301
|
Svec J, Onhajzer J, Korinek V. Origin, development and therapy of colorectal cancer from the perspective of a biologist and an oncologist. Crit Rev Oncol Hematol 2024; 204:104544. [PMID: 39490796 DOI: 10.1016/j.critrevonc.2024.104544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 10/22/2024] [Accepted: 10/22/2024] [Indexed: 11/05/2024] Open
Abstract
The intestinal epithelium, a rapidly renewing tissue, is characterized by a continuous cell turnover that occurs through a well-coordinated process of cell proliferation and differentiation. This dynamic is crucial for the long-term function of the gastrointestinal tract. Disruption of this process can lead to colorectal carcinoma, a common malignancy worldwide. The first part of the review focuses on the cellular composition of the epithelium and the molecular mechanisms that control its functions, and describes the pathways that lead to epithelial transformation and tumor progression. This forms the basis for understanding the development and progression of advanced colorectal cancer. The second part deals with current therapeutic approaches and presents the latest treatment options, ongoing clinical trials and new drugs. In addition, the biological and medical perspectives of the adverse effects of therapies and models of regeneration of the intestinal epithelium are highlighted and, finally, future treatment options are discussed.
Collapse
Affiliation(s)
- Jiri Svec
- Laboratory of Cell and Developmental Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic; Department of Oncology, Third Faculty of Medicine, Charles University, University Hospital Kralovske Vinohrady, Prague, Czech Republic
| | - Jakub Onhajzer
- Laboratory of Cell and Developmental Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Vladimir Korinek
- Laboratory of Cell and Developmental Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic.
| |
Collapse
|
302
|
Chen Q, Zhang C, Meng T, Yang K, Hu Q, Tong Z, Wang X. Prediction of clinical prognosis and drug sensitivity in hepatocellular carcinoma through the combination of multiple cell death pathways. Cell Biol Int 2024; 48:1816-1835. [PMID: 39192561 DOI: 10.1002/cbin.12235] [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: 03/30/2024] [Revised: 07/29/2024] [Accepted: 08/10/2024] [Indexed: 08/29/2024]
Abstract
Hepatocellular carcinoma (HCC) is the sixth most common malignant tumor, highlighting a significant need for reliable predictive models to assess clinical prognosis, disease progression, and drug sensitivity. Recent studies have highlighted the critical role of various programmed cell death pathways, including apoptosis, necroptosis, pyroptosis, ferroptosis, cuproptosis, entotic cell death, NETotic cell death, parthanatos, lysosome-dependent cell death, autophagy-dependent cell death, alkaliptosis, oxeiptosis, and disulfidptosis, in tumor development. Therefore, by investigating these pathways, we aimed to develop a predictive model for HCC prognosis and drug sensitivity. We analyzed transcriptome, single-cell transcriptome, genomic, and clinical information using data from the TCGA-LIHC, GSE14520, GSE45436, and GSE166635 datasets. Machine learning algorithms were used to establish a cell death index (CDI) with seven gene signatures, which was validated across three independent datasets, showing that high CDI correlates with poorer prognosis. Unsupervised clustering revealed three molecular subtypes of HCC with distinct biological processes. Furthermore, a nomogram integrating CDI and clinical information demonstrated good predictive performance. CDI was associated with immune checkpoint genes and tumor microenvironment components using single-cell transcriptome analysis. Drug sensitivity analysis indicated that patients with high CDI may be resistant to oxaliplatin and cisplatin but sensitive to axitinib and sorafenib. In summary, our model offers a precise prediction of clinical outcomes and drug sensitivity for patients with HCC, providing valuable insights for personalized treatment strategies.
Collapse
Affiliation(s)
- QingKun Chen
- Department of Graduate School, Bengbu Medical University, Bengbu, China
- Department of Hepatobiliary Surgery, The First People's Hospital of Hefei, Hefei, China
| | - ChenGuang Zhang
- Department of Graduate School, Bengbu Medical University, Bengbu, China
- Department of Hepatobiliary Surgery, The First People's Hospital of Hefei, Hefei, China
| | - Tao Meng
- Department of Hepatobiliary Surgery, The First People's Hospital of Hefei, Hefei, China
| | - Ke Yang
- Department of Hepatobiliary Surgery, The First People's Hospital of Hefei, Hefei, China
| | - QiLi Hu
- Department of Hepatobiliary Surgery, The First People's Hospital of Hefei, Hefei, China
| | - Zhong Tong
- Department of Hepatobiliary Surgery, The First People's Hospital of Hefei, Hefei, China
| | - XiaoGang Wang
- Department of Hepatobiliary Surgery, The First People's Hospital of Hefei, Hefei, China
| |
Collapse
|
303
|
Molla MD, Symonds EL, Winter JM, Debie A, Wassie MM. Metabolic risk factors of colorectal cancer: Umbrella review. Crit Rev Oncol Hematol 2024; 204:104502. [PMID: 39245299 DOI: 10.1016/j.critrevonc.2024.104502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 08/28/2024] [Accepted: 08/29/2024] [Indexed: 09/10/2024] Open
Abstract
BACKGROUND AND AIM The association between metabolic factors and colorectal cancer (CRC) risk is inconclusive. This umbrella review aimed to summarise and describe the association using existing systematic reviews and/or meta-analyses. METHOD Four databases (Medline, Scopus, Web of Science, and Cochrane Library) were searched for systematic reviews and/or meta-analyses of observational studies. Two independent authors extracted data on the summary estimated effect and heterogeneity of studies using I2 from the individual reviews. The Assessing the Methodological Quality of Systematic Reviews (AMSTAR 2) tool was used to evaluate the methodological quality. RESULTS 49 articles were included in this review. Although most included studies were graded with critically low methodological quality (81.6 %), we found a significant positive association between obesity (summary relative risk (SRR) range 1.19-1.49), diabetes mellitus (SRR range 1.20-1.37), hypertension (SRR range 1.07-1.62), metabolic syndrome (SRR range 1.25-1.36), non-alcoholic fatty liver disease (pooled odds ratio (POR) range 1.13-1.56), and risk of CRC. Higher serum high-density lipoprotein cholesterol levels were associated with a lower risk of CRC in 3/6 reviews, while others did not find any association. There was no clear association between high triglyceride levels, total cholesterol levels, low-density lipoprotein cholesterol levels, and risk of CRC. CONCLUSION This umbrella review identified that most metabolic factors are significantly associated with increased risk of CRC. Thus, people affected by metabolic factors may be benefited from CRC screening and surveillance.
Collapse
Affiliation(s)
- Meseret Derbew Molla
- Flinders University, College of Medicine and PublicHealth, Flinders Health and Medical Research Institute, Adelaide, South Australia, Australia; Department of Biochemistry, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia.
| | - Erin L Symonds
- Flinders University, College of Medicine and PublicHealth, Flinders Health and Medical Research Institute, Adelaide, South Australia, Australia; Gastroenterology and Hepatology Department, Flinders Medical Centre, Southern Adelaide Local Health Network, Bedford Park, South Australia, Australia
| | - Jean M Winter
- Flinders University, College of Medicine and PublicHealth, Flinders Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Ayal Debie
- Flinders University, College of Medicine and PublicHealth, Flinders Health and Medical Research Institute, Adelaide, South Australia, Australia; Department of Health Systems and Policy, Institute of Public Health, University of Gondar, Gondar, Ethiopia
| | - Molla M Wassie
- Flinders University, College of Medicine and PublicHealth, Flinders Health and Medical Research Institute, Adelaide, South Australia, Australia
| |
Collapse
|
304
|
Saranya I, Preetha D, Nivruthi S, Selvamurugan N. A comprehensive bioinformatic analysis of the role of TGF-β1-stimulated activating transcription factor 3 by non-coding RNAs during breast cancer progression. Comput Biol Chem 2024; 113:108208. [PMID: 39276678 DOI: 10.1016/j.compbiolchem.2024.108208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/01/2024] [Accepted: 09/06/2024] [Indexed: 09/17/2024]
Abstract
A potent growth inhibitor for normal mammary epithelial cells is transforming growth factor beta 1 (TGF-β1). When breast tissues lose the anti-proliferative activity of this factor, invasion and bone metastases increase. Human breast cancer (hBC) cells express more activating transcription factor 3 (ATF3) when exposed to TGF-β1, and this transcription factor is essential for BC development and bone metastases. Non-coding RNAs (ncRNAs), including circular RNAs (circRNAs) and microRNAs (miRNAs), have emerged as key regulators controlling several cellular processes. In hBC cells, TGF-β1 stimulated the expression of hsa-miR-4653-5p that putatively targets ATF3. Bioinformatics analysis predicted that hsa-miR-4653-5p targets several key signaling components and transcription factors, including NFKB1, STAT1, STAT3, NOTCH1, JUN, TCF3, p300, NRF2, SUMO2, and NANOG, suggesting the diversified role of hsa-miR-4653-5p under physiological and pathological conditions. Despite the high abundance of hsa-miR-4653-5p in hBC cells, the ATF3 level remained elevated, indicating other ncRNAs could inhibit hsa-miR-4653-5p's activity. In silico analysis identified several circRNAs having the binding sites for hsa-miR-4653-5p, indicating the sponging activity of circRNAs towards hsa-miR-4653-5p. The study's findings suggest that TGF-β1 regulates circRNAs and hsa-miR-4653-5p, which in turn affects ATF3 expression, thus influencing BC progression and bone metastasis. Therefore, focusing on the TGF-β1/circRNAs/hsa-miR-4653-5p/ATF3 network could lead to new ways of diagnosing and treating BC.
Collapse
Affiliation(s)
- Iyyappan Saranya
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603 203, India
| | - Dilipkumar Preetha
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603 203, India
| | - Sasi Nivruthi
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603 203, India
| | - Nagarajan Selvamurugan
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603 203, India.
| |
Collapse
|
305
|
Ge Y, Janson V, Liu H. Comprehensive review on leucine-rich pentatricopeptide repeat-containing protein (LRPPRC, PPR protein): A burgeoning target for cancer therapy. Int J Biol Macromol 2024; 282:136820. [PMID: 39476900 DOI: 10.1016/j.ijbiomac.2024.136820] [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: 09/03/2024] [Revised: 10/12/2024] [Accepted: 10/21/2024] [Indexed: 11/04/2024]
Abstract
Leucine-rich pentatricopeptide repeat-containing (LRPPRC), known as the gene mutations that cause Leigh Syndrome French Canadian, encodes a high molecular weight PPR protein (157,905 Da), LRPPRC. LRPPRC binds to DNA, RNA, and proteins to regulate transcription and translation, leading to changes in cell fate. Increasing evidence indicates that LRPPRC plays a pivotal role in various human diseases, particularly cancer in recent years. Here, we review the structure, function, molecular mechanism, as well as inhibitors of LRPPRC. LRPPRC expression elevates in most cancer types and high expression of LRPPRC predicts the poor prognosis of cancer patients. Targeting LRPPRC suppresses tumor progression by affecting several cancer hallmarks, including signal transduction, cancer metabolism, and immune regulation. LRPPRC is a promising target in cancer research, serving as both a biomarker and therapeutic target. Further studies are required to extend the understanding of LRPPRC function and molecular mechanism, as well as to refine novel therapeutic strategies targeting LRPPRC in cancer therapy.
Collapse
Affiliation(s)
- Yunxiao Ge
- Department of Pathophysiology, School of Basic Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, Henan 450001, China; China-US (Henan) Hormel Cancer Institute, No.127, Dongming Road, Jinshui District, Zhengzhou, Henan 450008, China
| | - Victor Janson
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Hui Liu
- Department of Pathophysiology, School of Basic Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, Henan 450001, China; Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden; China-US (Henan) Hormel Cancer Institute, No.127, Dongming Road, Jinshui District, Zhengzhou, Henan 450008, China.
| |
Collapse
|
306
|
Hao J, Lu M, Zhao X, Li C, Ge C, Zhang J, Tu L, Zhang Q. Exploring the underlying mechanism by transcriptome sequencing in rats with high-voltage electrical burns and the role of iron metabolism. Burns 2024; 50:107226. [PMID: 39368958 DOI: 10.1016/j.burns.2024.07.030] [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: 03/20/2024] [Revised: 05/24/2024] [Accepted: 07/28/2024] [Indexed: 10/07/2024]
Abstract
BACKGROUND Clinically, the condition of skeletal muscle injury is the key to the process of high voltage electrical burn (HVEB) wound repair. The aim of this study was to identify the potential mechanisms and intervention targets of skeletal muscle injury after HVEB. METHODS A skeletal muscle injury model in SD rats with HVEB was made. Pathological examination and transcriptome sequencing of injured skeletal muscles were performed, and the expression levels of key proteins and genes in related signaling pathways were verified. RESULTS Skeletal muscle injury was progressively aggravated within 48 h, then the injury was gradually repaired with scar formation occurring within 1 week. The mechanism of skeletal muscle injury is complex and varied, and ferroptosis is one of the mechanisms. The ferrous iron content in the injured skeletal muscle tissue of model rats increased significantly at 24 h after injury. After 24 h, damage to injured skeletal muscle tissue could be alleviated by increasing iron storage and blocking lysosomal phagocytosis of autophagy. CONCLUSIONS Skeletal muscle injury caused by HVEB is characterized by adjacent endangered tissue progression after injury. Ferroptosis is involved in the mechanism of HVEB, and iron metabolism-related proteins may be potential targets for preventing progressive skeletal muscle injury.
Collapse
Affiliation(s)
- Jiawen Hao
- Burn and Wound Repair Center, Hebei Medical University Third Hospital, Shijiazhuang, Hebei Province 050035, China
| | - Mengyuan Lu
- Burn and Wound Repair Center, Hebei Medical University Third Hospital, Shijiazhuang, Hebei Province 050035, China
| | - Xuegang Zhao
- Burn and Wound Repair Center, Hebei Medical University Third Hospital, Shijiazhuang, Hebei Province 050035, China
| | - Congying Li
- Burn and Wound Repair Center, Hebei Medical University Third Hospital, Shijiazhuang, Hebei Province 050035, China
| | - Chenyang Ge
- Burn and Wound Repair Center, Hebei Medical University Third Hospital, Shijiazhuang, Hebei Province 050035, China
| | - Jing Zhang
- Burn and Wound Repair Center, Hebei Medical University Third Hospital, Shijiazhuang, Hebei Province 050035, China
| | - Lihong Tu
- Burn and Wound Repair Center, Hebei Medical University Third Hospital, Shijiazhuang, Hebei Province 050035, China
| | - Qingfu Zhang
- Burn and Wound Repair Center, Hebei Medical University Third Hospital, Shijiazhuang, Hebei Province 050035, China.
| |
Collapse
|
307
|
Lou S, Yang W, Zhao Q, Ouyang Y, Cao L, Lin C. Identification of circRNA-mediated competing endogenous RNA network involved in the development of cervical cancer. Mol Cell Probes 2024; 78:101984. [PMID: 39307294 DOI: 10.1016/j.mcp.2024.101984] [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: 08/20/2024] [Revised: 09/20/2024] [Accepted: 09/20/2024] [Indexed: 10/13/2024]
Abstract
BACKGROUND The abnormal expression of circRNA may contribute to the progression of cervical cancer by influencing the biological processes. AIM This study aimed to identify the differentially expressed circRNAs in cervical cancer and validate the circ_0008193 ceRNA network in cervical cancer cells. METHODS Using the absolute log2 value of fold change >1 and p-value of <0.05, the differentially expressed circRNAs were obtained from GSE102686 and GSE113696 from cervical cancer tissues and cervical cancer cells with the help of the GEO2R tool. Downstream miRNAs and mRNAs were predicted using relevant informatics databases. The circRNA-miRNA-mRNA interaction network was conducted with the assistance of Cytoscape. Circ_0008193-miR-182-5p-PTEN axis was validated with expression level and cell function using RT-qPCR, a dual-luciferase reporter assay, and cellular experiments. RESULTS GSE102686 and GSE113696 databases overlapped 7 differentially expressed circRNAs and five circRNAs have the same expression pattern. Based on the literature and expression pattern, a circRNA-miRNA-mRNA network was conducted. The circ_0008193, miR-182-5p, and PTEN expression patterns were downregulation, upregulation, and downregulation, respectively. Overexpressed circ_0008193 suppressed proliferation, migration, and invasion of cervical cancer cells. MiR-182-5p diminished the inhibitory influence of circ_0008193 on cellular behaviors, while PTEN counteracted the effect of miR-182-5p. CONCLUSION This investigation revealed the existence of a circRNA-miRNA-mRNA network in cervical cancer, and preliminary verified the function of circ_0008193-miR-182-5p-PTEN axis in cervical cancer cells, which offers additional guidance on investigating the molecular mechanisms of cervical cancer.
Collapse
Affiliation(s)
- Shaosheng Lou
- Department of Pathology, School of Basic Medical Sciences, Xinjiang Medical University/Key Laboratory of Molecular Biology of Endemic Diseases, Xinjiang, 830000, China.
| | - Wang Yang
- Department of Pathology, School of Basic Medical Sciences, Xinjiang Medical University/Key Laboratory of Molecular Biology of Endemic Diseases, Xinjiang, 830000, China
| | - Qian Zhao
- Department of Pathology, School of Basic Medical Sciences, Xinjiang Medical University/Key Laboratory of Molecular Biology of Endemic Diseases, Xinjiang, 830000, China
| | - Yunshan Ouyang
- Department of Pathology, School of Basic Medical Sciences, Xinjiang Medical University/Key Laboratory of Molecular Biology of Endemic Diseases, Xinjiang, 830000, China
| | - Lingling Cao
- Department of Pathology, School of Basic Medical Sciences, Xinjiang Medical University/Key Laboratory of Molecular Biology of Endemic Diseases, Xinjiang, 830000, China
| | - Chen Lin
- Graduate School of Xinjiang Medical University, Xinjiang, 830000, China.
| |
Collapse
|
308
|
Zheng Y, Xiang G, Zeng L, Yang C, Ke J, Yu H, Zhang J. MiR-24-3p modulates cardiac function in doxorubicin -induced heart failure via the Sp1/PI3K signaling pathway. Cell Signal 2024; 124:111407. [PMID: 39278455 DOI: 10.1016/j.cellsig.2024.111407] [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: 07/02/2024] [Revised: 09/01/2024] [Accepted: 09/09/2024] [Indexed: 09/18/2024]
Abstract
PURPOSE The goal of this research was to explore the role of miR-24-3p in heart failure (HF), with a focus on its impact on the specificity protein 1 (Sp1)/phosphoinositide 3-kinase (PI3K) pathway. METHODS HF rat and HF cell models were established using doxorubicin(Dox). Cardiac function was assessed through echocardiography, while histological changes were observed via hematoxylin-eosin (HE) staining. To further investigate the underlying mechanisms, HF cell models were treated with either an Sp1 inhibitor or a PI3K inhibitor. Additionally, models with miR-24-3p overexpression or silencing were constructed. N-terminal pro-brain natriuretic peptide (NT-proBNP) levels were determined by ELISA. Cell apoptosis was evaluated using TUNEL staining, and lactate dehydrogenase (LDH) levels were measured by colorimetry. Reactive oxygen species (ROS) production was analyzed using flow cytometry. Related gene and protein expressions were assessed via qRT-PCR and Western blotting. Finally, the relationship between miR-24-3p and Sp1 was confirmed through dual-luciferase assays. RESULTS Dox treatment increased the left ventricular internal diameter (LVIDd) while decreasing ejection fraction (EF) and fractional shortening (FS), leading to disorganized cardiomyocyte arrangement, cellular edema, and necrosis in rats. In HF rats, NT-proBNP, Caspase-3, and miR-24-3p expression levels were elevated, whereas Sp1 and PI3K mRNA and protein expression levels were decreased. Similarly, Dox-induced damage in H9c2 cardiomyocytes resulted in increased NT-proBNP, apoptosis, Caspase-3, LDH, ROS, and miR-24-3p expression, along with decreased Sp1 and PI3K expression. Treatment with either Sp1 or PI3K inhibitors exacerbated the Dox-induced cardiomyocyte damage, further elevating NT-proBNP, apoptosis, Caspase-3, LDH, ROS, and miR-24-3p expression levels. Notably, Sp1 inhibition reduced PI3K expression, and PI3K inhibition, in turn, suppressed Sp1 expression. Overexpression of miR-24-3p worsened Dox-induced cardiomyocyte damage, characterized by increased NT-proBNP, apoptosis, Caspase-3, LDH, and ROS expression, alongside reduced Sp1 and PI3K expression. In contrast, silencing miR-24-3p mitigated these detrimental effects and increased Sp1 and PI3K expression. Dual-luciferase assays confirmed that miR-24-3p directly targets Sp1. CONCLUSION Dox induces cardiomyocyte damage, impairs cardiac function, and promotes cardiomyocyte apoptosis and oxidative stress. Silencing miR-24-3p offers a protective effect by activating the Sp1/PI3K signaling pathway in heart failure.
Collapse
Affiliation(s)
- Yonghong Zheng
- Provincial Clinical Medical College of Fujian Medical University, Fuzhou 350001, Fujian, China; Fuzhou University Affiliated Provincial Hospital, Fuzhou 350001, Fujian, China; Department of Cardiology, Fujian Provincial Hospital, Fuzhou 350001, Fujian, China
| | - Guojian Xiang
- Provincial Clinical Medical College of Fujian Medical University, Fuzhou 350001, Fujian, China; Fuzhou University Affiliated Provincial Hospital, Fuzhou 350001, Fujian, China; Department of Cardiology, Fujian Provincial Hospital, Fuzhou 350001, Fujian, China
| | - Linwen Zeng
- Provincial Clinical Medical College of Fujian Medical University, Fuzhou 350001, Fujian, China
| | - Chao Yang
- Provincial Clinical Medical College of Fujian Medical University, Fuzhou 350001, Fujian, China; Fuzhou University Affiliated Provincial Hospital, Fuzhou 350001, Fujian, China; Department of Intensive Care Medicine, Fujian Provincial Hospital, Fuzhou 350001, Fujian, China
| | - Jun Ke
- Provincial Clinical Medical College of Fujian Medical University, Fuzhou 350001, Fujian, China; Fuzhou University Affiliated Provincial Hospital, Fuzhou 350001, Fujian, China
| | - Huizhen Yu
- Provincial Clinical Medical College of Fujian Medical University, Fuzhou 350001, Fujian, China; Fuzhou University Affiliated Provincial Hospital, Fuzhou 350001, Fujian, China; Department of Cardiology in South Branch, Fujian Provincial Hospital, Fuzhou 350001, Fujian, China.
| | - Jiancheng Zhang
- Provincial Clinical Medical College of Fujian Medical University, Fuzhou 350001, Fujian, China; Fuzhou University Affiliated Provincial Hospital, Fuzhou 350001, Fujian, China; Department of Cardiology, Fujian Provincial Hospital, Fuzhou 350001, Fujian, China.
| |
Collapse
|
309
|
Zhang J, Xiang H, Jiang L, Wang M, Yang G. Construction of a novel platelet‑related gene risk model to predict the prognosis and drug response in virus‑related hepatocellular carcinoma. Oncol Lett 2024; 28:592. [PMID: 39417040 PMCID: PMC11481168 DOI: 10.3892/ol.2024.14725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 09/05/2024] [Indexed: 10/19/2024] Open
Abstract
Platelet activity in the tumor microenvironment (TME) is crucial for the development of tumors. However, the roles and clinical potential of platelet activity in the TME for virus-related hepatocellular carcinoma (HCC) remain unclear. The present study aimed to identify a novel signature based on platelet activity for prognostic prediction and treatment decisions in virus-related HCC. First, a novel platelet signature score (PSS) for each patient with virus-related HCC from The Cancer Genome Atlas was calculated using gene set variation analysis, and the patients were divided into two subgroups (high and low PSS). It was demonstrated that the patients with a high PSS had a worse prognosis, higher platelet activity, stronger inflammation and immunosuppression in TME than patients with a low PSS. Furthermore, 137 differentially expressed genes (DEGs; fold change >2; P<0.05) were identified using 'DESeq2' and 'edgeR' software. Subsequently, 3 genes (cyclin-J-Like protein, nuclear receptor subfamily 0 group B member 1 and tripartite motif containing 54) were identified from DEGs using univariate Cox and least absolute shrinkage and selection operator (LASSO) analyses. Risk score (RS) was calculated based on gene expression and coefficients from LASSO. Patients were divided into high and low RS groups according to the median value, and the 3-gene model was used to predict prognoses and drug responses. Notably, it was demonstrated that patients with a low RS may be better candidates for immune therapy due to lower levels of tumor immune dysfunction and exclusion scores. Moreover, patients with a high RS may be better candidates for nonimmune therapy due to lower half-maximal inhibitory concentration values of drugs (such as AKT inhibitors and gemcitabine). Finally, it was demonstrated that patients with a high PSS and RS had a higher platelet activity, inflammation status, tumor hallmarks and the worst prognosis than patients with a low PSS and RS. This helped to better find patients with these characteristics and suitable treatments using this method. Collectively, the findings of the present study indicate that PSS combined with RS has great potential to evaluate the prognosis of patients with virus -related HCC and assist in deciding treatment strategies.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Gastroenterology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Honglin Xiang
- Department of Orthopaedics, Laboratory of Biological Tissue Engineering and Digital Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Ling Jiang
- Department of Gastroenterology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Mei Wang
- Department of Gastroenterology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Guodong Yang
- Department of Gastroenterology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| |
Collapse
|
310
|
Zhou X, Liu H, Hou F, Zheng ZQ, Cao X, Wang Q, Jiang W. REMR: Identification of RNA Editing-mediated MiRNA Regulation in Cancers. Comput Struct Biotechnol J 2024; 23:3418-3429. [PMID: 39386942 PMCID: PMC11462282 DOI: 10.1016/j.csbj.2024.09.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 09/17/2024] [Accepted: 09/17/2024] [Indexed: 10/12/2024] Open
Abstract
Dysregulation of adenosine-to-inosine (A-to-I) RNA editing has been implicated in cancer progression. However, a comprehensive understanding of how A-to-I RNA editing is incorporated into miRNA regulation to modulate gene expression in cancer remains unclear, given the lack of effective identification methods. To this end, we introduced an information theory-based algorithm named REMR to systematically identify 12,006 A-to-I RNA editing-mediated miRNA regulatory triplets (RNA editing sites, miRNAs, and genes) across ten major cancer types based on multi-omics profiling data from The Cancer Genome Atlas (TCGA). Through analyses of functional enrichment, transcriptional regulatory networks, and protein-protein interaction (PPI) networks, we showed that RNA editing-mediated miRNA regulation potentially affects critical cancer-related functions, such as apoptosis, cell cycle, drug resistance, and immunity. Furthermore, triplets can serve as biomarkers for classifying cancer subtypes with distinct prognoses or drug responses, highlighting the clinical relevance of such regulation. In addition, an online resource (http://www.jianglab.cn/REMR/) was constructed to support the convenient retrieval of our findings. In summary, our study systematically dissected the RNA editing-mediated miRNA regulations, thereby providing a valuable resource for understanding the mechanism of RNA editing as an epitranscriptomic regulator in cancer.
Collapse
Affiliation(s)
- Xu Zhou
- Department of Biomedical Engineering, College of Automation Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing 211106, China
| | - Haizhou Liu
- Fujian Provincial Key Laboratory of Precision Medicine for Cancer, The First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
| | - Fei Hou
- Department of Biomedical Engineering, College of Automation Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing 211106, China
| | - Zong-Qing Zheng
- Fujian Provincial Key Laboratory of Precision Medicine for Cancer, The First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
- Department of Neurosurgery, Neurosurgery Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
- Department of Neurosurgery, Binhai Branch of National Regional Medical Center, The First Affiliated Hospital, Fujian Medical University, Fuzhou 350209, China
| | - Xinyu Cao
- Department of Biomedical Engineering, College of Automation Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing 211106, China
| | - Quan Wang
- Department of Biomedical Engineering, College of Automation Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing 211106, China
| | - Wei Jiang
- Department of Biomedical Engineering, College of Automation Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing 211106, China
- Fujian Provincial Key Laboratory of Precision Medicine for Cancer, The First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
| |
Collapse
|
311
|
Xu C, Wu M, Yu W, Xie D, Wang Q, Chen B, Xi Y, Yu L, Yan Y, Yamamoto T, Koyama H, Zhao H, Cheng J. High Uric Acid Orchestrates Ferroptosis to Promote Cardiomyopathy Via ROS-GPX4 Signaling. Antioxid Redox Signal 2024; 41:1134-1149. [PMID: 39113539 DOI: 10.1089/ars.2023.0473] [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] [Indexed: 08/29/2024]
Abstract
Aims: High uric acid (HUA), as a pro-oxidant, plays a significant role in the pathophysiology of cardiovascular disease. Studies have indicated that elevated uric acid levels can adversely affect cardiovascular health. Nevertheless, the impact of hyperuricemia on cardiomyopathy remains uncertain. Further research is needed to elucidate the relationship between HUA and cardiomyopathy, shedding light on its potential implications for heart health. Results: We demonstrated that uricase knockout (Uox-KO) mice accelerated the development of cardiomyopathy, causing significantly impaired cardiac function and myocardial fibrosis. Meanwhile, the mitochondrial morphology was destroyed, the lipid peroxidation products increased in number and the antioxidant function was weakened. In addition, we evaluated the effects of ferrostatin-1 (Fer-1), the ferroptosis inhibitor. Myocardial damage can be reversed by the Fer-1 treatment caused by HUA combined with doxorubicin (DOX) treatment. Benzbromarone, a uric acid-lowering drug, decreases myocardial fibrosis, and ferroptosis by alleviating hyperuricemia in Uox-KO mice by DOX administration. In vitro, we observed that the activity of cardiomyocytes treated with HUA combined with DOX decreased significantly, and lipid reactive oxygen species (ROS) increased significantly. Afterward, we demonstrated that HUA can promote oxidative stress in DOX, characterized by increased mitochondrial ROS, and downregulate protein levels of glutathione peroxidase 4 (GPX4). N-acetyl-l-cysteine, an antioxidant, inhibits the process by which HUA promotes DOX-induced ferroptosis by increasing the GPX4 expression. Innovation: We verified that HUA can exacerbate myocardial damage. This has clinical implications for the treatment of cardiac damage in patients with hyperuricemia. Conclusions: Our data suggested that HUA promotes the cardiomyopathy. HUA promotes DOX-induced ferroptosis by increasing oxidative stress and downregulating GPX4. Antioxid. Redox Signal. 41, 1134-1149.
Collapse
Affiliation(s)
- Chenxi Xu
- Department of Endocrinology, Xiang'an Hospital of Xiamen University, Xiamen, China
- Institute of Metabolism and Cell Death, Helmholtz Munich, 85764 Neuherberg, Bavaria, Germany
| | - Mengni Wu
- Department of Endocrinology, Xiang'an Hospital of Xiamen University, Xiamen, China
| | - Wei Yu
- Department of Endocrinology, Xiang'an Hospital of Xiamen University, Xiamen, China
| | - De Xie
- Department of Endocrinology, Xiang'an Hospital of Xiamen University, Xiamen, China
| | - Qiang Wang
- Department of Endocrinology, Xiang'an Hospital of Xiamen University, Xiamen, China
| | - Binyang Chen
- Department of Endocrinology, Xiang'an Hospital of Xiamen University, Xiamen, China
| | - Yuemei Xi
- Department of Endocrinology, Xiang'an Hospital of Xiamen University, Xiamen, China
| | - Linqian Yu
- Department of Endocrinology, Xiang'an Hospital of Xiamen University, Xiamen, China
| | - Yunbo Yan
- Department of Endocrinology, Xiang'an Hospital of Xiamen University, Xiamen, China
| | - Tetsuya Yamamoto
- Department of Diabetes, Endocrinology, and Clinical Immunology, Hyogo College of Medicine, Nishinomiya, Japan
| | - Hidenori Koyama
- Department of Diabetes, Endocrinology, and Clinical Immunology, Hyogo College of Medicine, Nishinomiya, Japan
| | - Hong Zhao
- Department of Endocrinology, Xiang'an Hospital of Xiamen University, Xiamen, China
- Xiamen Key Laboratory of Translational Medicine for Nucleic Acid Metabolism and Regulation, Xiamen, China
| | - Jidong Cheng
- Department of Endocrinology, Xiang'an Hospital of Xiamen University, Xiamen, China
- Department of Diabetes, Endocrinology, and Clinical Immunology, Hyogo College of Medicine, Nishinomiya, Japan
- Xiamen Key Laboratory of Translational Medicine for Nucleic Acid Metabolism and Regulation, Xiamen, China
| |
Collapse
|
312
|
Shi Y, Cao Z, Ge L, Lei L, Tao D, Zhong J, Xu D, Geng T, Li X, Li Z, Xing S, Wu X, Wang Z, Li L. Rotenone adaptation promotes migration and invasion of p53-wild-type colon cancer through lipid metabolism. Clin Transl Oncol 2024:10.1007/s12094-024-03785-x. [PMID: 39612123 DOI: 10.1007/s12094-024-03785-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 10/10/2024] [Indexed: 11/30/2024]
Abstract
BACKGROUND The association between mitochondrial dysfunction and multiple metabolic adaptations is increasingly being proven. We previously elucidated that mitochondrial complex I deficiency can promote glycolysis in mut-p53 SW480 cells. However, studies have revealed a phenotype with attenuated glycolysis but enhanced fatty acid oxidation (FAO) in invasive tumors. The interplay between complex I and FAO in carcinogenesis remains obscure. METHODS The p53 wild-type RKO cells were exposed to rotenone over at least 2 months to acquire rotenone adaptation cells. Then the transwell invasion assays and expression of metabolic enzymes were first detected in rotenone adaptation cells to illustrate whether rotenone adaptation is correlated with the invasion and FAO. The levels of epithelial-to-mesenchymal transition (EMT)-related proteins and acetyl-CoA in rotenone adaptation cells treated with etomoxir (ETO) and acetate were evaluated to verify the role of CPT1A in regulating invasion. Finally, the levels of reactive oxygen species (ROS) were detected. Meanwhile, the invasiveness and histone acetylation levels of rotenone adaptation cells were observed after adding an ROS inhibitor (N-acetyl-L-cysteine NAC) to demonstrate the molecular connection between FAO and invasion during rotenone adaptation. RESULTS We found long-term exposure to rotenone (a mitochondrial complex I inhibitor) led to EMT and high CPT1A expression in wt-p53 colon cancer. The inhibition of CPT1A suppressed the invasion and reduced histone acetylation, which was rescued by supplementing with acetate. Mechanistically, ROS is crucial for lipid metabolism remodeling. CONCLUSION Our study provides a novel understanding of the role of complex I in lipid reprogramming facilitating colon cancer invasion and metastasis.
Collapse
Affiliation(s)
- Yingying Shi
- Department of Oncology, Maternal and Child Health Hospital of Hubei Province, Wuhan, 430070, China
- Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, 430071, China
| | - Zhen Cao
- Department of Oncology, Maternal and Child Health Hospital of Hubei Province, Wuhan, 430070, China
| | - Ling Ge
- Department of Oncology, Maternal and Child Health Hospital of Hubei Province, Wuhan, 430070, China
| | - Lin Lei
- Department of Oncology, Maternal and Child Health Hospital of Hubei Province, Wuhan, 430070, China
| | - Dan Tao
- Department of Oncology, Maternal and Child Health Hospital of Hubei Province, Wuhan, 430070, China
| | - Juan Zhong
- Department of Oncology, Maternal and Child Health Hospital of Hubei Province, Wuhan, 430070, China
| | - Dan Xu
- Department of Oncology, Maternal and Child Health Hospital of Hubei Province, Wuhan, 430070, China
| | - Tao Geng
- Department of Oncology, Maternal and Child Health Hospital of Hubei Province, Wuhan, 430070, China
| | - Xuetao Li
- Department of Oncology, Maternal and Child Health Hospital of Hubei Province, Wuhan, 430070, China
| | - Ziwei Li
- Department of Oncology, Maternal and Child Health Hospital of Hubei Province, Wuhan, 430070, China
| | - Shuaishuai Xing
- Department of Oncology, Maternal and Child Health Hospital of Hubei Province, Wuhan, 430070, China
| | - Xinyu Wu
- Department of Oncology, Maternal and Child Health Hospital of Hubei Province, Wuhan, 430070, China
| | - Zhongxu Wang
- Department of Oncology, Maternal and Child Health Hospital of Hubei Province, Wuhan, 430070, China
| | - Linjun Li
- Department of Oncology, Maternal and Child Health Hospital of Hubei Province, Wuhan, 430070, China.
| |
Collapse
|
313
|
Ge A, Xiang W, Li Y, Zhao D, Chen J, Daga P, Dai CC, Yang K, Yan Y, Hao M, Zhang B, Xiao W. Broadening horizons: the multifaceted role of ferroptosis in breast cancer. Front Immunol 2024; 15:1455741. [PMID: 39664391 PMCID: PMC11631881 DOI: 10.3389/fimmu.2024.1455741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 10/21/2024] [Indexed: 12/13/2024] Open
Abstract
Breast cancer poses a serious threat to women's health globally. Current radiotherapy and chemotherapy regimens can induce drug-resistance effects in cancer tissues, such as anti-apoptosis, anti-pyroptosis, and anti-necroptosis, leading to poor clinical outcomes in the treatment of breast cancer. Ferroptosis is a novel programmed cell death modality characterized by iron overload, excessive generation of reactive oxygen species, and membrane lipid peroxidation. The occurrence of ferroptosis results from the imbalance between intracellular peroxidation mechanisms (executive system) and antioxidant mechanisms (defensive system), specifically involving iron metabolism pathways, amino acid metabolism pathways, and lipid metabolism pathways. In recent years, it has been found that ferroptosis is associated with the progression of various diseases, including tumors, hypertension, diabetes, and Alzheimer's disease. Studies have confirmed that triggering ferroptosis in breast cancer cells can significantly inhibit cancer cell proliferation and invasion, and improve cancer cell sensitivity to radiotherapy and chemotherapy, making induction of ferroptosis a potential strategy for the treatment of breast cancer. This paper reviews the development of the concept of ferroptosis, the mechanisms of ferroptosis (including signaling pathways such as GSH-GPX4, FSP1-CoQ1, DHODH-CoQ10, and GCH1-BH4) in breast cancer disease, the latest research progress, and summarizes the research on ferroptosis in breast cancer disease within the framework of metabolism, reactive oxygen biology, and iron biology. The key regulatory factors and mechanisms of ferroptosis in breast cancer disease, as well as important concepts and significant open questions in the field of ferroptosis and related natural compounds, are introduced. It is hoped that future research will make further breakthroughs in the regulatory mechanisms of ferroptosis and the use of ferroptosis in treating breast cancer cells. Meanwhile, natural compounds may also become a new direction for potential drug development targeting ferroptosis in breast cancer treatment. This provides a theoretical basis and opens up a new pathway for research and the development of drugs for the prevention and treatment of breast cancer.
Collapse
Affiliation(s)
- Anqi Ge
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Wang Xiang
- Department of Rheumatology, The First People’s Hospital Changde City, Changde, Hunan, China
| | - Yan Li
- People's Hospital of Ningxiang City, Ningxiang, China
| | - Da Zhao
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Junpeng Chen
- Psychosomatic Laboratory, Department of Psychiatry, Daqing Hospital of Traditional Chinese Medicine, Daqing, China
- Department of Physiology, School of Medicine, University of Louisville, Louisville, KY, United States
- Tong Jiecheng Studio, Hunan University of Science and Technology, Xiangtan, China
| | - Pawan Daga
- Department of Internal Medicine, University of Louisville, Louisville, KY, United States
| | - Charles C. Dai
- Department of Oral and Maxillofacial Surgery, University of Maryland School of Dentistry, Baltimore, MD, United States
- Fischell Department of Bioengineering, A. James Clark School of Engineering, University of Maryland, College Park, MD, United States
| | - Kailin Yang
- Psychosomatic Laboratory, Department of Psychiatry, Daqing Hospital of Traditional Chinese Medicine, Daqing, China
- Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Yexing Yan
- Psychosomatic Laboratory, Department of Psychiatry, Daqing Hospital of Traditional Chinese Medicine, Daqing, China
| | - Moujia Hao
- Psychosomatic Laboratory, Department of Psychiatry, Daqing Hospital of Traditional Chinese Medicine, Daqing, China
| | | | - Wei Xiao
- Department of Rheumatology, The First People’s Hospital Changde City, Changde, Hunan, China
| |
Collapse
|
314
|
Rowland EC, D’Antuono M, Jermakowicz A, Ayad NG. MAT2a and AHCY inhibition disrupts antioxidant metabolism and reduces glioblastoma cell survival. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.23.624981. [PMID: 39605416 PMCID: PMC11601785 DOI: 10.1101/2024.11.23.624981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Glioblastoma (GBM) is a highly aggressive primary malignant adult brain tumor that inevitably recurs with a fatal prognosis. This is due in part to metabolic reprogramming that allows tumors to evade treatment. We therefore must uncover the pathways mediating these adaptations to develop novel and effective treatments. We searched for genes that are essential in GBM cells as measured by a whole-genome pan-cancer CRISPR screen available from DepMap and identified the methionine metabolism genes MAT2A and AHCY. We conducted genetic knockdown, evaluated mitochondrial respiration, and performed targeted metabolomics to study the function of these genes in GBM. We demonstrate that MAT2A or AHCY knockdown induces oxidative stress, hinders cellular respiration, and reduces the survival of GBM cells. Furthermore, selective MAT2a or AHCY inhibition reduces GBM cell viability, impairs oxidative metabolism, and changes the metabolic profile of these cells towards oxidative stress and cell death. Mechanistically, MAT2a or AHCY regulates spare respiratory capacity, the redox buffer cystathionine, lipid and amino acid metabolism, and prevents DNA damage in GBM cells. Our results point to the methionine metabolic pathway as a novel vulnerability point in GBM.
Collapse
Affiliation(s)
- Emma C. Rowland
- Georgetown University, Lombardi Comprehensive Cancer Center, 3970 Reservoir Rd NW Washington D.C. 20007, United States of America
| | - Matthew D’Antuono
- Georgetown University, Lombardi Comprehensive Cancer Center, 3970 Reservoir Rd NW Washington D.C. 20007, United States of America
| | - Anna Jermakowicz
- Georgetown University, Lombardi Comprehensive Cancer Center, 3970 Reservoir Rd NW Washington D.C. 20007, United States of America
| | - Nagi G. Ayad
- Georgetown University, Lombardi Comprehensive Cancer Center, 3970 Reservoir Rd NW Washington D.C. 20007, United States of America
| |
Collapse
|
315
|
Péterffy B, Nádasi TJ, Krizsán S, Horváth A, Márk Á, Barna G, Timár B, Almási L, Müller J, Csanádi K, Rakonczai A, Nagy Z, Kállay K, Kertész G, Kriván G, Csóka M, Sebestyén A, Semsei ÁF, Kovács GT, Erdélyi DJ, Bödör C, Egyed B, Alpár D. Digital PCR-based quantification of miR-181a in the cerebrospinal fluid aids patient stratification in pediatric acute lymphoblastic leukemia. Sci Rep 2024; 14:28556. [PMID: 39558071 PMCID: PMC11574027 DOI: 10.1038/s41598-024-79733-0] [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: 08/29/2024] [Accepted: 11/12/2024] [Indexed: 11/20/2024] Open
Abstract
Despite remarkable improvements in the survival of pediatric acute lymphoblastic leukemia (ALL), sensitive detection and clinical management of central nervous system leukemia (CNSL) are still immensely challenging. Blast cells residing in the CNS but not circulating in the cerebrospinal fluid (CSF) remain undetected by current diagnostic methods, preventing a truly risk-adapted anti-leukemic treatment in this compartment. We examined the clinical applicability of the molecular marker microRNA (miR)-181a quantified in the cell-free CSF to evaluate the level of CNS involvement and to optimize patient stratification based on CNS status. Normalized copy number of miR-181a was longitudinally profiled using droplet digital PCR, and the results were compared with the degree of leukemic involvement of the CNS. After combining cytospin- and flow cytometry (FCM) data with miR-181a expression, we could stratify previously ambiguous cases and reclassify patients into a CNS-positive/miR-significant group (mean ± SE for miR-181a copies: 3300.70 ± 809.69) bearing remarkable infiltration as well as into CNS-minimal/miR-significant and CNS-minimal/miR-minimal groups differentiating putative, clinically significant occult CNSL cases (2503.50 ± 275.89 and 744.02 ± 86.81 copies, respectively, p = 1.13 × 10-6). In summary, miR-181a expression is a promising biomarker for CNSL detection, facilitating the robust identification of patients who could benefit from intensified CNS-directed therapy.
Collapse
Grants
- PD145889, FK134253, K137948, K139139 Hungarian National Research, Development and Innovation Office
- PD145889, FK134253, K137948, K139139 Hungarian National Research, Development and Innovation Office
- PD145889, FK134253, K137948, K139139 Hungarian National Research, Development and Innovation Office
- PD145889, FK134253, K137948, K139139 Hungarian National Research, Development and Innovation Office
- PD145889, FK134253, K137948, K139139 Hungarian National Research, Development and Innovation Office
- PD145889, FK134253, K137948, K139139 Hungarian National Research, Development and Innovation Office
- PD145889, FK134253, K137948, K139139 Hungarian National Research, Development and Innovation Office
- STIA-KFI-2022 Semmelweis Scientific and Innovation fund
- STIA-KFI-2022 Semmelweis Scientific and Innovation fund
- 739593 Horizon 2020 Framework Programme
- 739593 Horizon 2020 Framework Programme
- TKP2021-EGA-24, TKP2021-NVA-15 National Research, Development and Innovation Fund
- TKP2021-EGA-24, TKP2021-NVA-15 National Research, Development and Innovation Fund
- EFOP-3.6.3-VEKOP-16-2017-00009 Complementary Research Excellence Program of Semmelweis University
- BO/00125/22 János Bolyai Research Scholarship
Collapse
Affiliation(s)
- Borbála Péterffy
- HCEMM-SE, MTA-SE "Lendület" Molecular Oncohematology Research Group, Department of Pathology and Experimental Cancer Research, Semmelweis University, 26 Üllői Str, 1085, Budapest, Hungary
| | - Tamás J Nádasi
- Pediatric Center, Semmelweis University, 7-9 Tűzoltó Str, 1094, Budapest, Hungary
| | - Szilvia Krizsán
- HCEMM-SE, MTA-SE "Lendület" Molecular Oncohematology Research Group, Department of Pathology and Experimental Cancer Research, Semmelweis University, 26 Üllői Str, 1085, Budapest, Hungary
- Pediatric Center, Semmelweis University, 7-9 Tűzoltó Str, 1094, Budapest, Hungary
| | - Anna Horváth
- Department of Pathology and Experimental Cancer Research, Semmelweis University, 26 Üllői Str, 1085, Budapest, Hungary
| | - Ágnes Márk
- Department of Pathology and Experimental Cancer Research, Semmelweis University, 26 Üllői Str, 1085, Budapest, Hungary
| | - Gábor Barna
- Department of Pathology and Experimental Cancer Research, Semmelweis University, 26 Üllői Str, 1085, Budapest, Hungary
| | - Botond Timár
- HCEMM-SE, MTA-SE "Lendület" Molecular Oncohematology Research Group, Department of Pathology and Experimental Cancer Research, Semmelweis University, 26 Üllői Str, 1085, Budapest, Hungary
| | - Laura Almási
- Pediatric Center, Semmelweis University, 7-9 Tűzoltó Str, 1094, Budapest, Hungary
| | - Judit Müller
- Pediatric Center, Semmelweis University, 7-9 Tűzoltó Str, 1094, Budapest, Hungary
| | - Krisztina Csanádi
- Hemato-Oncology Unit, Heim Pál Children's Hospital, 86 Üllői Str, 1089, Budapest, Hungary
| | - Anna Rakonczai
- Department of Internal Medicine and Hematology, Semmelweis University, 46 Szentkirályi Str, 1088, Budapest, Hungary
| | - Zsolt Nagy
- Department of Internal Medicine and Hematology, Semmelweis University, 46 Szentkirályi Str, 1088, Budapest, Hungary
| | - Krisztián Kállay
- Department of Pediatric Hematology and Stem Cell Transplantation, Central Hospital of Southern Pest, National Institute of Hematology and Infectious Diseases, 5-7 Albert Flórián Str, 1097, Budapest, Hungary
| | - Gabriella Kertész
- Department of Pediatric Hematology and Stem Cell Transplantation, Central Hospital of Southern Pest, National Institute of Hematology and Infectious Diseases, 5-7 Albert Flórián Str, 1097, Budapest, Hungary
| | - Gergely Kriván
- Department of Pediatric Hematology and Stem Cell Transplantation, Central Hospital of Southern Pest, National Institute of Hematology and Infectious Diseases, 5-7 Albert Flórián Str, 1097, Budapest, Hungary
| | - Monika Csóka
- Pediatric Center, Semmelweis University, 7-9 Tűzoltó Str, 1094, Budapest, Hungary
| | - Anna Sebestyén
- Department of Pathology and Experimental Cancer Research, Semmelweis University, 26 Üllői Str, 1085, Budapest, Hungary
| | - Ágnes F Semsei
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, 4 Nagyvárad Square, 1089, Budapest, Hungary
| | - Gábor T Kovács
- Pediatric Center, Semmelweis University, 7-9 Tűzoltó Str, 1094, Budapest, Hungary
| | - Dániel J Erdélyi
- Pediatric Center, Semmelweis University, 7-9 Tűzoltó Str, 1094, Budapest, Hungary
| | - Csaba Bödör
- HCEMM-SE, MTA-SE "Lendület" Molecular Oncohematology Research Group, Department of Pathology and Experimental Cancer Research, Semmelweis University, 26 Üllői Str, 1085, Budapest, Hungary
| | - Bálint Egyed
- HCEMM-SE, MTA-SE "Lendület" Molecular Oncohematology Research Group, Department of Pathology and Experimental Cancer Research, Semmelweis University, 26 Üllői Str, 1085, Budapest, Hungary.
- Pediatric Center, Semmelweis University, 7-9 Tűzoltó Str, 1094, Budapest, Hungary.
| | - Donát Alpár
- HCEMM-SE, MTA-SE "Lendület" Molecular Oncohematology Research Group, Department of Pathology and Experimental Cancer Research, Semmelweis University, 26 Üllői Str, 1085, Budapest, Hungary
| |
Collapse
|
316
|
Ju S, Singh MK, Han S, Ranbhise J, Ha J, Choe W, Yoon KS, Yeo SG, Kim SS, Kang I. Oxidative Stress and Cancer Therapy: Controlling Cancer Cells Using Reactive Oxygen Species. Int J Mol Sci 2024; 25:12387. [PMID: 39596452 PMCID: PMC11595237 DOI: 10.3390/ijms252212387] [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: 09/26/2024] [Revised: 10/31/2024] [Accepted: 11/13/2024] [Indexed: 11/28/2024] Open
Abstract
Cancer is a multifaceted disease influenced by various mechanisms, including the generation of reactive oxygen species (ROS), which have a paradoxical role in both promoting cancer progression and serving as targets for therapeutic interventions. At low concentrations, ROS serve as signaling agents that enhance cancer cell proliferation, migration, and resistance to drugs. However, at elevated levels, ROS induce oxidative stress, causing damage to biomolecules and leading to cell death. Cancer cells have developed mechanisms to manage ROS levels, including activating pathways such as NRF2, NF-κB, and PI3K/Akt. This review explores the relationship between ROS and cancer, focusing on cell death mechanisms like apoptosis, ferroptosis, and autophagy, highlighting the potential therapeutic strategies that exploit ROS to target cancer cells.
Collapse
Affiliation(s)
- Songhyun Ju
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.); (M.K.S.); (S.H.); (J.R.); (J.H.); (W.C.); (K.-S.Y.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Manish Kumar Singh
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.); (M.K.S.); (S.H.); (J.R.); (J.H.); (W.C.); (K.-S.Y.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Sunhee Han
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.); (M.K.S.); (S.H.); (J.R.); (J.H.); (W.C.); (K.-S.Y.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Jyotsna Ranbhise
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.); (M.K.S.); (S.H.); (J.R.); (J.H.); (W.C.); (K.-S.Y.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Joohun Ha
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.); (M.K.S.); (S.H.); (J.R.); (J.H.); (W.C.); (K.-S.Y.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Wonchae Choe
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.); (M.K.S.); (S.H.); (J.R.); (J.H.); (W.C.); (K.-S.Y.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Kyung-Sik Yoon
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.); (M.K.S.); (S.H.); (J.R.); (J.H.); (W.C.); (K.-S.Y.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Seung Geun Yeo
- Department of Otorhinolaryngology—Head and Neck Surgery, College of Medicine, Kyung Hee University Medical Center, Kyung Hee University, Seoul 02453, Republic of Korea;
| | - Sung Soo Kim
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.); (M.K.S.); (S.H.); (J.R.); (J.H.); (W.C.); (K.-S.Y.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Insug Kang
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.); (M.K.S.); (S.H.); (J.R.); (J.H.); (W.C.); (K.-S.Y.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
317
|
Gao L, Zhang Y, Feng M, Shen M, Yang L, Wei B, Zhou Y, Zhang Z. HER3: Updates and current biology function, targeted therapy and pathologic detecting methods. Life Sci 2024; 357:123087. [PMID: 39366553 DOI: 10.1016/j.lfs.2024.123087] [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: 07/20/2024] [Revised: 09/18/2024] [Accepted: 09/28/2024] [Indexed: 10/06/2024]
Abstract
Being a member of the EGFR tyrosine kinase family, HER3 has been shown to be overexpressed in a number of cancers, including breast cancer (BC). The kinase activity of HER3 is extremely low, and it forms heterodimers with partners, HER2 in particular, that promote biological processes like cell migration, survival, and proliferation by activating downstream carcinogenic signaling pathways. The overexpression of HER3 is also directly linked to tumor invasion, metastasis, and a poor prognosis. Despite the relatively low expression of HER3 compared to EGFR and HER2, a lot of targeted drugs are making their way into clinical trials and seem to have a bright further. This review aims to summarize the relationship between HER3 overexpression, mutations, and carcinogenicity and drug resistance, starting from the unique structure and kinase activity of HER3. Simultaneously, numerous approaches to HER3 targeted therapy are enumerated, and the clinical detection methods for HER3 that are commonly employed in pathology are sorted and contrasted to offer physicians a range of options. We think that a better knowledge of the mechanisms underlying HER3 in tumors and the advancement of targeted HER3 therapy will contribute to an improved prognosis for cancer patients and an increase in the efficacy of anticancer therapies.
Collapse
Affiliation(s)
- Leyi Gao
- Department of Pathology, West China Hospital, Sichuan University, No.37, Guo Xue Xiang, Chengdu 610041, Sichuan, China
| | - Yu Zhang
- Department of Pathology, West China Hospital, Sichuan University, No.37, Guo Xue Xiang, Chengdu 610041, Sichuan, China
| | - Mengna Feng
- Department of Pathology, West China Hospital, Sichuan University, No.37, Guo Xue Xiang, Chengdu 610041, Sichuan, China
| | - Mengjia Shen
- Department of Pathology, West China Hospital, Sichuan University, No.37, Guo Xue Xiang, Chengdu 610041, Sichuan, China
| | - Libo Yang
- Department of Pathology, West China Hospital, Sichuan University, No.37, Guo Xue Xiang, Chengdu 610041, Sichuan, China
| | - Bing Wei
- Department of Pathology, West China Hospital, Sichuan University, No.37, Guo Xue Xiang, Chengdu 610041, Sichuan, China
| | - Yongjie Zhou
- Laboratory of Liver Transplantation, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Zhang Zhang
- Department of Pathology, West China Hospital, Sichuan University, No.37, Guo Xue Xiang, Chengdu 610041, Sichuan, China.
| |
Collapse
|
318
|
Huang P, Zhao H, Dai H, Li J, Pan X, Pan W, Xia C, Liu F. FXR deficiency induced ferroptosis via modulation of the CBP-dependent p53 acetylation to suppress breast cancer growth and metastasis. Cell Death Dis 2024; 15:826. [PMID: 39543094 PMCID: PMC11564727 DOI: 10.1038/s41419-024-07222-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 10/31/2024] [Accepted: 11/04/2024] [Indexed: 11/17/2024]
Abstract
Farnesoid X receptor (NR1H4/FXR) functions as a scavenger of lipid peroxide products and drives the proliferation and metastasis of various cancers. However, the underlying molecular mechanisms remain poorly understood. In our study, we found that the expression levels of FXR, vimentin and SLC7A11 were significantly higher in breast cancer tissues, particularly in metastatic cancer tissues compared to non-metastatic ones. Furthermore, the increased FXR expression was positively correlated with vimentin and SLC7A11 in clinical tumor specimens. In addition, a high level of FXR correlated with poor prognosis in patients with breast cancer. Both Z-Guggulsterone (Z-GS), as a pharmacological inhibitor of FXR, and silencing FXR curbed proliferation and migration of breast cancer cells by promoting ferroptosis. Notably, our results showed that FXR competitively bound to CREB-binding protein (CBP) to suppress the interaction between p53 and CBP in the nucleus, and thus prevented p53 acetylation at lys382, which was essential for upregulating the expression of SLC7A11. Conversely, FXR knockdown increased the interaction between p53 and CBP and promoted p53 acetylation, which ultimately led to facilitating ferroptosis in breast cancer cells. More importantly, we also found that Z-GS inhibited TGF-β1-induced tumor growth and metastasis of breast cancer primarily through ferroptosis via regulating CBP-dependent p53 acetylation in nude mice. In conclusion, the FXR was first reported as a tumor promoter that enhanced the proliferation and metastasis of breast cancer cells through regulating CBP-dependent p53 K382 acetylation. It proposes that FXR may serve as a potential therapeutic target for the treatment of breast cancer.
Collapse
Affiliation(s)
- Ping Huang
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, 330031, P. R. China
| | - Han Zhao
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, 330031, P. R. China
| | - Hua Dai
- Department of Pathology, the First Affiliated Hospital of Nanchang University, Nanchang, 330038, P. R. China
| | - Jinying Li
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, 330031, P. R. China
| | - Xiafang Pan
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, 330031, P. R. China
| | - Wentian Pan
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, 330031, P. R. China
| | - Chunhua Xia
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, 330031, P. R. China.
- Jiangxi Province Key Laboratory of New Drug Evaluation and Transformation, Nanchang, 330031, P. R. China.
| | - Fanglan Liu
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, 330031, P. R. China.
- Jiangxi Province Key Laboratory of New Drug Evaluation and Transformation, Nanchang, 330031, P. R. China.
| |
Collapse
|
319
|
Xu Q, Li Y, Qin X, Xin Y, Wang J, Zhang Y, Xu K, Yang X, Wang X. osa-miR168a, a Plant miRNA That Survives the Process of In Vivo Food Digestion, Attenuates Dextran Sulfate Sodium-Induced Colitis in Mice by Oral Administration. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:25146-25160. [PMID: 39480689 DOI: 10.1021/acs.jafc.4c07283] [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: 11/02/2024]
Abstract
Previous studies showed that osa-miR168a, a plant miRNA rich in fruits and vegetables, had cross-kingdom biological effects on immunocytes, silkworms, and rodents. In this study, the effects of miR168a on mouse colitis induced by dextran sulfate sodium (DSS) were investigated. The results showed that miR168a oligomers were resistant during the process of food digestion, ending up with a residual concentration of 67.8 ± 11.2 fM in mouse intestines 4 h after oral gavage. More importantly, direct oral administration of the miRNA to the colitis mice significantly ameliorated the progression of the disease, as evidenced by the reduction in DAI score, histopathological lesions, and proinflammatory cytokines. Repairing intestinal barrier function by promoting the regeneration of TJ proteins and the mucus layer, suppressing oxidative stress and colonic inflammation via modulating Nrf2 and NF-κB signaling pathways, and restoring the imbalanced gut microbiota caused by DSS are proposed mechanisms behind the anticolitis activity of miR168a. This study provided new evidence of the cross-kingdom regulatory effects of dietary miRNAs, suggesting the potential of the plant miRNA for the prevention and treatment of inflammatory bowel diseases.
Collapse
Affiliation(s)
- Qin Xu
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, Shaanxi 710062, China
| | - Ying Li
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, Shaanxi 710062, China
| | - Xinshu Qin
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, Shaanxi 710062, China
- Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
| | - Yirao Xin
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, Shaanxi 710062, China
| | - Jianing Wang
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, Shaanxi 710062, China
| | - Yi Zhang
- Department of Food Science, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Ke Xu
- Department of Joint Surgery, Hong Hui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710054, China
| | - Xingbin Yang
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, Shaanxi 710062, China
| | - Xingyu Wang
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, Shaanxi 710062, China
| |
Collapse
|
320
|
Sánchez-Castillo A, Kampen KR. Understanding serine and glycine metabolism in cancer: a path towards precision medicine to improve patient's outcomes. Discov Oncol 2024; 15:652. [PMID: 39538085 PMCID: PMC11561223 DOI: 10.1007/s12672-024-01544-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 11/06/2024] [Indexed: 11/16/2024] Open
Abstract
In this perspective, we highlight and reflect on the current knowledge with respect to serine/glycine metabolism in cancer, therapeutic resistance, and precision medicine opportunities for therapeutic targeting and treatment follow-up. Cancer subtypes with high mortality rates include lung cancer and glioblastomas. In order to improve future therapeutic opportunities, patient stratification need to be performed to select patients that might benefit from adjuvant serine/glycine targeting compounds. In an effort to identify the group of patients for stratification purposes, we analyzed publicly available TCGA patient datasets to test associations between serine/glycine metabolism enzyme expression and important cancer drivers in lung cancer and glioblastoma. These patients presenting serine/glycine pathway overexpression might benefit from adjuvant sertraline treatment in the future.
Collapse
Affiliation(s)
- Anaís Sánchez-Castillo
- Department of Radiation Oncology (MAASTRO), GROW School for Oncology and Reproduction, Maastricht University Medical Center, Maastricht University, Maastricht, The Netherlands
| | - Kim R Kampen
- Department of Radiation Oncology (MAASTRO), GROW School for Oncology and Reproduction, Maastricht University Medical Center, Maastricht University, Maastricht, The Netherlands.
- Department of Oncology, Laboratory for Disease Mechanisms in Cancer, KU Leuven and Leuven Cancer Institute (LKI), Louvain, Belgium.
| |
Collapse
|
321
|
Ou M, Deng Z, Shi Y, He J, Ye Z, Guo M, Cheng G, Wu J, Lv L. Mechanism of Apigenin against breast cancer stem cells: network pharmacology and experimental validation. Front Pharmacol 2024; 15:1496664. [PMID: 39605916 PMCID: PMC11598448 DOI: 10.3389/fphar.2024.1496664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Accepted: 10/31/2024] [Indexed: 11/29/2024] Open
Abstract
Apigenin (API), a traditionally sourced flavonoid, is recognized for its anti-neoplastic properties. Despite well-documented effects on tumorigenesis, the detailed therapeutic impact on breast cancer stem cells (BCSCs) and the associated molecular mechanisms are yet to be clarified. The objective of this study is to elucidate the therapeutic effects of API on BCSCs and to uncover its molecular mechanisms through network pharmacology and experimental validation. Interactions of API with candidate targets were examined through target screening, enrichment analysis, construction of protein-protein interaction networks, and molecular docking. MCF-7-derived BCSCs were utilized as a model system to investigate and substantiate the anti-BCSC effects of API and the underlying mechanism. Molecular docking studies have shown that API and TP53 exhibit favorable binding affinity. Compared with the negative control group, API effectively suppressed the expression of BCSC-related proteins such as ALDH1A1, NANOG, EpCAM, and MYC, downregulated p-PI3K and p-AKT, and upregulated p53. This study demonstrates that API can play an anti-BCSC role by regulating the PI3K/AKT/p53 pathway in BCSCs of MCF-7 cells, highlighting its potential as a therapeutic agent for targeting BCSCs.
Collapse
Affiliation(s)
- Mengdie Ou
- School of Pharmacy, Jinan University, Guangzhou, China
- Department of Pharmacy, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Zhicheng Deng
- Guangdong Provincial Key Laboratory of Cancer Pathogenesis and Precision Diagnosis and Treatment, Shenshan Medical Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Shanwei, China
| | - Yonghui Shi
- Department of Pharmacy, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jianxiong He
- Department of Pharmacy, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Zicong Ye
- Department of Pharmacy, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Ming Guo
- Department of Pharmacy, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Guohua Cheng
- School of Pharmacy, Jinan University, Guangzhou, China
| | - Junyan Wu
- Department of Pharmacy, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Li Lv
- Department of Pharmacy, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
322
|
Zhu J, Zhou W, Yao Y, Zhou X, Ma X, Zhang B, Yang Z, Tang B, Zhu H, Li N. Targeted Positron Emission Tomography-Tracked Biomimetic Codelivery Synergistically Amplifies Ferroptosis and Pyroptosis for Inducing Lung Cancer Regression and Anti-PD-L1 Immunotherapy Efficacy. ACS NANO 2024; 18:31401-31420. [PMID: 39475541 DOI: 10.1021/acsnano.4c11278] [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: 11/13/2024]
Abstract
The chemoresistance and systemic toxicity of cisplatin (CDDP) severely limit its application in the treatment of non-small cell lung cancer (NSCLC). Here, I-124 labeled cancer cell membrane biomimetic nanovesicles loading Polyphyllin VI (PPVI) and CDDP (termed 124I-P/C@CMLvs) were constructed to enhance the sensitivity and efficacy of CDDP. The radiochemical purity (RCP) of 124I-P/C@CMLvs reached more than 99% and maintained reliable stability in vitro. Micro-positron emission tomography (micro-PET) imaging of I-124 quantitatively revealed the distribution and specific homologous tumor targeting ability of 124I-P/C@CMLvs in vivo with superior diagnosis performance, beneficial for dynamically monitoring the efficacy against NSCLC. Loaded PPVI significantly strengthened the sensitivity of NSCLC to CDDP therapy and exerted synergistic anti-tumor effect in vitro and in vivo, which was achieved by PPVI promoting p53 deubiquitination and stimulating reactive oxygen species (ROS) production to trigger the crosstalk between the amplification of GPX4 signaling-mediated ferroptosis and NLRP3/GSDMD/Caspase-1 axis-mediated pyroptosis. 124I-P/C@CMLvs also significantly stimulated the infiltration of immune cells including dendritic cells, CD8+ T cells, and CD4+ T cells in tumor tissues (P < 0.05). The combination of 124I-P/C@CMLvs and anti-PD-L1 therapy further synergistically promoted NSCLC regression. Altogether, 124I-P/C@CMLvs provide a transformational solution to the challenge of improving CDDP sensitivity and realizing the integration of diagnosis, treatment, and monitoring of NSCLC.
Collapse
Affiliation(s)
- Jinyu Zhu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Peking University, Beijing 100142, China
| | - Wenyuan Zhou
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Peking University, Beijing 100142, China
| | - Yuan Yao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Peking University, Beijing 100142, China
| | - Xin Zhou
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Peking University, Beijing 100142, China
| | - Xiaokun Ma
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Peking University, Beijing 100142, China
| | - Baohui Zhang
- Department of Physiology, School of Life Science, China Medical University, Shenyang,Liaoning 110122, China
| | - Zhi Yang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Peking University, Beijing 100142, China
| | - Bufu Tang
- Department of Radiation Oncology, Zhongshan Hospital Affiliated to Fudan University, Shanghai 200032,China
| | - Hua Zhu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Peking University, Beijing 100142, China
| | - Nan Li
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Peking University, Beijing 100142, China
| |
Collapse
|
323
|
Li L, Li L, Wang Y, Wu B, Guan Y, Chen Y, Zhao J. Integration of Machine Learning and Experimental Validation to Identify Anoikis-Related Prognostic Signature for Predicting the Breast Cancer Tumor Microenvironment and Treatment Response. Genes (Basel) 2024; 15:1458. [PMID: 39596658 PMCID: PMC11594124 DOI: 10.3390/genes15111458] [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: 10/21/2024] [Revised: 11/07/2024] [Accepted: 11/11/2024] [Indexed: 11/29/2024] Open
Abstract
Background/Objectives: Anoikis-related genes (ANRGs) are crucial in the invasion and metastasis of breast cancer (BC). The underlying role of ANRGs in the prognosis of breast cancer patients warrants further study. Methods: The anoikis-related prognostic signature (ANRS) was generated using a variety of machine learning methods, and the correlation between the ANRS and the tumor microenvironment (TME), drug sensitivity, and immunotherapy was investigated. Moreover, single-cell analysis and spatial transcriptome studies were conducted to investigate the expression of prognostic ANRGs across various cell types. Finally, the expression of ANRGs was verified by RT-PCR and Western blot analysis (WB), and the expression level of PLK1 in the blood was measured by the enzyme-linked immunosorbent assay (ELISA). Results: The ANRS, consisting of five ANRGs, was established. BC patients within the high-ANRS group exhibited poorer prognoses, characterized by elevated levels of immune suppression and stromal scores. The low-ANRS group had a better response to chemotherapy and immunotherapy. Single-cell analysis and spatial transcriptomics revealed variations in ANRGs across cells. The results of RT-PCR and WB were consistent with the differential expression analyses from databases. NU.1025 and imatinib were identified as potential inhibitors for SPIB and PLK1, respectively. Additionally, findings from ELISA demonstrated increased expression levels of PLK1 in the blood of BC patients. Conclusions: The ANRS can act as an independent prognostic indicator for BC patients, providing significant guidance for the implementation of chemotherapy and immunotherapy in these patients. Additionally, PLK1 has emerged as a potential blood-based diagnostic marker for breast cancer patients.
Collapse
Affiliation(s)
- Longpeng Li
- Institute of Physical Education and Sport, Shanxi University, Taiyuan 030006, China; (L.L.)
| | - Longhui Li
- School of Kinesiology and Health, Capital University of Physical Education and Sports, Beijing 100191, China
| | - Yaxin Wang
- Institute of Physical Education and Sport, Shanxi University, Taiyuan 030006, China; (L.L.)
| | - Baoai Wu
- Institute of Physical Education and Sport, Shanxi University, Taiyuan 030006, China; (L.L.)
| | - Yue Guan
- Institute of Physical Education and Sport, Shanxi University, Taiyuan 030006, China; (L.L.)
| | - Yinghua Chen
- Institute of Physical Education and Sport, Shanxi University, Taiyuan 030006, China; (L.L.)
| | - Jinfeng Zhao
- Institute of Physical Education and Sport, Shanxi University, Taiyuan 030006, China; (L.L.)
| |
Collapse
|
324
|
Zhang LK, Li Y, Zhai L, Tang Y, Jiao Y, Mei Y, Yang R, You R, Yin L, Ni H, Ge J, Guan YQ. Natural Phycocyanin/Paclitaxel Micelle Delivery of Therapeutic P53 to Activate Apoptosis for HER2 or ER Positive Breast Cancer Therapy. ACS Biomater Sci Eng 2024; 10:6995-7004. [PMID: 39390952 DOI: 10.1021/acsbiomaterials.4c00756] [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: 10/12/2024]
Abstract
The P53 gene is commonly mutated in breast cancer, protein based the gene as anticancer drugs could provide efficient and stable advantages by restoring the function of the wild-type P53 protein. In this study, we describe the creation and utilization of a micelle composed by natural phycocyanin and paclitaxel and grafting anti-HER2 (PPH), which effectively packages and transports recombinant P53 protein with anti-ER (PE), resulting in a new entity designated as PE@PPH, to address localization obstacles and modify cellular tropism to the cell membrane or nucleus. The results indicate that PE@PPH has strong antitumor properties, even at low doses of PTX both in vitro and in vivo. These findings suggest that PE@PPH could be an enhancing micelle for delivering therapeutic proteins and promoting protein functional recovery, particularly in addressing the challenges posed by tumor heterogeneity in breast cancer.
Collapse
Affiliation(s)
- Ling-Kun Zhang
- School of Life Science, South China Normal University, Guangzhou 510631, China
- School of Engineering, Westlake University, Hangzhou 310030, China
- Institute of Advanced Technology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang 310024, China
| | - Yuan Li
- School of Life Science, South China Normal University, Guangzhou 510631, China
| | - Limin Zhai
- School of Life Science, South China Normal University, Guangzhou 510631, China
| | - Yunzhi Tang
- School of Life Science, South China Normal University, Guangzhou 510631, China
| | - Yuxuan Jiao
- School of Life Science, South China Normal University, Guangzhou 510631, China
| | - Yitong Mei
- School of Life Science, South China Normal University, Guangzhou 510631, China
| | - Runcai Yang
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong 510060, China
| | - Rong You
- School of Life Science, South China Normal University, Guangzhou 510631, China
| | - Liang Yin
- School of Life Science, South China Normal University, Guangzhou 510631, China
| | - He Ni
- School of Life Science, South China Normal University, Guangzhou 510631, China
| | - Jian Ge
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong 510060, China
| | - Yan-Qing Guan
- School of Life Science, South China Normal University, Guangzhou 510631, China
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, Guangdong Provincial Key Laboratory of Laser Life Science, Guangzhou Key Laboratory of Spectral Analysis and Functional Probes, College of Biophotonics, South China Normal University, Guangzhou 510631, China
| |
Collapse
|
325
|
López I, Valdivia IL, Vojtesek B, Fåhraeus R, Coates P. Re-appraising the evidence for the source, regulation and function of p53-family isoforms. Nucleic Acids Res 2024; 52:12112-12129. [PMID: 39404067 PMCID: PMC11551734 DOI: 10.1093/nar/gkae855] [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: 04/29/2024] [Revised: 09/12/2024] [Accepted: 09/19/2024] [Indexed: 11/12/2024] Open
Abstract
The p53 family of proteins evolved from a common ancestor into three separate genes encoding proteins that act as transcription factors with distinct cellular roles. Isoforms of each member that lack specific regions or domains are suggested to result from alternative transcription start sites, alternative splicing or alternative translation initiation, and have the potential to exponentially increase the functional repertoire of each gene. However, evidence supporting the presence of individual protein variants at functional levels is often limited and is inferred by mRNA detection using highly sensitive amplification techniques. We provide a critical appraisal of the current evidence for the origins, expression, functions and regulation of p53-family isoforms. We conclude that despite the wealth of publications, several putative isoforms remain poorly established. Future research with improved technical approaches and the generation of isoform-specific protein detection reagents is required to establish the physiological relevance of p53-family isoforms in health and disease. In addition, our analyses suggest that p53-family variants evolved partly through convergent rather than divergent evolution from the ancestral gene.
Collapse
Affiliation(s)
- Ignacio López
- Biochemistry, Faculty of Science, Universidad de la República, Iguá 4225, Montevideo 11400, Uruguay
- Cell Biology Unit, Institut Pasteur de Montevideo, Mataojo 2020, Montevideo 11400, Uruguay
| | - Irene Larghero Valdivia
- Biochemistry, Faculty of Science, Universidad de la República, Iguá 4225, Montevideo 11400, Uruguay
| | - Borivoj Vojtesek
- RECAMO, Masaryk Memorial Cancer Institute, Zluty kopec 7, Brno 65653, Czech Republic
| | - Robin Fåhraeus
- RECAMO, Masaryk Memorial Cancer Institute, Zluty kopec 7, Brno 65653, Czech Republic
- Inserm UMRS 1131, Institut de Génétique Moléculaire, Université de Paris Cité, 27 rue Juliette Dodu, Hôpital St. Louis, Paris F-75010, France
- Department of Medical Biosciences, Building 6M, Umeå University, Umeå 90185, Sweden
| | - Philip J Coates
- RECAMO, Masaryk Memorial Cancer Institute, Zluty kopec 7, Brno 65653, Czech Republic
| |
Collapse
|
326
|
Zarrabian M, Sherif SM. Silence is not always golden: A closer look at potential environmental and ecotoxicological impacts of large-scale dsRNA application. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 950:175311. [PMID: 39122031 DOI: 10.1016/j.scitotenv.2024.175311] [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: 06/05/2024] [Revised: 08/02/2024] [Accepted: 08/04/2024] [Indexed: 08/12/2024]
Abstract
RNA interference (RNAi) technology has emerged as a pivotal strategy in sustainable pest management, offering a targeted approach that significantly mitigates the environmental and health risks associated with traditional insecticides. Originally implemented through genetically modified organisms (GMOs) to produce specific RNAi constructs, the technology has evolved in response to public and regulatory concerns over GMOs. This evolution has spurred the development of non-transgenic RNAi applications such as spray-induced gene silencing (SIGS), which employs double-stranded RNA (dsRNA) to silence pest genes directly without altering the plant's genetic makeup. Despite its advantages in specificity and reduced ecological footprint, SIGS faces significant obstacles, particularly the instability of dsRNA in field conditions, which limits its practical efficacy. To overcome these limitations, innovative delivery mechanisms have been developed. These include nanotechnology-based systems, minicells, and nanovesicles, which are designed to protect dsRNA from degradation and enhance its delivery to target organisms. While these advancements have improved the stability and application efficiency of dsRNA, comprehensive assessments of their environmental safety and the potential for increased exposure risks to non-target organisms remain incomplete. This comprehensive review aims to elucidate the environmental fate of dsRNA and evaluate the potential risks associated with its widespread application on non-target organisms, encompassing soil microorganisms, beneficial insects, host plants, and mammals. The objective is to establish a more refined framework for RNAi risk assessment within environmental and ecotoxicological contexts, thereby fostering the development of safer, non-transgenic RNAi-based pest control strategies.
Collapse
Affiliation(s)
- Mohammad Zarrabian
- Virginia Tech, School of Plant and Environmental Sciences, Alson H. Smith Jr. Agricultural Research, and Extension Center, Winchester, VA 22602, United States
| | - Sherif M Sherif
- Virginia Tech, School of Plant and Environmental Sciences, Alson H. Smith Jr. Agricultural Research, and Extension Center, Winchester, VA 22602, United States.
| |
Collapse
|
327
|
Feng B, Pei Y, Zhang W, Zheng Q, Zhou Y. Asparaginase and isoaspartyl peptidase 1 RNA interference suppresses the growth of nasopharyngeal carcinoma cells. Discov Oncol 2024; 15:636. [PMID: 39520610 PMCID: PMC11550296 DOI: 10.1007/s12672-024-01228-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 08/08/2024] [Indexed: 11/16/2024] Open
Abstract
Nasopharyngeal carcinoma (NPC) is one of the common malignant tumors, and its pathogenesis has not been fully clarified. This study aims to explore the impact of RNA interference on the growth and invasion of NPC cells. Asparaginase and isoaspartyl peptidase 1 (ASRGL1)-short hairpin(sh) RNA expressing lentivirus was used to investigate the effect of ASRGL1 knockdown on NPC cells (C666-1 and SUN-1). The target shASRGL1 gene was determined by mRNA and protein expression in nasopharyngeal carcinoma cells; nasopharyngeal carcinoma cell proliferation viability, migration, invasion, apoptosis, ATP levels, and oxidative stress were examined. The results found that ASRGL1 was found to be highly expressed in NPC tissues and cell lines. shASRGL1 exhibited a high gene expression knockdown efficiency, downregulated the ASRGL1 protein expression in the nasopharyngeal carcinoma cells, suppressed proliferation viability of transfected nasopharyngeal carcinoma cells, inhibited their migration and invasion and ATP levels, promoted nasopharyngeal carcinoma cell apoptosis, ROS, and ferroptosis. shASRGL1 plays a role in protecting against NPC cell growth and invasion.
Collapse
Affiliation(s)
- Bo Feng
- Department of Otorhinolaryngology, The First Affiliated Hospital of Ningbo University, No.56 Liuting Street, Ningbo, 315020, Zhejiang, China.
| | - Yingying Pei
- Department of Otorhinolaryngology, The First Affiliated Hospital of Ningbo University, No.56 Liuting Street, Ningbo, 315020, Zhejiang, China
| | - Weiwei Zhang
- Department of Otorhinolaryngology, The First Affiliated Hospital of Ningbo University, No.56 Liuting Street, Ningbo, 315020, Zhejiang, China
| | - Qi Zheng
- Department of Otorhinolaryngology, The First Affiliated Hospital of Ningbo University, No.56 Liuting Street, Ningbo, 315020, Zhejiang, China
| | - Yan Zhou
- Department of Otorhinolaryngology, The First Affiliated Hospital of Ningbo University, No.56 Liuting Street, Ningbo, 315020, Zhejiang, China
| |
Collapse
|
328
|
Yang X, Liu Y, Wang Z, Jin Y, Gu W. Ferroptosis as a new tool for tumor suppression through lipid peroxidation. Commun Biol 2024; 7:1475. [PMID: 39521912 PMCID: PMC11550846 DOI: 10.1038/s42003-024-07180-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
As a newly defined type of programmed cell death, ferroptosis is considered a potent weapon against tumors due to its distinct mechanism from other types of programmed cell death. Ferroptosis is triggered by the uncontrolled accumulation of hydroperoxyl polyunsaturated fatty acid-containing phospholipids, also called lipid peroxidation. The lipid peroxidation, generated through enzymatic and non-enzymatic mechanisms, drives changes in cell morphology and the destruction of membrane integrity. Here, we dissect the mechanisms of ferroptosis induced enzymatically or non-enzymatically, summarize the major metabolism pathways in modulating lipid peroxidation, and provide insights into the relationship between ferroptosis and tumor suppression. In this review, we discuss the recent advances of ferroptosis in tumor microenvironments and the prospect of potential therapeutic application.
Collapse
Affiliation(s)
- Xin Yang
- Suzhou Ninth Hospital Affiliated to Soochow University, The Institutes of Biology and Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China.
- Institute for Cancer Genetics, and Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA.
| | - Yanqing Liu
- Institute for Cancer Genetics, and Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Zhe Wang
- Institute for Cancer Genetics, and Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Ying Jin
- Suzhou Ninth Hospital Affiliated to Soochow University, Suzhou Ninth People's Hospital, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Wei Gu
- Institute for Cancer Genetics, and Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA.
- Department of Pathology and Cell Biology, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA.
| |
Collapse
|
329
|
Jansen J, Dobbelstein M. MDM4 exon skipping upon dysfunctional ribosome assembly. Trends Cell Biol 2024:S0962-8924(24)00212-5. [PMID: 39516053 DOI: 10.1016/j.tcb.2024.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 10/15/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024]
Abstract
Recent studies revealed how nucleolar stress enhances MDM4 exon skipping and activates p53 via the ribosomal protein L22 (RPL22; eL22). Tumor-associated L22 mutations lead to full-length MDM4 synthesis, overcoming tumor suppression by p53. This forum article explores how MDM4 splicing patterns integrate stress signaling to take p53-dependent cell fate decisions.
Collapse
Affiliation(s)
- Jennifer Jansen
- Department of Molecular Oncology, Göttingen Center of Molecular Biosciences (GZMB), University Medical Center Göttingen, Justus-von-Liebig-Weg 11, 37077 Göttingen, Germany.
| | - Matthias Dobbelstein
- Department of Molecular Oncology, Göttingen Center of Molecular Biosciences (GZMB), University Medical Center Göttingen, Justus-von-Liebig-Weg 11, 37077 Göttingen, Germany; Max Planck Institute for Multidisciplinary Sciences, Am Fassberg 11, 37077 Göttingen, Germany.
| |
Collapse
|
330
|
Moretti M, Farina A, Angeloni A, Anastasi E. Emerging horizons on molecular and circulating biomarkers in pancreatic adenocarcinoma. Front Oncol 2024; 14:1483306. [PMID: 39575418 PMCID: PMC11578827 DOI: 10.3389/fonc.2024.1483306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 10/17/2024] [Indexed: 11/24/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the most aggressive and invasive type of pancreatic cancer and is expected to soon become the second leading cause of cancer-associated death. The high mortality rate is due to the clinical features that allow asymptomatic progression to advanced stages, a period when current therapeutic treatments have limited efficacy. To address these challenges, researchers are focused on identifying new molecular and circulating markers for early PDAC detection and precision medicine. In this mini-review, we report the most well-known and recently identified molecular and circulating biomarkers. This study aimed to emphasize the need for continued innovative research to develop diagnostic algorithms and therapies to improve the management of patients with PDAC.
Collapse
Affiliation(s)
| | | | | | - Emanuela Anastasi
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
331
|
Chen XL, Ojalill M, Jean C, Tancioni I, Jiang S, Boyer A, Ozmadenci D, Uryu S, Tarin D, Schlessinger J, Stupack DG, Schlaepfer DD. Inducible FAK Deletion but not FAK Inhibition in Endothelial Cells Activates p53 to Suppress Tumor Growth in PYK2-null Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.04.622008. [PMID: 39574770 PMCID: PMC11580918 DOI: 10.1101/2024.11.04.622008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2025]
Abstract
Focal adhesion kinase (FAK) functions as a signaling and scaffolding protein within endothelial cells (ECs) impacting blood vessel function and tumor growth. Interpretations of EC FAK-null phenotypes are complicated by related PYK2 (protein tyrosine kinase 2) expression, and to test this, we created PYK2 -/- FAK fl/fl mice with tamoxifen-inducible EC-specific Cre recombinase expression. At 11 weeks of age, EC FAK inactivation resulted in increased heart and lung mass and vascular leakage only on a PYK2 -/- background. Surprisingly, ∼90% of PYK2 -/- EC FAK -/- mice survived to 75 weeks of age. Syngeneic melanoma, breast, or lung carcinoma tumors did not grow in PYK2 -/- EC FAK -/- mice, but tumors grew normally in PYK2 -/- EC FAK fl/fl mice lacking Cre. This tumor inhibitory phenotype was associated with abortive EC vessel sprouting, enhanced EC p53 tumor suppressor and p21CIP1 (cyclin-dependent inhibitor 1) expression, and alterations in serum cytokine levels. To discern the role of FAK kinase versus scaffolding activity in ECs, we generated kinase defective (FAK K454R, KD) PYK2 -/- EC FAK fl/KD and PYK2 -/- EC FAK fl/WT (WT, wildtype) mice. Hemizygous EC FAK -/KD expression supported primary tumor growth but not metastasis, implicating EC FAK activity in tumor dissemination. In vitro , hemizygous expression of either WT or KD FAK suppressed EC p21CIP1 levels and cell death observed in primary PYK2 -/- EC FAK -/- ECs. Combined FAK and PYK2 knockdown in tumor cells also increased p21CIP1 and PARP1 (poly ADP-ribose polymerase 1) levels in a p53-associated manner impacting anchorage-independent growth. Together, these results underscore the linkage between PYK2 and FAK loss with p53 activation impacting tumor growth. Impact Statement PYK2-null combined with endothelial cell-specific FAK transgenic mouse models show that loss of FAK activity limits tumor spread and that genetic or chemical degradation preventing combined FAK-PYK2 expression may be an approach to induce a p53-associated anti-tumor response.
Collapse
|
332
|
Zuo X, Zhao X, Zhang X, Li Q, Jiang X, Huang S, Chen X, Chen X, Jia W, Zou H, Shi D, Qian X. PTPN20 promotes metastasis through activating NF-κB signaling in triple-negative breast cancer. Breast Cancer Res 2024; 26:155. [PMID: 39506852 PMCID: PMC11542355 DOI: 10.1186/s13058-024-01910-w] [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: 07/22/2024] [Accepted: 10/22/2024] [Indexed: 11/08/2024] Open
Abstract
BACKGROUND Cancer metastasis remains a major challenge in the clinical management of triple-negative breast cancer (TNBC). The NF-κB signaling pathway has been implicated as a crucial factor in the development of metastases, but the underlying molecular mechanisms remain largely unclear. METHODS PTPN20 expression was evaluated using data from the Sweden Cancerome Analysis Network-Breast and The Cancer Genome Atlas database, as well as by western blotting and immunohistochemistry in 88 TNBC patients. The ability of PTPN20 to activate NF-κB was assessed by luciferase reporter assays. The effects of PTPN20 overexpression and knockdown via short hairpin RNA were examined in TNBC cell lines by wound healing and transwell matrix penetration assays. Additionally, we analyzed the growth and metastasis abilitiy of 4T1 xenograft tumors in nude mice. RESULTS PTPN20 levels were elevated in TNBC cell lines and patient samples compared to controls, and higher protein levels correlated with metastasis-free survival. Overexpression of PTPN20 enhanced migration and invasion in vitro, and promoted lung metastasis in vivo. Our finding revealed that PTPN20 activates NF-κB signaling by dephosphorylating p65 at Ser468, preventing its binding to COMMD1, thereby protecting p65 from degradation. Downregulation of PTPN20 effectively inhibit, while p65 S468A mutant restored the migratory and invasive abilities of TNBC cells. CONCLUSIONS Collectively, our results demonstrate that PTPN20 plays a critical role in TNBC metastasis through the activation of NF-κB signaling. We propose that PTPN20 may serve as a novel prognostic marker and potential therapeutic target for the treatment of TNBC.
Collapse
Affiliation(s)
- Xiaoxiao Zuo
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
- Department of Radiotherapy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Xiaohan Zhao
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, China
| | - Xiaofei Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Qingyuan Li
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, China
| | - Xingyu Jiang
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, China
| | - Shumei Huang
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Xuwei Chen
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, China
| | - Xiangfu Chen
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, China
| | - Weihua Jia
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, China
- Department of Biobank, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, China
| | - Hequn Zou
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, China.
| | - Dongni Shi
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, China.
| | - Xueke Qian
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China.
| |
Collapse
|
333
|
Luo S, Xu J, Mo C, Gong W, Li C, Hou X, Ou M. High-throughput sequencing reveals twelve cell death pattern prognostic target genes as potential drug-response-associated genes in the treatment of colorectal cancer cells with palmatine hydrochloride. ONCOLOGIE 2024. [DOI: 10.1515/oncologie-2024-0378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
Abstract
Objectives
Palmatine Hydrochloride (PaH), an isoquinoline alkaloid from Phellodendron amurense and Coptis chinensis, has analgesic, anti-inflammatory, and anticancer properties. This study aimed to assess PaH’s effectiveness against SW480 colorectal cancer (CRC) cells and explore its molecular mechanisms.
Methods
PaH’s effects on SW480 CRC cells were evaluated using MTT assays for proliferation, scratch assays for migration, and flow cytometry for apoptosis. Differentially expressed genes (DEGs) were identified through high-throughput sequencing. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses assessed DEG roles. Prognostic significance related to programmed cell death (PCD) was analyzed using R-Package with TCGA data. RT-qPCR validated key genes identified.
Results
PaH significantly inhibited SW480 cell growth, invasion, and apoptosis. The MTT assay showed inhibition rates increased from 5.49 % at 25 μg/mL to 52.48 % at 400 μg/mL. Scratch assays indicated reduced cell invasion over 24, 48, and 72 h. Apoptosis rose from 12.36 % in controls to 45.54 % at 400 μg/mL. Sequencing identified 3,385 significant DEGs, primarily in cancer pathways (p=0.004). Among 35 PCD-related DEGs, Lasso Cox regression highlighted 12 key genes, including TERT, TGFBR1, WNT4, and TP53. RT-qPCR confirmed TERT and TGFBR1 downregulation (0.614-fold, p=0.008; 0.41-fold, p<0.001) and TP53 and WNT4 upregulation (5.634-fold, p<0.001; 5.124-fold, p=0.002).
Conclusions
PaH inhibits CRC cell proliferation, migration, and invasion by modulating key PCD genes, suggesting its potential as a CRC therapeutic agent.
Collapse
Affiliation(s)
- Sha Luo
- Laboratory Center, Guangxi Health Commission Key Laboratory of Glucose and Lipid Metabolism Disorders , The Second Affiliated Hospital of Guilin Medical University , Guilin, 541199 , China
- Laboratory Center, Guangxi Key Laboratory of Metabolic Reprogramming and Intelligent Medical Engineering for Chronic Diseases , The Second Affiliated Hospital of Guilin Medical University , Guilin, 541199 , China
| | - Jiajun Xu
- Laboratory Center, Guangxi Health Commission Key Laboratory of Glucose and Lipid Metabolism Disorders , The Second Affiliated Hospital of Guilin Medical University , Guilin, 541199 , China
- Laboratory Center, Guangxi Key Laboratory of Metabolic Reprogramming and Intelligent Medical Engineering for Chronic Diseases , The Second Affiliated Hospital of Guilin Medical University , Guilin, 541199 , China
| | - Chune Mo
- Laboratory Center, Guangxi Health Commission Key Laboratory of Glucose and Lipid Metabolism Disorders , The Second Affiliated Hospital of Guilin Medical University , Guilin, 541199 , China
- Laboratory Center, Guangxi Key Laboratory of Metabolic Reprogramming and Intelligent Medical Engineering for Chronic Diseases , The Second Affiliated Hospital of Guilin Medical University , Guilin, 541199 , China
| | - Weiwei Gong
- Laboratory Center, Guangxi Health Commission Key Laboratory of Glucose and Lipid Metabolism Disorders , The Second Affiliated Hospital of Guilin Medical University , Guilin, 541199 , China
- Laboratory Center, Guangxi Key Laboratory of Metabolic Reprogramming and Intelligent Medical Engineering for Chronic Diseases , The Second Affiliated Hospital of Guilin Medical University , Guilin, 541199 , China
| | - Chunhong Li
- Laboratory Center, Guangxi Health Commission Key Laboratory of Glucose and Lipid Metabolism Disorders , The Second Affiliated Hospital of Guilin Medical University , Guilin, 541199 , China
- Laboratory Center, Guangxi Key Laboratory of Metabolic Reprogramming and Intelligent Medical Engineering for Chronic Diseases , The Second Affiliated Hospital of Guilin Medical University , Guilin, 541199 , China
| | - Xianliang Hou
- Laboratory Center, Guangxi Health Commission Key Laboratory of Glucose and Lipid Metabolism Disorders , The Second Affiliated Hospital of Guilin Medical University , Guilin, 541199 , China
- Laboratory Center, Guangxi Key Laboratory of Metabolic Reprogramming and Intelligent Medical Engineering for Chronic Diseases , The Second Affiliated Hospital of Guilin Medical University , Guilin, 541199 , China
| | - Minglin Ou
- Laboratory Center, Guangxi Health Commission Key Laboratory of Glucose and Lipid Metabolism Disorders , The Second Affiliated Hospital of Guilin Medical University , Guilin, 541199 , China
- Laboratory Center, Guangxi Key Laboratory of Metabolic Reprogramming and Intelligent Medical Engineering for Chronic Diseases , The Second Affiliated Hospital of Guilin Medical University , Guilin, 541199 , China
| |
Collapse
|
334
|
Zhou R, Bai G, Zhu D, Xu Q, Zhang X, Li T, Qian Y, Bu C. Pump-free SERS microfluidic chip based on an identification-competition strategy for ultrasensitive and efficient simultaneous detection of liver cancer-related microRNAs. BIOMEDICAL OPTICS EXPRESS 2024; 15:6469-6485. [PMID: 39553886 PMCID: PMC11563321 DOI: 10.1364/boe.542523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 10/10/2024] [Accepted: 10/11/2024] [Indexed: 11/19/2024]
Abstract
In this study, we present a pump-free SERS microfluidic chip capable of detecting liver cancer-related miR-21 and miR-155 concurrently with ultra-sensitivity and high efficiency. We employed a Fe3O4@cDNA-AuNPs@Raman reporter@H composite structure and a recognition competition strategy. When the target miRNAs (miR-21 and miR-155) are present in the test liquid, they specifically compete with the nucleic acid complementary strand(H) of Fe3O4@cDNA-AuNPs@Raman reporter@H, causing AuNPs to competitively detach from the surface of Fe3O4, resulting in a decrease in the SERS signal. Consequently, this pump-free SERS microfluidic chip enables the detection of the target miRNAs more rapidly and accurately in complex environments. This method offers an approach for the simultaneous and efficient detection of miRNAs and holds promising applications in the early diagnosis of liver cancer.
Collapse
Affiliation(s)
- Ruoyu Zhou
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, China
| | - Guangfu Bai
- Affiliated Huishan Hospital of Medical College, Yangzhou University, Wuxi Huishan District People's Hospital, Wuxi 214187, Jiangsu Province, China
| | - Dongxu Zhu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, China
| | - Qiong Xu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, China
| | - Xudong Zhang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, China
| | - Tianran Li
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, China
| | - Yayun Qian
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, China
| | - Chiwen Bu
- Department of General Surgery, Guanyun County People's Hospital, Lianyungang 222200, China
| |
Collapse
|
335
|
Peng Y, Liu D, Huang D, Inuzuka H, Liu J. PROTAC as a novel anti-cancer strategy by targeting aging-related signaling. Semin Cancer Biol 2024; 106-107:143-155. [PMID: 39368654 DOI: 10.1016/j.semcancer.2024.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/18/2024] [Accepted: 09/20/2024] [Indexed: 10/07/2024]
Abstract
Aging and cancer share common cellular hallmarks, including cellular senescence, genomic instability, and abnormal cell death and proliferation, highlighting potential areas for therapeutic interventions. Recent advancements in targeted protein degradation technologies, notably Proteolysis-Targeting Chimeras (PROTACs), offer a promising approach to address these shared pathways. PROTACs leverage the ubiquitin-proteasome system to specifically degrade pathogenic proteins involved in cancer and aging, thus offering potential solutions to key oncogenic drivers and aging-related cellular dysfunction. This abstract summarizes the recent progress of PROTACs in targeting critical proteins implicated in both cancer progression and aging, and explores future perspectives in integrating these technologies for more effective cancer treatments.
Collapse
Affiliation(s)
- Yunhua Peng
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China; Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Donghua Liu
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China; Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an 710061, China
| | - Daoyuan Huang
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States
| | - Hiroyuki Inuzuka
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States.
| | - Jing Liu
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China; Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an 710061, China.
| |
Collapse
|
336
|
Lu J, Liu X, Cen A, Hong Y, Wang Y. HYPOXIA induces lncRNA HOTAIR for recruiting RELA in papillary thyroid cancer cells to upregulate miR-181a and promote angiogenesis. J Endocrinol Invest 2024; 47:2873-2884. [PMID: 38748197 DOI: 10.1007/s40618-024-02388-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 05/02/2024] [Indexed: 10/15/2024]
Abstract
BACKGROUND Papillary thyroid carcinoma (PTC) is one of the most common subtypes of thyroid carcinoma. Exosomal miR-181a plays an important role in the development of PTC. This study examined the regulatory mechanism of miR-181a under conditions of hypoxia and its impact on angiogenesis. METHODS A ribonucleoprotein immunoprecipitation (RIP) experiment was conducted to verify the interaction between HOTAIR and RELA. The relationship between RELA and the miR-181a promoter was detected by ChIP-qPCR. Short hairpin (sh) RNA was designed to knock down HOTAIR in TPC cells. The underlying mechanism of miR-181a was verified by use of dual-luciferase assays and rescue experiments. The regulatory effect of GATA6 on angiogenesis was studied using CCK8, EdU, Transwell, and western blot assays. RESULTS A RIP assay showed that HOTAIR could bind to RELA under hypoxic conditions. ChIP-qPCR and dual luciferase assays showed RELA could interact with the miR181a promoter and upregulate miR-181a. Knockdown of HOTAIR downregulated miR-181a in TPC-1 cells, and the downregulation could be rescued by RELA overexpression. MiR-181a downregulated GATA6 in HUVEC cells. Overexpression of GATA6 inhibited HUVEC proliferation, migration, tube formation, and EGFR expression. Exosomal miR-181a promoted angiogenesis by downregulating GATA6 expression. CONCLUSION HOTAIR activated RELA to upregulate miR-181a during hypoxia. Exosomal miR-181a promotes tumor angiogenesis by downregulating GATA6.
Collapse
MESH Headings
- Humans
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Thyroid Cancer, Papillary/genetics
- Thyroid Cancer, Papillary/pathology
- Thyroid Cancer, Papillary/metabolism
- Transcription Factor RelA/metabolism
- Transcription Factor RelA/genetics
- Neovascularization, Pathologic/genetics
- Neovascularization, Pathologic/metabolism
- Neovascularization, Pathologic/pathology
- Thyroid Neoplasms/genetics
- Thyroid Neoplasms/pathology
- Thyroid Neoplasms/metabolism
- Gene Expression Regulation, Neoplastic
- Cell Proliferation
- GATA6 Transcription Factor/genetics
- GATA6 Transcription Factor/metabolism
- Up-Regulation
- Cell Movement/genetics
- Cell Line, Tumor
- Hypoxia/metabolism
- Hypoxia/genetics
- Angiogenesis
Collapse
Affiliation(s)
- J Lu
- Department of Endocrinology, the First Affiliated Hospital of Jinan University, 613 W. Huangpu Avenue, Guangzhou, China
| | - X Liu
- Department of Endocrinology, the First Affiliated Hospital of Jinan University, 613 W. Huangpu Avenue, Guangzhou, China
| | - A Cen
- Department of Endocrinology, the People's Hospital of Jiangmen, Jiangmen, Guangdong, China
| | - Y Hong
- Department of Endocrinology, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, the Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Y Wang
- Department of Endocrinology, the First Affiliated Hospital of Jinan University, 613 W. Huangpu Avenue, Guangzhou, China.
| |
Collapse
|
337
|
Zeng Q, Zeng S, Dai X, Ding Y, Huang C, Ruan R, Xiong J, Tang X, Deng J. MDM2 inhibitors in cancer immunotherapy: Current status and perspective. Genes Dis 2024; 11:101279. [PMID: 39263534 PMCID: PMC11388719 DOI: 10.1016/j.gendis.2024.101279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 02/13/2024] [Accepted: 02/21/2024] [Indexed: 09/13/2024] Open
Abstract
Murine double minute 2 (MDM2) plays an essential role in the cell cycle, apoptosis, DNA repair, and oncogene activation through p53-dependent and p53-independent signaling pathways. Several preclinical studies have shown that MDM2 is involved in tumor immune evasion. Therefore, MDM2-based regulation of tumor cell-intrinsic immunoregulation and the immune microenvironment has attracted increasing research attention. In recent years, immune checkpoint inhibitors targeting PD-1/PD-L1 have been widely used in the clinic. However, the effectiveness of a single agent is only approximately 20%-40%, which may be related to primary and secondary drug resistance caused by the dysregulation of oncoproteins. Here, we reviewed the role of MDM2 in regulating the immune microenvironment, tumor immune evasion, and hyperprogression during immunotherapy. In addition, we summarized preclinical and clinical findings on the use of MDM2 inhibitors in combination with immunotherapy in tumors with MDM2 overexpression or amplification. The results reveal that the inhibition of MDM2 could be a promising strategy for enhancing immunotherapy.
Collapse
Affiliation(s)
- Qinru Zeng
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, Nanchang, Jiangxi 330006, China
| | - Shaocheng Zeng
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, Nanchang, Jiangxi 330006, China
| | - Xiaofeng Dai
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, Nanchang, Jiangxi 330006, China
| | - Yun Ding
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, Nanchang, Jiangxi 330006, China
| | - Chunye Huang
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, Nanchang, Jiangxi 330006, China
| | - Ruiwen Ruan
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, Nanchang, Jiangxi 330006, China
| | - Jianping Xiong
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, Nanchang, Jiangxi 330006, China
| | - Xiaomei Tang
- Department of Oncology, Jiangxi Chest Hospital, Nanchang, Jiangxi 330006, China
| | - Jun Deng
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, Nanchang, Jiangxi 330006, China
- Postdoctoral Innovation Practice Base, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| |
Collapse
|
338
|
Wu SY, Liao EC, Wen YF, Wang YS, Meng H, Chou HC, Chan HL. Exploring the effects of pemetrexed on drug resistance mechanisms in human lung adenocarcinoma and its association with PGRMC1. Chem Biol Interact 2024; 403:111259. [PMID: 39368770 DOI: 10.1016/j.cbi.2024.111259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 09/01/2024] [Accepted: 09/30/2024] [Indexed: 10/07/2024]
Abstract
According to the 2022 cancer statistics of the World Health Organization, lung cancer ranks among the top ten causes of death, with lung adenocarcinoma being the most prevalent type. Despite significant advancements in lung cancer therapeutics, many clinical limitations remain, primarily due to the development of drug resistance. The present study investigated the effects of pemetrexed on the drug resistance mechanisms in human lung adenocarcinoma and its association with progesterone receptor membrane component 1 (PGRMC1) expression. Given that KRAS-mutant lung adenocarcinoma cell lines (e.g., A549) exhibit a high folate synthesis activity, pemetrexed, which is structurally similar to folate, was selected as the therapeutic drug. The present study used a lung adenocarcinoma cell line (A549) and established a drug-resistant lung adenocarcinoma cell line (A549/PEM). The findings demonstrated that PGRMC1 expression was elevated in the A549/PEM cells. It has been hypothesized that PGRMC1 regulates iron absorption through heme binding, resulting in a preference for iron-related cell death pathways (ferroptosis). Our findings indicate that drug-resistant lung adenocarcinoma cells with high PGRMC1 levels exhibit elevated antioxidant activity on the cell membrane and increased reliance on iron-dependent cell death pathways. This suggests a correlation between PGRMC1 and pemetrexed-induced iron-dependent cell death. Our study contributes to the development of more effective therapeutic strategies to improve the prognosis of patients with lung adenocarcinoma, particularly those facing drug resistance challenges.
Collapse
Affiliation(s)
- Ssu-Yun Wu
- Institute of Bioinformatics and Structural Biology, National TsiFng Hua University, Hsinchu, Taiwan
| | - En-Chi Liao
- Institute of Bioinformatics and Structural Biology, National TsiFng Hua University, Hsinchu, Taiwan
| | - Yueh-Feng Wen
- Institute of Bioinformatics and Structural Biology, National TsiFng Hua University, Hsinchu, Taiwan; Department of Internal Medicine, National Taiwan University Hospital, Hsinchu Branch, Hsinchu, Taiwan
| | - Yi-Shiuan Wang
- Institute of Bioinformatics and Structural Biology, National TsiFng Hua University, Hsinchu, Taiwan
| | - Han Meng
- Institute of Bioinformatics and Structural Biology, National TsiFng Hua University, Hsinchu, Taiwan
| | - Hsiu-Chuan Chou
- Institute of Analytical and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Hong-Lin Chan
- Institute of Bioinformatics and Structural Biology, National TsiFng Hua University, Hsinchu, Taiwan; Department of Medical Sciences, National Tsing Hua University, Hsinchu, Taiwan.
| |
Collapse
|
339
|
Gao Q, Chen JM, Li CSZ, Zhan JY, Yin XD, Li BS, Dong HL, Luo LX, Li ZL. CDKN1A promotes Cis-induced AKI by inducing cytoplasmic ROS production and ferroptosis. Food Chem Toxicol 2024; 193:115003. [PMID: 39353481 DOI: 10.1016/j.fct.2024.115003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 08/28/2024] [Accepted: 09/12/2024] [Indexed: 10/04/2024]
Abstract
BACKGROUND AND OBJECTIVE This study focuses on investigating the role of CDKN1A in cisplatin-induced AKI (acute kidney injury, AKI) and its potential as a biomarker for early diagnosis and therapeutic intervention by integrating bioinformatics analysis, machine learning, and experimental validation. METHODS We analyzed the GSE85957 dataset to find genes that changed between control and cisplatin-treated rats. Using bioinformatics and machine learning, we found 13 important genes related to ferroptosis and the P53 pathway. The key gene, CDKN1A, was identified using various algorithms. We then tested how reducing CDKN1A in human kidney cells affected cell health, ROS, and iron levels. We also checked how CDKN1A changes the levels of proteins linked to ferroptosis using Q-PCR and Western Blot. RESULTS CDKN1A was found to negatively regulate the G1/S phase transition and was associated with ferroptosis in p53 signaling. Experiments in human renal tubular epithelial cells (HK-2) and rat NRK-52E cells showed that CDKN1A knockdown mitigated cisplatin-induced cell injury by reducing oxidative stress and ferroptosis. CONCLUSION Our integrated approach identified CDKN1A as a biomarker for cisplatin-induced AKI. Its regulation could be key in AKI pathogenesis, offering new therapeutic insights and aiding in early diagnosis and intervention.
Collapse
Affiliation(s)
- Qian Gao
- Department of Pharmacy, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China; School of Medicine, Shanghai Jiao Tong University, Shanghai, 200125, China
| | - Jun-Ming Chen
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, 524023, Guangdong, China
| | - Chen-Sui-Zi Li
- Department of Pharmacy, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China; School of Medicine, Shanghai Jiao Tong University, Shanghai, 200125, China
| | - Jia-Yi Zhan
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, 524023, Guangdong, China
| | - Xue-Dong Yin
- Department of Pharmacy, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Ben-Shang Li
- Department of Hematology/Oncology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai, China
| | - Hong-Liang Dong
- Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200120, China.
| | - Lian-Xiang Luo
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, 524023, Guangdong, China.
| | - Zhi-Ling Li
- Department of Pharmacy, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
| |
Collapse
|
340
|
Gao Y, Li J, Ma M, Fu W, Ma L, Sui Y, Wang Y. Prognostic prediction of m6A and ferroptosis-associated lncRNAs in liver hepatocellular carcinoma. J Transl Int Med 2024; 12:526-529. [PMID: 39513037 PMCID: PMC11538885 DOI: 10.1515/jtim-2024-0023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2024] Open
Affiliation(s)
- Yuchen Gao
- Epidemiology and Health Statistics, Shenyang Medical College, Shenyang110034, Liaoning Province, China
| | - Jingxiao Li
- Epidemiology and Health Statistics, Shenyang Medical College, Shenyang110034, Liaoning Province, China
| | - Mingyue Ma
- Department of Toxicology, School of Public Heath, Shenyang Medical College, Shenyang110034, Liaoning Province, China
| | - Wenting Fu
- Integrated Business Department, Shenyang Center for Disease Control and Preventior, Shenyang110034, Liaoning Province, China
| | - Lin Ma
- Chengdu Kegene Biotechnology Co., Ltd, Chengdu610072, Sichuan Province, China
| | - Yi Sui
- Department of Neurology and Neurosurgery, Shenyang First People’s Hospital, Shenyang Medical College Affiliated Brain Hospital, Shenyang110041, Liaoning Province, China
| | - Yu Wang
- School of Public Heath, Shenyang Medical College, Shenyang110034, Liaoning Province, China
| |
Collapse
|
341
|
Wang Y, Wu Z, Wang C, Wu N, Wang C, Hu S, Shi J. The role of WWP1 and WWP2 in bone/cartilage development and diseases. Mol Cell Biochem 2024; 479:2907-2919. [PMID: 38252355 DOI: 10.1007/s11010-023-04917-7] [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: 08/18/2023] [Accepted: 12/11/2023] [Indexed: 01/23/2024]
Abstract
Bone and cartilage diseases are often associated with trauma and senescence, manifested as pain and limited mobility. The repair of bone and cartilage lesion by mesenchymal stem cells is regulated by various transcription factors. WW domain-containing protein 1 (WWP1) and WW domain-containing protein 2 (WWP2) are named for WW domain which recognizes PPXY (phono Ser Pro and Pro Arg) motifs of substrate. WWP1and WWP2 are prominent components of the homologous to the E6-AP carboxyl terminus (HECT) subfamily, a group of the ubiquitin ligase. Recently, some studies have found that WWP1 and WWP2 play an important role in the pathogenesis of bone and cartilage diseases and regulate the level and the transactivation of various transcription factors through ubiquitination. Therefore, this review summarizes the distribution and effects of WWP1 and WWP2 in the development of bone and cartilage, discusses the potential mechanism and therapeutic drugs in bone and cartilage diseases such as osteoarthritis, fracture, and osteoporosis.
Collapse
Affiliation(s)
- Ying Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310016, China
| | - Zuping Wu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310016, China
| | - Cunyi Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310016, China
| | - Na Wu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310016, China
| | - Chenyu Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310016, China
| | - Shiyu Hu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310016, China
| | - Jiejun Shi
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310016, China.
| |
Collapse
|
342
|
Xin Z, Hu C, Zhang C, Liu M, Li J, Sun X, Hu Y, Liu X, Wang K. LncRNA-HMG incites colorectal cancer cells to chemoresistance via repressing p53-mediated ferroptosis. Redox Biol 2024; 77:103362. [PMID: 39307047 PMCID: PMC11447409 DOI: 10.1016/j.redox.2024.103362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 09/11/2024] [Accepted: 09/17/2024] [Indexed: 10/06/2024] Open
Abstract
Upon chemotherapy, excessive reactive oxygen species (ROS) often lead to the production of massive lipid peroxides in cancer cells and induce cell death, namely ferroptosis. The elimination of ROS is pivotal for tumor cells to escape from ferroptosis and acquire drug resistance. Nevertheless, the precise functions of long non-coding RNAs (lncRNAs) in ROS metabolism and tumor drug-resistance remain elusive. In this study, we identify LncRNA-HMG as a chemoresistance-related lncRNA in colorectal cancer (CRC) by high-throughput screening. Abnormally high expression of LncRNA-HMG predicts poorer prognosis in CRC patients. Concurrently, we found that LncRNA-HMG protects CRC cells from ferroptosis upon chemotherapy, thus enhancing drug resistance of CRC cells. LncRNA-HMG binds to p53 and facilitates MDM2-mediated degradation of p53. Decreased p53 induces upregulation of SLC7A11 and VKORC1L1, which contribute to increase the supply of reducing agents and eliminate excessive ROS. Consequently, CRC cells escape from ferroptosis and acquire chemoresistance. Importantly, inhibition of LncRNA-HMG by anti-sense oligo (ASO) dramatically sensitizes CRC cells to chemotherapy in patient-derived xenograft (PDX) model. LncRNA-HMG is also a transcriptional target of β-catenin/TCF and activated Wnt signals trigger the marked upregulation of LncRNA-HMG. Collectively, these findings demonstrate that LncRNA-HMG promotes CRC chemoresistance and might be a prognostic or therapeutic target for CRC.
Collapse
Affiliation(s)
- Zechang Xin
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Hepatopancreatobiliary Surgery Department I, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Chenyu Hu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Hepatopancreatobiliary Surgery Department I, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Chunfeng Zhang
- Department of Medical Genetics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Ming Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Hepatopancreatobiliary Surgery Department I, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Juan Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Hepatopancreatobiliary Surgery Department I, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Xiaoyan Sun
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Hepatopancreatobiliary Surgery Department I, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Yang Hu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Hepatopancreatobiliary Surgery Department I, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Xiaofeng Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Hepatopancreatobiliary Surgery Department I, Peking University Cancer Hospital & Institute, Beijing, 100142, China.
| | - Kun Wang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Hepatopancreatobiliary Surgery Department I, Peking University Cancer Hospital & Institute, Beijing, 100142, China.
| |
Collapse
|
343
|
Wang M, Wang X, Wang Y, Gai Y, Ye J, Xu X, You X. Advances in the study of the mechanism of action of miR‑22 in liver lesions (Review). Oncol Lett 2024; 28:541. [PMID: 39310022 PMCID: PMC11413475 DOI: 10.3892/ol.2024.14674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 08/15/2024] [Indexed: 09/25/2024] Open
Abstract
Globally, nearly 2 million deaths annually are attributed to the development of liver diseases, with liver cancer and cirrhosis being particularly prominent, which makes liver disease a significant global health concern. Cirrhosis is closely linked to the evolution of hepatitis, hepatic fibrosis and fatty liver. However, most liver diseases have an insidious onset, are challenging to treat and the prognosis and efficacy of current therapies are unsatisfactory, which can result in irreversible functional damage to the liver. Therefore, there is an urgent need to explore the molecular mechanisms underlying liver disease and identify new biomarkers and therapeutic targets. In previous years, microRNAs (miRs), a class of short non-coding RNAs comprising 17-25 nucleotides, have attracted attention for their roles in various types of liver diseases. Among them, miR-22 serves a unique role in mediating multiple pathway mechanisms and epigenetic modifications and can act both as an inhibitor of liver cancer and a metabolic blocker. Given its close association with the liver, several studies have reported that the differential expression of miR-22 regulates the metabolic process of liver cancer and is involved in the evolution of hepatic fibrosis and steatohepatitis, making it a potential target for early diagnosis and treatment. The present manuscript aimed to comprehensively review the key role of miR-22 in the evolution of liver diseases and offer valuable references and guidance for subsequent studies by identifying its specific mechanism of action and future development prospects.
Collapse
Affiliation(s)
- Minghe Wang
- College of Second Clinical Medical, Jining Medical University, Jining, Shandong 272067, P.R. China
| | - Xuejing Wang
- College of Second Clinical Medical, Jining Medical University, Jining, Shandong 272067, P.R. China
| | - Yanqi Wang
- College of Clinical Medical, Jining Medical University, Jining, Shandong 272067, P.R. China
| | - Yikuo Gai
- College of Second Clinical Medical, Jining Medical University, Jining, Shandong 272067, P.R. China
| | - Jingran Ye
- College of Second Clinical Medical, Jining Medical University, Jining, Shandong 272067, P.R. China
| | - Xinyan Xu
- College of Second Clinical Medical, Jining Medical University, Jining, Shandong 272067, P.R. China
| | - Xue You
- Lin He's Academician Workstation of New Medicine and Clinical Translation, Jining Medical University, Jining, Shandong 272067, P.R. China
| |
Collapse
|
344
|
Zhu H, Xu S. SOX4 inhibits ferroptosis and promotes proliferation of endometrial cancer cells via the p53/SLC7A11 signaling. J Obstet Gynaecol Res 2024; 50:2093-2106. [PMID: 39318043 DOI: 10.1111/jog.16094] [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: 05/24/2024] [Accepted: 09/07/2024] [Indexed: 09/26/2024]
Abstract
AIM Sex-determining region Y-related high-mobility group box 4 (SOX4) has been reported to play a carcinogenic role in endometrial cancer (EC). However, the biological function and regulatory mechanisms of SOX4 in ferroptosis during the progression of EC are still unknown. METHODS The mRNA and protein levels were scrutinized by quantitative reverse-transcription polymerase chain reaction and western blot, respectively. The cell viability and proliferative capability were determined by cell counting kit-8 assay and 5-ethynyl-2'-deoxyuridine (EdU) assay. Transcriptional regulation of gene expression was investigated by dual-luciferase reporter assay and chromatin immunoprecipitation. Ferroptosis was evaluated by detection of reactive oxygen species, malondialdehyde, Fe2+, and ferroptosis-related proteins. The mice test was implemented to confirm the influence of SOX4 on EC tumor growth and ferroptosis in vivo. RESULTS We here discovered the elevation of SOX4 in EC tissues and cells. Functionally, SOX4 knockdown hampered proliferation and promoted ferroptosis of EC cells. Mechanistically, SOX4 bound to p53 promoter and inhibited its transcriptional activity in EC cells. In addition, p53 transcriptionally suppressed SLC7A11 expression in EC cells. Downregulation of p53 reverses the effect of SOX4 knockdown on proliferation and ferroptosis of EC cells. Finally, in vivo experiments demonstrated that SOX4 depletion hindered tumor growth and triggered ferroptosis in EC. CONCLUSIONS These findings collectively suggested that SOX4 inhibited ferroptosis and promoted proliferation of EC cells via the p53/SLC7A11 signaling. Our research unveiled a novel regulatory mechanism of ferroptosis in EC, offering promising perspectives for the development of EC therapies.
Collapse
Affiliation(s)
- Hongli Zhu
- Department of Obstetrics and Gynecology, Affliated Hangzhou First People's Hospital of Zhejiang University, Hangzhou, Zhejiang, China
| | - Song Xu
- Department of Obstetrics and Gynecology, Affliated Hangzhou First People's Hospital of Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
345
|
Cui X, Zheng Z, Rahman MU, Hong X, Ji X, Li Z, Chen HF. Drude2019IDPC polarizable force field reveals structure-function relationship of insulin. Int J Biol Macromol 2024; 280:136256. [PMID: 39366599 DOI: 10.1016/j.ijbiomac.2024.136256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 09/30/2024] [Accepted: 10/01/2024] [Indexed: 10/06/2024]
Abstract
Intrinsically disordered proteins (IDPs) lack stable tertiary structures under physiological conditions, yet play key roles in biological processes and associated with human complex diseases. Their conformational characteristics and high content of charged residues make the use of polarizable force fields an advantageous for simulating IDPs. The Drude2019IDP polarizable force field, previously introduced, has demonstrated comprehensive enhancements and improvements in dipeptides, short peptides, and IDPs, achieving a balanced sampling between IDPs and structured proteins. However, the performance in simulating 5 dipeptides was found to be underestimate. Therefore, we individually performed reweighting and grid-based energy correction map (CMAP) optimization for these 5 dipeptides, resulting in the enhanced Drude2019IDPC force field. The performance of Drude2019IDPC was evaluated with 5 dipeptides, 5 disordered short peptides, and a representative IDP. The results demonstrated a marked improvement comparing with original Drude2019IDP. To further substantiate the capabilities of Drude2019IDPC, MD simulation and Markov state model (MSM) were applied to wild type and mutant for insulin, to elucidate the difference of conformational characteristics and transition path. The findings reveal that mutation can maintain the monomorphic characteristics, providing insights for engineered insulin development. These results indicate that Drude2019IDPC could be used to reveal the structure-function relationship for other proteins.
Collapse
Affiliation(s)
- Xiaochen Cui
- State Key Laboratory of Microbial metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, Department of Bioinformatics and Biostatistics, National Experimental Teaching Center for Life Sciences and Biotechnology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Zhuoqi Zheng
- State Key Laboratory of Microbial metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, Department of Bioinformatics and Biostatistics, National Experimental Teaching Center for Life Sciences and Biotechnology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Mueed Ur Rahman
- State Key Laboratory of Microbial metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, Department of Bioinformatics and Biostatistics, National Experimental Teaching Center for Life Sciences and Biotechnology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xiaokun Hong
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian 350116, China
| | - Xiaoyue Ji
- State Key Laboratory of Microbial metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, Department of Bioinformatics and Biostatistics, National Experimental Teaching Center for Life Sciences and Biotechnology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Zhengxin Li
- State Key Laboratory of Microbial metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, Department of Bioinformatics and Biostatistics, National Experimental Teaching Center for Life Sciences and Biotechnology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Hai-Feng Chen
- State Key Laboratory of Microbial metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, Department of Bioinformatics and Biostatistics, National Experimental Teaching Center for Life Sciences and Biotechnology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China.
| |
Collapse
|
346
|
Xu H, Wang T, Nie H, Sun Q, Jin C, Yang S, Chen Z, Wang X, Tang J, Feng Y, Sun Y. USP36 promotes colorectal cancer progression through inhibition of p53 signaling pathway via stabilizing RBM28. Oncogene 2024; 43:3442-3455. [PMID: 39343961 PMCID: PMC11573713 DOI: 10.1038/s41388-024-03178-y] [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: 05/30/2024] [Revised: 09/19/2024] [Accepted: 09/23/2024] [Indexed: 10/01/2024]
Abstract
Colorectal cancer (CRC) stands as the second most common cause of cancer-related mortality globally and p53, a widely recognized tumor suppressor, contributes to the development of CRC. Ubiquitin-specific protease 36 (USP36), belonging to the deubiquitinating enzyme family, is involved in tumor progression across multiple cancers. However, the underlying molecular mechanism in which USP36 regulates p53 signaling pathway in CRC is unclear. Here, our study revealed that USP36 was increased in CRC tissues and associated with unfavorable prognosis. Functionally, elevated USP36 could promote proliferation, migration, and invasion of CRC cells in vitro and in vivo. Mechanistically, USP36 could interact with and stabilize RBM28 via deubiquitination at K162 residue. Further, upregulated RBM28 could bind with p53 to suppress its transcriptional activity and therefore inactivate p53 signaling pathway. Collectively, our investigation identified the novel USP36/RBM28/p53 axis and its involvement in promoting cell proliferation and metastasis in CRC, which presents a promising therapeutic strategy for CRC treatment.
Collapse
Affiliation(s)
- Hengjie Xu
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China
- The Colorectal Institute of Nanjing Medical University, Nanjing, China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational Medicine, Nanjing, China
| | - Tuo Wang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China
- The Colorectal Institute of Nanjing Medical University, Nanjing, China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational Medicine, Nanjing, China
| | - Hongxu Nie
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China
- The Colorectal Institute of Nanjing Medical University, Nanjing, China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational Medicine, Nanjing, China
| | - Qingyang Sun
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China
- The Colorectal Institute of Nanjing Medical University, Nanjing, China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational Medicine, Nanjing, China
| | - Chi Jin
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China
- The Colorectal Institute of Nanjing Medical University, Nanjing, China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational Medicine, Nanjing, China
| | - Sheng Yang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China
- The Colorectal Institute of Nanjing Medical University, Nanjing, China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational Medicine, Nanjing, China
| | - Zhihao Chen
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China
- The Colorectal Institute of Nanjing Medical University, Nanjing, China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational Medicine, Nanjing, China
| | - Xiaowei Wang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China
- The Colorectal Institute of Nanjing Medical University, Nanjing, China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational Medicine, Nanjing, China
| | - Junwei Tang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China.
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China.
- The Colorectal Institute of Nanjing Medical University, Nanjing, China.
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational Medicine, Nanjing, China.
| | - Yifei Feng
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China.
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China.
- The Colorectal Institute of Nanjing Medical University, Nanjing, China.
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational Medicine, Nanjing, China.
| | - Yueming Sun
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China.
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China.
- The Colorectal Institute of Nanjing Medical University, Nanjing, China.
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational Medicine, Nanjing, China.
| |
Collapse
|
347
|
Zhang Y, Liu Y, Xing X, Liu H, Guan W. Genome-Wide Analysis of p53 Targets Reveals SCN2A as a Novel Player in p53-Induced Cell Arrest in HPV-Positive Cells. Viruses 2024; 16:1725. [PMID: 39599840 PMCID: PMC11598893 DOI: 10.3390/v16111725] [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: 10/07/2024] [Revised: 10/29/2024] [Accepted: 10/29/2024] [Indexed: 11/29/2024] Open
Abstract
The host transcription factor p53 is a critical tumor suppressor in HPV-induced carcinogenesis, regulating target genes involved in cell cycle arrest and apoptosis. However, the p53 targets have not been thoroughly analyzed in HPV-infected cells. In this study, p53 signaling in HPV16 and HPV18 cells was activated by depleting the viral oncoprotein E6. Subsequently, p53-regulated genes were identified by comparing them with genes altered in p53-silenced cells. True p53 targets were defined as genes with at least one overlapping p53 binding site and ChIP peak near their locus. Our analysis revealed that while some p53 targets were common to both the HPV16 and HPV18 cells, the majority of the targets differed between these two types, potentially contributing to the varying prevalence of HPV16 and HPV18 in cervical cancer. Additionally, we identified SCN2A as a novel p53 target involved in p53-induced cell cycle arrest in HPV-related carcinogenesis. This study provides new insights into the mechanisms by which p53 inhibits HPV-induced carcinogenesis.
Collapse
Affiliation(s)
- Yudi Zhang
- Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430207, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yi Liu
- Hubei Jiangxia Laboratory, Wuhan 430200, China; (Y.L.)
| | - Xueyan Xing
- Hubei Jiangxia Laboratory, Wuhan 430200, China; (Y.L.)
| | - Haibin Liu
- Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430207, China
- Hubei Jiangxia Laboratory, Wuhan 430200, China; (Y.L.)
| | - Wuxiang Guan
- Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430207, China
- Hubei Jiangxia Laboratory, Wuhan 430200, China; (Y.L.)
| |
Collapse
|
348
|
Cheng H, Wang X, Yao J, Yang C, Liu J. Mitophagy and Ferroptosis in Sepsis-Induced ALI/ARDS: Molecular Mechanisms, Interactions and Therapeutic Prospects of Medicinal Plants. J Inflamm Res 2024; 17:7819-7835. [PMID: 39494205 PMCID: PMC11531397 DOI: 10.2147/jir.s488655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 10/17/2024] [Indexed: 11/05/2024] Open
Abstract
Sepsis is a common critical illness characterized by high mortality rates and a significant disease burden. In the context of sepsis-induced organ dysfunction, the lungs are among the initial organs affected, which may progress to acute lung injury (ALI) and acute respiratory distress syndrome (ARDS). Recent studies have highlighted the crucial roles of mitophagy and ferroptosis in the development and progression of sepsis-induced ALI/ARDS. Identifying key convergence points in these processes may provide valuable insights for the treatment of this condition. In recent years, certain herbs and their bioactive compounds have demonstrated unique benefits in managing sepsis-induced ALI/ARDS by modulating mitophagy or ferroptosis. This review summary the mechanisms of mitophagy and ferroptosis, explores their interactions, and emphasizes their regulatory roles in the progression of sepsis-induced ALI/ARDS. Additionally, it offers a novel perspective on treatment strategies by summarizing various herbs and their bioactive compounds relevant to this condition.
Collapse
Affiliation(s)
- Huixin Cheng
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu Province, People’s Republic of China
| | - Xuehan Wang
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu Province, People’s Republic of China
| | - Juyi Yao
- Traditional Chinese Medicine Hospital of Xinjiang Uygur Autonomous Region, Urumqi, People’s Republic of China
| | - Chunbo Yang
- Department of Critical Medicine Center, First Affiliated Hospital of Xinjiang Medical University, Urumqi, People’s Republic of China
| | - Jian Liu
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu Province, People’s Republic of China
- Department of Intensive Care Unit, Gansu Provincial Maternity and Child Health Hospital/Gansu Provincial General Hospital, Lan Zhou, Gansu Province, People’s Republic of China
| |
Collapse
|
349
|
Gozdz A, Maksym RB, Ścieżyńska A, Götte M, Kieda C, Włodarski PK, Malejczyk J. Expression of Reversion-Inducing Cysteine-Rich Protein with Kazal Motifs ( RECK) Gene and Its Regulation by miR200b in Ovarian Endometriosis. Int J Mol Sci 2024; 25:11594. [PMID: 39519143 PMCID: PMC11547164 DOI: 10.3390/ijms252111594] [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: 09/04/2024] [Revised: 10/22/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024] Open
Abstract
Endometriosis is a common chronic disorder characterized by the growth of endometrium-like tissue outside the uterine cavity. The disease is associated with chronic inflammation and pelvic pain and may have an impact on the patient's fertility. The causative factors and pathophysiology of the disease are still poorly recognized. The dysregulation of the immune system, aberrant tissue remodeling, and angiogenesis contribute to the disease progression. In endometriosis patients, the proteins regulating the breakdown and reorganization of the connective tissue, e.g., collagenases, and other proteases, as well as their inhibitors, show an incorrect pattern of expression. Here, we report that the expression of reversion-inducing cysteine-rich protein with Kazal motifs (RECK), one of the inhibitors of connective tissue proteases, is elevated in endometrioma cysts as compared to normal endometrium from unaffected women. We also demonstrate a reduced level of miR200b in endometriotic tissue that correlates with RECK mRNA levels. Furthermore, we employ the 12Z cell line, derived from a peritoneal endometriotic lesion, and the Ishikawa cell line, originating from endometrial adenocarcinoma to identify RECK as a direct target of miR200b. The described effect of miR200b on RECK, together with the aberrant expression of both genes in endometrioma, may help to understand the role played by the tissue remodeling system in the pathogenesis of endometriosis.
Collapse
Affiliation(s)
- Agata Gozdz
- Department of Histology and Embryology, Center of Biostructure Research, Medical University of Warsaw, ul. T. Chałubińskiego 5, 02-004 Warsaw, Poland; (R.B.M.); (A.Ś.); (P.K.W.)
| | - Radosław B. Maksym
- Department of Histology and Embryology, Center of Biostructure Research, Medical University of Warsaw, ul. T. Chałubińskiego 5, 02-004 Warsaw, Poland; (R.B.M.); (A.Ś.); (P.K.W.)
- 1st Department of Obstetrics and Gynecology, Centre for Postgraduate Medical Education, ul. Żelazna 90, 01-004 Warsaw, Poland
- Center for Molecular Biophysics UPR 4301 CNRS, 45071 Orleans, France;
| | - Aneta Ścieżyńska
- Department of Histology and Embryology, Center of Biostructure Research, Medical University of Warsaw, ul. T. Chałubińskiego 5, 02-004 Warsaw, Poland; (R.B.M.); (A.Ś.); (P.K.W.)
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine-National Research Institute, 04-141 Warsaw, Poland
| | - Martin Götte
- Department of Obstetrics and Gynecology, University Hospital Münster, Albert-Schweitzer-Campus 1, 48149 Münster, Germany;
- Cells-in-Motion Interfaculty Centre (CiMIC), University of Münster, 48149 Münster, Germany
| | - Claudine Kieda
- Center for Molecular Biophysics UPR 4301 CNRS, 45071 Orleans, France;
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine-National Research Institute, 04-141 Warsaw, Poland
| | - Paweł K. Włodarski
- Department of Histology and Embryology, Center of Biostructure Research, Medical University of Warsaw, ul. T. Chałubińskiego 5, 02-004 Warsaw, Poland; (R.B.M.); (A.Ś.); (P.K.W.)
| | - Jacek Malejczyk
- Department of Histology and Embryology, Center of Biostructure Research, Medical University of Warsaw, ul. T. Chałubińskiego 5, 02-004 Warsaw, Poland; (R.B.M.); (A.Ś.); (P.K.W.)
| |
Collapse
|
350
|
Pasculli E, Gadaleta RM, Arconzo M, Cariello M, Moschetta A. The Role of Exogenous microRNAs on Human Health: The Plant-Human Trans-Kingdom Hypothesis. Nutrients 2024; 16:3658. [PMID: 39519491 PMCID: PMC11547593 DOI: 10.3390/nu16213658] [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: 10/10/2024] [Revised: 10/25/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
MicroRNAs (miRNAs) are small, endogenous, single-stranded RNAs that act on gene silencing at the post-transcriptional level by binding to a target messenger RNA (mRNA), leading to its degradation or inhibiting translation into functional proteins. The key role of miRNAs in development, proliferation, differentiation andapoptosis has been deeply investigated, revealing that deregulation in their expression is critical in various diseases, such as metabolic disorders and cancer. Since these small molecules initially evolved as a mechanism of protection against viruses and transposable elements, the fascinating hypothesis that they can move between organisms both of the same or different species has been postulated. Trans-kingdom is the term used to define the migration that occurs between species. This mechanism has been well analyzed between plants and their pests, in order to boost defense and increase pathogenicity, respectively. Intriguingly, in the last decades, the plant-human trans-kingdom migration via food intake hypothesis arose. In particular, various studies highlighted the ability of exogenous miRNAs, abundant in the mainly consumed plant-derived food, to enter the human body affecting gene expression. Notably, plant miRNAs can resist the strict conditions of the gastrointestinal tract through a methylation step that occurs during miRNA maturation, conferring high stability to these small molecules. Recent studies observed the anti-tumoral, immune modulator and anti-inflammatory abilities of trans-kingdom interaction between plant and human. Here, we depict the existing knowledge and discuss the fascinating plant-human trans-kingdom interaction, highlighting first the eventual role of plant miRNAs from foods on our somatic gene identity card and then the potential impact of using plant miRNAs as novel therapeutic avenues.
Collapse
Affiliation(s)
- Emanuela Pasculli
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (E.P.); (R.M.G.); (M.A.)
| | - Raffaella Maria Gadaleta
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (E.P.); (R.M.G.); (M.A.)
- INBB National Institute for Biostructure and Biosystems, Viale delle Medaglie d’Oro 305, 00136 Rome, Italy
| | - Maria Arconzo
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (E.P.); (R.M.G.); (M.A.)
| | - Marica Cariello
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (E.P.); (R.M.G.); (M.A.)
- INBB National Institute for Biostructure and Biosystems, Viale delle Medaglie d’Oro 305, 00136 Rome, Italy
| | - Antonio Moschetta
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (E.P.); (R.M.G.); (M.A.)
- INBB National Institute for Biostructure and Biosystems, Viale delle Medaglie d’Oro 305, 00136 Rome, Italy
| |
Collapse
|