregulators, enabling oncogenic SHR-mediated signaling, which has the potential for novel therapeutic interventions. With a focus on breast and prostate cancer, we describe in this review recent developments in, and insights into, the complex interplay between SHR signaling and the DDR, highlighting opportunities for future clinical interventions.
Collapse
Affiliation(s)
- Bim de Klein
- Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Nils Eickhoff
- Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Wilbert Zwart
- Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands; Department of Biomedical Engineering, Eindhoven University of Technology, 5600, MB, Eindhoven, The Netherlands.
| |
Collapse
|
30
|
Zhou T, Niu Y, Li Y. Advances in research on malignant tumors and targeted agents for TOP2A (Review). Mol Med Rep 2025; 31:50. [PMID: 39670307 DOI: 10.3892/mmr.2024.13415] [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: 07/18/2024] [Accepted: 11/28/2024] [Indexed: 12/14/2024] Open
Abstract
The DNA topoisomerase isoform topoisomerase IIα (TOP2A) is essential for the condensation and segregation of cellular mitotic chromosomes and the structural maintenance. It has been demonstrated that TOP2A is highly expressed in various malignancies, including lung adenocarcinoma (LUAD), hepatocellular carcinoma (HCC) and breast cancer (BC), associating with poor prognosis and aggressive tumor behavior. Additionally, TOP2A has emerged as a promising target for cancer therapy, with widespread clinical application of associated chemotherapeutic agents. The present study explored the impact of TOP2A on malignant tumor growth and the advancements in research on its targeted drugs. The fundamental mechanisms of TOP2A have been detailed, its specific roles in tumor cells are analyzed, and its potential as a biomarker for tumor prognosis and therapeutic targeting is highlighted. Additionally, the present review compiles findings from the latest clinical trials of relevant targeted agents, information on newly developed inhibitors, and discusses future research directions and clinical application strategies in cancer therapy, aiming to propose novel ideas and methods.
Collapse
Affiliation(s)
- Tao Zhou
- Department of Hepatobiliary Surgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi 030032, P.R. China
| | - Yiting Niu
- Department of Hepatobiliary Surgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi 030032, P.R. China
| | - Yanjun Li
- Department of Hepatobiliary Surgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi 030032, P.R. China
| |
Collapse
|
31
|
Huang Y, Xi X, Ye Z, Zhang C, Jiang Y, Yu F, Huang G. MYBL2 promotes proliferation of clear cell renal cell carcinoma by regulating TOP2A and activating AKT/mTOR signaling pathway. FASEB J 2025; 39:e70330. [PMID: 39831843 DOI: 10.1096/fj.202401910r] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 12/28/2024] [Accepted: 01/08/2025] [Indexed: 01/22/2025]
Abstract
Renal cell carcinoma (RCC) is one of the most common malignancies in the urinary system, and clear cell renal cell carcinoma (ccRCC) is the most common subtype. MYBL2 has been reported to be overexpressed in various tumors and associated with poor prognosis in patients, but its biological role in ccRCC remains unclear. In this study, we investigated the mRNA and protein expression levels of MYBL2 in ccRCC samples and evaluated the prognostic value of MYBL2 using TCGA dataset. In vitro functional assays were performed using CCK-8, EdU, colony formation, cell scratch, and transwell assays, as well as in vivo tumorigenesis assays to investigate the biological functions of MYBL2 in ccRCC. Additionally, gene set enrichment analysis (GSEA) was used to explore the downstream pathways of MYBL2, which were further validated. Finally, we predicted the target genes of MYBL2 using bioinformatics and validated them using ChIP and dual-luciferase reporter gene assays. MYBL2 expression was significantly higher in ccRCC than in adjacent normal tissues and was associated with poor prognosis. MYBL2 expression was positively correlated with the pathological tumor grade and clinical TNM stage of ccRCC patients. Knockdown of MYBL2 significantly inhibited the proliferation of renal cancer cells in vitro and in vivo, and knockdown of MYBL2 could inhibit cell invasion and migration, while overexpression of MYBL2 had the opposite effect. GSEA revealed that MYBL2 was associated with the mTOR signaling pathway and cell cycle pathway, which was confirmed by our study. Finally, we found that TOP2A was a target gene of MYBL2, and MYBL2 could bind to the TOP2A promoter to regulate its transcriptional activity, promoting the proliferation of clear cell renal cell carcinoma cells. MYBL2 emerges as a highly expressed factor that significantly correlates with adverse patient prognosis in ccRCC. Mechanistically, MYBL2 transcriptionally upregulates TOP2A, thereby modulating the proliferation of ccRCC cells. Furthermore, MYBL2 activates the mTOR signaling pathway, a critical node in the progression of ccRCC. Collectively, these findings position MYBL2 as a promising candidate for both a biological marker and a therapeutic target in the management of ccRCC.
Collapse
Affiliation(s)
- Yawei Huang
- Department of Urology, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xiaoqing Xi
- Department of Urology, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zhenfeng Ye
- Department of Urology, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Chiyu Zhang
- Department of Urology, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yi Jiang
- Department of Urology, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Fanfan Yu
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Gaomin Huang
- Department of Urology, Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
32
|
Li Z, Zhang W, Zhang Z, Mao G, Qi L, Wang Y, Yang H, Ye H. PICH, A protein that maintains genomic stability, can promote tumor growth. Gene 2025; 935:149074. [PMID: 39491600 DOI: 10.1016/j.gene.2024.149074] [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: 07/26/2024] [Revised: 10/22/2024] [Accepted: 10/30/2024] [Indexed: 11/05/2024]
Abstract
Genomic instability is regardedas a hallmark of cancer cells. It can be presented in many ways, among which chromosome instability has received attention. Ultrafine anaphase bridges are a typeof chromatin bridges, the untimely resolution of which can also lead to chromosome instability. PICH can play a role in maintaining chromosome stability by regulating chromosome morphologyand resolving ultrafine anaphase bridges. Recently, PICH has been found to be overexpressed in various cancers. Overexpression of PICH is related to the proliferation of tumors and poor prognosis. In this article, we consider that PICH can maintain genomic stability by regulating appropriate chromosome structure, ensuring proper chromosome segregation, and facilitating replication fork reversal. We summarize how PICH regulates chromosome stability, how PICH resolves Ultrafine anaphase bridges with other proteins, and how PICH promotes tumor progression.
Collapse
Affiliation(s)
- Zeyuan Li
- The Second Clinical Medical School, Lanzhou University, Lanzhou, People's Republic of China
| | - Wentao Zhang
- The Second Clinical Medical School, Lanzhou University, Lanzhou, People's Republic of China
| | - Zihan Zhang
- The Second Clinical Medical School, Lanzhou University, Lanzhou, People's Republic of China
| | - Guoming Mao
- The Second Clinical Medical School, Lanzhou University, Lanzhou, People's Republic of China
| | - Linping Qi
- The Second Clinical Medical School, Lanzhou University, Lanzhou, People's Republic of China
| | - Yubin Wang
- Laboratory Medicine Center Gansu Provincial Natural Science, Lanzhou University Second Hospital, People's Republic of China
| | - Hanteng Yang
- The Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, People's Republic of China.
| | - Huili Ye
- The Second Clinical Medical School, Lanzhou University, Lanzhou, People's Republic of China; Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, People's Republic of China; Biobank of Tumors from Plateau of Gansu Province, Lanzhou University Second Hospital, Lanzhou, People's Republic of China.
| |
Collapse
|
33
|
Liu T, Wang Y, An XZ, Liu J, Wu Y, Xiang Y, Zhang YJ, Huang L, Li JC, Li YZP, Yu J. Identification of a novel TOP2B::AFF2 fusion gene in B-cell acute lymphoblastic leukemia. Sci Rep 2025; 15:3280. [PMID: 39863655 PMCID: PMC11763246 DOI: 10.1038/s41598-025-86865-4] [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: 08/01/2024] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
Genetic alterations play a pivotal role in leukemic clonal transformation, significantly influencing disease pathogenesis and clinical outcomes. Here, we report a novel fusion gene and investigate its pathogenic role in acute lymphoblastic leukemia (ALL). We engineer a transposon transfection system expressing the TOP2B::AFF2 transcript and introduce it into Ba/F3 cells. Functional studies, including proliferation, cell cycle, and apoptosis assays, were conducted to assess the fusion gene's impact. In vitro assays reveal that the TOP2B::AFF2 fusion significantly enhances Ba/F3 cell proliferation and G1/S phase transition while suppressing differentiation and apoptosis. This study identifies TOP2B::AFF2 as a potential oncogenic driver. However, further validation through in vivo studies are warranted to fully elucidate the fusion gene's role in leukemogenesis.
Collapse
Affiliation(s)
- Tao Liu
- Department of Hematology and Oncology, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Children's Hospital of Chongqing Medical University, No 136 Zhongshan 2 road, YuZhong district, Chongqing, 400014, China
| | - Yang Wang
- Department of Hematology and Oncology, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Children's Hospital of Chongqing Medical University, No 136 Zhongshan 2 road, YuZhong district, Chongqing, 400014, China
- Department of Hematology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Xi-Zhou An
- Department of Hematology and Oncology, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Children's Hospital of Chongqing Medical University, No 136 Zhongshan 2 road, YuZhong district, Chongqing, 400014, China
| | - Jiaqi Liu
- Shanghai Cinopath Medical Testing Co Ltd, Shanghai, China
| | - Yuqin Wu
- Laboratory Medicine Department, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Yan Xiang
- Department of Hematology and Oncology, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Children's Hospital of Chongqing Medical University, No 136 Zhongshan 2 road, YuZhong district, Chongqing, 400014, China
| | - Yong-Jie Zhang
- Department of Hematology and Oncology, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Children's Hospital of Chongqing Medical University, No 136 Zhongshan 2 road, YuZhong district, Chongqing, 400014, China
| | - Lan Huang
- Department of Hematology and Oncology, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Children's Hospital of Chongqing Medical University, No 136 Zhongshan 2 road, YuZhong district, Chongqing, 400014, China
| | - Jia-Cheng Li
- Department of Hematology and Oncology, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Children's Hospital of Chongqing Medical University, No 136 Zhongshan 2 road, YuZhong district, Chongqing, 400014, China
| | - Yu-Zhuo-Pu Li
- Department of Hematology and Oncology, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Children's Hospital of Chongqing Medical University, No 136 Zhongshan 2 road, YuZhong district, Chongqing, 400014, China
| | - Jie Yu
- Department of Hematology and Oncology, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Children's Hospital of Chongqing Medical University, No 136 Zhongshan 2 road, YuZhong district, Chongqing, 400014, China.
| |
Collapse
|
34
|
Yao Q, Zhu L, Shi Z, Banerjee S, Chen C. Topoisomerase-modulated genome-wide DNA supercoiling domains colocalize with nuclear compartments and regulate human gene expression. Nat Struct Mol Biol 2025; 32:48-61. [PMID: 39152238 DOI: 10.1038/s41594-024-01377-5] [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: 10/10/2023] [Accepted: 07/24/2024] [Indexed: 08/19/2024]
Abstract
DNA supercoiling is a biophysical feature of the double helix with a pivotal role in biological processes. However, understanding of DNA supercoiling in the chromatin remains limited. Here, we developed azide-trimethylpsoralen sequencing (ATMP-seq), a DNA supercoiling assay offering quantitative accuracy while minimizing genomic bias and background noise. Using ATMP-seq, we directly visualized transcription-dependent negative and positive twin-supercoiled domains around genes and mapped kilobase-resolution DNA supercoiling throughout the human genome. Remarkably, we discovered megabase-scale supercoiling domains (SDs) across all chromosomes that are modulated mainly by topoisomerases I and IIβ. Transcription activities, but not the consequent supercoiling accumulation in the local region, contribute to SD formation, indicating the long-range propagation of transcription-generated supercoiling. Genome-wide SDs colocalize with A/B compartments in both human and Drosophila cells but are distinct from topologically associating domains (TADs), with negative supercoiling accumulation at TAD boundaries. Furthermore, genome-wide DNA supercoiling varies between cell states and types and regulates human gene expression, underscoring the importance of supercoiling dynamics in chromatin regulation and function.
Collapse
Affiliation(s)
- Qian Yao
- Laboratory of Biochemistry and Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Linying Zhu
- Laboratory of Biochemistry and Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Zhen Shi
- Laboratory of Biochemistry and Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Subhadra Banerjee
- Laboratory of Genome Integrity, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Chongyi Chen
- Laboratory of Biochemistry and Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
35
|
Zhang K, Zheng X, Sun Y, Feng X, Wu X, Liu W, Gao C, Yan Y, Tian W, Wang Y. TOP2A modulates signaling via the AKT/mTOR pathway to promote ovarian cancer cell proliferation. Cancer Biol Ther 2024; 25:2325126. [PMID: 38445610 PMCID: PMC10936659 DOI: 10.1080/15384047.2024.2325126] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 02/26/2024] [Indexed: 03/07/2024] Open
Abstract
Ovarian cancer (OC) is a form of gynecological malignancy that is associated with worse patient outcomes than any other cancer of the female reproductive tract. Topoisomerase II α (TOP2A) is commonly regarded as an oncogene that is associated with malignant disease progression in a variety of cancers, its mechanistic functions in OC have yet to be firmly established. We explored the role of TOP2A in OC through online databases, clinical samples, in vitro and in vivo experiments. And initial analyses of public databases revealed high OC-related TOP2A expression in patient samples that was related to poorer prognosis. This was confirmed by clinical samples in which TOP2A expression was elevated in OC relative to healthy tissue. Kaplan-Meier analyses further suggested that higher TOP2A expression levels were correlated with worse prognosis in OC patients. In vitro, TOP2A knockdown resulted in the inhibition of OC cell proliferation, with cells entering G1 phase arrest and undergoing consequent apoptotic death. In rescue assays, TOP2A was confirmed to regulate cell proliferation and cell cycle through AKT/mTOR pathway activity. Mouse model experiments further affirmed the key role that TOP2A plays as a driver of OC cell proliferation. These data provide strong evidence supporting TOP2A as an oncogenic mediator and prognostic biomarker related to OC progression and poor outcomes. At the mechanistic level, TOP2A can control tumor cell growth via AKT/mTOR pathway modulation. These preliminary results provide a foundation for future research seeking to explore the utility of TOP2A inhibitor-based combination treatment regimens in platinum-resistant recurrent OC patients.
Collapse
Affiliation(s)
- Kaiwen Zhang
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Xingyu Zheng
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Yiqing Sun
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Xinyu Feng
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Xirong Wu
- Department of Gynecology and Obstetrics, Affiliated Hospital of Nantong University, Nantong, China
| | - Wenlu Liu
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Chao Gao
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Ye Yan
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Wenyan Tian
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Yingmei Wang
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
36
|
Borella F, Fucina S, Seminara Y, Denti P, Ferraioli D, Bertero L, Gallio N, Cusato J, Valabrega G, Revelli A, Marozio L, Cosma S. Targeting TOP2A in Ovarian Cancer: Biological and Clinical Implications. Curr Oncol 2024; 31:8054-8074. [PMID: 39727717 DOI: 10.3390/curroncol31120594] [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: 10/31/2024] [Revised: 12/09/2024] [Accepted: 12/19/2024] [Indexed: 12/28/2024] Open
Abstract
The enzyme topoisomerase II alpha (TOP2A) plays a critical role in DNA replication and cell proliferation, making it a promising target for cancer therapy. In epithelial ovarian cancer (EOC), TOP2A overexpression is associated with poor prognosis and resistance to conventional treatments. This review explores the biological functions of TOP2A in EOC and discusses its potential as a therapeutic target. We highlight studies on the mechanisms through which TOP2A contributes to tumor progression and recurrence. Additionally, we evaluate the clinical implications of targeting TOP2A, including the use of TOP2A inhibitors and their combination with novel drugs. We provide a comprehensive overview of the current understanding and future directions for targeting TOP2A in the management of EOC.
Collapse
Affiliation(s)
- Fulvio Borella
- Gynecology and Obstetrics 1U, Department of Surgical Sciences, University of Turin, 10126 Turin, Italy
| | - Stefano Fucina
- Gynecology and Obstetrics 1U, Department of Surgical Sciences, University of Turin, 10126 Turin, Italy
| | - Ylenia Seminara
- Gynecology and Obstetrics 1U, Department of Surgical Sciences, University of Turin, 10126 Turin, Italy
| | - Pietro Denti
- Gynecology and Obstetrics 1U, Department of Surgical Sciences, University of Turin, 10126 Turin, Italy
| | - Domenico Ferraioli
- Department of Gynecology, Léon Bérard, Comprehensive Cancer Centre, 69008 Lyon, France
| | - Luca Bertero
- Pathology Unit, Department of Medical Sciences, University of Turin, 10126 Turin, Italy
| | - Niccolò Gallio
- Gynecology and Obstetrics 2U, Department of Surgical Sciences, University of Turin, 10126 Turin, Italy
| | - Jessica Cusato
- Laboratory of Clinical Pharmacology and Pharmacogenetics, Department of Medical Sciences, University of Turin, 10149 Turin, Italy
| | - Giorgio Valabrega
- Department of Oncology, University of Turin, Medical Oncology, Ordine Mauriziano Hospital, 10128 Turin, Italy
| | - Alberto Revelli
- Gynecology and Obstetrics 2U, Department of Surgical Sciences, University of Turin, 10126 Turin, Italy
| | - Luca Marozio
- Gynecology and Obstetrics 1U, Department of Surgical Sciences, University of Turin, 10126 Turin, Italy
| | - Stefano Cosma
- Gynecology and Obstetrics 1U, Department of Surgical Sciences, University of Turin, 10126 Turin, Italy
| |
Collapse
|
37
|
Alsaleh AN, Aziz IM, Aljowaie RM, Alshalan RM, Alkubaisi NA, Aboul-Soud MAM. In Vitro Evaluation, Chemical Profiling, and In Silico ADMET Prediction of the Pharmacological Activities of Artemisia absinthium Root Extract. Pharmaceuticals (Basel) 2024; 17:1646. [PMID: 39770489 PMCID: PMC11728498 DOI: 10.3390/ph17121646] [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: 10/21/2024] [Revised: 11/21/2024] [Accepted: 12/05/2024] [Indexed: 01/16/2025] Open
Abstract
Artemisia absinthium L., is a plant with established pharmacological properties, but the A. absinthium root extract (AARE) remains unexplored. The aim of this study was to examine the chemical composition of AARE and assess its biological activity, which included antidiabetic, antibacterial, anticancer, and antioxidant properties. GC-MS was used to analyze the chemical components. The antioxidant activity of the total phenolic and flavonoid content was evaluated. Antibacterial activity and cytotoxic effects were identified. Enzyme inhibition experiments were performed to determine its antidiabetic potential. Molecular docking was utilized to evaluate the potential antioxidant, antibacterial, and anticancer activities of the compounds from AARE using Maestro 11.5 from the Schrödinger suite. AARE exhibited moderate antioxidant activity in DPPH (IC50: 172.41 ± 3.15 μg/mL) and ABTS (IC50: 378.94 ± 2.18 μg/mL) assays. Cytotoxicity tests on MCF-7 and HepG2 cancer cells demonstrated significant anticancer effects, with IC50 values of 150.12 ± 0.74 μg/mL and 137.11 ± 1.33 μg/mL, respectively. Apoptotic studies indicated an upregulation of pro-apoptotic genes (caspase-3, 8, 9, Bax) and a downregulation of anti-apoptotic markers (Bcl-2 and Bcl-Xl). AARE also inhibited α-amylase and α-glucosidase, suggesting potential antidiabetic effects, with IC50 values of 224.12 ± 1.17 μg/mL and 243.35 ± 1.51 μg/mL. Antibacterial assays revealed strong activity against Gram-positive bacteria. Molecular docking and pharmacokinetic analysis identified promising inhibitory effects of key AARE compounds on NADPH oxidase, E. coli Gyrase B, and Topoisomerase IIα, with favorable drug-like properties. These findings suggest AARE's potential in treating cancer, diabetes, and bacterial infections, warranting further in vivo and clinical studies.
Collapse
Affiliation(s)
- Asma N. Alsaleh
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia; (A.N.A.); (R.M.A.); (R.M.A.); (N.A.A.)
| | - Ibrahim M. Aziz
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia; (A.N.A.); (R.M.A.); (R.M.A.); (N.A.A.)
| | - Reem M. Aljowaie
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia; (A.N.A.); (R.M.A.); (R.M.A.); (N.A.A.)
| | - Rawan M. Alshalan
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia; (A.N.A.); (R.M.A.); (R.M.A.); (N.A.A.)
| | - Noorah A. Alkubaisi
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia; (A.N.A.); (R.M.A.); (R.M.A.); (N.A.A.)
| | - Mourad A. M. Aboul-Soud
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, P.O. Box 10219, Riyadh 11433, Saudi Arabia;
| |
Collapse
|
38
|
Yu J, Shang C, Deng X, Jia J, Shang X, Wang Z, Zheng Y, Zhang R, Wang Y, Zhang H, Liu H, Liu WJ, Li H, Cao B. Time-resolved scRNA-seq reveals transcription dynamics of polarized macrophages with influenza A virus infection and antigen presentation to T cells. Emerg Microbes Infect 2024; 13:2387450. [PMID: 39129565 PMCID: PMC11370681 DOI: 10.1080/22221751.2024.2387450] [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: 03/25/2024] [Revised: 07/02/2024] [Accepted: 07/30/2024] [Indexed: 08/13/2024]
Abstract
Throughout history, the influenza A virus has caused numerous devastating global pandemics. Macrophages, as pivotal innate immune cells, exhibit a wide range of immune functions characterized by distinct polarization states, reflecting their intricate heterogeneity. In this study, we employed the time-resolved single-cell sequencing technique coupled with metabolic RNA labelling to elucidate the dynamic transcriptional changes in distinct polarized states of bone marrow-derived macrophages (BMDMs) upon infection with the influenza A virus. Our approach not only captures the temporal dimension of transcriptional activity, which is lacking in conventional scRNA-seq methods, but also reveals that M2-polarized Arg1_macrophage cluster is the sole state supporting successful replication of influenza A virus. Furthermore, we identified distinct antigen presentation capabilities to CD4+ T and CD8+ T cells across diverse polarized states of macrophages. Notably, the M1 phenotype, exhibited by (BMDMs) and murine alveolar macrophages (AMs), demonstrated superior conventional and cross-presentation abilities for exogenous antigens, with a particular emphasis on cross-presentation capacity. Additionally, as CD8+ T cell differentiation progressed, M1 polarization exhibited an enhanced capacity for cross-presentation. All three phenotypes of BMDMs, including M1, demonstrated robust presentation to CD4+ regulatory T cells, while displaying limited ability to present to naive CD4+ T cells. These findings offer novel insights into the immunological regulatory mechanisms governing distinct polarized states of macrophages, particularly their roles in restricting the replication of influenza A virus and modulating antigen-specific T cell responses through innate immunity.
Collapse
Affiliation(s)
- Jiapei Yu
- Department of Pulmonary and Critical Care Medicine, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Center of Respiratory Medicine, China–Japan Friendship Hospital, Beijing, People’s Republic of China
- Institute of Respiratory Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
| | - Congcong Shang
- Department of Pulmonary and Critical Care Medicine, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Center of Respiratory Medicine, China–Japan Friendship Hospital, Beijing, People’s Republic of China
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, People’s Republic of China
| | - Xiaoyan Deng
- THU-PKU Joint Center for Life Sciences, Tsinghua University, Beijing, People’s Republic of China
| | - Ju Jia
- Department of Infectious Disease, Beijing Friendship Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Xiao Shang
- THU-PKU Joint Center for Life Sciences, Tsinghua University, Beijing, People’s Republic of China
| | - Zeyi Wang
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, People’s Republic of China
| | - Ying Zheng
- Department of Pulmonary and Critical Care Medicine, Clinical Center for Pulmonary Infections, Capital Medical University, Beijing, People’s Republic of China
| | - Rongling Zhang
- Department of Pulmonary and Critical Care Medicine, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Center of Respiratory Medicine, China–Japan Friendship Hospital, Beijing, People’s Republic of China
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, People’s Republic of China
| | - Yeming Wang
- Department of Pulmonary and Critical Care Medicine, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Center of Respiratory Medicine, China–Japan Friendship Hospital, Beijing, People’s Republic of China
- Institute of Respiratory Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
| | - Hui Zhang
- Department of Pulmonary and Critical Care Medicine, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Center of Respiratory Medicine, China–Japan Friendship Hospital, Beijing, People’s Republic of China
- Department of Pulmonary and Critical Care Medicine, Clinical Center for Pulmonary Infections, Capital Medical University, Beijing, People’s Republic of China
| | - Hongyu Liu
- Department of Pulmonary and Critical Care Medicine, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Center of Respiratory Medicine, China–Japan Friendship Hospital, Beijing, People’s Republic of China
- Department of Pulmonary and Critical Care Medicine, Clinical Center for Pulmonary Infections, Capital Medical University, Beijing, People’s Republic of China
| | - William J. Liu
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention,Beijing, People’s Republic of China
| | - Hui Li
- Department of Pulmonary and Critical Care Medicine, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Center of Respiratory Medicine, China–Japan Friendship Hospital, Beijing, People’s Republic of China
- Institute of Respiratory Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
| | - Bin Cao
- Department of Pulmonary and Critical Care Medicine, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Center of Respiratory Medicine, China–Japan Friendship Hospital, Beijing, People’s Republic of China
- Institute of Respiratory Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, People’s Republic of China
- Department of Infectious Disease, Beijing Friendship Hospital, Capital Medical University, Beijing, People’s Republic of China
- Department of Pulmonary and Critical Care Medicine, Clinical Center for Pulmonary Infections, Capital Medical University, Beijing, People’s Republic of China
| |
Collapse
|
39
|
Liu H, Zhang W, Zhang Y, Adegboro AA, Fasoranti DO, Dai L, Pan Z, Liu H, Xiong Y, Li W, Peng K, Wanggou S, Li X. Mime: A flexible machine-learning framework to construct and visualize models for clinical characteristics prediction and feature selection. Comput Struct Biotechnol J 2024; 23:2798-2810. [PMID: 39055398 PMCID: PMC11269309 DOI: 10.1016/j.csbj.2024.06.035] [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: 03/06/2024] [Revised: 06/27/2024] [Accepted: 06/27/2024] [Indexed: 07/27/2024] Open
Abstract
The widespread use of high-throughput sequencing technologies has revolutionized the understanding of biology and cancer heterogeneity. Recently, several machine-learning models based on transcriptional data have been developed to accurately predict patients' outcome and clinical response. However, an open-source R package covering state-of-the-art machine-learning algorithms for user-friendly access has yet to be developed. Thus, we proposed a flexible computational framework to construct a machine learning-based integration model with elegant performance (Mime). Mime streamlines the process of developing predictive models with high accuracy, leveraging complex datasets to identify critical genes associated with prognosis. An in silico combined model based on de novo PIEZO1-associated signatures constructed by Mime demonstrated high accuracy in predicting the outcomes of patients compared with other published models. Furthermore, the PIEZO1-associated signatures could also precisely infer immunotherapy response by applying different algorithms in Mime. Finally, SDC1 selected from the PIEZO1-associated signatures demonstrated high potential as a glioma target. Taken together, our package provides a user-friendly solution for constructing machine learning-based integration models and will be greatly expanded to provide valuable insights into current fields. The Mime package is available on GitHub (https://github.com/l-magnificence/Mime).
Collapse
Affiliation(s)
- Hongwei Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Wei Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Yihao Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Abraham Ayodeji Adegboro
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Deborah Oluwatosin Fasoranti
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Luohuan Dai
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Zhouyang Pan
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Hongyi Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Yi Xiong
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Wang Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Kang Peng
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Siyi Wanggou
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Xuejun Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| |
Collapse
|
40
|
Longo GMC, Sayols S, Stefanova ME, Xie T, Elsayed W, Panagi A, Stavridou AI, Petrosino G, Ing-Simmons E, Melo US, Gothe HJ, Vaquerizas JM, Kotini AG, Papantonis A, Mundlos S, Roukos V. Type II topoisomerases shape multi-scale 3D chromatin folding in regions of positive supercoils. Mol Cell 2024; 84:4267-4281.e8. [PMID: 39486417 DOI: 10.1016/j.molcel.2024.10.007] [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: 02/26/2024] [Revised: 08/19/2024] [Accepted: 10/04/2024] [Indexed: 11/04/2024]
Abstract
Type II topoisomerases (TOP2s) resolve torsional stress accumulated during various cellular processes and are enriched at chromatin loop anchors and topologically associated domain (TAD) boundaries, where, when trapped, can lead to genomic instability promoting the formation of oncogenic fusions. Whether TOP2s relieve topological constraints at these positions and/or participate in 3D chromosome folding remains unclear. Here, we combine 3D genomics, imaging, and GapRUN, a method for the genome-wide profiling of positive supercoiling, to assess the role of TOP2s in shaping chromosome organization in human cells. Acute TOP2 depletion led to the emergence of new, large-scale contacts at the boundaries between active, positively supercoiled, and lamina-associated domains. TOP2-dependent changes at the higher-order chromatin folding were accompanied by remodeling of chromatin-nuclear lamina interactions and of gene expression, while at the chromatin loop level, TOP2 depletion predominantly remodeled transcriptionally anchored, positively supercoiled loops. We propose that TOP2s act as a fine regulator of chromosome folding at multiple scales.
Collapse
Affiliation(s)
- Gabriel M C Longo
- Institute of Molecular Biology gGmbH, Ackermannweg 4, 55128 Mainz, Germany
| | - Sergi Sayols
- Institute of Molecular Biology gGmbH, Ackermannweg 4, 55128 Mainz, Germany
| | - Maria E Stefanova
- Charité-Universitätsmedizin Berlin, Lindenberger Weg 80, 13125 Berlin, Germany; Max Planck Institute for Molecular Genetics, Ihnestr. 63-73, 14195 Berlin, Germany
| | - Ting Xie
- Institute of Pathology, University Medical Center Göttingen, Robert-Koch-Str. 40, 37075 Göttingen, Germany
| | - Waheba Elsayed
- Institute of Molecular Biology gGmbH, Ackermannweg 4, 55128 Mainz, Germany
| | - Anastasia Panagi
- Department of General Biology, Medical School, University of Patras, Rio, Patras 26500 Greece
| | - Amalia I Stavridou
- Department of General Biology, Medical School, University of Patras, Rio, Patras 26500 Greece
| | - Giuseppe Petrosino
- Institute of Molecular Biology gGmbH, Ackermannweg 4, 55128 Mainz, Germany
| | - Elizabeth Ing-Simmons
- MRC London Institute of Medical Sciences, Du Cane Rd., London W12 0HS, United Kingdom; Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, Du Cane Road, London, W12 0NN, UK
| | - Uirá Souto Melo
- Max Planck Institute for Molecular Genetics, Ihnestr. 63-73, 14195 Berlin, Germany
| | - Henrike J Gothe
- Institute of Molecular Biology gGmbH, Ackermannweg 4, 55128 Mainz, Germany
| | - Juan M Vaquerizas
- MRC London Institute of Medical Sciences, Du Cane Rd., London W12 0HS, United Kingdom; Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, Du Cane Road, London, W12 0NN, UK
| | - Andriana G Kotini
- Department of General Biology, Medical School, University of Patras, Rio, Patras 26500 Greece
| | - Argyris Papantonis
- Institute of Pathology, University Medical Center Göttingen, Robert-Koch-Str. 40, 37075 Göttingen, Germany.
| | - Stefan Mundlos
- Max Planck Institute for Molecular Genetics, Ihnestr. 63-73, 14195 Berlin, Germany.
| | - Vassilis Roukos
- Institute of Molecular Biology gGmbH, Ackermannweg 4, 55128 Mainz, Germany; Department of General Biology, Medical School, University of Patras, Rio, Patras 26500 Greece.
| |
Collapse
|
41
|
Roszkowska M. Multilevel Mechanisms of Cancer Drug Resistance. Int J Mol Sci 2024; 25:12402. [PMID: 39596466 PMCID: PMC11594576 DOI: 10.3390/ijms252212402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 11/14/2024] [Accepted: 11/17/2024] [Indexed: 11/28/2024] Open
Abstract
Cancer drug resistance represents one of the most significant challenges in oncology and manifests through multiple interconnected molecular and cellular mechanisms. Objective: To provide a comprehensive analysis of multilevel processes driving treatment resistance by integrating recent advances in understanding genetic, epigenetic, and microenvironmental factors. This is a systematic review of the recent literature focusing on the mechanisms of cancer drug resistance, including genomic studies, clinical trials, and experimental research. Key findings include the following: (1) Up to 63% of somatic mutations can be heterogeneous within individual tumors, contributing to resistance development; (2) cancer stem cells demonstrate enhanced DNA repair capacity and altered metabolic profiles; (3) the tumor microenvironment, including cancer-associated fibroblasts and immune cell populations, plays a crucial role in promoting resistance; and (4) selective pressure from radiotherapy drives the emergence of radioresistant phenotypes through multiple adaptive mechanisms. Understanding the complex interplay between various resistance mechanisms is essential for developing effective treatment strategies. Future therapeutic approaches should focus on combination strategies that target multiple resistance pathways simultaneously, guided by specific biomarkers.
Collapse
Affiliation(s)
- Malgorzata Roszkowska
- Department of Clinical Neuropsychology, Collegium Medicum, Nicolaus Copernicus University, 85-067 Bydgoszcz, Poland
| |
Collapse
|
42
|
Qian K, Feng Q, Wang JR, Zhu JD, Wang P, Guo Y, Zhou T, Zhu QW, Cai L, Zhang Z, He GH. Identification and validation of a novel prognostic signature and key genes related to development of anaplastic thyroid carcinoma. Discov Oncol 2024; 15:680. [PMID: 39562412 DOI: 10.1007/s12672-024-01563-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 11/11/2024] [Indexed: 11/21/2024] Open
Abstract
BACKGROUND Anaplastic thyroid carcinoma (ATC) is a rare but the most aggressive type of thyroid carcinoma. Nevertheless, limited advances were made to reduce mortality and improve survival over the last decades. Therefore, identifying novel diagnostic biomarkers and therapeutic targets for ATC patients is still needed. MATERIALS AND METHODS RNA sequencing data and corresponding clinical features were available from GEO and TCGA databases. We integrated WGCNA and PPI network analysis to identify hub genes associated with ATC development, and RT-qPCR was employed for data verification. Univariate and LASSO Cox regression analyses were used to generate prognostic signatures. RESULTS Based on PPI and WGCNA, 6 hub genes were identified, namely KIF2C, PBK, TOP2A, CDK1, KIF20A, and ASPM, which play vital roles in ATC development. Subsequently, RT-qPCR experiments showed that most of these genes were significantly upregulated in CAL-62 cells compared to Nthy-ori 3-1 cells. Moreover, a prognostic signature featuring GPSM2, FGF5, ASXL3, CYP4B1, CLMP, and DUXAP9 was generated, which was also verified by RT-qPCR results and proved as an independent predictor of poorer prognosis of ATC. Additionally, a nomogram incorporating the risk score and clinicopathological parameters was further constructed for accurate prediction of 1-, 3- and 5-year survival probabilities of ATC. CONCLUSIONS Our study identified 6 key genes critical to ATC development and constructed a prognostic signature. These findings provide reliable biomarkers and a relatively comprehensive tumorigenesis profile of ATC, which may inform future strategies for clinical diagnosis and pharmaceutical design.
Collapse
Affiliation(s)
- Kai Qian
- Department of Clinical Pharmacy, 920th Hospital of Joint Logistics Support Force of People's Liberation Army, Kunming, China
- College of Pharmacy, Dali University, Dali, China
| | - Qiang Feng
- Department of Pathology, 920th Hospital of Joint Logistics Support Force of People's Liberation Army, Kunming, China
| | - Jia-Rui Wang
- Oncology Department, Kunming Tongren Hospital, Kunming, China
| | - Jia-De Zhu
- Department of Clinical Pharmacy, 920th Hospital of Joint Logistics Support Force of People's Liberation Army, Kunming, China
- College of Pharmacy, Dali University, Dali, China
| | - Ping Wang
- Department of Clinical Pharmacy, 920th Hospital of Joint Logistics Support Force of People's Liberation Army, Kunming, China
| | - Yu Guo
- Department of Clinical Pharmacy, 920th Hospital of Joint Logistics Support Force of People's Liberation Army, Kunming, China
- College of Pharmacy, Dali University, Dali, China
| | - Tao Zhou
- Department of Clinical Pharmacy, 920th Hospital of Joint Logistics Support Force of People's Liberation Army, Kunming, China
| | - Qian-Wei Zhu
- College of Pharmacy, Dali University, Dali, China
| | - Liao Cai
- Department of Clinical Pharmacy, 920th Hospital of Joint Logistics Support Force of People's Liberation Army, Kunming, China
- College of Pharmacy, Dali University, Dali, China
| | - Zheng Zhang
- Medical Engineering Section, The 306th Hospital of People's Liberation Army (PLA), Beijing, China.
| | - Gong-Hao He
- Department of Clinical Pharmacy, 920th Hospital of Joint Logistics Support Force of People's Liberation Army, Kunming, China.
| |
Collapse
|
43
|
Liu G, Lin W, Zhang K, Chen K, Niu G, Zhu Y, Liu Y, Li P, Li Z, An Y. Elucidating the prognostic and therapeutic significance of TOP2A in various malignancies. Cancer Genet 2024; 288-289:68-81. [PMID: 39454521 DOI: 10.1016/j.cancergen.2024.10.005] [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: 07/24/2024] [Revised: 09/26/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024]
Abstract
Topoisomerase IIα (TOP2A) is a crucial enzyme that plays a vital role in DNA replication and transcription mechanisms. Dysregulated expression of TOP2A has been associated with various malignancies, including hepatocellular carcinoma, prostate cancer, colon cancer, lung cancer and breast cancer. In this review, we summarized the prognostic relevances of TOP2A in various types of cancer. The increased expression of TOP2A has been linked to resistance to therapy and reduced survival rates. Therefore, evaluating TOP2A levels could assist in identifying patients who may derive advantages from molecular targeted therapy. The amplification of TOP2A has been linked to a positive response to chemotherapy regimens that contain anthracycline. Nevertheless, the overexpression of TOP2A also indicates a heightened likelihood of disease recurrence and unfavorable prognosis. The prognostic significance of TOP2A has been extensively studied in various types of cancer. The increased expression of TOP2A is associated with poor clinical outcomes, indicating its potential as a valuable biomarker for assessing risk and stratifying treatment in these malignancies. However, further investigation is needed to elucidate the underlying mechanisms by which TOP2A influences cancer progression and to explore its potential as a therapeutic target.
Collapse
Affiliation(s)
- Guangchao Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; School of Stomatology, Henan University, Kaifeng, 475004, China
| | - Wenlong Lin
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; School of Stomatology, Henan University, Kaifeng, 475004, China
| | - Kaifeng Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; School of Stomatology, Henan University, Kaifeng, 475004, China
| | - Kangxu Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; School of Stomatology, Henan University, Kaifeng, 475004, China
| | - Guanglin Niu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; School of Stomatology, Henan University, Kaifeng, 475004, China
| | - Yonghao Zhu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; School of Stomatology, Henan University, Kaifeng, 475004, China
| | - Yixuan Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key laboratory of cell signal transduction, Henan University, Kaifeng, 475004, China
| | - Pengkun Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key laboratory of cell signal transduction, Henan University, Kaifeng, 475004, China
| | - Zhihao Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key laboratory of cell signal transduction, Henan University, Kaifeng, 475004, China
| | - Yang An
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key laboratory of cell signal transduction, Henan University, Kaifeng, 475004, China.
| |
Collapse
|
44
|
Da Silva AJ, Hästbacka HSE, Luoto JC, Gough RE, Coelho-Rato LS, Laitala LM, Goult BT, Imanishi SY, Sistonen L, Henriksson E. Proteomic profiling identifies a direct interaction between heat shock transcription factor 2 and the focal adhesion adapter talin-1. FEBS J 2024; 291:4830-4848. [PMID: 39285620 DOI: 10.1111/febs.17271] [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: 08/03/2023] [Revised: 06/13/2024] [Accepted: 09/02/2024] [Indexed: 11/02/2024]
Abstract
Heat shock factor 2 (HSF2) is a versatile transcription factor that regulates gene expression under stress conditions, during development, and in disease. Despite recent advances in characterizing HSF2-dependent target genes, little is known about the protein networks associated with this transcription factor. In this study, we performed co-immunoprecipitation coupled with mass spectrometry analysis to identify the HSF2 interactome in mouse testes, where HSF2 is required for normal sperm development. Endogenous HSF2 was discovered to form a complex with several adhesion-associated proteins, a finding substantiated by mass spectrometry analysis conducted in human prostate carcinoma PC-3 cells. Notably, this group of proteins included the focal adhesion adapter protein talin-1 (TLN1). Through co-immunoprecipitation and proximity ligation assays, we demonstrate the conservation of the HSF2-TLN1 interaction from mouse to human. Additionally, employing sequence alignment analyses, we uncovered a TLN1-binding motif in the HSF2 C terminus that binds directly to multiple regions of TLN1 in vitro. We provide evidence that the 25 C-terminal amino acids of HSF2, fused to EGFP, are sufficient to establish a protein complex with TLN1 and modify cell-cell adhesion in human cells. Importantly, this TLN1-binding motif is absent in the C-terminus of a closely related HSF family member, HSF1, which does not form a complex with TLN1. These results highlight the unique molecular characteristics of HSF2 in comparison to HSF1. Taken together, our data unveil the protein partners associated with HSF2 in a physiologically relevant context and identifies TLN1 as the first adhesion-related HSF2-interacting partner.
Collapse
Affiliation(s)
- Alejandro J Da Silva
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Turku, Finland
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Hendrik S E Hästbacka
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Turku, Finland
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Jens C Luoto
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Turku, Finland
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | | | - Leila S Coelho-Rato
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Turku, Finland
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Leena M Laitala
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Turku, Finland
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | | | | | - Lea Sistonen
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Turku, Finland
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Eva Henriksson
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Turku, Finland
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| |
Collapse
|
45
|
Sitarek P, Merecz-Sadowska A, Sikora J, Dudzic M, Wiertek-Płoszaj N, Picot L, Śliwiński T, Kowalczyk T. Flavonoids and their derivatives as DNA topoisomerase inhibitors with anti-cancer activity in various cell models: Exploring a novel mode of action. Pharmacol Res 2024; 209:107457. [PMID: 39389401 DOI: 10.1016/j.phrs.2024.107457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 09/25/2024] [Accepted: 10/07/2024] [Indexed: 10/12/2024]
Abstract
Flavonoids, a diverse group of plant-derived secondary metabolites, have garnered significant attention for their potential anti-cancer properties. This review explores the role of flavonoids as inhibitors of DNA topoisomerases, key enzymes essential for DNA replication, transcription, and cell division. The article offers a comprehensive overview of flavonoid classification, biosynthesis, and their widespread natural occurrence. It further delves into the molecular mechanisms through which flavonoids exert their anti-cancer effects, emphasizing their interactions with topoisomerases. The review provides a thorough analysis of both in vitro and in vivo studies that highlight the topoisomerase inhibitory activities of various flavonoids and their derivatives. Key findings demonstrate that flavonoids can function as catalytic inhibitors, poisons, or DNA intercalators, affecting both type I and type II topoisomerases. The structure-activity relationships of flavonoids concerning their topoisomerase inhibitory potency are also examined. This review underscores the potential of flavonoids as promising lead compounds for the development of novel topoisomerase inhibitors, which could have important implications for cancer therapy. However, it also acknowledges the need for further research to fully understand the intricate interactions between flavonoids and topoisomerases within the cellular environment.
Collapse
Affiliation(s)
- Przemysław Sitarek
- Department of Medical Biology, Medical University of Lodz, Muszynskiego 1, Lodz 90-151, Poland.
| | - Anna Merecz-Sadowska
- Department of Economic and Medical Informatics, University of Lodz, Lodz 90-214, Poland
| | - Joanna Sikora
- Department of Bioinorganic Chemistry, Medical University of Lodz, Muszynskiego 1, Lodz 90-151, Poland
| | - Malwina Dudzic
- Students Research Group, Department of Medical Biology, Medical University of Lodz, Lodz 90-151, Poland
| | - Natasza Wiertek-Płoszaj
- Students Research Group, Department of Molecular Biotechnology and Genetics, Faculty of Biology and Environmental Protection, University of Lodz, Banacha 12/16, Lodz 90-237, Poland
| | - Laurent Picot
- Littoral Environnement et Sociétés UMRi CNRS 7266 LIENSs, La Rochelle Université, La Rochelle 17042, France
| | - Tomasz Śliwiński
- Department of Molecular Genetics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, Lodz 90-236, Poland
| | - Tomasz Kowalczyk
- Department of Molecular Biotechnology and Genetics, Faculty of Biology and Environmental Protection, University of Lodz, Banacha 12/16, Lodz 90-237, Poland
| |
Collapse
|
46
|
Liu F, Tian L, Zhang Y, Deng W, Xu X, Zou Y, An R. DIA proteomic and PRM validation through human granulose cells profiles screen suitable biomarkers for polycystic ovary syndrome patients. J Proteomics 2024; 309:105332. [PMID: 39424224 DOI: 10.1016/j.jprot.2024.105332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 10/12/2024] [Accepted: 10/12/2024] [Indexed: 10/21/2024]
Abstract
The aim of this study is to identify differentially expressed proteins (DEPs) in granulose cells (GCs) from women with or withoutpolycystic ovary syndrome (PCOS) via data independent acquisition (DIA) proteomic analysis.A total of 63 women were recruited for this study, 34 PCOS patients as experimental group (P), and 29 women without PCOS as Normal group (NP). DIA-based proteomic analysis was performed to identify DEPs in GCs between the P and NP samples. Certain typical DEPs were further validated by Parallel reaction monitoring (PRM), and correlation analysis was performed between these DEPs and the clinical characteristics.Cell vitality was measured by CCK-8 assay. DIA analysis revealed 174 significantly DEPs, of which 7 were upregulated and 167 downregulated. Bioinformatics analysis was performed to analysis the significantly DEPs. The PRM experiment confirmed TOP2A and SPHKAP were upregulated significantly in P by comparing to NP, while GM2A, MRPS16, APOA2 and FGF2 were downregulated significantly. Most notably, Correlation analysis revealed that TOP2A, SPHKAP, MRPS16 and FGF2were positively correlated with TG, AMH and Age, but negatively correlated with Menarche age, DBIL, FT3, Basal serum FSH and LH.Meanwhile, CCK-8 assay has shown that downregulation of FGF2 could weaken cell viability. Finally, a panel of DEPs were identified in the GCs of patients with PCOS, of which certain significant DEPs might play essential roles in the pathogenesis of PCOS, could be regarded as candidate biomarkers for PCOS.
Collapse
Affiliation(s)
- Faying Liu
- Department of Obstetrics and Gynecology, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Central Laboratory, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Reproductive Health, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi 330006, China
| | - Lifeng Tian
- Reproductive Medicine Center, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi 330006, China
| | - Ying Zhang
- Central Laboratory, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi 330006, China
| | - Wei Deng
- Department of Obstetrics and Gynecology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang 321099, China
| | - Xiaoyun Xu
- Department of Quality Control, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi 330006, China
| | - Yang Zou
- Central Laboratory, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Reproductive Health, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi 330006, China
| | - Ruifang An
- Department of Obstetrics and Gynecology, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China.
| |
Collapse
|
47
|
Tan M, Sun S, Liu Y, Perreault AA, Phanstiel DH, Dou L, Pang B. Targeting the 3D genome by anthracyclines for chemotherapeutic effects. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.15.614434. [PMID: 39463926 PMCID: PMC11507702 DOI: 10.1101/2024.10.15.614434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
The chromatins are folded into three-dimensional (3D) structures inside cells, which coordinates the regulation of gene transcription by the non-coding regulatory elements. Aberrant chromatin 3D folding has been shown in many diseases, such as acute myeloid leukemia (AML), and may contribute to tumorigenesis. The anthracycline topoisomerase II inhibitors can induce histone eviction and DNA damage. We performed genome-wide high-resolution mapping of the chemotherapeutic effects of various clinically used anthracycline drugs. ATAC-seq was used to profile the histone eviction effects of different anthracyclines. TOP2A ChIP-seq was used to profile the potential DNA damage regions. Integrated analyses show that different anthracyclines have distinct target selectivity on epigenomic regions, based on their respective ATAC-seq and ChIP-seq profiles. We identified the underlying molecular mechanism that unique anthracycline variants selectively target chromatin looping anchors via disrupting CTCF binding, suggesting an additional potential therapeutic effect on the 3D genome. We further performed Hi-C experiments, and data from K562 cells treated with the selective anthracycline drugs indicate that the 3D chromatin organization is disrupted. Furthermore, AML patients receiving anthracycline drugs showed altered chromatin structures around potential looping anchors, which linked to distinct clinical outcomes. Our data indicate that anthracyclines are potent and selective epigenomic targeting drugs and can target the 3D genome for anticancer therapy, which could be used for personalized medicine to treat tumors with aberrant 3D chromatin structures.
Collapse
|
48
|
Huang Y, Urban C, Hubel P, Stukalov A, Pichlmair A. Protein turnover regulation is critical for influenza A virus infection. Cell Syst 2024; 15:911-929.e8. [PMID: 39368468 DOI: 10.1016/j.cels.2024.09.004] [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: 12/19/2023] [Revised: 08/16/2024] [Accepted: 09/13/2024] [Indexed: 10/07/2024]
Abstract
The abundance of a protein is defined by its continuous synthesis and degradation, a process known as protein turnover. Here, we systematically profiled the turnover of proteins in influenza A virus (IAV)-infected cells using a pulse-chase stable isotope labeling by amino acids in cell culture (SILAC)-based approach combined with downstream statistical modeling. We identified 1,798 virus-affected proteins with turnover changes (tVAPs) out of 7,739 detected proteins (data available at pulsechase.innatelab.org). In particular, the affected proteins were involved in RNA transcription, splicing and nuclear transport, protein translation and stability, and energy metabolism. Many tVAPs appeared to be known IAV-interacting proteins that regulate virus propagation, such as KPNA6, PPP6C, and POLR2A. Notably, our analysis identified additional IAV host and restriction factors, such as the splicing factor GPKOW, that exhibit significant turnover rate changes while their total abundance is minimally affected. Overall, we show that protein turnover is a critical factor both for virus replication and antiviral defense.
Collapse
Affiliation(s)
- Yiqi Huang
- Institute of Virology, Technical University of Munich, School of Medicine, Munich, Germany
| | - Christian Urban
- Institute of Virology, Technical University of Munich, School of Medicine, Munich, Germany
| | - Philipp Hubel
- Core Facility Hohenheim, Universität Hohenheim, Stuttgart, Germany
| | - Alexey Stukalov
- Institute of Virology, Technical University of Munich, School of Medicine, Munich, Germany
| | - Andreas Pichlmair
- Institute of Virology, Technical University of Munich, School of Medicine, Munich, Germany; Institute of Virology, Helmholtz Munich, Munich, Germany; German Centre for Infection Research (DZIF), Partner Site, Munich, Germany.
| |
Collapse
|
49
|
Samantaray S, Joshi N, Vasa S, Shibu S, Kaloni A, Parekh B, Modi A. Integrated bioinformatics reveals genetic links between visceral obesity and uterine tumors. Mol Genet Genomics 2024; 299:93. [PMID: 39368016 DOI: 10.1007/s00438-024-02184-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 09/11/2024] [Indexed: 10/07/2024]
Abstract
Visceral obesity (VO), characterized by excess fat around internal organs, is a recognized risk factor for gynecological tumors, including benign uterine leiomyoma (ULM) and malignant uterine leiomyosarcoma (ULS). Despite this association, the shared molecular mechanisms remain underexplored. This study utilizes an integrated bioinformatics approach to elucidate common molecular pathways and identify potential therapeutic targets linking VO, ULM, and ULS. We analyzed gene expression datasets from the Gene Expression Omnibus (GEO) to identify differentially expressed genes (DEGs) in each condition. We found 101, 145, and 18 DEGs in VO, ULM, and ULS, respectively, with 37 genes overlapping across all three conditions. Functional enrichment analysis revealed that these overlapping DEGs were significantly enriched in pathways related to cell proliferation, immune response, and transcriptional regulation, suggesting shared biological processes. Protein-protein interaction network analysis identified 14 hub genes, of which TOP2A, APOE, and TYMS showed significant differential expression across all three conditions. Drug-gene interaction analysis identified 26 FDA-approved drugs targeting these hub genes, highlighting potential therapeutic opportunities. In conclusion, this study uncovers shared molecular pathways and actionable drug targets across VO, ULM, and ULS. These findings deepen our understanding of disease etiology and offer promising avenues for drug repurposing. Experimental validation is needed to translate these insights into clinical applications and innovative treatments.
Collapse
Affiliation(s)
- Swayamprabha Samantaray
- School of Applied Sciences and Technology, Gujarat Technological University, Ahmedabad, Gujarat, 382424, India
| | - Nidhi Joshi
- School of Applied Sciences and Technology, Gujarat Technological University, Ahmedabad, Gujarat, 382424, India
| | - Shrinal Vasa
- School of Applied Sciences and Technology, Gujarat Technological University, Ahmedabad, Gujarat, 382424, India
| | - Shan Shibu
- School of Applied Sciences and Technology, Gujarat Technological University, Ahmedabad, Gujarat, 382424, India
| | - Aditi Kaloni
- School of Applied Sciences and Technology, Gujarat Technological University, Ahmedabad, Gujarat, 382424, India
| | - Bhavin Parekh
- School of Applied Sciences and Technology, Gujarat Technological University, Ahmedabad, Gujarat, 382424, India.
- Department of Validation Indic Knowledge Through Advanced Research, Gujarat University, Ahmedabad, Gujarat, 380009, India.
| | - Anupama Modi
- School of Applied Sciences and Technology, Gujarat Technological University, Ahmedabad, Gujarat, 382424, India.
| |
Collapse
|
50
|
Nitiss KC, Bandak A, Berger JM, Nitiss JL. Genome Instability Induced by Topoisomerase Misfunction. Int J Mol Sci 2024; 25:10247. [PMID: 39408578 PMCID: PMC11477040 DOI: 10.3390/ijms251910247] [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: 08/28/2024] [Revised: 09/19/2024] [Accepted: 09/20/2024] [Indexed: 10/20/2024] Open
Abstract
Topoisomerases alter DNA topology by making transient DNA strand breaks (DSBs) in DNA. The DNA cleavage reaction mechanism includes the formation of a reversible protein/DNA complex that allows rapid resealing of the transient break. This mechanism allows changes in DNA topology with minimal risks of persistent DNA damage. Nonetheless, small molecules, alternate DNA structures, or mutations in topoisomerase proteins can impede the resealing of the transient breaks, leading to genome instability and potentially cell death. The consequences of high levels of enzyme/DNA adducts differ for type I and type II topoisomerases. Top1 action on DNA containing ribonucleotides leads to 2-5 nucleotide deletions in repeated sequences, while mutant Top1 enzymes can generate large deletions. By contrast, small molecules that target Top2, or mutant Top2 enzymes with elevated levels of cleavage lead to small de novo duplications. Both Top1 and Top2 have the potential to generate large rearrangements and translocations. Thus, genome instability due to topoisomerase mis-function is a potential pathogenic mechanism especially leading to oncogenic progression. Recent studies support the potential roles of topoisomerases in genetic changes in cancer cells, highlighting the need to understand how cells limit genome instability induced by topoisomerases. This review highlights recent studies that bear on these questions.
Collapse
Affiliation(s)
- Karin C. Nitiss
- Pharmaceutical Sciences Department, University of Illinois Chicago, Rockford, IL 61107, USA;
| | - Afif Bandak
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins School of Medicine, Baltimore, MD 20215, USA; (A.B.); (J.M.B.)
| | - James M. Berger
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins School of Medicine, Baltimore, MD 20215, USA; (A.B.); (J.M.B.)
| | - John L. Nitiss
- Pharmaceutical Sciences Department, University of Illinois Chicago, Rockford, IL 61107, USA;
| |
Collapse
|