1
|
Yang Y, Zhang C, Li H, He Q, Xie J, Liu H, Cui F, Lei Z, Qin X, Liu Y, Xu M, Huang S, Zhang X. A review of molecular interplay between inflammation and cancer: The role of lncRNAs in pathogenesis and therapeutic potential. Int J Biol Macromol 2025; 309:142824. [PMID: 40187457 DOI: 10.1016/j.ijbiomac.2025.142824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 03/31/2025] [Accepted: 04/02/2025] [Indexed: 04/07/2025]
Abstract
The inflammatory microenvironment (IME) has been demonstrated to facilitate the initiation and progression of tumors throughout the inflammatory process. Simultaneously, cancer can initiate or intensify the inflammatory response, thereby promoting tumor progression. This review examines the dual role of long non-coding RNAs (lncRNAs) in the interplay between inflammation and cancer. LncRNA modulate inflammation-induced cancer by influencing the activation of signaling pathways (NF-κB, Wnt/β-catenin, mTOR, etc), microRNA (miRNA) sponging, protein interactions, interactions with immune cells, and encoding short peptides. In contrast, lncRNAs also impact cancer-induced inflammatory processes by regulating cytokine expression, mediating tumor-derived extracellular vesicles (EVs), modulating intracellular reactive oxygen species (ROS) levels, and facilitating metabolic reprogramming. Furthermore, the therapeutic potential of lncRNA and the challenges of clinical translation were explicitly discussed as well. Overall, this review aims to provide a comprehensive and systematic resource for future researchers investigating the impact of lncRNAs on inflammation and cancer.
Collapse
Affiliation(s)
- Yan Yang
- Department of Pharmacy, The Third People's Hospital of Chengdu, Chengdu, China; School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Chuxi Zhang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Huacui Li
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China; Tangshan Institute of Southwest Jiaotong University, Tangshan, China
| | - Qin He
- Department of Pharmacy, The Third People's Hospital of Chengdu, Chengdu, China
| | - Jiang Xie
- Department of Pediatrics, The Third People's Hospital of Chengdu, Chengdu, China
| | - Hongmei Liu
- Department of Pharmacy, The Third People's Hospital of Chengdu, Chengdu, China
| | - Fenfang Cui
- Department of Pharmacy, The Third People's Hospital of Chengdu, Chengdu, China
| | - Ziqin Lei
- Department of Pharmacy, The Third People's Hospital of Chengdu, Chengdu, China
| | - Xiaoyan Qin
- Department of Pharmacy, The Third People's Hospital of Chengdu, Chengdu, China
| | - Ying Liu
- Department of Pharmacy, The Third People's Hospital of Chengdu, Chengdu, China
| | - Min Xu
- Department of Pharmacy, The Third People's Hospital of Chengdu, Chengdu, China.
| | - Shuai Huang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China.
| | - Xu Zhang
- Department of Pharmacy, Chengdu Integrated TCM & Western Medicine Hospital, Chengdu University of TCM, Chengdu, China.
| |
Collapse
|
2
|
Zhang M, Li L, Li S. The Role of miR-150-5p/SOCS1 Pathway in Arsenic-Induced Pyroptosis of LX-2 Cells. Biol Trace Elem Res 2025; 203:822-834. [PMID: 38689138 DOI: 10.1007/s12011-024-04211-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 04/25/2024] [Indexed: 05/02/2024]
Abstract
This study aims to explore the mechanism of pyroptosis of human hepatocyte LX-2 cells induced by NaAsO2 through the miR-150-5p/SOCS1 pathway. LX-2 cells were transfected with different concentrations of NaAsO2, miR-150-5p inhibitor, and SOCS1 agonist. Cell activity, cell pyroptosis, and the expression of related genes and proteins were detected by scanning electron microscopy, CCK-8, qRT-PCR, western blot, and immunofluorescence. Compared with the control group, 10 µmol/L and 20 µmol/L NaAsO2 significantly elevated the protein expression levels of the pyroptosis-related proteins NLRP3, GSDMD, GSDMD-N, caspase1, and cleaved caspase1 as well as the mRNA levels of NLRP3, GSDMD, caspase1, IL-18, and IL-1β. The typical pyroptosis with swelling and rupture of the plasma membrane was observed through scanning electron microscopy. The expression of miR-150-5p of the NaAsO2 intervention group increased, while the expression of SOCS1 decreased; then the level of NF-κB p65 elevated. With co-treatment of miR-150-5p inhibitor, SOCS1 agonist, and NaAsO2, the cell pyroptosis was attenuated, and the expressions of NLRP3, caspase1, GSDMD, GSDMD-N, IL-18, IL-1β, p65 of the group of miR-150-5p inhibitor and NaAsO2 group, and of the group of SOCS1 agonist and NaAsO2 reduced compared with the NaAsO2 group. Arsenic exposure promotes miR-150-5p, inhibits the expression of SOCS1, and activates the NF-κB/NLRP3 pathway in LX-2 cell pyroptosis.
Collapse
Affiliation(s)
- Mengyao Zhang
- Department of Preventive Medicine, College of Medicine, Shihezi University, Shihezi, 832000, China
- Key Laboratory for Prevention and Control of Emerging Infectious Diseases and Public Health Security, the Xinjiang Production and Construction Corps, Shihezi, China
| | - Linzhi Li
- Department of Preventive Medicine, College of Medicine, Shihezi University, Shihezi, 832000, China
- Key Laboratory for Prevention and Control of Emerging Infectious Diseases and Public Health Security, the Xinjiang Production and Construction Corps, Shihezi, China
| | - Shugang Li
- School of Public Health, Capital Medical University, Beijing, China.
| |
Collapse
|
3
|
Mathews L, Appukuttan D, Victor DJ, Venkadassalapathy S, Subramanian S, Prakash PSG. Role of miRNA-155 in macrophage polarisation in stage III/IV periodontitis with type II diabetes mellitus: An analytical case-control study. Hum Immunol 2025; 86:111214. [PMID: 39667205 DOI: 10.1016/j.humimm.2024.111214] [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: 07/15/2024] [Revised: 12/01/2024] [Accepted: 12/02/2024] [Indexed: 12/14/2024]
Abstract
AIM To evaluate the role of miR-155 in macrophage polarisation in stage III/IV periodontitis with Type 2 diabetes mellitus (T2DM). MATERIALS AND METHODS Sixty four patients were recruited and categorized into Group I-systemically and Periodontally healthy (n = 16), Group II-systemically healthy with Stage III/IV Periodontitis(n = 16), Group III-Periodontally healthy with T2DM (n = 16) and Group IV- Stage III/IV Periodontitis with T2DM(n = 16).Gingival tissue samples were collected and Real time-PCR was carried out for microRNA-155, TNF- α(marker for M1 phenotype) and Arg-1(marker for M2 phenotype) gene expression. RESULTS Group IV showed the highest increase in miR-155 fold change (FC) (11.17 ± 10.44), followed by groups III and II(10.35 ± 15.87, 5.1 ± 5.17 respectively) when compared to Group I. Likewise, the highest FC for TNF-α expression was observed in the group IV (10.11 ± 12.14). Groups II and III showed an almost similar increase in the FC(4.36 ± 4.48 and 4.79 ± 6.91, respectively). Periodontally healthy subjects demonstrated higher levels of Arg-1 gene expression(4.41 ± 5.17), followed by reduced expression in the groups II and IV (2.46 ± 2.21 and 2.65 ± 3.25, respectively). The TNF-α:Arg-1 ratio indicated that group I had higher Arg-1 expression, while group III and IV individuals had higher TNF-α expression. Compared to miR-155 and Arg-1, which demonstrated poor Area under the curve(AUC), sensitivity and specificity, TNF-α was able to distinguish between the groups III & IV (AUC = 0.74, p = 0.02, sensitivity 92 %, specificity 83 %, cut off = 27.28) and I & IV (AUC = 0.6, p = 0.03, sensitivity 81 %, specificity 77 %, cut off = 26.04). CONCLUSION Dysregulated miR-155 contributes to hyperinflammatory state in T2DM associated periodontitis by favouring macrophage polarisation towards the M1 phenotype.However, the strength of this relationship and the association with severity of periodontal disease could not be confirmed in this study.
Collapse
Affiliation(s)
- Leya Mathews
- Postgraduate Student, Department of Periodontics, SRM Dental College, Bharathi Salai, Chennai, India.
| | - Devapriya Appukuttan
- Professor, Department of Periodontics, SRM Dental College, Bharathi Salai,Chennai, India.
| | - Dhayanand John Victor
- Professor and Head of the Department, Department of Periodontics, SRM Dental College, Bharathi Salai, Chennai, India
| | | | - Sangeetha Subramanian
- Professor, Department of Periodontics, SRM Dental College, Bharathi Salai,Chennai, India
| | - P S G Prakash
- Professor, Department of Periodontics, SRM Dental College, Bharathi Salai,Chennai, India
| |
Collapse
|
4
|
Gu Q, Qi A, Wang N, Zhou Z, Zhou X. Macrophage dynamics in prostate cancer: Molecular to therapeutic insights. Biomed Pharmacother 2024; 177:117002. [PMID: 38960836 DOI: 10.1016/j.biopha.2024.117002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 06/08/2024] [Accepted: 06/17/2024] [Indexed: 07/05/2024] Open
Abstract
This review provides an in-depth examination of the role that tumor-associated macrophages (TAMs) play in the progression of prostate cancer (PCa), with a particular focus on the factors influencing the polarization of M1 and M2 macrophages and the implications of targeting these cells for cancer progression. The development and prognosis of PCa are significantly influenced by the behavior of macrophages within the tumor microenvironment. M1 macrophages typically exhibit anti-tumor properties by secreting pro-inflammatory cytokines such as interferon-gamma (IFN-γ) and tumor necrosis factor-alpha (TNF-α), thereby enhancing the immune response. Conversely, M2 macrophages contribute to tumor cell migration and invasion through the production of factors like arginase-1 (Arg1) and interleukin-10 (IL-10). This review not only explores the diverse factors that affect macrophage polarization but also delves into the potential therapeutic strategies targeting macrophage polarization, including the critical roles of non-coding RNA and exosomes in regulating this process. The polarization state of macrophages is highlighted as a key determinant in PCa progression, offering a novel perspective for clinical treatment. Future research should concentrate on gaining a deeper understanding of the molecular mechanisms underlying macrophage polarization and on developing effective targeted therapeutic strategies. The exploration of the potential of combination therapies to improve treatment efficacy is also emphasized. By emphasizing the importance of macrophages as a therapeutic target in PCa, this review aims to provide valuable insights and research directions for clinicians and researchers.
Collapse
Affiliation(s)
- Qiannan Gu
- China Pharmaceutical University, School of Basic Medicine and Clinical Pharmacy, Nanjing, Jiangsu 210009, China
| | - Anning Qi
- Medical Laboratory, Liuhe People's Hospital of Jiangsu Province, Nanjing, Jiangsu 211500, China
| | - Ne Wang
- Jiangning Hospital Tiandi New City Branch, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, Jiangsu Province 211198, China
| | - Zhenxian Zhou
- Nanjing Second People's Hospital, Jiangsu Province 211103, China
| | - Xiaohui Zhou
- China Pharmaceutical University, School of Basic Medicine and Clinical Pharmacy, Nanjing, Jiangsu 210009, China; Jiangning Outpatient Department of China Pharmaceutical University, Nanjing 211198, China.
| |
Collapse
|
5
|
Li H, Wang S, Yang Z, Meng X, Niu M. Nanomaterials modulate tumor-associated macrophages for the treatment of digestive system tumors. Bioact Mater 2024; 36:376-412. [PMID: 38544737 PMCID: PMC10965438 DOI: 10.1016/j.bioactmat.2024.03.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 02/25/2024] [Accepted: 03/03/2024] [Indexed: 11/25/2024] Open
Abstract
The treatment of digestive system tumors presents challenges, particularly in immunotherapy, owing to the advanced immune tolerance of the digestive system. Nanomaterials have emerged as a promising approach for addressing these challenges. They provide targeted drug delivery, enhanced permeability, high bioavailability, and low toxicity. Additionally, nanomaterials target immunosuppressive cells and reshape the tumor immune microenvironment (TIME). Among the various cells in the TIME, tumor-associated macrophages (TAMs) are the most abundant and play a crucial role in tumor progression. Therefore, investigating the modulation of TAMs by nanomaterials for the treatment of digestive system tumors is of great significance. Here, we present a comprehensive review of the utilization of nanomaterials to modulate TAMs for the treatment of gastric cancer, colorectal cancer, hepatocellular carcinoma, and pancreatic cancer. We also investigated the underlying mechanisms by which nanomaterials modulate TAMs to treat tumors in the digestive system. Furthermore, this review summarizes the role of macrophage-derived nanomaterials in the treatment of digestive system tumors. Overall, this research offers valuable insights into the development of nanomaterials tailored for the treatment of digestive system tumors.
Collapse
Affiliation(s)
- Hao Li
- Department of Interventional Radiology, First Hospital of China Medical University, Shenyang, China
| | - Shuai Wang
- Department of Interventional Radiology, First Hospital of China Medical University, Shenyang, China
| | - Zhengqiang Yang
- Department of Interventional Therapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xianwei Meng
- Laboratory of Controllable Preparation and Application of Nanomaterials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, China
| | - Meng Niu
- China Medical University, Shenyang, China
| |
Collapse
|
6
|
Peng W, Yang Y, Lu H, Shi H, Jiang L, Liao X, Zhao H, Wang W, Liu J. Network pharmacology combines machine learning, molecular simulation dynamics and experimental validation to explore the mechanism of acetylbinankadsurin A in the treatment of liver fibrosis. JOURNAL OF ETHNOPHARMACOLOGY 2024; 323:117682. [PMID: 38169205 DOI: 10.1016/j.jep.2023.117682] [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: 10/03/2023] [Revised: 12/10/2023] [Accepted: 12/26/2023] [Indexed: 01/05/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The Kadsura coccinea (Lem.) A. C. Smith is known as "Heilaohu" of the Tujia ethnomedicine in China. It has anti-tumor, anti-oxidation, anti-HIV, anti-inflammatory and liver protective effects, used to treat diseases such as rheumatoid arthritis, cancer, gastritis and hepatitis. In this research, we investigated the anti-fibrotic effect and possible mechanisms of acetylbinankadsurin A (ACBA) in vitro and in vivo, which is a natural compound derived from roots of K. coccinea. AIM OF THE STUDY We try to evaluate the efficacy of ACBA in the treatment of liver fibrosis and to explore the underlying mechanisms of ACBA by network pharmacology, machine learning, molecular docking, molecular dynamics simulations, and experimental assessment. MATERIALS AND METHODS ACBA was isolated from the CH2Cl2 layer of the roots of K. coccinea through column chromatographic techniques. The structure of ACBA was determined by using 1D and 2D NMR. CCl4-induced C57BL/6 mouse liver fibrosis models were established to evaluate the anti-fibrosis effects of ACBA in vivo. The molecular targets of ACBA and liver fibrosis were obtained from various databases, then constructed a protein-protein interaction (PPI) networks through the STRING database. Gene ontology (GO) enrichment and kyoto encyclopedia of genes and genomes (KEGG) analysis were applied using the "clusterProfiler" R package. Furthermore, the key genes for ACBA treatment of liver fibrosis were identified by the least absolute shrinkage and selection operator (LASSO). Molecular docking and molecular dynamics simulations were also carried out. Finally, the target and pathway of ACBA were verified by immunofluorescence staining, RT-PCR and Western blot. RESULT First, ACBA attenuated CCl4-induced liver injury and fibrosis in vivo. These findings were accompanied by decreased expression of α-SMA and collagen I. Second, ACBA significantly decreased serum levels of ALT, AST, TNF-α and IL-6. Then, we identified 133 potential targets of ACBA and 7987 targets of liver fibrosis. KEGG analysis showed that ACBA could regulate the drug metabolism - cytochrome P450, fructose and mannose metabolism, IL-17 and NF-κB signaling pathways. Next, six core targets was screened by LASSO analysis including AKR1B1, PFKFB3, EPHA3, CDK1, CCR1 and CYP3A4. Molecular docking showed that ACBA has a good binding affinity for CCR1. Furthermore, compared with CCR1 inhibitor BX-471, The results of molecular simulation dynamics showed that ACBA was stable in binding with CCR1. Consistently, ACBA remarkably downregulated the expression of CCR1, p-NF-κBp65, p-IκBα, p-STAT1 and TNF-α proteins, which were upregulated in CCl4-induced hepatic fibrosis and LPS-THP-1 cells. CONCLUSION Our results suggest that ACBA significantly attenuated CCl4-induced liver fibrosis in histopathological and in serum level. This effect may be mediated by CCR1, NF-κB and STAT1 signalling.
Collapse
Affiliation(s)
- Wangxia Peng
- Science and Technology Innovation Center of Hunan University of Traditional Chinese Medicine, Innovation Base of Hunan State Key Laboratory of Innovative Medicine and Traditional Chinese Medicine, Changsha, 410208, China
| | - Yupei Yang
- TCM and Ethnomedicine Innovation & Development International Laboratory, Innovative Materia Medica Research Institute, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Huaguan Lu
- Science and Technology Innovation Center of Hunan University of Traditional Chinese Medicine, Innovation Base of Hunan State Key Laboratory of Innovative Medicine and Traditional Chinese Medicine, Changsha, 410208, China
| | - Huan Shi
- Science and Technology Innovation Center of Hunan University of Traditional Chinese Medicine, Innovation Base of Hunan State Key Laboratory of Innovative Medicine and Traditional Chinese Medicine, Changsha, 410208, China
| | - Lihong Jiang
- Science and Technology Innovation Center of Hunan University of Traditional Chinese Medicine, Innovation Base of Hunan State Key Laboratory of Innovative Medicine and Traditional Chinese Medicine, Changsha, 410208, China
| | - Xiaolin Liao
- Science and Technology Innovation Center of Hunan University of Traditional Chinese Medicine, Innovation Base of Hunan State Key Laboratory of Innovative Medicine and Traditional Chinese Medicine, Changsha, 410208, China
| | - Hongqing Zhao
- Science and Technology Innovation Center of Hunan University of Traditional Chinese Medicine, Innovation Base of Hunan State Key Laboratory of Innovative Medicine and Traditional Chinese Medicine, Changsha, 410208, China
| | - Wei Wang
- TCM and Ethnomedicine Innovation & Development International Laboratory, Innovative Materia Medica Research Institute, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China.
| | - Jianjun Liu
- Science and Technology Innovation Center of Hunan University of Traditional Chinese Medicine, Innovation Base of Hunan State Key Laboratory of Innovative Medicine and Traditional Chinese Medicine, Changsha, 410208, China.
| |
Collapse
|
7
|
Topchieva LV, Kurbatova IV, Dudanova OP, Vasileva AV, Zhulai GA. Immune cell balance as potential biomarker of progressing non-alcoholic fatty liver disease. GENES & CELLS 2024; 19:105-125. [DOI: 10.17816/gc610252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a widespread chronic, slowly progressive metabolic multifactorial disease. It is represented by several clinical and morphological forms: steatosis, nonalcoholic steatohepatitis (NASH) (with or without fibrosis), and liver cirrhosis. The search for minimally invasive and cost-effective biomarkers of NAFLD is a key task in the diagnosis, staging of progression, and long-term monitoring of NAFLD. This article discusses the possibility of using immune cell balance as potential minimally invasive peripheral markers of NAFLD progression. In the progression of NASH from steatosis to fibrosis and cirrhosis, inflammation plays an important role because of the activation of Kupffer cells and increased migration of monocytes, dendritic cells, neutrophils, and activated T lymphocytes into the tissues. Macrophages originating from monocytes, with NASH progression, gradually begin to prevail over the pool of resident macrophages. The risk of NASH and fibrosis development in patients with NAFLD increases with the ratio of neutrophils/lymphocytes in the liver. An increase in the Th17 cell count and a decrease in T-regulatory cell count can contribute to increased hepatic steatosis and inflammation development in NAFLD and accelerate the transition from simple steatosis to steatohepatitis and fibrosis. Information on the participation of noncoding RNAs in the regulation of the balance of immune cells in NAFLD is presented, which also allows us to consider them as additional, along with cellular, markers of disease progression.
Collapse
|