1
|
Huang SM, Chen HQ, Liu LT, Zhang YT, Wang J, Zhou DH, Fang JP, Xu LH. Clinical characteristics and prognostic analysis of CDKN2A/2B gene in pediatric acute lymphoblastic leukemia: a retrospective case-control study. Hematology 2025; 30:2439606. [PMID: 39676312 DOI: 10.1080/16078454.2024.2439606] [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: 08/07/2024] [Accepted: 12/03/2024] [Indexed: 12/17/2024] Open
Abstract
In this retrospective case-control study involving 424 pediatric patients diagnosed with Pediatric Acute Lymphoblastic Leukemia (ALL), the investigation focused on analyzing the clinical characteristics and prognosis associated with the Cyclin-dependent kinase inhibitor 2A/2B (CDKN2A/2B) gene. Treatment and evaluation followed the South China Children's Leukemia Group-ALL-2016 protocol (SCCLG-ALL-2016). Among the cohort, 92 patients (21.7%) exhibited CDKN2A/2B gene deletions, with 11.1% homozygous and 10.6% heterozygous deletions. Notably, ALL patients that do have CDKN2A/2B gene deletions tended to present at an older age (P = 0.001), demonstrate hepatosplenomegaly on palpation (P < 0.001), and exhibit a higher incidence of Central nervous system leukemia (CNSL) (P = 0.037) and T-ALL (P = 0.007). A significant correlation was observed between ALL that does have CDKN2A/2B gene deletions and ETV6::RUNX1-positive (8.7% vs. 19.3%, P = 0.017) and IKZF1 gene deletions (20.7% vs. 8.4%, P = 0.001). Survival analysis of 392 patients revealed no significant differences in 5-year relapse, Overall survival (OS), or Event-free survival (EFS) between ALL that does/ does not have CDKN2A/2B gene deletions. Subgroup analysis highlighted poorer prognosis among hepatosplenomegaly patients in the CDKN2A/2B gene deletion group, with a 5-year EFS of 81.8%, 95%CI (0.695-0.963), P = 0.05. Hepatosplenomegaly emerged as the most significant prognostic factor for EFS [HR = 2.306, 95%CI (1.192-4.461), P = 0.013]. Cox regression analyses identified covariates influencing prognosis, ALL with the CDKN2A/2B gene showing no significant impact on outcomes. In conclusion, while ALL that does have CDKN2A/2B gene deletions is associated with certain clinical characteristics and genetic aberrations, they did not significantly impact OS or EFS. Furthermore, subgroup analysis revealed a potential prognostic role of ALL that does have CDKN2A/2B deletions presenting with hepatosplenomegaly on palpation, emphasizing the importance of comprehensive risk stratification in treatment decision-making for this subgroup.
Collapse
Affiliation(s)
- Shi-Mei Huang
- Children's Medical Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Hui-Qin Chen
- Department of Pediatrics, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Li-Ting Liu
- Department of Pediatrics, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Ya-Ting Zhang
- Children's Medical Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Jian Wang
- Children's Medical Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Dun-Hua Zhou
- Children's Medical Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Jian-Pei Fang
- Children's Medical Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Lu-Hong Xu
- Children's Medical Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| |
Collapse
|
2
|
Fu Z, Jin C, Yu S, Xu H, Zhang H, Qiu M, Dong J, Duan S. Unraveling the role of miR-767 in tumor progression: Mechanisms and clinical implications. Gene 2025; 949:149366. [PMID: 40023339 DOI: 10.1016/j.gene.2025.149366] [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: 08/04/2024] [Revised: 01/05/2025] [Accepted: 02/24/2025] [Indexed: 03/04/2025]
Abstract
MicroRNAs (miRNAs), a distinctive class of small single-stranded non-coding RNA molecules typically spanning between 21 and 23 nucleotides, hold a pivotal position within the intricate regulatory network governing gene expression. Notably, miR-767, located on chromosome Xq28, has emerged as a significant player in tumor development, with its two mature products, miR-767-3p and miR-767-5p, garnering considerable attention in scientific inquiry. Extensive investigations reveal aberrant expression patterns of miR-767 across a spectrum of cancers affecting neurological, digestive, reproductive, urinary, and respiratory systems. Remarkably, miR-767 exhibits substantial upregulation in 13 distinct cancer types and demonstrates precise targeting of at least 14 pivotal protein-coding genes (PCGs) crucial for regulating cellular processes including the cell cycle, proliferation, epithelial-mesenchymal transition (EMT), invasion, and migration. Moreover, the expression level of miR-767 bears significant implications for cancer patient diagnosis, prognosis, and drug sensitivity, thus offering novel insights for clinical tumor management. At the mechanistic level, miR-767-5p and miR-767-3p intricately participate in the regulation of key signaling pathways, with miR-767-5p influencing JAK/STAT, EPK1/2, and PI3K/Akt pathways, while miR-767-3p predominantly affects TGF-β and PI3K/Akt pathways. Notably, both miRNAs converge on the PI3K/Akt pathway, underscoring its pivotal role in their joint regulation. This review provides a comprehensive analysis of the intricate mechanisms underlying miR-767-mediated tumor progression through the modulation of diverse target genes, and explores the potential correlation between host gene GABRA3 transcription and the expression of these miRNAs. Furthermore, the review systematically delineates the binding sites of miR-767-5p and miR-767-3p with circRNA and target genes, alongside the PCGs regulated by miR-767, offering profound insights into their multifaceted roles in tumor development. In essence, this review not only comprehensively elucidates the pivotal role of miR-767 in tumor progression but also provides valuable cues and avenues for future research, thereby fostering deeper scientific inquiry within the realm of cancer research.
Collapse
Affiliation(s)
- Zheng Fu
- Department of Clinical Medicine, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Chenghong Jin
- Department of Clinical Medicine, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Sihan Yu
- Department of Clinical Medicine, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Hening Xu
- Department of Clinical Medicine, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Haoyu Zhang
- Department of Clinical Medicine, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Mingxiao Qiu
- Department of Rehabilitation Physical Therapy, Guangzhou Medical University, Guangdong 511400, China
| | - Jingyin Dong
- Department of Clinical Medicine, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China.
| | - Shiwei Duan
- Department of Clinical Medicine, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China; Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China.
| |
Collapse
|
3
|
Wei X, Ning G, Ma H, Yin Y, Ma J, Han L, Chen D, Shi Z. Assessment of in vitro assays and quantitative determination of selectivity and modality of inhibitors targeting the cell cycle regulating, oncogenic cyclin-dependent kinases. Arch Biochem Biophys 2025; 767:110349. [PMID: 39978616 DOI: 10.1016/j.abb.2025.110349] [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: 11/11/2024] [Revised: 01/23/2025] [Accepted: 02/17/2025] [Indexed: 02/22/2025]
Abstract
At the heart of cancer pathology lies the dysregulated cell cycle, which is often driven by aberrant activities of the cell cycle regulating, cyclin-dependent kinases (CDKs). Efforts to harness the therapeutic potential of modulating CDK activities have led to the development of inhibitors with tailored CDK selectivity. However, uniformity in the methods used to evaluate CDK inhibitor selectivity has been lacking and consequently, direct comparison and interpretation of selectivity profiles determined under different assay conditions is difficult. Determination of the inhibition modalities crucial to profiling selectivity of a CDK inhibitor requires thorough kinetic analysis carried out under comparable assay conditions. In this study, we employed a streamlined series of in vitro assays for profiling CDK inhibitors wherein intrinsic inhibition constants and cellular binding parameters were measured by using strategically designed enzymatic inhibition and complementary biophysical assays. Our findings demonstrate the effectiveness of this strategy in determining and quantitatively analyzing the selectivity and inhibition modality of a set of representative CDK inhibitors towards the major oncogenic, cell cycle CDKs. In addition, the assay results provide insights into the inhibitor-target interactions that extend beyond potency and selectivity.
Collapse
Affiliation(s)
- Xiaolu Wei
- Guangdong Provincial Key Laboratory of Advanced Drug Delivery, Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, School of Pharmacy, The Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Guidan Ning
- School of Health, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Huitong Ma
- Department of Biology, Division of Discovery Research, Ionova Biotherapeutics Co., Inc, Foshan, 528000, PR China
| | - Yujiao Yin
- Department of Biology, Division of Discovery Research, Ionova Biotherapeutics Co., Inc, Foshan, 528000, PR China
| | - Jianchun Ma
- The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Liang Han
- School of Health, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China.
| | - Danqi Chen
- Department of Biology, Division of Discovery Research, Ionova Biotherapeutics Co., Inc, Foshan, 528000, PR China.
| | - Zhongfeng Shi
- Guangdong Provincial Key Laboratory of Advanced Drug Delivery, Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, School of Pharmacy, The Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China.
| |
Collapse
|
4
|
Chen YL, Reddy S, Suzuki A. Reversible and effective cell cycle synchronization method for studying stage-specific processes. Life Sci Alliance 2025; 8:e202403000. [PMID: 40037894 PMCID: PMC11880160 DOI: 10.26508/lsa.202403000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 02/20/2025] [Accepted: 02/21/2025] [Indexed: 03/06/2025] Open
Abstract
The cell cycle is a crucial process for cell proliferation, differentiation, and development. Numerous genes and proteins play pivotal roles at specific cell cycle stages to ensure precise regulation of these events. Understanding the stage-specific regulations of the cell cycle requires the accumulation of cell populations at desired cell cycle stages, typically achieved through cell cycle synchronization using kinase and protein inhibitors. However, suboptimal concentrations of these inhibitors can result in inefficiencies, irreversibility, and unintended cellular defects. In this study, we have optimized effective and reversible cell cycle synchronization protocols for human RPE1 cells by combining high-precision cell cycle identification techniques with high-temporal resolution live-cell imaging. These reproducible synchronization methods offer powerful tools for dissecting cell cycle stage-specific regulatory mechanisms.
Collapse
Affiliation(s)
- Yu-Lin Chen
- https://ror.org/01y2jtd41 McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison, Madison, WI, USA
| | - Syon Reddy
- https://ror.org/01y2jtd41 McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison, Madison, WI, USA
| | - Aussie Suzuki
- https://ror.org/01y2jtd41 McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison, Madison, WI, USA
- https://ror.org/01y2jtd41 Carbone Comprehensive Cancer Center, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
5
|
Sulaiman NF, Zulkifli SZ, Saaidin AS, Lekkala R, Izzaty Hassan N, Pungot NH. Exploring β-carboline hybrids and their derivatives: A review on synthesis and anticancer efficiency. Eur J Med Chem 2025; 288:117412. [PMID: 39987835 DOI: 10.1016/j.ejmech.2025.117412] [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: 11/20/2024] [Revised: 01/28/2025] [Accepted: 02/15/2025] [Indexed: 02/25/2025]
Abstract
β-Carboline is a crucial compound in medicinal chemistry known for its versatile pharmacological activities. Recent research has focused on hybrid molecules incorporating a β-carboline scaffold linked to other pharmacophore moieties. These hybrid compounds have demonstrated diverse therapeutic properties, including anticancer, antianxiety, antimalarial, antidepressant, anti-inflammatory, antileishmanial, and antioxidant effects. This review highlights studies conducted from 2014 to the present with a particular emphasis on the development of β-carboline hybrid compounds and their derivatives as potent anticancer agents. The structure-activity relationship (SAR) analysis reveals that these hybrids exhibit significant cytotoxicity against various cancer cell lines. This review aims to inspire further research into the novel synthesis and evolution of β-carboline hybrids and their derivatives, potentially leading to new therapeutic advancements.
Collapse
Affiliation(s)
- Nur Fatihah Sulaiman
- Faculty of Applied Sciences, Universiti Teknologi MARA, Shah Alam, Selangor, 40450, Malaysia
| | - Siti Zafirah Zulkifli
- Faculty of Applied Sciences, Universiti Teknologi MARA, Shah Alam, Selangor, 40450, Malaysia; Organic Synthesis Laboratory, Institute of Science, Universiti Teknologi MARA, Cawangan Selangor, Bandar Puncak Alam, Selangor, 42300, Malaysia
| | - Aimi Suhaily Saaidin
- Organic Synthesis Laboratory, Institute of Science, Universiti Teknologi MARA, Cawangan Selangor, Bandar Puncak Alam, Selangor, 42300, Malaysia
| | - Ravindar Lekkala
- Department of Chemical Sciences, Faculty of Science & Technology, Universiti Kebangsaan Malaysia, Bandar Baru Bangi, Selangor, 43600, Malaysia
| | - Nurul Izzaty Hassan
- Department of Chemical Sciences, Faculty of Science & Technology, Universiti Kebangsaan Malaysia, Bandar Baru Bangi, Selangor, 43600, Malaysia
| | - Noor Hidayah Pungot
- Faculty of Applied Sciences, Universiti Teknologi MARA, Shah Alam, Selangor, 40450, Malaysia; Organic Synthesis Laboratory, Institute of Science, Universiti Teknologi MARA, Cawangan Selangor, Bandar Puncak Alam, Selangor, 42300, Malaysia.
| |
Collapse
|
6
|
Bhattacharya R, Avdieiev SS, Bukkuri A, Whelan CJ, Gatenby RA, Tsai KY, Brown JS. The Hallmarks of Cancer as Eco-Evolutionary Processes. Cancer Discov 2025; 15:685-701. [PMID: 40170539 DOI: 10.1158/2159-8290.cd-24-0861] [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: 06/20/2024] [Revised: 11/19/2024] [Accepted: 01/28/2025] [Indexed: 04/03/2025]
Abstract
SIGNIFICANCE Viewing the hallmarks as a sequence of adaptations captures the "why" behind the "how" of the molecular changes driving cancer. This eco-evolutionary view distils the complexity of cancer progression into logical steps, providing a framework for understanding all existing and emerging hallmarks of cancer and developing therapeutic interventions.
Collapse
Affiliation(s)
- Ranjini Bhattacharya
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Cancer Biology, University of South Florida, Tampa, Florida
| | - Stanislav S Avdieiev
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Anuraag Bukkuri
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania
- Center for Evolutionary Biology and Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Christopher J Whelan
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois
| | - Robert A Gatenby
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Radiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Kenneth Y Tsai
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Tumor Microenvironment & Metastasis, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Pathology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Joel S Brown
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
7
|
Jiang S, Li C, Liu D, Zeng F, Wei W, He T, Yang W. Role, mechanisms and effects of Radix Bupleuri in anti‑breast cancer (Review). Oncol Lett 2025; 29:166. [PMID: 39963320 PMCID: PMC11831725 DOI: 10.3892/ol.2025.14912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 12/30/2024] [Indexed: 02/20/2025] Open
Abstract
The prevalence of breast cancer among women has led to a growing need for innovative anti-breast cancer medications and an in-depth investigation into their molecular mechanisms of action, both of which are essential tactics in clinical intervention. In the clinical practice of Traditional Chinese Medicine, Radix Bupleuri and its active components have shown promise as potential anti-breast cancer agents due to their ability to target multiple pathways, exhibit synergistic effects and reduce toxicity. These compounds are considered to enhance the prognosis of patients with cancer, prolong survival and combat chemotherapy resistance. The present review aimed to delve into the anti-breast cancer properties of Radix Bupleuri and its active ingredients, highlighting their mechanisms, such as inhibition of cell proliferation, promotion of apoptosis, metastasis prevention, microenvironment improvement and synergy with certain chemotherapeutic agents. These findings may provide a scientific rationale for combining Radix Bupleuri and its active components with traditional chemotherapy agents for the management of breast cancer.
Collapse
Affiliation(s)
- Shiting Jiang
- Institute for Cancer Medicine, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Chengxia Li
- Institute for Cancer Medicine, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Dan Liu
- Institute for Cancer Medicine, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Fancai Zeng
- Laboratory of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Wenli Wei
- Institute for Cancer Medicine, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Tao He
- Institute for Cancer Medicine, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Wenli Yang
- Institute for Cancer Medicine, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| |
Collapse
|
8
|
Hagar FF, Abbas SH, Atef E, Abdelhamid D, Abdel-Aziz M. Benzimidazole scaffold as a potent anticancer agent with different mechanisms of action (2016-2023). Mol Divers 2025; 29:1821-1849. [PMID: 39031290 PMCID: PMC11909089 DOI: 10.1007/s11030-024-10907-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Accepted: 05/30/2024] [Indexed: 07/22/2024]
Abstract
Benzimidazole scaffolds have potent anticancer activity due to their structure similarity to nucleoside. In addition, benzimidazoles could function as hydrogen donors or acceptors and bind to different drug targets that participate in cancer progression. The literature had many anticancer agents containing benzimidazole cores that gained much interest. Provoked by our endless interest in benzimidazoles as anticancer agents, we summarized the successful trials of the benzimidazole scaffolds in this concern. Moreover, we discuss the substantial opportunities in cancer treatment using benzimidazole-based drugs that may direct medicinal chemists for a compelling future design of more active chemotherapeutic agents with potential clinical applications. The uniqueness of this work lies in the highlighted benzimidazole scaffold hybridization with different molecules and benzimidazole-metal complexes, detailed mechanisms of action, and the IC50 of the developed compounds determined by different laboratories after 2015.
Collapse
Affiliation(s)
- Fatma Fouad Hagar
- Medicinal Chemistry Department, Faculty of Pharmacy, Minia University, Minia, Egypt
| | - Samar H Abbas
- Medicinal Chemistry Department, Faculty of Pharmacy, Minia University, Minia, Egypt.
| | - Eman Atef
- College of Pharmacy, West Coast University, Los Angeles, CA, USA
| | - Dalia Abdelhamid
- Medicinal Chemistry Department, Faculty of Pharmacy, Minia University, Minia, Egypt.
- Raabe College of Pharmacy, Ohio Northern University, Ohio, USA.
| | - Mohamed Abdel-Aziz
- Medicinal Chemistry Department, Faculty of Pharmacy, Minia University, Minia, Egypt
| |
Collapse
|
9
|
Zhao R, Zhang X, Geng Y, Lu D, Wang Y, Xie H, Zhang X, Xu S, Cao Y. SPRY1 regulates macrophage M1 polarization in skin aging and melanoma prognosis. Transl Oncol 2025; 54:102331. [PMID: 40023001 PMCID: PMC11915026 DOI: 10.1016/j.tranon.2025.102331] [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: 11/28/2024] [Revised: 01/28/2025] [Accepted: 02/10/2025] [Indexed: 03/04/2025] Open
Abstract
INTRODUCTION Skin aging is a complex, multifactorial process involving cellular damage, inflammation, and increased susceptibility to diseases. Despite its importance, the role of SPRY1 in skin aging remains poorly understood. This study aims to investigate the function of SPRY1 in skin aging, particularly its impact on macrophage M1 polarization, and explore its potential as a therapeutic target for mitigating skin aging and melanoma. METHODS Bioinformatics analyses were performed using datasets from the GTEx and GEO databases, alongside in vitro cellular experiments. These included Weighted Gene Co-expression Network Analysis (WGCNA), single-cell sequencing, and various cellular assays in RAW264.7 murine monocyte/macrophage leukemia cells and NIH/3T3 mouse skin fibroblasts. The assays comprised gene transfection, Cell Counting Kit-8 (CCK-8) assays, quantitative real-time PCR (qRT-PCR), and measurements of reactive oxygen species (ROS) and superoxide dismutase (SOD) activity. RESULTS SPRY1 was identified as a key gene within modules linked to skin aging. Single-cell sequencing revealed its enrichment in macrophages and keratinocytes. Knockdown of SPRY1 in RAW264.7 cells resulted in a shift from M1 to M2 macrophage polarization, reduced oxidative stress, and decreased expression of inflammatory markers. In NIH/3T3 cells, SPRY1 knockdown reduced cell viability and lowered the expression of inflammatory genes. Additionally, SPRY1 expression was downregulated in melanoma, and its reduced levels were associated with poorer survival outcomes. CONCLUSIONS SPRY1 accelerates skin aging by promoting macrophage M1 polarization and may serve as a promising therapeutic target. Future research should focus on in vivo validation and further exploration of its regulatory networks to develop novel treatments.
Collapse
Affiliation(s)
- Rongxin Zhao
- Department of Dermatology, Pudong New Area People's Hospital, 490 Chuanhuang South Road, Pudong New Area, Shanghai, China
| | - Xun Zhang
- Digestive Endoscopy Center, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, 1111 XianXia Road, Shanghai 200336, China
| | - Yingnan Geng
- Department of Burns and Plastic Surgery, Second Affiliated Hospital of Naval Medical University, 415 Fengyang Road, Huangpu District, Shanghai 200003, China
| | - Dan Lu
- Department of Dermatology, Pudong New Area People's Hospital, 490 Chuanhuang South Road, Pudong New Area, Shanghai, China
| | - Yuqing Wang
- Department of Dermatology, Xuzhou Huamei Cosmetology Hospital, Jiangsu, West Huaihai Road, Quanshan District, Xuzhou, Jiangsu, China
| | - Han Xie
- The Fifth People's Hospital of Shanghai, Fudan University, No. 128, Ruili Road, Minhang District, Shanghai, China
| | - Xiaofei Zhang
- Shanghai Xinmei Medical Beauty Outpatient Department, 202A, No.285, Jianguo West Road, Xuhui District, Shanghai, China.
| | - Shunming Xu
- Department of Dermatology, Pudong New Area People's Hospital, 490 Chuanhuang South Road, Pudong New Area, Shanghai, China.
| | - Yanyun Cao
- Department of Dermatology, Pudong New Area People's Hospital, 490 Chuanhuang South Road, Pudong New Area, Shanghai, China.
| |
Collapse
|
10
|
Sun J, Sun X, Yin L, Jin S, Huang Q, Dong Y, Gu X, Zhang Y, Jin Y, Zhu R, Wang G. Dual Functional Radioactive Gel-Microspheres for Combinatorial Radioembolization and Photothermal Therapy of Hepatocellular Carcinoma. Adv Healthc Mater 2025; 14:e2401057. [PMID: 40025905 DOI: 10.1002/adhm.202401057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 02/17/2025] [Indexed: 03/04/2025]
Abstract
Transarterial radioembolization (TARE) is an established clinical therapy for treating patients with intermediate to advanced hepatocellular carcinoma (HCC) or those who cannot undergo radical treatment. However, the delivery of a high radiation dose is associated with several adverse effects, such as radiation pneumonitis. Additionally, the available radioactive microspheres (MSs) are dense and unsuitable for interventional delivery. This study proposes the use of commercial CalliSpheres polyvinyl alcohol (PVA) gel MSs coated with polydopamine (PDA) as a carrier for radioactive iodine (131I) labeled using the iodogen method, denoted as 131I-PDA@PVA MSs, which can be for radioembolization combined photothermal therapy (PTT) of HCC. In vitro experiments have demonstrated that 131I-PDA@PVA MSs have high radiolabeling stability and photothermal properties. Single photon emission computed tomography (SPECT)/computed tomography (CT) imaging and biodistribution experiments have shown that 131I-PDA@PVA MSs remain stable in vivo without any radioactive leakage. The results of the antitumor study suggest that 131I-PDA@PVA MSs are an effective treatment for inhibiting tumor growth through a combination of radioembolization and PTT while avoiding significant side effects. These multifunctional MSs have great potential for clinical application in the treatment of HCC.
Collapse
Affiliation(s)
- Jing Sun
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, P. R. China
| | - Xingwei Sun
- Department of Interventional, The Second Affiliated Hospital of Soochow University, Suzhou, 215006, P. R. China
| | - Liang Yin
- Department of Interventional, The Second Affiliated Hospital of Soochow University, Suzhou, 215006, P. R. China
| | - Shuo Jin
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, P. R. China
| | - Qian Huang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, P. R. China
| | - Yi Dong
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, P. R. China
| | - Xiaolei Gu
- Suzhou Hengrui Medical Devices Co., Ltd, Suzhou, 215127, P. R. China
| | - Yujuan Zhang
- Experimental Center of Soochow University, Department of Medicine, Soochow University, Suzhou, 215123, P. R. China
| | - Yong Jin
- Department of Interventional, The Second Affiliated Hospital of Soochow University, Suzhou, 215006, P. R. China
| | - Ran Zhu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, P. R. China
| | - Guanglin Wang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, P. R. China
| |
Collapse
|
11
|
Niu S, Huang S, Shi M, Luo Z, Shao Z, Hong B, Tian W. Discovery of new aphidicolin diterpenoids from the deep-sea-derived fungus Botryotinia fuckeliana with cytotoxic activity against human bladder cancer cells. Bioorg Chem 2025; 157:108311. [PMID: 40022848 DOI: 10.1016/j.bioorg.2025.108311] [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: 12/24/2024] [Revised: 02/19/2025] [Accepted: 02/21/2025] [Indexed: 03/04/2025]
Abstract
Aphidicolin, characterized by a highly fused 6/6/5/6 tetracyclic diterpenoid skeleton, had been explored as a potential anticancer drug in clinical trials. However, its development has been constrained by poor solubility. The discovery of new aphidicolin derivatives offers promising prospects for anticancer drug development. In the present study, 37 new aphidicolin derivatives, designated as aphidicolins B1-B37 (1-37), together with 34 known analogues (38-71), were discovered from the ethyl acetate (EtOAc) extract of the deep-sea-derived fungus Botryotinia fuckeliana. Their structures, including absolute configurations, were determined by extensive analyses of spectroscopic data, the phenylglycine methyl ester (PGME) method, modified Mosher's method, and comparison of experimental and calculated electronic circular dichroism (ECD) data. Notably, aphidicolin B12 (12) features a 6/6/5/6/6 pentacyclic framework with an unprecedented γ-lactone ring E, while compound 31 contains a novel 6/6/5/6/5/5 hexacyclic system bearing an unprecedented tetrahydrofuran ring formed by an ether bridge between C-3 and C-6. All isolated aphidicolins were evaluated for their cytotoxic effects against T24 human bladder cancer cells. Among them, 11 diterpenoids showed stronger inhibitory activity than aphidicolin (60), especially compound 32, which exhibited an IC50 value of 1.9 μM, significantly more potent than 60 (IC50 = 27.6 μM). The structure-bioactivity relationships were also discussed. Further mechanistic studies revealed that 32 inhibits T24 cells proliferation by inducing cell cycle arrest in the G0/G1 phase, suggesting its potential as a therapeutic agent for bladder cancer treatment.
Collapse
Affiliation(s)
- Siwen Niu
- Technology Innovation Center for Exploitation of Marine Biological Resources, Third Institute of Oceanography, Ministry of Natural Resources, Xiamen 361005, PR China.
| | - Shuhuan Huang
- Technology Innovation Center for Exploitation of Marine Biological Resources, Third Institute of Oceanography, Ministry of Natural Resources, Xiamen 361005, PR China
| | - Menglei Shi
- Technology Innovation Center for Exploitation of Marine Biological Resources, Third Institute of Oceanography, Ministry of Natural Resources, Xiamen 361005, PR China
| | - Zhuhua Luo
- Technology Innovation Center for Exploitation of Marine Biological Resources, Third Institute of Oceanography, Ministry of Natural Resources, Xiamen 361005, PR China
| | - Zongze Shao
- Technology Innovation Center for Exploitation of Marine Biological Resources, Third Institute of Oceanography, Ministry of Natural Resources, Xiamen 361005, PR China
| | - Bihong Hong
- Technology Innovation Center for Exploitation of Marine Biological Resources, Third Institute of Oceanography, Ministry of Natural Resources, Xiamen 361005, PR China.
| | - Wenjing Tian
- Fujian Provincial Key Laboratory of Innovative Drug Target, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, PR China.
| |
Collapse
|
12
|
Bhattacharyya T, Das P, Ansari A, Mohan AA, Chandra Y, Narayan KP, Banerjee R. Glucocorticoid Receptor-Targeted Nanoliposome for STAT3 Inhibition-Led Myeloid-Derived Suppressor Cell Modulation and Efficient Colon Cancer Treatment. ACS APPLIED BIO MATERIALS 2025. [PMID: 40162961 DOI: 10.1021/acsabm.5c00002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
STAT3 is an important protein responsible for cellular proliferation, motility, and immune tolerance and is hyperactive in colorectal cancer, instigating metastasis, cellular proliferation, migration, as well as inhibition. It helps in proliferation of myeloid-derived suppressor cells (MDSCs), which within the tumor microenvironment (TME) suppress T cells to encourage tumor growth, metastasis, and resistance to immunotherapy, besides playing dynamic role in regulating macrophages within the tumor. Thus, MDSC is a potential target to augment immune surveillance within the TME. Herein, we report targeting both colorectal cancer and MDSCs using a glucocorticoid receptor (GR)-targeted nanoliposomal formulation carrying GR-ligand, dexamethasone (Dex), and a STAT3 inhibitor, niclosamide (N). Our main objective was to selectively inhibit STAT3, the key immunomodulatory factor in most TME-associated cells including MDSCs, and also repurpose the use of this antihelminthic, low-cost drug N for cancer treatment. The resultant formulation D1XN exhibited better tumor regression and survivability compared to GR nontargeted formulation. Further, bone marrow cell-derived MDSCs were engineered by D1XN treatment ex vivo and were inoculated back to tumor-bearing mice. Significant tumor growth inhibition with enhanced antiproliferative immune cell signatures, such as T cell infiltration, decrease in Treg cells, and increased M1/M2 macrophage ratio within the TME were observed. This reveals the effectiveness of engineered MDSCs to modulate tumor surveillance besides reversing the aggressiveness of the tumor. Therefore, D1XN and D1XN-mediated engineered MDSCs alone or in combination can be considered as potent selective chemo-immunotherapeutic nanoliposomal agent(s) against colorectal cancer.
Collapse
Affiliation(s)
- Tithi Bhattacharyya
- Division of Oils, Lipids Science and Technology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Pritam Das
- Division of Oils, Lipids Science and Technology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Aasia Ansari
- Division of Oils, Lipids Science and Technology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Adrij A Mohan
- Department of Biotechnology, Manipal Institute of Technology, Manipal, Karnataka 576104, India
| | - Yogesh Chandra
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
| | - Kumar Pranav Narayan
- Department of Biological Sciences, BITS Pilani, Hyderabad Campus, Jawahar Nagar, Hyderabad 500078, India
| | - Rajkumar Banerjee
- Division of Oils, Lipids Science and Technology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
13
|
Loycano MA, Pienta KJ, Amend SR. Temporal myc dynamics permit mitotic bypass, promoting polyploid phenotypes. Cancer Lett 2025; 613:217526. [PMID: 39909233 PMCID: PMC11924244 DOI: 10.1016/j.canlet.2025.217526] [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: 10/17/2024] [Revised: 01/29/2025] [Accepted: 02/02/2025] [Indexed: 02/07/2025]
Abstract
High Myc phenotypes are extensively documented in the hyperproliferative cell cycle of cancer cells, as well as non-proliferative endoreplication cycles engaged during normal development and stress response. Notably, endoreplication in cancer produces chemotherapy resistant polyploid cells, necessitating a clearer understanding of altered cell cycle regulation that uncouples DNA replication and mitotic cell division. The c-Myc oncogene is a well-established transcriptional regulator of cell cycle progression and has been extensively published as an essential driver of the G1/S transition. Beyond S phase, Myc transcriptionally activates the proteins that drive mitotic entry. Sustained activation of Myc through the cell cycle transcriptionally couples DNA replication and mitotic cell division. Based on the literature in this field, we propose a new model of temporal regulation of Myc activity that serves to either couple or uncouple these two processes, determining cell cycle fate - proliferation or polyploidy. The mitotic cell cycle requires two pulses of Myc activity - the first driving the G1/S transition and the second driving the G2/M transition. During mitosis, Myc activity must be silenced to achieve high-fidelity division. Absence of the second activity pulse during G2 results in the downregulation of the proteins essential for mitotic entry and permits premature activation of APC/C, inducing mitotic bypass. A subsequent rise of Myc activity following mitotic bypass permits genome re-replication, driving polyploid phenotypes. This model serves to provide a new level of understanding to the global regulation of S phase-mitosis coupling, as well as a new lens to view low Myc phenotypes.
Collapse
Affiliation(s)
- Michael A Loycano
- Cancer Ecology Center, James Buchanan Brady Urological Institute, Johns Hopkins Medical Institute, Baltimore, MD, USA; Cellular and Molecular Medicine Graduate Training Program, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| | - Kenneth J Pienta
- Cancer Ecology Center, James Buchanan Brady Urological Institute, Johns Hopkins Medical Institute, Baltimore, MD, USA; Cellular and Molecular Medicine Graduate Training Program, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Sarah R Amend
- Cancer Ecology Center, James Buchanan Brady Urological Institute, Johns Hopkins Medical Institute, Baltimore, MD, USA; Cellular and Molecular Medicine Graduate Training Program, Johns Hopkins School of Medicine, Baltimore, MD, USA
| |
Collapse
|
14
|
Benhassoun R, Morel AP, Jacquot V, Puisieux A, Ouzounova M. The epipliancy journey: Tumor initiation at the mercy of identity crisis and epigenetic drift. Biochim Biophys Acta Rev Cancer 2025; 1880:189307. [PMID: 40174706 DOI: 10.1016/j.bbcan.2025.189307] [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: 10/25/2024] [Revised: 03/05/2025] [Accepted: 03/27/2025] [Indexed: 04/04/2025]
Abstract
Cellular pliancy refers to the unique disposition of different stages of cellular differentiation to transform when exposed to specific oncogenic insults. This concept highlights a strong interconnection between cellular identity and tumorigenesis, and implies overcoming of epigenetic barriers defining cellular states. Emerging evidence suggests that the cell-type-specific response to intrinsic and extrinsic stresses is modulated by accessibility to certain areas of the genome. Understanding the interplay between epigenetic mechanisms, cellular differentiation, and oncogenic insults is crucial for deciphering the complex nature of tumorigenesis and developing targeted therapies. Hence, cellular pliancy relies on a dynamic cooperation between the cellular identity and the cellular context through epigenetic control, including the reactivation of cellular mechanisms, such as epithelial-to-mesenchymal transition (EMT). Such mechanisms and pathways confer plasticity to the cell allowing it to adapt to a hostile environment in a context of tumor initiation, thus changing its cellular identity. Indeed, growing evidence suggests that cancer is a disease of cell identity crisis, whereby differentiated cells lose their defined identity and gain progenitor characteristics. The loss of cell fate commitment is a central feature of tumorigenesis and appears to be a prerequisite for neoplastic transformation. In this context, EMT-inducing transcription factors (EMT-TFs) cooperate with mitogenic oncoproteins to foster malignant transformation. The aberrant activation of EMT-TFs plays an active role in tumor initiation by alleviating key oncosuppressive mechanisms and by endowing cancer cells with stem cell-like properties, including the ability to self-renew, thus changing the course of tumorigenesis. This highly dynamic phenotypic change occurs concomitantly to major epigenome reorganization, a key component of cell differentiation and cancer cell plasticity regulation. The concept of pliancy was initially proposed to address a fundamental question in cancer biology: why are some cells more likely to become cancerous in response to specific oncogenic events at particular developmental stages? We propose the concept of epipliancy, whereby a difference in epigenetic configuration leads to malignant transformation following an oncogenic insult. Here, we present recent studies furthering our understanding of how the epigenetic landscape may impact the modulation of cellular pliancy during early stages of cancer initiation.
Collapse
Affiliation(s)
- Rahma Benhassoun
- Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Cancer Research Center of Lyon, France; LabEx DEVweCAN, Université de Lyon, France
| | - Anne-Pierre Morel
- Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Cancer Research Center of Lyon, France; LabEx DEVweCAN, Université de Lyon, France
| | - Victoria Jacquot
- Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Cancer Research Center of Lyon, France
| | - Alain Puisieux
- Equipe labellisée Ligue contre le cancer, U1339 Inserm - UMR3666 CNRS, Paris, France; Institut Curie, PSL Research University, Paris, France
| | - Maria Ouzounova
- Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Cancer Research Center of Lyon, France; LabEx DEVweCAN, Université de Lyon, France.
| |
Collapse
|
15
|
Tang W, Zhang D, Liu D, Liu Z, Xiao K, Lei C, Yang Y, Zhou Q, Wang X. E2F7 upregulates MCM4 and fatty acid metabolism to advance lung adenocarcinoma metastasis. Prostaglandins Other Lipid Mediat 2025; 178:106988. [PMID: 40158794 DOI: 10.1016/j.prostaglandins.2025.106988] [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: 11/14/2024] [Revised: 03/18/2025] [Accepted: 03/25/2025] [Indexed: 04/02/2025]
Abstract
BACKGROUND MCM4, a key protein in MCM, is frequently overexpressed in cancers, but its specific role in lung adenocarcinoma (LUAD) metastasis is unclear. METHODS Bioinformatics revealed the mRNA expression pattern of MCM4 in LUAD, which we confirmed in both normal lung epithelial and adenocarcinoma cell lines using qRT-PCR and western blot (WB). Cellular proliferation was gauged by cell counting kit-8 and colony formation assays, and the expression of epithelial-mesenchymal transition markers along with fatty acid synthase (FASN) was probed via WB. We employed Transwell to assess cellular migration and invasion, and utilized kits for quantifying intracellular triglycerides and phospholipids. Bioinformatics identified E2F7 as a potential transcriptional regulator of MCM4, prompting us to explore its relationship with MCM4, including predicted binding sites and E2F7 mRNA expression in LUAD. Chromatin immunoprecipitation and dual-luciferase reporter assays were conducted to validate the regulatory effects of E2F7 on MCM4. RESULTS MCM4 was found to be overexpressed in LUAD, and its knockdown inhibited cancer cell proliferation, migration, invasion, and metastasis, along with decreased FASN expression and declined levels of triglycerides and phospholipids within cells. Mechanistically, E2F7 transcriptionally activated MCM4, regulating fatty acid metabolism and promoting LUAD progression and metastasis. CONCLUSION Our study elucidates the mechanism by which E2F7 transcriptionally controls MCM4 to activate fatty acid metabolism, fueling LUAD metastasis. These discoveries emphasize the pivotal function of lipid metabolism in LUAD development and suggests new therapeutic targets for LUAD treatment.
Collapse
Affiliation(s)
- WuAsen Tang
- Department of Cardiothoracic and Macrovascular Surgery, Jingzhou Hospital Affiliated to Yangtze University, No.26 Chuyuan Avenue, Jingzhou District, Jingzhou City, Hubei province 434020, China
| | - Deming Zhang
- Department of Cardiothoracic and Macrovascular Surgery, Jingzhou Hospital Affiliated to Yangtze University, No.26 Chuyuan Avenue, Jingzhou District, Jingzhou City, Hubei province 434020, China.
| | - Di Liu
- Department of Cardiothoracic and Macrovascular Surgery, Jingzhou Hospital Affiliated to Yangtze University, No.26 Chuyuan Avenue, Jingzhou District, Jingzhou City, Hubei province 434020, China
| | - Zikang Liu
- Department of Cardiothoracic and Macrovascular Surgery, Jingzhou Hospital Affiliated to Yangtze University, No.26 Chuyuan Avenue, Jingzhou District, Jingzhou City, Hubei province 434020, China
| | - Kuang Xiao
- Department of Cardiothoracic and Macrovascular Surgery, Jingzhou Hospital Affiliated to Yangtze University, No.26 Chuyuan Avenue, Jingzhou District, Jingzhou City, Hubei province 434020, China
| | - Chenggang Lei
- Department of Cardiothoracic and Macrovascular Surgery, Jingzhou Hospital Affiliated to Yangtze University, No.26 Chuyuan Avenue, Jingzhou District, Jingzhou City, Hubei province 434020, China
| | - Yalun Yang
- Department of Cardiothoracic and Macrovascular Surgery, Jingzhou Hospital Affiliated to Yangtze University, No.26 Chuyuan Avenue, Jingzhou District, Jingzhou City, Hubei province 434020, China
| | - Qian Zhou
- Department of Cardiothoracic and Macrovascular Surgery, Jingzhou Hospital Affiliated to Yangtze University, No.26 Chuyuan Avenue, Jingzhou District, Jingzhou City, Hubei province 434020, China
| | - Xianghui Wang
- Department of Cardiothoracic and Macrovascular Surgery, Jingzhou Hospital Affiliated to Yangtze University, No.26 Chuyuan Avenue, Jingzhou District, Jingzhou City, Hubei province 434020, China.
| |
Collapse
|
16
|
Lu B, Winnall S, Cross W, Barnes CP. Cell-cycle dependent DNA repair and replication unifies patterns of chromosome instability. Nat Commun 2025; 16:3033. [PMID: 40155604 PMCID: PMC11953314 DOI: 10.1038/s41467-025-58245-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 03/17/2025] [Indexed: 04/01/2025] Open
Abstract
Chromosomal instability (CIN) is pervasive in human tumours and often leads to structural or numerical chromosomal aberrations. Somatic structural variants (SVs) are intimately related to copy number alterations but the two types of variant are often studied independently. Additionally, despite numerous studies on detecting various SV patterns, there are still no general quantitative models of SV generation. To address this issue, we develop a computational cell-cycle model for the generation of SVs from end-joining repair and replication after double-strand break formation. Our model provides quantitative information on the relationship between breakage fusion bridge cycle, chromothripsis, seismic amplification, and extra-chromosomal circular DNA. Given whole-genome sequencing data, the model also allows us to infer important parameters in SV generation with Bayesian inference. Our quantitative framework unifies disparate genomic patterns resulted from CIN, provides a null mutational model for SV, and reveals deeper insights into the impact of genome rearrangement on tumour evolution.
Collapse
Affiliation(s)
- Bingxin Lu
- Department of Cell and Developmental Biology, University College London, Gower Street, London, UK.
- UCL Genetics Institute, University College London, Gower Street, London, UK.
- School of Biosciences, University of Surrey, Stag Hill, Guildford, UK.
- Surrey Institute for People-Centred Artificial Intelligence, University of Surrey, Stag Hill, Guildford, UK.
| | - Samuel Winnall
- Department of Cell and Developmental Biology, University College London, Gower Street, London, UK
| | - William Cross
- Department of Cell and Developmental Biology, University College London, Gower Street, London, UK
- Rare Malignancies and Cancer Evolution Laboratory, School of Biological Sciences, University of Reading, Whiteknights, Reading, UK
| | - Chris P Barnes
- Department of Cell and Developmental Biology, University College London, Gower Street, London, UK.
- UCL Genetics Institute, University College London, Gower Street, London, UK.
| |
Collapse
|
17
|
Zhou M, Yang Y, He S, Xu Q, Du C, Tian W, Chen H. Ingenane Diterpenoids from Euphorbia peplus as Potential New CHK1 Inhibitors That Sensitize Cancer Cells to Chemotherapy. JOURNAL OF NATURAL PRODUCTS 2025; 88:688-705. [PMID: 40056138 DOI: 10.1021/acs.jnatprod.4c01343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/10/2025]
Abstract
Phosphorylation of checkpoint kinase 1 at Ser-345 (p-CHK1(S345)) mediates the replication stress response in cancer cells, leading to chemotherapy resistance. Therefore, finding inhibitors of p-CHK1(S345) is currently a promising strategy to prevent acquired drug resistance. In this study, 14 ingenane diterpenoids (1-14), involving two undescribed compounds possessing an unprecedented exocyclic double bond Δ6(20), were identified from Euphorbia peplus. The inhibitory effects of the isolated compounds on p-CHK1(S345) and their structure-activity relationship (SAR) were investigated. Compounds 7 and 8 presented the strongest inhibitory effects, abrogated cell cycle arrest, and caused the accumulation of DNA damage, improving the sensitivity of cancer cells to chemotherapeutic drugs. An in vivo assay confirmed the enhancement of 8 on the anticancer effect of topotecan via blocking of p-CHK1(S345). Mechanistically, 8 increased CHK1 ubiquitination to inhibit p-CHK1(S345) via activation of protein kinase C (PKC). PKC activation was first found to enhance CHK1 ubiquitination to block p-CHK1(S345). Above all, this finding not only indicates that compound 8 could be developed as a novel CHK1 inhibitor but also reveals a previously unrecognized role of PKC in regulating cancer chemotherapy sensitivity.
Collapse
Affiliation(s)
- Mi Zhou
- School of Pharmaceutical Science, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361002, China
- Guangxi Key Laboratory of Marine Drugs, Institute of Marine Drugs, Guangxi University of Chinese Medicine, Nanning 530200, China
| | - Yanlan Yang
- School of Pharmaceutical Science, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361002, China
| | - Shoulun He
- School of Pharmaceutical Science, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361002, China
| | - Qiannan Xu
- School of Pharmaceutical Science, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361002, China
| | - Chunchun Du
- School of Pharmaceutical Science, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361002, China
| | - Wenjing Tian
- School of Pharmaceutical Science, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361002, China
| | - Haifeng Chen
- School of Pharmaceutical Science, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361002, China
| |
Collapse
|
18
|
Yang C, Wang S, Qi Y, Jin Y, Guan R, Huang Z. Mechanisms of adipocyte regulation: Insights from HADHB gene modulation. PLoS One 2025; 20:e0319384. [PMID: 40146690 PMCID: PMC11949335 DOI: 10.1371/journal.pone.0319384] [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: 09/20/2024] [Accepted: 01/31/2025] [Indexed: 03/29/2025] Open
Abstract
The HADHB gene encodes the beta-subunit of 3-hydroxy acyl-CoA dehydrogenase, closely related to energy metabolism, fatty acid synthesis, and catabolism. This study aimed to investigate the effect of the HADHB gene on the proliferation and differentiation of bovine preadipocytes and to gain new insights into the mechanisms of adipocyte regulation. RNA was extracted from adipose tissue of yellow cattle and the HADHB gene CDS region was cloned. Meanwhile, isolation and cultivation of preadipocytes were used for siRNA and adenovirus overexpression, quantitative real-time PCR, transcriptome sequencing, and cell proliferation and cell cycle were measured by oil red staining, CCK8 assay, and flow cytometry. Subsequently, the transcriptome data were analyzed using bioinformatics. The results showed that the HADHB gene modulates significantly the expression of critical genes involved in proliferation (CDK2 and PCNA) and differentiation (PPARγ and CEBPα), influencing preadipocyte proliferation and differentiation and altering cell cycle progression. The results of transcriptome sequencing demonstrated that the overexpression of the HADHB gene markedly altered the transcriptional profile of preadipocytes, with 170 genes exhibiting a significant increase in expression and 113 genes displaying a decrease. The HADHB gene exerts a regulatory influence on the differentiation process of precursor adipocytes by modulating the expression of key genes involved in proliferation and differentiation.These findings highlight the central role of the HADHB gene in adipocyte regulation and provide new insights into the regulatory mechanisms governing adipocyte biology.
Collapse
Affiliation(s)
- Chaoyun Yang
- College of Animal Science and Technology, Xichang University, Xichang, China
| | - Shuzhe Wang
- College of Animal Science and Technology, Xichang University, Xichang, China
| | - Yunxia Qi
- College of Animal Science and Technology, Xichang University, Xichang, China
| | - Yadong Jin
- College of Animal Science and Technology, Xichang University, Xichang, China
| | - Ran Guan
- College of Animal Science and Technology, Xichang University, Xichang, China
| | - Zengwen Huang
- College of Animal Science and Technology, Xichang University, Xichang, China
| |
Collapse
|
19
|
Park J, Lee DH. Loss of protein phosphatase 4 inhibitory protein leads to genomic instability and heightens vulnerability to replication stress. Biochim Biophys Acta Gen Subj 2025; 1869:130797. [PMID: 40157551 DOI: 10.1016/j.bbagen.2025.130797] [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: 11/20/2024] [Revised: 03/09/2025] [Accepted: 03/22/2025] [Indexed: 04/01/2025]
Abstract
Protein phosphatase 4 inhibitory protein (PP4IP) has recently emerged as a key player in cellular processes, particularly in DNA double-strand break repair and telomere maintenance, although research on its functions remains limited. To further investigate the cellular pathways involving PP4IP, we conducted transcriptomic analysis via RNA sequencing in PP4IP-knockout cells and observed an upregulation of p21 expression. This upregulation was linked to an increased population of p21-positive G1-phase cells in the absence of PP4IP. Prior studies have suggested that unresolved under-replicated DNA in mother cells, transmitted to daughter cells, can trigger a quiescent G1 phase characterized by p21 expression and the formation of p53-binding protein 1 (53BP1) nuclear bodies. Consistent with this, we found a higher proportion of 53BP1 nuclear bodies-positive G1 cells in PP4IP-knockout cells compared to controls. Additionally, PP4IP-deficient cells displayed an increased occurrence of anaphase bridges-indicative of incomplete DNA replication-without a corresponding increase in lagging chromosomes. Furthermore, PP4IP-knockout cells exhibited a heightened susceptibility to replication stress, as evidenced by an elevated frequency of replication stress-induced chromatid breaks and increased sensitivity to such stress. Collectively, these results suggest that PP4IP plays a critical role in safeguarding cells from replication stress and ensuring genomic stability.
Collapse
Affiliation(s)
- Jaehong Park
- Department of Biological Sciences, College of Natural Sciences, Chonnam National University, Gwangju, Republic of Korea.
| | - Dong-Hyun Lee
- Department of Biological Sciences, College of Natural Sciences, Chonnam National University, Gwangju, Republic of Korea; Research Center of Ecomimetics, Chonnam National University, Gwangju, Republic of Korea; Institute of Sustainable Ecological Environment, Chonnam National Univesity, Gwangju, Republic of Korea.
| |
Collapse
|
20
|
Jiang J, Zhan L, Jiang B, Pan J, Hong C, Chen Z, Yang L. Anticancer therapy-induced peripheral neuropathy in solid tumors: diagnosis, mechanisms, and treatment strategies. Cancer Lett 2025; 620:217679. [PMID: 40154913 DOI: 10.1016/j.canlet.2025.217679] [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: 11/26/2024] [Revised: 03/21/2025] [Accepted: 03/25/2025] [Indexed: 04/01/2025]
Abstract
Anticancer therapy-induced peripheral neuropathy (PN) is a common adverse event during the diagnosis and treatment of solid tumors. The drug class, cumulative dose, and individual susceptibility affect the incidence and severity of PN. Owing to the lack of specific biomarkers and imaging tests, the diagnostic criteria for PN remain unclear. Moreover, the available and effective clinical treatment strategies are very limited, and most of the current drugs focus on symptom management rather than fundamental reversal of the disease course. The morbidity mechanisms of PN are diverse, including direct neurotoxicity, mitochondrial dysfunction, and disruption of axonal transport. Here, we summarize the diagnosis, mechanisms, and neuroprotective strategies of PN and discuss potential intervention treatments.
Collapse
Affiliation(s)
- Jiahong Jiang
- Cancer Center, Department of Medical Oncology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Luying Zhan
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Boyang Jiang
- The Clinical Medical College, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Jingyi Pan
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Chaojin Hong
- Cancer Center, Department of Medical Oncology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Zheling Chen
- Cancer Center, Department of Medical Oncology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China.
| | - Liu Yang
- Cancer Center, Department of Medical Oncology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China.
| |
Collapse
|
21
|
Poirier A, Utecht T, Villot R, Gélinas Y, Mouchiroud M, Kordahi M, Kolnohuz A, Pasteur C, Roy J, Beaulieu MJ, Orain M, Samson N, Blanchet MR, Joubert P, Laplante M. ZNF768 loss amplifies p53 action and reduces lung tumorigenesis in mice. Oncogene 2025:10.1038/s41388-025-03352-w. [PMID: 40133474 DOI: 10.1038/s41388-025-03352-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 02/17/2025] [Accepted: 03/11/2025] [Indexed: 03/27/2025]
Abstract
Cell proliferation is a fundamental process required for organismal development, growth, and maintenance. Failure to control this process leads to several diseases, including cancer. Zinc finger protein 768 (ZNF768) is an emerging transcription factor that plays key roles in driving proliferation. In addition to controlling a gene network supporting cell division, ZNF768 physically interacts and inhibits the activity of the tumor suppressor p53. Although the importance of ZNF768 in promoting cell proliferation has been well demonstrated in vitro, the physiological and pathological roles of ZNF768 in vivo are still unknown. Here, we report the generation and characterization of a ZNF768 null mouse model. ZNF768 null mice are viable but show a growth defect early in life. Mouse embryonic fibroblasts (MEFs) isolated from ZNF768 null embryos exhibit higher p53 levels, premature senescence, and higher sensitivity to genotoxic stress. In line with these findings, ZNF768 null mice showed increased radiosensitivity. This effect was associated not only with higher expression of a subset of p53 target genes, but also with alterations in genes regulating transmembrane receptor signaling, cell adhesion, and growth. Because ZNF768 levels are elevated in tumors, we tested the impact of ZNF768 loss on cancer development in mice. Here, we show that ZNF768 deletion was sufficient to repress lung tumor development in a KRASG12D-induced cancer mouse model. Overall, our findings establish ZNF768 as an important protein controlling cell proliferation that could potentially be targeted to reduce tumorigenesis.
Collapse
Affiliation(s)
- Audrey Poirier
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
- Centre de recherche sur le cancer de l'Université Laval, Université Laval, Québec, QC, Canada
| | - Timon Utecht
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
| | - Romain Villot
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
| | - Yves Gélinas
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
| | - Mathilde Mouchiroud
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
| | - Manal Kordahi
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
| | - Alona Kolnohuz
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
- Centre de recherche sur le cancer de l'Université Laval, Université Laval, Québec, QC, Canada
| | - Coline Pasteur
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
| | - Joanny Roy
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
| | - Marie-Josée Beaulieu
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
| | - Michèle Orain
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
| | - Nolwenn Samson
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
| | - Marie-Renée Blanchet
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
- Faculté de médecine, Université Laval, Québec, QC, Canada
| | - Philippe Joubert
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
- Centre de recherche sur le cancer de l'Université Laval, Université Laval, Québec, QC, Canada
- Faculté de médecine, Université Laval, Québec, QC, Canada
| | - Mathieu Laplante
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada.
- Centre de recherche sur le cancer de l'Université Laval, Université Laval, Québec, QC, Canada.
- Faculté de médecine, Université Laval, Québec, QC, Canada.
| |
Collapse
|
22
|
Ismail A, Hassan NW, Saudi MN, Abdel-Ghany YS, Labib HF, El-Deeb NM, El-Hawash SA. Exploring Thieno/Furo[2,3-b]pyridines as new scaffolds for potential FAK inhibition: Design, synthesis, biological evaluation and in silico studies. Bioorg Chem 2025; 159:108392. [PMID: 40139117 DOI: 10.1016/j.bioorg.2025.108392] [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: 01/03/2025] [Revised: 03/02/2025] [Accepted: 03/15/2025] [Indexed: 03/29/2025]
Abstract
Focal adhesion kinase (FAK) is a non-receptor tyrosine kinase that plays a vital role in regulating cancer cell survival, proliferation, migration, and angiogenesis. Aiming to explore new potent inhibitors, a series of thieno/furo[2,3-b]pyridine derivatives was designed and synthesized. The newly synthesized compounds were evaluated for their in vitro anti-proliferative activity against human liver (HUH-7), lung (A549) and breast (MCF-7) cancer cell lines, in addition to their cytotoxic activity against normal lung cell line (WI-38) to predict their safety profile. Seven compounds (4a, 4c, 5, 6, 10c, 11 and 12) displayed significant anti-proliferative activity as well as high selectivity towards the tested cancer cell lines (SI > 2). Among them, two compounds (4a and 4c) potently inhibited FAK enzyme with IC50 values of 54.96 and 50.98 nM, respectively. Flow cytometric cell cycle analysis indicated that compounds 4a and 4c caused cell cycle arrest at G1 phase. Compound 4c also exhibited an increase in the expression level of caspase-3 enzyme. Moreover, molecular docking study of the most promising compounds into FAK's active site was performed to elucidate their possible binding modes and to provide a structural basis for the further structural guidance design of FAK inhibitors.
Collapse
Affiliation(s)
- Azza Ismail
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Nayera W Hassan
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt.
| | - Manal N Saudi
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Yasser S Abdel-Ghany
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Hala F Labib
- Department of Pharmaceutical Chemistry, College of Pharmacy, Arab Academy of Science Technology and Maritime Transport, Alexandria, Egypt
| | - Nehal M El-Deeb
- Pharmaceutical Bioproducts Research Department, Genetic Engineering and Biotechnology Research Institute, City of Scientific Research and Technological Applications (SRTA-City), New Borg El-Arab City, 21934, Alexandria, Egypt.; Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, K1H 8 M5, Canada
| | - Soad A El-Hawash
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt.
| |
Collapse
|
23
|
Saif A, Islam MT, Raihan MO, Yousefi N, Rahman MA, Faridi H, Hasan AR, Hossain MM, Saleem RM, Albadrani GM, Al-Ghadi MQ, Ahasan Setu MA, Kamel M, Abdel-Daim MM, Aktaruzzaman M. Pan-cancer analysis of CDC7 in human tumors: Integrative multi-omics insights and discovery of novel marine-based inhibitors through machine learning and computational approaches. Comput Biol Med 2025; 190:110044. [PMID: 40120182 DOI: 10.1016/j.compbiomed.2025.110044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 03/14/2025] [Accepted: 03/16/2025] [Indexed: 03/25/2025]
Abstract
Cancer remains a significant global health challenge, with the Cell Division Cycle 7 (CDC7) protein emerging as a potential therapeutic target due to its critical role in tumor proliferation, survival, and resistance. However, a comprehensive analysis of CDC7 across multiple cancers is lacking, and existing therapeutic options have come with limited clinical success. The aim of this is to integrate a comprehensive pan-cancer analysis of CDC7 with the identification of novel marine-derived inhibitors, bridging the understanding of CDC7's role as a prognostic biomarker and therapeutic target across diverse cancer types. In this study, we conducted a pan-cancer analysis of CDC7 across 33 tumor types using publicly available datasets to evaluate its expression, genetic alterations, immune interactions, survival, and prognostic significance. Additionally, a marine-derived compound library of 31,492 molecules was screened to identify potential CDC7 inhibitors using chemoinformatics and machine learning. The top candidates underwent rigorous evaluations, including molecular docking, pharmacokinetics, toxicity, Density Functional Theory (DFT) calculations, and Molecular Dynamics (MD) simulations. The findings revealed that CDC7 is overexpressed in several cancers and is associated with poor survival outcomes and unfavorable prognosis. Enrichment analysis linked CDC7 to critical DNA replication pathways, while its role in modulating tumor-immune interactions highlighted its potential as a target for immunotherapy. Among all tested compounds, Tetrahydroaltersolanol D (CMNPD21999) exhibited the strongest binding affinity and stability, along with better drug-likeness and zero toxicity. These attributes highlight its potential as a promising drug candidate for CDC7 inhibition and future cancer treatment development. Furthermore, additional in vitro and in vivo studies are required to confirm the effectiveness of this drug candidate against the CDC7 protein.
Collapse
Affiliation(s)
- Ahmed Saif
- Department of Pharmacy, Faculty of Science, University of Rajshahi, Rajshahi, 6205, Bangladesh; Laboratory of Advanced Computational Biology, Biological Research on the Brain (BRB), Jashore, 7408, Bangladesh.
| | - Md Tarikul Islam
- Department of Genetic Engineering and Biotechnology, Faculty of Biological Science and Technology, Jashore University of Science and Technology, Jashore, 7408, Bangladesh; Laboratory of Advanced Computational Biology, Biological Research on the Brain (BRB), Jashore, 7408, Bangladesh.
| | - Md Obayed Raihan
- Laboratory of Advanced Computational Biology, Biological Research on the Brain (BRB), Jashore, 7408, Bangladesh; Department of Pharmaceutical Sciences, College of Health Sciences and Pharmacy, Chicago State University, Chicago, IL, USA.
| | - Niloofar Yousefi
- Department of Industrial Engineering and Management Systems, University of Central Florida, USA, Orlando, FL, USA
| | - Md Ajijur Rahman
- Department of Pharmacy, Faculty of Science, University of Rajshahi, Rajshahi, 6205, Bangladesh
| | - Hafeez Faridi
- Department of Pharmaceutical Sciences, College of Health Sciences and Pharmacy, Chicago State University, Chicago, IL, USA
| | - Al Riyad Hasan
- Laboratory of Advanced Computational Biology, Biological Research on the Brain (BRB), Jashore, 7408, Bangladesh; Department of Pharmacy, Faculty of Biological Science and Technology, Jashore University of Science and Technology, Jashore, 7408, Bangladesh
| | - Mirza Mahfuj Hossain
- Laboratory of Advanced Computational Biology, Biological Research on the Brain (BRB), Jashore, 7408, Bangladesh; Department of Computer Science and Engineering, Faculty of Engineering and Technology, Jashore University of Science and Technology, Jashore, 7408, Bangladesh
| | - Rasha Mohammed Saleem
- Department of Laboratory Medicine, Faculty of Applied Medical Sciences, Al-Baha University, Al-Baha, 65431, Saudi Arabia
| | - Ghadeer M Albadrani
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, 84428, Riyadh, 11671, Saudi Arabia
| | - Muath Q Al-Ghadi
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh, 11451, Saudi Arabia
| | - Md Ali Ahasan Setu
- Laboratory of Advanced Computational Biology, Biological Research on the Brain (BRB), Jashore, 7408, Bangladesh; Department of Microbiology, Faculty of Biological Science and Technology, Jashore University of Science and Technology, Jashore, 7408, Bangladesh
| | - Mohamed Kamel
- Department of Medicine and Infectious Diseases, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt
| | - Mohamed M Abdel-Daim
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, Jeddah, 21442, Saudi Arabia; Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, 41522, Egypt
| | - Md Aktaruzzaman
- Laboratory of Advanced Computational Biology, Biological Research on the Brain (BRB), Jashore, 7408, Bangladesh; Department of Pharmacy, Faculty of Biological Science and Technology, Jashore University of Science and Technology, Jashore, 7408, Bangladesh.
| |
Collapse
|
24
|
Zhang Z, Rao C, Hu M, Yan W, Du Z. Highly expressed GCN1 is associated with cancer progression and poor prognosis in hepatocellular carcinoma patients. Cancer Cell Int 2025; 25:107. [PMID: 40114124 PMCID: PMC11927180 DOI: 10.1186/s12935-025-03732-y] [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: 07/21/2024] [Accepted: 03/06/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND General control non-derepressible protein 1 (GCN1), a ribosome-binding protein, has been implicated in the development and progression of multiple cancers. However, the potential role of GCN1 in hepatocellular carcinoma (HCC) has not yet been investigated. METHODS The expression of GCN1 in HCC was analyzed using multiple databases. Bioinformatics analysis was employed to investigate the correlation of GCN1 expression with clinical significance and immune infiltration in HCC. Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis, gene set enrichment analysis (GSEA), and in vitro experiments were conducted to study the function and potential mechanisms of GCN1 in HCC. RESULTS GCN1 was significantly upregulated in HCC, which was associated with worse clinicopathological features and poorer prognosis of the patients. GCN1 expression was closely associated with immune cell infiltration in HCC. GSEA analysis showed that GCN1 was involved in several tumor-related signaling pathways, including cell cycle, DNA replication, and Wnt signaling pathway. Knockdown of GCN1 inhibited the proliferation, invasion and migration of HCC cells, and also down-regulated the expression levels of cell cycle protein cyclin B1 (CCNB1), cyclin D1 (CCND1), and Wnt signaling pathway-related proteins Wnt3A and β-catenin. CONCLUSION GCN1 overexpression was associated with HCC progression and poor prognosis, and GCN1 knockdown could suppress the proliferation, migration and invasion ability of HCC cells by regulating Wnt signaling pathway, suggesting the potential of GCN1 as a prognostic and therapeutic target for HCC.
Collapse
Affiliation(s)
- Zhongchao Zhang
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, Hubei, 430000, China
| | - Caijun Rao
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mingcun Hu
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Yan
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, Hubei, 430000, China.
| | - Zhipeng Du
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, Hubei, 430000, China.
| |
Collapse
|
25
|
Chandrasekaran J, Gopal D, Sureshkumar LV, Santhiyagu IX, Senthil Kumar V, Munuswamy B, Gani BFHMY, Agrawal M. Integrated AI and machine learning pipeline identifies novel WEE1 kinase inhibitors for targeted cancer therapy. Mol Divers 2025:10.1007/s11030-025-11157-y. [PMID: 40106128 DOI: 10.1007/s11030-025-11157-y] [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: 01/28/2025] [Accepted: 03/04/2025] [Indexed: 03/22/2025]
Abstract
The dysregulation of the cell cycle in cancer underscores the therapeutic potential of targeting WEE1 kinase, a key regulator of the G2/M checkpoint. This study harnessed artificial intelligence (AI)-driven methodologies, particularly the MORLD platform, to identify novel WEE1 inhibitors. Starting with clinically validated WEE1 inhibitors as references, we generated 20,000 structurally diverse compounds optimized for binding affinity, synthetic accessibility, and drug-likeness. A rigorous cheminformatics pipeline-comprising PAINS filtering, physicochemical property assessments, and molecular fingerprinting-refined this library to 242 promising candidates. Dimensionality reduction using UMAP and clustering via K-means enabled the prioritization of structurally unique leads. Molecular docking studies highlighted two compounds, MORLD5036 and MORLD6305, with exceptional binding affinities and interactions with key WEE1 active site residues. Molecular dynamics simulations and MM-GBSA binding free energy calculations further validated MORLD5036 as the most stable and potent inhibitor. Scaffold analysis revealed novel chemotypes distinct from existing inhibitors, enhancing potential for intellectual property. Comprehensive ADME profiling confirmed favorable pharmacokinetics, while synthetic accessibility evaluations indicated practicality for experimental validation. The identified lead compound, MORLD5036, exhibits favorable pharmacokinetics and novel chemotypes, positioning it as a potential therapeutic candidate for cancers reliant on WEE1-mediated cell cycle control. This integrated, AI-driven pipeline expedites the identification of next-generation WEE1 inhibitors, paving the way for advancements in precision oncology. Unlike traditional methods reliant on pre-existing datasets, this study leverages MORLD's reinforcement learning framework to autonomously generate inhibitors, enabling exploration of uncharted chemical space. These findings establish MORLD5036 as a computationally promising WEE1 inhibitor candidate warranting further experimental validation.
Collapse
Affiliation(s)
- Jaikanth Chandrasekaran
- Department of Pharmacology, Sri Ramachandra Faculty of Pharmacy, Sri Ramachandra Institute of Higher Education and Research (Deemed to be University), Chennai, Tamil Nadu, 600116, India.
| | - Dhanushya Gopal
- Department of Pharmacology, Sri Ramachandra Faculty of Pharmacy, Sri Ramachandra Institute of Higher Education and Research (Deemed to be University), Chennai, Tamil Nadu, 600116, India
| | - Lokesh Vishwa Sureshkumar
- Department of Pharmacology, Sri Ramachandra Faculty of Pharmacy, Sri Ramachandra Institute of Higher Education and Research (Deemed to be University), Chennai, Tamil Nadu, 600116, India
| | - Infant Xavier Santhiyagu
- Department of Pharmacology, Sri Ramachandra Faculty of Pharmacy, Sri Ramachandra Institute of Higher Education and Research (Deemed to be University), Chennai, Tamil Nadu, 600116, India
| | - Varsha Senthil Kumar
- Department of Pharmacology, Sri Ramachandra Faculty of Pharmacy, Sri Ramachandra Institute of Higher Education and Research (Deemed to be University), Chennai, Tamil Nadu, 600116, India
| | - Bhuvaneshwari Munuswamy
- Department of Pharmacology, Sri Ramachandra Faculty of Pharmacy, Sri Ramachandra Institute of Higher Education and Research (Deemed to be University), Chennai, Tamil Nadu, 600116, India
| | - Beevi Fathima Harshatha Mohamed Yousuf Gani
- Department of Pharmacology, Sri Ramachandra Faculty of Pharmacy, Sri Ramachandra Institute of Higher Education and Research (Deemed to be University), Chennai, Tamil Nadu, 600116, India
| | - Mohit Agrawal
- Department of Pharmacology, School of Medical and Allied Sciences, K.R. Mangalam University, Gurugram, Haryana, India
| |
Collapse
|
26
|
Cui J, Ruan S, Zhang Z, Wang H, Yan Q, Chen Y, Yang J, Fang J, Wu Q, Chen S, Huang S, Zhang C, Hou B. Pan-cancer analysis of the prognosis and immune infiltration of NSUN7 and its potential function in renal clear cell carcinoma. Discov Oncol 2025; 16:345. [PMID: 40100478 PMCID: PMC11920486 DOI: 10.1007/s12672-025-02061-w] [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: 10/18/2024] [Accepted: 03/05/2025] [Indexed: 03/20/2025] Open
Abstract
BACKGROUND NSUN7, an enzyme responsible for the RNA m5c modification, has been recognized as a valuable indicator for predicting and diagnosing an array of cancer. Nevertheless, there is still a scarcity of thorough analyses exploring its diagnostic, predictive, and immune system-related importance in various types of cancer. METHODS We integrated multiple publicly available databases, including TCGA, TISIDB, TISCH2, and UALCAN, to comprehensively investigate the role of NSUN7 in pan-cancer across various omics data types. The research included examining survival rates, genetic mutations, immune cell presence in tumors, analyzing differences in gene expression, and studying individual cells, among other things. RESULTS NSUN7 expression showed an increase across 12 cancer types and a decrease in another 12 types. NSUN7 was discovered to be linked with enhanced survival rates in bladder urothelial carcinoma (BLCA), kidney renal clear cell carcinoma (KIRC), kidney renal papillary cell carcinoma (KIRP), lung adenocarcinoma (LUAD), pheochromocytoma and paraganglioma (PCPG), skin cutaneous melanoma (SKCM), and uveal melanoma (UVM).On the other hand, NSUN7 seemed to have a detrimental impact on the prognosis of glioblastoma multiforme/brain lower grade glioma (GBMLGG), adrenocortical carcinoma (ACC),acute myeloid leukemia (LAML), stomach adenocarcinoma (STAD), and brain lower grade glioma (LGG). Furthermore, our experimental validation confirmed the inhibitory effect of NSUN7 on proliferation of renal clear cell carcinoma while elucidating its specific part in blocking cell cycle progression. CONCLUSIONS The findings underscore the potential utility of NSUN7 as a valuable prognostic indicator for patients and offer insights into the mechanisms underlying cancer initiation and progression.
Collapse
Affiliation(s)
- Jinwei Cui
- South China University of Technology School of Medicine, Guangzhou, 51000, China
- Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Guangzhou, 510080, China
| | - Shiye Ruan
- Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Guangzhou, 510080, China
| | - Zhongyan Zhang
- Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Guangzhou, 510080, China
| | - Hailiang Wang
- Department of Hepatobiliary Surgery, Weihai Central Hospital, Qingdao University, Weihai, 264400, China
| | - Qian Yan
- Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Guangzhou, 510080, China
| | - Yubin Chen
- Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Guangzhou, 510080, China
| | - Jiayu Yang
- Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Guangzhou, 510080, China
| | - Jike Fang
- Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Guangzhou, 510080, China
| | - Qianlong Wu
- Department of General Surgery, Heyuan People's Hospital, Heyuan, 517000, China
- Heyuan Key Laboratory of Molecular Diagnosis and Disease Prevention and Treatment, Heyuan People's Hospital, Heyuan, 517000, China
| | - Sheng Chen
- South China University of Technology School of Medicine, Guangzhou, 51000, China
- Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Guangzhou, 510080, China
| | - Shanzhou Huang
- South China University of Technology School of Medicine, Guangzhou, 51000, China.
- Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Guangzhou, 510080, China.
| | - Chuanzhao Zhang
- South China University of Technology School of Medicine, Guangzhou, 51000, China.
- Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Guangzhou, 510080, China.
| | - Baohua Hou
- South China University of Technology School of Medicine, Guangzhou, 51000, China.
- Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Guangzhou, 510080, China.
| |
Collapse
|
27
|
Cui B, Tu S, Li H, Zeng Z, Xiao R, Guo J, Liang X, Liu C, Pan L, Chen W, Ge M, Zhong X, Ye L, Chen H, Zhang Q, Xu Y. METTL3 knockout accelerates hepatocarcinogenesis via inhibiting endoplasmic reticulum stress response. FEBS Open Bio 2025. [PMID: 40103332 DOI: 10.1002/2211-5463.70023] [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: 09/21/2024] [Revised: 02/24/2025] [Accepted: 03/06/2025] [Indexed: 03/20/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is among the most common causes of cancer-related deaths worldwide. Previous studies showed that N6-methyladenosine (m6A), the most abundant chemical modification in eukaryotic RNAs, is implicated in HCC progression. Using liver-specific conditional knockout mice, we found that the loss of METTL3, the core catalytic subunit of m6A methyltransferase, significantly promoted hepatic tumor initiation under various oncogenic challenges, contrary to the previously reported oncogenic role of METTL3 in liver cancer cell lines or xenograft models. Mechanistically, we hypothesized that METTL3 deficiency accelerated HCC initiation by inhibiting m6A deposition on MANF transcripts, impairing nuclear export and thus MANF protein levels, which led to insufficient endoplasmic reticulum (ER) stress response pathway activation. Our findings suggest a tumor-suppressive role for METTL3 in the early stages of HCC, emphasizing the importance of understanding the dynamic role of epigenetic regulation in tumorigenesis and targeted therapy.
Collapse
Affiliation(s)
- Bo Cui
- Biotherapy Centre, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Silin Tu
- Biotherapy Centre, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Haibo Li
- Department of Hepatic Surgery and Liver Transplantation Center, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zhancheng Zeng
- Biotherapy Centre, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ruiqi Xiao
- Biotherapy Centre, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jing Guo
- Biotherapy Centre, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaoqi Liang
- Cell-Gene Therapy Translational Medicine Research Centre, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Chang Liu
- Biotherapy Centre, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Lijie Pan
- Laboratory Animal Center, Sun Yat-sen University, Guangzhou, China
| | - Wenjie Chen
- Biotherapy Centre, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Cell-Gene Therapy Translational Medicine Research Centre, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Liver Disease Research, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Mian Ge
- Department of Anesthesiology, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaofen Zhong
- Biotherapy Centre, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Linsen Ye
- Department of Hepatic Surgery and Liver Transplantation Center, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Liver Disease Research, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Huaxin Chen
- Biotherapy Centre, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qi Zhang
- Biotherapy Centre, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Cell-Gene Therapy Translational Medicine Research Centre, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Liver Disease Research, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yan Xu
- Biotherapy Centre, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
28
|
Choe H, Wang Z, Huang J, Yang Y, Zhao Z, Jo H, Pak H, Ali T, Ding K, Ma J, Li L, Shang D, Zhang L. Broussoflavonol B induces S-phase arrest and apoptosis in pancreatic cancer cells by modulating the cell cycle checkpoint through inhibition of the AURKA/PLK1 pathway. Cancer Cell Int 2025; 25:100. [PMID: 40098146 PMCID: PMC11916852 DOI: 10.1186/s12935-025-03717-x] [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: 11/10/2024] [Accepted: 02/25/2025] [Indexed: 03/19/2025] Open
Abstract
BACKGROUND Broussoflavonol B (Bf-B), a flavonoid compound identified in the roots of Daphne giraldii Nitsche, has been extensively investigated for its potential anti-inflammatory, antioxidant, and anticancer properties. However, the precise mechanism underlying the regulation of AURKA/PLK1 pathway-mediated cell cycle arrest by Bf-B in pancreatic cancer remains poorly understood. PURPOSE The objective of this study was to investigate the inhibitory effect of Bf-B on pancreatic ductal adenocarcinoma (PDAC) and its underlying mechanism. METHODS A CCK8 assay was conducted to identify the flavonoids with the highest inhibitory activity against PANC-1, the pancreatic cancer cell line among the 25 flavonoids. Through bioinformatics analysis and molecular docking, the pathogenic targets of pancreatic cancer and flavonoid-related targets were explored, and the key targets and signaling pathways of drug intervention in pancreatic cancer were analyzed. The viability and migration ability of pancreatic cancer cells were assessed following treatment with Bf-B via the CCK8, colony formation, and wound healing assays. The cell cycle distribution and cell apoptosis were analyzed through flow cytometry and Hoechst staining. Western blotting and qPCR were employed to investigate the expression of relevant proteins and genes. For in vivo experiments, we employed a xenograft mouse model to evaluate the anticancer efficacy of Bf-B. Immunohistochemistry and immunofluorescence assays were employed to investigate the expression of relevant proteins. RESULTS In this study, the structure‒activity relationships of 25 flavonoids were evaluated. The results demonstrated that Bf-B with diisopentenyl has potent cytotoxic effects on PANC-1 cancer cells. AURKA, PLK1, and MET might serve as key targets for Bf-B inhibition of disease progression in PDAC patients. The results demonstrated that Bf-B inhibits the proliferation and migration of PANC-1 and BXPC-3 cells and induces cell cycle S-phase arrest, apoptosis, and DNA damage. Moreover, the results of western blot and qPCR experiments indicated that Bf-B exerts anticancer effects by downregulating the expression of the genes encoding AURKA/PLK1, the cell cycle checkpoint kinase ATR/CHK1/CDC25C, and Cyclin B1/CDK1 signaling pathway-related proteins and upregulating the expression of PP53, P21, and histone H2A. XS139ph expression. In xenograft-bearing mice, AURKA/PLK1 expression was reduced in a dose-dependent manner, accompanied by an increase in histone H2A. XS139ph expression. CONCLUSION Bf-B might be a potent therapeutic agent for pancreatic cancer because of its ability to suppress the expression of AURKA/PLK1.
Collapse
Affiliation(s)
- HyokChol Choe
- Institute of Integrative Medicine, Dalian Medical University, Dalian, 0411, China
- Department of Clinical Medicine, Sinuiju Medical University, Sinuiju, Korea
| | - Zhen Wang
- Institute of Integrative Medicine, Dalian Medical University, Dalian, 0411, China
- Department of General Surgery, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jianhua Huang
- Institute of Integrative Medicine, Dalian Medical University, Dalian, 0411, China
| | - Yutong Yang
- Institute of Integrative Medicine, Dalian Medical University, Dalian, 0411, China
| | - Zhihao Zhao
- Institute of Integrative Medicine, Dalian Medical University, Dalian, 0411, China
| | - HyonSu Jo
- Pyongyang University of Medical Sciences, Pyongyang, Korea
| | - HyonU Pak
- Pyongyang University of Medical Sciences, Pyongyang, Korea
| | - Tanveer Ali
- Institute of Integrative Medicine, Dalian Medical University, Dalian, 0411, China
| | - Kaiyue Ding
- Institute of Integrative Medicine, Dalian Medical University, Dalian, 0411, China
| | - Junnan Ma
- Institute of Integrative Medicine, Dalian Medical University, Dalian, 0411, China
| | - Lingzhi Li
- The College of Traditional Chinese Medicine, Shenyang Pharmaceutical University, Shenyang, China
| | - Dong Shang
- Institute of Integrative Medicine, Dalian Medical University, Dalian, 0411, China.
- Department of General Surgery, First Affiliated Hospital of Dalian Medical University, Dalian, China.
| | - Lin Zhang
- Institute of Integrative Medicine, Dalian Medical University, Dalian, 0411, China.
| |
Collapse
|
29
|
Attaie Y, Storey KB. MicroRNA-mediated regulation in anoxic Lithobates sylvaticus liver. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2025; 55:101487. [PMID: 40106882 DOI: 10.1016/j.cbd.2025.101487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 03/11/2025] [Accepted: 03/16/2025] [Indexed: 03/22/2025]
Abstract
The wood frog, Lithobates sylvaticus (formerly Rana sylvatica), endures various physiological stressors including freezing, anoxia, dehydration, and hyperglycemia during winter, relying on metabolic stress response to survive. microRNA (miRNA)-mediated gene regulation is a well-documented stress response, however, its role during anoxia in L. sylvaticus liver remains underexplored. This research identifies seven miRNAs with significant differential expression during anoxia. Bioinformatic analyses predicted downregulation of monounsaturated fatty acid biosynthesis and membrane fluidity pathways, alongside upregulation of insulin signalling pathways (PI3K/Akt, mTOR). These results suggest that miRNAs contribute to gene regulation in L. sylvaticus during anoxia.
Collapse
Affiliation(s)
- Yasser Attaie
- Department of Biology and Institute of Biochemistry, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario K1S 5B6, Canada.
| | - Kenneth B Storey
- Department of Biology and Institute of Biochemistry, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario K1S 5B6, Canada
| |
Collapse
|
30
|
Kale Bakir E, Deveci Ozkan A, Erman G, Isik S, Yuzugullu Karakus Y. Anticancer potential of purified laccase enzyme from Trametes versicolor: specific cytotoxicity against thyroid and endometrial cancer cells. Mol Biol Rep 2025; 52:312. [PMID: 40085415 DOI: 10.1007/s11033-025-10416-3] [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: 01/27/2025] [Accepted: 03/05/2025] [Indexed: 03/16/2025]
Abstract
BACKGROUND Cancer is one of the leading causes of death worldwide, highlighting the need to develop novel therapeutic strategies that are more effective and have fewer side effects than conventional treatments. Enzymatic cancer therapy is a promising approach due to its high specificity and minimal toxicity. Among the various enzymes, laccase, a widely used biocatalyst, has shown significant potential for anti-cancer applications due to its proliferation inhibitory properties. METHODS AND RESULTS In this study, the enzyme laccase from Trametes versicolor was purified by three-phase partitioning and then its cytotoxic, genotoxic and apoptotic effects on thyroid cancer (TT) and endometrial cancer (Ishikawa) cell lines were investigated. Laccase exhibited IC50 values of 88.63 µM in TT cells and 1.68 µM in Ishikawa cells. The enzyme triggered apoptosis in Ishikawa cells and induced cell cycle arrest in S phase, while significantly increasing DNA damage in both cancer cell lines. Treatment with laccase led to downregulation of the anti-apoptotic gene Bcl-2 and upregulation of the pro-apoptotic gene Bax and the DNA damage repair genes Rad51 and ATM. CONCLUSIONS Our results emphasize the specific cytotoxicity and molecular mechanisms underlying the anti-cancer effect of laccase and demonstrate that laccase is capable of selectively targeting cancer cells and causing apoptosis and DNA damage. This study demonstrates the potential of laccase as a novel enzymatic therapeutic for the treatment of thyroid and endometrial cancer and warrants further investigation into its clinical application and efficacy.
Collapse
Affiliation(s)
- Elif Kale Bakir
- Department of Biology, Faculty of Arts and Sciences, Kocaeli University, Kocaeli, 41001, Turkey
| | - Asuman Deveci Ozkan
- Department of Medical Biology, Faculty of Medicine, Sakarya University, Sakarya, 54050, Turkey
| | - Gulay Erman
- Health Services Education Research and Application Centre, Sakarya University, Sakarya, 54050, Turkey
- Department of Medical Biochemistry, Institute of Health Science, Sakarya University, Sakarya, 54050, Turkey
| | - Semih Isik
- Department of Biology, Institute of Natural and Applied Sciences, Kocaeli University, Kocaeli, 41001, Turkey
| | - Yonca Yuzugullu Karakus
- Department of Biology, Faculty of Arts and Sciences, Kocaeli University, Kocaeli, 41001, Turkey.
| |
Collapse
|
31
|
Wu P, Wen Z. ATM is associated with the prognosis of colorectal cancer: a systematic review. Front Oncol 2025; 15:1470939. [PMID: 40144209 PMCID: PMC11936800 DOI: 10.3389/fonc.2025.1470939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 02/12/2025] [Indexed: 03/28/2025] Open
Abstract
Objective Chemosensitivity and radiosensitivity are associated with the prognosis of colorectal cancer, and the expression of the ataxia-telangiectasia mutated (ATM) protein plays an essential role in these processes. The present study examined the relationship between ATM expression and the survival outcomes of colorectal cancer patients and explored the underlying mechanism and promising therapeutic strategies. Method A search including medical subject headings (MeSH), free terms, and combined words was conducted using Pubmed, EMBASE, and Cochrane. Studies had to meet the inclusion criteria as well as include processes such as data extraction and quality evaluation. The survival outcomes were assessed using hazard ratio (HR) and 95% confidence interval (CI). Heterogeneity, and publication bias were analyzed, and a P value <0.05 was considered statistically significant. Results Nine studies with 2883 patients were included in the meta-analysis. Low ATM expression level was related to poor overall survival (HR=0.542, 95% CI=0.447-0.637; P=0.000). Disease-free, progression-free, and recurrence-free survival rates were lower in patients with low ATM expression than in those with high ATM expression. There was no significant difference between Stage I-II and Stage III-IV colorectal cancer patients [risk ratio (RR)=1.173, 95% CI=0.970-1.417, P=0.690]. Conclusions Low ATM expression level may be a marker of poor survival in colorectal cancer and contributes to resistance to therapy. Targeting related factors in these pathways to sensitize tumors to treatment is a potential therapeutic strategy, and monitoring ATM status could be a valuable guide independent of the immunotherapy or chemotherapy strategy used.
Collapse
Affiliation(s)
- Pei Wu
- Department of Gastrointestinal Surgery, Yongchuan Hospital of Chongqing Medical University, Chongqing, China
| | - Zelin Wen
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
32
|
Zhang L, Zhuge Y, Ni J. BUB1 serves as a biomarker for poor prognosis in liver hepatocellular carcinoma. BMC Immunol 2025; 26:20. [PMID: 40069598 PMCID: PMC11895216 DOI: 10.1186/s12865-025-00698-4] [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: 12/01/2024] [Accepted: 02/27/2025] [Indexed: 03/15/2025] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is the most frequent kind of liver cancer with high morbidity and mortality rates worldwide. Altered expression of BUB1 (budding uninhibited by benzimidazole 1) gene leads to chromosome instability and aneuploidy. This study investigated the expression of BUB1 and its prognostic value as well as its correlation with immune cell infiltration and immune checkpoints in HCC. RESULTS Using the Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA) databases, we found that BUB1 was up-regulated in HCC, thus prompting us to validate this observation by immunohistochemistry on 57 HCC paraffin embedded tissues from Wuxi No.2 People's Hospital. Kaplan-Meier survival analysis revealed that HCC patients with high BUB1 expression had shorter overall survival (OS) time as well as progression-free interval (PFI), and disease-specific survival (DSS) time compared to the patients with low BUB1 expression. Besides, STRING database showed that the top 10 co-expression genes were mainly involved in the regulation of cell division during the mitosis. Gene Ontology (GO) analysis and the Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis showed that BUB1 had a connection to cancer related pathways. Lastly, The Tumor Immune Estimation Resource (TIMER) analysis found that BUB1 was positively related to immune cell infiltration and some immune checkpoint gene in HCC. CLINICAL TRIAL NUMBER Not applicable. CONCLUSIONS Our present study demonstrated that BUB1 is a potential prognostic biomarker, and BUB1 may play a role in the tumor immune microenvironment in HCC.
Collapse
Affiliation(s)
- Lili Zhang
- Department of Gastroenterology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, Jiangsu Province, China.
- Department of Gastroenterology, Jiangnan University Medical Center, Wuxi No.2 People's Hospital, Wuxi, Jiangsu Province, China.
| | - Yuzheng Zhuge
- Department of Gastroenterology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, Jiangsu Province, China.
| | - Jingbin Ni
- Department of Gastroenterology, Jiangnan University Medical Center, Wuxi No.2 People's Hospital, Wuxi, Jiangsu Province, China
| |
Collapse
|
33
|
Diao X, Qi G, Li X, Tian Y, Li J, Jin Y. Label-Free Exosomal SERS Detection Assisted by Machine Learning for Accurately Discriminating Cell Cycle Stages and Revealing the Molecular Mechanisms during the Mitotic Process. Anal Chem 2025; 97:5093-5101. [PMID: 39999424 DOI: 10.1021/acs.analchem.4c06240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2025]
Abstract
Cell cycle analysis is crucial for disease diagnosis and treatment, especially for investigating cell heterogeneity and regulating cell behaviors. Exosomes are highly appealing as noninvasive biomarkers for monitoring real-time changes in the cell cycle due to their abundant molecular information inherited from their metrocyte cells and reflecting the state of these cells to some extent. However, to our knowledge, the relationship between exosomes and the cell cycle has not been reported. Herein, we successfully monitored the variation of exosomal surface-enhanced Raman spectroscopy (SERS) spectra to discriminate different cell cycle stages (G0/G1, S, and G2/M phases) based on label-free surface-enhanced Raman spectroscopy (SERS) combined with the machine learning method of linear discriminant analysis (LDA). An average accuracy of 85% based on the trained SERS spectra of exosomes from different cell cycle stages confirmed the high reliability of the support vector machine (SVM) algorithm for analyzing dynamic changes in the cell cycle at different time points. Importantly, the related molecular mechanisms among mitotic processes (prometaphase, metaphase, and anaphase/telophase) and unique biomolecular events between cancerous (HeLa) and normal (H8) cells were also revealed by the present label-free SERS detection platform. Based on SERS analysis, the content of phenylalanine (Phe) within HeLa cells increased, and some structures of proteins containing Phe and tryptophan (Trp) residues may be transformed during the mitotic process. Notably, the α-helix and β-sheet of proteins coexisted in HeLa cells; meanwhile, the α-helix of the proteins was more dominant in H8 cells than in HeLa cells. The strategy is effective for discriminating cell cycle stages and elucidating the associated molecular events during the cell mitotic process and will provide potential application value for guiding the cell cycle treatment strategies of cancer in the future.
Collapse
Affiliation(s)
- Xingkang Diao
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, P. R. China
| | - GuoHua Qi
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
- Guangdong Key Laboratory of Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, P. R. China
| | - Xinli Li
- College of Instrumentation and Electrical Engineering, Jilin University, Changchun 130061, P. R. China
| | - Yu Tian
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
| | - Jing Li
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, P. R. China
| | - Yongdong Jin
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
- Guangdong Key Laboratory of Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, P. R. China
| |
Collapse
|
34
|
Ippen FM, Hielscher T, Friedel D, Göbel K, Reuss D, Herold-Mende C, Krieg S, Deimling AV, Wick W, Sahm F, Suwala AK. The prognostic impact of CDKN2A/B hemizygous deletions in IDH-mutant glioma. Neuro Oncol 2025; 27:743-754. [PMID: 39530475 PMCID: PMC11889711 DOI: 10.1093/neuonc/noae238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Homozygous deletions of CDKN2A/B are known to predict poor prognosis in gliomas, but the impact of hemizygous deletions is less clear. This study aimed to evaluate the prognostic significance of hemizygous CDKN2A/B deletions in IDH-mutant low-grade astrocytomas and oligodendrogliomas. METHODS Tissue samples diagnosed as astrocytoma, IDH-mutant and oligodendroglioma, IDH-mutant, 1p/19q co-deleted CNS WHO grade 2 and 3 were collected from the archives of the Institute of Neuropathology in Heidelberg. DNA methylation analysis was performed on formalin-fixed paraffin-embedded samples. Evaluation of the CDKN2A/B locus was performed by visual inspection of copy-number plots derived from methylation-array data for each case. Hemizygous and homozygous losses were assessed in relation to whole chromosomal or larger segmental losses and gains in the chromosomal profile. Survival probabilities were assessed using the Kaplan-Meier method. RESULTS A total of 334 low-grade glioma cases were identified, including 173 astrocytomas and 161 oligodendrogliomas. Hemizygous deletions in CDKN2A/B (37/173 in astrocytomas, 15/161 in oligodendrogliomas) did not confer significantly worse survival outcomes compared to CDKN2A/B wild-type cases in neither low-grade astrocytoma (log-rank P = .2556; HR 2.29, 95% CI [0.76; 6.40], P = .135) nor oligodendroglioma (log-rank P = .2760; HR 0.17; 95% CI [0.01; 5.05]; P = .305), regardless of CNS WHO grade, which was further demonstrated on a subgroup of astrocytoma, IDH mutant CNS WHO 4 cases (log-rank P = .1680; HR 4.55, 95% CI [0.88; 24.51], P = .0689). CONCLUSIONS Hemizygous CDKN2A/B deletions do not significantly worsen OS or progression-free survival in IDH-mutant astrocytomas and oligodendrogliomas, CNS WHO grades 2 and 3.
Collapse
Affiliation(s)
- Franziska M Ippen
- National Center for Tumor Diseases (NCT), NCT Heidelberg, A Partnership Between DKFZ and University Hospital Heidelberg, Heidelberg, Germany
- Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
- Department of Neurology, University Hospital Heidelberg, Heidelberg, Germany
| | - Thomas Hielscher
- Department of Biostatistics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Dennis Friedel
- Faculty of Bioscience, Heidelberg University, Heidelberg, Germany
- Department of Neuropathology, University Hospital Heidelberg, Heidelberg, Germany
- Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
| | - Kirsten Göbel
- Department of Neuropathology, University Hospital Heidelberg, Heidelberg, Germany
- Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
| | - David Reuss
- Department of Neuropathology, University Hospital Heidelberg, Heidelberg, Germany
- Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
| | - Christel Herold-Mende
- Department of Neurosurgery, University Hospital Heidelberg, Heidelberg, Germany
- Department of Neurosurgery, Division of Experimental Neurosurgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Sandro Krieg
- Department of Neurosurgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Andreas v Deimling
- Department of Neuropathology, University Hospital Heidelberg, Heidelberg, Germany
- Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
| | - Wolfgang Wick
- Clinical Cooperation Unit Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neurology, University Hospital Heidelberg, Heidelberg, Germany
| | - Felix Sahm
- Department of Neuropathology, University Hospital Heidelberg, Heidelberg, Germany
- Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
| | - Abigail K Suwala
- Department of Neuropathology, University Hospital Heidelberg, Heidelberg, Germany
- Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
| |
Collapse
|
35
|
Rust K, Schubert A, Peralta JM, Nystul TG. Independent signaling pathways provide a fail-safe mechanism to prevent tumorigenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.28.640798. [PMID: 40093137 PMCID: PMC11908167 DOI: 10.1101/2025.02.28.640798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Controlled signaling activity is vital for normal tissue homeostasis and oncogenic signaling activation facilitates tumorigenesis. Here we use single-cell transcriptomics to investigate the effects of pro-proliferative signaling on epithelial homeostasis using the Drosophila follicle cell lineage. Notably, EGFR-Ras overactivation induces cell cycle defects by activating the transcription factors Pointed and E2f1 and impedes differentiation. Hh signaling simultaneously promotes an undifferentiated state and induces differentiation via activation of EMT-associated transcription factors zfh1 and Mef2. As a result, overactivation of Hh signaling generates a transcriptional hybrid state comparable to epithelial-mesenchymal-transition. Co-overactivation of Hh signaling with EGFR-Ras signaling blocks differentiation and induces key characteristics of tumor cells including a loss of tissue architecture caused by reduced expression of cell adhesion molecules, sustained proliferation and an evasion of cell cycle checkpoints. These findings provide new insight into how non-interacting signaling pathways converge at the transcriptional level to prevent malignant cell behavior.
Collapse
Affiliation(s)
- Katja Rust
- Institute of Physiology and Pathophysiology, Dept. of Molecular Cell Physiology, Philipps University Marburg, Germany
| | - Andrea Schubert
- Institute of Physiology and Pathophysiology, Dept. of Molecular Cell Physiology, Philipps University Marburg, Germany
| | - Jobelle M Peralta
- UCSF, Department of Anatomy, 513 Parnassus Ave, San Francisco, CA 94143, USA
- UCSF, Department of OB-GYN/RS, 513 Parnassus Ave, San Francisco, CA 94143, USA
- Broad Center of Regeneration Medicine and Stem Cell Research, 513 Parnassus Ave, San Francisco, CA 94143, USA
| | - Todd G Nystul
- UCSF, Department of Anatomy, 513 Parnassus Ave, San Francisco, CA 94143, USA
- UCSF, Department of OB-GYN/RS, 513 Parnassus Ave, San Francisco, CA 94143, USA
- Broad Center of Regeneration Medicine and Stem Cell Research, 513 Parnassus Ave, San Francisco, CA 94143, USA
| |
Collapse
|
36
|
Mishan MA, Choo YM, Winkler J, Hamann MT, Karan D. Manzamine A: A promising marine-derived cancer therapeutic for multi-targeted interactions with E2F8, SIX1, AR, GSK-3β, and V-ATPase - A systematic review. Eur J Pharmacol 2025; 990:177295. [PMID: 39863145 DOI: 10.1016/j.ejphar.2025.177295] [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: 08/12/2024] [Revised: 01/21/2025] [Accepted: 01/22/2025] [Indexed: 01/27/2025]
Abstract
Manzamine A, a natural compound derived from various sponge genera, features a β-carboline structure and exhibits a range of biological activities, including anti-inflammatory and antimalarial effects. Its potential as an anticancer agent has been explored in several tumor models, both in vitro and in vivo, showing effects through mechanisms such as cytotoxicity, regulation of the cell cycle, inhibition of cell migration, epithelial-to-mesenchymal transition (EMT), autophagy, and apoptosis through multi-target interactions of E2F transcriptional factors, ribosomal S6 kinases, androgen receptor (AR), SIX1, GSK-3β, v-ATPase, and p53/p21/p27 cascades. This systematic review evaluates existing literature on the potential application of this marine alkaloid as a novel cancer therapy, highlighting its promising ability to inhibit cancer cell growth while causing minimal side effects.
Collapse
Affiliation(s)
- Mohammad Amir Mishan
- Department of Urology, Brown Cancer Center, 505 S Hancock Street, Louisville, KY, USA
| | - Yeun-Mun Choo
- Department of Chemistry, Faculty of Science, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Jeffery Winkler
- Department of Chemistry, The University of Pennsylvania, Philadelphia, PA, USA
| | - Mark T Hamann
- Department of Drug Discovery and Biomedical Sciences and Public Health, Colleges of Pharmacy and Medicine, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Dev Karan
- Department of Urology, Brown Cancer Center, 505 S Hancock Street, Louisville, KY, USA.
| |
Collapse
|
37
|
Fan Y, Wang Q, Zhang Y, Wang Y, Li W, Jiang S, Duan JN. Mechanism of Guishao Yigong decoction in treating colorectal cancer based on network pharmacology and experimental validation. J Pharm Pharmacol 2025; 77:430-445. [PMID: 39352002 DOI: 10.1093/jpp/rgae045] [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: 11/17/2023] [Accepted: 04/05/2024] [Indexed: 03/06/2025]
Abstract
OBJECTIVES To explore the effective components of Guishao Yigong decoction (GYD) in the treatment of colorectal cancer and reveal its potential mechanism of action. METHODS Through network pharmacology, the main target and signaling pathway of GYD therapy for colorectal cancer (CRC) were found. Subsequently, the effect of GYD was verified by in vitro cell viability measurements, colony formation, and scratch healing tests. The effects of GYD on metabolic pathways in vivo were found through plasma metabolomics. Finally, flow cytometry and qPCR experiments were used to verify the cycle-blocking effect of GYD on CRC cells. KEY FINDINGS Based on the network pharmacological analysis and molecular docking technology, it was found that GYD could restrain the growth of CRC cells by affecting lipid metabolic pathways and mitogen-activated protein kinase (MAPK) signaling pathways. A series of cell experiments showed that GYD could inhibit the proliferation, migration and clonogenic ability of CRC cells. Furthermore, the plasma metabolomics results showed that GYD could affect the production of unsaturated fatty acids in mice. Flow cytometry and qPCR experiments further proved that GYD blocked the CRC cells in the G1 phase and modulated the expression of cell cycle-related targets, such as AKT, TP53, CDKN1A, and CDK2. CONCLUSIONS All the results indicated that GYD could regulate the related metabolism of unsaturated fatty acids. Thus, the cell cycle was blocked and the expressions of the key proteins such as AKT and TP53 were regulated, which achieved the purpose of intervention in colorectal cancer.
Collapse
Affiliation(s)
- Yuwen Fan
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, China
- College of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, China
| | - Quyi Wang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, China
- College of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, China
| | - Yun Zhang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, China
- College of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, China
| | - Yu Wang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, China
- College of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, China
| | - Wenwen Li
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, China
- College of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, China
| | - Shu Jiang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, China
- College of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, China
| | - Ji-Nao Duan
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, China
- College of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, China
| |
Collapse
|
38
|
Zhou T, Zhu X, Ji X, He J, Zhao K. Histone acetylation activated-IGF2BP3 regulates cyclin D1 mRNA stability to drive cell cycle transition and tumor progression of hepatocellular carcinoma. Int J Biol Macromol 2025; 306:141678. [PMID: 40037458 DOI: 10.1016/j.ijbiomac.2025.141678] [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: 12/30/2024] [Revised: 02/19/2025] [Accepted: 02/28/2025] [Indexed: 03/06/2025]
Abstract
Insulin-like growth factor 2 mRNA binding protein 3 (IGF2BP3) is an oncofetal protein, is strongly associated with tumor initiation and progression due to its upregulation. However, the regulatory mechanisms driving IGF2BP3 upregulation and its contribution to the development and progression in hepatocellular carcinoma (HCC) remain unclear. In this study, we demonstrated that IGF2BP3 is re-expressed in HCC mouse models, with elevated levels correlating with a poor prognosis in patients with HCC. Our data revealed that histone acetylation at the IGF2BP3 promoter region drives transcription activation of IGF2BP3 in primary hepatocytes. Notably, histone acetylation and transcriptional reactivation of IGF2BP3 were observed in human HCC tissues as well. Mechanistically, IGF2BP3 knockdown modulated the cell cycle and cell proliferation by limiting G1/S phase transition, which is dependent on cyclin D1. We further showed that IGF2BP3 maintains CCND1 mRNA stability by directly interacting with its 3'UTR. Importantly, IGF2BP3 recruits the RNA stabilizer PABPC1 to potentiate CCND1 mRNA stability. These two proteins synergistically protect CCND1 mRNA from degradation. Furthermore, IGF2BP3-depleted HCC cells were unable to form tumors in the xenograft model. High IGF2BP3 and CCND1 levels predicted poor outcomes in patients. Collectively, our findings highlight the pivotal role of the IGF2BP3/cyclin D1 axis and reveal a new regulatory mechanism for IGF2BP3 re-expression via transcriptional activation during hepatocarcinogenesis. These results indicate that the IGF2BP3/CCND1 axis is a promising prognostic biomarker and potential therapeutic target for HCC.
Collapse
Affiliation(s)
- Tao Zhou
- School of Public Health, Qingdao University, Qingdao, Shandong Province, China, 266071
| | - Xiaoxiao Zhu
- School of Public Health, Qingdao University, Qingdao, Shandong Province, China, 266071
| | - Xiaoying Ji
- School of Public Health, Qingdao University, Qingdao, Shandong Province, China, 266071
| | - Jinli He
- School of Public Health, Qingdao University, Qingdao, Shandong Province, China, 266071
| | - Kunming Zhao
- School of Public Health, Qingdao University, Qingdao, Shandong Province, China, 266071.
| |
Collapse
|
39
|
Handa S, Puri S, Chatterjee M, Puri V. Bioinformatics-Driven Investigations of Signature Biomarkers for Triple-Negative Breast Cancer. Bioinform Biol Insights 2025; 19:11779322241271565. [PMID: 40034579 PMCID: PMC11873876 DOI: 10.1177/11779322241271565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 06/29/2024] [Indexed: 03/05/2025] Open
Abstract
Breast cancer is a highly heterogeneous disorder characterized by dysregulated expression of number of genes and their cascades. It is one of the most common types of cancer in women posing serious health concerns globally. Recent developments and discovery of specific prognostic biomarkers have enabled its application toward developing personalized therapies. The basic premise of this study was to investigate key signature genes and signaling pathways involved in triple-negative breast cancer using bioinformatics approach. Microarray data set GSE65194 from the National Center for Biotechnology Information (NCBI) Gene Expression Omnibus was used for identification of differentially expressed genes (DEGs) using R software. Gene ontology and Kyoto Encyclopedia of Genes and Genome (KEGG) pathway enrichment analyses were carried out using the ClueGO plugin in Cytoscape software. The up-regulated DEGs were primarily engaged in the regulation of cell cycle, overexpression of spindle assembly checkpoint, and so on, whereas down-regulated DEGs were employed in alteration to major signaling pathways and metabolic reprogramming. The hub genes were identified using cytoHubba from protein-protein interaction (PPI) network for top up-regulated and down-regulated DEG's plugin in Cytoscape software. The hub genes were validated as potential signature biomarkers by evaluating the overall survival percentage in breast cancer patients.
Collapse
Affiliation(s)
- Shristi Handa
- Biotechnology Engineering, University Institute of Engineering and Technology, Panjab University, Chandigarh, India
| | - Sanjeev Puri
- Biotechnology Engineering, University Institute of Engineering and Technology, Panjab University, Chandigarh, India
| | - Mary Chatterjee
- Biotechnology Engineering, University Institute of Engineering and Technology, Panjab University, Chandigarh, India
| | - Veena Puri
- Centre for Systems Biology and Bioinformatics, Panjab University, Chandigarh, India
| |
Collapse
|
40
|
Gao J, Zheng J, Chen S, Lin S, Duan S. RMRP variants inhibit the cell cycle checkpoints pathway in cartilage‑hair hypoplasia. Mol Med Rep 2025; 31:81. [PMID: 39886981 PMCID: PMC11800184 DOI: 10.3892/mmr.2025.13446] [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: 09/12/2024] [Accepted: 01/06/2025] [Indexed: 02/01/2025] Open
Abstract
Cartilage‑hair hypoplasia (CHH) is an autosomal recessive form of metaphyseal chondrodysplasia caused by RNA component of mitochondrial RNA processing endoribonuclease (RMRP) gene variants; however, its molecular etiology remains unclear. Whole‑exome sequencing was performed to detect possible pathogenic variants in a patient with a typical short stature and sparse hair. A co‑segregation analysis was also conducted and variants in the family members of the patient were confirmed by Sanger sequencing. A novel compound heterozygous variant in RMRP (NR_003051.4: n.‑21_‑2dup and n.197C>T) was identified in the affected patient. Data from 2 years and 4 months of follow‑up showed a positive effect of growth hormone (GH) therapy on height. Subsequently, two gene expression profiles associated with CHH were obtained from the EMBL‑EBI ENA and ArrayExpress databases. Differentially expressed genes between patients with CHH and healthy controls were selected using R software and were subjected to core analysis using ingenuity pathway analysis (IPA) software. IPA core analysis showed that the 'cell cycle checkpoints' was the most prominent canonical pathway, and the top enriched diseases and functions included various types of cancer, immunological diseases, development disorders and respiratory diseases. The integrative analysis displayed that RMRP can regulate the aberrant expression of downstream targets mainly via the transcription factor TP53, which results in the inhibition of 'cell cycle checkpoints'; eventually, functions associated with the CHH phenotype, such as 'growth failure or short stature' are activated. In conclusion, novel disease‑causing genetic variants of RMRP expand the genetic etiology of CHH, which must be clinically differentiated from achondroplasia. The findings of the present study provide new insights into the mechanisms underlying CHH.
Collapse
Affiliation(s)
- Jian Gao
- Laboratory of Molecular Medicine, Institute of Maternal and Child Medicine Research, Shenzhen Maternity and Child Healthcare Hospital, Shenzhen, Guangdong 518040, P.R. China
- Shenzhen Key Laboratory of Maternal and Child Health and Diseases, Shenzhen Maternity and Child Healthcare Hospital, Shenzhen, Guangdong 518040, P.R. China
| | - Junge Zheng
- Laboratory of Molecular Medicine, Institute of Maternal and Child Medicine Research, Shenzhen Maternity and Child Healthcare Hospital, Shenzhen, Guangdong 518040, P.R. China
| | - Shiguo Chen
- Laboratory of Molecular Medicine, Institute of Maternal and Child Medicine Research, Shenzhen Maternity and Child Healthcare Hospital, Shenzhen, Guangdong 518040, P.R. China
| | - Sheng Lin
- Laboratory of Medical Genetics, Shenzhen Health Development Research and Data Management Center, Shenzhen, Guangdong 518028, P.R. China
| | - Shan Duan
- Laboratory of Molecular Medicine, Institute of Maternal and Child Medicine Research, Shenzhen Maternity and Child Healthcare Hospital, Shenzhen, Guangdong 518040, P.R. China
- Shenzhen Key Laboratory of Maternal and Child Health and Diseases, Shenzhen Maternity and Child Healthcare Hospital, Shenzhen, Guangdong 518040, P.R. China
| |
Collapse
|
41
|
Huang Y, Gao Y, Lin Z, Miao H. Involvement of the ubiquitin-proteasome system in the regulation of the tumor microenvironment and progression. Genes Dis 2025; 12:101240. [PMID: 39759114 PMCID: PMC11697063 DOI: 10.1016/j.gendis.2024.101240] [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: 06/29/2023] [Revised: 10/11/2023] [Accepted: 12/05/2023] [Indexed: 01/07/2025] Open
Abstract
The tumor microenvironment is a complex environment comprising tumor cells, non-tumor cells, and other critical non-cellular components. Some studies about tumor microenvironment have recently achieved remarkable progress in tumor treatment. As a substantial part of post-translational protein modification, ubiquitination is a crucial player in maintaining protein stability in cell signaling, cell growth, and a series of cellular life activities, which are also essential for regulating tumor cells or other non-tumor cells in the tumor microenvironment. This review focuses on the role and function of ubiquitination and deubiquitination modification in the tumor microenvironment while discussing the prospect of developing inhibitors targeting ubiquity-related enzymes, thereby providing ideas for future research in cancer therapy.
Collapse
Affiliation(s)
- Yulan Huang
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing 400038, China
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Yuan Gao
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing 400038, China
| | - Zhenghong Lin
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Hongming Miao
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing 400038, China
- School of Life Sciences, Chongqing University, Chongqing 401331, China
- Jinfeng Laboratory, Chongqing 401329, China
| |
Collapse
|
42
|
Rehmutulla M, Zhang S, Yin J, Huang J, Xiao Y, Hu Z, Tong Q, Zhang Y. Ustusolate E and 11α-Hydroxy-Ustusolate E induce apoptosis in cancer cell lines by regulating the PI3K/AKT/mTOR and p-53 pathways. Chin J Nat Med 2025; 23:346-353. [PMID: 40122664 DOI: 10.1016/s1875-5364(25)60840-5] [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: 12/08/2023] [Revised: 04/23/2024] [Accepted: 05/06/2024] [Indexed: 03/25/2025]
Abstract
Cancer represents a significant disease that profoundly impacts human health and longevity. Projections indicate a 47% increase in the global cancer burden by 2040 compared to 2020, accompanied by a further rise in the associated economic burden. Consequently, there is an urgent need to discover and develop new alternative drugs to mitigate the global impact of cancer. Natural products (NPs) play a crucial role in the identification and development of anticancer therapeutics. This study identified ustusolate E (UE) and its analog 11α-hydroxy-ustusolate E (HUE) from strain Aspergilluscalidoustus TJ403-EL05, and examined their antitumor activities and mechanisms of action. The findings demonstrate that both compounds significantly inhibited the proliferation and colony formation of AGS (human gastric cancer cells) and 786-O (human renal clear cell carcinoma cells), induced irreversible DNA damage, blocked the cell cycle at the G2/M phase, and further induced apoptosis in tumor cells. To the best of the authors' knowledge, this is the first report on the anticancer effects of UE and HUE and their underlying mechanisms. The present study suggests that HUE and UE could serve as lead compounds for the development of novel anticancer drugs.
Collapse
Affiliation(s)
- Mewlude Rehmutulla
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Sitian Zhang
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jie Yin
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jianzheng Huang
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yang Xiao
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhengxi Hu
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Qingyi Tong
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Yonghui Zhang
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
43
|
Hao J, Wang Z, Ren J, Cao S, Xie Z, Yang J, Li J, Ding W, Li J, Han Z, Yuan Y, Hai T, Ding S, Zhang MQ, Shi M. Single-cell multi-omics deciphers hepatocyte dedifferentiation and illuminates maintenance strategies. Cell Prolif 2025; 58:e13772. [PMID: 39810466 PMCID: PMC11882756 DOI: 10.1111/cpr.13772] [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: 07/11/2024] [Revised: 10/09/2024] [Accepted: 10/29/2024] [Indexed: 01/16/2025] Open
Abstract
Due to the similarity to human hepatocytes, porcine hepatocytes play an important role in hepatic research and drug evaluation. However, once hepatocytes were cultured in vitro, it was often prone to dedifferentiate, resulting in the loss of their characteristic features and normal functions, which impede their application in liver transplantation and hepatotoxic drugs evaluation. Up to now, this process has yet to be thoroughly investigated from the single-cell resolution and multi-omics perspective. In this study, we utilized 10× multiome technology to dissect the heterogeneity of porcine hepatocytes at different time points (Days 0, 1, 3, 5 and 7) during dedifferentiation. We comprehensively investigated cell heterogeneity, cellular dynamics, signalling pathways, potential gene targets, enhancer-driven gene regulatory networks, cell-cell communications of these cells and the conservation of mechanisms across species. We found that a series of critical signalling pathways driven by ERK, PI3K, Src and TGF-β were activated during this process, especially in the early stage of dedifferentiation. Based on these discoveries, we constructed a chemical combination targeting these pathways, which effectively inhibited the dedifferentiation of porcine hepatocytes in vitro. To validate the effectiveness of this combination, we transplanted such treated hepatocytes into FRGN mice, and the results demonstrated that these cells could effectively repopulate the liver and improve the survival of mice.
Collapse
Affiliation(s)
- Jie Hao
- School of Pharmaceutical SciencesTsinghua UniversityBeijingChina
| | - Zhenyi Wang
- MOE Key Laboratory of Bioinformatics, Beijing National Research Center for Information Science and Technology, Bioinformatics DivisionTsinghua UniversityBeijingChina
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai)Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Jilong Ren
- Key Laboratory of Organ Regeneration and ReconstructionState Key Laboratory of Stem Cell and Reproductive BiologyInstitute of ZoologyChinese Academy of SciencesBeijingChina
- Beijing Farm Animal Research Center, Institute of ZoologyChinese Academy of SciencesBeijingChina
| | - Shenghao Cao
- Institute of Image Processing and Pattern RecognitionShanghai Jiao Tong UniversityShanghaiChina
- Key Laboratory of System Control and Information ProcessingMinistry of Education of ChinaShanghaiChina
| | - Zhongchen Xie
- School of Life SciencesTsinghua UniversityBeijingChina
| | - Jinghuan Yang
- School of Pharmaceutical SciencesTsinghua UniversityBeijingChina
| | - Jiachen Li
- State Key Laboratory of Biopharmaceutical Preparation and DeliveryInstitute of Process EngineeringChinese Academy of SciencesBeijingChina
| | - Weizhe Ding
- School of Pharmaceutical SciencesTsinghua UniversityBeijingChina
| | - Jie Li
- Key Laboratory of Organ Regeneration and ReconstructionState Key Laboratory of Stem Cell and Reproductive BiologyInstitute of ZoologyChinese Academy of SciencesBeijingChina
- Beijing Farm Animal Research Center, Institute of ZoologyChinese Academy of SciencesBeijingChina
| | - Zhiqiang Han
- Key Laboratory of Organ Regeneration and ReconstructionState Key Laboratory of Stem Cell and Reproductive BiologyInstitute of ZoologyChinese Academy of SciencesBeijingChina
- Beijing Farm Animal Research Center, Institute of ZoologyChinese Academy of SciencesBeijingChina
| | - Ye Yuan
- State Key Laboratory of Biopharmaceutical Preparation and DeliveryInstitute of Process EngineeringChinese Academy of SciencesBeijingChina
| | - Tang Hai
- Key Laboratory of Organ Regeneration and ReconstructionState Key Laboratory of Stem Cell and Reproductive BiologyInstitute of ZoologyChinese Academy of SciencesBeijingChina
- Beijing Farm Animal Research Center, Institute of ZoologyChinese Academy of SciencesBeijingChina
| | - Sheng Ding
- School of Pharmaceutical SciencesTsinghua UniversityBeijingChina
| | - Michael Q. Zhang
- MOE Key Laboratory of Bioinformatics, Beijing National Research Center for Information Science and Technology, Bioinformatics DivisionTsinghua UniversityBeijingChina
- Department of Biological Sciences, Center for Systems BiologyThe University of TexasRichardsonTexasUSA
| | - Minglei Shi
- MOE Key Laboratory of Bioinformatics, Beijing National Research Center for Information Science and Technology, Bioinformatics DivisionTsinghua UniversityBeijingChina
| |
Collapse
|
44
|
Glaviano A, Singh SK, Lee EHC, Okina E, Lam HY, Carbone D, Reddy EP, O'Connor MJ, Koff A, Singh G, Stebbing J, Sethi G, Crasta KC, Diana P, Keyomarsi K, Yaffe MB, Wander SA, Bardia A, Kumar AP. Cell cycle dysregulation in cancer. Pharmacol Rev 2025; 77:100030. [PMID: 40148026 DOI: 10.1016/j.pharmr.2024.100030] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Accepted: 11/12/2024] [Indexed: 03/29/2025] Open
Abstract
Cancer is a systemic manifestation of aberrant cell cycle activity and dysregulated cell growth. Genetic mutations can determine tumor onset by either augmenting cell division rates or restraining normal controls such as cell cycle arrest or apoptosis. As a result, tumor cells not only undergo uncontrolled cell division but also become compromised in their ability to exit the cell cycle accurately. Regulation of cell cycle progression is enabled by specific surveillance mechanisms known as cell cycle checkpoints, and aberrations in these signaling pathways often culminate in cancer. For instance, DNA damage checkpoints, which preclude the generation and augmentation of DNA damage in the G1, S, and G2 cell cycle phases, are often defective in cancer cells, allowing cell division in spite of the accumulation of genetic errors. Notably, tumors have evolved to become dependent on checkpoints for their survival. For example, checkpoint pathways such as the DNA replication stress checkpoint and the mitotic checkpoint rarely undergo mutations and remain intact because any aberrant activity could result in irreparable damage or catastrophic chromosomal missegregation leading to cell death. In this review, we initially focus on cell cycle control pathways and specific functions of checkpoint signaling involved in normal and cancer cells and then proceed to examine how cell cycle control and checkpoint mechanisms can provide new therapeutic windows that can be exploited for cancer therapy. SIGNIFICANCE STATEMENT: DNA damage checkpoints are often defective in cancer cells, allowing cell division in spite of the accumulation of genetic errors. Conversely, DNA replication stress and mitotic checkpoints rarely undergo mutations because any aberrant activity could result in irreparable damage or catastrophic chromosomal missegregation, leading to cancer cell death. This review focuses on the checkpoint signaling mechanisms involved in cancer cells and how an emerging understanding of these pathways can provide new therapeutic opportunities for cancer therapy.
Collapse
Affiliation(s)
- Antonino Glaviano
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Palermo, Italy
| | - Samarendra K Singh
- School of Biotechnology, Institute of Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - E Hui Clarissa Lee
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Elena Okina
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Hiu Yan Lam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Daniela Carbone
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Palermo, Italy
| | - E Premkumar Reddy
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Mark J O'Connor
- Discovery Centre, AstraZeneca, Francis Crick Avenue, Cambridge CB2 0AA, United Kingdom
| | - Andrew Koff
- Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York
| | - Garima Singh
- School of Biotechnology, Institute of Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Justin Stebbing
- School of Life Sciences, Anglia Ruskin University, Cambridge, United Kingdom
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Karen Carmelina Crasta
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; Centre for Healthy Longevity, National University Health System, Singapore, Singapore
| | - Patrizia Diana
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Palermo, Italy
| | - Khandan Keyomarsi
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Michael B Yaffe
- MIT Center for Precision Cancer Medicine, Koch Institute for Integrative Cancer Research, Broad Institute, Massachusetts Institute of Technology, Cambridge, Boston, Massachusetts
| | - Seth A Wander
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Aditya Bardia
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
45
|
Zhang H, Li Y, Li N, Miao Y, Sun S, Gu L, Xiong B. Nicotinamide mononucleotide enhances the developmental potential of mouse early embryos exposed to perfluorooctanoic acid. Reprod Toxicol 2025; 132:108762. [PMID: 39613165 DOI: 10.1016/j.reprotox.2024.108762] [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: 10/09/2024] [Revised: 11/17/2024] [Accepted: 11/26/2024] [Indexed: 12/01/2024]
Abstract
Perfluorooctanoic acid (PFOA) exposure severely affects the health of animals and humans, including early embryonic development, but the effective approaches to improve the quality of embryos exposed to PFOA have not been explored. Here, we report that nicotinamide mononucleotide (NMN) can be used to attenuate the impairment of mouse early embryos caused by PFOA exposure. We find that NMN supplementation maintains the normal spindle assembly and proper chromosome alignment by restoring the acetylation level of microtubule to enhance the mitotic capacity of embryos at zygotic cleavage stage under PFOA exposure. In addition, NMN exerts its beneficial effect by enhancing mitochondrial function and eliminating accumulated reactive oxygen species (ROS), which in turn alleviates DNA damage and apoptosis in PFOA-exposed 2-cell embryos. Moreover, NMN ameliorates the quality of PFOA-exposed blastocysts via recovering the octamer-binding transcription factor 4 (Oct4) expression, the actin dynamics, and the total number of cells. Collectively, our findings demonstrate that supplementation with NMN is a feasible strategy to restore the compromised early embryonic development under PFOA exposure, providing a scientific basis for application of NMN to increase the female fertility.
Collapse
Affiliation(s)
- Hanwen Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Yu Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Na Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Yilong Miao
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Shaochen Sun
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Ling Gu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Bo Xiong
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China.
| |
Collapse
|
46
|
Hu S, Lu Y, Yu G, Zheng Z, Wang W, Ni K, Giri A, Zhang J, Zhang Y, Watanabe K, Yao G, Xing J. Epithelial-mesenchymal transition couples with cell cycle arrest at various stages. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.24.639880. [PMID: 40060597 PMCID: PMC11888286 DOI: 10.1101/2025.02.24.639880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
Numerous computational approaches have been developed to infer cell state transition trajectories from snapshot single-cell data. Most approaches first require projecting high-dimensional data onto a low-dimensional representation, raising the question of whether the dynamics of the system become distorted. Using epithelial-to-mesenchymal transition (EMT) as a test system, we show that both biology-guided low-dimensional representations and stochastic trajectory simulations in high-dimensional state space, not representations obtained with brute force dimension-reduction methods, reveal multiple distinct paths of TGF-β-induced EMT. The paths arise from coupling between EMT and cell cycle arrest at either the G1/S, G2/M or M checkpoints, contributing to cell-cycle related EMT heterogeneity. The present study emphasizes that caution should be taken when inferring transition dynamics from snapshot single-cell data in two- or three-dimensional representations, and that incorporating dynamical information can improve prediction accuracy.
Collapse
Affiliation(s)
- Sophia Hu
- Department of Computational and Systems Biology, University of Pittsburgh, USA
- Joint CMU-Pitt Ph.D. Program in Computational Biology, University of Pittsburgh, USA
| | - Yong Lu
- Department of Computational and Systems Biology, University of Pittsburgh, USA
| | - Gaohan Yu
- Department of Physics and Astronomy, University of Pittsburgh, USA
| | - Zhiqian Zheng
- Department of Computational and Systems Biology, University of Pittsburgh, USA
| | - Weikang Wang
- CAS Key Laboratory for Theoretical Physics, Institute of Theoretical Physics, Chinese Academy of Sciences, Beijing 100190, China
- School of Physical Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ke Ni
- Department of Computational and Systems Biology, University of Pittsburgh, USA
- Joint CMU-Pitt Ph.D. Program in Computational Biology, University of Pittsburgh, USA
| | - Amitava Giri
- Department of Computational and Systems Biology, University of Pittsburgh, USA
| | - Jingyu Zhang
- Department of Computational and Systems Biology, University of Pittsburgh, USA
- Joint CMU-Pitt Ph.D. Program in Computational Biology, University of Pittsburgh, USA
| | - Yan Zhang
- Department of Computational and Systems Biology, University of Pittsburgh, USA
| | | | - Guang Yao
- Department of Molecular & Cellular Biology, University of Arizona, Tucson, AZ 85721, USA
- Arizona Cancer Center, University of Arizona, Tucson, AZ 85719, USA
| | - Jianhua Xing
- Department of Computational and Systems Biology, University of Pittsburgh, USA
- Department of Physics and Astronomy, University of Pittsburgh, USA
- UPMC-Hillman Cancer Center, University of Pittsburgh, USA
| |
Collapse
|
47
|
Jones T, Feng J, Luyties O, Cozzolino K, Sanford L, Rimel JK, Ebmeier CC, Shelby GS, Watts LP, Rodino J, Rajagopal N, Hu S, Brennan F, Maas ZL, Alnemy S, Richter WF, Koh AF, Cronin NB, Madduri A, Das J, Cooper E, Hamman KB, Carulli JP, Allen MA, Spencer S, Kotecha A, Marineau JJ, Greber BJ, Dowell RD, Taatjes DJ. TFIIH kinase CDK7 drives cell proliferation through a common core transcription factor network. SCIENCE ADVANCES 2025; 11:eadr9660. [PMID: 40020069 PMCID: PMC11870056 DOI: 10.1126/sciadv.adr9660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 01/28/2025] [Indexed: 03/03/2025]
Abstract
How cyclin-dependent kinase 7 (CDK7) coordinately regulates the cell cycle and RNA polymerase II transcription remains unclear. Here, high-resolution cryo-electron microscopy revealed how two clinically relevant inhibitors block CDK7 function. In cells, CDK7 inhibition rapidly suppressed transcription, but constitutively active genes were disproportionately affected versus stimulus-responsive. Distinct transcription factors (TFs) regulate constitutive versus stimulus-responsive genes. Accordingly, stimulus-responsive TFs were refractory to CDK7 inhibition whereas constitutively active "core" TFs were repressed. Core TFs (n = 78) are predominantly promoter associated and control cell cycle and proliferative gene expression programs across cell types. Mechanistically, rapid suppression of core TF function can occur through CDK7-dependent phosphorylation changes in core TFs and RB1. Moreover, CDK7 inhibition depleted core TF protein levels within hours, consistent with durable target gene suppression. Thus, a major but unappreciated biological function for CDK7 is regulation of a TF cohort that drives proliferation, revealing an apparent universal mechanism by which CDK7 coordinates RNAPII transcription with cell cycle CDK regulation.
Collapse
Affiliation(s)
- Taylor Jones
- Department of Biochemistry, University of Colorado, Boulder, CO 80303, USA
| | - Junjie Feng
- Institute for Cancer Research, Chester Beatty Laboratories, 237 Fulham Road, London SW3 6JB, UK
| | - Olivia Luyties
- Department of Biochemistry, University of Colorado, Boulder, CO 80303, USA
| | - Kira Cozzolino
- Department of Biochemistry, University of Colorado, Boulder, CO 80303, USA
| | - Lynn Sanford
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, CO 80303, USA
- BioFrontiers Institute, University of Colorado, Boulder, CO 80303, USA
| | - Jenna K. Rimel
- Department of Biochemistry, University of Colorado, Boulder, CO 80303, USA
| | | | - Grace S. Shelby
- Department of Biochemistry, University of Colorado, Boulder, CO 80303, USA
| | - Lotte P. Watts
- Department of Biochemistry, University of Colorado, Boulder, CO 80303, USA
- BioFrontiers Institute, University of Colorado, Boulder, CO 80303, USA
| | - Jessica Rodino
- Department of Biochemistry, University of Colorado, Boulder, CO 80303, USA
| | | | - Shanhu Hu
- Syros Pharmaceuticals, Cambridge, MA 02140, USA
| | - Finn Brennan
- Department of Biochemistry, University of Colorado, Boulder, CO 80303, USA
| | - Zachary L. Maas
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, CO 80303, USA
- BioFrontiers Institute, University of Colorado, Boulder, CO 80303, USA
| | | | - William F. Richter
- Department of Biochemistry, University of Colorado, Boulder, CO 80303, USA
| | - Adrian F. Koh
- Materials and Structural Analysis Division, Thermo Fisher Scientific, Achtseweg Noord 5, 5651 Eindhoven, Netherlands
| | - Nora B. Cronin
- London Consortium for High-Resolution Cryo-EM, The Francis Crick Institute, London NW1 1AT, UK
| | | | - Jhuma Das
- Syros Pharmaceuticals, Cambridge, MA 02140, USA
| | | | | | | | - Mary A. Allen
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, CO 80303, USA
- BioFrontiers Institute, University of Colorado, Boulder, CO 80303, USA
| | - Sabrina Spencer
- Department of Biochemistry, University of Colorado, Boulder, CO 80303, USA
- BioFrontiers Institute, University of Colorado, Boulder, CO 80303, USA
| | - Abhay Kotecha
- Materials and Structural Analysis Division, Thermo Fisher Scientific, Achtseweg Noord 5, 5651 Eindhoven, Netherlands
| | | | - Basil J. Greber
- Institute for Cancer Research, Chester Beatty Laboratories, 237 Fulham Road, London SW3 6JB, UK
| | - Robin D. Dowell
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, CO 80303, USA
- BioFrontiers Institute, University of Colorado, Boulder, CO 80303, USA
| | - Dylan J. Taatjes
- Department of Biochemistry, University of Colorado, Boulder, CO 80303, USA
| |
Collapse
|
48
|
Giles KA, Taberlay PC, Cesare AJ, Jones MJK. Roles for the 3D genome in the cell cycle, DNA replication, and double strand break repair. Front Cell Dev Biol 2025; 13:1548946. [PMID: 40083661 PMCID: PMC11903485 DOI: 10.3389/fcell.2025.1548946] [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: 12/20/2024] [Accepted: 02/10/2025] [Indexed: 03/16/2025] Open
Abstract
Large eukaryotic genomes are packaged into the restricted area of the nucleus to protect the genetic code and provide a dedicated environment to read, copy and repair DNA. The physical organisation of the genome into chromatin loops and self-interacting domains provides the basic structural units of genome architecture. These structural arrangements are complex, multi-layered, and highly dynamic and influence how different regions of the genome interact. The role of chromatin structures during transcription via enhancer-promoter interactions is well established. Less understood is how nuclear architecture influences the plethora of chromatin transactions during DNA replication and repair. In this review, we discuss how genome architecture is regulated during the cell cycle to influence the positioning of replication origins and the coordination of DNA double strand break repair. The role of genome architecture in these cellular processes highlights its critical involvement in preserving genome integrity and cancer prevention.
Collapse
Affiliation(s)
- Katherine A. Giles
- Children’s Medical Research Institute, University of Sydney, Westmead, NSW, Australia
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Phillippa C. Taberlay
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Anthony J. Cesare
- Children’s Medical Research Institute, University of Sydney, Westmead, NSW, Australia
| | - Mathew J. K. Jones
- Faculty of Medicine, Frazer Institute, University of Queensland, Brisbane, QLD, Australia
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
49
|
Shi M, Ma G, Yang X. Artesunate: A Review of Its Potential Therapeutic Effects and Mechanisms in Digestive Diseases. Pharmaceutics 2025; 17:299. [PMID: 40142963 PMCID: PMC11945051 DOI: 10.3390/pharmaceutics17030299] [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: 01/13/2025] [Revised: 01/31/2025] [Accepted: 02/10/2025] [Indexed: 03/28/2025] Open
Abstract
Artesunate (ART), an artemisinin-derived semi-synthetic sesquiterpene lactone distinguished by its unique endoperoxide group, has become a cornerstone of clinical antimalarial therapy. Recent research has demonstrated its broad pharmacological profile, including its potent antimalarial, anti-inflammatory, anti-tumor, antidiabetic, immunomodulatory, and anti-fibrotic properties. These discoveries have significantly broadened the therapeutic scope of ART and offer new perspectives for its potential use in treating gastrointestinal disorders. Mechanistically, ART exerts significant therapeutic effects against diverse gastrointestinal pathologies-such as gastric ulcers, ulcerative colitis (UC), hepatic fibrosis (HF), gastric cancer, hepatocellular carcinoma, and colorectal cancer-via multimodal mechanisms, including cell cycle modulation, apoptosis induction, the suppression of tumor cell invasion and migration, proliferation inhibition, ferroptosis activation, and immune regulation. This review evaluates existing evidence on ART's therapeutic applications and molecular mechanisms in digestive diseases, intending to elucidate its clinical translation potential. ART emerges as a promising multi-target agent with significant prospects for improving the management of gastrointestinal disorders.
Collapse
Affiliation(s)
| | | | - Xiulan Yang
- Department of Pharmacology, The School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou 434023, China; (M.S.)
| |
Collapse
|
50
|
Stein CS, Linzer CR, Heer CD, Witmer NH, Cochran JD, Spitz DR, Boudreau RL. Mitoregulin Promotes Cell Cycle Progression in Non-Small Cell Lung Cancer Cells. Int J Mol Sci 2025; 26:1939. [PMID: 40076565 PMCID: PMC11899852 DOI: 10.3390/ijms26051939] [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: 12/23/2024] [Revised: 02/18/2025] [Accepted: 02/20/2025] [Indexed: 03/14/2025] Open
Abstract
Mitoregulin (MTLN) is a 56-amino-acid mitochondrial microprotein known to modulate mitochondrial energetics. MTLN gene expression is elevated broadly across most cancers and has been proposed as a prognostic biomarker for non-small cell lung cancer (NSCLC). In addition, lower MTLN expression in lung adenocarcinoma (LUAD) correlates with significantly improved patient survival. In our studies, we have found that MTLN silencing in A549 NSCLC cells slowed proliferation and, in accordance with this, we observed the following: (1) increased proportion of cells in the G1 phase of cell cycle; (2) protein changes consistent with G1 arrest (e.g., reduced levels and/or reduced phosphorylation of ERK, MYC, CDK2, and RB, and elevated p27Kip1); (3) reduction in clonogenic cell survival and; (4) lower steady-state cytosolic and mitochondrial H2O2 levels as indicated by use of the roGFP2-Orp1 redox sensor. Conflicting with G1 arrest, we observed a boost in cyclin D1 abundance. We also tested MTLN silencing in combination with buthionine sulfoximine (BSO) and auranofin (AF), drugs that inhibit GSH synthesis and thioredoxin reductase, respectively, to elevate the reactive oxygen species (ROS) amount to a toxic range. Interestingly, clonogenic survival after drug treatment was greater for MTLN-silenced cultures versus the control cultures. Lower H2O2 output and reduced vulnerability to ROS damage due to G1 status may have jointly contributed to the partial BSO + AF resistance. Overall, our results provide evidence that MTLN fosters H2O2 signaling to propel G1/S transition and suggest MTLN silencing as a therapeutic strategy to limit NSCLC growth.
Collapse
Affiliation(s)
- Colleen S. Stein
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA; (C.R.L.); (N.H.W.); (J.D.C.)
| | - Connor R. Linzer
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA; (C.R.L.); (N.H.W.); (J.D.C.)
| | - Collin D. Heer
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA 52242, USA; (C.D.H.); (D.R.S.)
| | - Nathan H. Witmer
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA; (C.R.L.); (N.H.W.); (J.D.C.)
| | - Jesse D. Cochran
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA; (C.R.L.); (N.H.W.); (J.D.C.)
| | - Douglas R. Spitz
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA 52242, USA; (C.D.H.); (D.R.S.)
| | - Ryan L. Boudreau
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA; (C.R.L.); (N.H.W.); (J.D.C.)
| |
Collapse
|