1
|
Ruban M, Pozhidaeva E, Bolotina L, Kaprin A. The Role of Diet and Nutrition in Cancer Development and Management: From Molecular Mechanisms to Personalized Interventions. Foods 2025; 14:1788. [PMID: 40428567 PMCID: PMC12110988 DOI: 10.3390/foods14101788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2025] [Revised: 05/08/2025] [Accepted: 05/16/2025] [Indexed: 05/29/2025] Open
Abstract
Diet plays a crucial role in cancer development and progression, beyond traditional risk factors. This review aims to summarize current evidence on the role of diet and specific nutrients in cancer development and progression, focusing on molecular mechanisms. We also discuss the potential of personalized dietary interventions, based on tumor and patient characteristics, in enhancing cancer prevention and treatment strategies. The review covers the impact of calories, protein, sugar, and other dietary components on signaling pathways and growth factors involved in carcinogenesis. We examine the influence of obesity, insulin resistance, and other metabolic factors on cancer risk and outcomes. The article also explores current dietary strategies, including calorie restriction, ketogenic diets, and the role of the gut microbiome in modulating response to anticancer therapies. Finally, we highlight the need for further research to develop targeted, personalized dietary recommendations based on an individual's tumor profile, stage of disease, and other clinical factors. Integrating such personalized dietary approaches into cancer prevention and treatment holds promise for improving patient outcomes and quality of life.
Collapse
Affiliation(s)
- Maxim Ruban
- P. Hertsen Moscow Oncology Research Institute—Branch of the National Medical Research Radiological Centre, 105425 Moscow, Russia
| | - Elizaveta Pozhidaeva
- P. Hertsen Moscow Oncology Research Institute—Branch of the National Medical Research Radiological Centre, 105425 Moscow, Russia
| | - Larisa Bolotina
- P. Hertsen Moscow Oncology Research Institute—Branch of the National Medical Research Radiological Centre, 105425 Moscow, Russia
| | - Andrey Kaprin
- P. Hertsen Moscow Oncology Research Institute—Branch of the National Medical Research Radiological Centre, 105425 Moscow, Russia
- Department of Urology and Operative Nephrology, Peoples’ Friendship University of Russia (RUDN University), Mikluho-Maklaya St., 6, 117198 Moscow, Russia
| |
Collapse
|
2
|
Chen X, Li R, Qiu Y, Lin F, Chen S, Li X, Sun H, Jiang G, Fang H, Qin J, Fang M. Design, synthesis, and biological evaluation of N-(2-amino-phenyl)-5-(4-aryl- pyrimidin-2-yl) amino)-1H-indole-2-carboxamide derivatives as novel inhibitors of CDK9 and class I HDACs for cancer treatment. Bioorg Chem 2025; 162:108577. [PMID: 40383016 DOI: 10.1016/j.bioorg.2025.108577] [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: 02/14/2025] [Revised: 05/06/2025] [Accepted: 05/09/2025] [Indexed: 05/20/2025]
Abstract
The mechanisms underlying transcriptional dysregulation in tumorigenesis have received considerable attention as promising therapeutic targets to combat human cancer. Cyclin-dependent kinase 9 (CDK9) and class I histone deacetylases (HDACs) are significant therapeutic targets due to their pivotal roles in the dysregulated transcriptional programs characteristic of many cancers. Furthermore, the combinatorial transcriptional therapy with CDK9 and class I HDAC inhibitors has been shown to have a synergistic anticancer effect. In this study, a series of novel N-(2-amino-phenyl)-5-(4-aryl-pyrimidin-2-yl) amino)-1H-indole-2-carboxamide derivatives were designed and synthesized as novel dual-functional inhibitors targeting CDK9 and HDAC signaling pathways for cancer treatment. Among the synthesized compounds, 13ea demonstrated potent anti-proliferative activities (IC50 < 5.0 μM) in various cancer cell lines (HeLa, MDA-MB-231, HepG2). In addition, 13ea was found to significantly inhibit the phosphorylation function of CDK9 and the deacetylation function of class I HDACs. Furthermore, 13ea was found to inhibit the protein activity of CDK9 (IC50 = 0.17 μM), HDAC1 (IC50 = 1.73 μM), and HDAC3 (IC50 = 1.11 μM). The docking studies predicted the binding patterns of 13ea in the active pockets of CDK9 and HDAC1/3. The cellular assays revealed that 13ea induced mitochondria-related apoptosis and G2/M phase arrest in cancer cells, showing superior activities compared to those of AZD-5438 (a CDK9 inhibitor) and Mocetinostat (an inhibitor of class I HDACs). Notably, the in vivo assay demonstrated that 13ea (30 mg/kg) exhibited significant inhibition on MDA-MB-231 xenograft tumor growth, with a tumor shrinkage rate of 76.83 %. In summary, we have identified 13ea as a novel CDK9/HDAC inhibitor with excellent anticancer activity in vitro and in vivo.
Collapse
Affiliation(s)
- Xiaohui Chen
- Department of Clinical laboratory, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, China; Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Rongna Li
- Department of Clinical laboratory, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, China
| | - Yingkun Qiu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Fanhong Lin
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Shutong Chen
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Xiaodan Li
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Hui Sun
- Department of Clinical laboratory, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, China
| | - Guanmin Jiang
- Department of Clinical laboratory, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, China
| | - Hua Fang
- Technical Innovation Center for Utilization of Marine Biological Resources, Third Institute of Oceanography, Ministry of Natural Resources, Xiamen 361005, China.
| | - Jingbo Qin
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China; Guangdong Provincial Engineering Research Center of Molecular Imaging and Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine Foundation of Guangdong Province, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong 519000, China.
| | - Meijuan Fang
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China.
| |
Collapse
|
3
|
Hao Q, Zhao W, Li Z, Lai Y, Wang Y, Yang Q, Zhang L. Combination therapy and dual-target inhibitors based on cyclin-dependent kinases (CDKs): Emerging strategies for cancer therapy. Eur J Med Chem 2025; 289:117465. [PMID: 40037064 DOI: 10.1016/j.ejmech.2025.117465] [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: 01/04/2025] [Revised: 02/22/2025] [Accepted: 02/26/2025] [Indexed: 03/06/2025]
Abstract
Cyclin-dependent kinases (CDKs) are pivotal regulators of the cell cycle and transcriptional machinery, making them attractive targets for cancer therapy. While CDK inhibitors have demonstrated promising clinical outcomes, they also face challenges in enhancing efficacy, particularly in overcoming drug resistance. Combination therapies have emerged as a key strategy to augment the effectiveness of CDK inhibitors when used alongside other kinase inhibitors or non-kinase-targeted agents. Dual-target inhibitors that simultaneously inhibit CDKs and other oncogenic drivers are gaining attention, offering novel avenues to optimize cancer therapy. Based on the structural characterization and biological functions of CDKs, this review comprehensively examines the structure-activity relationship (SAR) of existing dual-target CDK inhibitors from a drug design perspective. We also thoroughly investigate the preclinical studies and clinical translational potential of combination therapies and dual-target inhibitors. Tailoring CDK inhibitors to specific cancer subtypes and therapeutic settings will inspire innovative approaches for the next generation of CDK-related therapies, ultimately improving patient survival.
Collapse
Affiliation(s)
- Qi Hao
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Wenzhe Zhao
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Zhijia Li
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Yue Lai
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Yan Wang
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Qianqian Yang
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Lan Zhang
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China; Institute of Precision Drug Innovation and Cancer Center, Second Affiliated Hospital of Dalian Medical University, Dalian, 116023, China.
| |
Collapse
|
4
|
Bhuia MS, Chowdhury R, Afroz M, Akbor MS, Al Hasan MS, Ferdous J, Hasan R, de Alencar MVOB, Mubarak MS, Islam MT. Therapeutic Efficacy Studies on the Monoterpenoid Hinokitiol in the Treatment of Different Types of Cancer. Chem Biodivers 2025; 22:e202401904. [PMID: 39776341 DOI: 10.1002/cbdv.202401904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 12/10/2024] [Accepted: 12/15/2024] [Indexed: 01/11/2025]
Abstract
Hinokitiol (HK), a monoterpenoid that naturally occurs in plants belonging to the Cupressaceae family, possesses important biological activities, including an anticancer effect. This review summarizes its anticancer potential and draws possible molecular interventions. In addition, it evaluates the biopharmaceutical, toxicological properties, and clinical application of HK to establish its viability for future advancement as a dependable anticancer medication. The assessment is based on the most recent information available from various databases. Findings demonstrate that HK possesses substantial therapeutic advantages against diverse types of cancer (colon, cervical, breast, bone, endometrial, liver, prostate, oral, and skin) through various molecular mechanisms. HK induces oxidative stress, cytotoxicity, apoptosis, cell-cycle arrest at the G and S phases, and autophagy through modulation of phosphatidylinositol 3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR), p38/ERK/MAPK, nuclear factor kappa B, and c-Jun N-terminal kinase signaling pathways. Furthermore, this compound exhibits good oral bioavailability with excellent plasma clearance. Clinical uses of HK demonstrate therapeutic advantages without any significant negative effects. A thorough study of the pertinent data suggests that HK may serve as a viable candidate for developing novel cancer therapies. Consequently, more extensive studies are necessary to evaluate its cancer treatment efficacy, safety, and possible long-term hazards.
Collapse
Affiliation(s)
- Md Shimul Bhuia
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Dhaka, Bangladesh
- Phytochemistry and Biodiversity Research Laboratory, BioLuster Research Center Ltd, Gopalganj, Dhaka, Bangladesh
| | - Raihan Chowdhury
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Dhaka, Bangladesh
- Phytochemistry and Biodiversity Research Laboratory, BioLuster Research Center Ltd, Gopalganj, Dhaka, Bangladesh
| | - Meher Afroz
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Dhaka, Bangladesh
| | - Md Showkot Akbor
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Dhaka, Bangladesh
| | - Md Sakib Al Hasan
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Dhaka, Bangladesh
- Phytochemistry and Biodiversity Research Laboratory, BioLuster Research Center Ltd, Gopalganj, Dhaka, Bangladesh
| | - Jannatul Ferdous
- Department of Biotechnology and Genetic Engineering, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Dhaka, Bangladesh
| | - Rubel Hasan
- Phytochemistry and Biodiversity Research Laboratory, BioLuster Research Center Ltd, Gopalganj, Dhaka, Bangladesh
| | | | | | - Muhammad Torequl Islam
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Dhaka, Bangladesh
- Phytochemistry and Biodiversity Research Laboratory, BioLuster Research Center Ltd, Gopalganj, Dhaka, Bangladesh
- Pharmacy Discipline, Khulna University, Khulna, Dhaka, Bangladesh
| |
Collapse
|
5
|
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] [Download PDF] [Figures] [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
|
6
|
Zhou Y, Zhu H, Zhao L, Zhao G, Sun J. Bidirectional Mendelian randomization and potential mechanistic insights into the causal relationship between gut microbiota and malignant mesothelioma. Medicine (Baltimore) 2025; 104:e42245. [PMID: 40295238 PMCID: PMC12040020 DOI: 10.1097/md.0000000000042245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 04/07/2025] [Accepted: 04/08/2025] [Indexed: 04/30/2025] Open
Abstract
Malignant mesothelioma (MM) is a rare but aggressive cancer originating from mesothelial cells, which presents significant challenges to patients' physical and psychological well-being. The gut-lung axis underscores the connection between gut microbiota and respiratory diseases, with emerging evidence suggesting a strong association between gut microbiota and the development of MM. In this study, we conducted a two-sample Mendelian randomization (MR) analysis to investigate the potential causal relationship between gut microbiota and MM, while also exploring the underlying mechanisms through bioinformatics approaches. Gut microbiota summary data were obtained from the MiBioGen consortium, while MM data were sourced from the FinnGen R11 dataset. Causality was examined using the inverse variance weighted method as the primary analysis. Additional methods, including the weighted median, simple mode, MR-Egger, and weighted mode, were also employed. The robustness of the findings was validated through sensitivity analyses, and reverse causality was considered to further strengthen the MR results. Moreover, bioinformatics analyses were conducted on genetic loci associated with both gut microbiota and MM to explore potential underlying mechanisms. Our study suggests that genetically predicted increases in class.Bacilli, family.Rikenellaceae, genus.Clostridium innocuum group, and order.Lactobacillales were suggestively associated with a higher risk of MM, whereas increases in genus.Ruminococcaceae UCG004, genus.Flavonifractor, phylum.Firmicutes, genus.Anaerofilum, genus.Clostridium sensu stricto 1, and genus.Lactobacillus appeared to confer protective effects. Bioinformatics analysis indicated that differentially expressed genes near loci associated with gut microbiota might affect MM by modulating pathways and the tumor microenvironment. The results of this study point to a potential genetic predisposition linking gut microbiota to MM. Further experimental validation is crucial to confirm these candidate microbes, establish causality, and elucidate the underlying mechanisms.
Collapse
Affiliation(s)
- Yinjie Zhou
- Department of Thoracic Surgery, Ningbo No. 2 Hospital, Ningbo, Zhejiang, China
| | - Huangkai Zhu
- Department of Thoracic Surgery, Ningbo No. 2 Hospital, Ningbo, Zhejiang, China
| | - Long Zhao
- Department of Cardiovascular Surgery, Ningbo No. 2 Hospital, Ningbo, Zhejiang, China
| | - Guofang Zhao
- Department of Thoracic Surgery, Ningbo No. 2 Hospital, Ningbo, Zhejiang, China
| | - Jiaen Sun
- Department of Cardiovascular Surgery, Ningbo No. 2 Hospital, Ningbo, Zhejiang, China
| |
Collapse
|
7
|
Ahmed MZ, Billah MM, Ferdous J, Antar SI, Al Mamun A, Hossain MJ. Pan-cancer analysis reveals immunological and prognostic significance of CCT5 in human tumors. Sci Rep 2025; 15:14405. [PMID: 40274875 PMCID: PMC12022336 DOI: 10.1038/s41598-025-88339-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 01/28/2025] [Indexed: 04/26/2025] Open
Abstract
The chaperonin containing TCP1 subunit 5 (CCT5) is believed to function as a tumor driver. However, a systematic pan-cancer analysis of CCT5 is still lacking. Therefore, this study aimed to identify the potential role of CCT5 in different types of tumors. This study comprehensively investigated the gene expression, proteomic expression, immune infiltration, DNA methylation, genetic alterations, correlation with TMB and MSI, drug sensitivity, enrichment analysis, and prognostic significance of CCT5 in 33 different tumors based on the TIMER2.0, GEPIA2, UALCAN, SMART, cBioPortal, GSCA databases, and TCGAplot R package. The results revealed significant CCT5 overexpression in most tumors and was significantly associated with poor OS and DFS in different tumor types. Reduced promoter and N-shore methylation of CCT5, indicating its potential oncogenic and epigenetic roles. Amplification was the most common type of CCT5 alterations. Immune infiltration analysis revealed a strong correlation between CCT5 and different immune cells. CCT5 exhibited a significant correlation with TMB and MSI in KIRC and STAD. Furthermore, enrichment analysis revealed associations between CCT5 and cell cycle pathway and various cellular functions. These findings suggested that CCT5 might serve as a potential prognostic biomarker and target for immunotherapy in various cancers.
Collapse
Affiliation(s)
- Md Zabir Ahmed
- Big Bioinformatics Lab (BigBio Lab), Center for Health Innovation, Research, Action, and Learning- Bangladesh (CHIRAL Bangladesh), Dhaka, Bangladesh
- Department of Microbiology, Jagannath University, Dhaka, Bangladesh
| | - Md Mohtasim Billah
- Big Bioinformatics Lab (BigBio Lab), Center for Health Innovation, Research, Action, and Learning- Bangladesh (CHIRAL Bangladesh), Dhaka, Bangladesh
- Department of Microbiology, Jagannath University, Dhaka, Bangladesh
| | - Jannatul Ferdous
- Big Bioinformatics Lab (BigBio Lab), Center for Health Innovation, Research, Action, and Learning- Bangladesh (CHIRAL Bangladesh), Dhaka, Bangladesh
- Department of Microbiology, Jagannath University, Dhaka, Bangladesh
| | - Shoriful Islam Antar
- Big Bioinformatics Lab (BigBio Lab), Center for Health Innovation, Research, Action, and Learning- Bangladesh (CHIRAL Bangladesh), Dhaka, Bangladesh
- Department of Microbiology, Jahangirnagar University, Savar, Dhaka, Bangladesh
| | - Abdullah Al Mamun
- Big Bioinformatics Lab (BigBio Lab), Center for Health Innovation, Research, Action, and Learning- Bangladesh (CHIRAL Bangladesh), Dhaka, Bangladesh
- Department of Animal Science and Veterinary Medicine, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Bangladesh
| | - Md Jubayer Hossain
- Center for Health Innovation, Research, Action, and Learning-Bangladesh (CHIRAL Bangladesh), Dhaka, Bangladesh.
| |
Collapse
|
8
|
Lan Y, Sun J, Xu J, Chen X. Anti-lung cancer activity of lotusine in non-small cell lung cancer HCC827 via reducing proliferation, oxidative stress, induction of apoptosis, and G0/G1 cell cycle arrest via suppressing EGFR-Akt-ERK signalling. In Vitro Cell Dev Biol Anim 2025; 61:450-458. [PMID: 40392483 DOI: 10.1007/s11626-025-01048-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Accepted: 04/10/2025] [Indexed: 05/22/2025]
Abstract
Non-small cell lung cancer (NSCLC) is one of the leading causes of cancer-related deaths worldwide, with resistance to targeted therapies and the need for novel therapeutic agents driving ongoing research. In this study, we investigated the anti-lung cancer activity of lotusine, a natural alkaloid, in the A549 (non-EGFR mutant), and EGFR-mutant HCC827 NSCLC cell line (deletion in exon 19). Our results demonstrated that lotusine significantly inhibited cell proliferation in a concentration- and time-dependent manner of HCC827 cells in comparison to A549 cells. Mechanistic analysis revealed that lotusine induced apoptosis in HCC827 cells, as evidenced by increased expression of pro-apoptotic markers (Bax and cleaved caspase-3) and decreased levels of anti-apoptotic proteins (Bcl-2). Cell cycle analysis indicated that lotusine caused G0/G1 phase arrest. Importantly, lotusine exerted its effects through the inhibition of the epidermal growth factor receptor (EGFR) EGFR-Akt-ERK signaling pathway, as evidenced by reduction of p-EGFR, p-Akt, and p-ERK in a western blot analysis in HCC827 cells. These findings suggest that lotusine exerts potent anti-cancer effects via a multifaceted mechanism, including inhibition of proliferation, apoptosis induction, and cell cycle arrest, predominantly mediated by EGFR suppression. This study highlights lotusine as a promising therapeutic candidate for the treatment of EGFR-mutant NSCLC and provides insights into its molecular mechanisms of action, paving the way for further preclinical and clinical evaluations.
Collapse
Affiliation(s)
- Yuanmin Lan
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen and Longgang District People's Hospital of Shenzhen, Guangdong, Guangdong, 518172, China
| | - Jing Sun
- Department Of Oncology, The Fifth People's Hospital Of Dalian, Dalian Liaoning, 116021, China
| | - Jiqing Xu
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen and Longgang District People's Hospital of Shenzhen, Guangdong, Guangdong, 518172, China
| | - Xiaoying Chen
- Department of Respiratory and Critical Care Medicine, Lishui Second People's Hospital, Lishui Zhejiang, 323000, China.
| |
Collapse
|
9
|
Ceccaldi R, Cejka P. Mechanisms and regulation of DNA end resection in the maintenance of genome stability. Nat Rev Mol Cell Biol 2025:10.1038/s41580-025-00841-4. [PMID: 40133633 DOI: 10.1038/s41580-025-00841-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/24/2025] [Indexed: 03/27/2025]
Abstract
DNA end resection is a crucial early step in most DNA double-strand break (DSB) repair pathways. Resection involves the nucleolytic degradation of 5' ends at DSB sites to generate 3' single-stranded DNA overhangs. The first, short-range resection step is catalysed by the nuclease MRE11, acting as part of the MRE11-RAD50-NBS1 complex. Subsequent long-range resection is catalysed by the nucleases EXO1 and/or DNA2. Resected DNA is necessary for homology search and the priming of DNA synthesis in homologous recombination. DNA overhangs may also mediate DNA annealing in the microhomology-mediated end-joining and single-strand annealing pathways, and activate the DNA damage response. By contrast, DNA end resection inhibits DSB repair by non-homologous end-joining. In this Review, we discuss the importance of DNA end resection in various DSB repair pathways, the molecular mechanisms of end resection and its regulation, focusing on phosphorylation and other post-translational modifications that control resection throughout the cell cycle and in response to DNA damage.
Collapse
Affiliation(s)
- Raphael Ceccaldi
- INSERM U830, PSL Research University, Institut Curie, Paris, France.
| | - Petr Cejka
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland.
| |
Collapse
|
10
|
Alamillo-Vásquez JA, Pérez-Torres CA, Ibarra-Laclette E, Ramón-Farías F, Nicasio-Torres P, Alatorre-Cobos F. Secondary Metabolites from Croton Species and Their Biological Activity on Cell Cycle Regulators. Metabolites 2025; 15:216. [PMID: 40278345 PMCID: PMC12029420 DOI: 10.3390/metabo15040216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 03/16/2025] [Accepted: 03/21/2025] [Indexed: 04/26/2025] Open
Abstract
Plant-based traditional medicine integrates beliefs, knowledge, and practices to prevent and treat multiple diseases. Croton is a large and worldwide-spread genus belonging to Euphorbiaceae, a family well known for comprising many species with medicinal properties due to its high diversity of phytochemical constituents with biological activities. Among the various benefits of Croton species in traditional medicine, its use in cancer treatment has recently received significant attention from the scientific community. This review provides a general overview of different studies on the Croton genus in the research for alternative cancer treatments and the impact of its secondary metabolite catalog on cell cycle targets. Our analysis indicates that just under 30 secondary metabolites have been identified so far in latex and extracts obtained from leaves, twigs, or bark from 22 different Croton species. Based on standard assays using cell lines or human platelets, these molecules show multiple biological activities mainly compromising cell viability and cell cycle progression, supporting the ethnobotanical use of Croton species for cancer treatment. Several studies indicate that Croton metabolites target CDK-cyclin complexes and signaling routes that trigger apoptosis; however, further studies are needed to better understand the molecular mechanisms underlying Croton metabolites' effects and their accurate future applications in cancer treatment.
Collapse
Affiliation(s)
- Jorge Augusto Alamillo-Vásquez
- Red de Estudios Moleculares Avanzados (REMAV), Instituto de Ecología, A.C. (INECOL), Carretera Antigua a Coatepec 351, Col. El Haya, Xalapa 91073, Veracruz, Mexico; (J.A.A.-V.); (E.I.-L.)
| | - Claudia-Anahí Pérez-Torres
- SECIHTI-Red de Estudios Moleculares Avanzados (REMAV), Instituto de Ecología, A.C. (INECOL), Carretera Antigua a Coatepec 351, Col. El Haya, Xalapa 91073, Veracruz, Mexico
| | - Enrique Ibarra-Laclette
- Red de Estudios Moleculares Avanzados (REMAV), Instituto de Ecología, A.C. (INECOL), Carretera Antigua a Coatepec 351, Col. El Haya, Xalapa 91073, Veracruz, Mexico; (J.A.A.-V.); (E.I.-L.)
| | - Feliza Ramón-Farías
- Facultad de Ciencias Biológicas y Agropecuarias, Universidad Veracruzana, Córdoba 94500, Veracruz, Mexico;
| | - Pilar Nicasio-Torres
- Centro de Investigación Biomédica del Sur (CIBIS), Instituto Mexicano del Seguro Social (IMSS), Xochitepec 62790, Morelos, Mexico;
| | - Fulgencio Alatorre-Cobos
- SECIHTI-Unidad de Biología Integrativa Centro de Investigación Científica de Yucatán, A.C., Calle 43 No. 130 × 32 y 34, Col. Chuburná de Hidalgo, Mérida 97205, Yucatán, Mexico
| |
Collapse
|
11
|
Nizami ZN, Al Azzani M, Khaldi S, Wali AF, Magramane R, Samad SA, Eid AH, Arafat K, Al Dhaheri Y, Attoub S, Iratni R. Rhus coriaria (Sumac) induces autophagic cell death and inhibits mTOR, p38MAPK and STAT3 pathways in 5fluorouracil-resistant colorectal cancer cells. Front Pharmacol 2025; 16:1542204. [PMID: 40176890 PMCID: PMC11962434 DOI: 10.3389/fphar.2025.1542204] [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: 12/09/2024] [Accepted: 02/24/2025] [Indexed: 04/05/2025] Open
Abstract
Introduction Colorectal cancer is a leading cause of cancer related-death worldwide, and resistance to 5-fluorouracil (5FU, a key component of chemotherapy regimens, is a major clinical concern. We have previously elucidated the effects of Rhus coriaria ethanolic extract (RCE) in triple-negative breast cancer, CRC, and pancreatic cancer cells. Here, we explored the anticancer effects of RCE in parental (HCT-116-WT) and 5FU-resistant HCT-116 (HCT-116-5FU-R) CRC cells. Methods MTT assay was used to assess cell viability. Muse analyzer was used to assess cell viability, cell cycle distribution, and apoptosis. Additionally, colony formation and growth assays and western blots were performed. In vivo effects of RCE were assessed by an in ovo chick embryo tumor growth assay. Results We found that RCE inhibited the viability and colony formation and growth capacities of HCT-116-WT and HCT-116-5FU-R cells. The antiproliferative effects were attributed to DNA damage-mediated impairment of cell cycle at S phase, and induction of Beclin-1-independent autophagy in both cell lines. Mechanistically, inhibition of the mTOR, STAT3 and p38 MAPK pathways was implicated in the latter. Additionally, RCE induced caspase-7-independent apoptosis in HCT-116-WT cells. However, HCT-116-5FU-R cells were resistant to apoptosis through upregulation of survivin, and downregulation of Bax. Using autophagy and proteasome inhibitors, we clarified that autophagy and the proteasome pathway contributed to RCE-mediated cell death in HCT-116-WT and HCT-116-5FU-R cells. Lastly, we confirmed RCE inhibited the growth of both HCT-116-WT and HCT-116-5FU-R xenografts in a chick embryo model. Discussion Collectively, our findings highlight that RCE is a source of phytochemicals that can be used as anticancer agents for 5FU-resistant CRC.
Collapse
Affiliation(s)
- Zohra Nausheen Nizami
- Department of Biology, College of Science, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - Mazoun Al Azzani
- Department of Biology, College of Science, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - Samah Khaldi
- Department of Biology, College of Science, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - Adil Farooq Wali
- Department of Pharmaceutical Chemistry, RAK College of Pharmacy, RAK Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates
| | - Rym Magramane
- Department of Biology, College of Science, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - Shamaa Abdul Samad
- Department of Biology, College of Science, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - Ali H. Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Kholoud Arafat
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - Yusra Al Dhaheri
- Department of Biology, College of Science, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - Samir Attoub
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - Rabah Iratni
- Department of Biology, College of Science, United Arab Emirates University, Al-Ain, United Arab Emirates
| |
Collapse
|
12
|
Xiong S, Jin J, Zhao X, Zhao Y, He Z, Guo H, Gong C, Yu J, Guo L, Liang T. Cell Cycle-Based Molecular Features via Synthetic Lethality and Non-Coding RNA Interactions in Cancer. Genes (Basel) 2025; 16:310. [PMID: 40149461 PMCID: PMC11941865 DOI: 10.3390/genes16030310] [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: 01/22/2025] [Revised: 02/21/2025] [Accepted: 03/03/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND The cell cycle, a critical and intricate biological process, comprises various phases, and its dysregulation plays a pivotal role in tumorigenesis and metastasis. The exploration of cell cycle-based molecular subtypes across pan-cancers, along with the application of synthetic lethality concepts, holds promise for advancing cancer therapies. METHODS A pan-cancer analysis was conducted to assess the cell cycle serves as a reliable signature for classifying molecular subtypes and to understand the potential clinical application of genes as potential drug targets based on synthetic lethality. RESULTS Molecular subtypes derived from cell cycle features in certain cancers, particularly kidney-related malignancies, exhibited distinct immune characteristics. Synthetic lethal interactions within the cell cycle pathway were common, with significant genetic interactions further identifying potential drug targets through the exploitation of genetic relationships with key driver genes. Additionally, miRNAs and lncRNAs may influence the cell cycle through miRNA:mRNA interactions and ceRNA networks, thereby enriching the genetic interaction landscape. CONCLUSIONS These findings suggest that the cell cycle pathway could serve as a promising molecular subtype signature to enhance cancer prognostication and offer potential targets for anticancer drug development through synthetic lethality.
Collapse
Affiliation(s)
- Shizheng Xiong
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China; (S.X.); (J.J.); (X.Z.); (Y.Z.); (Z.H.); (C.G.)
| | - Jiaming Jin
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China; (S.X.); (J.J.); (X.Z.); (Y.Z.); (Z.H.); (C.G.)
| | - Xinmiao Zhao
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China; (S.X.); (J.J.); (X.Z.); (Y.Z.); (Z.H.); (C.G.)
| | - Yang Zhao
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China; (S.X.); (J.J.); (X.Z.); (Y.Z.); (Z.H.); (C.G.)
| | - Zhiheng He
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China; (S.X.); (J.J.); (X.Z.); (Y.Z.); (Z.H.); (C.G.)
| | - Haochuan Guo
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, Nanjing Normal University, Nanjing 210023, China;
| | - Chengjun Gong
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China; (S.X.); (J.J.); (X.Z.); (Y.Z.); (Z.H.); (C.G.)
| | - Jiafeng Yu
- Shandong Provincial Key Laboratory of Biophysics, Institute of Biophysics, Dezhou University, Dezhou 253023, China;
| | - Li Guo
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China; (S.X.); (J.J.); (X.Z.); (Y.Z.); (Z.H.); (C.G.)
| | - Tingming Liang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, Nanjing Normal University, Nanjing 210023, China;
| |
Collapse
|
13
|
Sun J, Liang S, Liu X, Zhang S, Li M, Zhang Q, Chen J. Insights into the selectivity of a brain-penetrant CDK4/6 vs CDK1/2 inhibitor for glioblastoma used in multiple replica molecular dynamics simulations. J Biomol Struct Dyn 2025; 43:2223-2242. [PMID: 38112295 DOI: 10.1080/07391102.2023.2294175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 11/23/2023] [Indexed: 12/21/2023]
Abstract
Cyclin dependent kinases (CDKs) play an important role in cell cycle regulation and their dysfunction is associated with many cancers. That is why CDKs have been attractive targets for the treatment of cancer. Glioblastoma is a cancer caused by the aberrant expression of CDK4/6, so exploring the mechanism of the selection of CDK4/6 toward inhibitors relative to the other family members CDK1/2 is essential. In this work, multiple replica molecular dynamics (MRMD) simulations, principal component analysis (PCA), free energy landscapes (FELs), molecular mechanics Poisson-Boltzmann/Generalized Born surface area (MM-PB/GBSA) and other methods were integrated to decipher the selectively binding mechanism of the inhibitor N1J to CDK4/6 and CDK1/2. Molecular electrostatic potential (MESP) analysis provides an explanation for the N1J selectivity. Residue-based free energy decomposition reveals that most of the hot residues are located at the same location of CDKs proteins, but the different types of residues in different proteins cause changes in binding energy, which is considered as a potential developmental direction to improve the selectivity of inhibitors to CDK4/6. These results provide insights into the source of inhibitor and CDK4/6 selectivity for the future development of more selective inhibitors.
Collapse
Affiliation(s)
- Jiahao Sun
- School of Physics and Electronics, Shandong Normal University, Jinan, China
| | - Shanshan Liang
- School of Physics and Electronics, Shandong Normal University, Jinan, China
| | - Xinguo Liu
- School of Physics and Electronics, Shandong Normal University, Jinan, China
| | - Shaolong Zhang
- School of Physics and Electronics, Shandong Normal University, Jinan, China
| | - Meng Li
- School of Physics and Electronics, Shandong Normal University, Jinan, China
| | - Qinggang Zhang
- School of Physics and Electronics, Shandong Normal University, Jinan, China
| | - Jianzhong Chen
- School of Science, Shandong Jiaotong University, Jinan, China
| |
Collapse
|
14
|
Babu M, Rao RM, Babu A, Jerom JP, Gogoi A, Singh N, Seshadri M, Ray A, Shelley BP, Datta A. Antioxidant Effect of Naringin Demonstrated Through a Bayes' Theorem Driven Multidisciplinary Approach Reveals its Prophylactic Potential as a Dietary Supplement for Ischemic Stroke. Mol Neurobiol 2025; 62:3918-3933. [PMID: 39352635 DOI: 10.1007/s12035-024-04525-6] [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: 01/31/2024] [Accepted: 09/23/2024] [Indexed: 01/03/2025]
Abstract
Naringin (NAR), a flavanone glycoside, occurs widely in citrus fruits, vegetables, and alcoholic beverages. Despite evidence of the neuroprotective effects of NAR on animal models of ischemic stroke, brain cell-type-specific data about the antioxidant efficacy of NAR and possible protein targets of such beneficial effects are limited. Here, we demonstrate the brain cell type-specific prophylactic role of NAR, an FDA-listed food additive, in an in vitro oxygen-glucose deprivation (OGD) model of cerebral ischemia using MTT and DCFDA assays. Using Bayes' theorem-based predictive model, we first ranked the top-10 protein targets (ALDH2, ACAT1, CTSB, FASN, LDHA, PTGS1, CTSD, LGALS1, TARDBP, and CDK1) from a curated list of 289 NAR-interacting proteins in neurons that might be mediating its antioxidant effect in the OGD model. When preincubated with NAR for 2 days, N2a and CTX-TNA2 cells could withstand up to 8 h of OGD without a noticeable decrease in cell viability. This cerebroprotective effect is partly mediated by reducing intracellular ROS production in the above two brain cell types. The antioxidant effect of NAR was comparable with the equimolar (50 µM) concentration of clinically used ROS-scavenger and neuroprotective edaravone. Molecular docking of NAR with the top-10 protein targets from Bayes' analysis showed the lowest binding energy for CDK1 (- 8.8 kcal/M). Molecular dynamics simulation analysis showed that NAR acts by inhibiting CDK1 by stably occupying its ATP-binding cavity. Considering diet has been listed as a risk factor for stroke, NAR may be explored as a component of functional food for stroke or related neurological disorders.
Collapse
Affiliation(s)
- Manju Babu
- Laboratory of Translational Neuroscience, Division of Neuroscience, Yenepoya Research Center, Yenepoya (Deemed to be University), University Road, Deralakatte, Mangalore, 575018, Karnataka, India
| | - Rajas M Rao
- Division of Data Analytics, Bioinformatics and Structural Biology, Yenepoya Research Center, Yenepoya (Deemed to be University), University Road, Deralakatte, Mangalore, 575018, Karnataka, India
| | - Anju Babu
- Department of Biological Sciences, Indian Institute of Science Education and Research, Bhopal, 462066, MP, India
| | | | - Anaekshi Gogoi
- Laboratory of Translational Neuroscience, Division of Neuroscience, Yenepoya Research Center, Yenepoya (Deemed to be University), University Road, Deralakatte, Mangalore, 575018, Karnataka, India
| | - Nikhil Singh
- Laboratory of Translational Neuroscience, Division of Neuroscience, Yenepoya Research Center, Yenepoya (Deemed to be University), University Road, Deralakatte, Mangalore, 575018, Karnataka, India
| | - Meenakshi Seshadri
- Department of Pharmacology, Yenepoya Pharmacy College and Research Center, Naringana, Deralakatte, Mangalore, 575018, Karnataka, India
| | - Animikh Ray
- Father Muller Research Center, Father Muller Medical College, Mangalore, 575002, Karnataka, India
| | - Bhaskara P Shelley
- Department of Neurology, Yenepoya Medical College, Yenepoya (Deemed to be University), University Road, Deralakatte, Mangalore, 575018, Karnataka, India
| | - Arnab Datta
- Laboratory of Translational Neuroscience, Division of Neuroscience, Yenepoya Research Center, Yenepoya (Deemed to be University), University Road, Deralakatte, Mangalore, 575018, Karnataka, India.
- Department of Pharmacology, Yenepoya Pharmacy College and Research Center, Naringana, Deralakatte, Mangalore, 575018, Karnataka, India.
| |
Collapse
|
15
|
Avolio E, Bassani B, Campanile M, Mohammed KA, Muti P, Bruno A, Spinetti G, Madeddu P. Shared molecular, cellular, and environmental hallmarks in cardiovascular disease and cancer: Any place for drug repurposing? Pharmacol Rev 2025; 77:100033. [PMID: 40148035 DOI: 10.1016/j.pharmr.2024.100033] [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: 12/04/2024] [Accepted: 12/17/2024] [Indexed: 03/29/2025] Open
Abstract
Cancer and cardiovascular disease (CVD) are the 2 biggest killers worldwide. Specific treatments have been developed for the 2 diseases. However, mutual therapeutic targets should be considered because of the overlap of cellular and molecular mechanisms. Cancer research has grown at a fast pace, leading to an increasing number of new mechanistic treatments. Some of these drugs could prove useful for treating CVD, which realizes the concept of cancer drug repurposing. This review provides a comprehensive outline of the shared hallmarks of cancer and CVD, primarily ischemic heart disease and heart failure. We focus on chronic inflammation, altered immune response, stromal and vascular cell activation, and underlying signaling pathways causing pathological tissue remodeling. There is an obvious scope for targeting those shared mechanisms, thereby achieving reciprocal preventive and therapeutic benefits. Major attention is devoted to illustrating the logic, advantages, challenges, and viable examples of drug repurposing and discussing the potential influence of sex, gender, age, and ethnicity in realizing this approach. Artificial intelligence will help to refine the personalized application of drug repurposing for patients with CVD. SIGNIFICANCE STATEMENT: Cancer and cardiovascular disease (CVD), the 2 biggest killers worldwide, share several underlying cellular and molecular mechanisms. So far, specific therapies have been developed to tackle the 2 diseases. However, the development of new cardiovascular drugs has been slow compared with cancer drugs. Understanding the intersection between pathological mechanisms of the 2 diseases provides the basis for repurposing cancer therapeutics for CVD treatment. This approach could allow the rapid development of new drugs for patients with CVDs.
Collapse
Affiliation(s)
- Elisa Avolio
- Bristol Heart Institute, Laboratory of Experimental Cardiovascular Medicine, Translational Health Sciences, Bristol Medical School, University of Bristol, United Kingdom.
| | - Barbara Bassani
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry, and Immunology, IRCCS MultiMedica, Milan, Italy
| | - Marzia Campanile
- Laboratory of Cardiovascular Pathophysiology - Regenerative Medicine, IRCCS MultiMedica, Milan, Italy; Department of Biosciences, University of Milan, Milan, Italy
| | - Khaled Ak Mohammed
- Bristol Heart Institute, Laboratory of Experimental Cardiovascular Medicine, Translational Health Sciences, Bristol Medical School, University of Bristol, United Kingdom; Department of Cardiothoracic Surgery, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Paola Muti
- IRCCS MultiMedica, Milan, Italy; Department of Biomedical, Surgical and Dental Health Sciences, University of Milan, Italy
| | - Antonino Bruno
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry, and Immunology, IRCCS MultiMedica, Milan, Italy; Laboratory of Immunology and General Pathology, Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy.
| | - Gaia Spinetti
- Laboratory of Cardiovascular Pathophysiology - Regenerative Medicine, IRCCS MultiMedica, Milan, Italy.
| | - Paolo Madeddu
- Bristol Heart Institute, Laboratory of Experimental Cardiovascular Medicine, Translational Health Sciences, Bristol Medical School, University of Bristol, United Kingdom.
| |
Collapse
|
16
|
Gai S, Cao P, Zhong X, Lin Y, Lin B, Jiang M. Designing an anticancer Pd(II) complex as poly(ADP-ribose) polymerase 1 inhibitor. Int J Biol Macromol 2025; 297:139885. [PMID: 39818402 DOI: 10.1016/j.ijbiomac.2025.139885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 01/01/2025] [Accepted: 01/13/2025] [Indexed: 01/18/2025]
Abstract
Targeting DNA repair mechanisms, particularly PARP-1 inhibition, has emerged as a promising strategy for developing anticancer therapies. we designed and synthesized two 2-thiazolecarboxaldehyde thiosemicarbazone palladium(II) complexes (C1 and C2), and evaluated their anti-cancer activities. These Pd(II) complexes exhibited potent PARP-1 enzyme inhibition and demonstrated considerable antiproliferative activity against various cancer cell lines. In vivo studies using the A549 tumor xenograft model revealed that C2 effectively suppressed tumor growth and exhibited minimal systemic toxicity. Mechanistically, C2 induced A549 cell death through multiple pathways: cell cycle arrest, elevated intracellular reactive oxygen species (ROS) levels, DNA damage induction, exacerbated DNA double-strand breakage via PARP-1 inhibition, mitochondrial membrane potential reduction, and ultimately apoptosis. These findings provide a new design strategy for developing safe and highly effective PARP-1 inhibitors.
Collapse
Affiliation(s)
- Shuangshuang Gai
- School of Biological and Food Engineering, Guangxi Science & Technology Normal University, Laibin, Guangxi 546199, China
| | - Peng Cao
- School of Food and Health, Guilin Tourism University, Guilin, Guangxi 541006, China
| | - Xuwei Zhong
- School of Biological and Food Engineering, Guangxi Science & Technology Normal University, Laibin, Guangxi 546199, China
| | - YiCan Lin
- School of Biological and Food Engineering, Guangxi Science & Technology Normal University, Laibin, Guangxi 546199, China
| | - Benxin Lin
- School of Biological and Food Engineering, Guangxi Science & Technology Normal University, Laibin, Guangxi 546199, China
| | - Ming Jiang
- School of Biological and Food Engineering, Guangxi Science & Technology Normal University, Laibin, Guangxi 546199, China.
| |
Collapse
|
17
|
Li N, Xu J, Li Y, Elango J, Wu W. Polyethylene Glycolylation of the Purified Basic Protein (Protamine) of Squid ( Symplectoteuthis oualaniensis): Structural Changes and Evaluation of Proliferative Effects on Fibroblast. Int J Mol Sci 2025; 26:1869. [PMID: 40076495 PMCID: PMC11899872 DOI: 10.3390/ijms26051869] [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: 01/11/2025] [Revised: 02/18/2025] [Accepted: 02/20/2025] [Indexed: 03/14/2025] Open
Abstract
In recent years, arginine-rich basic proteins have garnered significant attention due to their essential roles in various biological processes. However, the potential of marine-derived proteins in this domain remains largely unexplored. This study presents, for the first time, the isolation and purification of a 14.3 kDa protamine (SOP) from the mature spermatogonial tissues of Symplectoteuthis oualaniensis. Additionally, we obtained an 18.5 kDa PEGylated derivative, SOP-PEG. The physicochemical properties of both SOP and SOP-PEG were comprehensively characterized using SEM, FTIR, CD, and TGA. PEGylation markedly altered the surface morphology, secondary structure, and thermal stability of SOP. In vitro studies demonstrated that PEGylation significantly enhanced the biocompatibility of SOP, leading to improved proliferation of L-929 fibroblasts. Furthermore, both SOP and its PEGylated derivative (SOP-PEG) regulated the cell cycle, activated the PI3K-Akt signaling pathway, and modulated anti-apoptotic mechanisms, suggesting their potential to support cell survival and facilitate tissue regeneration. Notably, SOP-PEG exhibited superior bioactivity, likely attributable to its enhanced delivery efficiency conferred by PEGylation. Collectively, these findings underscore the promising applications of SOP and SOP-PEG in regenerative medicine and highlight the pivotal role of PEGylation in augmenting the bioactivity of SOP.
Collapse
Affiliation(s)
- Na Li
- Department of Marine Pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China; (N.L.); (J.X.); (Y.L.)
| | - Jiren Xu
- Department of Marine Pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China; (N.L.); (J.X.); (Y.L.)
| | - Yu Li
- Department of Marine Pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China; (N.L.); (J.X.); (Y.L.)
| | - Jeevithan Elango
- Department of Marine Pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China; (N.L.); (J.X.); (Y.L.)
- Department of Biomaterials Engineering, Faculty of Health Sciences, UCAM-Universidad Católica San Antonio de Murcia, Guadalupe, 30107 Murcia, Spain
- Center of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600077, India
| | - Wenhui Wu
- Department of Marine Pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China; (N.L.); (J.X.); (Y.L.)
- Marine Biomedical Science and Technology Innovation Platform of Lin-gang Special Area, Shanghai 201306, China
- Putuo Branch of International Combined Research Center for Marine Biological Sciences, Zhoushan 316104, China
| |
Collapse
|
18
|
Ahmadi Y, Faiq T, Abolhasani S. Impact of G1 phase kinetics on the acquisition of stemness in cancer cells: the critical role of cyclin D. Mol Biol Rep 2025; 52:230. [PMID: 39951181 DOI: 10.1007/s11033-025-10351-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 02/07/2025] [Indexed: 05/09/2025]
Abstract
Cancer stem cells (CSCs) represent a unique subpopulation of cells with the ability to self-renew and differentiate, thereby sustaining tumor growth and contributing to disease recurrence. Although CSCs predominantly reside in the G0 phase, their stem-like properties, such as the expression of specific CD markers, self-renewal, differentiation potential, tumor initiation, drug resistance, and increased invasive and metastatic potential, manifest during their active proliferative phase. Rapidly dividing cells exhibit alterations in their cell cycle, often characterized by shortened or bypassed G1 phases, a phenomenon observed in both embryonic stem cells and cancerous cells. Dysregulation of cell cycle control is a hallmark of cancer, leading to uncontrolled cellular proliferation and tumorigenesis. Disruption in key regulatory proteins, signaling pathways, and cell cycle checkpoints-particularly during the G1 phase-enables cancer cells to escape normal proliferation restrictions. The rapid cell-cycle progression can impair the timely degradation of proteins critical for cell cycle regulation, particularly cyclin D, thereby compromising proper cell cycle control. Therefore these proteins may be passed to daughter cells, promoting further rounds of rapid cycles. Additionally, cyclin D is often overexpressed in cancer cells, further exacerbating uncontrolled proliferation. These mechanisms may underpin key properties of CSCs, including rapid proliferation and their stem-like traits. This review examines the relationship between G1 phase kinetics and the acquisition of stem-like characteristics, emphasizing how rapid G1 phase progression and transitions between dormancy and active proliferation contribute to the emergence of CSC traits.
Collapse
Affiliation(s)
- Yasin Ahmadi
- Department of Medical Laboratory Science, Komar University of Science and Technology, Sulaymaniyah, Kurdistan Region, Iraq.
| | - Tahran Faiq
- Department of Medical Laboratory Science, Komar University of Science and Technology, Sulaymaniyah, Kurdistan Region, Iraq
| | - Sakhavat Abolhasani
- Department of Basic Sciences and Health, Sarab Faculty of Medical Sciences, Sarab, East Azerbaijan, Iran.
- Sarab School of Medical Sciences and Health Services, Sarab, East Azerbaijan, Iran.
| |
Collapse
|
19
|
Xing Y, Jia D, Zhu X, Yang J, Gao Z, Meng N, Xu H, Wang M, Chang S, Zhao M, Zhang S, Mu Z, Tang Q, Zhao W. Inotodiol induces hepatocellular carcinoma apoptosis by activation of MAPK/ERK pathway. PLoS One 2025; 20:e0318450. [PMID: 39879230 PMCID: PMC11778785 DOI: 10.1371/journal.pone.0318450] [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: 09/12/2024] [Accepted: 01/14/2025] [Indexed: 01/31/2025] Open
Abstract
Hepatocellular carcinoma(HCC) has a high mortality and morbidity rate and seriously jeopardizes human life. Chemicals and chemotherapeutic agents have been experiencing problems such as side effects and drug resistance in the treatment of HCC, which cannot meet the needs of clinical treatment. Therefore, finding novel low-toxicity and high-efficiency anti-hepatocellular carcinoma drugs and exploring their mechanisms of action have become the current problems to be solved in the treatment of HCC. Several studies have reported anticancer effects of inotodiol. This study focuses on the anticancer effect of inotodiol in HCC cells and its molecular mechanism, aiming to explore its anticancer effect in depth. The CCK8 assay was utilized to assess cell viability, the scratch assay was utilized to detect migration ability, the clone formation assay was utilized to detect clonogenic ability, and flow cytometry was utilized to analyze apoptosis and cell cycle. Animal experiments was utilized to verify the inhibitory effect of inotodiol on HCC. Meanwhile, western blotting was utilized to detect proteins associated with apoptosis, cell cycle and MAPK/ERK pathway. These results showed that inotodiol has the ability to promote apoptosis, as well as inhibit the ability of cell proliferation, migration, and clonogenic ability. The cell cycle was arrested in G1 phase, when the expression of CDK2, CDK4, CDK6 and Cyclin D were inhibited. In addition, inotodiol showed to induce apoptosis, characterized by an increase in Bax expression, a decrease in Bcl-2, Bcl-XL and MCL1 expression, the initiation of cleaved PARP1 and cleaved caspase 3, and inhibition of the MAPK/ERK pathway. Animal studies demonstrated that inotodiol possessed the ability to suppress tumor growth in nude mice models, at the same time, there was no significant impact on the body weight and organs of the mice. In conclusion, the findings presented herein compellingly suggest that inotodiol may serve as a promising candidate for the treatment of hepatocellular carcinoma (HCC).
Collapse
Affiliation(s)
- Yushuang Xing
- Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang, China
- Graduate Department, Qiqihar Medical University, Qiqihar, Heilongjiang, China
| | - Di Jia
- Department of Medical Technology, Qiqihar Medical University, Qiqihar, Heilongjiang, China
- The Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Xinping Zhu
- Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang, China
| | - Jialu Yang
- Department of Medical Technology, Qiqihar Medical University, Qiqihar, Heilongjiang, China
| | - Zhipeng Gao
- Department of Medical Technology, Qiqihar Medical University, Qiqihar, Heilongjiang, China
| | - Nana Meng
- Basic Medical Science College, Qiqihar Medical University, Qiqihar, Heilongjiang, China
| | - Haohao Xu
- Department of Medical Technology, Qiqihar Medical University, Qiqihar, Heilongjiang, China
| | - Mengxiao Wang
- Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang, China
| | - Shijun Chang
- Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang, China
| | - Mingqian Zhao
- Department of Medical Technology, Qiqihar Medical University, Qiqihar, Heilongjiang, China
| | - Shanbo Zhang
- Department of Medical Technology, Qiqihar Medical University, Qiqihar, Heilongjiang, China
| | - Zichen Mu
- The Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Qiang Tang
- Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang, China
- Rehabilitation Center, The Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Weiming Zhao
- Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang, China
| |
Collapse
|
20
|
Toson EA, El-Fallal AA, Oransa MA, El-Gharabawy HM. In vitro antitumor effects of methanolic extracts of three Ganoderema mushrooms. Sci Rep 2025; 15:2274. [PMID: 39824924 PMCID: PMC11748650 DOI: 10.1038/s41598-025-86162-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 01/08/2025] [Indexed: 01/20/2025] Open
Abstract
Ganoderma mushrooms have a variety of pharmacological activities and may have antitumor effects. Therefore, the antitumor activity of the methanolic fruiting body extracts of three Ganoderma spp. will be evaluated by estimating cell viability, cell cycle parameters and the mode of cellular death. In this regard, Sulfo-rhodamine B staining and flow cytometry were used. Hepatocellular carcinoma (HepG2) and breast ductal carcinoma (T-47D) cell lines were used as cancer models, while mouse normal liver (BNL) and oral epithelial cell (OEC) lines were used as respective controls. The results revealed that Ganoderma resinaceum extract decreased the viability of BNL at an IC50 > 100 µg/mL but not that of HepG2 at an IC50 of 72.32 µg/mL. Additionally, Ganoderma australe and Ganoderma mbrekobenum decreased the viability of OEC cell line at an IC50 of 328.29 and 271.56 µg/ mL, respectively. On the other hand, the IC50 of T-47D were 221.95 and 236.45 µg/mL, respectively. The three extracts arrested the cell life cycle at the G1 phase in each case. G. resinaceum extract stimulated total apoptosis (Q2 + Q4) of 19.99% with low necrosis (Q1). However, the percentages of total cell necrosis in the T-47D cell line treated with the other two extracts were 31.10% and 18.28%, respectively while the percentages of total cell apoptosis were 6.83% and 1.78%, respectively. Thus, G. resinaceum significantly inhibited the viability of the HepG2 cell line, while both the G. australe and G. mbrekobenum extracts significantly decreased the viability of the T-47D cell line. These results may encourage speculation about their possible use for the therapeutic management of hepatocellular carcinoma and breast ductal carcinoma after further investigation.
Collapse
Affiliation(s)
- Elshahat A Toson
- Chemistry Department, Faculty of Science, Damietta University, New Damietta, 34517, Egypt.
| | - Amira A El-Fallal
- Botany and Microbiology Department, Faculty of Science, Damietta University, New Damietta, 34517, Egypt
| | - Marwa A Oransa
- Botany and Microbiology Department, Faculty of Science, Damietta University, New Damietta, 34517, Egypt
| | - Hoda M El-Gharabawy
- Botany and Microbiology Department, Faculty of Science, Damietta University, New Damietta, 34517, Egypt.
| |
Collapse
|
21
|
Pandey AK, Trivedi V. Heat shock protein HSPA8 impedes hemin-induced cellular-toxicity in liver. Toxicol In Vitro 2025; 102:105959. [PMID: 39486598 DOI: 10.1016/j.tiv.2024.105959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 08/27/2024] [Accepted: 10/28/2024] [Indexed: 11/04/2024]
Abstract
Accumulation of hemin in cells, tissues, and organs is one of the major pathological conditions linked to hemolytic diseases like malaria. Pro-oxidant hemin confers high toxicity following its accumulation. We tested the cellular toxicity of hemin on HepG2 cells by exploring modulation in various cellular characteristics. Hemin reduces the viability of HepG2 cells and brings about visible morphological changes. Hemin causes perforations on the surface of HepG2 cells observed through SEM. Hemin leads to the extracellular release of liver enzymes and reduces the wound-healing potential of HepG2 cells. Hemin leads to the fragmentation of HepG2 DNA, arrests the cell cycle progression in the S-phase and induces apoptosis in these cells. Western blot analysis revealed that hemin triggers both the extrinsic and intrinsic pathways of apoptosis in HepG2 cells. We have already shown that the cytoprotective protein HSPA8 can polymerize hemin and minimize its toxicity. Similar experiments with hemin in the presence and absence of HSPA8 showed that HSPA8 reverses all the tested toxic effects of hemin on HepG2 cells. The protection from hemin toxicity in HepG2 cells appeared to be due to the extracellular polymerization of hemin by HSPA8.
Collapse
Affiliation(s)
- Alok Kumar Pandey
- Malaria Research Group, Department of Bioscience and Bioengineering, Indian Institute of Technology-Guwahati, Guwahati 781039, Assam, India
| | - Vishal Trivedi
- Malaria Research Group, Department of Bioscience and Bioengineering, Indian Institute of Technology-Guwahati, Guwahati 781039, Assam, India.
| |
Collapse
|
22
|
Migliaccio I, Guarducci C, Malorni L. CDK4/6 Inhibitor Resistance in ER+ Breast Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1464:475-493. [PMID: 39821039 DOI: 10.1007/978-3-031-70875-6_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
The cyclin-dependent kinases 4 and 6 inhibitors are the mainstay of treatment for patients with hormone receptor-positive and HER2-negative breast cancer. The ability of these drugs to improve the outcome of patients both in the metastatic and the early setting has been largely demonstrated. However, resistance, either de novo or acquired, represents a major clinical challenge. In the past years, efforts have been made to identify biomarkers that might help in a better selection of patients or to unravel the mechanisms leading to resistance in order to develop new therapeutic strategies to overcome it. Alterations of cell cycle-related genes and proteins are among the best characterized markers of resistance, and pathways impacting the cell cycle, including nuclear and growth factor receptors signaling, have been thoroughly investigated. Despite this, to date, cyclin-dependent kinases 4 and 6 inhibitors are administered based only on the hormone receptor and HER2 status of the tumor, and patients progressing on therapy are managed with currently available treatments. Here we summarize present knowledge on the cyclin-dependent kinases 4 and 6 inhibitors' mechanisms of action, efficacy data, and mechanisms of resistance.
Collapse
Affiliation(s)
- Ilenia Migliaccio
- Translational Research Unit, Hospital of Prato, AUSL Toscana Centro, Prato, Italy
| | - Cristina Guarducci
- Translational Research Unit, Hospital of Prato, AUSL Toscana Centro, Prato, Italy
| | - Luca Malorni
- Translational Research Unit, Hospital of Prato, AUSL Toscana Centro, Prato, Italy.
| |
Collapse
|
23
|
Teodoro L, Carreira ACO, Sogayar MC. Exploring the Complexity of Pan-Cancer: Gene Convergences and in silico Analyses. BREAST CANCER (DOVE MEDICAL PRESS) 2024; 16:913-934. [PMID: 39691553 PMCID: PMC11651076 DOI: 10.2147/bctt.s489246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 11/06/2024] [Indexed: 12/19/2024]
Abstract
Cancer is a complex and multifaceted group of diseases characterized by highly intricate mechanisms of tumorigenesis and tumor progression, which complicates diagnosis, prognosis, and treatment. In recent years, targeted therapies have gained prominence by focusing on specific mutations and molecular features unique to each tumor type, offering more effective and personalized treatment options. However, it is equally critical to explore the genetic commonalities across different types of cancer, which has led to the rise of pan-cancer studies. These approaches help identify shared therapeutic targets across various tumor types, enabling the development of broader and potentially more widely applicable treatment strategies. This review aims to provide a comprehensive overview of key concepts related to tumors, including tumorigenesis processes, the tumor microenvironment, and the role of extracellular vesicles in tumor biology. Additionally, we explore the molecular interactions and mechanisms driving tumor progression, with a particular focus on the pan-cancer perspective. To achieve this, we conducted an in silico analysis using publicly available datasets, which facilitated the identification of both common and divergent genetic and molecular patterns across different tumor types. By integrating these diverse areas, this review offers a clearer and deeper understanding of the factors influencing tumorigenesis and highlights potential therapeutic targets.
Collapse
Affiliation(s)
- Leandro Teodoro
- Cell and Molecular Therapy NUCEL Group, School of Medicine, University of São Paulo, São Paulo, São Paulo, 01246-903, Brazil
- Biochemistry Department, Chemistry Institute, University of São Paulo, São Paulo, São Paulo, 05508-900, Brazil
| | - Ana Claudia O Carreira
- Cell and Molecular Therapy NUCEL Group, School of Medicine, University of São Paulo, São Paulo, São Paulo, 01246-903, Brazil
- Center of Human and Natural Sciences, Federal University of ABC, Santo André, São Paulo, 09280-560, Brazil
| | - Mari C Sogayar
- Cell and Molecular Therapy NUCEL Group, School of Medicine, University of São Paulo, São Paulo, São Paulo, 01246-903, Brazil
- Biochemistry Department, Chemistry Institute, University of São Paulo, São Paulo, São Paulo, 05508-900, Brazil
| |
Collapse
|
24
|
Kalitin N, Koroleva N, Lushnikova A, Babaeva M, Samoylenkova N, Savchenko E, Smirnova G, Borisova Y, Kostarev A, Karamysheva A, Pavlova G. N-Glycoside of Indolo[2,3- a]pyrrolo[3,4- c]carbazole LCS1269 Exerts Anti-Glioblastoma Effects by G2 Cell Cycle Arrest and CDK1 Activity Modulation: Molecular Docking Studies, Biological Investigations, and ADMET Prediction. Pharmaceuticals (Basel) 2024; 17:1642. [PMID: 39770484 PMCID: PMC11676706 DOI: 10.3390/ph17121642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 11/28/2024] [Accepted: 11/29/2024] [Indexed: 01/04/2025] Open
Abstract
Background/Objectives: Indolo[2,3-a]pyrrolo[3,4-c]carbazole scaffold is successfully used as an efficient structural motif for the design and development of different antitumor agents. In this study, we investigated the anti-glioblastoma therapeutic potential of glycosylated indolocarbazole analog LCS1269 utilizing in vitro, in vivo, and in silico approaches. Methods: Cell viability was estimated by an MTT assay. The distribution of cell cycle phases was monitored using flow cytometry. Mitotic figures were visualized by fluorescence microscopy. Quantitative RT-PCR was used to evaluate the gene expression. The protein expression was assessed by Western blotting. Molecular docking and computational ADMET were approved for the probable protein target simulations and predicted pharmacological assessments, respectively. Results: Our findings clearly suggest that LCS1269 displayed a significant cytotoxic effect against diverse glioblastoma cell lines and patient-derived glioblastoma cultures as well as strongly suppressed xenograft growth in nude mice. LCS1269 exhibited more potent anti-proliferative activity toward glioblastoma cell lines and patient-derived glioblastoma cultures compared to conventional drug temozolomide. We further demonstrated that LCS1269 treatment caused the severe G2 phase arrest of cell cycle in a dose-dependent manner. Mechanistically, we proposed that LCS1269 could affect the CDK1 activity both by targeting active site of this enzyme and indirectly, in particular through the modulation of the Wee1/Myt1 and FOXM1/Plk1 signaling pathways, and via p21 up-regulation. LCS1269 also showed favorable pharmacological characteristics in in silico ADME prediction in comparison with staurosporine, rebeccamycin, and becatecarin as reference drugs. Conclusions: Further investigations of LCS1269 as an anti-glioblastoma medicinal agent could be very promising.
Collapse
Affiliation(s)
- Nikolay Kalitin
- Laboratory of Tumor Cell Genetics, N.N. Blokhin National Medical Research Center of Oncology, Kashirskoe Shosse 24, 115478 Moscow, Russia;
| | - Natalia Koroleva
- Laboratory of Oncogenomics, N.N. Blokhin National Medical Research Center of Oncology, 115478 Moscow, Russia; (N.K.); (A.L.)
| | - Anna Lushnikova
- Laboratory of Oncogenomics, N.N. Blokhin National Medical Research Center of Oncology, 115478 Moscow, Russia; (N.K.); (A.L.)
| | - Maria Babaeva
- Molecular Medicine, Universitätsmedizin Berlin, 10117 Berlin, Germany;
| | - Nadezhda Samoylenkova
- Laboratory of Molecular and Cellular Neurogenetics, N.N. Burdenko National Medical Research Center of Neurosurgery, 125047 Moscow, Russia; (N.S.); (E.S.); (G.P.)
| | - Ekaterina Savchenko
- Laboratory of Molecular and Cellular Neurogenetics, N.N. Burdenko National Medical Research Center of Neurosurgery, 125047 Moscow, Russia; (N.S.); (E.S.); (G.P.)
| | - Galina Smirnova
- Laboratory of Biochemical Pharmacology and Tumor Models, N.N. Blokhin National Medical Research Center of Oncology, 115478 Moscow, Russia; (G.S.); (Y.B.)
| | - Yulia Borisova
- Laboratory of Biochemical Pharmacology and Tumor Models, N.N. Blokhin National Medical Research Center of Oncology, 115478 Moscow, Russia; (G.S.); (Y.B.)
| | - Alexander Kostarev
- Max Planck Institute for Biology, University of Tübingen, 72074 Tübingen, Germany;
| | - Aida Karamysheva
- Laboratory of Tumor Cell Genetics, N.N. Blokhin National Medical Research Center of Oncology, Kashirskoe Shosse 24, 115478 Moscow, Russia;
| | - Galina Pavlova
- Laboratory of Molecular and Cellular Neurogenetics, N.N. Burdenko National Medical Research Center of Neurosurgery, 125047 Moscow, Russia; (N.S.); (E.S.); (G.P.)
- Laboratory of Neurogenetics and Developmental Genetics, Institute of Higher Nervous Activity and Neurophysiology of RAS, 117485 Moscow, Russia
| |
Collapse
|
25
|
Saha S, Ray R, Paul S. Depside and depsidone-rich hydroalcoholic extract, resourced from the lichen Parmelinella wallichiana (Taylor) Elix & Hale selectively restricts Non-Small Cell Lung Cancer by modulating p53, FOXO1 and PALLADIN genes. Fitoterapia 2024; 179:106211. [PMID: 39277022 DOI: 10.1016/j.fitote.2024.106211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 08/30/2024] [Accepted: 09/10/2024] [Indexed: 09/17/2024]
Abstract
The non-specificity of contemporary cancer therapeutics has enticed us to develop safer, anticancer alternatives from natural resources. Lichens are unique natural entities which have long been neglected for explorations in cancer therapy, despite their vast potential. Our present study aims to investigate the anti-cancer potential of a wild lichen Parmelinella wallichiana. The anti-proliferative efficacy of the lichen extracts were screened through MTT assay against a panel of cell lines and the potent hydroalcoholic extract was selected for further evaluation against the most sensitive lung-cancer cell line A549 by implementing a wide range of microscopic and flow cytometric applications. The observations suggest that the extract could selectively induce apoptosis by augmenting ROS and disrupting the mitochondrial membrane potentiality. It was also found that the lichen-induced apoptosis was regulated by two crucial tumor suppressor genes, FOXO1, and p53, along with cell cycle inhibitor p21 which ultimately resulted in robust apoptosis through the up-regulation of pro-apoptotic BAX expression. Moreover, the extract also restricted the cancer progression by down-regulating the PALLADIN expression. Further, an LC-MS-based metabolomic profile highlighted a number of depsides, depsidones and dibenzofurans, which included atranorin, physodalic acid, salazinic acid, constictic acid and usnic acid. Then, an in silico docking with these lichen-derived metabolites against the PI3Kα receptor predicted these compounds has a binding affinity close to a standard PI3Kα inhibitor copanlisib. The study concludes that the extract restricts lung cancer possibly through the PI3Kα/FOXO1 axis and thus Parmelinella wallichiana represents a potential resource for anti-lung cancer drug development in future.
Collapse
Affiliation(s)
- Saparja Saha
- Laboratory of Cell and Molecular Biology, Department of Botany, Centre of Advanced Study, University of Calcutta, Kolkata 700 019, West Bengal, India
| | - Ribhu Ray
- Laboratory of Cell and Molecular Biology, Department of Botany, Centre of Advanced Study, University of Calcutta, Kolkata 700 019, West Bengal, India
| | - Santanu Paul
- Laboratory of Cell and Molecular Biology, Department of Botany, Centre of Advanced Study, University of Calcutta, Kolkata 700 019, West Bengal, India.
| |
Collapse
|
26
|
Hirsch GE, Parisi MM, Martins LAM, Costa-Beber LC, Andrade CMB, Barbé Tuana FM, Terra SR, Ferrão TDS, Wagner R, Emanuelli T, Guma FTCR. Cytotoxic properties of Thuya occidentalis hydroalcoholic extract on androgen unresponsive prostate cancer cells. Arch Physiol Biochem 2024:1-11. [PMID: 39564984 DOI: 10.1080/13813455.2024.2430488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 08/18/2024] [Accepted: 11/10/2024] [Indexed: 11/21/2024]
Abstract
Background: Androgen independent phase in prostate cancer (PCa) commonly limits the therapeutic efficacy. Thuya occidentalis through its main active compound, α-thujone, appears to be an option, considering its anti-proliferative, anti-metastatic and pro-apoptotic effects on hepatocellular carcinoma. However, studies on PCa are limited. Objective: To evaluate if T. occidentalis could be useful against androgen responsive and unresponsive PCa cells. Methods: Androgen responsive (LNCaP) and unresponsive (DU145 and PC3) cell lines were exposed to T. occidentalis hydroalcoholic extract (0.05 mL/mL) for different periods. Further, α-thujone was measured in the extract and tested in the cell lines. Results: T. occidentalis and α-thujone showed the highest cytotoxicity on LNCaP cells. In androgen unresponsive cells, T. occidentalis decreased cell viability and density, and promoted apoptosis, necrosis and cell cycle arrest, possibly associated with Cav-1 downregulation. The α-thujone present in the extract significantly LNCaP cells density, but did not affect DU145 and PC3 cells, suggesting that other compounds may also be cytotoxic to androgen unresponsive cells.
Collapse
Affiliation(s)
- Gabriela Elisa Hirsch
- Postgraduate Program in Biological Sciences: Biochemistry, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Mariana Migliorini Parisi
- Postgraduate Program in Biological Sciences: Biochemistry, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
- Biomedicine Course, Cruz Alta University, Cruz Alta, Brazil
| | - Leo Anderson Meira Martins
- Postgraduate Program in Biological Sciences: Biochemistry, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Lílian Corrêa Costa-Beber
- Postgraduate Program in Biological Sciences: Biochemistry, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | | | - Florencia Mária Barbé Tuana
- Postgraduate Program in Biological Sciences: Biochemistry, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
- Postgraduate Program in Cellular and Molecular Biology, School of Sciences, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil
| | - Silvia Resende Terra
- Postgraduate Program in Biological Sciences: Biochemistry, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | | | - Roger Wagner
- Food Science and Technology Department, Federal University of Santa Maria, Santa Maria, Brazil
| | - Tatiana Emanuelli
- Food Science and Technology Department, Federal University of Santa Maria, Santa Maria, Brazil
| | | |
Collapse
|
27
|
Chen YL, Chen YC, Suzuki A. ImmunoCellCycle-ID - a high-precision immunofluorescence-based method for cell cycle identification. J Cell Sci 2024; 137:jcs263414. [PMID: 39564775 DOI: 10.1242/jcs.263414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 10/18/2024] [Indexed: 11/21/2024] Open
Abstract
The cell cycle is a fundamental process essential for cell proliferation, differentiation and development. It consists of four major phases: G1, S, G2 and M. These phases collectively drive the reproductive cycle and are meticulously regulated by various proteins that play crucial roles in both the prevention and progression of cancer. Traditional methods for studying these functions, such as flow cytometry, require a substantial number of cells to ensure accuracy. In this study, we have developed a user-friendly immunofluorescence-based method for identifying cell cycle stages, providing single-cell resolution and precise identification of G1, early/mid S, late S, early/mid G2, late G2, and each sub-stage of the M phase using fluorescence microscopy called ImmunoCellCycle-ID. This method provides high-precision cell cycle identification and can serve as an alternative to, or in combination with, traditional flow cytometry to dissect detailed sub-stages of the cell cycle in a variety of cell lines.
Collapse
Affiliation(s)
- Yu-Lin Chen
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison, Madison, Wisconsin, 53705, USA
| | - Yu-Chia Chen
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison, Madison, Wisconsin, 53705, USA
- Molecular and Cellular Pharmacology Graduate Program, University of Wisconsin-Madison, Madison, Wisconsin, 53705, USA
| | - Aussie Suzuki
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison, Madison, Wisconsin, 53705, USA
- Molecular and Cellular Pharmacology Graduate Program, University of Wisconsin-Madison, Madison, Wisconsin, 53705, USA
- Carbone Comprehensive Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin, 53705, USA
| |
Collapse
|
28
|
Hanna DH, Al-Atmani AK, AlRashidi AA, Shafee EE. Camellia sinensis methanolic leaves extract: Phytochemical analysis and anticancer activity against human liver cancer cells. PLoS One 2024; 19:e0309795. [PMID: 39541389 PMCID: PMC11563400 DOI: 10.1371/journal.pone.0309795] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 08/19/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND The study's primary goal is to ascertain whether there is a relationship between the processed green tea methanolic extract's (GTME) phytochemical components and its potential effectiveness against human liver cancer cells. The GTME's phytochemical composition was identified using gas chromatography-mass spectrometry, and the extract's capacity to lower cellular proliferation and cause apoptosis in HepG2 cancerous liver cell lines was checked. RESULTS The findings of the gas chromatography-mass chromatogram showed that GTME included bioactive antioxidants and anticancer substances. Additionally, utilizing the MTT, comet assay, and acridine assay, GTME revealed a selective cytotoxic impact with a significant IC50 value (27.3 µg/ml) on HepG2 cells without any harmful effects on WI-38 healthy cells. Also, compared to untreated cells, the extract-treated HepG2 cells had an upsurge in the proportion of cells that have undergone apoptosis and displayed a comet nucleus, which is a sign of DNA damage. In addition, HepG2 cells treated with GTME revealed a stop in the G1 phase and sub-G1 apoptotic cells (37.32%) in a flow cytometry analysis. Furthermore, reactive oxygen species were shown to be responsible for HepG2 apoptosis, and the tested extract significantly reduced their levels in the treated cells. Lastly, compared to untreated cells in treated HepG2 cells, GTME significantly changed protein expression levels linked with cell cycle arrest in the G1 phase and apoptosis. CONCLUSION These findings provided information about the processes through which the GTME inhibited the growth of HepG2. Therefore, it has potential as an effective natural therapy for the treatment of human liver cancer. However, to validate these findings, animal models must be used for in vivo studies.
Collapse
Affiliation(s)
- Demiana H. Hanna
- Faculty of Science, Department of Chemistry, Cairo University, Giza, Egypt
| | - Ahlam K. Al-Atmani
- Faculty of Science, Department of Chemistry, Cairo University, Giza, Egypt
| | | | - E. El. Shafee
- Faculty of Science, Department of Chemistry, Cairo University, Giza, Egypt
| |
Collapse
|
29
|
Łupicka-Słowik A, Cossu F, Sieńczyk M. Palbociclib as an Antitumor Drug: A License to Kill. Molecules 2024; 29:5334. [PMID: 39598723 PMCID: PMC11596203 DOI: 10.3390/molecules29225334] [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: 09/27/2024] [Revised: 11/08/2024] [Accepted: 11/09/2024] [Indexed: 11/29/2024] Open
Abstract
Neoplastic cells are characterized by uncontrolled cell divisions caused by cell cycle dysregulation. Key regulatory proteins governing the transition from the G1 to the S phase are the CDK4 and CDK6 kinases, which are controlled by D-type cyclins. The CDK4/6 kinases enable the use of these proteins as targets for anticancer therapy because they prevent the growth and the development of malignant cells by inhibiting their activity. This paper surveys the clinical trial results concerning palbociclib, the first in-class FDA-approved anticancer drug for hormone-dependent breast cancer. It discusses the therapeutic applications in breast cancer as well as in solid tumors and hematopoietic malignancies. Additionally, the paper presents an analysis of palbociclib resistance acquired during therapy and explores new approaches, such as modifications to palbociclib that enhance its desired activity or open up new therapeutic possibilities (PROTACs).
Collapse
Affiliation(s)
- Agnieszka Łupicka-Słowik
- Division of Organic and Medicinal Chemistry, Faculty of Chemistry, Wroclaw University of Science and Technology, Wybrzeze Wyspianskiego 27, 50-370 Wroclaw, Poland;
| | - Federica Cossu
- National Research Council, Institute of Biophysics (IBF-CNR), Milan Unit, Via Corti, 12, 20133 Milan, Italy;
| | - Marcin Sieńczyk
- Division of Organic and Medicinal Chemistry, Faculty of Chemistry, Wroclaw University of Science and Technology, Wybrzeze Wyspianskiego 27, 50-370 Wroclaw, Poland;
| |
Collapse
|
30
|
Kuzmanov A, Salemi S, Eberli D, Kranzbühler B. Regulation of prostate-specific membrane antigen (PSMA) expression in prostate cancer cells after treatment with dutasteride and lovastatin. Neoplasia 2024; 57:101045. [PMID: 39236400 PMCID: PMC11405815 DOI: 10.1016/j.neo.2024.101045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 07/31/2024] [Accepted: 08/26/2024] [Indexed: 09/07/2024]
Abstract
PSMA expression gradually increases from benign prostatic hyperplasia to adenocarcinoma of the prostate and is therefore used for the development of improved diagnostic (PSMA)-based prostate cancer imaging tools. Pharmacological induction of PSMA is therefore eminent to further improve the detection rate of PSMA-based imaging. Our previous studies have demonstrated that lovastatin (Lova) and dutasteride (Duta) are able to induce PSMA expression. However, the mechanisms by which PSMA is regulated in prostate cancer remain poorly understood. Androgen receptor (AR) and homeobox B13 (HOXB13) are the best known regulators of PSMA, hence in the present study we aimed to explore the PSMA regulation by HOXB13 and AR signaling in LNCaP and VCaP cells following treatments with Lova and Duta. Furthermore, our previous research revealed a growth arrest in prostate cancer cells after Lova, but not after Duta treatment. To understand this discrepancy, we explored the influence of Lova and Duta on well known tumor growth promoters, such as AR, the mTOR/Akt signaling pathways and Cyclin D1. Our results showed that treatment with Lova leads to a significant inhibition of the investigated tumor promoters and results in growth regression of LNCaP and VCaP cells. In contrast, Duta does not show these effects. Furthermore, we confirm the cooperative effect of HOXB13 and AR in regulating PSMA in LNCaP cells, and extend the investigations to an additional prostate cancer cell line (VCaP).
Collapse
Affiliation(s)
- Aleksandar Kuzmanov
- Department of Urology, University Hospital Zürich, University of Zurich, Laboratory for Urologic Oncology and Stem Cell Therapy, Zurich, Switzerland
| | - Souzan Salemi
- Department of Urology, University Hospital Zürich, University of Zurich, Laboratory for Urologic Oncology and Stem Cell Therapy, Zurich, Switzerland
| | - Daniel Eberli
- Department of Urology, University Hospital Zürich, University of Zurich, Laboratory for Urologic Oncology and Stem Cell Therapy, Zurich, Switzerland
| | - Benedikt Kranzbühler
- Department of Urology, University Hospital Zürich, University of Zurich, Laboratory for Urologic Oncology and Stem Cell Therapy, Zurich, Switzerland.
| |
Collapse
|
31
|
Advani D, Kumar P. Uncovering Cell Cycle Dysregulations and Associated Mechanisms in Cancer and Neurodegenerative Disorders: A Glimpse of Hope for Repurposed Drugs. Mol Neurobiol 2024; 61:8600-8630. [PMID: 38532240 DOI: 10.1007/s12035-024-04130-7] [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: 12/25/2023] [Accepted: 03/19/2024] [Indexed: 03/28/2024]
Abstract
The cell cycle is the sequence of events orchestrated by a complex network of cell cycle proteins. Unlike normal cells, mature neurons subsist in a quiescent state of the cell cycle, and aberrant cell cycle activation triggers neuronal death accompanied by neurodegeneration. The periodicity of cell cycle events is choreographed by various mechanisms, including DNA damage repair, oxidative stress, neurotrophin activity, and ubiquitin-mediated degradation. Given the relevance of cell cycle processes in cancer and neurodegeneration, this review delineates the overlapping cell cycle events, signaling pathways, and mechanisms associated with cell cycle aberrations in cancer and the major neurodegenerative disorders. We suggest that dysregulation of some common fundamental signaling processes triggers anomalous cell cycle activation in cancer cells and neurons. We discussed the possible use of cell cycle inhibitors for neurodegenerative disorders and described the associated challenges. We propose that a greater understanding of the common mechanisms driving cell cycle aberrations in cancer and neurodegenerative disorders will open a new avenue for the development of repurposed drugs.
Collapse
Affiliation(s)
- Dia Advani
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly Delhi College of Engineering), Shahbad Daulatpur, Bawana Road, New Delhi, Delhi, 110042, India
| | - Pravir Kumar
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly Delhi College of Engineering), Shahbad Daulatpur, Bawana Road, New Delhi, Delhi, 110042, India.
| |
Collapse
|
32
|
Hao J, Han G, Liang X, Ruan Y, Huang C, Sa N, Hu H, Hu B, Li Z, Zhang K, Gao P, Dong X. PELO regulates erythroid differentiation through interaction with MYC to upregulate KLF10. FEBS J 2024; 291:4714-4731. [PMID: 39206622 DOI: 10.1111/febs.17254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 03/27/2024] [Accepted: 08/14/2024] [Indexed: 09/04/2024]
Abstract
Erythropoiesis is a multistep process of erythroid cell production that is controlled by multiple regulatory factors. Ribosome rescue factor PELO plays a crucial role in cell meiotic division and mice embryonic development. However, the function of PELO in erythroid differentiation remains unclear. Here, we showed that knockdown of PELO increased hemin-induced erythroid differentiation of K562 and HEL cells, exhibiting a higher number of benzidine-positive cells and increased mRNA levels of erythroid genes. PELO knockdown inhibited the proliferation and cell cycle progression and promoted apoptosis of K562 cells. Mechanistically, PELO could regulate the expression of KLF10 through interaction with MYC. Moreover, KLF10 knockdown also enhanced erythroid differentiation of K562 and HEL cells induced by hemin. Collectively, our results demonstrated that PELO regulates erythroid differentiation and increases KLF10 expression levels by interacting with MYC.
Collapse
Affiliation(s)
- Jinglan Hao
- College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Guiqin Han
- College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Xin Liang
- College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Yongtong Ruan
- College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Chen Huang
- College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Naer Sa
- College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Hang Hu
- College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Bixi Hu
- College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Zhongqi Li
- College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Kai Zhang
- Hansoh Bio, 9600 Medical Center drive, Rockville, USA
| | - Ping Gao
- College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Xiaoming Dong
- College of Life Sciences, Shaanxi Normal University, Xi'an, China
| |
Collapse
|
33
|
Park SY, Mun JG, Lee YS, Lee SB, Kim SJ, Jang JH, Kim HY, Hong SH, Kee JY. Inhibitory Effect of Alnustone on Survival and Lung Metastasis of Colorectal Cancer Cells. Nutrients 2024; 16:3737. [PMID: 39519569 PMCID: PMC11547205 DOI: 10.3390/nu16213737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 10/24/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND/OBJECTIVES Alnustone (Aln) is an effective compound of Alpinia katsumadae Hayata. Aln possesses various pharmacological activities such as antibacterial, anti-inflammatory, and anti-cancer effects. However, the inhibitory effect of Aln on colorectal cancer (CRC) has not yet been identified. Thus, research was conducted to clarify whether Aln can suppress the proliferative and metastatic ability of CRC cells. METHODS A cell viability assay was performed to confirm the decrease in CRC cell viability following Aln treatment. Flow cytometry was carried out to evaluate the effects of Aln on cell cycle arrest, autophagy, and apoptosis in CRC cells. In addition, a lung metastasis animal model was used to check the inhibitory effect of Aln on the metastasis of CRC cells. RESULTS Aln remarkably diminished the viability and colony-forming ability of several CRC cell lines. In addition, Aln led to a halt at the G0/G1 phase through downregulating cyclin D1-CDK4 in CRC cells. The upregulation of LC3B and p62 expression by Aln triggered autophagy of CRC cells. Moreover, Aln promoted mitochondrial depolarization, resulting in apoptosis of CRC cells. Oral administration of Aln significantly restrained the metastasized lung tumor nodules. CONCLUSIONS This study demonstrated that Aln can suppress the survival and lung metastasis of CRC cells by promoting cell cycle arrest, autophagy, and apoptosis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Seung-Heon Hong
- Department of Oriental Pharmacy, College of Pharmacy, Wonkwang-Oriental Medicines Research Institute, Wonkwang University, 460 Iksandae-ro, Iksan 54538, Jeonbuk, Republic of Korea; (S.-Y.P.); (J.-G.M.); (Y.-S.L.); (S.-B.L.); (S.-J.K.); (J.-H.J.); (H.-Y.K.)
| | - Ji-Ye Kee
- Department of Oriental Pharmacy, College of Pharmacy, Wonkwang-Oriental Medicines Research Institute, Wonkwang University, 460 Iksandae-ro, Iksan 54538, Jeonbuk, Republic of Korea; (S.-Y.P.); (J.-G.M.); (Y.-S.L.); (S.-B.L.); (S.-J.K.); (J.-H.J.); (H.-Y.K.)
| |
Collapse
|
34
|
Lichawska-Cieslar A, Szukala W, Ylla G, Machaj G, Ploskonka F, Chlebicka I, Szepietowski JC, Jura J. MCPIP1 modulates the miRNA‒mRNA landscape in keratinocyte carcinomas. J Exp Clin Cancer Res 2024; 43:290. [PMID: 39428471 PMCID: PMC11492624 DOI: 10.1186/s13046-024-03211-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 10/10/2024] [Indexed: 10/22/2024] Open
Abstract
BACKGROUND Monocyte Chemotactic Protein 1-Induced Protein 1 (MCPIP1, also called Regnase-1) is a negative modulator of inflammation with tumor-suppressive properties. Mice with keratinocyte-specific deletion of the Zc3h12a gene, encoding MCPIP1, (Mcpip1eKO mice) are more susceptible to the development of epidermal papillomas initiated by 7,12-dimethylbenz[a]-anthracene (DMBA) and promoted by 2-O-tetradecanoylphorbol-13-acetate (TPA). METHODS The aim of this study was to investigate the MCPIP1 RNase-dependent microRNA (miRNA)‒mRNA regulatory network in chemically induced squamous cell carcinoma (SCC)-like skin papillomas. Next-generation sequencing (NGS) coupled with bioinformatic analysis was used to shortlist the MCPIP1-dependent changes in protein-coding genes and miRNAs. The expression levels of the selected miRNAs were analyzed by quantitative PCR in human keratinocytes with MCPIP1 silencing. Functional studies were performed in human keratinocytes transfected with appropriate miRNA mimics. The DIANA-microT-CDS algorithm and DIANA-TarBase v7 database were used to predict potential target genes and identify the experimentally validated targets of differentially expressed (DE) miRNAs. RESULTS RNA sequencing (RNA-Seq) analysis of control and Mcpip1eKO DMBA/TPA-induced papillomas revealed transcriptome changes, with 2400 DE protein-coding genes and 33 DE miRNAs. The expression of miR-223-3p, miR-376c-3p, and miR-139-5p was confirmed to be dependent on MCPIP1 activity in both murine and human models. We showed that MCPIP1 directly regulates the expression of miR-376c-3p via direct cleavage of the corresponding precursor miRNA. The pro-proliferative activity of miR-223-3p, miR-376c-3p, and miR-139-5p was experimentally confirmed in SCC-like keratinocytes. Bioinformatic prediction of the mRNA targets of the DE-miRNAs revealed 416 genes as putative targets of the 18 upregulated miRNAs and 425 genes as putative targets of the 15 downregulated miRNAs. Further analyses revealed the murine interactions that are conserved in humans. Functional analysis indicated that during the development of cutaneous SCC, the most important pathways/processes mediated by the miRNA‒mRNA MCPIP1-dependent network are the regulation of inflammatory processes, epithelial cell proliferation, Wnt signaling, and miRNA transcription. CONCLUSIONS Loss of MCPIP1 modulates the expression profiles of 33 miRNAs in chemically induced Mcpip1eKO papillomas, and these changes directly affect the miRNA‒mRNA network and the modulation of pathways and processes related to carcinogenesis.
Collapse
Affiliation(s)
- Agata Lichawska-Cieslar
- Faculty of Biochemistry, Biophysics and Biotechnology, Department of General Biochemistry, Jagiellonian University, Gronostajowa 7, Krakow, 30-387, Poland
| | - Weronika Szukala
- Faculty of Biochemistry, Biophysics and Biotechnology, Department of General Biochemistry, Jagiellonian University, Gronostajowa 7, Krakow, 30-387, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, Lojasiewicza 11, Krakow, 30- 348, Poland
| | - Guillem Ylla
- Faculty of Biochemistry, Biophysics and Biotechnology, Laboratory of Bioinformatics and Genome Biology, Jagiellonian University, Gronostajowa 7, Krakow, 30-387, Poland
| | - Gabriela Machaj
- Faculty of Biochemistry, Biophysics and Biotechnology, Laboratory of Bioinformatics and Genome Biology, Jagiellonian University, Gronostajowa 7, Krakow, 30-387, Poland
| | - Faustyna Ploskonka
- Faculty of Biochemistry, Biophysics and Biotechnology, Department of General Biochemistry, Jagiellonian University, Gronostajowa 7, Krakow, 30-387, Poland
| | - Iwona Chlebicka
- Department of Dermatology, Venereology and Allergology, Wroclaw Medical University, Chalubinskiego 1, Wroclaw, 50-368, Poland
- Faculty of Medicine, Wroclaw University of Science and Technology, Grunwaldzki sq. 11, Wroclaw, 51-377, Polska
| | - Jacek C Szepietowski
- Department of Dermatology, Venereology and Allergology, Wroclaw Medical University, Chalubinskiego 1, Wroclaw, 50-368, Poland
- Faculty of Medicine, Wroclaw University of Science and Technology, Grunwaldzki sq. 11, Wroclaw, 51-377, Polska
| | - Jolanta Jura
- Faculty of Biochemistry, Biophysics and Biotechnology, Department of General Biochemistry, Jagiellonian University, Gronostajowa 7, Krakow, 30-387, Poland.
| |
Collapse
|
35
|
Kim T, Lee K, Cheon M, Yu W. GAN-WGCNA: Calculating gene modules to identify key intermediate regulators in cocaine addiction. PLoS One 2024; 19:e0311164. [PMID: 39361596 PMCID: PMC11449371 DOI: 10.1371/journal.pone.0311164] [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: 05/24/2024] [Accepted: 09/13/2024] [Indexed: 10/05/2024] Open
Abstract
Understanding time-series interplay of genes is essential for diagnosis and treatment of disease. Spatio-temporally enriched NGS data contain important underlying regulatory mechanisms of biological processes. Generative adversarial networks (GANs) have been used to augment biological data to describe hidden intermediate time-series gene expression profiles during specific biological processes. Developing a pipeline that uses augmented time-series gene expression profiles is needed to provide an unbiased systemic-level map of biological processes and test for the statistical significance of the generated dataset, leading to the discovery of hidden intermediate regulators. Two analytical methods, GAN-WGCNA (weighted gene co-expression network analysis) and rDEG (rescued differentially expressed gene), interpreted spatiotemporal information and screened intermediate genes during cocaine addiction. GAN-WGCNA enables correlation calculations between phenotype and gene expression profiles and visualizes time-series gene module interplay. We analyzed a transcriptome dataset of two weeks of cocaine self-administration in C57BL/6J mice. Utilizing GAN-WGCNA, two genes (Alcam and Celf4) were selected as missed intermediate significant genes that showed high correlation with addiction behavior. Their correlation with addictive behavior was observed to be notably significant in aspect of statistics, and their expression and co-regulation were comprehensively mapped in terms of time, brain region, and biological process.
Collapse
Affiliation(s)
- Taehyeong Kim
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science & Technology, Daegu, South Korea
| | - Kyoungmin Lee
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science & Technology, Daegu, South Korea
| | - Mookyung Cheon
- Dementia Research Group, Korean Brain Research Institute, Daegu, South Korea
| | - Wookyung Yu
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science & Technology, Daegu, South Korea
| |
Collapse
|
36
|
Contreras A, Sánchez SA, Rodríguez-Medina C, Botero JE. The role and impact of viruses on cancer development. Periodontol 2000 2024; 96:170-184. [PMID: 38641954 DOI: 10.1111/prd.12566] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 02/13/2024] [Accepted: 03/16/2024] [Indexed: 04/21/2024]
Abstract
This review focuses on three major aspects of oncoviruses' role in cancer development. To begin, we discuss their geographic distribution, revealing that seven oncoviruses cause 20% of all human cancers worldwide. Second, we investigate the primary carcinogenic mechanisms, looking at how these oncogenic viruses can induce cellular transformation, angiogenesis, and local and systemic inflammation. Finally, we investigate the possibility of SARS-CoV-2 infection reactivating latent oncoviruses, which could increase the risk of further disease. The development of oncovirus vaccines holds great promise for reducing cancer burden. Many unanswered questions about the host and environmental cofactors that contribute to cancer development and prevention remain, which ongoing research is attempting to address.
Collapse
Affiliation(s)
| | - Sandra Amaya Sánchez
- Advanced Periodontology Program, Escuela de Odontología, Universidad del Valle, Cali, Colombia
| | | | | |
Collapse
|
37
|
Sergeeva SV, Loshchenova PS, Oshchepkov DY, Orishchenko KE. Crosstalk between BER and NHEJ in XRCC4-Deficient Cells Depending on hTERT Overexpression. Int J Mol Sci 2024; 25:10405. [PMID: 39408734 PMCID: PMC11476898 DOI: 10.3390/ijms251910405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 09/21/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
Targeting DNA repair pathways is an important strategy in anticancer therapy. However, the unrevealed interactions between different DNA repair systems may interfere with the desired therapeutic effect. Among DNA repair systems, BER and NHEJ protect genome integrity through the entire cell cycle. BER is involved in the repair of DNA base lesions and DNA single-strand breaks (SSBs), while NHEJ is responsible for the repair of DNA double-strand breaks (DSBs). Previously, we showed that BER deficiency leads to downregulation of NHEJ gene expression. Here, we studied BER's response to NHEJ deficiency induced by knockdown of NHEJ scaffold protein XRCC4 and compared the knockdown effects in normal (TIG-1) and hTERT-modified cells (NBE1). We investigated the expression of the XRCC1, LIG3, and APE1 genes of BER and LIG4; the Ku70/Ku80 genes of NHEJ at the mRNA and protein levels; as well as p53, Sp1 and PARP1. We found that, in both cell lines, XRCC4 knockdown leads to a decrease in the mRNA levels of both BER and NHEJ genes, though the effect on protein level is not uniform. XRCC4 knockdown caused an increase in p53 and Sp1 proteins, but caused G1/S delay only in normal cells. Despite the increased p53 protein, p21 did not significantly increase in NBE1 cells with overexpressed hTERT, and this correlated with the absence of G1/S delay in these cells. The data highlight the regulatory function of the XRCC4 scaffold protein and imply its connection to a transcriptional regulatory network or mRNA metabolism.
Collapse
Affiliation(s)
- Svetlana V. Sergeeva
- Institute of Cytology and Genetics, Russian Academy of Sciences, Lavrentieva 10, Novosibirsk 630090, Russia; (P.S.L.); (K.E.O.)
- Department of Genetic Technologies, Novosibirsk State University, Pirogova 2, Novosibirsk 630090, Russia
| | - Polina S. Loshchenova
- Institute of Cytology and Genetics, Russian Academy of Sciences, Lavrentieva 10, Novosibirsk 630090, Russia; (P.S.L.); (K.E.O.)
- Department of Genetic Technologies, Novosibirsk State University, Pirogova 2, Novosibirsk 630090, Russia
| | - Dmitry Yu. Oshchepkov
- Institute of Cytology and Genetics, Russian Academy of Sciences, Lavrentieva 10, Novosibirsk 630090, Russia; (P.S.L.); (K.E.O.)
| | - Konstantin E. Orishchenko
- Institute of Cytology and Genetics, Russian Academy of Sciences, Lavrentieva 10, Novosibirsk 630090, Russia; (P.S.L.); (K.E.O.)
- Department of Genetic Technologies, Novosibirsk State University, Pirogova 2, Novosibirsk 630090, Russia
| |
Collapse
|
38
|
Wang FL, Chang X, Shi Y, Yang T, Li J, Dong H, Wang Q, Zhang S, Liu J. β-Ionone enhances the inhibitory effects of 5-fluorouracil on the proliferation of gastric adenocarcinoma cells by the GSK-3β signaling pathway. PLoS One 2024; 19:e0309014. [PMID: 39241034 PMCID: PMC11379261 DOI: 10.1371/journal.pone.0309014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 08/03/2024] [Indexed: 09/08/2024] Open
Abstract
5-Fluorouracil (5-FU) is widely used in the treatment of gastric cancer, and the emergence of drug resistance and toxic effects has limited its application. Therefore, there is an urgent need for safe and effective novel drugs or new therapies. β-Ionone (BI) is found in vegetables and fruits and possesses an inhibitory proliferation of tumor cells in vitro and in vivo. In this study, we investigated whether BI could enhance the inhibitory effects of 5-FU on the proliferation of gastric adenocarcinoma cells and the growth of gastric cancer cell xenografts in a mouse model. The effects of BI and 5-FU alone or their combination on the cell viability, apoptosis, and mitochondrial membrane potential, the cell cycle, and its related proteins-Cyclin D1, and CDK4 as well as PCNA and GSK-3β were evaluated in SGC-7901 cells and MKN45 cells by MTT, MB, flow cytometry and Western blot. In addition, the effects of BI and 5-FU alone or their combination on the growth of SGC-7901 cell xenografts in nude mice were investigated. The results showed that BI significantly enhanced the sensitivity of gastric adenocarcinoma cells to 5-FU in vitro and in vivo, i.e. proliferation inhibited, apoptosis induced and GSK-3β protein activated. Therefore, our results suggest that BI increases the antitumor effect of 5-FU on gastric adenocarcinoma cells, at least partly from an activated GSK-3β signaling pathway.
Collapse
Affiliation(s)
- Fa-Lin Wang
- Department of Clinical Laboratory, The Forth Affiliated Hospital of Harbin Medical University, Nangang District, Harbin, China
| | - Xiaoxia Chang
- Department of Clinical Laboratory, Xi'an No. 9 Hospital, Beilin District, Xi'an City, China
| | - Yuanyang Shi
- Department of Laboratory, Shaoyang Central Hospital, Daxiang District, Shaoyang City, China
| | - Tingting Yang
- Department of Clinical Laboratory, The Forth Affiliated Hospital of Harbin Medical University, Nangang District, Harbin, China
| | - Juan Li
- Department of Clinical Laboratory, The Forth Affiliated Hospital of Harbin Medical University, Nangang District, Harbin, China
| | - Hongwei Dong
- Public Health College, Harbin Medical University, Nangang District, Harbin, China
| | - Qi Wang
- Public Health College, Harbin Medical University, Nangang District, Harbin, China
| | - Shujun Zhang
- Department of Pathology, The Forth Affiliated Hospital of Harbin Medical University, Nangang District, Harbin, China
| | - Jiaren Liu
- Department of Clinical Laboratory, The Forth Affiliated Hospital of Harbin Medical University, Nangang District, Harbin, China
| |
Collapse
|
39
|
Chen YL, Reddy S, Suzuki A. Reversible and effective cell cycle synchronization method for studying stage-specific investigations. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.02.610832. [PMID: 39282459 PMCID: PMC11398389 DOI: 10.1101/2024.09.02.610832] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/22/2024]
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 regulate these events precisely. Studying the stage-specific functions of the cell cycle requires accumulating cell populations at the desired cell cycle stage. Cell synchronization, achieved through the use of cell cycle kinase and protein inhibitors, is often employed for this purpose. However, suboptimal concentrations of these inhibitors can result in reduced efficiency, irreversibility, and undesirable cell cycle defects. In this study, we have optimized effective and reversible techniques to synchronize the cell cycle at each stage in human RPE1 cells, utilizing both fixed high-precision cell cycle identification methods and high-temporal live-cell imaging. These reproducible synchronization methods are invaluable for investigating the regulatory mechanisms specific to each cell cycle stage.
Collapse
Affiliation(s)
- Yu-Lin Chen
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Syon Reddy
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Aussie Suzuki
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Molecular and Cellular Pharmacology Graduate Program, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Carbone Comprehensive Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
40
|
Kajiho H, Sakisaka T. Degradation of STIM1 through FAM134B-mediated ER-phagy is potentially involved in cell proliferation. J Biol Chem 2024; 300:107674. [PMID: 39128711 PMCID: PMC11414581 DOI: 10.1016/j.jbc.2024.107674] [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: 04/16/2024] [Revised: 08/02/2024] [Accepted: 08/05/2024] [Indexed: 08/13/2024] Open
Abstract
Autophagy is classified as nonselective or selective depending on the types of degrading substrates. Endoplasmic reticulum (ER)-phagy is a form of selective autophagy for transporting the ER-resident proteins to autolysosomes. FAM134B, a member of the family with sequence similarity 134, is a well-known ER-phagy receptor. Dysfunction of FAM134B results in several diseases including viral infection, inflammation, neurodegenerative disorder, and cancer, indicating that FAM134B has crucial roles in various kinds of intracellular functions. However, how FAM134B-mediated ER-phagy regulates intracellular functions is not well understood. In this study, we found that FAM134B knockdown in mammalian cells accelerated cell proliferation. FAM134B knockdown increased the protein amount of stromal interaction molecule 1 (STIM1), an ER Ca2+ sensor protein mediating the store-operated Ca2+ entry involved in G1 to S phase transition. FAM134B bound to STIM1 through its C-terminal cytosolic region. FAM134B knockdown reduced transport of STIM1 from the ER to autolysosomes. Finally, FAM134B knockdown accelerated G1 to S phase transition. These results suggest that FAM134B is involved in cell proliferation possibly through degradation of STIM1 via ER-phagy.
Collapse
Affiliation(s)
- Hiroaki Kajiho
- Division of Membrane Dynamics, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe, Japan; Department of Biochemical Pathophysiology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan.
| | - Toshiaki Sakisaka
- Division of Membrane Dynamics, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
41
|
Drainas AP, Hsu WH, Dallas AE, Poltorack CD, Kim JW, He A, Coles GL, Baron M, Bassik MC, Sage J. GCN2 is a determinant of the response to WEE1 kinase inhibition in small-cell lung cancer. Cell Rep 2024; 43:114606. [PMID: 39120974 PMCID: PMC11407228 DOI: 10.1016/j.celrep.2024.114606] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 06/28/2024] [Accepted: 07/24/2024] [Indexed: 08/11/2024] Open
Abstract
Patients with small-cell lung cancer (SCLC) are in dire need of more effective therapeutic options. Frequent disruption of the G1 checkpoint in SCLC cells creates a dependency on the G2/M checkpoint to maintain genomic integrity. Indeed, in pre-clinical models, inhibiting the G2/M checkpoint kinase WEE1 shows promise in inhibiting SCLC growth. However, toxicity and acquired resistance limit the clinical effectiveness of this strategy. Here, using CRISPR-Cas9 knockout screens in vitro and in vivo, we identified multiple factors influencing the response of SCLC cells to the WEE1 kinase inhibitor AZD1775, including the GCN2 kinase and other members of its signaling pathway. Rapid activation of GCN2 upon AZD1775 treatment triggers a stress response in SCLC cells. Pharmacological or genetic activation of the GCN2 pathway enhances cancer cell killing by AZD1775. Thus, activation of the GCN2 pathway represents a promising strategy to increase the efficacy of WEE1 inhibitors in SCLC.
Collapse
Affiliation(s)
- Alexandros P Drainas
- Department of Pediatrics, Stanford University, Stanford, CA, USA; Department of Genetics, Stanford University, Stanford, CA, USA
| | - Wen-Hao Hsu
- Department of Pediatrics, Stanford University, Stanford, CA, USA; Department of Genetics, Stanford University, Stanford, CA, USA
| | - Alec E Dallas
- Department of Pediatrics, Stanford University, Stanford, CA, USA; Department of Genetics, Stanford University, Stanford, CA, USA
| | - Carson D Poltorack
- Department of Pediatrics, Stanford University, Stanford, CA, USA; Department of Genetics, Stanford University, Stanford, CA, USA
| | - Jun W Kim
- Department of Pediatrics, Stanford University, Stanford, CA, USA; Department of Genetics, Stanford University, Stanford, CA, USA
| | - Andy He
- Department of Pediatrics, Stanford University, Stanford, CA, USA; Department of Genetics, Stanford University, Stanford, CA, USA
| | - Garry L Coles
- Department of Pediatrics, Stanford University, Stanford, CA, USA; Department of Genetics, Stanford University, Stanford, CA, USA
| | - Maya Baron
- Department of Pediatrics, Stanford University, Stanford, CA, USA; Department of Genetics, Stanford University, Stanford, CA, USA
| | | | - Julien Sage
- Department of Pediatrics, Stanford University, Stanford, CA, USA; Department of Genetics, Stanford University, Stanford, CA, USA.
| |
Collapse
|
42
|
Wang Z, Wang W, Luo Q, Song G. Plectin: Dual Participation in Tumor Progression. Biomolecules 2024; 14:1050. [PMID: 39334817 PMCID: PMC11430127 DOI: 10.3390/biom14091050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 08/20/2024] [Accepted: 08/21/2024] [Indexed: 09/30/2024] Open
Abstract
The plectin gene can encode a cytoskeletal linking protein, plectin, known for its interaction with three critical components of the cellular cytoskeleton: intermediate filaments, microtubules, and actin filaments. In recent years, more and more studies have reported that plectin is closely related to tumorigenesis and development, exhibiting both tumor-suppressive and tumor-promoting functions. Here, we first introduce the molecular structure and function of plectin, and then we summarize the current understanding of the crucial role of plectin in cancer progression. Finally, we also discuss the possible reasons for the different roles of plectin expression in various types of cancer and highlight the double-edged sword role of plectin in tumor progression. The review aims to deepen the comprehensive understanding of plectin's role in cancer and further help to develop novel therapeutic strategies and drug targets.
Collapse
Affiliation(s)
- Zhihui Wang
- College of Bioengineering, Chongqing University, Chongqing 400030, China
- Key Laboratory of Biorheological Science & Technology, Ministry of Education, Chongqing University, Chongqing 400030, China
| | - Wenbin Wang
- College of Bioengineering, Chongqing University, Chongqing 400030, China
- Key Laboratory of Biorheological Science & Technology, Ministry of Education, Chongqing University, Chongqing 400030, China
| | - Qing Luo
- College of Bioengineering, Chongqing University, Chongqing 400030, China
- Key Laboratory of Biorheological Science & Technology, Ministry of Education, Chongqing University, Chongqing 400030, China
| | - Guanbin Song
- College of Bioengineering, Chongqing University, Chongqing 400030, China
- Key Laboratory of Biorheological Science & Technology, Ministry of Education, Chongqing University, Chongqing 400030, China
| |
Collapse
|
43
|
Chen YL, Chen YC, Suzuki A. ImmunoCellCycle-ID: A high-precision immunofluorescence-based method for cell cycle identification. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.14.607961. [PMID: 39185179 PMCID: PMC11343203 DOI: 10.1101/2024.08.14.607961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
The cell cycle is a fundamental process essential for cell proliferation, differentiation, and development. It consists of four major phases: G1, S, G2, and M. These phases collectively drive the reproductive cycle and are meticulously regulated by various proteins that play critical roles in both the prevention and progression of cancer. Traditional methods for studying these functions, such as flow cytometry, require a substantial number of cells to ensure accuracy. In this study, we have developed a user-friendly, immunofluorescence-based method for identifying cell cycle stages, providing single-cell resolution and precise identification of G1, early S, late S, early G2, late G2, and each sub-stage of the M phase using fluorescence microscopy. This method provides high-precision cell cycle identification and can serve as an alternative to, or in combination with, traditional flow cytometry to dissect detailed substages of the cell cycle in a variety of cell lines.
Collapse
Affiliation(s)
- Yu-Lin Chen
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Yu-Chia Chen
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Molecular and Cellular Pharmacology Graduate Program, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Aussie Suzuki
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Molecular and Cellular Pharmacology Graduate Program, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Carbone Comprehensive Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
44
|
Dutta T, Chakraborty B, Nigam A, Minocha S, Koner AL. A small-molecule probe to decipher stress-induced ER microenvironments and ER-Golgi communication. J Mater Chem B 2024; 12:7848-7857. [PMID: 38808376 DOI: 10.1039/d4tb00572d] [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: 05/30/2024]
Abstract
Cellular stress is a crucial factor in regulating and maintaining both organismal and microenvironmental homeostasis. It induces a response that also affects the micropolarity of specific cellular compartments, which is essential for early disease diagnosis. In this contribution, we present a quantitative study of micropolarity changes inside the endoplasmic reticulum (ER) during the G1/S and G2/M phases, using a biocompatible small-molecule fluorophore called ER-Oct. This probe is selectively driven to the ER by its hydrophobicity, and it has the fastest diffusion properties among a series of analogous probes. We found that induced ER stress caused cell cycle arrests leading to an increase in ER micropolarity which is well supported by lambda scanning experiments and fluorescence lifetime imaging microscopy (FLIM) as well. ER-Oct is a versatile staining agent that could effectively stain the ER in various living/fixed mammalian cells, isolated ER, Caenorhabditis elegans, and mice tissues. Furthermore, we used this probe to visualize a well-known biological event, ER to Golgi transport, by live-cell fluorescence microscopy. Our exhaustive investigation of micropolarity using ER-staining dye provides a new way to study ER stress, which could provide a deeper understanding of proteostasis in model systems and even in fixed patient samples.
Collapse
Affiliation(s)
- Tanoy Dutta
- Bionanotechnology Lab, Department of Chemistry, Indian Institute of Science Education and Research Bhopal, Bhopal Bypass Road, Bhauri, Bhopal, Madhya Pradesh - 462066, India.
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | - Barsha Chakraborty
- Bionanotechnology Lab, Department of Chemistry, Indian Institute of Science Education and Research Bhopal, Bhopal Bypass Road, Bhauri, Bhopal, Madhya Pradesh - 462066, India.
| | - Aditya Nigam
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, Hauz Khas, New Delhi - 110016, India
| | - Shilpi Minocha
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, Hauz Khas, New Delhi - 110016, India
| | - Apurba Lal Koner
- Bionanotechnology Lab, Department of Chemistry, Indian Institute of Science Education and Research Bhopal, Bhopal Bypass Road, Bhauri, Bhopal, Madhya Pradesh - 462066, India.
| |
Collapse
|
45
|
Ibrahim JN, El-Hakim S, Semaan J, Ghosn S, El Ayoubi H, Elnar AA, Tohme N, El Boustany C. Sodium Butyrate (NaB) and Sodium Propionate (NaP) Reduce Cyclin A2 Expression, Inducing Cell Cycle Arrest and Proliferation Inhibition of Different Breast Cancer Subtypes, Leading to Apoptosis. Biomedicines 2024; 12:1779. [PMID: 39200243 PMCID: PMC11351769 DOI: 10.3390/biomedicines12081779] [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: 07/15/2024] [Revised: 07/27/2024] [Accepted: 08/02/2024] [Indexed: 09/02/2024] Open
Abstract
Sodium butyrate (NaB) and sodium propionate (NaP) have recently garnered attention for their role in regulating inflammation and controlling signaling pathways of cell growth and apoptosis, potentially preventing cancer development. However, their therapeutic effect and the underlying mechanisms involved remain elusive in breast cancer. This study aims at investigating the anticancer role of NaB and NaP in different types of breast cancer by assessing their antiproliferative effect on MCF-7 and MDA-MB-231 cells (through an MTT assay), as well as their ability to alter the cell cycle and cyclin expression (using flow cytometry and RT-qPCR, respectively), and to promote apoptosis (using Annexin V-FITC conjugated and sub-G1 phase techniques). MDA-MB-231 cell proliferation was inhibited by NaB and NaP in a dose- and time-dependent manner with respective IC50 values of 2.56 mM and 6.49 mM. Treatment induced cell arrest in the G1 phase which was further supported by the significant reduction in cyclin A2 and cyclin B1 expressions. Finally, NaB, and less significantly NaP, induced apoptosis in a dose-dependent manner with higher concentrations required for MDA-MB-231 than MCF-7. Our findings elucidate the cyclin-dependent inhibitory effect of NaB and NaP on the progression of different breast cancer subtypes, thus highlighting their therapeutic potential in breast cancer.
Collapse
Affiliation(s)
- José-Noel Ibrahim
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University (LAU), Beirut 1102, Lebanon
| | - Sandy El-Hakim
- College of Engineering and Technology, American University of the Middle East, Egaila 54200, Kuwait;
| | - Josiane Semaan
- Department of Laboratory Science, Faculty of Public Health—Branch 2, Lebanese University, Fanar 2611, Lebanon; (J.S.); (S.G.); (H.E.A.); (A.A.E.); (N.T.); (C.E.B.)
| | - Stéphanie Ghosn
- Department of Laboratory Science, Faculty of Public Health—Branch 2, Lebanese University, Fanar 2611, Lebanon; (J.S.); (S.G.); (H.E.A.); (A.A.E.); (N.T.); (C.E.B.)
| | - Hiba El Ayoubi
- Department of Laboratory Science, Faculty of Public Health—Branch 2, Lebanese University, Fanar 2611, Lebanon; (J.S.); (S.G.); (H.E.A.); (A.A.E.); (N.T.); (C.E.B.)
| | - Arpiné Ardzivian Elnar
- Department of Laboratory Science, Faculty of Public Health—Branch 2, Lebanese University, Fanar 2611, Lebanon; (J.S.); (S.G.); (H.E.A.); (A.A.E.); (N.T.); (C.E.B.)
| | - Najat Tohme
- Department of Laboratory Science, Faculty of Public Health—Branch 2, Lebanese University, Fanar 2611, Lebanon; (J.S.); (S.G.); (H.E.A.); (A.A.E.); (N.T.); (C.E.B.)
| | - Charbel El Boustany
- Department of Laboratory Science, Faculty of Public Health—Branch 2, Lebanese University, Fanar 2611, Lebanon; (J.S.); (S.G.); (H.E.A.); (A.A.E.); (N.T.); (C.E.B.)
| |
Collapse
|
46
|
Sachdeva A, Roy A, Gupta MK, Mandal S. Pharmacological inhibition of protein kinase D2/Aurora kinase A signalling axis suppresses G2/M cell cycle progression and proliferation of epithelial ovarian cancer cells. Pathol Res Pract 2024; 260:155390. [PMID: 38878668 DOI: 10.1016/j.prp.2024.155390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 05/29/2024] [Accepted: 06/05/2024] [Indexed: 08/09/2024]
Abstract
Epithelial ovarian cancer (EOC) is the deadliest gynecological malignancy with poor prognosis and patient survival outcome. Protein kinase D2 (PKD2) belongs to Ca++/calmodulin-dependent serine/threonine kinase family and its aberrant expression is associated with many cellular and physiological functions associated with tumorigenesis including cell proliferation. We show that PKD2 is activated during G2/M cell cycle transition and its catalytic inactivation by small molecule inhibitor CRT0066101 or genetic knockdown caused suppression of EOC cell proliferation followed by a delay into mitotic entry. Our RNASeq analysis of PKD2-inactivated EOC cells revealed significant downregulation of genes associated with cell cycle including Aurora kinase A, a critical mitotic regulator. Mechanistically, PKD2 positively regulated Aurora kinase A stability at both transcriptional and post-translational levels by interfering with the function of Fbxw7, drove G2/M cell cycle transition and EOC cell proliferation. Moreover, pharmacological inhibition of Aurora kinase A by small molecule CD532 or its shRNA-mediated genetic knockdown suppressed EOC cell proliferation, induced G2/M cell cycle arrest and mitotic catastrophe followed by apoptosis. Taken together, our results indicated that PKD2 positively regulates Aurora kinase A during G2/M cell cycle entry and pharmacological targeting of PKD2/Aurora kinase A signalling axis could serve as a novel therapeutic intervention against a lethal pathology like EOC.
Collapse
Affiliation(s)
- Abha Sachdeva
- Amity Institute of Molecular Medicine & Stem Cell Research, Amity University, Sector 125, Noida, Uttar Pradesh 201303, India
| | - Adhiraj Roy
- Amity Institute of Molecular Medicine & Stem Cell Research, Amity University, Sector 125, Noida, Uttar Pradesh 201303, India.
| | - Manoj Kumar Gupta
- Amity Institute of Molecular Medicine & Stem Cell Research, Amity University, Sector 125, Noida, Uttar Pradesh 201303, India
| | - Supratim Mandal
- Department of Microbiology, University of Kalyani, Kalyani, Nadia, West Bengal 741235, India
| |
Collapse
|
47
|
El Mahi Y, Nizami ZN, Wali AF, Al Neyadi A, Magramane M, Al Azzani M, Arafat K, Attoub S, Eid AH, Iratni R. Rhus coriaria induces autophagic and apoptotic cell death in pancreatic cancer cells. Front Pharmacol 2024; 15:1412565. [PMID: 39139643 PMCID: PMC11319293 DOI: 10.3389/fphar.2024.1412565] [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: 04/05/2024] [Accepted: 07/10/2024] [Indexed: 08/15/2024] Open
Abstract
Background:Pancreatic cancer is a leading cause of cancer-related mortality worldwide with increasing global incidence. We previously reported the anticancer effect of Rhus coriaria ethanolic extract (RCE) in triple negative breast and colon cancer cells. Herein, we investigated the anticancer effect of RCE on human pancreatic cancer cells. Methods: Cell viability was measured using Cell Titer-Glo and staining of viable and dead cells based on differential permeability to two DNA binding dyes. Cell cycle distribution and annexin V staining was carried out in Muse cell analyzer. Protein level was determined by Western blot. Tumor growth was assessed by in ovo chick embryo chorioallantoic membrane assay. Results: We found that RCE significantly inhibited the viability and colony growth of pancreatic cancer cells (Panc-1, Mia-PaCa-2, S2-013, AsPC-1). The antiproliferative effects of RCE in pancreatic cancer cells (Panc-1 and Mia-PaCa-2) were mediated through induction of G1 cell cycle arrest, Beclin-1-independent autophagy, and apoptosis. RCE activated both the extrinsic and intrinsic pathways of apoptosis and regulated the Bax/Bcl-2 apoptotic switch. Mechanistically, we found that RCE inhibited the AKT/mTOR pathway, downstream of which, inactivation of the cell cycle regulator p70S6K and downregulation of the antiapoptotic protein survivin was observed. Additionally, we found that RCE-induced autophagy preceded apoptosis. Further, we confirmed the anticancer effect of RCE in a chick embryo xenograft model and found that RCE inhibited the growth of pancreatic cancer xenografts without affecting embryo survival. Conclusion: Collectively, our findings demonstrate that Rhus coriaria exerts potent anti-pancreatic cancer activity though cell cycle impairment, autophagy, and apoptosis, and is hence a promising source of anticancer phytochemicals.
Collapse
Affiliation(s)
- Yassine El Mahi
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Zohra Nausheen Nizami
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Adil Farooq Wali
- Department of Pharmaceutical Chemistry, RAK College of Pharmacy, RAK Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates
| | - Aysha Al Neyadi
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Mohamed Magramane
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Mazoun Al Azzani
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Kholoud Arafat
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Samir Attoub
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Ali H. Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Rabah Iratni
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| |
Collapse
|
48
|
Sutter SO, Tobler K, Seyffert M, Lkharrazi A, Zöllig J, Schraner EM, Vogt B, Büning H, Fraefel C. Interferon-γ inducible factor 16 (IFI16) restricts adeno-associated virus type 2 (AAV2) transduction in an immune-modulatory independent way. J Virol 2024; 98:e0011024. [PMID: 38837381 PMCID: PMC11338077 DOI: 10.1128/jvi.00110-24] [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: 01/20/2024] [Accepted: 04/28/2024] [Indexed: 06/07/2024] Open
Abstract
We determined the transcription profile of adeno-associated virus type 2 (AAV2)-infected primary human fibroblasts. Subsequent analysis revealed that cells respond to AAV infection through changes in several significantly affected pathways, including cell cycle regulation, chromatin modulation, and innate immune responses. Various assays were performed to validate selected differentially expressed genes and to confirm not only the quality but also the robustness of the raw data. One of the genes upregulated in AAV2-infected cells was interferon-γ inducible factor 16 (IFI16). IFI16 is known as a multifunctional cytosolic and nuclear innate immune sensor for double-stranded as well as single-stranded DNA, exerting its effects through various mechanisms, such as interferon response, epigenetic modifications, or transcriptional regulation. IFI16 thereby constitutes a restriction factor for many different viruses among them, as shown here, AAV2 and thereof derived vectors. Indeed, the post-transcriptional silencing of IFI16 significantly increased AAV2 transduction efficiency, independent of the structure of the virus/vector genome. We also show that IFI16 exerts its inhibitory effect on AAV2 transduction in an immune-modulatory independent way by interfering with Sp1-dependent transactivation of wild-type AAV2 and AAV2 vector promoters. IMPORTANCE Adeno-associated virus (AAV) vectors are among the most frequently used viral vectors for gene therapy. The lack of pathogenicity of the parental virus, the long-term persistence as episomes in non-proliferating cells, and the availability of a variety of AAV serotypes differing in their cellular tropism are advantageous features of this biological nanoparticle. To deepen our understanding of virus-host interactions, especially in terms of antiviral responses, we present here the first transcriptome analysis of AAV serotype 2 (AAV2)-infected human primary fibroblasts. Our findings indicate that interferon-γ inducible factor 16 acts as an antiviral factor in AAV2 infection and AAV2 vector-mediated cell transduction in an immune-modulatory independent way by interrupting the Sp1-dependent gene expression from viral or vector genomes.
Collapse
Affiliation(s)
| | - Kurt Tobler
- Institute of Virology,
University of Zurich,
Zurich, Switzerland
| | - Michael Seyffert
- Institute of Virology,
University of Zurich,
Zurich, Switzerland
| | - Anouk Lkharrazi
- Institute of Virology,
University of Zurich,
Zurich, Switzerland
| | - Joël Zöllig
- Institute of Virology,
University of Zurich,
Zurich, Switzerland
| | | | - Bernd Vogt
- Institute of Virology,
University of Zurich,
Zurich, Switzerland
| | - Hildegard Büning
- Institute of
Experimental Hematology, Hannover Medical
School, Hannover,
Germany
| | - Cornel Fraefel
- Institute of Virology,
University of Zurich,
Zurich, Switzerland
| |
Collapse
|
49
|
Kawaue H, Matsubara T, Nagano K, Ikedo A, Rojasawasthien T, Yoshimura A, Nakatomi C, Imai Y, Kakuta Y, Addison WN, Kokabu S. KIF22 regulates mitosis and proliferation of chondrocyte cells. iScience 2024; 27:110151. [PMID: 38989461 PMCID: PMC11233920 DOI: 10.1016/j.isci.2024.110151] [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: 09/04/2023] [Revised: 03/12/2024] [Accepted: 05/28/2024] [Indexed: 07/12/2024] Open
Abstract
Point mutations in KIF22 have been linked to spondyloepimetaphyseal dysplasia with joint laxity, type 2 (SEMDJL2). Skeletal features of SEMDJL2 include short stature and joint laxity. Mechanisms underlying these limb abnormalities are unknown. Here in this manuscript, we have investigated the function of KIF22 in chondrocytes. Quantitative PCR and immunostaining revealed that Kif22 was highly expressed in proliferating-zone growth-plate chondrocytes. Kif22 knockdown resulted in defective mitotic spindle formation and reduced cell proliferation. Forced expression of SEMDJL-associated mutant Kif22 constructs likewise induced abnormal mitotic spindle morphology and reduced proliferation. Mice expressing a KIF22 truncation mutant had shorter growth plates and shorter tibial bones compared to wild-type mice. These results suggest that KIF22 regulates mitotic spindle formation in proliferating chondrocytes thereby linking the stunted longitudinal bone growth observed in SEMDJL2 to failures of chondrocyte division.
Collapse
Affiliation(s)
- Hiroka Kawaue
- Division of Molecular Signaling and Biochemistry, Department of Health Improvement, Kyushu Dental University, Kitakyushu, Fukuoka 803-8580, Japan
| | - Takuma Matsubara
- Division of Molecular Signaling and Biochemistry, Department of Health Improvement, Kyushu Dental University, Kitakyushu, Fukuoka 803-8580, Japan
| | - Kenichi Nagano
- Department of Oral Pathology, Institute of Biomedical Sciences, Nagasaki University, Nagasaki, Nagasaki 852-8588, Japan
| | - Aoi Ikedo
- Division of Integrative Pathophysiology, Proteo-Science Center, Ehime University, Toon, Ehime 791-0295, Japan
| | - Thira Rojasawasthien
- Division of Molecular Signaling and Biochemistry, Department of Health Improvement, Kyushu Dental University, Kitakyushu, Fukuoka 803-8580, Japan
| | - Anna Yoshimura
- Division of Molecular Signaling and Biochemistry, Department of Health Improvement, Kyushu Dental University, Kitakyushu, Fukuoka 803-8580, Japan
| | - Chihiro Nakatomi
- Division of Physiology, Department of Health Improvement, Kyushu Dental University, Manazuru, Kitakyushu, Fukuoka 803-8580, Japan
| | - Yuuki Imai
- Division of Integrative Pathophysiology, Proteo-Science Center, Ehime University, Toon, Ehime 791-0295, Japan
| | - Yoshimitsu Kakuta
- Laboratory of Structural Biology, Graduate School of Systems Life Sciences, Kyushu University, Fukuoka, Fukuoka 819-0395, Japan
| | - William N. Addison
- Division of Molecular Signaling and Biochemistry, Department of Health Improvement, Kyushu Dental University, Kitakyushu, Fukuoka 803-8580, Japan
| | - Shoichiro Kokabu
- Division of Molecular Signaling and Biochemistry, Department of Health Improvement, Kyushu Dental University, Kitakyushu, Fukuoka 803-8580, Japan
| |
Collapse
|
50
|
Jagtap YA, Kumar P, Dubey AR, Kinger S, Choudhary A, Karmakar S, Lal G, Kumar A, Kumar A, Prasad A, Mishra A. Acetaminophen induces mitochondrial apoptosis through proteasome dysfunctions. Life Sci 2024; 349:122732. [PMID: 38768775 DOI: 10.1016/j.lfs.2024.122732] [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: 11/15/2023] [Revised: 03/12/2024] [Accepted: 05/15/2024] [Indexed: 05/22/2024]
Abstract
Acetaminophen is a known antipyretic and non-opioid analgesic for mild pain and fever. Numerous studies uncover their hidden chemotherapeutics applications, including chronic cancer pain management. Acetaminophen also represents an anti-proliferative effect in some cancer cells. Few studies also suggest that the use of Acetaminophen can trigger apoptosis and impede cellular growth. However, Acetaminophen's molecular potential and precise mechanism against improper cellular proliferation and use as an effective anti-proliferative agent still need to be better understood. Here, our current findings show that Acetaminophen induces proteasomal dysfunctions, resulting in aberrant protein accumulation and mitochondrial abnormalities, and consequently induces cell apoptosis. We observed that the Acetaminophen treatment leads to improper aggregation of ubiquitylated expanded polyglutamine proteins, which may be due to the dysfunctions of proteasome activities. Our in-silico analysis suggests the interaction of Acetaminophen and proteasome. Furthermore, we demonstrated the accumulation of proteasome substrates and the depletion of proteasome activities after treating Acetaminophen in cells. Acetaminophen induces proteasome dysfunctions and mitochondrial abnormalities, leading to pro-apoptotic morphological changes and apoptosis successively. These results suggest that Acetaminophen can induce cell death and may retain a promising anti-proliferative effect. These observations can open new possible molecular strategies in the near future for developing and designing specific and effective proteasome inhibitors, which can be helpful in conjugation with other anti-tumor drugs for their better efficiency.
Collapse
Affiliation(s)
- Yuvraj Anandrao Jagtap
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Rajasthan, 342037, India
| | - Prashant Kumar
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Rajasthan, 342037, India
| | - Ankur Rakesh Dubey
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Rajasthan, 342037, India
| | - Sumit Kinger
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Rajasthan, 342037, India
| | - Akash Choudhary
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Rajasthan, 342037, India
| | - Surojit Karmakar
- National Centre for Cell Science (NCCS), Ganeshkhind, Pune, Maharashtra, 411007, India
| | - Girdhari Lal
- National Centre for Cell Science (NCCS), Ganeshkhind, Pune, Maharashtra, 411007, India
| | - Awanish Kumar
- Department of Biotechnology, National Institute of Technology, Raipur, Chhattisgarh, 492010, India
| | - Amit Kumar
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, Indore, Madhya Pradesh, 453552, India
| | - Amit Prasad
- School of Biosciences and Bioengineering, Indian Institute of Technology Mandi, Mandi, Himachal Pradesh, 175005, India
| | - Amit Mishra
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Rajasthan, 342037, India.
| |
Collapse
|