1
|
Wei C, Liu X, Miao Z, Zhang H, Wang Y, Qi G. TWEAK/Fn14 axis may promote vascular smooth muscle cell senescence via p38 signaling pathway: preliminary evidence. Future Sci OA 2025; 11:2455906. [PMID: 39840833 PMCID: PMC11756581 DOI: 10.1080/20565623.2025.2455906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 12/12/2024] [Indexed: 01/23/2025] Open
Abstract
AIM The primary objective of this study is to investigate the impact of tumor necrosis factor-like weak inducer of apoptosis (TWEAK) and its functional receptor, fibroblast growth factor-inducible 14 (Fn14), on the process of vascular smooth muscle cell (VSMC) senescence. METHODS Rat arterial VSMCs were cultured with angiotensin II to establish a model of premature senescence. The effects of TWEAK and Fn14 on senescent VSMCs were evaluated. Additionally, the role of p38 phosphorylation pathway in the effect of TWEAK on VSMCs senescence was assessed. RESULTS Expressions of TWEAK and Fn14 were significantly elevated in senescent VSMCs. TWEAK activated the p38 phosphorylation pathway and promoted the SA-β-gal staining and P53 expression. CONCLUSION These preliminary findings suggest that the TWEAK/Fn14 axis may play a crucial role in promoting VSMC senescence.
Collapse
Affiliation(s)
- Chunyang Wei
- Department of Gerontology, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Xiaoying Liu
- Department of Gerontology, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Zhuang Miao
- Department of Anesthesiology, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Hua Zhang
- Department of Gerontology, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Yanfu Wang
- Department of Gerontology, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Guoxian Qi
- Department of Gerontology, the First Affiliated Hospital, China Medical University, Shenyang, China
| |
Collapse
|
2
|
Antonyan L, Zhang X, Ni A, Peng H, Alsuwaidi S, Fleming P, Zhang Y, Semenak A, Macintosh J, Wu H, Hettige NC, Jefri M, Ernst C. Reciprocal and non-reciprocal effects of clinically relevant SETBP1 protein dosage changes. Hum Mol Genet 2025:ddaf003. [PMID: 39825586 DOI: 10.1093/hmg/ddaf003] [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: 04/23/2024] [Revised: 12/17/2024] [Accepted: 01/06/2025] [Indexed: 01/20/2025] Open
Abstract
Many genes in the human genome encode proteins that are dosage sensitive, meaning they require protein levels within a narrow range to properly execute function. To investigate if clinically relevant variation in protein levels impacts the same downstream pathways in human disease, we generated cell models of two SETBP1 syndromes: Schinzel-Giedion Syndrome (SGS) and SETBP1 haploinsufficiency disease (SHD), where SGS is caused by too much protein, and SHD is caused by not enough SETBP1. Using patient and sex-matched healthy first-degree relatives from both SGS and SHD SETBP1 cases, we assessed how SETBP1 protein dosage affects downstream pathways in human forebrain progenitor cells. We find that extremes of SETBP1 protein dose reciprocally influence important signalling molecules such as AKT, suggesting that the SETBP1 protein operates within a narrow dosage range and that extreme doses are detrimental. We identified SETBP1 nuclear bodies as interacting with the nuclear lamina and suggest that SETBP1 may organize higher order chromatin structure via links to the nuclear envelope. SETBP1 protein doses may exert significant influence on global gene expression patterns via these SETBP1 nuclear bodies. This work provides evidence for the importance of SETBP1 protein dose in human brain development, with implications for two neurodevelopmental disorders.
Collapse
Affiliation(s)
- Lilit Antonyan
- Department of Human Genetics, McGill University, 3666 McTavish Street, Montreal, QC H3A 1Y2, Canada
- Rare Neurodevelopmental Disorders Laboratory, Montreal Neurological Institute, 3801 University Street, Montreal, QC H3A 2B4, Canada
| | - Xin Zhang
- Rare Neurodevelopmental Disorders Laboratory, Montreal Neurological Institute, 3801 University Street, Montreal, QC H3A 2B4, Canada
| | - Anjie Ni
- Department of Human Genetics, McGill University, 3666 McTavish Street, Montreal, QC H3A 1Y2, Canada
- Rare Neurodevelopmental Disorders Laboratory, Montreal Neurological Institute, 3801 University Street, Montreal, QC H3A 2B4, Canada
| | - Huashan Peng
- Rare Neurodevelopmental Disorders Laboratory, Montreal Neurological Institute, 3801 University Street, Montreal, QC H3A 2B4, Canada
| | - Shaima Alsuwaidi
- Rare Neurodevelopmental Disorders Laboratory, Montreal Neurological Institute, 3801 University Street, Montreal, QC H3A 2B4, Canada
- Integrated Program in Neuroscience, McGill University, 1033 Pine Ave. W., Montreal, QC H3A 1A1, Canada
| | - Peter Fleming
- Rare Neurodevelopmental Disorders Laboratory, Montreal Neurological Institute, 3801 University Street, Montreal, QC H3A 2B4, Canada
- Integrated Program in Neuroscience, McGill University, 1033 Pine Ave. W., Montreal, QC H3A 1A1, Canada
| | - Ying Zhang
- Rare Neurodevelopmental Disorders Laboratory, Montreal Neurological Institute, 3801 University Street, Montreal, QC H3A 2B4, Canada
| | - Amelia Semenak
- Rare Neurodevelopmental Disorders Laboratory, Montreal Neurological Institute, 3801 University Street, Montreal, QC H3A 2B4, Canada
- Integrated Program in Neuroscience, McGill University, 1033 Pine Ave. W., Montreal, QC H3A 1A1, Canada
| | - Julia Macintosh
- Rare Neurodevelopmental Disorders Laboratory, Montreal Neurological Institute, 3801 University Street, Montreal, QC H3A 2B4, Canada
- Integrated Program in Neuroscience, McGill University, 1033 Pine Ave. W., Montreal, QC H3A 1A1, Canada
| | - Hanrong Wu
- Rare Neurodevelopmental Disorders Laboratory, Montreal Neurological Institute, 3801 University Street, Montreal, QC H3A 2B4, Canada
| | - Nuwan C Hettige
- Department of Human Genetics, McGill University, 3666 McTavish Street, Montreal, QC H3A 1Y2, Canada
- Rare Neurodevelopmental Disorders Laboratory, Montreal Neurological Institute, 3801 University Street, Montreal, QC H3A 2B4, Canada
| | - Malvin Jefri
- Rare Neurodevelopmental Disorders Laboratory, Montreal Neurological Institute, 3801 University Street, Montreal, QC H3A 2B4, Canada
- Integrated Program in Neuroscience, McGill University, 1033 Pine Ave. W., Montreal, QC H3A 1A1, Canada
| | - Carl Ernst
- Department of Human Genetics, McGill University, 3666 McTavish Street, Montreal, QC H3A 1Y2, Canada
- Rare Neurodevelopmental Disorders Laboratory, Montreal Neurological Institute, 3801 University Street, Montreal, QC H3A 2B4, Canada
- Integrated Program in Neuroscience, McGill University, 1033 Pine Ave. W., Montreal, QC H3A 1A1, Canada
| |
Collapse
|
3
|
Du Y, Hu P, Ding X, Wang D, Luo J, Le S, Ren L, Chen M, Ye P, Xia J. Deficiency of ATF3 facilitates both angiotensin II-induced and spontaneously formed aortic aneurysm and dissection development by activating cGAS-STING pathway. Clin Transl Med 2025; 15:e70147. [PMID: 39731276 DOI: 10.1002/ctm2.70147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 12/03/2024] [Accepted: 12/09/2024] [Indexed: 12/29/2024] Open
Abstract
BACKGROUND Sporadic aortic aneurysm and dissection (AAD) is a critical condition characterised by the progressive loss of vascular smooth muscle cells (VSMCs) and the breakdown of the extracellular matrix. However, the molecular mechanisms responsible for the phenotypic switch and loss of VSMCs in AAD are not fully understood. METHODS AND RESULTS In this study, we employed a discovery-driven, unbiased approach. This approach encourages us to explore the unknown functions of activating transcription factor 3 (ATF3) rather than merely confirming existing hypotheses, while no assumptions were made about ATF3 prior to the experiments. We ensured the unbiased nature of our assessment by conducting morphological evaluations with two independent observers in a blinded manner. We identified elevated expression of ATF3 in both human sporadic AAD tissues and mouse AAD models. VSMC-specific ATF3 conditional knockout (Atf3 cKO) mice showed notable enlargement, dissection and rupture in both thoracic and abdominal aortic regions after exposure to Ang II. Interestingly, older Atf3 cKO mice exhibited spontaneous aortic dissections and senescence of the aortic wall. Mechanistically, ATF3 deficiency led to the degradation of P21 through ubiquitination. Impaired DNA repair in VSMCs resulted in micronuclei formation in the cytoplasm, activating the cyclicGMP-AMP synthase- stimulator of interferon genes (cGAS-STING) pathway and inducing VSMC phenotypic switching and apoptosis. Finally, both pharmacological complementation of P21 function and knockdown of STING expression alleviated ATF3 deficiency-induced AAD. CONCLUSIONS Our study indicates that ATF3 is essential for genomic DNA stability in VSMCs through the P21-cGAS-STING pathway, suggesting that enhancing ATF3 expression in VSMCs could help prevent sporadic AAD. KEY POINTS ATF3 deficiency led to degradation of P21 through ubiquitination, which abolished the G1 phase arrest. VSMCs had no time window to repair the damaged DNA, leading to generation of micronuclei in cytoplasm. Cytoplasmic micronuclei facilitating the activation of cGAS-STING pathway, thus inducing the phenotypic switch and apoptosis of VSMCs.
Collapse
Affiliation(s)
- Yifan Du
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Poyi Hu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiangchao Ding
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Dashuai Wang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Henan Province, China
| | - Jingjing Luo
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sheng Le
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Lingyun Ren
- Department of Anesthesiology, Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Manhua Chen
- Department of Cardiology, Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ping Ye
- Department of Cardiology, Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiahong Xia
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
4
|
Lin L, Ding J, Liu S, Liu C, Li Q, Gao X, Niu Y, Tong WM. Protein Phosphatase 2ACα Regulates ATR-Mediated Endogenous DNA Damage Response Against Microcephaly. Mol Neurobiol 2025; 62:1266-1281. [PMID: 38976130 DOI: 10.1007/s12035-024-04301-6] [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: 11/29/2022] [Accepted: 06/11/2024] [Indexed: 07/09/2024]
Abstract
Protein phosphatase 2A (PP2A) is an abundant heterotrimeric holoenzyme in eukaryotic cells coordinating with specific kinases to regulate spatial-temporal protein dephosphorylation in various biological processes. However, the function of PP2A in cortical neurogenesis remains largely unknown. Here, we report that neuronal-specific deletion of Pp2acα in mice displayed microcephaly, with significantly smaller brains and defective learning and memory ability. Mechanistically, neuronal Pp2acα deficiency resulted in elevated endogenous DNA damage and activation of ATR/CHK1 signaling. It was further induced by the loss of direct interaction between PP2AC and ATR as well as the function of PP2AC to dephosphorylate ATR. Importantly, ATR/CHK1 signaling dysregulation altered both the expression and activity of several critical downstream factors including P53, P21, Bcl2, and Bax, which led to decreased proliferation of cortical progenitor cells and increased apoptosis in developing cortical neurons. Taken together, our results indicate an essential function of PP2ACα in endogenous DNA damage response-mediated ATR signaling during neurogenesis, and defective PP2ACα in neurons contributes to microcephaly.
Collapse
Affiliation(s)
- Lin Lin
- Department of Pathology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Jing Ding
- Department of Pathology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Simeng Liu
- Department of Pathology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
- Department of Pathology, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Chunying Liu
- Department of Pathology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Qing Li
- Department of Pathology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Xiang Gao
- Model Animal Research Center and MOE Key Laboratory of Model Animal for Disease Study, Nanjing University, Nanjing, China
| | - Yamei Niu
- Department of Pathology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China.
- Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Wei-Min Tong
- Department of Pathology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China.
- Molecular Pathology Research Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
5
|
Zhang K, Zheng X, Sun Y, Feng X, Wu X, Liu W, Gao C, Yan Y, Tian W, Wang Y. TOP2A modulates signaling via the AKT/mTOR pathway to promote ovarian cancer cell proliferation. Cancer Biol Ther 2024; 25:2325126. [PMID: 38445610 PMCID: PMC10936659 DOI: 10.1080/15384047.2024.2325126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 02/26/2024] [Indexed: 03/07/2024] Open
Abstract
Ovarian cancer (OC) is a form of gynecological malignancy that is associated with worse patient outcomes than any other cancer of the female reproductive tract. Topoisomerase II α (TOP2A) is commonly regarded as an oncogene that is associated with malignant disease progression in a variety of cancers, its mechanistic functions in OC have yet to be firmly established. We explored the role of TOP2A in OC through online databases, clinical samples, in vitro and in vivo experiments. And initial analyses of public databases revealed high OC-related TOP2A expression in patient samples that was related to poorer prognosis. This was confirmed by clinical samples in which TOP2A expression was elevated in OC relative to healthy tissue. Kaplan-Meier analyses further suggested that higher TOP2A expression levels were correlated with worse prognosis in OC patients. In vitro, TOP2A knockdown resulted in the inhibition of OC cell proliferation, with cells entering G1 phase arrest and undergoing consequent apoptotic death. In rescue assays, TOP2A was confirmed to regulate cell proliferation and cell cycle through AKT/mTOR pathway activity. Mouse model experiments further affirmed the key role that TOP2A plays as a driver of OC cell proliferation. These data provide strong evidence supporting TOP2A as an oncogenic mediator and prognostic biomarker related to OC progression and poor outcomes. At the mechanistic level, TOP2A can control tumor cell growth via AKT/mTOR pathway modulation. These preliminary results provide a foundation for future research seeking to explore the utility of TOP2A inhibitor-based combination treatment regimens in platinum-resistant recurrent OC patients.
Collapse
Affiliation(s)
- Kaiwen Zhang
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Xingyu Zheng
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Yiqing Sun
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Xinyu Feng
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Xirong Wu
- Department of Gynecology and Obstetrics, Affiliated Hospital of Nantong University, Nantong, China
| | - Wenlu Liu
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Chao Gao
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Ye Yan
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Wenyan Tian
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Yingmei Wang
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
6
|
Zhong Q, Huang Y, Sha Y, Wei Q, Long K, Xiao J, Liu Z, Wei X. Halobenzoquinone-induced potential carcinogenicity associated with p53-mediated cell cycle pathway. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 363:125230. [PMID: 39489323 DOI: 10.1016/j.envpol.2024.125230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 10/13/2024] [Accepted: 10/30/2024] [Indexed: 11/05/2024]
Abstract
2,6-Dibromo-1,4-benzoquinone (2,6-DBBQ) and 2,6-dichloro-1,4-benzoquinone (2,6-DCBQ), two emerging halobenzoquinones (HBQs), have the highest detection frequencies and levels in drinking water among all HBQs. They are more toxic than the regulated disinfection byproducts. Quantitative structure toxicity relationship analysis predicted that HBQs are a class of potential bladder carcinogens. However, direct experimental evidence for the carcinogenicity of 2,6-DBBQ and 2,6-DCBQ is lacking and the associated toxicity mechanisms remain unclear. In this study, we confirmed the potential carcinogenicity of 2,6-DBBQ and 2,6-DCBQ using an in vitro malignant transformation assay, evaluated their cytotoxicity and genotoxicity, and investigated their toxicity mechanisms. The results showed that 2,6-DBBQ and 2,6-DCBQ significantly decreased the viability of human uroepithelial SV-HUC-1 cells and induced DNA damage in SV-HUC-1 cells, and chromosomal damage in HepG2 cells, and malignant transformation of SV-HUC-1 cells. Moreover, transcriptome sequencing revealed that 2,6-DBBQ and 2,6-DCBQ activated the p53-mediated cell cycle pathway in bladder cancer. In the p53-mediated cell cycle pathway, 2,6-DBBQ and 2,6-DCBQ induced cell cycle arrest at the S phase by downregulating p53 and upregulating p21. Additionally, 2,6-DBBQ and 2,6-DCBQ may have produced excessive reactive oxygen species, damaging DNA and chromosomes. These results not only first confirm the potential carcinogenicity of 2,6-DBBQ and 2,6-DCBQ but also provide an important reference for exploring the cytotoxicity and genotoxicity mechanisms of these HBQs.
Collapse
Affiliation(s)
- Qing Zhong
- Department of Occupational and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Yuwen Huang
- Department of Occupational and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Yujie Sha
- Department of Occupational and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Qiuyan Wei
- Department of Occupational and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Kunling Long
- Department of Occupational and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Jingyi Xiao
- Department of Occupational and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Zhanmou Liu
- Department of Occupational and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Xiao Wei
- Department of Occupational and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi, 530021, China.
| |
Collapse
|
7
|
Oshiiwa B, da Silva AP, Alves GR, Filho VC, Niero R, O'Neill de Mascarenhas Gaivão I, de Oliveira LM, de Lima LVA, Mantovani MS, Maistro EL. Risk assessment of 2β,3β-19α-trihydroxyursolic acid from Rubus imperialis (Rosaceae) in HepG2/C3A cells via genotoxicity, metabolism, and cell growth. J Appl Toxicol 2024; 44:1886-1896. [PMID: 39128859 DOI: 10.1002/jat.4684] [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: 06/04/2024] [Revised: 07/24/2024] [Accepted: 07/28/2024] [Indexed: 08/13/2024]
Abstract
Rubus imperialis (Rosaceae) is a Brazilian medicinal plant that already exhibited therapeutical perspectives. However, previous studies revealed cellular and/or genetic toxicity of extracts from aerial parts of this plant, as well as other species of the Rubus genus. Being 2β,3β-19α-trihydroxyursolic acid (2B) one of the major compounds of this plant, with proven pharmacological effect, it is important to investigate the biosafety of this isolated compound. Therefore, in the present study, (2B) was tested by several cytogenotoxic endpoints up to 20 μg/ml in human hepatoma HepG2/C3A cells. The test compound did not produce any decreased cell viability, DNA damage, chromosomal mutations, cell cycle changes, or apoptotic effects in the tested cells. Additionally, RT-qPCR analysis revealed the downregulation of CYP3A4 (metabolism), M-TOR (cell death), and CDKN1A (cell cycle) genes. Under the experimental conditions used, the 2B compound did not show cytogenotoxic activity after a single exposure to HepG2/C3A human cells.
Collapse
Affiliation(s)
- Bruna Oshiiwa
- Faculty of Philosophy and Sciences, Speech and Hearing Therapy Department, São Paulo State University (UNESP), Marília, São Paulo State, Brazil
| | - Aline Pereira da Silva
- Postgraduate Program in Health and Aging, Marilia Medical School (FAMEMA), Marilia, São Paulo State, Brazil
| | - Greice Rafaele Alves
- Programa de Pós-Graduação em Ciências Farmacêuticas e Núcleo de Investigações Químico-Farmacêuticas (NIQFAR), Vale do Itajaí University (UNIVALI), Itajaí, Santa Catarina State, Brazil
| | - Valdir Cechinel Filho
- Programa de Pós-Graduação em Ciências Farmacêuticas e Núcleo de Investigações Químico-Farmacêuticas (NIQFAR), Vale do Itajaí University (UNIVALI), Itajaí, Santa Catarina State, Brazil
| | - Rivaldo Niero
- Programa de Pós-Graduação em Ciências Farmacêuticas e Núcleo de Investigações Químico-Farmacêuticas (NIQFAR), Vale do Itajaí University (UNIVALI), Itajaí, Santa Catarina State, Brazil
| | | | | | | | - Mário Sérgio Mantovani
- Department of General Biology, Londrina State University (UEL), Londrina, Paraná State, Brazil
| | - Edson Luis Maistro
- Faculty of Philosophy and Sciences, Speech and Hearing Therapy Department, São Paulo State University (UNESP), Marília, São Paulo State, Brazil
- Postgraduate Program in Health and Aging, Marilia Medical School (FAMEMA), Marilia, São Paulo State, Brazil
| |
Collapse
|
8
|
Erdogan MK, Sever A, Gundogdu R, Toy Y, Gecibesler IH, Yapar Y, Behcet L, Zengin G. Verbascum gimgimense an Endemic Turkish Plant: Evaluation of In Vitro Anticancer, Antioxidant, Enzyme Inhibitory Activities, and Phytochemical Profile. Cell Biochem Funct 2024; 42:e70023. [PMID: 39632482 DOI: 10.1002/cbf.70023] [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: 08/23/2024] [Revised: 10/21/2024] [Accepted: 11/14/2024] [Indexed: 12/07/2024]
Abstract
The Verbascum genus has gained significant attention in the pharmaceutical field, particularly in recent years, due to its valuable medicinal properties, which are well-recognized in complementary and alternative medicine. Certain species within this genus contain essential compounds and exhibit a wide range of therapeutic activities. In this study, the ethanolic extract of Verbascum gimgimense (VG) was analyzed for its cytotoxic, apoptotic, antioxidant, and enzyme inhibitory properties, as well as its phenolic and lipophilic compounds. The phenolic compounds in the extract were identified using Exactive Plus Orbitrap HPLC-HRMS, while the lipophilic components were characterized by GC-MS analysis. The Neutral Red Uptake (NRU) cell viability assay and colony formation assay were performed to assess the antiproliferative and anti-colony survival effects of VG on the A549 human lung adenocarcinoma cell line. Additionally, a wound healing assay measured cell migration, and the apoptotic process was evaluated using Caspase-3 ELISA and acridine orange/ethidium bromide staining. Protein expression levels were determined by western blot analysis. DPPH, ABTS FRAP, and CUPRAC assays were used to determine free radical scavenging, reducing power, and metal chelating activities, respectively. VG was rich in dominant phenolic components, including benzoic acid (6.809 mg/g extract), phloretic acid (1.279 mg/g extract), luteolin 7-rutinoside (2.799 mg/g extract), luteoloside (3.300 mg/g extract), kuromanine (3.456 mg/g extract), and rutin hydrate (2.015 mg/g extract). Major fatty acids identified in VG included palmitic acid (17.3%), stearic acid (2.99%), linoleic acid (9.44%), and α-linolenic acid (26.48%). VG treatment significantly reduced colony formation ability, decreased wound closure, and increased both apoptotic cell count and caspase-3 activity compared to the control group. Protein levels of c-PARP, p53, and p21 were substantially elevated compared to controls. In addition to its strong free radical scavenging, reducing power and metal chelating activity, VG exhibited strong inhibitory effects on α-amylase, α-glucosidase, AChE, BChE, and tyrosinase. Our study demonstrates that VG possesses antiproliferative, apoptotic, antioxidant, and enzyme-inhibitory properties. V. gimgimense emerges as a promising natural antioxidant source with potentially significant regulatory effects on key enzymes and proteins, which could contribute to managing various human diseases and inspire the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Mehmet Kadir Erdogan
- Department of Molecular Biology and Genetics, Faculty of Arts and Sciences, Bingol University, Bingol, Türkiye
| | - Aydın Sever
- Department of Pharmacy Services, Vocational School of Health Services, Bingol University, Bingol, Türkiye
| | - Ramazan Gundogdu
- Department of Pharmacy Services, Vocational School of Health Services, Bingol University, Bingol, Türkiye
| | - Yusuf Toy
- Department of Molecular Biology and Genetics, Faculty of Arts and Sciences, Bingol University, Bingol, Türkiye
| | - Ibrahim Halil Gecibesler
- Department of Occupational Health and Safety, Faculty of Health Science, Bingol University, Bingol, Türkiye
| | - Yakup Yapar
- Department of Molecular Biology and Genetics, Faculty of Arts and Sciences, Bingol University, Bingol, Türkiye
| | - Lutfi Behcet
- Department of Molecular Biology and Genetics, Faculty of Arts and Sciences, Bingol University, Bingol, Türkiye
| | - Gokhan Zengin
- Department of Biology, Science Faculty, Selcuk University, Konya, Türkiye
| |
Collapse
|
9
|
Pedrosa P, Zhang Z, Nuñez-Quintela V, Macias D, Ge J, Denholm M, Dyas A, Estevez-Souto V, Lado-Fernandez P, Gonzalez P, Gomez M, Martin JE, Da Silva-Alvarez S, Collado M, Muñoz-Espín D. Inhibition of lung tumorigenesis by transient reprogramming in cancer cells. Cell Death Dis 2024; 15:857. [PMID: 39587064 PMCID: PMC11589828 DOI: 10.1038/s41419-024-07207-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 10/25/2024] [Accepted: 10/31/2024] [Indexed: 11/27/2024]
Abstract
Oncogenic transformation and Oct4, Sox2, Klf4 and c-Myc (OSKM)-mediated induction of pluripotency are two independent and incompatible cellular fates. While continuous expression of OSKM can convert normal somatic cells into teratogenic pluripotent cells, it remains speculative what is the impact of transient OSKM expression in cancer cells. Here, we find that OSKM expression limits the growth of transformed lung cells by inducing apoptosis and senescence. We identify Oct4 and Klf4 as the main individual reprogramming factors responsible for this effect. Mechanistically, the induction of cell cycle inhibitor p21 downstream of the reprogramming factors acts as mediator of cell death and senescence. Using a variety of in vivo systems, including allografts, orthotopic transplantation and KRAS-driven lung cancer mouse models, we demonstrate that transient reprogramming by OSKM expression in cancer cells impairs tumor growth and reduces tumor burden. Altogether, our results show that the induction of transient reprogramming in cancer cells is antitumorigenic opening novel potential therapeutic avenues in oncology.
Collapse
Affiliation(s)
- Pablo Pedrosa
- Cell Senescence, Cancer and Aging Laboratory, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Zhenguang Zhang
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Victor Nuñez-Quintela
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, UK
| | - David Macias
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Jianfeng Ge
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Mary Denholm
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, UK
- Department of Oncology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Anna Dyas
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Valentin Estevez-Souto
- Cell Senescence, Cancer and Aging Laboratory, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Patricia Lado-Fernandez
- Cell Senescence, Cancer and Aging Laboratory, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
- Department of Physiology and Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidad de Santiago de Compostela, Santiago de Compostela, Spain
| | - Patricia Gonzalez
- Histopathology Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Maria Gomez
- Histopathology Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Jose Ezequiel Martin
- CMDL, Department of Oncology, SMCL, Department of Medical Genetics, University of Cambridge, Cambridge, UK
| | - Sabela Da Silva-Alvarez
- Cell Senescence, Cancer and Aging Laboratory, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Manuel Collado
- Cell Senescence, Cancer and Aging Laboratory, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain.
- Department of Immunology and Oncology (DIO), Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain.
| | - Daniel Muñoz-Espín
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, UK.
- CRUK Cambridge Centre Thoracic Cancer Programme, University of Cambridge, Cambridge, UK.
| |
Collapse
|
10
|
Zhai LH, Jia XL, Chen YL, Liu MY, Zhang JD, Ma SJ, Wang XJ, Cheng WH, He JL, Zhou JJ, Zuo LY, Zhang MQ, Yuan Q, Xu MH, Ji J, Tan MJ, Liu B. Comprehensive multi-omics analysis elucidates colchicine-induced toxicity mechanisms and unveils the therapeutic potential of MLN4924 and kinase inhibitors. Acta Pharmacol Sin 2024:10.1038/s41401-024-01422-5. [PMID: 39567751 DOI: 10.1038/s41401-024-01422-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 11/05/2024] [Indexed: 11/22/2024] Open
Abstract
Colchicine is a widely prescribed anti-inflammatory drug for the treatment of gout, familial Mediterranean fever and pericarditis, but its narrow therapeutic window presents a significant risk of severe toxicity. Despite its clinical relevance, the molecular mechanisms underlying colchicine's pharmacological effects and associated toxicity and explored potential therapeutic interventions to mitigate its adverse effects. We showed the colchicine's impact on cellular morphology in human umbilical vein endothelial cells (HUVEC) and HeLa cells including cell rounding and detachment following 24 h of exposure that revealed pronounced cytotoxic effects. We then established a large-scale screening model to identify small molecules capable of reversing colchicine-induced cellular toxicity, and identified MLN4924, an inhibitor of the Cullin-RING E3 ligase (CRL) system, as a promising candidate for mitigating colchicine-induced cellular injury. Through a comprehensive multi-omics approach including transcriptomics, proteomics, phosphoproteomics and ubiquitinomics, we systematically characterized the molecular perturbations caused by colchicine and delineated the protective mechanisms of MLN4924. We found that MLN4924 exerted its protective effects by modulating critical cellular pathways, specifically preventing the dysregulation of cell cycle progression, mitotic disruption and microtubule destabilization triggered by colchicine. Furthermore, proteomic and phosphoproteomic analyses revealed significant alterations in kinase signaling networks, with combined inhibition of CDK1 and PAK1 emerging as an effective strategy to counteract colchicine-induced cellular dysfunction. These results not only provide a detailed molecular characterization of colchicine toxicity but also identify key therapeutic targets, laying the groundwork for the development of targeted interventions to mitigate colchicine-induced adverse effects in clinical practice.
Collapse
Affiliation(s)
- Lin-Hui Zhai
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
- Translational Research Institute of Brain and Brain-like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, China
| | - Xing-Long Jia
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Yu-Lu Chen
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Mu-Yin Liu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Jing-Dan Zhang
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, 528400, China
| | - Shao-Jie Ma
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Xiu-Jun Wang
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Wen-Hao Cheng
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Jing-Liang He
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Jiao-Jiao Zhou
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Ling-Yi Zuo
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Mei-Qi Zhang
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Qing Yuan
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Meng-Han Xu
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Jing Ji
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Min-Jia Tan
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China.
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, 528400, China.
| | - Bin Liu
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China.
| |
Collapse
|
11
|
Sun YD, Li GH, Zhang F, Cheng T, Zhang JP, Zhang XB. A p21 reporter iPSC line for evaluating CRISPR-Cas9 and vector-induced stress responses. Stem Cells 2024; 42:992-1005. [PMID: 39283950 PMCID: PMC11541227 DOI: 10.1093/stmcls/sxae056] [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: 05/02/2024] [Accepted: 08/23/2024] [Indexed: 11/08/2024]
Abstract
CRISPR-Cas9 editing triggers activation of the TP53-p21 pathway, but the impacts of different editing components and delivery methods have not been fully explored. In this study, we introduce a p21-mNeonGreen reporter iPSC line to monitor TP53-p21 pathway activation. This reporter enables dynamic tracking of p21 expression via flow cytometry, revealing a strong correlation between p21 expression and indel frequencies, and highlighting its utility in guide RNA screening. Our findings show that p21 activation is significantly more pronounced with double-stranded oligodeoxynucleotides (ODNs) or adeno-associated viral vectors (AAVs) compared to their single-stranded counterparts. Lentiviral vectors (LVs) and integrase-defective lentiviral vectors induce notably lower p21 expression than AAVs, suggesting their suitability for gene therapy in sensitive cells such as hematopoietic stem cells or immune cells. Additionally, specific viral promoters like SFFV significantly amplify p21 activation, emphasizing the critical role of promoter selection in vector development. Thus, the p21-mNeonGreen reporter iPSC line is a valuable tool for assessing the potential adverse effects of gene editing methodologies and vectors. Highlights Established a p21-mNeonGreen reporter iPSC line to track activation of the TP53-p21 pathway. Found a direct correlation between p21-mNeonGreen expression and indel frequencies, aiding in gRNA screening. Showed that LVs are preferable over AAVs for certain cells due to lower p21 activation, with viral promoter choice impacting p21 response.
Collapse
Affiliation(s)
- Yi-Dan Sun
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, People’s Republic of China
- Tianjin Institutes of Health Science, Tianjin 301600, People’s Republic of China
| | - Guo-Hua Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, People’s Republic of China
- Tianjin Institutes of Health Science, Tianjin 301600, People’s Republic of China
| | - Feng Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, People’s Republic of China
- Tianjin Institutes of Health Science, Tianjin 301600, People’s Republic of China
| | - Tao Cheng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, People’s Republic of China
- Tianjin Institutes of Health Science, Tianjin 301600, People’s Republic of China
| | - Jian-Ping Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, People’s Republic of China
- Tianjin Institutes of Health Science, Tianjin 301600, People’s Republic of China
| | - Xiao-Bing Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, People’s Republic of China
- Tianjin Institutes of Health Science, Tianjin 301600, People’s Republic of China
| |
Collapse
|
12
|
Wang N, Yang S, Li Y, Gou F, Lv Y, Zhao X, Wang Y, Xu C, Zhou B, Dong F, Ju Z, Cheng T, Cheng H. p21/Zbtb18 repress the expression of cKit to regulate the self-renewal of hematopoietic stem cells. Protein Cell 2024; 15:840-857. [PMID: 38721703 PMCID: PMC11528518 DOI: 10.1093/procel/pwae022] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 04/01/2024] [Indexed: 11/03/2024] Open
Abstract
The maintenance of hematopoietic stem cells (HSCs) is a complex process involving numerous cell-extrinsic and -intrinsic regulators. The first member of the cyclin-dependent kinase family of inhibitors to be identified, p21, has been reported to perform a wide range of critical biological functions, including cell cycle regulation, transcription, differentiation, and so on. Given the previous inconsistent results regarding the functions of p21 in HSCs in a p21-knockout mouse model, we employed p21-tdTomato (tdT) mice to further elucidate its role in HSCs during homeostasis. The results showed that p21-tdT+ HSCs exhibited increased self-renewal capacity compared to p21-tdT- HSCs. Zbtb18, a transcriptional repressor, was upregulated in p21-tdT+ HSCs, and its knockdown significantly impaired the reconstitution capability of HSCs. Furthermore, p21 interacted with ZBTB18 to co-repress the expression of cKit in HSCs and thus regulated the self-renewal of HSCs. Our data provide novel insights into the physiological role and mechanisms of p21 in HSCs during homeostasis independent of its conventional role as a cell cycle inhibitor.
Collapse
Affiliation(s)
- Nini Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
- CAMS Center for Stem Cell Medicine, PUMC Department of Stem Cell and Regenerative Medicine, Tianjin 300020, China
| | - Shangda Yang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
- CAMS Center for Stem Cell Medicine, PUMC Department of Stem Cell and Regenerative Medicine, Tianjin 300020, China
| | - Yu Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
- CAMS Center for Stem Cell Medicine, PUMC Department of Stem Cell and Regenerative Medicine, Tianjin 300020, China
| | - Fanglin Gou
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Cell Biology, Tianjin Medical University, Tianjin 300270, China
| | - Yanling Lv
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
- CAMS Center for Stem Cell Medicine, PUMC Department of Stem Cell and Regenerative Medicine, Tianjin 300020, China
| | - Xiangnan Zhao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
- CAMS Center for Stem Cell Medicine, PUMC Department of Stem Cell and Regenerative Medicine, Tianjin 300020, China
| | - Yifei Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
- CAMS Center for Stem Cell Medicine, PUMC Department of Stem Cell and Regenerative Medicine, Tianjin 300020, China
| | - Chang Xu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
- CAMS Center for Stem Cell Medicine, PUMC Department of Stem Cell and Regenerative Medicine, Tianjin 300020, China
| | - Bin Zhou
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Fang Dong
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
- CAMS Center for Stem Cell Medicine, PUMC Department of Stem Cell and Regenerative Medicine, Tianjin 300020, China
| | - Zhenyu Ju
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Tao Cheng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
- CAMS Center for Stem Cell Medicine, PUMC Department of Stem Cell and Regenerative Medicine, Tianjin 300020, China
| | - Hui Cheng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
- CAMS Center for Stem Cell Medicine, PUMC Department of Stem Cell and Regenerative Medicine, Tianjin 300020, China
| |
Collapse
|
13
|
Kim D, Cooper JA, Helfman DM. Loss of myosin light chain kinase induces the cellular senescence associated secretory phenotype to promote breast epithelial cell migration. Sci Rep 2024; 14:25786. [PMID: 39468273 PMCID: PMC11519378 DOI: 10.1038/s41598-024-76868-y] [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: 05/22/2024] [Accepted: 10/17/2024] [Indexed: 10/30/2024] Open
Abstract
Overexpression or activation of oncogenes or loss of tumor-suppressor genes can induce cellular senescence as a defense mechanism against tumor development, thereby maintaining cellular homeostasis. However, cancer cells can circumvent this senescent state and continue to spread. Myosin light chain kinase (MLCK) is downregulated in many breast cancers. Here we report that downregulation of MLCK in normal breast epithelial cells induces a senescence-associated secretory phenotype and stimulates migration. The reduction of MLCK results in increased p21Cip1 expression, dependent on p53 and the AKT-mammalian target of rapamycin pathway. Subsequently, p21Cip1 promotes the secretion of soluble ICAM-1, IL-1α, IL-6 and IL-8, thereby enhancing collective cell migration in a non-cell-autonomous manner. These findings provide new mechanistic insights into the role of MLCK in cellular senescence and cancer progression.
Collapse
Affiliation(s)
- Dayoung Kim
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA.
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, South Korea.
| | - Jonathan A Cooper
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - David M Helfman
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| |
Collapse
|
14
|
Zhang W, Zheng Z, Wang T, Yang X, Zhao J, Zhong Y, Peng X, Zhou Y. Succinylated Type I Collagen Regulates Ferroptosis to Attenuate Skin Photoaging. ACS APPLIED MATERIALS & INTERFACES 2024; 16:56744-56761. [PMID: 39392263 DOI: 10.1021/acsami.4c11952] [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: 10/12/2024]
Abstract
During the process of photoaging in the skin, Succinylated type I collagen has a significant effect on reversing the damage caused by UVB radiation, with the regulation of cellular ferroptosis being one of its important pathophysiological mechanisms. Specifically, Succinylated type I collagen reduces the expression of key cell cycle regulators P16, P21, and P53, as well as the ferroptosis-related factor Acyl-CoA Synthetase Long Chain Family Member 4 (ACSL4), induced by UVB radiation in cells and tissues. Meanwhile, it increases the expression of key factors Glutathione Peroxidase 4 (GPX4) and Solute Carrier Family 7 Member 11 (SLC7A11), which inhibit ferroptosis. Additionally, our study also reveals the impact of Succinylated type I collagen on the levels of malondialdehyde (MDA), glutathione (GSH), and reactive oxygen species (ROS) in cells and tissues, directly affecting the cells' ability to cope with oxidative stress. This further suggests that Succinylated type I collagen may improve skin photoaging through various pathways, including regulating ferroptosis, antioxidation, promoting collagen synthesis, protecting the skin barrier, reducing pigmentation, and inhibiting inflammatory responses, contributing to maintaining healthy and youthful skin.
Collapse
Affiliation(s)
- Wenwen Zhang
- Department of Pathophysiology, Guangdong Medical University, Dongguan 523808, China
| | - Zetai Zheng
- Department of Pathophysiology, Guangdong Medical University, Dongguan 523808, China
| | - Tingyu Wang
- Department of Pathophysiology, Guangdong Medical University, Dongguan 523808, China
| | - Xiangjie Yang
- School of Public Health, Guangdong Medical University, Dongguan 523808, China
| | - Jiayi Zhao
- The Second Clinical Medical College of Guangdong Medical University, Dongguan 523808, China
| | - Yuesong Zhong
- The Second Clinical Medical College of Guangdong Medical University, Dongguan 523808, China
| | - Xinsheng Peng
- School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Yanfang Zhou
- Department of Pathophysiology, Guangdong Medical University, Dongguan 523808, China
- The Second Affiliated Hospital of Guangdong Medical University, Dongguan 523808, China
| |
Collapse
|
15
|
Gautam P, Ciuta I, Teif VB, Sinha SK. Predicting p53-dependent cell transitions from thermodynamic models. J Chem Phys 2024; 161:135101. [PMID: 39356070 DOI: 10.1063/5.0225166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Accepted: 09/18/2024] [Indexed: 10/03/2024] Open
Abstract
A cell's fate involves transitions among its various states, each defined by a distinct gene expression profile governed by the topology of gene regulatory networks, which are affected by 3D genome organization. Here, we develop thermodynamic models to determine the fate of a malignant cell as governed by the tumor suppressor p53 signaling network, taking into account long-range chromatin interactions in the mean-field approximation. The tumor suppressor p53 responds to stress by selectively triggering one of the potential transcription programs that influence many layers of cell signaling. These range from p53 phosphorylation to modulation of its DNA binding affinity, phase separation phenomena, and internal connectivity among cell fate genes. We use the minimum free energy of the system as a fundamental property of biological networks that influences the connection between the gene network topology and the state of the cell. We constructed models based on network topology and equilibrium thermodynamics. Our modeling shows that the binding of phosphorylated p53 to promoters of target genes can have properties of a first order phase transition. We apply our model to cancer cell lines ranging from breast cancer (MCF-7), colon cancer (HCT116), and leukemia (K562), with each one characterized by a specific network topology that determines the cell fate. Our results clarify the biological relevance of these mechanisms and suggest that they represent flexible network designs for switching between developmental decisions.
Collapse
Affiliation(s)
- Pankaj Gautam
- Theoretical and Computational Biophysical Chemistry Group, Department of Chemistry, Indian Institute of Technology Ropar, Rupnagar, Punjab 140001, India
| | - Isabella Ciuta
- School of Life Sciences, University of Essex, Wivenhoe Park, Colchester CO4 3SQ, United Kingdom
| | - Vladimir B Teif
- School of Life Sciences, University of Essex, Wivenhoe Park, Colchester CO4 3SQ, United Kingdom
| | - Sudipta Kumar Sinha
- Theoretical and Computational Biophysical Chemistry Group, Department of Chemistry, Indian Institute of Technology Ropar, Rupnagar, Punjab 140001, India
| |
Collapse
|
16
|
Schubert SA, Ruano D, Joruiz SM, Stroosma J, Glavak N, Montali A, Pinto LM, Rodríguez-Girondo M, Barge-Schaapveld DQCM, Nielsen M, van Nesselrooij BPM, Mensenkamp AR, van Leerdam ME, Sharp TH, Morreau H, Bourdon JC, de Miranda NFCC, van Wezel T. Germline variant affecting p53β isoforms predisposes to familial cancer. Nat Commun 2024; 15:8208. [PMID: 39294166 PMCID: PMC11410958 DOI: 10.1038/s41467-024-52551-8] [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: 05/08/2020] [Accepted: 09/06/2024] [Indexed: 09/20/2024] Open
Abstract
Germline and somatic TP53 variants play a crucial role during tumorigenesis. However, genetic variations that solely affect the alternatively spliced p53 isoforms, p53β and p53γ, are not fully considered in the molecular diagnosis of Li-Fraumeni syndrome and cancer. In our search for additional cancer predisposing variants, we identify a heterozygous stop-lost variant affecting the p53β isoforms (p.*342Serext*17) in four families suspected of an autosomal dominant cancer syndrome with colorectal, breast and papillary thyroid cancers. The stop-lost variant leads to the 17 amino-acid extension of the p53β isoforms, which increases oligomerization to canonical p53α and dysregulates the expression of p53's transcriptional targets. Our study reveals the capacity of p53β mutants to influence p53 signalling and contribute to the susceptibility of different cancer types. These findings underscore the significance of p53 isoforms and the necessity of comprehensive investigation into the entire TP53 gene in understanding cancer predisposition.
Collapse
Affiliation(s)
- Stephanie A Schubert
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Dina Ruano
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Jordy Stroosma
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Nikolina Glavak
- School of Medicine, University of Dundee, Dundee, UK
- Croatian Institute of Transfusion Medicine, Zagreb, Croatia
| | - Anna Montali
- School of Medicine, University of Dundee, Dundee, UK
| | - Lia M Pinto
- School of Medicine, University of Dundee, Dundee, UK
| | - Mar Rodríguez-Girondo
- Department of Biomedical Data Sciences, Section of Medical Statistics, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Maartje Nielsen
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Arjen R Mensenkamp
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Monique E van Leerdam
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Thomas H Sharp
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
- School of Biochemistry, University of Bristol, Bristol, UK
| | - Hans Morreau
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | - Tom van Wezel
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands.
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands.
| |
Collapse
|
17
|
Romaldini A, Spanò R, Veronesi M, Grimaldi B, Bandiera T, Sabella S. Human Multi-Lineage Liver Organoid Model Reveals Impairment of CYP3A4 Expression upon Repeated Exposure to Graphene Oxide. Cells 2024; 13:1542. [PMID: 39329726 PMCID: PMC11429598 DOI: 10.3390/cells13181542] [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: 07/08/2024] [Revised: 09/06/2024] [Accepted: 09/09/2024] [Indexed: 09/28/2024] Open
Abstract
Three-dimensional hepatic cell cultures can provide an important advancement in the toxicity assessment of nanomaterials with respect to 2D models. Here, we describe liver organoids (LOs) obtained by assembling multiple cell lineages in a fixed ratio 1:1:0.2. These are upcyte® human hepatocytes, UHHs, upcyte® liver sinusoidal endothelial cells, LSECs, and human bone marrow-derived mesenchymal stromal cells, hbmMSCs. The structural and functional analyses indicated that LOs reached size stability upon ca. 10 days of cultivation (organoid maturation), showing a surface area of approximately 10 mm2 and the hepatic cellular lineages, UHHs and LSECs, arranged to form both primitive biliary networks and sinusoid structures, alike in vivo. LOs did not show signs of cellular apoptosis, senescence, or alteration of hepatocellular functions (e.g., dis-regulation of CYP3A4 or aberrant production of Albumin) for the entire culture period (19 days since organoid maturation). After that, LOs were repeatedly exposed for 19 days to a single or repeated dose of graphene oxide (GO: 2-40 µg/mL). We observed that the treatment did not induce any macroscopic signs of tissue damage, apoptosis activation, and alteration of cell viability. However, in the repeated dose regimen, we observed a down-regulation of CYP3A4 gene expression. Notably, these findings are in line with recent in vivo data, which report a similar impact on CYP3A4 when mice were repeatedly exposed to GO. Taken together, these findings warn of the potential detrimental effects of GO in real-life exposure (e.g., occupational scenario), where its progressive accumulation is likely expected. More in general, this study highlights that LOs formed by many cell lineages can enable repeated exposure regimens (suitable to mimic accumulation); thus, they can be suitably considered alternative or complementary in vitro systems to animal models.
Collapse
Affiliation(s)
- Alessio Romaldini
- Nanoregulatory Group, D3 PharmaChemistry, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy; (A.R.); (R.S.)
| | - Raffaele Spanò
- Nanoregulatory Group, D3 PharmaChemistry, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy; (A.R.); (R.S.)
| | - Marina Veronesi
- Structural Biophysics Facility, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy;
- D3 PharmaChemistry, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy
| | - Benedetto Grimaldi
- Molecular Medicine, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy
| | - Tiziano Bandiera
- Nanoregulatory Group, D3 PharmaChemistry, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy; (A.R.); (R.S.)
| | - Stefania Sabella
- Nanoregulatory Group, D3 PharmaChemistry, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy; (A.R.); (R.S.)
| |
Collapse
|
18
|
Iordache F, Petcu ACI, Alexandru DM. Genetic and Epigenetic Interactions Involved in Senescence of Stem Cells. Int J Mol Sci 2024; 25:9708. [PMID: 39273655 PMCID: PMC11396476 DOI: 10.3390/ijms25179708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/04/2024] [Accepted: 09/05/2024] [Indexed: 09/15/2024] Open
Abstract
Cellular senescence is a permanent condition of cell cycle arrest caused by a progressive shortening of telomeres defined as replicative senescence. Stem cells may also undergo an accelerated senescence response known as premature senescence, distinct from telomere shortening, as a response to different stress agents. Various treatment protocols have been developed based on epigenetic changes in cells throughout senescence, using different drugs and antioxidants, senolytic vaccines, or the reprogramming of somatic senescent cells using Yamanaka factors. Even with all the recent advancements, it is still unknown how different epigenetic modifications interact with genetic profiles and how other factors such as microbiota physiological conditions, psychological states, and diet influence the interaction between genetic and epigenetic pathways. The aim of this review is to highlight the new epigenetic modifications that are involved in stem cell senescence. Here, we review recent senescence-related epigenetic alterations such as DNA methylation, chromatin remodeling, histone modification, RNA modification, and non-coding RNA regulation outlining new possible targets for the therapy of aging-related diseases. The advantages and disadvantages of the animal models used in the study of cellular senescence are also briefly presented.
Collapse
Affiliation(s)
- Florin Iordache
- Biochemistry Disciplines, Faculty of Veterinary Medicine, University of Agronomic Sciences and Veterinary Medicine, 050097 Bucharest, Romania
- Advanced Research Center for Innovative Materials, Products and Processes CAMPUS, Politehnica University, 060042 Bucharest, Romania
| | - Adriana Cornelia Ionescu Petcu
- Biochemistry Disciplines, Faculty of Veterinary Medicine, University of Agronomic Sciences and Veterinary Medicine, 050097 Bucharest, Romania
| | - Diana Mihaela Alexandru
- Pharmacology and Pharmacy Disciplines, Faculty of Veterinary Medicine, University of Agronomic Sciences and Veterinary Medicine, 050097 Bucharest, Romania
| |
Collapse
|
19
|
Myeza N, Slabber C, Munro OQ, Sookai S, Zacharias SC, Martins-Furness C, Harmse L. An 8-aminoquinoline-naphthyl copper complex causes apoptotic cell death by modulating the expression of apoptotic regulatory proteins in breast cancer cells. Eur J Pharmacol 2024; 978:176764. [PMID: 38908670 DOI: 10.1016/j.ejphar.2024.176764] [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: 02/27/2024] [Revised: 06/08/2024] [Accepted: 06/19/2024] [Indexed: 06/24/2024]
Abstract
Breast cancer is one of the most common cancers globally and a leading cause of cancer-related deaths among women. Despite the combination of chemotherapy with targeted therapy, including monoclonal antibodies and kinase inhibitors, drug resistance and treatment failure remain a common occurrence. Copper, complexed to various organic ligands, has gained attention as potential chemotherapeutic agents due to its perceived decreased toxicity to normal cells. The cytotoxic efficacy and the mechanism of cell death of an 8-aminoquinoline-naphthyl copper complex (Cu8AqN) in MCF-7 and MDA-MB-231 breast cancer cell lines was investigated. The complex inhibited the growth of MCF-7 and MDA-MB-231 cells with IC50 values of 2.54 ± 0.69 μM and 3.31 ± 0.06 μM, respectively. Nuclear fragmentation, annexin V binding, and increased caspase-3/7 activity indicated apoptotic cell death. The loss of mitochondrial membrane potential, an increase in caspase-9 activity, the absence of active caspase-8 and a decrease of tumour necrosis factor receptor 1(TNFR1) expression supported activation of the intrinsic apoptotic pathway. Increased ROS formation and increased expression of haem oxygenase-1 (HMOX-1) indicated activation of cellular stress pathways. Expression of p21 protein in the nuclei was increased indicating cell cycle arrest, whilst the expression of inhibitor of apoptosis proteins (IAPs); cIAP1, XIAP and survivin were decreased, creating a pro-apoptotic environment. Phosphorylated p53 species; phospho-p53(S15), phospho-p53(S46), and phospho-p53(S392) accumulated in MCF-7 cells indicating the potential of Cu8AqN to restore p53 function in the cells. In combination, the data indicates that Cu8AqN is a useful lead molecule worthy of further exploration as a potential anti-cancer drug.
Collapse
Affiliation(s)
- Nonzuzo Myeza
- Division of Pharmacology, Department of Pharmacy and Pharmacology, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, 2193, South Africa
| | - Cathy Slabber
- Molecular Sciences Institute, School of Chemistry, University of the Witwatersrand, 1 Jan Smut Ave, Braamfontein, Johannesburg, 2017, South Africa
| | - Orde Q Munro
- Molecular Sciences Institute, School of Chemistry, University of the Witwatersrand, 1 Jan Smut Ave, Braamfontein, Johannesburg, 2017, South Africa; School of Chemistry, University of Leeds, Woodhouse Lane, Leeds, LS2 9JT, UK
| | - Sheldon Sookai
- Molecular Sciences Institute, School of Chemistry, University of the Witwatersrand, 1 Jan Smut Ave, Braamfontein, Johannesburg, 2017, South Africa
| | - Savannah C Zacharias
- School of Chemistry and Physics, University of KwaZulu-Natal, King Edward Drive, Pietermaritzburg, Scottsville, 3209, South Africa
| | - Carla Martins-Furness
- Division of Pharmacology, Department of Pharmacy and Pharmacology, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, 2193, South Africa
| | - Leonie Harmse
- Division of Pharmacology, Department of Pharmacy and Pharmacology, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, 2193, South Africa.
| |
Collapse
|
20
|
Yu GT, Gomez PT, Prata LG, Lehman JS, Tchkonia T, Kirkland JL, Meves A, Wyles SP. Clinicopathological and cellular senescence biomarkers in chronic stalled wounds. Int J Dermatol 2024; 63:1227-1235. [PMID: 38351588 PMCID: PMC11323232 DOI: 10.1111/ijd.17072] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 01/15/2024] [Accepted: 01/19/2024] [Indexed: 08/16/2024]
Abstract
BACKGROUND Chronic wounds have been associated with an elevated burden of cellular senescence, a state of essentially irreversible cell cycle arrest, resistance to apoptosis, and a secretory phenotype. However, whether senescent cells contribute to wound chronicity in humans remains unclear. The objective of this article is to assess the role of clinicopathological characteristics and cellular senescence in the time-to-healing of chronic wounds. METHODS A cohort of 79 patients with chronic wounds was evaluated in a single-center academic practice from February 1, 2005, to February 28, 2015, and followed for up to 36 months. Clinical characteristics and wound biopsies were obtained at baseline, and time-to-healing was assessed. Wound biopsies were analyzed histologically for pathological characteristics and molecularly for markers of cellular senescence. In addition, biopsy slides were stained for p16INK4a expression. RESULTS No clinical or pathological characteristics were found to have significant associations with time-to-healing. A Cox proportional hazard ratio model revealed increased CDKN1A (p21CIP1/WAF1) expression to predict longer time-to-healing, and a model adjusted for gender and epidermal hyperplasia revealed increased CDKN1A expression and decreased PAPPA expression to predict longer time-to-healing. Increased p16INK4a staining was observed in diabetic wounds compared to non-diabetic wounds, and the same association was observed in the context of high dermal fibrosis. CONCLUSIONS The findings of this pilot study suggest that senescent cells contribute to wound chronicity in humans, especially in diabetic wounds.
Collapse
Affiliation(s)
- Grace Tianen Yu
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic Alix School of Medicine, and Mayo Clinic Medical Scientist Training Program, Rochester, MN
| | - Paul T. Gomez
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN
| | - Larissa G. Prata
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN
| | - Julia Scott Lehman
- Department of Dermatology, Mayo Clinic, Rochester, MN
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Tamar Tchkonia
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN
| | - James L. Kirkland
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN
- Division of General Internal Medicine, Department of Medicine, Mayo Clinic, Rochester, MN
| | | | - Saranya P. Wyles
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN
- Department of Dermatology, Mayo Clinic, Rochester, MN
| |
Collapse
|
21
|
Erdogan MK, Gundogdu R, Toy Y, Halil Gecibesler I, Yapar Y, Behcet L, Zengin G. Comparison of Anticancer, Antioxidant, Enzyme Inhibitory Effects and Phytochemical Contents Between Edible Lettuce (Lactuca sativa) and a New Wild Species (Lactuca anatolica). Chem Biodivers 2024; 21:e202400552. [PMID: 38958194 DOI: 10.1002/cbdv.202400552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 07/01/2024] [Accepted: 07/02/2024] [Indexed: 07/04/2024]
Abstract
In this study, the bioactive components, enzyme inhibitory, antioxidant and anticancer potentials of edible (L. sativa) and a new species (L. anatolica) of Lactuca were evaluated and compared. The quantitative analyzes of the bioactive components of L. sativa (LS) and L. anatolica (LA) were analyzed quantitatively by GC-MS and Orbitrab HPLC-HRMS. Antioxidant, enzyme inhibitory and anticancer properties were analyzed by various assays. In general, LA exhibited more stronger antioxidant properties compared to LS. The extracts showed similar inhibitory effects on these enzymes. It was determined that LS was dominant in terms of linoleic acid (23.71 %), while LA contained a high level of α-linolenic acid (31.70 %). LA and LS inhibited the viability of A549 and MCF-7 cells in a dose-dependent manner. IC50 values for LA, LS and cisplatin were determined as 120.3, 197.5, 4.3 μg/mL in A549 cell line and 286.2, 472.8, 7.2 μg/mL in MCF-7 cell line, respectively. It was revealed that LA and LS treatment at 50 μg/mL concentrations in A549 cells completely suppressed the colony forming capacity, and treatment with IC50 doses inhibited cell migration, and triggered apoptosis by regulating caspase-3, cPARP, p53 and p21. The findings of this study suggested that these species have significant pharmacological potential.
Collapse
Affiliation(s)
- Mehmet Kadir Erdogan
- Department of Molecular Biology and Genetics, Faculty of Arts and Sciences, Bingol University, Bingol, Turkiye
| | - Ramazan Gundogdu
- Department of Pharmacy Services, Vocational School of Health Services, Bingol University, Bingol, Turkiye
- Current address: Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Yusuf Toy
- Department of Molecular Biology and Genetics, Faculty of Arts and Sciences, Bingol University, Bingol, Turkiye
| | - Ibrahim Halil Gecibesler
- Department of Occupational Health and Safety, Faculty of Health Science, Bingol University, Bingol, Turkiye
| | - Yakup Yapar
- Department of Molecular Biology and Genetics, Faculty of Arts and Sciences, Bingol University, Bingol, Turkiye
| | - Lutfi Behcet
- Department of Molecular Biology and Genetics, Faculty of Arts and Sciences, Bingol University, Bingol, Turkiye
| | - Gokhan Zengin
- Department of Biology, Science Faculty, Selcuk University, Konya, Turkiye
| |
Collapse
|
22
|
Abdel Halim AS, Ali MAM, Inam F, Alhalwan AM, Daoush WM. Fe 3O 4-Coated CNTs-Gum Arabic Nano-Hybrid Composites Exhibit Enhanced Anti-Leukemia Potency Against AML Cells via ROS-Mediated Signaling. Int J Nanomedicine 2024; 19:7323-7352. [PMID: 39055376 PMCID: PMC11269411 DOI: 10.2147/ijn.s467733] [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: 03/07/2024] [Accepted: 07/03/2024] [Indexed: 07/27/2024] Open
Abstract
Background Prior studies on magnetite (Fe3O4) NPs and carbon nanotubes (CNTs) cytotoxic effects against acute myeloid leukemia (AML) are inconclusive rather than definitive. Purpose Investigation of the effects of Gum Arabic (GA)-stabilized/destabilized Fe3O4 NPs and CNTs, alone or in combination, on AML cell proliferation. Methods Hybrid NPs were synthesized, characterized, and assessed for their cytotoxicity against Kasumi-1, HL-60, and THP-1 in comparison to normal primary bone marrow CD34+ cells. The molecular pathways of nanostructures' cytotoxicity were also investigated. Results The Fe3O4 NPs were effectively synthesized and attached to the surface of the CNTs, resulting in the formation of a novel hybrid through their interaction with the GA colloidal solution in an aqueous media. Although the evaluated nanostructured nanoparticles had significant growth suppression ability against the leukemia cell lines, with IC50 values ranging from 42.437 to 189.842 μg/mL, they exhibited comparatively modest toxicity towards normal hematopoietic cells (IC50: 113.529‒162.656 μg/mL). The incorporation of Fe3O4 NPs with CNTs in a hybrid nanocomposite significantly improved their effectiveness against leukemia cells, with the extent of improvement varying depending on the specific cell type. The nanostructured particles were stabilized by GA, which enhances their ability to inhibit cell proliferation in a manner that depends on the specific cell type. Also, nanoparticles exhibit cytotoxicity due to their capacity to stimulate the production of intracellular ROS, halt the cell cycle at the G1 phase, and induce apoptosis. This is supported by the activation of p53, BAX, cytochrome C, and caspase-3, which are triggered by ROS. The nanostructures lead to an increase in the expression of genes encoding proteins related to oxidative stress (SIRT1, FOXO3, NFE2L2, and MAP3K5) and cyclin-dependent kinase inhibitors (CDKN1A and CDKN1B) in response to ROS. Conclusion We provide an effective Fe3O4 NPs/CNTs nano-hybrid composite that induces apoptosis and has strong anti-leukemic capabilities. This hybrid nanocomposite is promising for in vivo testing and validation.
Collapse
Affiliation(s)
- Alyaa S Abdel Halim
- Department of Biochemistry, Faculty of Science, Ain Shams University, Cairo, 11566, Egypt
| | - Mohamed A M Ali
- Department of Biochemistry, Faculty of Science, Ain Shams University, Cairo, 11566, Egypt
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, 11623, Kingdom of Saudi Arabia
| | - Fawad Inam
- Department of Engineering and Computing, School of Architecture, Computing and Engineering, University of East London, London, UK
- Executive Principal Office, Oxford Business College, Oxford, OX1 2EP, UK
| | - Abdulrahman M Alhalwan
- Department of Chemistry, College of Science, Imam Mohammad Ibn Saud Islamic University, Riyadh, 11623, Kingdom of Saudi Arabia
| | - Walid M Daoush
- Department of Chemistry, College of Science, Imam Mohammad Ibn Saud Islamic University, Riyadh, 11623, Kingdom of Saudi Arabia
- Department of Production Technology, Faculty of Technology and Education, Helwan University, Cairo, 11281, Egypt
| |
Collapse
|
23
|
Al-Hayali MZ, Nge CE, Lim KH, Collins HM, Kam TS, Bradshaw TD. Conofolidine: A Natural Plant Alkaloid That Causes Apoptosis and Senescence in Cancer Cells. Molecules 2024; 29:2654. [PMID: 38893527 PMCID: PMC11173856 DOI: 10.3390/molecules29112654] [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: 01/02/2024] [Revised: 05/27/2024] [Accepted: 05/28/2024] [Indexed: 06/21/2024] Open
Abstract
Natural products contribute substantially to anticancer therapy; the plant kingdom provides an important source of molecules. Conofolidine is a novel Aspidosperma-Aspidosperma bisindole alkaloid isolated from the Malayan plant Tabernaemontana corymbosa. Herein, we report conofolidine's broad-spectrum anticancer activity together with that of three other bisindoles-conophylline, leucophyllidine, and bipleiophylline-against human-derived breast, colorectal, pancreatic, and lung carcinoma cell lines. Remarkably, conofolidine was able to induce apoptosis (e.g., in MDA-MB-468 breast) or senescence (e.g., in HT-29 colorectal) in cancer cells. Annexin V-FITC/PI, caspase activation, and PARP cleavage confirmed the former while positive β-gal staining corroborated the latter. Cell cycle perturbations were evident, comprising S-phase depletion, accompanied by downregulated CDK2, and cyclins (A2, D1) with p21 upregulation. Confocal imaging of HCT-116 cells revealed an induction of aberrant mitotic phenotypes-membrane blebbing, DNA-fragmentation with occasional multi-nucleation. DNA integrity assessment in HCT-116, MDA-MB-468, MIAPaCa-2, and HT-29 cells showed increased fluorescent γ-H2AX during the G1 cell cycle phase; γ-H2AX foci were validated in HCT-116 and MDA-MB-468 cells by confocal microscopy. Conofolidine increased oxidative stress, preceding apoptosis- and senescence-induction in most carcinoma cell lines as seen by enhanced ROS levels accompanied by increased NQO1 expression. Collectively, we present conofolidine as a putative potent anticancer agent capable of inducing heterogeneous modes of cancerous cell death in vitro, encouraging further preclinical evaluations of this natural product.
Collapse
Affiliation(s)
- Mohammed Zuhair Al-Hayali
- School of Pharmacy, Al-Kitab University, Kirkuk 36015, Iraq
- School of Pharmacy, Biodiscovery Institute, University of Nottingham, University Park, Nottingham NG7 2RD, UK;
| | - Choy-Eng Nge
- Department of Chemistry, Faculty of Science, University of Malaya, Kuala Lumpur 50603, Malaysia; (C.-E.N.); (T.-S.K.)
| | - Kuan Hon Lim
- School of Pharmacy, University of Nottingham Malaysia, Jalan Broga, Semenyih 43500, Malaysia;
| | - Hilary M. Collins
- School of Pharmacy, Biodiscovery Institute, University of Nottingham, University Park, Nottingham NG7 2RD, UK;
| | - Toh-Seok Kam
- Department of Chemistry, Faculty of Science, University of Malaya, Kuala Lumpur 50603, Malaysia; (C.-E.N.); (T.-S.K.)
| | - Tracey D. Bradshaw
- School of Pharmacy, Biodiscovery Institute, University of Nottingham, University Park, Nottingham NG7 2RD, UK;
| |
Collapse
|
24
|
Subramaniyam K, Harihar S. An Overview on the Emerging Role of the Plasma Protease Inhibitor Protein ITIH5 as a Metastasis Suppressor. Cell Biochem Biophys 2024; 82:399-409. [PMID: 38355846 DOI: 10.1007/s12013-024-01227-7] [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: 01/30/2023] [Accepted: 02/02/2024] [Indexed: 02/16/2024]
Abstract
Most cancers are not detected until they have progressed to the point of becoming malignant and life-threatening. Chemotherapy and conventional medicines are often ineffective against cancer. Although we have made significant progress, new conceptual discoveries are still required to investigate new treatments. The role of metastasis suppressor genes as a therapeutic option for limiting tumor progression and metastasis has been on the anvil for some time. In this review, we discuss the role of ITIH5 as a metastasis suppressor gene and catalog its involvement in different cancers. We further shed light on the mode of action of ITIH5 based on the available data. The review will provide a new perspective on ITIH5 as an anti-metastatic protein and hopefully serve as an impetus for future studies towards the application of ITIH5 for clinical intervention in targeting metastatic cancers.
Collapse
Affiliation(s)
- Krishnaveni Subramaniyam
- Department of Genetic Engineering, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, 603203, Tamil Nadu, India
| | - Sitaram Harihar
- Department of Biotechnology, GITAM School of Science, GITAM (Deemed to be) University, Visakhapatnam, 530045, Andhra Pradesh, India.
| |
Collapse
|
25
|
Xu S, Li Z, Xin X, An F. Curdepsidone A Induces Intrinsic Apoptosis and Inhibits Protective Autophagy via the ROS/PI3K/AKT Signaling Pathway in HeLa Cells. Mar Drugs 2024; 22:227. [PMID: 38786619 PMCID: PMC11123476 DOI: 10.3390/md22050227] [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: 04/10/2024] [Revised: 04/25/2024] [Accepted: 04/25/2024] [Indexed: 05/25/2024] Open
Abstract
Among female oncology patients, cervical cancer stands as the fourth most prevalent malignancy, exerting significant impacts on their health. Over 600,000 women received the diagnosis of cervical cancer in 2020, and the illness claimed over 300,000 lives globally. Curdepsidone A, a derivative of depsidone, was isolated from the secondary metabolites of Curvularia sp. IFB-Z10. In this study, we revised the molecular structure of curdepsidone A and investigated the fundamental mechanism of the anti-tumor activity of curdepsidone A in HeLa cells for the first time. The results demonstrated that curdepsidone A caused G0/G1 phase arrest, triggered apoptosis via a mitochondrial apoptotic pathway, blocked the autophagic flux, suppressed the PI3K/AKT pathway, and increased the accumulation of reactive oxygen species (ROS) in HeLa cells. Furthermore, the PI3K inhibitor (LY294002) promoted apoptosis induced by curdepsidone A, while the PI3K agonist (IGF-1) eliminated such an effect. ROS scavenger (NAC) reduced curdepsidone A-induced cell apoptosis and the suppression of autophagy and the PI3K/AKT pathway. In conclusion, our results revealed that curdepsidone A hindered cell growth by causing cell cycle arrest, and promoted cell apoptosis by inhibiting autophagy and the ROS-mediated PI3K/AKT pathway. This study provides a molecular basis for the development of curdepsidone A as a new chemotherapy drug for cervical cancer.
Collapse
Affiliation(s)
- Sunjie Xu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Mei Long Road, Shanghai 200237, China; (S.X.); (Z.L.); (X.X.)
| | - Zhimin Li
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Mei Long Road, Shanghai 200237, China; (S.X.); (Z.L.); (X.X.)
| | - Xiujuan Xin
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Mei Long Road, Shanghai 200237, China; (S.X.); (Z.L.); (X.X.)
| | - Faliang An
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Mei Long Road, Shanghai 200237, China; (S.X.); (Z.L.); (X.X.)
- Marine Biomedical Science and Technology Innovation Platform of Lin-Gang Special Area, No. 4, Lane 218, Haiji Sixth Road, Shanghai 201306, China
| |
Collapse
|
26
|
Nasri E, Torrence DE, Vasilopoulos T, Knapik JA, Lagmay JP, Reith JD, Gibbs CP. Cell Cycle Checkpoints p16 and p21-Strong Predictors of Clinicopathologic Outcomes in High-Grade Osteosarcoma. Cancer J 2024; 30:133-139. [PMID: 38753746 DOI: 10.1097/ppo.0000000000000714] [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: 05/18/2024]
Abstract
PURPOSE In this study, we used a series of immunohistochemical measurements of 2 cell cycle regulators, p16 and p21, to evaluate their prognostic value, separately and in combination, for the disease outcomes. METHOD A total of 101 patients with high-grade osteosarcoma were included in this study. Clinicopathologic data were collected, and immunohistochemistry for p16 and p21 was performed and interpreted by 3 independent pathologists. Statistical analysis was performed to assess the strength of each of these markers relative to disease outcome. RESULTS Our results indicate that more than 90% expression (high) of p16 by immunohistochemistry on the initial biopsy has a strong predictive value for good histologic response to chemotherapy. The patients are also more likely to survive the past 5 years and less likely to develop metastasis than patients with less than 90% p16 (low) expression. The results for p21, on the other hand, show a unique pattern of relationship to the clinicopathologic outcomes of the disease. Patients with less than 1% (low) or more than 50% (high) expression of p21 by immunohistochemistry show a higher chance of metastasis, poor necrotic response to chemotherapy, and an overall decreased survival rate when compared with p21 expression between 1% and 50% (moderate). Our results also showed that the expression of p16 and combined p16 and p21 demonstrates a stronger predictive relationship to 5-year survival than tumor histologic necrosis and p21 alone. DISCUSSION The results of this study, once proven to be reproducible by a larger number of patients, will be valuable in the initial assessment and risk stratification of the patients for treatment and possibly the clinical trials.
Collapse
Affiliation(s)
- Elham Nasri
- From the Department of Pathology, Immunology, and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL
| | | | - Terrie Vasilopoulos
- Departments of Anesthesiology and Orthopaedic Surgery, University of Florida College of Medicine, Gainesville, FL
| | - Jacquelyn A Knapik
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL
| | - Joanne P Lagmay
- Department of Pediatrics, Pediatric Hematology/Oncology, University of Florida College of Medicine, Gainesville, FL
| | - John D Reith
- Department of Pathology, L25, Cleveland Clinic, Cleveland, OH
| | - Charles Parker Gibbs
- Department of Orthopaedics and Sport Medicine, University of Florida College of Medicine, Gainesville, FL
| |
Collapse
|
27
|
Wang P, Nie J, Li J, Ye C, Chen J, Zhang Z, Li B. VDRA downregulate β-catenin/Smad3 and DNA damage and repair associated with improved prognosis in ccRCC patients. Int J Biol Macromol 2024; 263:130405. [PMID: 38403213 DOI: 10.1016/j.ijbiomac.2024.130405] [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: 07/31/2023] [Revised: 02/21/2024] [Accepted: 02/21/2024] [Indexed: 02/27/2024]
Abstract
The clear cell renal cell carcinoma (ccRCC) spotlighted the poorest survival, while chromophobe renal cell carcinoma (chRCC) was associated with the best survival. Earlier studies corroborated vitamin D receptor (VDR) was a promising molecular for improving the prognosis of RCC. In contrast to VDRA, the one of VDR isoforms, VDRB1 (VDR isoform B1) has an N-terminal extension of 50 amino acids and is less ligand-dependent. However, the functional differences between VDRA and VDRB1, and their roles in the prognosis of ccRCC and chRCC, have not been investigated. In the present study, we uncovered that the transcripts related to vitamin D pathway and cellular calcium signaling were effectively decreased in the context of ccRCC, yet failed to exert a comparable effect within chRCC. Specially, minimally levels of VDRA wherein kidneys of patients suffering from ccRCC predict shorter survival time. In addition, the protein expressions for β-catenin/Smad3 pathway and DNA damage and repair pathways were obviously impeded in VDRA-overexpressed ccRCC cells, yet this inhibitory effect was conspicuously absent in enable VDRB1 cells. Our results provide a new idea to improve the prognosis of ccRCC via VDRA upregulation.
Collapse
Affiliation(s)
- Ping Wang
- Department of Occupational and Environmental Health, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Jin Nie
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Soochow University, Suzhou, China
| | - Jiafu Li
- Department of Occupational and Environmental Health, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Caiyong Ye
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Soochow University, Suzhou, China
| | - Jianwu Chen
- Fujian Key Laboratory of Intelligent Imaging and Precision Radiotherapy for Tumors (Fujian Medical University), Fuzhou, Fujian Province, China.
| | - Zengli Zhang
- Department of Occupational and Environmental Health, School of Public Health, Medical College of Soochow University, Suzhou, China.
| | - Bingyan Li
- Deparment of Nutrition and Food Hygiene, Medical College of Soochow University, Suzhou, China.
| |
Collapse
|
28
|
Csergeová L, Krbušek D, Janoštiak R. CIP/KIP and INK4 families as hostages of oncogenic signaling. Cell Div 2024; 19:11. [PMID: 38561743 PMCID: PMC10985988 DOI: 10.1186/s13008-024-00115-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 03/25/2024] [Indexed: 04/04/2024] Open
Abstract
CIP/KIP and INK4 families of Cyclin-dependent kinase inhibitors (CKIs) are well-established cell cycle regulatory proteins whose canonical function is binding to Cyclin-CDK complexes and altering their function. Initial experiments showed that these proteins negatively regulate cell cycle progression and thus are tumor suppressors in the context of molecular oncology. However, expanded research into the functions of these proteins showed that most of them have non-canonical functions, both cell cycle-dependent and independent, and can even act as tumor enhancers depending on their posttranslational modifications, subcellular localization, and cell state context. This review aims to provide an overview of canonical as well as non-canonical functions of CIP/KIP and INK4 families of CKIs, discuss the potential avenues to promote their tumor suppressor functions instead of tumor enhancing ones, and how they could be utilized to design improved treatment regimens for cancer patients.
Collapse
Affiliation(s)
- Lucia Csergeová
- BIOCEV-First Faculty of Medicine, Charles University, Prague, Czechia
| | - David Krbušek
- BIOCEV-First Faculty of Medicine, Charles University, Prague, Czechia
| | | |
Collapse
|
29
|
Chi F, Griffiths JI, Nath A, Bild AH. Paradoxical cancer cell proliferation after FGFR inhibition through decreased p21 signaling in FGFR1-amplified breast cancer cells. Breast Cancer Res 2024; 26:54. [PMID: 38553760 PMCID: PMC10979625 DOI: 10.1186/s13058-024-01808-7] [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: 09/27/2023] [Accepted: 03/13/2024] [Indexed: 04/02/2024] Open
Abstract
Fibroblast growth factors (FGFs) control various cellular functions through fibroblast growth factor receptor (FGFR) activation, including proliferation, differentiation, migration, and survival. FGFR amplification in ER + breast cancer patients correlate with poor prognosis, and FGFR inhibitors are currently being tested in clinical trials. By comparing three-dimensional spheroid growth of ER + breast cancer cells with and without FGFR1 amplification, our research discovered that FGF2 treatment can paradoxically decrease proliferation in cells with FGFR1 amplification or overexpression. In contrast, FGF2 treatment in cells without FGFR1 amplification promotes classical FGFR proliferative signaling through the MAPK cascade. The growth inhibitory effect of FGF2 in FGFR1 amplified cells aligned with an increase in p21, a cell cycle inhibitor that hinders the G1 to S phase transition in the cell cycle. Additionally, FGF2 addition in FGFR1 amplified cells activated JAK-STAT signaling and promoted a stem cell-like state. FGF2-induced paradoxical effects were reversed by inhibiting p21 or the JAK-STAT pathway and with pan-FGFR inhibitors. Analysis of patient ER + breast tumor transcriptomes from the TCGA and METABRIC datasets demonstrated a strong positive association between expression of FGF2 and stemness signatures, which was further enhanced in tumors with high FGFR1 expression. Overall, our findings reveal a divergence in FGFR signaling, transitioning from a proliferative to stemness state driven by activation of JAK-STAT signaling and modulation of p21 levels. Activation of these divergent signaling pathways in FGFR amplified cancer cells and paradoxical growth effects highlight a challenge in the use of FGFR inhibitors in cancer treatment.
Collapse
Affiliation(s)
- Feng Chi
- Department of Medical Oncology and Therapeutics, City of Hope Comprehensive Cancer Institute, 1218 S Fifth Ave, Monrovia, CA, 91016, USA
| | - Jason I Griffiths
- Department of Medical Oncology and Therapeutics, City of Hope Comprehensive Cancer Institute, 1218 S Fifth Ave, Monrovia, CA, 91016, USA
| | - Aritro Nath
- Department of Medical Oncology and Therapeutics, City of Hope Comprehensive Cancer Institute, 1218 S Fifth Ave, Monrovia, CA, 91016, USA
| | - Andrea H Bild
- Department of Medical Oncology and Therapeutics, City of Hope Comprehensive Cancer Institute, 1218 S Fifth Ave, Monrovia, CA, 91016, USA.
| |
Collapse
|
30
|
Afaloniati H, Aindelis G, Spyridopoulou K, Lagou MK, Tsingotjidou A, Chlichlia K, Erdman SE, Poutahidis T, Angelopoulou K. Peri-weaning cholera toxin consumption suppresses chemically-induced carcinogenesis in mice. Int J Cancer 2024; 154:1097-1110. [PMID: 38095490 DOI: 10.1002/ijc.34816] [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: 08/29/2023] [Revised: 10/27/2023] [Accepted: 11/17/2023] [Indexed: 01/23/2024]
Abstract
Gastrointestinal bacteria are known to have an impact on local and systemic immunity, and consequently either promote or suppress cancer development. Following the notion that perinatal bacterial exposure might confer immune system competency for life, we investigated whether early-life administration of cholera-toxin (CT), a protein exotoxin of the small intestine pathogenic bacterium Vibrio cholerae, may shape local and systemic immunity to impart a protective effect against tumor development in epithelia distantly located from the gut. For that, newborn mice were orally treated with low non-pathogenic doses of CT and later challenged with the carcinogen 7,12-dimethylbenzanthracene (DMBA), known to cause mainly mammary, but also skin, lung and stomach cancer. Our results revealed that CT suppressed the overall incidence and multiplicity of tumors, with varying efficiencies among cancer types, and promoted survival. Harvesting mouse tissues at an earlier time-point (105 instead of 294 days), showed that CT does not prevent preneoplastic lesions per se but it rather hinders their evolution into tumors. CT pretreatment universally increased apoptosis in the cancer-prone mammary, lung and nonglandular stomach, and altered the expression of several cancer-related molecules. Moreover, CT had a long-term effect on immune system cells and factors, the most prominent being the systemic neutrophil decrease. Finally, CT treatment significantly affected gut bacterial flora composition, leading among others to a major shift from Clostridia to Bacilli class abundance. Overall, these results support the notion that early-life CT consumption is able to affect host's immune, microbiome and gene expression profiles toward the prevention of cancer.
Collapse
Affiliation(s)
- Hara Afaloniati
- Laboratory of Biochemistry and Toxicology, School of Veterinary Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Georgios Aindelis
- Department of Molecular Biology and Genetics, Democritus University of Thrace, University Campus Dragana, Alexandroupolis, Greece
| | - Katerina Spyridopoulou
- Department of Molecular Biology and Genetics, Democritus University of Thrace, University Campus Dragana, Alexandroupolis, Greece
| | - Maria K Lagou
- Laboratory of Pathology, School of Veterinary Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Anastasia Tsingotjidou
- Laboratory of Anatomy, Histology and Embryology, School of Veterinary Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Katerina Chlichlia
- Department of Molecular Biology and Genetics, Democritus University of Thrace, University Campus Dragana, Alexandroupolis, Greece
| | - Suzan E Erdman
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Theofilos Poutahidis
- Laboratory of Pathology, School of Veterinary Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Katerina Angelopoulou
- Laboratory of Biochemistry and Toxicology, School of Veterinary Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
31
|
Li J, Liu T, Tang N, Lin S, Zhang F, Yuan W, Zhang T, Deng SH, Wu DM, Xu Y. Cyclin-dependent kinase inhibitor 1A inhibits pyroptosis to enhance human lung adenocarcinoma cell radioresistance by promoting DNA repair. Heliyon 2024; 10:e26975. [PMID: 38468925 PMCID: PMC10926078 DOI: 10.1016/j.heliyon.2024.e26975] [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: 10/10/2023] [Revised: 01/26/2024] [Accepted: 02/22/2024] [Indexed: 03/13/2024] Open
Abstract
Purpose One of the best anticancer treatments available is radiotherapy, which can be used either alone or in conjunction with other forms of treatment including chemotherapy and surgery. Nevertheless, a number of biochemical and physiological processes that react to ionizing radiation might provide tumor cells radioresistance, which makes radiotherapy ineffective. It has been found that CDKN1A regulates DNA damage repair, which contributes to tumor radioresistance. However, the precise mechanism is still unknown. Therefore, this study aimed to explore the mechanisms underlying CDKN1A-enhanced radioresistance in tumor cells. Methods Cells were irradiated with 4 Gy after CDKN1A overexpression or knockdown. CDKN1A expression was measured using real-time PCR, cell viability was evaluated using cell counting kit-8 and colony formation assays, and cytotoxicity was assessed using a lactate dehydrogenase assay. Pyroptosis in cells was analyzed using caspase-1 activity assay, enzyme-linked immunosorbent assay, and flow cytometry. Inflammation activation was detected through a co-immunoprecipitation assay. Activation of pyroptosis-related proteins was analyzed using immunohistochemistry, Western blot, and immunofluorescence. Tumor radioresistance in vivo was evaluated in a mouse xenograft model. Results Radiotherapy upregulated CDKN1A expression, which promoted lung adenocarcinoma cell survival. CDKN1A influenced radiation-induced pyroptosis in A549, which mainly depended on inhibiting the activation of the AIM2 inflammasome by promoting DNA repair. Additionally, CDKN1A upregulation enhanced A549 xenograft tumor radioresistance by inhibiting radiation-induced pyroptosis in vivo. Conclusions CDKN1A inhibits pyroptosis to enhance the radioresistance of lung adenocarcinoma cells by promoting DNA repair. This study may serve as a reference for developing novel targeted therapies against cancer.
Collapse
Affiliation(s)
- Jing Li
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, 610500, PR China
- The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, PR China
| | - Teng Liu
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, 610500, PR China
- The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, PR China
| | - Ning Tang
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, 610500, PR China
- The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, PR China
| | - Sheng Lin
- The First People's Hospital of Ziyang City, Ziyang, Sichuan, PR China
| | - Feng Zhang
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, 610500, PR China
- The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, PR China
| | - Wei Yuan
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, 610500, PR China
- The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, PR China
| | - Ting Zhang
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, 610500, PR China
- The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, PR China
| | - Shi-hua Deng
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, 610500, PR China
- The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, PR China
| | - Dong-ming Wu
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, 610500, PR China
- The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, PR China
| | - Ying Xu
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, 610500, PR China
- The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, PR China
| |
Collapse
|
32
|
Yang SF, Nelson CB, Wells JK, Fernando M, Lu R, Allen JAM, Malloy L, Lamm N, Murphy VJ, Mackay JP, Deans AJ, Cesare AJ, Sobinoff AP, Pickett HA. ZNF827 is a single-stranded DNA binding protein that regulates the ATR-CHK1 DNA damage response pathway. Nat Commun 2024; 15:2210. [PMID: 38472229 PMCID: PMC10933417 DOI: 10.1038/s41467-024-46578-0] [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: 06/15/2022] [Accepted: 03/04/2024] [Indexed: 03/14/2024] Open
Abstract
The ATR-CHK1 DNA damage response pathway becomes activated by the exposure of RPA-coated single-stranded DNA (ssDNA) that forms as an intermediate during DNA damage and repair, and as a part of the replication stress response. Here, we identify ZNF827 as a component of the ATR-CHK1 kinase pathway. We demonstrate that ZNF827 is a ssDNA binding protein that associates with RPA through concurrent binding to ssDNA intermediates. These interactions are dependent on two clusters of C2H2 zinc finger motifs within ZNF827. We find that ZNF827 accumulates at stalled forks and DNA damage sites, where it activates ATR and promotes the engagement of homologous recombination-mediated DNA repair. Additionally, we demonstrate that ZNF827 depletion inhibits replication initiation and sensitizes cancer cells to the topoisomerase inhibitor topotecan, revealing ZNF827 as a therapeutic target within the DNA damage response pathway.
Collapse
Affiliation(s)
- Sile F Yang
- Telomere Length Regulation Unit, Children's Medical Research Institute, Faculty of Medicine and Health, University of Sydney, Westmead, NSW, 2145, Australia
| | - Christopher B Nelson
- Telomere Length Regulation Unit, Children's Medical Research Institute, Faculty of Medicine and Health, University of Sydney, Westmead, NSW, 2145, Australia
| | - Jadon K Wells
- Telomere Length Regulation Unit, Children's Medical Research Institute, Faculty of Medicine and Health, University of Sydney, Westmead, NSW, 2145, Australia
| | - Madushan Fernando
- Telomere Length Regulation Unit, Children's Medical Research Institute, Faculty of Medicine and Health, University of Sydney, Westmead, NSW, 2145, Australia
| | - Robert Lu
- Telomere Length Regulation Unit, Children's Medical Research Institute, Faculty of Medicine and Health, University of Sydney, Westmead, NSW, 2145, Australia
| | - Joshua A M Allen
- Telomere Length Regulation Unit, Children's Medical Research Institute, Faculty of Medicine and Health, University of Sydney, Westmead, NSW, 2145, Australia
| | - Lisa Malloy
- Telomere Length Regulation Unit, Children's Medical Research Institute, Faculty of Medicine and Health, University of Sydney, Westmead, NSW, 2145, Australia
| | - Noa Lamm
- Nuclear Dynamics Group, Children's Medical Research Institute, Faculty of Medicine and Health, University of Sydney, Westmead, NSW, 2145, Australia
| | - Vincent J Murphy
- Genome Stability Unit, St Vincent's Institute, Fitzroy, VIC, 3065, Australia
| | - Joel P Mackay
- School of Life and Environmental Sciences, University of Sydney, Sydney, NSW, 2006, Australia
| | - Andrew J Deans
- Genome Stability Unit, St Vincent's Institute, Fitzroy, VIC, 3065, Australia
- Department of Medicine (St Vincent's), University of Melbourne, Fitzroy, VIC, 3065, Australia
| | - Anthony J Cesare
- Genome Integrity Unit, Children's Medical Research Institute, Faculty of Medicine and Health, University of Sydney, Westmead, NSW, 2145, Australia
| | - Alexander P Sobinoff
- Telomere Length Regulation Unit, Children's Medical Research Institute, Faculty of Medicine and Health, University of Sydney, Westmead, NSW, 2145, Australia
| | - Hilda A Pickett
- Telomere Length Regulation Unit, Children's Medical Research Institute, Faculty of Medicine and Health, University of Sydney, Westmead, NSW, 2145, Australia.
| |
Collapse
|
33
|
Gao W, Liu YF, Zhang YX, Wang Y, Jin YQ, Yuan H, Liang XY, Ji XY, Jiang QY, Wu DD. The potential role of hydrogen sulfide in cancer cell apoptosis. Cell Death Discov 2024; 10:114. [PMID: 38448410 PMCID: PMC10917771 DOI: 10.1038/s41420-024-01868-w] [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: 07/12/2023] [Revised: 02/05/2024] [Accepted: 02/14/2024] [Indexed: 03/08/2024] Open
Abstract
For a long time, hydrogen sulfide (H2S) has been considered a toxic compound, but recent studies have found that H2S is the third gaseous signaling molecule which plays a vital role in physiological and pathological conditions. Currently, a large number of studies have shown that H2S mediates apoptosis through multiple signaling pathways to participate in cancer occurrence and development, for example, PI3K/Akt/mTOR and MAPK signaling pathways. Therefore, the regulation of the production and metabolism of H2S to mediate the apoptotic process of cancer cells may improve the effectiveness of cancer treatment. In this review, the role and mechanism of H2S in cancer cell apoptosis in mammals are summarized.
Collapse
Affiliation(s)
- Wei Gao
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Ya-Fang Liu
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Yan-Xia Zhang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Yan Wang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Yu-Qing Jin
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Hang Yuan
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Xiao-Yi Liang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Xin-Ying Ji
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan, 475004, China.
- Faculty of Basic Medical Subjects, Shu-Qing Medical College of Zhengzhou, Zhengzhou, Henan, 450064, China.
| | - Qi-Ying Jiang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan, 475004, China.
| | - Dong-Dong Wu
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan, 475004, China.
- School of Stomatology, Henan University, Kaifeng, Henan, 475004, China.
- Department of Stomatology, Huaihe Hospital of Henan University, Kaifeng, Henan, 475000, China.
| |
Collapse
|
34
|
Sánchez-Castillo A, Heylen E, Hounjet J, Savelkouls KG, Lieuwes NG, Biemans R, Dubois LJ, Reynders K, Rouschop KM, Vaes RDW, De Keersmaecker K, Lambrecht M, Hendriks LEL, De Ruysscher DKM, Vooijs M, Kampen KR. Targeting serine/glycine metabolism improves radiotherapy response in non-small cell lung cancer. Br J Cancer 2024; 130:568-584. [PMID: 38160212 PMCID: PMC10876524 DOI: 10.1038/s41416-023-02553-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 12/01/2023] [Accepted: 12/11/2023] [Indexed: 01/03/2024] Open
Abstract
BACKGROUND Lung cancer is the most lethal cancer, and 85% of cases are classified as non-small cell lung cancer (NSCLC). Metabolic rewiring is a cancer hallmark that causes treatment resistance, and lacks insights into serine/glycine pathway adaptations upon radiotherapy. METHODS We analyzed radiotherapy responses using mass-spectrometry-based metabolomics in NSCLC patient's plasma and cell lines. Efficacy of serine/glycine conversion inhibitor sertraline with radiotherapy was investigated by proliferation, clonogenic and spheroid assays, and in vivo using a serine/glycine dependent NSCLC mouse model by assessment of tumor growth, metabolite and cytokine levels, and immune signatures. RESULTS Serine/glycine pathway metabolites were significantly consumed in response to radiotherapy in NSCLC patients and cell models. Combining sertraline with radiotherapy impaired NSCLC proliferation, clonogenicity and stem cell self-renewal capacity. In vivo, NSCLC tumor growth was reduced solely in the sertraline plus radiotherapy combination treatment group. Tumor weights linked to systemic serine/glycine pathway metabolite levels, and were inhibited in the combination therapy group. Interestingly, combination therapy reshaped the tumor microenvironment via cytokines associated with natural killer cells, supported by eradication of immune checkpoint galectin-1 and elevated granzyme B levels. CONCLUSION Our findings highlight that targeting serine/glycine metabolism using sertraline restricts cancer cell recovery from radiotherapy and provides tumor control through immunomodulation in NSCLC.
Collapse
Affiliation(s)
- Anaís Sánchez-Castillo
- Department of Radiation Oncology (MAASTRO), GROW School for Oncology and Reproduction, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Elien Heylen
- Department of Oncology, Laboratory for Disease Mechanisms in Cancer, KU Leuven, and Leuven Cancer Institute (LKI), Herestraat 49, 3000, Leuven, Belgium
| | - Judith Hounjet
- Department of Radiation Oncology (MAASTRO), GROW School for Oncology and Reproduction, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Kim G Savelkouls
- Department of Radiation Oncology (MAASTRO), GROW School for Oncology and Reproduction, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Natasja G Lieuwes
- Department of Precision Medicine, The M-Lab, GROW School for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands
| | - Rianne Biemans
- Department of Precision Medicine, The M-Lab, GROW School for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands
| | - Ludwig J Dubois
- Department of Precision Medicine, The M-Lab, GROW School for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands
| | - Kobe Reynders
- Department of Radiation Oncology (MAASTRO), GROW School for Oncology and Reproduction, Maastricht University Medical Center+, Maastricht, The Netherlands
- Department of Oncology, Experimental Radiation Oncology, KU Leuven, and Leuven Cancer Institute (LKI), Herestraat 49, 3000, Leuven, Belgium
| | - Kasper M Rouschop
- Department of Radiation Oncology (MAASTRO), GROW School for Oncology and Reproduction, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Rianne D W Vaes
- Department of Radiation Oncology (MAASTRO), GROW School for Oncology and Reproduction, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Kim De Keersmaecker
- Department of Oncology, Laboratory for Disease Mechanisms in Cancer, KU Leuven, and Leuven Cancer Institute (LKI), Herestraat 49, 3000, Leuven, Belgium
| | - Maarten Lambrecht
- Department of Radiation Oncology, University Hospital Leuven, Leuven, Belgium
| | - Lizza E L Hendriks
- Department of Pulmonology, GROW School for Oncology and Reproduction, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Dirk K M De Ruysscher
- Department of Radiation Oncology (MAASTRO), GROW School for Oncology and Reproduction, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Marc Vooijs
- Department of Radiation Oncology (MAASTRO), GROW School for Oncology and Reproduction, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Kim R Kampen
- Department of Radiation Oncology (MAASTRO), GROW School for Oncology and Reproduction, Maastricht University Medical Center+, Maastricht, The Netherlands.
- Department of Oncology, Laboratory for Disease Mechanisms in Cancer, KU Leuven, and Leuven Cancer Institute (LKI), Herestraat 49, 3000, Leuven, Belgium.
| |
Collapse
|
35
|
Dai W, Liu Z, Yan M, Nian X, Hong F, Zhou Z, Wang C, Fu X, Li X, Jiang M, Zhu Y, Huang Q, Lu X, Hou L, Yan N, Wang Q, Hu J, Mo W, Zhang X, Zhang L. Nucleoporin Seh1 controls murine neocortical development via transcriptional repression of p21 in neural stem cells. Dev Cell 2024; 59:482-495.e6. [PMID: 38272027 DOI: 10.1016/j.devcel.2024.01.002] [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: 11/08/2022] [Revised: 06/21/2023] [Accepted: 01/05/2024] [Indexed: 01/27/2024]
Abstract
Mutations or dysregulation of nucleoporins (Nups) are strongly associated with neural developmental diseases, yet the underlying mechanisms remain poorly understood. Here, we show that depletion of Nup Seh1 in radial glial progenitors results in defective neural progenitor proliferation and differentiation that ultimately manifests in impaired neurogenesis and microcephaly. This loss of stem cell proliferation is not associated with defects in the nucleocytoplasmic transport. Rather, transcriptome analysis showed that ablation of Seh1 in neural stem cells derepresses the expression of p21, and knockdown of p21 partially restored self-renewal capacity. Mechanistically, Seh1 cooperates with the NuRD transcription repressor complex at the nuclear periphery to regulate p21 expression. Together, these findings identified that Nups regulate brain development by exerting a chromatin-associated role and affecting neural stem cell proliferation.
Collapse
Affiliation(s)
- Wenxiu Dai
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Department of Neuroscience, the First Affiliated Hospital, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen 361102, Fujian, China
| | - Zhixiong Liu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Department of Neuroscience, the First Affiliated Hospital, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen 361102, Fujian, China; Guangdong Institute of Intelligence Science and Technology, Hengqin, Zhuhai 519031, China
| | - Minbiao Yan
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Department of Neuroscience, the First Affiliated Hospital, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen 361102, Fujian, China
| | - Ximing Nian
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Department of Neuroscience, the First Affiliated Hospital, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen 361102, Fujian, China
| | - Fan Hong
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Department of Neuroscience, the First Affiliated Hospital, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen 361102, Fujian, China
| | - Zhihao Zhou
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Department of Neuroscience, the First Affiliated Hospital, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen 361102, Fujian, China
| | - Chaomeng Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Department of Neuroscience, the First Affiliated Hospital, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen 361102, Fujian, China
| | - Xing Fu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Department of Neuroscience, the First Affiliated Hospital, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen 361102, Fujian, China
| | - Xuewen Li
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Department of Neuroscience, the First Affiliated Hospital, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen 361102, Fujian, China
| | - Mengyun Jiang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Department of Neuroscience, the First Affiliated Hospital, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen 361102, Fujian, China
| | - Yanqin Zhu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Department of Neuroscience, the First Affiliated Hospital, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen 361102, Fujian, China
| | - Qiuying Huang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Department of Neuroscience, the First Affiliated Hospital, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen 361102, Fujian, China
| | - Xiaoyun Lu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Department of Neuroscience, the First Affiliated Hospital, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen 361102, Fujian, China
| | - Lichao Hou
- Department of Anesthesiology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361102, Fujian, China
| | - Ning Yan
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Qin Wang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jin Hu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Department of Neuroscience, the First Affiliated Hospital, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen 361102, Fujian, China
| | - Wei Mo
- Sir Run Run Shaw Hospital, Department of Immunology, School of Basic Medical Science, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, Hangzhou 311121, China
| | - Xueqin Zhang
- Department of Gynaecology and Obstetrics, Women and Children's Hospital Affiliated to Xiamen University, Xiamen University, Xiamen 361102, Fujian, China
| | - Liang Zhang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Department of Neuroscience, the First Affiliated Hospital, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen 361102, Fujian, China; Department of Gynaecology and Obstetrics, Women and Children's Hospital Affiliated to Xiamen University, Xiamen University, Xiamen 361102, Fujian, China.
| |
Collapse
|
36
|
Saba KH, Difilippo V, Kovac M, Cornmark L, Magnusson L, Nilsson J, van den Bos H, Spierings DC, Bidgoli M, Jonson T, Sumathi VP, Brosjö O, Staaf J, Foijer F, Styring E, Nathrath M, Baumhoer D, Nord KH. Disruption of the TP53 locus in osteosarcoma leads to TP53 promoter gene fusions and restoration of parts of the TP53 signalling pathway. J Pathol 2024; 262:147-160. [PMID: 38010733 DOI: 10.1002/path.6219] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 08/24/2023] [Accepted: 09/19/2023] [Indexed: 11/29/2023]
Abstract
TP53 is the most frequently mutated gene in human cancer. This gene shows not only loss-of-function mutations but also recurrent missense mutations with gain-of-function activity. We have studied the primary bone malignancy osteosarcoma, which harbours one of the most rearranged genomes of all cancers. This is odd since it primarily affects children and adolescents who have not lived the long life thought necessary to accumulate massive numbers of mutations. In osteosarcoma, TP53 is often disrupted by structural variants. Here, we show through combined whole-genome and transcriptome analyses of 148 osteosarcomas that TP53 structural variants commonly result in loss of coding parts of the gene while simultaneously preserving and relocating the promoter region. The transferred TP53 promoter region is fused to genes previously implicated in cancer development. Paradoxically, these erroneously upregulated genes are significantly associated with the TP53 signalling pathway itself. This suggests that while the classical tumour suppressor activities of TP53 are lost, certain parts of the TP53 signalling pathway that are necessary for cancer cell survival and proliferation are retained. In line with this, our data suggest that transposition of the TP53 promoter is an early event that allows for a new normal state of genome-wide rearrangements in osteosarcoma. © 2023 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Karim H Saba
- Department of Laboratory Medicine, Division of Clinical Genetics, Lund University, Lund, Sweden
| | - Valeria Difilippo
- Department of Laboratory Medicine, Division of Clinical Genetics, Lund University, Lund, Sweden
| | - Michal Kovac
- Bone Tumour Reference Centre at the Institute of Pathology, University Hospital and University of Basel, Basel, Switzerland
- Faculty of Informatics and Information Technologies, Slovak University of Technology, Bratislava, Slovakia
| | - Louise Cornmark
- Department of Laboratory Medicine, Division of Clinical Genetics, Lund University, Lund, Sweden
| | - Linda Magnusson
- Department of Laboratory Medicine, Division of Clinical Genetics, Lund University, Lund, Sweden
| | - Jenny Nilsson
- Department of Laboratory Medicine, Division of Clinical Genetics, Lund University, Lund, Sweden
| | - Hilda van den Bos
- European Research Institute for the Biology of Ageing, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Diana Cj Spierings
- European Research Institute for the Biology of Ageing, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Mahtab Bidgoli
- Department of Clinical Genetics and Pathology, Laboratory Medicine, Medical Services, Skåne University Hospital, Lund, Sweden
| | - Tord Jonson
- Department of Clinical Genetics and Pathology, Laboratory Medicine, Medical Services, Skåne University Hospital, Lund, Sweden
| | - Vaiyapuri P Sumathi
- Department of Musculoskeletal Pathology, Royal Orthopaedic Hospital, Birmingham, UK
| | - Otte Brosjö
- Department of Orthopedics, Karolinska University Hospital, Stockholm, Sweden
| | - Johan Staaf
- Department of Clinical Sciences, Division of Oncology and Pathology, Lund University, Lund, Sweden
| | - Floris Foijer
- European Research Institute for the Biology of Ageing, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Emelie Styring
- Department of Orthopedics, Lund University, Skåne University Hospital, Lund, Sweden
| | - Michaela Nathrath
- Children's Cancer Research Centre and Department of Pediatrics, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
- Department of Pediatric Oncology, Klinikum Kassel, Kassel, Germany
| | - Daniel Baumhoer
- Bone Tumour Reference Centre at the Institute of Pathology, University Hospital and University of Basel, Basel, Switzerland
| | - Karolin H Nord
- Department of Laboratory Medicine, Division of Clinical Genetics, Lund University, Lund, Sweden
| |
Collapse
|
37
|
Li K, You G, Jiang K, Wang R, Li W, Meng Y, Fang Y, Chen W, Zhu G, Song J, Wang W, Su H, Hu B, Sun F, Jia Z, Li C, Zhu J. Root extract of Hemsleya amabilis Diels suppresses renal cell carcinoma cell growth through inducing apoptosis and G 2/M phase arrest via PI3K/AKT signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:117014. [PMID: 37557938 DOI: 10.1016/j.jep.2023.117014] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 06/16/2023] [Accepted: 08/07/2023] [Indexed: 08/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Hemsleya amabilis Diels, belongs to cucurbitaceae, was traditional Chinese medicine (TCM). It is widely used to treat various diseases. However, these diseases may contribute to the development of RCC. AIM OF THE STUDY investigated the anticancer activities of root extract of Hemsleya amabilis Diels (HRE), and elucidated the underlying molecular mechanism in vivo and in vitro. MATERIALS AND METHODS Dried Hemsleya amabilis Diels roots were extracted by ethyl acetate and used to treat RCC4, OS-RC-2 and ACHN cells. UHPLC-MS was used to analyze the chemical composition of the extract. CCK-8 and colony formation assay were used to investigate proliferation. PI staining was used to detect cell cycle. Annexin-V-FITC, AO/EB and TEM were used to evaluate apoptosis. Transwell and wound healing assays were used to evaluate migration and invasion. RNA-seq, Network pharmacology, autodocking for virtual screening and molecular dynamics simulation were used to analyze potential molecular mechanisms and active components of HRE inhibiting proliferation of RCC. LY294002 and UC2288 were used to inhibit PI3K and P21 expression, respectively. IGF-1 was used to activate PI3K. Xenograft tumor model was established to evaluate its anti-tumor potential in vivo. Immunohistochemistry and Western blot were used to test protein expression levels. H&E staining was used to explore the side effects of HRE in vivo. Applying bioinformatics to analyze the effect of P21 on RCC. RESULTS HRE consists of 739 compounds. CCK-8 and colony formation assay showed that HRE significantly inhibited RCC cells proliferation. PI staining indicated that HRE caused G2/M phase arrest. Annexin-V-FITC, AO/EB and TEM experiments revealed that HRE significantly promoted apoptosis of RCC cells. Transwell and wound healing assays showed that HRE can inhibit the migration and invasion of RCC cells. RNA-seq showed that HRE induced 230 gene changes. Network pharmacology analysis found the relationship between HRE-component-target-RCC. Auto-docking found that Epitulipinolide diepoxide in HRE can stably bind to PIK3CA (-7.22 kJ/mol), and molecular dynamics simulation verified the combination between Epitulipinolide diepoxide of PIK3CA. In RCC4 cells, pretreatment with IGF-1, attenuated HRE-induced apoptosis and G2/M arrest. When pretreated with PIK3 inhibitor LY294002, the opposite result appears. Pretreatment with CDKN1A (P21) inhibitor UC2288 attenuated HRE-induced G2/M arrest. Xenograft tumor model showed that HRE inhibited tumor growth. Western blot analysis indicated that HRE can regulating Bax, Bcl-2, PARP, cleared-PARP, Caspase-9, Caspase-8, Caspase-3, Survivin, Cyclin-B1, CDK1, N-cadherin, snail, slug, E-cadherin, MMP-9. Immunohistochemical staining showed that in the treated group, expression of E-cadherin, Bax, P21 was up-regulated, while N-cadherin, PI3K, AKT and Bcl-2 were down-regulated. H&E staining showed that compared to control groups, the main organs in the HRE-treated groups showed no histological abnormalities. The overall survival rate of RCC patients in the high-expression group of P21 was higher than in the low-expression group of P21 on bioinformatics analysis. CONCLUSIONS HRE inhibited RCC migration and invasion through EMT, and inhibited proliferation in vivo and in vitro. In addition, HRE inhibited proliferation through promoting apoptosis and P21-induced G2/M phase arrest via PI3K/AKT signaling pathway. Overall, these results suggest that HRE may be a promising chemotherapy agent for RCC.
Collapse
Affiliation(s)
- Kai Li
- Guizhou Provincial Key Laboratory for Rare Animal and Economic Insect of the Mountainous Region, College of Biology and Environmental Engineering, Guiyang University, Guiyang, 550025, China
| | - Ganhua You
- The Second People's Hospital of Guizhou Province, Guiyang, 550002, China
| | - Kehua Jiang
- Department of Urology, Guizhou Provincial People's Hospital, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou Province, 550002, China
| | - Rongpin Wang
- Department of Radiology, Guizhou Provincial People's Hospital, Guiyang, Guizhou, 550002, China
| | - Wuchao Li
- Department of Radiology, Guizhou Provincial People's Hospital, Guiyang, Guizhou, 550002, China
| | - Yonglu Meng
- Guizhou Provincial Key Laboratory for Rare Animal and Economic Insect of the Mountainous Region, College of Biology and Environmental Engineering, Guiyang University, Guiyang, 550025, China
| | - Yinyi Fang
- Medical College of Guizhou University, Guiyang, Guizhou Province, 550025, China
| | - Weiming Chen
- Medical College of Guizhou University, Guiyang, Guizhou Province, 550025, China
| | - Guohua Zhu
- Department of Pedictric, Guizhou Provincial People's Hospital, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou Province, 550002, China
| | - Jukun Song
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Guizhou Medical University, Guiyang, China
| | - Wei Wang
- Department of Pedictric, Guizhou Provincial People's Hospital, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou Province, 550002, China
| | - Hao Su
- Department of Urology, Guizhou Provincial People's Hospital, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou Province, 550002, China
| | - Bin Hu
- Department of Urology, Kweichow Moutai Hospital, Renhuai, China
| | - Fa Sun
- Department of Urology, Guizhou Provincial People's Hospital, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou Province, 550002, China.
| | - Zhenyu Jia
- University of California of Riverside, Riverside, CA, 92521, USA.
| | - Can Li
- Guizhou Provincial Key Laboratory for Rare Animal and Economic Insect of the Mountainous Region, College of Biology and Environmental Engineering, Guiyang University, Guiyang, 550025, China.
| | - Jianguo Zhu
- Guizhou Provincial Key Laboratory for Rare Animal and Economic Insect of the Mountainous Region, College of Biology and Environmental Engineering, Guiyang University, Guiyang, 550025, China; Department of Urology, Guizhou Provincial People's Hospital, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou Province, 550002, China.
| |
Collapse
|
38
|
Baik R, Cromer MK, Glenn SE, Vakulskas CA, Chmielewski KO, Dudek AM, Feist WN, Klermund J, Shipp S, Cathomen T, Dever DP, Porteus MH. Transient inhibition of 53BP1 increases the frequency of targeted integration in human hematopoietic stem and progenitor cells. Nat Commun 2024; 15:111. [PMID: 38169468 PMCID: PMC10762240 DOI: 10.1038/s41467-023-43413-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 11/08/2023] [Indexed: 01/05/2024] Open
Abstract
Genome editing by homology directed repair (HDR) is leveraged to precisely modify the genome of therapeutically relevant hematopoietic stem and progenitor cells (HSPCs). Here, we present a new approach to increasing the frequency of HDR in human HSPCs by the delivery of an inhibitor of 53BP1 (named "i53") as a recombinant peptide. We show that the use of i53 peptide effectively increases the frequency of HDR-mediated genome editing at a variety of therapeutically relevant loci in HSPCs as well as other primary human cell types. We show that incorporating the use of i53 recombinant protein allows high frequencies of HDR while lowering the amounts of AAV6 needed by 8-fold. HDR edited HSPCs were capable of long-term and bi-lineage hematopoietic reconstitution in NSG mice, suggesting that i53 recombinant protein might be safely integrated into the standard CRISPR/AAV6-mediated genome editing protocol to gain greater numbers of edited cells for transplantation of clinically meaningful cell populations.
Collapse
Affiliation(s)
- Ron Baik
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Lorry I. Lokey Stem Cell Research Building, 265 Campus Drive, Stanford, CA, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Molecular Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - M Kyle Cromer
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Lorry I. Lokey Stem Cell Research Building, 265 Campus Drive, Stanford, CA, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Steve E Glenn
- Integrated DNA Technologies, Inc., Coralville, IA, USA
| | | | - Kay O Chmielewski
- Institute for Transfusion Medicine and Gene Therapy, Medical Center - University of Freiburg, 79106, Freiburg, Germany
- Center for Chronic Immunodeficiency, University of Freiburg, 79106, Freiburg, Germany
- Ph.D. Program, Faculty of Biology, University of Freiburg, 79104, Freiburg, Germany
| | - Amanda M Dudek
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Lorry I. Lokey Stem Cell Research Building, 265 Campus Drive, Stanford, CA, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - William N Feist
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Lorry I. Lokey Stem Cell Research Building, 265 Campus Drive, Stanford, CA, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Julia Klermund
- Institute for Transfusion Medicine and Gene Therapy, Medical Center - University of Freiburg, 79106, Freiburg, Germany
- Center for Chronic Immunodeficiency, University of Freiburg, 79106, Freiburg, Germany
| | - Suzette Shipp
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Lorry I. Lokey Stem Cell Research Building, 265 Campus Drive, Stanford, CA, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Toni Cathomen
- Institute for Transfusion Medicine and Gene Therapy, Medical Center - University of Freiburg, 79106, Freiburg, Germany
- Center for Chronic Immunodeficiency, University of Freiburg, 79106, Freiburg, Germany
| | - Daniel P Dever
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Lorry I. Lokey Stem Cell Research Building, 265 Campus Drive, Stanford, CA, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Matthew H Porteus
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Lorry I. Lokey Stem Cell Research Building, 265 Campus Drive, Stanford, CA, USA.
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
39
|
Fan N, Zhang L, Wang Z, Ding H, Yue Z. Ivermectin Inhibits Bladder Cancer Cell Growth and Induces Oxidative Stress and DNA Damage. Anticancer Agents Med Chem 2024; 24:348-357. [PMID: 38375808 DOI: 10.2174/0118715206274095231106042833] [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: 07/25/2023] [Revised: 10/05/2023] [Accepted: 10/16/2023] [Indexed: 02/21/2024]
Abstract
BACKGROUND Bladder cancer is the most common malignant tumor of the urinary system. Nevertheless, current therapies do not provide satisfactory results. It is imperative that novel strategies should be developed for treating bladder cancer. OBJECTIVES To evaluate the effect of a broad-spectrum anti-parasitic agent, Ivermectin, on bladder cancer cells in vitro and in vivo. METHODS CCK-8 and EdU incorporation assays were used to evaluate cell proliferation. Apoptosis was detected by flow cytometry, TUNEL assay, and western blotting. Flow cytometry and DCFH-DA assay were used to analyze the reactive oxygen species (ROS) levels. DNA damage was determined by Neutral COMET assay and γ H2AX expression. Proteins related to apoptosis and DNA damage pathways were determined by WB assay. Xenograft tumor models in nude mice were used to investigate the anti-cancer effect of Ivermectin in vivo. RESULTS Our study showed that in vitro and in vivo, Ivermectin inhibited the growth of bladder cancer cells. In addition, Ivermectin could induce apoptosis, ROS production, DNA damage, and activate ATM/P53 pathwayrelated proteins in bladder cancer cells. CONCLUSIONS According to these findings, Ivermectin may be a potential therapeutic candidate against bladder cancer due to its significant anti-cancer effect.
Collapse
Affiliation(s)
- Ning Fan
- Institute of Urology, Key Laboratory of Gansu Urological Diseases, Gansu Nephro-Urological Clinical Center, Department of Urology, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Lixiu Zhang
- Department of Clinical Laboratory, Maternal and Child Health Hospital of Gansu. Lanzhou, 730050, China
| | - Zhiping Wang
- Institute of Urology, Key Laboratory of Gansu Urological Diseases, Gansu Nephro-Urological Clinical Center, Department of Urology, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Hui Ding
- Institute of Urology, Key Laboratory of Gansu Urological Diseases, Gansu Nephro-Urological Clinical Center, Department of Urology, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Zhongjin Yue
- Institute of Urology, Key Laboratory of Gansu Urological Diseases, Gansu Nephro-Urological Clinical Center, Department of Urology, Lanzhou University Second Hospital, Lanzhou, 730030, China
| |
Collapse
|
40
|
Sun F, Sutovsky P, Patterson AL, Balboula AZ. Mechanisms of DNA Damage Response in Mammalian Oocytes. ADVANCES IN ANATOMY, EMBRYOLOGY, AND CELL BIOLOGY 2024; 238:47-68. [PMID: 39030354 DOI: 10.1007/978-3-031-55163-5_3] [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: 07/21/2024]
Abstract
DNA damage poses a significant challenge to all eukaryotic cells, leading to mutagenesis, genome instability and senescence. In somatic cells, the failure to repair damaged DNA can lead to cancer development, whereas, in oocytes, it can lead to ovarian dysfunction and infertility. The response of the cell to DNA damage entails a series of sequential and orchestrated events including sensing the DNA damage, activating DNA damage checkpoint, chromatin-related conformational changes, activating the DNA damage repair machinery and/or initiating the apoptotic cascade. This chapter focuses on how somatic cells and mammalian oocytes respond to DNA damage. Specifically, we will discuss how and why fully grown mammalian oocytes differ drastically from somatic cells and growing oocytes in their response to DNA damage.
Collapse
Affiliation(s)
- Fei Sun
- Division of Animal Sciences, University of Missouri, Columbia, MO, USA
| | - Peter Sutovsky
- Division of Animal Sciences, University of Missouri, Columbia, MO, USA
- Department of Obstetrics, Gynecology and Women's Health, University of Missouri, Columbia, MO, USA
| | - Amanda L Patterson
- Division of Animal Sciences, University of Missouri, Columbia, MO, USA
- Department of Obstetrics, Gynecology and Women's Health, University of Missouri, Columbia, MO, USA
| | - Ahmed Z Balboula
- Division of Animal Sciences, University of Missouri, Columbia, MO, USA.
| |
Collapse
|
41
|
Li M, Yang J, Li J, Zhou Y, Li X, Ma Z, Li X, Ma H, Ye X. Epiberberine induced p53/p21-dependent G2/M cell cycle arrest and cell apoptosis in gastric cancer cells by activating γ-aminobutyric acid receptor- β3. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 123:155198. [PMID: 38006806 DOI: 10.1016/j.phymed.2023.155198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 11/03/2023] [Accepted: 11/07/2023] [Indexed: 11/27/2023]
Abstract
BACKGROUND AND PURPOSE Epiberberine (EPI) is one of the most important bioalkaloid found in the rhizome of Coptis chinensis, which has been observed to exhibit pharmaceutical effects against gastric cancer (GC). Nevertheless, the potential mechanism of EPI against GC cells still remains unclear. This study aimed to identify the core receptor on GC cells through which EPI inhibited the growth of GC cells and to explore the underlying inhibitory mechanisms. METHODS To identify hub receptor targets that respond to EPI treatment, RNA sequencing (RNA-Seq) data from a tumor-bearing mouse model were analyzed using bioinformatics method and molecular docking. The binding interaction between EPI and GABRB3 was validated through western blotting based-cellular thermal shift assay (WB-CETSA). To further verify the binding region between EPI and GABRB3 through circular dichroism (CD) chromatography, fragments of the extracellular and transmembrane domains of the GABRB3 protein were expressed and purified in vitro. Stable cell lines with the overexpression or knockdown of GABRB3 were established using the recombinant lentivirus system. MTT ((3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyl tetrazolium bromide)) assay, colony formation assay, invasion and migration experiments, and flow cytometry were conducted to validate the inhibitory effect of EPI on the GC cells via GABRB3. Additionally, western blotting was utilized to explore the potential inhibitory mechanisms. RESULTS Through the combination of multiple bioinformatics methods and molecular docking, we found that the γ-aminobutyric acid type A receptor subunit -β3 (GABRB3) might be the critical receptor target in response to EPI treatment. The results of WB-CETSA analysis indicated that EPI significantly promoted the thermostability of the GABRB3 protein. Importantly, EPI could directly bind to GABRB3 and alter the secondary structure of GABRB3 fragments similar to the natural agonist, γ-aminobutyric acid (GABA). The EPI-induced suppression of the malignant phenotype of GC cells was dependent on the presence of GABRB3. GABRB3 expression was positively correlated with TP53 in patients with GC. The binding of EPI to GABRB3 stimulated p53 accumulation in GC cells. This activated the p21/CDK1/cyclinB1 pathway, resulting in G2/M cell cycle arrest, and induced the Bcl-2/BAX/Caspase axis-dependent cell apoptosis. CONCLUSION This study revealed the target receptor for EPI in GC cells and provided new insights into its anticancer mechanisms.
Collapse
Affiliation(s)
- Mengmeng Li
- Engineering Research Center of Coptis Development and Utilization (Ministry of Education), School of Life Sciences, Southwest University, Chongqing 400715, China
| | - Jiaye Yang
- Engineering Research Center of Coptis Development and Utilization (Ministry of Education), School of Life Sciences, Southwest University, Chongqing 400715, China
| | - Juan Li
- School of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China
| | - Yuan Zhou
- Engineering Research Center of Coptis Development and Utilization (Ministry of Education), School of Life Sciences, Southwest University, Chongqing 400715, China
| | - Xiaoduo Li
- Engineering Research Center of Coptis Development and Utilization (Ministry of Education), School of Life Sciences, Southwest University, Chongqing 400715, China
| | - Zhengcai Ma
- Engineering Research Center of Coptis Development and Utilization (Ministry of Education), School of Life Sciences, Southwest University, Chongqing 400715, China
| | - Xuegang Li
- School of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China
| | - Hang Ma
- School of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China.
| | - Xiaoli Ye
- Engineering Research Center of Coptis Development and Utilization (Ministry of Education), School of Life Sciences, Southwest University, Chongqing 400715, China.
| |
Collapse
|
42
|
Liu L, Du Y, Du Y, Yan W, Li Y, Cui K, Li Z, Yu P, Zhang W, Feng J, Ma W, Zhao H. Exopolysaccharide from Weissella confusa J4-1 inhibits colorectal cancer via induction of cell cycle arrest. Int J Biol Macromol 2023; 253:127625. [PMID: 37884233 DOI: 10.1016/j.ijbiomac.2023.127625] [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: 09/17/2022] [Revised: 10/19/2023] [Accepted: 10/21/2023] [Indexed: 10/28/2023]
Abstract
Exopolysaccharide (EPS), a bioproduct of lactic acid bacteria (LAB), has various health-promoting biological activities that may be beneficial for cancer therapy. This in vivo and in vitro study aimed to elucidate the anti-colorectal cancer (CRC) capacity of a homopolysaccharide EPS obtained from Weissella confusa J4-1 (EPSJ4-1) isolated from the faeces of healthy infants. We confirmed that EPSJ4-1 contained glucose and effectively suppressed the proliferation, migration, and invasion of CRC cells. EPSJ4-1 treatment significantly retarded the growth of HT-29 tumour xenografts without causing cytotoxicity to normal organs. EPSJ4-1 exerts an inhibitory effect on cell proliferation by inducing G0/G1 phase cell cycle arrest in CRC cells. Furthermore, EPSJ4-1 upregulated p21 levels and downregulated mutant p53 and cyclin kinase 2 levels. This is the first study to demonstrate the antitumour effects of EPS from W. confusa on CRC via cell cycle arrest and inhibition of cell migration and invasion, suggesting that EPSJ4-1 has the potential to be developed as a nutraceutical or pharmaceutical drug to prevent and treat CRC.
Collapse
Affiliation(s)
- Lei Liu
- Oncology department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Yurong Du
- Oncology department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Yabing Du
- Oncology department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Weiliang Yan
- Oncology department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Yuanzhe Li
- Department of Pediatrics, Children's Hospital Affiliated of Zhengzhou University, Zhengzhou, China
| | - Kang Cui
- Oncology department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Zhen Li
- National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Heart Center of Henan Provincial People's Hospital, Central China Fuwai Hospital of Zhengzhou University, Fuwai Central China Cardiovascular Hospital & Central China Branch of National Center for Cardiovascular Diseases, Zhengzhou, Henan 451464, China
| | - Pu Yu
- Oncology department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - WanCun Zhang
- Department of Pediatrics, Children's Hospital Affiliated of Zhengzhou University, Zhengzhou, China
| | - Jianguo Feng
- Department of Anesthesiology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, China
| | - Wang Ma
- Oncology department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| | - Huan Zhao
- Oncology department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| |
Collapse
|
43
|
He MT, Nguyen QN, Cho EJ, Kim SH, Park S, Park JY, Lee S, Kang KS. Aloe-Emodin Isolated from Rheum Undulatum L. Regulates Cell Cycle Distribution and Cellular Senescence in Human Prostate Cancer LNCaP Cells. J Diet Suppl 2023; 21:389-407. [PMID: 38062982 DOI: 10.1080/19390211.2023.2284985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2023]
Abstract
Senescence can promote hyperplastic pathologies, such as cancer. Prostate cancer is the second most common type of cancer in men. The p21-mediate cellular senescence, facilitated through the tumor suppressor p53-dependent pathway, is considered the primary mechanism for cancer treatment. Aloe-emodin, has been reported to exert anticancer effects in various types of cancers. This study aimed to investigate the bioactivity of aloe-emodin in LNCaP cells via the activation of p21-mediated cellular senescence. Aloe-emodin treatment increased the percentage of cells in the G1 phase while decreasing the percentage in the S phase. This effect was reflected in the expression levels of proteins associated with cell cycle progression, such as p21CIP, retinoblastoma protein, and cyclin-dependent kinase2/4 in LNCaP cells. However, aloe-emodin-treated LNCaP cells did not induce cell cycle arrest at G2/M checkpoint. Moreover, increased senescence-associated-galactosidase activity was observed in a dose-dependent manner following treatment with aloe-emodin. Aloe-emodin also induced DNA damage by modulating the expression of histone H2AX and lamin B1. Furthermore, aloe-emodin inhibited the proliferation of LNCaP cells, contrasting with the exponential growth observed in the nontreated cells. Importantly, this inhibition did not impact the immune system, as evidenced by the increased proliferation of splenocytes isolated from mice. These findings provide preliminary evidence of the anticancer effect of aloe-emodin in LNCaP cells, necessitating further investigations into the underlying mechanisms in vivo and human subjects.
Collapse
Affiliation(s)
- Mei Tong He
- College of Korean Medicine, Gachon University, Seongnam, South Korea
| | - Quynh Nhu Nguyen
- College of Korean Medicine, Gachon University, Seongnam, South Korea
| | - Eun Ju Cho
- Department of Food Science & Nutrition, Pusan National University, Busan, South Korea
| | - Seung Hyun Kim
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon, South Korea
| | - SeonJu Park
- Chuncheon Center, Korea Basic Science Institute (KBSI), Chuncheon, South Korea
| | - Jun Yeon Park
- Department of Food Science and Biotechnology, Kyonggi University, Suwon, South Korea
| | - Sullim Lee
- Department of Life Science, College of Bio-Nano Technology, Gachon University, Seongnam, South Korea
| | - Ki Sung Kang
- College of Korean Medicine, Gachon University, Seongnam, South Korea
| |
Collapse
|
44
|
Chiang YC, Leu WJ, Chen YC, Ye PC, Hsu YT, Hsiao YC, Hsu JL, Chan SH, Hsu LC, Huang HS, Guh JH. Mechanistic study of dual-function inhibitors targeting topoisomerase II and Rad51-mediated DNA repair pathway against castration-resistant prostate cancer. Prostate 2023; 83:1549-1563. [PMID: 37583103 DOI: 10.1002/pros.24613] [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: 02/22/2023] [Revised: 06/17/2023] [Accepted: 07/24/2023] [Indexed: 08/17/2023]
Abstract
BACKGROUND Castration-resistant prostate cancer (CRPC) is refractory to hormone treatment and the therapeutic options are continuously advancing. This study aims to discover the anti-CRPC effects and underlying mechanisms of small-molecule compounds targeting topoisomerase (TOP) II and cellular components of DNA damage repair. METHODS Cell proliferation was determined in CRPC PC-3 and DU-145 cells using anchorage-dependent colony formation, sulforhodamine B assay and flow cytometric analysis of CFSE staining. Flow cytometric analyses of propidium iodide staining and JC-1 staining were used to examine the population of cell-cycle phases and mitochondrial membrane potential, respectively. Nuclear extraction was performed to detect the nuclear localization of cellular components in DNA repair pathways. Protein expressions were determined using Western blot analysis. RESULTS A series of azathioxanthone-based derivatives were synthesized and examined for bioactivities in which WC-A13, WC-A14, WC-A15, and WC-A16 displayed potent anti-CRPC activities in both PC-3 and DU-145 cell models. These WC-A compounds selectively downregulated both TOP IIα and TOP IIβ but not TOP I protein expression. WC-A13, WC-A14, and WC-A15 were more potent than WC-A16 on TOP II inhibition, mitochondrial dysfunction, and induction of caspase cascades indicating the key role of amine-containing side chain of the compounds in determining anti-CRPC activities. Furthermore, WC-A compounds induced an increase of γH2AX and activated ATR-Chk1 and ATM-Chk2 signaling pathways. P21 protein expression was also upregulated by WC-A compounds in which WC-A16 showed the least activity. Notably, WC-A compounds exhibited different regulation on Rad51, a major protein in homologous recombination of DNA in double-stranded break repair. WC-A13, WC-A14, and WC-A15 inhibited, whereas WC-A16 induced, the nuclear translocation of Rad51. CONCLUSION The data suggest that WC-A compounds exhibit anti-CRPC effects through the inhibition of TOP II activities, leading to mitochondrial stress-involved caspase activation and apoptosis. Moreover, WC-A13, WC-A14, and WC-A15 but not WC-A16 display inhibitory activities of Rad51-mediated DNA repair pathway which may increase apoptotic effect of CRPC cells.
Collapse
Affiliation(s)
- Yi-Chang Chiang
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Wohn-Jenn Leu
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yi-Chin Chen
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Pei-Chen Ye
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yu-Tung Hsu
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yu-Chi Hsiao
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Jui-Ling Hsu
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Pharmacy, New Taipei Municipal TuCheng Hospital, Chang Gung Memorial Hospital, New Taipei City, Taiwan
| | - She-Hung Chan
- Department of Cosmetic Science, Providence University, Taichung, Taiwan
| | - Lih-Ching Hsu
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Hsu-Shan Huang
- PhD Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei, Taiwan
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
- School of Pharmacy, National Defense Medical Center, Taipei, Taiwan
- PhD Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Jih-Hwa Guh
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
45
|
Melo ML, Fonseca R, Pauli F, Zavan B, Hanemann JAC, Miyazawa M, Caixeta ES, Nacif JLM, Aissa AF, Barreiro EJ, Ionta M. N-acylhydrazone derivative modulates cell cycle regulators promoting mitosis arrest and apoptosis in estrogen positive MCF-7 breast cancer cells. Toxicol In Vitro 2023; 93:105686. [PMID: 37652252 DOI: 10.1016/j.tiv.2023.105686] [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: 04/26/2023] [Revised: 07/14/2023] [Accepted: 08/28/2023] [Indexed: 09/02/2023]
Abstract
Breast cancer is the leading cause of cancer death among women worldwide. About 75% of all diagnosed cases are hormone-positive, which are treated with hormone therapy. However, many patients are refractory or become resistant to the drugs used in therapeutic protocols. In this scenario, it is essential to identify new substances with pharmacological potential against breast cancer. VEGFR2 inhibitors are considered promising antitumor agents not only due to their antiangiogenic activity but also by inhibiting the proliferation of tumor cells. Thus, the present study aimed to evaluate the effects of N-acylhydrazone derivative LASSBio-2029 on the proliferative behavior of MCF-7 cells. We observed a promising antitumor potential of this substance due to its ability to modulate critical cell cycle regulators including mitotic kinases (CDK1, AURKA, AURKB, and PLK1) and CDK inhibitor (CDKN1A). Increased frequencies of abnormal mitosis and apoptotic cells were observed in response to treatment. A molecular docking analysis predicts that LASSBio-2029 could bind to the proto-oncoprotein ABL1, which participates in cell cycle control, interacting with other controller proteins and regulating centrosome-associated tubulins. Finally, we created a gene signature with the downregulated genes, whose reduced expression is associated with a higher relapse-free survival probability in breast cancer patients.
Collapse
Affiliation(s)
- Melissa Lúcia Melo
- Institute of Biomedical Sciences, Federal University of Alfenas, MG 37130-001, Brazil
| | - Rafael Fonseca
- Institute of Biomedical Sciences, Federal University of Alfenas, MG 37130-001, Brazil
| | - Fernanda Pauli
- Institute of Chemistry, Fluminense Federal University, Niterói, RJ 24020-140, Brazil
| | - Bruno Zavan
- Institute of Biomedical Sciences, Federal University of Alfenas, MG 37130-001, Brazil
| | - João Adolfo Costa Hanemann
- Department of Clinic and Surgery, School of Dentistry. Federal University of Alfenas, 37130-001, MG, Brazil
| | - Marta Miyazawa
- Department of Clinic and Surgery, School of Dentistry. Federal University of Alfenas, 37130-001, MG, Brazil
| | | | | | - Alexandre Ferro Aissa
- Institute of Biomedical Sciences, Federal University of Alfenas, MG 37130-001, Brazil.
| | - Eliezer J Barreiro
- Laboratory of Evaluation and Synthesis of Bioactive Substances (LASSBio), Institute of Biomedical Sciences, Federal University of Rio de Janeiro, CCS, Rio de Janeiro, RJ, Brazil.
| | - Marisa Ionta
- Institute of Biomedical Sciences, Federal University of Alfenas, MG 37130-001, Brazil.
| |
Collapse
|
46
|
Ben-Oz BM, Machour FE, Nicola M, Argoetti A, Polyak G, Hanna R, Kleifeld O, Mandel-Gutfreund Y, Ayoub N. A dual role of RBM42 in modulating splicing and translation of CDKN1A/p21 during DNA damage response. Nat Commun 2023; 14:7628. [PMID: 37993446 PMCID: PMC10665399 DOI: 10.1038/s41467-023-43495-6] [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: 06/07/2022] [Accepted: 11/10/2023] [Indexed: 11/24/2023] Open
Abstract
p53-mediated cell cycle arrest during DNA damage is dependent on the induction of p21 protein, encoded by the CDKN1A gene. p21 inhibits cyclin-dependent kinases required for cell cycle progression to guarantee accurate repair of DNA lesions. Hence, fine-tuning of p21 levels is crucial to preserve genomic stability. Currently, the multilayered regulation of p21 levels during DNA damage is not fully understood. Herein, we identify the human RNA binding motif protein 42 (RBM42) as a regulator of p21 levels during DNA damage. Genome-wide transcriptome and interactome analysis reveals that RBM42 alters the expression of p53-regulated genes during DNA damage. Specifically, we demonstrate that RBM42 facilitates CDKN1A splicing by counteracting the splicing inhibitory effect of RBM4 protein. Unexpectedly, we also show that RBM42, underpins translation of various splicing targets, including CDKN1A. Concordantly, transcriptome-wide mapping of RBM42-RNA interactions using eCLIP further substantiates the dual function of RBM42 in regulating splicing and translation of its target genes, including CDKN1A. Collectively, our data show that RBM42 couples splicing and translation machineries to fine-tune gene expression during DNA damage response.
Collapse
Affiliation(s)
- Bella M Ben-Oz
- Department of Biology, Technion - Israel Institute of Technology, Haifa, 3200003, Israel
| | - Feras E Machour
- Department of Biology, Technion - Israel Institute of Technology, Haifa, 3200003, Israel
| | - Marian Nicola
- Department of Biology, Technion - Israel Institute of Technology, Haifa, 3200003, Israel
| | - Amir Argoetti
- Department of Biology, Technion - Israel Institute of Technology, Haifa, 3200003, Israel
| | - Galia Polyak
- Department of Biology, Technion - Israel Institute of Technology, Haifa, 3200003, Israel
| | - Rawad Hanna
- Department of Biology, Technion - Israel Institute of Technology, Haifa, 3200003, Israel
| | - Oded Kleifeld
- Department of Biology, Technion - Israel Institute of Technology, Haifa, 3200003, Israel
| | - Yael Mandel-Gutfreund
- Department of Biology, Technion - Israel Institute of Technology, Haifa, 3200003, Israel
| | - Nabieh Ayoub
- Department of Biology, Technion - Israel Institute of Technology, Haifa, 3200003, Israel.
| |
Collapse
|
47
|
Wang DX, Dong ZJ, Deng SX, Tian YM, Xiao YJ, Li X, Ma XR, Li L, Li P, Chang HZ, Liu L, Wang F, Wu Y, Gao X, Zheng SS, Gu HM, Zhang YN, Wu JB, Wu F, Peng Y, Zhang XW, Zhan RY, Gao LX, Sun Q, Guo X, Zhao XD, Luo JH, Zhou R, Han L, Shu Y, Zhao JW. GDF11 slows excitatory neuronal senescence and brain ageing by repressing p21. Nat Commun 2023; 14:7476. [PMID: 37978295 PMCID: PMC10656444 DOI: 10.1038/s41467-023-43292-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 11/06/2023] [Indexed: 11/19/2023] Open
Abstract
As a major neuron type in the brain, the excitatory neuron (EN) regulates the lifespan in C. elegans. How the EN acquires senescence, however, is unknown. Here, we show that growth differentiation factor 11 (GDF11) is predominantly expressed in the EN in the adult mouse, marmoset and human brain. In mice, selective knock-out of GDF11 in the post-mitotic EN shapes the brain ageing-related transcriptional profile, induces EN senescence and hyperexcitability, prunes their dendrites, impedes their synaptic input, impairs object recognition memory and shortens the lifespan, establishing a functional link between GDF11, brain ageing and cognition. In vitro GDF11 deletion causes cellular senescence in Neuro-2a cells. Mechanistically, GDF11 deletion induces neuronal senescence via Smad2-induced transcription of the pro-senescence factor p21. This work indicates that endogenous GDF11 acts as a brake on EN senescence and brain ageing.
Collapse
Affiliation(s)
- Di-Xian Wang
- Department of Pathology of Sir Run Run Shaw Hospital, and Department of Human Anatomy, Histology and Embryology, System Medicine Research Center, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, 310058, Hangzhou, Zhejiang, China
- Center of Cryo-Electron Microscopy, Zhejiang University, 310058, Hangzhou, Zhejiang, China
| | - Zhao-Jun Dong
- Department of Pathology of Sir Run Run Shaw Hospital, and Department of Human Anatomy, Histology and Embryology, System Medicine Research Center, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, 310058, Hangzhou, Zhejiang, China
- Center of Cryo-Electron Microscopy, Zhejiang University, 310058, Hangzhou, Zhejiang, China
| | - Sui-Xin Deng
- Department of Neurosurgery, Jinshan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Fudan University, 201508, Shanghai, China
| | | | - Yu-Jie Xiao
- Department of Neurosurgery, Jinshan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Fudan University, 201508, Shanghai, China
| | - Xinran Li
- The Global Scientific and Technological Innovation Center and the MOE Key Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, 310058, Hangzhou, Zhejiang, China
| | - Xiao-Ru Ma
- Department of Pathology of Sir Run Run Shaw Hospital, and Department of Human Anatomy, Histology and Embryology, System Medicine Research Center, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, 310058, Hangzhou, Zhejiang, China
- Center of Cryo-Electron Microscopy, Zhejiang University, 310058, Hangzhou, Zhejiang, China
| | - Liang Li
- Department of Neurosurgery, Jinshan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Fudan University, 201508, Shanghai, China
| | - Pengxiao Li
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai; Center for Systems Biomedicine, Shanghai Jiao Tong University, 200240, Shanghai, China
| | | | | | - Fan Wang
- Department of Pathology of Sir Run Run Shaw Hospital, and Department of Human Anatomy, Histology and Embryology, System Medicine Research Center, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, 310058, Hangzhou, Zhejiang, China
- Center of Cryo-Electron Microscopy, Zhejiang University, 310058, Hangzhou, Zhejiang, China
| | - Yang Wu
- Department of Pathology of Sir Run Run Shaw Hospital, and Department of Human Anatomy, Histology and Embryology, System Medicine Research Center, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, 310058, Hangzhou, Zhejiang, China
- Center of Cryo-Electron Microscopy, Zhejiang University, 310058, Hangzhou, Zhejiang, China
| | - Xiang Gao
- Department of Pathology of Sir Run Run Shaw Hospital, and Department of Human Anatomy, Histology and Embryology, System Medicine Research Center, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, 310058, Hangzhou, Zhejiang, China
- Center of Cryo-Electron Microscopy, Zhejiang University, 310058, Hangzhou, Zhejiang, China
| | - Shuang-Shuang Zheng
- Department of Pathology of Sir Run Run Shaw Hospital, and Department of Human Anatomy, Histology and Embryology, System Medicine Research Center, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, 310058, Hangzhou, Zhejiang, China
- Center of Cryo-Electron Microscopy, Zhejiang University, 310058, Hangzhou, Zhejiang, China
| | - Hui-Min Gu
- Department of Pathology of Sir Run Run Shaw Hospital, and Department of Human Anatomy, Histology and Embryology, System Medicine Research Center, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, 310058, Hangzhou, Zhejiang, China
- Center of Cryo-Electron Microscopy, Zhejiang University, 310058, Hangzhou, Zhejiang, China
| | - Ya-Nan Zhang
- Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Jian-Bin Wu
- Department of Pathology of Sir Run Run Shaw Hospital, and Department of Human Anatomy, Histology and Embryology, System Medicine Research Center, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, 310058, Hangzhou, Zhejiang, China
- Center of Cryo-Electron Microscopy, Zhejiang University, 310058, Hangzhou, Zhejiang, China
| | - Fan Wu
- Department of Neurosurgery, the First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, 310003, Hangzhou, China
| | - Yonglin Peng
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai; Center for Systems Biomedicine, Shanghai Jiao Tong University, 200240, Shanghai, China
| | - Xiao-Wen Zhang
- Department of Pathology of Sir Run Run Shaw Hospital, and Department of Human Anatomy, Histology and Embryology, System Medicine Research Center, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, 310058, Hangzhou, Zhejiang, China
- Center of Cryo-Electron Microscopy, Zhejiang University, 310058, Hangzhou, Zhejiang, China
| | - Ren-Ya Zhan
- Department of Neurosurgery, the First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, 310003, Hangzhou, China
| | - Li-Xia Gao
- Department of Neurology of the Second Affiliated Hospital, Interdisciplinary Institute of Neuroscience and Technology, Zhejiang University School of Medicine, 310020, Hangzhou, China
| | - Qiming Sun
- Department of Biochemistry, and Department of Cardiology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xing Guo
- Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Xiao-Dong Zhao
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai; Center for Systems Biomedicine, Shanghai Jiao Tong University, 200240, Shanghai, China
| | - Jian-Hong Luo
- Department of Neurobiology and Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 310058, Hangzhou, Zhejiang, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Zhejiang, China
| | - Ruhong Zhou
- Institute of Quantitative Biology, College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Lei Han
- BGI Research, 310030, Hangzhou, China.
| | - Yousheng Shu
- Department of Neurosurgery, Jinshan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Fudan University, 201508, Shanghai, China.
| | - Jing-Wei Zhao
- Department of Pathology of Sir Run Run Shaw Hospital, and Department of Human Anatomy, Histology and Embryology, System Medicine Research Center, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, 310058, Hangzhou, Zhejiang, China.
- Center of Cryo-Electron Microscopy, Zhejiang University, 310058, Hangzhou, Zhejiang, China.
| |
Collapse
|
48
|
Manohar S, Estrada ME, Uliana F, Vuina K, Alvarez PM, de Bruin RAM, Neurohr GE. Genome homeostasis defects drive enlarged cells into senescence. Mol Cell 2023; 83:4032-4046.e6. [PMID: 37977116 PMCID: PMC10659931 DOI: 10.1016/j.molcel.2023.10.018] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 06/30/2023] [Accepted: 10/16/2023] [Indexed: 11/19/2023]
Abstract
Cellular senescence refers to an irreversible state of cell-cycle arrest and plays important roles in aging and cancer biology. Because senescence is associated with increased cell size, we used reversible cell-cycle arrests combined with growth rate modulation to study how excessive growth affects proliferation. We find that enlarged cells upregulate p21, which limits cell-cycle progression. Cells that re-enter the cell cycle encounter replication stress that is well tolerated in physiologically sized cells but causes severe DNA damage in enlarged cells, ultimately resulting in mitotic failure and permanent cell-cycle withdrawal. We demonstrate that enlarged cells fail to recruit 53BP1 and other non-homologous end joining (NHEJ) machinery to DNA damage sites and fail to robustly initiate DNA damage-dependent p53 signaling, rendering them highly sensitive to genotoxic stress. We propose that an impaired DNA damage response primes enlarged cells for persistent replication-acquired damage, ultimately leading to cell division failure and permanent cell-cycle exit.
Collapse
Affiliation(s)
- Sandhya Manohar
- Institute for Biochemistry, Department of Biology, ETH Zürich 8093, Zürich, Zürich, Switzerland
| | - Marianna E Estrada
- Institute for Biochemistry, Department of Biology, ETH Zürich 8093, Zürich, Zürich, Switzerland
| | - Federico Uliana
- Institute for Biochemistry, Department of Biology, ETH Zürich 8093, Zürich, Zürich, Switzerland
| | - Karla Vuina
- Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, UK
| | - Patricia Moyano Alvarez
- Institute for Biochemistry, Department of Biology, ETH Zürich 8093, Zürich, Zürich, Switzerland
| | - Robertus A M de Bruin
- Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, UK; UCL Cancer Institute, University College London, London WC1E 6BT, UK
| | - Gabriel E Neurohr
- Institute for Biochemistry, Department of Biology, ETH Zürich 8093, Zürich, Zürich, Switzerland.
| |
Collapse
|
49
|
Wang CC, Lu DD, Shen MH, Chen RL, Zhang ZH, Lv JH. Clinical value of Cyclin D1 and P21 in the differential diagnosis of papillary thyroid carcinoma. Diagn Pathol 2023; 18:123. [PMID: 37951919 PMCID: PMC10638720 DOI: 10.1186/s13000-023-01410-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 10/29/2023] [Indexed: 11/14/2023] Open
Abstract
BACKGROUND With the continuous discovery of new borderline thyroid lesions and benign and malignant "gray areas", coupled with the limitations of traditional immune indicators, the differential diagnosis of papillary thyroid carcinoma (PTC) has become more difficult. Cyclin D1 and P21 are cell cycle regulators involved in the occurrence and metastasis of multiple tumors, including PTC, but their specific functions are unclear. METHODS In our study, immunohistochemical staining was used to explore the expression of Cyclin D1 and P21 in PTC, paracancerous tissue, follicular adenoma (FA) and papillary thyroid hyperplasia. In addition, their relationship with the clinicopathological features of PTC and their differential diagnostic value in distinguishing between intralymph node PTC metastases and intralymph node ectopic thyroid tissue were studied. RESULTS Among 200 primary PTC lesions, Cyclin D1 and P21 were found to be expressed in 186 (93.00%) and 177 (88.50%), respectively, and their expression levels were significantly higher in PTC tissue than in adjacent tissue, FA tissue and papillary thyroid hyperplasia tissue (P < 0.05). The expression levels of Cyclin D1 and P21 were positively correlated with tumor size and lymph node metastasis (P < 0.05) but not with sex, age, number of tumor lesions, histological subtype, chronic lymphocytic thyroiditis or TNM stage (P < 0.05). The expression levels of Cyclin D1 and P21 were significantly correlated (P < 0.05). The positivity rates of Cyclin D1 and P21 in intralymph node PTC metastases were 97.96% (48/49) and 89.80% (44/49), respectively, which were significantly higher than those in intralymph node ectopic thyroid tissue (P < 0.05). The sensitivity (Se) and negative predictive value (NPV) of Cyclin D1 and P21 detection alone or in combination were higher than those of the combined detection of the classical antibody markers CK19, HBME-1 and Galectin-3. Besides, the Se, Sp, PPV and NPV of Cyclin D1 and P21 in differentiating intralymph node PTC metastases and intralymph node ectopic thyroid tissue were higher. CONCLUSIONS The results of our study show that Cyclin D1 and P21 are highly sensitive and specific markers for the diagnosis of PTC that are superior to traditional classical antibodies. And, these two markers are of great value in the differential diagnosis of intralymph node PTC metastases and intralymph node ectopic thyroid tissue.
Collapse
Affiliation(s)
- Chen-Chen Wang
- Department of Pathology, Gusu School, the Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Nanjing Medical University, Suzhou, China
| | - Dan-Dan Lu
- Department of Pathology, Gusu School, the Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Nanjing Medical University, Suzhou, China
| | - Ming-Hong Shen
- Department of Pathology, Gusu School, the Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Nanjing Medical University, Suzhou, China
| | - Ru-Lei Chen
- Department of Pathology, Gusu School, the Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Nanjing Medical University, Suzhou, China
| | - Zhi-Hong Zhang
- Department of Pathology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | - Jing-Huan Lv
- Department of Pathology, Gusu School, the Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Nanjing Medical University, Suzhou, China.
| |
Collapse
|
50
|
Dar AA, Kim DD, Gordon SM, Klinzing K, Rosen S, Guha I, Porter N, Ortega Y, Forsyth KS, Roof J, Fazelinia H, Spruce LA, Eisenlohr LC, Behrens EM, Oliver PM. c-Myc uses Cul4b to preserve genome integrity and promote antiviral CD8 + T cell immunity. Nat Commun 2023; 14:7098. [PMID: 37925424 PMCID: PMC10625626 DOI: 10.1038/s41467-023-42765-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 10/17/2023] [Indexed: 11/06/2023] Open
Abstract
During infection, virus-specific CD8+ T cells undergo rapid bursts of proliferation and differentiate into effector cells that kill virus-infected cells and reduce viral load. This rapid clonal expansion can put T cells at significant risk for replication-induced DNA damage. Here, we find that c-Myc links CD8+ T cell expansion to DNA damage response pathways though the E3 ubiquitin ligase, Cullin 4b (Cul4b). Following activation, c-Myc increases the levels of Cul4b and other members of the Cullin RING Ligase 4 (CRL4) complex. Despite expressing c-Myc at high levels, Cul4b-deficient CD8+ T cells do not expand and clear the Armstrong strain of lymphocytic choriomeningitis virus (LCMV) in vivo. Cul4b-deficient CD8+ T cells accrue DNA damage and succumb to proliferative catastrophe early after antigen encounter. Mechanistically, Cul4b knockout induces an accumulation of p21 and Cyclin E2, resulting in replication stress. Our data show that c-Myc supports cell proliferation by maintaining genome stability via Cul4b, thereby directly coupling these two interdependent pathways. These data clarify how CD8+ T cells use c-Myc and Cul4b to sustain their potential for extraordinary population expansion, longevity and antiviral responses.
Collapse
Affiliation(s)
- Asif A Dar
- Division of Protective Immunity, The Children's Hospital of Philadelphia, Philadelphia, PA, USA.
| | - Dale D Kim
- Division of Protective Immunity, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Scott M Gordon
- Division of Neonatology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Kathleen Klinzing
- Division of Protective Immunity, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Siera Rosen
- Division of Protective Immunity, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Ipsita Guha
- Division of Protective Immunity, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Nadia Porter
- Division of Protective Immunity, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Yohaniz Ortega
- Division of Protective Immunity, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Katherine S Forsyth
- Division of Protective Immunity, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Jennifer Roof
- Division of Cell Pathology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Hossein Fazelinia
- Division of Cell Pathology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Lynn A Spruce
- Division of Cell Pathology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Laurence C Eisenlohr
- Division of Protective Immunity, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pathology, University of Pennsylvania, Philadelphia, PA, USA
| | - Edward M Behrens
- Division of Rheumatology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Paula M Oliver
- Division of Protective Immunity, The Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Department of Pathology, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|