1
|
Chang Q, Mao H, Feng J, Rao L, Tang W, Zhang Z, Hu Z. Sodium butyrate increases USP5-mediated ubiquitination degradation of GPX4 and enhances anti-cancer efficacy of anti-PD-1 antibody. Biochem Pharmacol 2025; 237:116927. [PMID: 40216263 DOI: 10.1016/j.bcp.2025.116927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 03/25/2025] [Accepted: 04/02/2025] [Indexed: 04/21/2025]
Abstract
Hepatocellular carcinoma (HCC), a prevalent malignancy associated with a dismal prognosis, necessitates the urgent exploration of novel therapeutic avenues. Ferroptosis, an iron-mediated, lipid peroxidation-induced form of regulated cell death (RCD), has emerged as a promising target for cancer therapy. Sodium butyrate (NaBu), a short-chain fatty acid sodium salt, has demonstrated antitumor efficacy against diverse cancers, yet its specific role and mechanisms in HCC treatment remain elusive. Our findings reveal that NaBu not only impedes HCC cell growth and epithelial-mesenchymal transition (EMT) but also triggers ferroptosis by enhancing Fe2+ accumulation, reactive oxygen species (ROS) generation, and malondialdehyde (MDA) production. Notably, these effects are effectively mitigated by Ferrostatin-1 (Fer-1), underscoring the ferroptosis-inducing capacity of NaBu. Mechanistically, NaBu exerts its action by diminishing the level of ubiquitin-specific protease 5 (USP5), which subsequently leads to the ubiquitination and destabilization of glutathione peroxidase 4 (GPX4), a crucial suppressor in ferroptosis. In a preclinical setting, NaBu significantly inhibits tumor xenograft growth in nude mice, highlighting its in vivo efficacy. When paired with an anti-programmed death 1 (PD-1) antibody, NaBu exhibits a potent synergistic antitumor effect, suggesting a potential role in enhancing immunotherapy response. Collectively, our results underscore the potential of NaBu as a novel therapeutic agent for HCC, through its ability to inhibit USP5 and indirectly downregulate GPX4, thereby stimulating ferroptosis.
Collapse
Affiliation(s)
- Qimeng Chang
- Department of Hepatobiliary and Pancreatic Surgery, Minhang Hospital, Fudan University, Shanghai 201199, China; Key Laboratory of Whole-Period Monitoring and Precise Intervention of Digestive Cancer, Shanghai Municipal Health Commission, Minhang Hospital, Fudan University, Shanghai 201199, China; Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai 201199, China
| | - Huarong Mao
- Department of Hepatobiliary and Pancreatic Surgery, Minhang Hospital, Fudan University, Shanghai 201199, China; Key Laboratory of Whole-Period Monitoring and Precise Intervention of Digestive Cancer, Shanghai Municipal Health Commission, Minhang Hospital, Fudan University, Shanghai 201199, China; Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai 201199, China
| | - Jinfeng Feng
- Department of Hepatobiliary and Pancreatic Surgery, Minhang Hospital, Fudan University, Shanghai 201199, China; Key Laboratory of Whole-Period Monitoring and Precise Intervention of Digestive Cancer, Shanghai Municipal Health Commission, Minhang Hospital, Fudan University, Shanghai 201199, China; Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai 201199, China
| | - Longhua Rao
- Department of Hepatobiliary and Pancreatic Surgery, Minhang Hospital, Fudan University, Shanghai 201199, China; Key Laboratory of Whole-Period Monitoring and Precise Intervention of Digestive Cancer, Shanghai Municipal Health Commission, Minhang Hospital, Fudan University, Shanghai 201199, China; Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai 201199, China
| | - Weiguo Tang
- Department of Hepatobiliary and Pancreatic Surgery, Minhang Hospital, Fudan University, Shanghai 201199, China; Key Laboratory of Whole-Period Monitoring and Precise Intervention of Digestive Cancer, Shanghai Municipal Health Commission, Minhang Hospital, Fudan University, Shanghai 201199, China; Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai 201199, China
| | - Ziping Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Minhang Hospital, Fudan University, Shanghai 201199, China; Key Laboratory of Whole-Period Monitoring and Precise Intervention of Digestive Cancer, Shanghai Municipal Health Commission, Minhang Hospital, Fudan University, Shanghai 201199, China; Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai 201199, China
| | - Zhiqiu Hu
- Department of Hepatobiliary and Pancreatic Surgery, Minhang Hospital, Fudan University, Shanghai 201199, China; Key Laboratory of Whole-Period Monitoring and Precise Intervention of Digestive Cancer, Shanghai Municipal Health Commission, Minhang Hospital, Fudan University, Shanghai 201199, China; Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai 201199, China.
| |
Collapse
|
2
|
Zhang Y, Ma T, Lu X, Hua H, Wu L, Chen Z. Mechanical mechanics-reclaiming a new battlefield for chronic liver disease. J Adv Res 2025:S2090-1232(25)00346-7. [PMID: 40379238 DOI: 10.1016/j.jare.2025.05.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Revised: 04/17/2025] [Accepted: 05/12/2025] [Indexed: 05/19/2025] Open
Abstract
BACKGROUND In the 21st century, significant breakthroughs have been made in the research of chronic liver disease. New biochemical markers of pathogenicity and corresponding drugs continue to emerge. However, current treatment strategies remain unsatisfactory due to complex pathological changes in the liver, including vascular dysfunction, myofibroblast-like transition, and local tissue necrosis in liver sinusoids. These challenges have created an urgent need for innovative, synergistic treatments. Mechanical mechanics is a growing field, with increasing evidence suggesting that mechanical signals play a role similar to that of biochemical markers. These signals influence response speed, conduction intensity, and functional diversity in regulating cell activities. AIM OF REVIEW This review summarizes the three main mechanical characteristics involved in the progression of "liver fibrosis-cirrhosis-hepatocellular carcinoma" and provides an in-depth interpretation of several mechanically-related targets. Finally, current and cutting-edge therapeutic strategies are proposed from a cellular perspective. Despite the many challenges that remain, this review is both relevant and significant.
Collapse
Affiliation(s)
- Yiheng Zhang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Tianle Ma
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - XingXing Lu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Haibing Hua
- Department of Gastroenterology, Jiangyin Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Jiangyin 214400, China.
| | - Li Wu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; Engineering Center of State Ministry of Education for Standardization of Chinese Medicine Processing, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Zhipeng Chen
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; Engineering Center of State Ministry of Education for Standardization of Chinese Medicine Processing, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
3
|
Zhang F, Li J, Liu X, Pan X, He S, Zhang J, Shen H, Tang S, Song S. Radiosynthesis and Evaluation of a Novel 68Ga-Labeled Peptide for PD-L1-Targeted PET Imaging. Mol Pharm 2025; 22:2694-2702. [PMID: 40257351 DOI: 10.1021/acs.molpharmaceut.5c00193] [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: 04/22/2025]
Abstract
Although programmed death-ligand 1 (PD-L1)-targeted immunotherapy has demonstrated favorable therapeutic effects, concern regarding a low response rate persists. Our study aimed to develop a novel peptide probe for PD-L1 targeting positron emission tomography (PET)/computed tomography (CT) imaging as an alternative for assessing PD-L1 expression and exploring its potential role in guiding PD-L1 immunotherapy in vivo. The probe targeting PD-L1 was obtained by modifying 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA) with the peptide CCC, followed by labeling with 68Ga. Radiochemical purity, log P, pharmacokinetics, and stability of the probe were evaluated in vitro and in vivo. The systematic evaluation of the probe performance included microPET/CT imaging, biodistribution, biosafety, and an investigation of its use in immunotherapy monitoring. 68Ga-DOTA-CCC was successfully synthesized with over 99% radiochemical purity, eliminating the need for purification. The probe exhibited good hydrophilicity and stability and was rapidly metabolized with a short blood clearance half-life of 16.1 ± 0.5 min. Additionally, the probe exhibited an excellent PD-L1 targeting ability, with tumor uptake positively correlating with PD-L1 expression levels in both cellular experiments and microPET/CT imaging. Moreover, the dynamic expression of PD-L1 was assessed using 68Ga-DOTA-CCC during atezolizumab administration. 68Ga-DOTA-CCC accurately reflects PD-L1 expression and holds promise for precisely guiding PD-L1-targeted immunotherapy.
Collapse
Affiliation(s)
- Fengsheng Zhang
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China
- Department of Nuclear Medicine, Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China
- Department of Oncology, Shanghai Medical College and Center for Biomedical Imaging, Fudan University, Shanghai 200032, P.R. China
- Center for Biomedical imaging, Fudan University, Shanghai 200032, P.R. China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai 200032, P.R. China
- Department of Nuclear Medicine, Shanghai Proton and Heavy Ion Center, Shanghai 200032, China
- Key Laboratory of Nuclear Physics and Ion-beam Application (MOE), Institute of Modern Physics, Fudan University, Shanghai 200433, China
| | - Jindian Li
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China
- Department of Nuclear Medicine, Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China
- Department of Oncology, Shanghai Medical College and Center for Biomedical Imaging, Fudan University, Shanghai 200032, P.R. China
- Center for Biomedical imaging, Fudan University, Shanghai 200032, P.R. China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai 200032, P.R. China
- Department of Nuclear Medicine, Shanghai Proton and Heavy Ion Center, Shanghai 200032, China
| | - Xuwei Liu
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China
- Department of Nuclear Medicine, Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China
- Department of Oncology, Shanghai Medical College and Center for Biomedical Imaging, Fudan University, Shanghai 200032, P.R. China
- Center for Biomedical imaging, Fudan University, Shanghai 200032, P.R. China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai 200032, P.R. China
- Department of Nuclear Medicine, Shanghai Proton and Heavy Ion Center, Shanghai 200032, China
| | - Xiaoyu Pan
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China
- Department of Nuclear Medicine, Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China
- Department of Oncology, Shanghai Medical College and Center for Biomedical Imaging, Fudan University, Shanghai 200032, P.R. China
- Center for Biomedical imaging, Fudan University, Shanghai 200032, P.R. China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai 200032, P.R. China
- Department of Nuclear Medicine, Shanghai Proton and Heavy Ion Center, Shanghai 200032, China
| | - Simin He
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China
- Department of Nuclear Medicine, Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China
- Department of Oncology, Shanghai Medical College and Center for Biomedical Imaging, Fudan University, Shanghai 200032, P.R. China
- Center for Biomedical imaging, Fudan University, Shanghai 200032, P.R. China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai 200032, P.R. China
- Department of Nuclear Medicine, Shanghai Proton and Heavy Ion Center, Shanghai 200032, China
| | - Jianping Zhang
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China
- Department of Nuclear Medicine, Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China
- Department of Oncology, Shanghai Medical College and Center for Biomedical Imaging, Fudan University, Shanghai 200032, P.R. China
- Center for Biomedical imaging, Fudan University, Shanghai 200032, P.R. China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai 200032, P.R. China
- Department of Nuclear Medicine, Shanghai Proton and Heavy Ion Center, Shanghai 200032, China
| | - Hao Shen
- Key Laboratory of Nuclear Physics and Ion-beam Application (MOE), Institute of Modern Physics, Fudan University, Shanghai 200433, China
| | - Shuang Tang
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China
- Department of Nuclear Medicine, Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China
- Department of Oncology, Shanghai Medical College and Center for Biomedical Imaging, Fudan University, Shanghai 200032, P.R. China
- Center for Biomedical imaging, Fudan University, Shanghai 200032, P.R. China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai 200032, P.R. China
- Department of Nuclear Medicine, Shanghai Proton and Heavy Ion Center, Shanghai 200032, China
| | - Shaoli Song
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China
- Department of Nuclear Medicine, Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China
- Department of Oncology, Shanghai Medical College and Center for Biomedical Imaging, Fudan University, Shanghai 200032, P.R. China
- Center for Biomedical imaging, Fudan University, Shanghai 200032, P.R. China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai 200032, P.R. China
- Department of Nuclear Medicine, Shanghai Proton and Heavy Ion Center, Shanghai 200032, China
- Key Laboratory of Nuclear Physics and Ion-beam Application (MOE), Institute of Modern Physics, Fudan University, Shanghai 200433, China
| |
Collapse
|
4
|
Zhang G, Zhang G, Zhao Y, Wan Y, Jiang B, Wang H. Unveiling the nexus of p53 and PD-L1: insights into immunotherapy resistance mechanisms in hepatocellular carcinoma. Am J Cancer Res 2025; 15:1410-1435. [PMID: 40371157 PMCID: PMC12070102 DOI: 10.62347/brto3272] [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: 01/26/2025] [Accepted: 03/25/2025] [Indexed: 05/16/2025] Open
Abstract
Hepatocellular carcinoma (HCC), the predominant form of primary liver cancer worldwide, continues to pose a substantial health challenge with limited treatment options for advanced stages. Despite progress in therapies such as surgery, transplantation, and targeted treatments, prognosis remains bleak for many patients. The advent of immunotherapy has revolutionized the landscape of advanced HCC treatment, offering hope for improved outcomes. However, its efficacy is limited, with a modest response rate of approximately 20% as a single-agent therapy, underscoring the urgent need to decipher mechanisms of immunotherapy resistance. Tumor protein 53 gene (TP53), a pivotal tumor suppressor gene, and Programmed death ligand 1 (PD-L1), a crucial immune checkpoint ligand, play central roles in HCC's evasion of immune responses. Understanding how tumor protein 53 (p53) influences PD-L1 expression and immune system interactions is essential for unraveling the complexities of immunotherapy resistance mechanisms. Elucidating these molecular interactions not only enhances our understanding of HCC's underlying mechanisms but also lays the foundation for developing targeted treatments that may improve outcomes for patients with advanced-stage liver cancer. Ultimately, deciphering the nexus of p53 and PD-L1 in immunotherapy resistance promises to advance treatment strategies and outcomes in the challenging landscape of HCC. This review delves into the intricate relationship between p53 and PD-L1 concerning immunotherapy resistance in HCC, offering insights that could pave the way for novel therapeutic strategies aimed at enhancing treatment efficacy and overcoming resistance in advanced stages of the disease.
Collapse
Affiliation(s)
- Guoyuan Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Taihe Hospital, Hubei University of MedicineShiyan 442000, Hubei Province, China
- Department of Hepatobiliary and Pancreatic Surgery, Hubei Provincial Clinical Research Center for Precision Diagnosis and Treatment of Liver Cancer, Taihe Hospital, Hubei University of MedicineShiyan 442000, Hubei, China
| | - Gan Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Taihe Hospital, Hubei University of MedicineShiyan 442000, Hubei Province, China
- Department of Hepatobiliary and Pancreatic Surgery, Hubei Provincial Clinical Research Center for Precision Diagnosis and Treatment of Liver Cancer, Taihe Hospital, Hubei University of MedicineShiyan 442000, Hubei, China
| | - Yixuan Zhao
- Department of Hepatobiliary and Pancreatic Surgery, Taihe Hospital, Hubei University of MedicineShiyan 442000, Hubei Province, China
- Department of Hepatobiliary and Pancreatic Surgery, Hubei Provincial Clinical Research Center for Precision Diagnosis and Treatment of Liver Cancer, Taihe Hospital, Hubei University of MedicineShiyan 442000, Hubei, China
| | - Yunyan Wan
- Department of Hepatobiliary and Pancreatic Surgery, Taihe Hospital, Hubei University of MedicineShiyan 442000, Hubei Province, China
- Department of Hepatobiliary and Pancreatic Surgery, Hubei Provincial Clinical Research Center for Precision Diagnosis and Treatment of Liver Cancer, Taihe Hospital, Hubei University of MedicineShiyan 442000, Hubei, China
| | - Bin Jiang
- Department of Hepatobiliary and Pancreatic Surgery, Taihe Hospital, Hubei University of MedicineShiyan 442000, Hubei Province, China
- Department of Hepatobiliary and Pancreatic Surgery, Hubei Provincial Clinical Research Center for Precision Diagnosis and Treatment of Liver Cancer, Taihe Hospital, Hubei University of MedicineShiyan 442000, Hubei, China
| | - Huaxiang Wang
- Department of Hepatobiliary and Pancreatic Surgery, Taihe Hospital, Hubei University of MedicineShiyan 442000, Hubei Province, China
- Department of Hepatobiliary and Pancreatic Surgery, Hubei Provincial Clinical Research Center for Precision Diagnosis and Treatment of Liver Cancer, Taihe Hospital, Hubei University of MedicineShiyan 442000, Hubei, China
| |
Collapse
|
5
|
Guo R, Rao PG, Liao BZ, Luo X, Yang WW, Lei XH, Ye JM. Melatonin suppresses PD-L1 expression and exerts antitumor activity in hepatocellular carcinoma. Sci Rep 2025; 15:8451. [PMID: 40069331 PMCID: PMC11897332 DOI: 10.1038/s41598-025-93486-4] [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: 06/19/2024] [Accepted: 03/07/2025] [Indexed: 03/14/2025] Open
Abstract
Melatonin, also known as the pineal hormone, is secreted by the pineal gland and primarily regulates circadian rhythms. Additionally, it possesses immunomodulatory properties and anticancer effects. However, its specific mechanism in hepatocellular carcinoma (HCC) remains unclear, particularly regarding its effect on HCC-mediated immune escape through PD-L1 expression.In this study, in vitro experiments were conducted using Huh7 and HepG2 HCC cells. Melatonin treatment was applied to both cell types to observe changes in malignant phenotypes. Additionally, melatonin-pretreated Huh7 or HepG2 cells were co-cultured with T cells to simulate the tumor microenvironment. The results showed that melatonin inhibited cancer cell proliferation, migration, and invasion, as well as reduced PD-L1 expression in cancer cells, exhibiting similar anti-cancer effects in the co-culture system. In vivo experiments involved establishing ascitic HCC mouse models using H22 cells, followed by subcutaneous tumor models in Balb/c nude and Balb/c wild-type mice. Melatonin inhibited tumor growth and suppressed PD-L1 expression in cancer tissues in both subcutaneous tumor models, and it increased T lymphocyte activity in the spleen of Balb/c wild-type mice. Overall, the in vitro and in vivo experiments demonstrated that melatonin has dual anti-cancer effects in HCC: direct intrinsic anti-cancer activity and enhancement of anti-tumor immunity by reducing PD-L1 expression thereby inhibiting cancer immune escape. Furthermore, a decrease in the expression of the upstream molecule HIF-1α of PD-L1 and an increase in the expression levels of JNK, P38, and their phosphorylated forms were detected. Thus, the mechanism by which melatonin reduces PD-L1 may involve the downregulation of HIF-1α expression or the activation of the MAPK-JNK and MAPK-P38 pathways. This provides new insights and strategies for HCC treatment.
Collapse
Affiliation(s)
- Rui Guo
- Suzhou Medical College of Soochow University, Suzhou, China.
- Department of Anesthesiology, First Affiliated Hospital of Gannan Medical University, No.23 Qingnian Road, Ganzhou City, Jiangxi Province, China.
| | - Pan-Guo Rao
- Gannan Medical University, Ganzhou, Jiangxi, China
| | | | - Xin Luo
- Gannan Medical University, Ganzhou, Jiangxi, China
| | - Wen-Wen Yang
- Gannan Medical University, Ganzhou, Jiangxi, China
| | | | - Jun-Ming Ye
- Suzhou Medical College of Soochow University, Suzhou, China.
- Gannan Medical University, Ganzhou, Jiangxi, China.
| |
Collapse
|
6
|
Kumar A, Rajput DS, Gupta MK, Kumar V, Singh H, Mishra AK, Chopra S, Chopra H. A novel phosphodiesterase target as a therapeutic approach: inhibiting DEN-induced hepatocellular carcinoma progression. EXCLI JOURNAL 2025; 24:407-429. [PMID: 40166422 PMCID: PMC11956523 DOI: 10.17179/excli2024-7941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 03/03/2025] [Indexed: 04/02/2025]
Abstract
Hepatocellular Carcinoma (HCC) is one of the most common and fatal types of liver cancer worldwide; in this sense, Diethylnitrosamine (DEN) has been established as a potent carcinogen affecting the development and progression of this disease. The present work focused on determining whether phosphodiesterase (PDE) enzymes, especially PDE5, may serve as targets in the therapeutic treatment of DEN-induced HCC. PDE5 inhibitors, widely used as therapeutic drugs for cardiovascular diseases and erectile dysfunction, have recently been found to be promising in preclinical cancer models through the modulation of key signaling pathways implicated in the progression of tumors, such as the cGMP-PKG, JNK, and MAPK pathways. These pathways are very important for cell proliferation, apoptosis and metastasis, and their dysregulation contributes to the aggressive nature of HCC. This study assessed the potential of PDE5 inhibitors to suppress proliferation, induce apoptosis, and alter the tumor microenvironment, thus potentially improving standard chemotherapy and immunotherapy interventions. By inhibiting certain PDE isoforms with these drugs, an anticancer response might occur as part of a complex mechanism that acts on both cancer cells and the microenvironment favorable for tumor growth. A preliminary review indicated that PDE inhibitors may be a promising therapeutic approach for overcoming some of the shortcomings of current treatments, particularly the development of resistance and the toxic effects of these treatments. Additional clinical investigations are necessary to determine the safety profile, appropriate amount of Osage, and long-term efficacy of these agents in the treatment of HCC, particularly in DEN-induced animal models. This study contributes to the expanding body of evidence supporting the use of PDE inhibitors in cancer treatment.
Collapse
Affiliation(s)
- Anil Kumar
- Faculty of Medical and Paramedical Sciences, Madhyanchal Professional University, Bhopal-462044, Madhya Pradesh, India
| | - Dharmendra Singh Rajput
- Faculty of Medical and Paramedical Sciences, Madhyanchal Professional University, Bhopal-462044, Madhya Pradesh, India
| | - Mandeep Kumar Gupta
- Moradabad Educational Trust Group of Institutions Faculty of Pharmacy, Moradabad-244001, Uttar Pradesh, India
| | - Vivek Kumar
- Moradabad Educational Trust Group of Institutions Faculty of Pharmacy, Moradabad-244001, Uttar Pradesh, India
| | - Harpreet Singh
- School of Pharmaceutical Sciences (Faculty of Pharmacy), IFTM University, Moradabad, Uttar Pradesh-244102, India
| | - Arun Kumar Mishra
- SOS School of Pharmacy (Faculty of Pharmacy), IFTM University, Moradabad, Uttar Pradesh-244102, India
| | - Shivani Chopra
- Department of Biosciences, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Chennai - 602105, Tamil Nadu, India
| | - Hitesh Chopra
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India
| |
Collapse
|
7
|
Garrido NJ, González-Martínez F, Torres AM, Blasco-Segura P, Losada S, Plaza A, Mateo J. Role of Artificial Intelligence in Identifying Vital Biomarkers with Greater Precision in Emergency Departments During Emerging Pandemics. Int J Mol Sci 2025; 26:722. [PMID: 39859435 PMCID: PMC11765594 DOI: 10.3390/ijms26020722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 01/12/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
The COVID-19 pandemic has accelerated advances in molecular biology and virology, enabling the identification of key biomarkers to differentiate between severe and mild cases. Furthermore, the use of artificial intelligence (AI) and machine learning (ML) to analyze large datasets has been crucial for rapidly identifying relevant biomarkers for disease prognosis, including COVID-19. This approach enhances diagnostics in emergency settings, allowing for more accurate and efficient patient management. This study demonstrates how machine learning algorithms in emergency departments can rapidly identify key biomarkers for the vital prognosis in an emerging pandemic using COVID-19 as an example by analyzing clinical, epidemiological, analytical, and radiological data. All consecutively admitted patients were included, and more than 89 variables were processed using the Random Forest (RF) algorithm. The RF model achieved the highest balanced accuracy at 92.61%. The biomarkers most predictive of mortality included procalcitonin (PCT), lactate dehydrogenase (LDH), and C-reactive protein (CRP). Additionally, the system highlighted the significance of interstitial infiltrates in chest X-rays and D-dimer levels. Our results demonstrate that RF is crucial in identifying critical biomarkers in emerging diseases, accelerating data analysis, and optimizing prognosis and personalized treatment, emphasizing the importance of PCT and LDH in high-risk patients.
Collapse
Affiliation(s)
- Nicolás J. Garrido
- Internal Medicine, Virgen de la Luz Hospital, 16002 Cuenca, Spain
- Expert Medical Analysis Group, Institute of Technology, University of Castilla-La Mancha, 16071 Cuenca, Spain
- Expert Medical Analysis Group, Instituto de Investigación Sanitaria de Castilla-La Mancha (IDISCAM), 45071 Toledo, Spain
| | - Félix González-Martínez
- Expert Medical Analysis Group, Institute of Technology, University of Castilla-La Mancha, 16071 Cuenca, Spain
- Expert Medical Analysis Group, Instituto de Investigación Sanitaria de Castilla-La Mancha (IDISCAM), 45071 Toledo, Spain
- Department of Emergency Medicine, Virgen de la Luz Hospital, 16002 Cuenca, Spain
| | - Ana M. Torres
- Expert Medical Analysis Group, Institute of Technology, University of Castilla-La Mancha, 16071 Cuenca, Spain
- Expert Medical Analysis Group, Instituto de Investigación Sanitaria de Castilla-La Mancha (IDISCAM), 45071 Toledo, Spain
| | | | - Susana Losada
- Department of Emergency Medicine, Virgen de la Luz Hospital, 16002 Cuenca, Spain
| | - Adrián Plaza
- Department of Emergency Medicine, Virgen de la Luz Hospital, 16002 Cuenca, Spain
| | - Jorge Mateo
- Expert Medical Analysis Group, Institute of Technology, University of Castilla-La Mancha, 16071 Cuenca, Spain
- Expert Medical Analysis Group, Instituto de Investigación Sanitaria de Castilla-La Mancha (IDISCAM), 45071 Toledo, Spain
| |
Collapse
|
8
|
Zheng Q, Wu X, Peng S. The immunotherapy mechanism of Hedyotis Diffusae Herba in treating liver cancer: a study based on network pharmacology, bioinformatics, and experimental validation. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:951-965. [PMID: 39093467 DOI: 10.1007/s00210-024-03312-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 07/16/2024] [Indexed: 08/04/2024]
Abstract
Liver cancer is a malignant tumor that develops on or inside the liver. Hedyotis diffusa Willd (HDW) plays a significant role in anti-tumor activities; however, its mechanism against liver cancer remains unclear. This study aims to evaluate the immunotherapeutic mechanism of HDW in treating liver cancer through network pharmacology, bioinformatics analysis, and experimental validation. Network pharmacology was utilized to identify the active components and potential targets of HDW from the TCMSP database. A potential target protein-protein interaction (PPI) network was constructed using the STRING database, followed by function and pathway enrichment analysis of the targets using GO and KEGG methods. In addition, the key targets for HDW against liver cancer were identified using five different algorithms in Cytoscape. The TCGA and HPA databases were used to assess the mRNA and protein expression of core target genes in normal liver and liver cancer tissues and their relationship with overall survival in liver cancer, as well as their role in immune infiltration. Molecular docking between the core components of HDW and the core targets was performed using PyMOL software. The effects of HDW on the proliferation and apoptosis of liver cancer cells were examined using MTT and flow cytometry. The regulatory effects of the core component quercetin on core targets were validated using RT-qPCR and Western blot. A total of 163 potential targets were identified by searching for intersections among 7 types of active components and all potential and liver cancer targets. PPI network analysis revealed the core targets IL6 and TNF. GO enrichment analysis involved 2089 biological processes, 76 cellular components, and 196 molecular functions. KEGG enrichment analysis suggested that the anti-cancer effects of HDW might be mediated by the AGE-RAGE signaling pathway, IL-17 signaling pathway, TNF signaling pathway, PI3K-Akt signaling pathway, and NF-κB signaling pathway. Database validation of key targets showed that mRNA and protein expression results for the IL6 gene were contradictory, while those for the TNF gene were consistent, both being underexpressed in liver cancer. Importantly, the expression of IL6 and TNF was related to the infiltration of 24 types of immune cells, with the highest correlation with macrophages. Molecular docking showed that IL6 and TNF had high binding stability with quercetin, with binding energies of - 7.4 and - 6.0 kJ∙mol-1, respectively. Experimental validation showed that quercetin inhibited liver cancer cell proliferation and promoted apoptosis in a dose-dependent manner, with protein results indicating that quercetin downregulated the mRNA and protein expression of IL6 and TNF, and upregulated key proteins in the AGE-RAGE signaling pathway, AGEs, and RAGE. This study comprehensively elucidates the activity, potential targets, and molecular mechanisms of HDW against liver cancer, providing a promising strategy for the scientific basis and treatment mechanism of traditional Chinese medicine in treating liver cancer.
Collapse
Affiliation(s)
- Qingsheng Zheng
- Department of General Surgery, The Affiliated People's Hospital of Fujian University of Traditional Chinese Medicine, 602 Ba Yi Qi Zhong Road, Taijiang District, Fuzhou, 350108, Fujian, China
| | - Xueying Wu
- Department of General Surgery, The Affiliated People's Hospital of Fujian University of Traditional Chinese Medicine, 602 Ba Yi Qi Zhong Road, Taijiang District, Fuzhou, 350108, Fujian, China
| | - Shuai Peng
- Department of General Surgery, The Affiliated People's Hospital of Fujian University of Traditional Chinese Medicine, 602 Ba Yi Qi Zhong Road, Taijiang District, Fuzhou, 350108, Fujian, China.
| |
Collapse
|
9
|
Yu D, Guo F, Zhang Q, Yu H, Wang W, Chen Y. ABCA1 promote tumor environment heterogeneity via epithelial mesenchymal transition in Huh7 and HepG2 liver cancer cell. Front Pharmacol 2024; 15:1498528. [PMID: 39749197 PMCID: PMC11693991 DOI: 10.3389/fphar.2024.1498528] [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: 09/19/2024] [Accepted: 12/03/2024] [Indexed: 01/04/2025] Open
Abstract
In this study, we delve into the intrinsic mechanisms of cell communication in hepatocellular carcinoma (HCC). Initially, employing single-cell sequencing, we analyze multiple malignant cell subpopulations and cancer-associated fibroblast (CAF) subpopulations, revealing their interplay through receptor-ligand interactions, with a particular focus on SPP1. Subsequently, employing unsupervised clustering analysis, we delineate two clusters, C1 and C2, and compare their infiltration characteristics using various tools and metrics, uncovering heightened cytotoxicity and overall invasion abundance in C1. Furthermore, our gene risk scoring model indicates heightened activity of the immune therapeutic pathway in C1. Lastly, employing a formulated scoring system, we stratify patients into high and low-risk groups, revealing notably poorer outcomes in the high-risk cohort on Kaplan-Meier curves. Risk scores exhibit a negative correlation with model genes and immune cell infiltration scores, indicating poor prognosis in the high-risk group. Further characterization elucidates the regulatory landscape of the high and low-risk groups across various signaling pathways. In addition, we used wet lab experiments to prove that ABCA1 plays a pro-oncogenic role in hepatocellular carcinoma cells by promoting proliferation, invasion, migration, and reducing apoptosis. In summary, these findings provide crucial insights, offering valuable clues and references for understanding HCC pathogenesis and patient prognosis.
Collapse
Affiliation(s)
- Dinglai Yu
- Department of Hepatobiliary Pancreatic Surgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Fang Guo
- Department of Gynecology, Wenzhou People’s Hospital, Wenzhou, China
| | - Qiyu Zhang
- Department of Hepatobiliary Pancreatic Surgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Huajun Yu
- Department of Hepatobiliary Pancreatic Surgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Wenmin Wang
- The Yangtze River Delta Biological Medicine Research and Development Center of Zhejiang Province (Yangtze Delta Region Institution of Tsinghua University), Hangzhou, Zhejiang, China
| | - Yunzhi Chen
- Department of Hepatobiliary Pancreatic Surgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
10
|
Montagner A, Arleo A, Suzzi F, D’Assoro AB, Piscaglia F, Gramantieri L, Giovannini C. Notch Signaling and PD-1/PD-L1 Interaction in Hepatocellular Carcinoma: Potentialities of Combined Therapies. Biomolecules 2024; 14:1581. [PMID: 39766289 PMCID: PMC11674819 DOI: 10.3390/biom14121581] [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: 10/01/2024] [Revised: 12/03/2024] [Accepted: 12/06/2024] [Indexed: 01/11/2025] Open
Abstract
Immunotherapy has shown significant improvement in the survival of patients with hepatocellular carcinoma (HCC) compared to TKIs as first-line treatment. Unfortunately, approximately 30% of HCC exhibits intrinsic resistance to ICIs, making new therapeutic combinations urgently needed. The dysregulation of the Notch signaling pathway observed in HCC can affect immune cell response, reducing the efficacy of cancer immunotherapy. Here, we provide an overview of how Notch signaling regulates immune responses and present the therapeutic rationale for combining Notch signaling inhibition with ICIs to improve HCC treatment. Moreover, we propose using exosomes as non-invasive tools to assess Notch signaling activation in hepatic cancer cells, enabling accurate stratification of patients who can benefit from combined strategies.
Collapse
Affiliation(s)
- Annapaola Montagner
- Department of Medical and Surgical Sciences, Bologna University, 40138 Bologna, Italy; (A.A.); (F.S.); (F.P.); (C.G.)
- Department of Oncology, Mayo Clinic College of Medicine, Rochester, MN 55902, USA;
| | - Andrea Arleo
- Department of Medical and Surgical Sciences, Bologna University, 40138 Bologna, Italy; (A.A.); (F.S.); (F.P.); (C.G.)
| | - Fabrizia Suzzi
- Department of Medical and Surgical Sciences, Bologna University, 40138 Bologna, Italy; (A.A.); (F.S.); (F.P.); (C.G.)
| | - Antonino B. D’Assoro
- Department of Oncology, Mayo Clinic College of Medicine, Rochester, MN 55902, USA;
| | - Fabio Piscaglia
- Department of Medical and Surgical Sciences, Bologna University, 40138 Bologna, Italy; (A.A.); (F.S.); (F.P.); (C.G.)
- Division of Internal Medicine, Hepatobiliary and Immunoallergic Diseases, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
| | - Laura Gramantieri
- Division of Internal Medicine, Hepatobiliary and Immunoallergic Diseases, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
| | - Catia Giovannini
- Department of Medical and Surgical Sciences, Bologna University, 40138 Bologna, Italy; (A.A.); (F.S.); (F.P.); (C.G.)
- Division of Internal Medicine, Hepatobiliary and Immunoallergic Diseases, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
| |
Collapse
|
11
|
Abida, Altamimi ASA, Ghaboura N, Balaraman AK, Rajput P, Bansal P, Rawat S, Alanazi FJ, Alruwaili AN, Aldhafeeri NA, Ali H, Deb PK. Therapeutic Potential of lncRNAs in Regulating Disulfidptosis for Cancer Treatment. Pathol Res Pract 2024; 263:155657. [PMID: 39437641 DOI: 10.1016/j.prp.2024.155657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 10/10/2024] [Accepted: 10/13/2024] [Indexed: 10/25/2024]
Abstract
Non-coding RNAs (lncRNAs) play critical roles in various cellular processes, including a novel form of regulated cell death known as disulfidptosis, characterized by accumulating protein disulfide bonds and severe endoplasmic reticulum stress. This review highlights the therapeutic potential of lncRNAs in regulating disulfidptosis for cancer treatment, emphasizing their influence on key pathway components such as GPX4, SLC7A11, and PDIA family members. Recent studies have demonstrated that targeting specific lncRNAs can sensitize cancer cells to disulfidptosis, offering a promising approach to cancer therapy. The regulation of disulfidptosis by lncRNAs involves various signaling pathways, including oxidative stress, ER stress, and calcium signaling. This review also discusses the molecular mechanisms underlying lncRNA regulation of disulfidptosis, the challenges of developing lncRNA-based therapies, and the future potential of this rapidly advancing field in cancer research.
Collapse
Affiliation(s)
- Abida
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia
| | - Abdulmalik S A Altamimi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al Kharj 11942, Saudi Arabia
| | - Nehmat Ghaboura
- Department of Pharmacy Practice, Pharmacy Program, Batterjee Medical College, PO Box 6231, Jeddah 21442, Saudi Arabia
| | - Ashok Kumar Balaraman
- Research and Enterprise, University of Cyberjaya, Persiaran Bestari, Cyber 11, Cyberjaya, Selangor 63000, Malaysia
| | - Pranchal Rajput
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali 140307, Punjab, India.
| | - Pooja Bansal
- Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan 303012, India
| | - Sushama Rawat
- Department of Biotechnology, Graphic Era (Deemed to be University), Clement Town, Dehradun 248002, India
| | - Fadiyah Jadid Alanazi
- Public Health Nursing Department, College of Nursing, Northern Border University, Arar, Saudi Arabia; Center for Health Research, Northern Border University, Arar, Saudi Arabia
| | - Abeer Nuwayfi Alruwaili
- Department of Nursing Administration and Education, College of Nursing, Jouf University, Al Jouf 72388, Saudi Arabia
| | - Nouf Afit Aldhafeeri
- College of Nursing, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia; King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Haider Ali
- Center for Global health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Department of Pharmacology, Kyrgyz State Medical College, Bishkek, Kyrgyzstan
| | - Pran Kishore Deb
- Department of Pharmaceutical Sciences and Technology, Birla Institue of Technology (BIT), Mesra, Ranchi, Jharkhand 835215, India
| |
Collapse
|
12
|
Thangavelu L, Altamimi ASA, Ghaboura N, Babu MA, Roopashree R, Sharma P, Pal P, Choudhary C, Prasad GVS, Sinha A, Balaraman AK, Rawat S. Targeting the p53-p21 axis in liver cancer: Linking cellular senescence to tumor suppression and progression. Pathol Res Pract 2024; 263:155652. [PMID: 39437639 DOI: 10.1016/j.prp.2024.155652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 10/09/2024] [Accepted: 10/11/2024] [Indexed: 10/25/2024]
Abstract
Liver cancer is a major health epidemic worldwide, mainly due to its high mortality rates and limited treatment options. The association of cellular senescence to tumorigenesis and the cancer hallmarks remains a subject of interest in cancer biology. The p53-p21 signalling axis is an important regulator in restoring the cell's balance by supporting tumor suppression and tumorigenesis in liver cancer. We review the novel molecular mechanisms that p53 and its downstream effector, p21, employ to induce cellular senescence, making it last longer, and halt the proliferation of damaged hepatocytes to become tumorous cells. We also examine how dysregulation of this pathway contributes to HCC pathogenesis, proliferation, survival, acquired resistance to apoptosis, and increased invasiveness. Furthermore, we comprehensively describe the molecular cross-talk between the p53-p21 signalling axis and major cell cycle signalling pathways, including Wnt/β-catenin, PI3K/Akt, and TGF-β in liver cancer and provide an overview of promising candidates for chemoprevention and future therapeutic strategies. This review article explores the roles of the p53-p21 pathway in liver cancer, examining its function in promoting cellular senescence under normal conditions and its potential role in cancer progression. It also highlights novel therapeutic drugs and drug targets within the pathway and discusses the implications for treatment strategies and prognosis in liver cancer.
Collapse
Affiliation(s)
- Lakshmi Thangavelu
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Abdulmalik S A Altamimi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam bin Abdulaziz University, Alkharj 11942, Saudi Arabia
| | - Nehmat Ghaboura
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, Jeddah 21442, Saudi Arabia
| | - M Arockia Babu
- Institute of Pharmaceutical Research, GLA UNIVERSITY, Mathura, UP 281406, India.
| | - R Roopashree
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Pawan Sharma
- Department of Sciences, Vivekananda Global University, Jaipur, Rajasthan 303012, India
| | - Pusparghya Pal
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - Chhavi Choudhary
- Chandigarh Pharmacy College, Chandigarh Group of College, Jhanjeri, Mohali, Punjab 140307, India
| | - G V Siva Prasad
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh 531162, India
| | - Aashna Sinha
- School of Applied and Life Sciences, Division of Research and Innovation, Uttaranchal University, Dehradun, India
| | - Ashok Kumar Balaraman
- Research and Enterprise, University of Cyberjaya, Persiaran Bestari, Cyber 11, Cyberjaya, Selangor 63000, Malaysia
| | - Sushama Rawat
- Department of Biotechnology, Graphic Era (Deemed to be University), Clement Town, Dehradun 248002, India
| |
Collapse
|
13
|
Montagna DR, Todero MF, Postma GC, Trigo R, Bernal A, Bustuoabad O, Vermeulen M, Ruggiero R, Duarte A. Resistance against the development of diethylnitrosamine-induced hepatocellular carcinoma in female C3H mice: an experimental model. Exp Anim 2024; 73:399-411. [PMID: 39098024 PMCID: PMC11534494 DOI: 10.1538/expanim.23-0149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 05/24/2024] [Indexed: 08/06/2024] Open
Abstract
Histopathological features of hepatocellular carcinoma (HCC) induced by diethylnitrosamine (DEN) in mice display strong similarities with those seen in humans, including the higher tumor prevalence in males than in females. Previous studies have demonstrated that continual production of the pro-inflammatory IL-6 by Kupffer cells is involved in the initiation and progression of DEN-induced HCC and that estrogen-mediated reduction of IL-6 secretion would decrease its incidence in females. Given the predominant utilization of male mice in hepatic carcinogenesis research, the objective of this study was to examine histopathological and immunological parameters in the DEN-induced liver carcinogenesis model in female C3H mice. We observed a significant prevalence of hepatocellular hyperplasias and adenomas alongside a minimal infiltration of inflammatory cells and a scarcity of senescent areas in females. Further, a low expression of immunosuppression markers is observed in females - such as neutrophil/lymphocyte ratio, PD-1 expression in CD8 T cells, and PD-L1 in myeloid cells - compared to males. Comparative studies between susceptible and resistant hosts to chemical carcinogenesis may help to unveil novel therapeutic strategies against cancer.
Collapse
Affiliation(s)
- Daniela Romina Montagna
- Instituto de Medicina Experimental (IMEX-CONICET), Academia Nacional de Medicina de Buenos Aires, Pacheco de Melo 3081, 1425, Buenos Aires, Argentina
| | - María Florencia Todero
- Instituto de Medicina Experimental (IMEX-CONICET), Academia Nacional de Medicina de Buenos Aires, Pacheco de Melo 3081, 1425, Buenos Aires, Argentina
| | - Gabriela Cintia Postma
- University of Buenos Aires, Faculty of Veterinary Sciences, Department of Pathology, Avenue Chorroarin 280, C1427CWO, Argentina
| | - Roberto Trigo
- University of Buenos Aires, Faculty of Veterinary Sciences, Department of Pathology, Avenue Chorroarin 280, C1427CWO, Argentina
| | - Alan Bernal
- Instituto de Medicina Experimental (IMEX-CONICET), Academia Nacional de Medicina de Buenos Aires, Pacheco de Melo 3081, 1425, Buenos Aires, Argentina
| | - Oscar Bustuoabad
- Instituto de Medicina Experimental (IMEX-CONICET), Academia Nacional de Medicina de Buenos Aires, Pacheco de Melo 3081, 1425, Buenos Aires, Argentina
| | - Mónica Vermeulen
- Instituto de Medicina Experimental (IMEX-CONICET), Academia Nacional de Medicina de Buenos Aires, Pacheco de Melo 3081, 1425, Buenos Aires, Argentina
| | - Raúl Ruggiero
- Instituto de Medicina Experimental (IMEX-CONICET), Academia Nacional de Medicina de Buenos Aires, Pacheco de Melo 3081, 1425, Buenos Aires, Argentina
| | - Alejandra Duarte
- Instituto de Medicina Experimental (IMEX-CONICET), Academia Nacional de Medicina de Buenos Aires, Pacheco de Melo 3081, 1425, Buenos Aires, Argentina
- Fundación Héctor Alejandro (H.A.) Barceló, Instituto Universitario de Ciencias de la Salud, Larrea 770, C1030AAP, Buenos Aires, Argentina
| |
Collapse
|
14
|
Zhang Y, Zhang C, Wu N, Feng Y, Wang J, Ma L, Chen Y. The role of exosomes in liver cancer: comprehensive insights from biological function to therapeutic applications. Front Immunol 2024; 15:1473030. [PMID: 39497820 PMCID: PMC11532175 DOI: 10.3389/fimmu.2024.1473030] [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: 07/30/2024] [Accepted: 09/24/2024] [Indexed: 11/07/2024] Open
Abstract
In recent years, cancer, especially primary liver cancer (including hepatocellular carcinoma and intrahepatic cholangiocarcinoma), has posed a serious threat to human health. In the field of liver cancer, exosomes play an important role in liver cancer initiation, metastasis and interaction with the tumor microenvironment. Exosomes are a class of nanoscale extracellular vesicles (EVs)secreted by most cells and rich in bioactive molecules, including RNA, proteins and lipids, that mediate intercellular communication during physiological and pathological processes. This review reviews the multiple roles of exosomes in liver cancer, including the initiation, progression, and metastasis of liver cancer, as well as their effects on angiogenesis, epithelial-mesenchymal transformation (EMT), immune evasion, and drug resistance. Exosomes have great potential as biomarkers for liver cancer diagnosis and prognosis because they carry specific molecular markers that facilitate early detection and evaluation of treatment outcomes. In addition, exosomes, as a new type of drug delivery vector, have unique advantages in the targeted therapy of liver cancer and provide a new strategy for the treatment of liver cancer. The challenges and prospects of exosome-based immunotherapy in the treatment of liver cancer were also discussed. However, challenges such as the standardization of isolation techniques and the scalability of therapeutic applications remain significant hurdles.
Collapse
Affiliation(s)
- Yinghui Zhang
- College of Rehabilitation Medicine, Henan University of Traditional Chinese Medicine, Zhengzhou, Henan, China
| | - Congcong Zhang
- College of Rehabilitation Medicine, Henan University of Traditional Chinese Medicine, Zhengzhou, Henan, China
| | - Nan Wu
- College of Rehabilitation Medicine, Henan University of Traditional Chinese Medicine, Zhengzhou, Henan, China
| | - Yuan Feng
- College of Rehabilitation Medicine, Henan University of Traditional Chinese Medicine, Zhengzhou, Henan, China
| | - Jiayi Wang
- College of Rehabilitation Medicine, Henan University of Traditional Chinese Medicine, Zhengzhou, Henan, China
| | - Liangliang Ma
- Rehabilitation Center, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Yulong Chen
- College of Rehabilitation Medicine, Henan University of Traditional Chinese Medicine, Zhengzhou, Henan, China
| |
Collapse
|
15
|
Liu T, Meng G, Ma S, You J, Yu L, He R, Zhao X, Cui Y. Progress of immune checkpoint inhibitors in the treatment of advanced hepatocellular carcinoma. Front Immunol 2024; 15:1455716. [PMID: 39185414 PMCID: PMC11341420 DOI: 10.3389/fimmu.2024.1455716] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 07/30/2024] [Indexed: 08/27/2024] Open
Abstract
Among primary liver cancers, hepatocellular carcinoma is the most common pathological type. Its onset is insidious, and most patients have no obvious discomfort in the early stage, so it is found late, and the opportunity for surgical radical treatment is lost, resulting in a poor prognosis. With the introduction of molecular-targeted drugs represented by sorafenib, patients with middle- and late-stage liver cancer have regained the light of day. However, their therapeutic efficacy is relatively low due to the limited target of drug action, toxic side effects, and other reasons. At this time, the emergence of immunotherapy represented by immune checkpoint inhibitors (ICIs) well breaks this embarrassing situation, which mainly achieves the anti-tumor purpose by improving the tumor immune microenvironment. Currently, ICI monotherapy, as well as combination therapy, has been widely used in the clinic, further prolonging the survival of patients with advanced hepatocellular carcinoma. This article reviews the development of monotherapy and combination therapy for ICIs in advanced hepatocellular carcinoma and the latest research progress.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Yunfu Cui
- Department of Hepatopancreatobiliary Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
16
|
Zhang Y, Xu W, Peng C, Ren S, Zhang C. Intricate effects of post-translational modifications in liver cancer: mechanisms to clinical applications. J Transl Med 2024; 22:651. [PMID: 38997696 PMCID: PMC11245821 DOI: 10.1186/s12967-024-05455-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Accepted: 06/29/2024] [Indexed: 07/14/2024] Open
Abstract
Liver cancer is a significant global health challenge, with hepatocellular carcinoma (HCC) being the most prevalent form, characterized by high incidence and mortality rates. Despite advances in targeted therapies and immunotherapies, the prognosis for advanced liver cancer remains poor. This underscores the urgent need for a deeper understanding of the molecular mechanisms underlying HCC to enable early detection and the development of novel therapeutic strategies. Post-translational modifications (PTMs) are crucial regulatory mechanisms in cellular biology, affecting protein functionality, interactions, and localization. These modifications, including phosphorylation, acetylation, methylation, ubiquitination, and glycosylation, occur after protein synthesis and play vital roles in various cellular processes. Recent advances in proteomics and molecular biology have highlighted the complex networks of PTMs, emphasizing their critical role in maintaining cellular homeostasis and disease pathogenesis. Dysregulation of PTMs has been associated with several malignant cellular processes in HCC, such as altered cell proliferation, migration, immune evasion, and metabolic reprogramming, contributing to tumor growth and metastasis. This review aims to provide a comprehensive understanding of the pathological mechanisms and clinical implications of various PTMs in liver cancer. By exploring the multifaceted interactions of PTMs and their impact on liver cancer progression, we highlight the potential of PTMs as biomarkers and therapeutic targets. The significance of this review lies in its potential to inform the development of novel therapeutic approaches and improve prognostic tools for early intervention in the fight against liver cancer.
Collapse
Affiliation(s)
- Yu Zhang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Weihao Xu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Chuanhui Peng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Shenli Ren
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Cheng Zhang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| |
Collapse
|