1
|
Li J, Gu J, Pan S, Deng N, Khan M, Li L, Wu X, Li Y. Synergic effect of the combination of isoliquiritigenin and arsenic trioxide in HepG2 liver cancer cells. Cell Signal 2025; 131:111752. [PMID: 40107478 DOI: 10.1016/j.cellsig.2025.111752] [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: 10/21/2024] [Revised: 02/06/2025] [Accepted: 03/16/2025] [Indexed: 03/22/2025]
Abstract
Despite continuous therapeutic interventions, the prognosis of hepatocellular carcinoma (HCC) remains very poor. Thus, quest for novel treatment strategies to improve therapeutic window of HCC therapy is paramount. Arsenic trioxide (ATO) is commonly used as the first-line treatment for acute promyelocytic leukemia (APL). Isoliquiritigenin (ISL) is a potential plant-based bioactive molecule with versatile biological and pharmacological effects including anticancer effect. The present study aimed to investigate the potential synergistic effects of combination of ISL and ATO in HCC cells. The data revealed that the combination of ISL and ATO synergistically inhibited HCC cell proliferation. The collective data demonstrate that synergistic anticancer effect of combined treatment of ISL + ATO was achieved via cooperative induction of mitochondrial apoptosis through ROS generation and inhibition of PI3K/Akt/mTOR pathway. In addition, ROS generation and suppression of PI3K/Akt/mTOR pathway were found to be two independent events in induction of apoptosis. Finally, we observed that combination treatment effectively suppressed tumor growth in nude mice xenograft model through induction of intrinsic apoptosis and inhibition of PI3K/Akt/mTOR pathway. In conclusion, the findings of this study suggest that both drugs work synergistically to exert anti-tumor effect in HCC, both in-vitro and in-vivo and could offer novel strategy for liver cancer treatment.
Collapse
Affiliation(s)
- Jingjing Li
- School of Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Juan Gu
- School of Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Sijia Pan
- School of Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Nuo Deng
- School of Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Muhammad Khan
- Cancer Research Lab, Institute of Zoology, University of the Punjab, Quaid-e-Azam Campus, Lahore, Pakistan.
| | - Lingyan Li
- School of Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xiao Wu
- School of Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Yongming Li
- School of Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
2
|
Huang X, Feng L, Lu X, Yang F, Liu S, Wei X, Huang J, Wang Y, Huang D, Huang T. Development and optimization of a self micro-emulsifying drug delivery system (SMEDDS) for co-administration of sorafenib and curcumin. Drug Deliv Transl Res 2025; 15:1609-1625. [PMID: 39207633 DOI: 10.1007/s13346-024-01699-3] [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] [Accepted: 08/06/2024] [Indexed: 09/04/2024]
Abstract
In this study, we developed a novel co-administration of curcumin and sorafenib using a Self micro-emulsifying Drug Delivery System (SMEDDS) called Sorafenib-Curcumin Self micro-emulsifying Drug Delivery System (SOR-CUR-SMEDDS). The formulation was optimized using star point design-response surface methodology, and in vitro cellular experiments were conducted to evaluate the delivery ratio and anti-tumor efficacy of the curcumin and sorafenib combination. The SOR-CUR-SMEDDS exhibited a small size distribution of 13.48 ± 0.61 nm, low polydispersity index (PDI) of 0.228 ± 0.05, and negative zeta potential (ZP) of - 12.4 mV. The half maximal inhibitory concentration (IC50) of the SOR-CUR-SMEDDS was 3-fold lower for curcumin and 5-fold lower for sorafenib against HepG2 cells (human hepatocellular carcinoma cells). Transmission electron microscopy (TEM) and particle size detection confirmed that the SOR-CUR-SMEDDS droplets were uniformly round and within the nano-emulsion particle size range of 10-20 nm. The SMEDDS were characterized then studied for drug release in vitro via dialysis membranes. Curcumin was released more completely in the combined delivery system, showing the largest in vitro drug release (79.20%) within 7 days in the medium, while the cumulative release rate of sorafenib in the release medium was low, reaching 58.96% on the 7 day. In vitro pharmacokinetic study, it demonstrated a significant increase in oral bioavailability of sorafenib (1239.88-fold) and curcumin (3.64-fold) when administered in the SMEDDS. These findings suggest that the SMEDDS formulation can greatly enhance drug solubility, improve drug absorption and prolong circulation in vivo, leading to increased oral bioavailability of sorafenib and curcumin.
Collapse
Affiliation(s)
- Xingzhen Huang
- School of Pharmacy, Guangxi Medical University, Nanning, Guangxi, 530000, PR China.
- Guangxi Key Laboratory of Bioactive Molecules Research and Evaluation, Nanning, Guangxi, 530000, PR China.
| | - Lizhen Feng
- School of Pharmacy, Guangxi Medical University, Nanning, Guangxi, 530000, PR China
| | - Xuefang Lu
- Department of Pharmacy, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530000, PR China
| | - Fan Yang
- Hechi Food and Drug Inspection Institute, Hechi, Guangxi, 547000, PR China
| | - Shengjun Liu
- School of Pharmacy, Guangxi Medical University, Nanning, Guangxi, 530000, PR China
| | - Xueqian Wei
- Hechi Food and Drug Inspection Institute, Hechi, Guangxi, 547000, PR China
| | - Jinping Huang
- School of Pharmacy, Guangxi Medical University, Nanning, Guangxi, 530000, PR China
| | - Yao Wang
- School of Pharmacy, Guangxi Medical University, Nanning, Guangxi, 530000, PR China
| | - Dongyi Huang
- School of Pharmacy, Guangxi Medical University, Nanning, Guangxi, 530000, PR China
| | - Tingting Huang
- School of Pharmacy, Guangxi Medical University, Nanning, Guangxi, 530000, PR China
| |
Collapse
|
3
|
Wang X, Li J, Nong J, Deng X, Chen Y, Han B, Zeng L, Huang X. MiR-518b Promotes the Tumorigenesis of Hepatocellular Carcinoma by Targeting EGR1 to Regulate PI3K/AKT/mTOR Signaling Pathway. Cell Biochem Biophys 2025:10.1007/s12013-025-01752-z. [PMID: 40221539 DOI: 10.1007/s12013-025-01752-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/02/2025] [Indexed: 04/14/2025]
Abstract
Hepatocellular carcinoma (HCC) is a prevalent malignancy originating from hepatocytes and is characterized by high invasiveness and fatality. Dysregulation of microRNAs (miRNAs) is frequently observed during HCC progression. This study aimed to investigate the role of miR-518b in HCC cell malignancy and tumor growth. MiR-518b expression in HCC cells was measured by RT-qPCR. The proliferative, migratory and invasive capabilities of Hep3B and SNU-387 were assessed by colony formation, wound healing and transwell assays, respectively. RNA immunoprecipitation and luciferase reporter assays were utilized to verify the binding between miR-518b and its target gene, early growth response factor 1 (EGR1). Results revealed that miR-518b was highly expressed while EGR1 was downregulated in HCC cells. Knockdown of miR-518b significantly repressed cell proliferation, migration and invasion. Moreover, miR-518b bound to 3'untranslated region of EGR1 and negatively regulated its expression in HCC cells. EGR1 knockdown counteracted the inhibitory impact of miR-518b inhibition on malignant cell behaviors. In addition, the silencing of EGR1 activated the PI3K/AKT/mTOR signaling in HCC cells, while miR-518b depletion had the opposite effect. Importantly, the suppressive impact of miR-518b on the pathway was rescued by EGR1 knockdown. In vivo experiments demonstrated that inhibition of miR-518b suppressed HCC tumor growth, reduced EGR1 and Ki67 (a proliferation marker) expression, and inactivated the PI3K/AKT/mTOR signaling. In conclusion, miR-518b promotes HCC tumorigenesis by targeting EGR1 and regulating the PI3K/AKT/mTOR signaling pathway.
Collapse
Affiliation(s)
- Xinyuan Wang
- College of Zhuang Medicine, Guangxi University of Chinese Medicine, Nanning, China
| | - Juan Li
- Department of pediatrics, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, China
| | - Jiao Nong
- College of Osteopathy, Guangxi University of Chinese Medicine, Nanning, China
| | - Xin Deng
- School of basic medicine, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, China
| | - Yiping Chen
- Emergency department, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, China
| | - Bing Han
- Guangxi University of Chinese Medicine, Nanning, China
| | - Lin Zeng
- Guangxi University of Chinese Medicine, Nanning, China
| | - Xiabing Huang
- Emergency department, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, China.
| |
Collapse
|
4
|
Nagashima S, Kobayashi S, Tsunoda S, Yamachika Y, Tozuka Y, Fukushima T, Morimoto M, Ueno M, Furuse J, Maeda S. A case of complete remission by cabozantinib as an end-line treatment for advanced hepatocellular carcinoma. Clin J Gastroenterol 2025; 18:125-129. [PMID: 39616585 PMCID: PMC11785699 DOI: 10.1007/s12328-024-02062-2] [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: 07/31/2024] [Accepted: 11/07/2024] [Indexed: 02/01/2025]
Abstract
Cabozantinib is a multi-kinase inhibitor targeting multiple tyrosine kinases. It improves overall survival and progression-free survival in patients previously treated with sorafenib for advanced hepatocellular carcinoma (HCC) compared to the placebo in the phase 3 CELESTIAL trial. A 71-year-old man presented to our hospital for treatment of HCC with chronic hepatitis C. He was refractory to sorafenib, lenvatinib, regorafenib, and ramucirumab and started atezolizumab and bevacizumab therapy in November 2020. After administering the second cycle on December 10, 2020, the patient was diagnosed with progressive disease in January 2021. Therefore, cabozantinib (60 mg/day) was initiated on January 14, 2021. As the grade 3 aspartate aminotransferase and alanine aminotransferase levels increased, grade 3 anorexia and a decline in performance status were observed in the first week, and cabozantinib was terminated. His performance status and anorexia gradually improved, and contrast-enhanced computed tomography (CT) in June 2021 showed complete remission (CR) according to the modified Response Evaluation Criteria in Solid Tumors. The patient did not show disease progression for 11 months without receiving any treatment for HCC. To the best of our knowledge, this is the first report of CR with cabozantinib in advanced HCC.
Collapse
Affiliation(s)
- Shuhei Nagashima
- Department of Gastroenterology, Kanagawa Cancer Center, 2-3-2 Nakao Asahi-ku, Yokohama, 241-8515, Japan.
- Department of Gastroenterology, Yokohama City University Graduate School of Medicine, 3-9, Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan.
| | - Satoshi Kobayashi
- Department of Gastroenterology, Kanagawa Cancer Center, 2-3-2 Nakao Asahi-ku, Yokohama, 241-8515, Japan
| | - Shotaro Tsunoda
- Department of Gastroenterology, Kanagawa Cancer Center, 2-3-2 Nakao Asahi-ku, Yokohama, 241-8515, Japan
| | - Yui Yamachika
- Department of Gastroenterology, Kanagawa Cancer Center, 2-3-2 Nakao Asahi-ku, Yokohama, 241-8515, Japan
| | - Yuichiro Tozuka
- Department of Gastroenterology, Kanagawa Cancer Center, 2-3-2 Nakao Asahi-ku, Yokohama, 241-8515, Japan
| | - Taito Fukushima
- Department of Gastroenterology, Kanagawa Cancer Center, 2-3-2 Nakao Asahi-ku, Yokohama, 241-8515, Japan
| | - Manabu Morimoto
- Department of Gastroenterology, Kanagawa Cancer Center, 2-3-2 Nakao Asahi-ku, Yokohama, 241-8515, Japan
| | - Makoto Ueno
- Department of Gastroenterology, Kanagawa Cancer Center, 2-3-2 Nakao Asahi-ku, Yokohama, 241-8515, Japan
| | - Junji Furuse
- Department of Gastroenterology, Kanagawa Cancer Center, 2-3-2 Nakao Asahi-ku, Yokohama, 241-8515, Japan
| | - Shin Maeda
- Department of Gastroenterology, Yokohama City University Graduate School of Medicine, 3-9, Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| |
Collapse
|
5
|
Jiang X, Tao L, Cao S, Xu Z, Zheng S, Zhang H, Xu X, Qu X, Liu X, Yu J, Chen X, Wu J, Liang X. Porous Silicon Particle-Assisted Mass Spectrometry Technology Unlocks Serum Metabolic Fingerprints in the Progression From Chronic Hepatitis B to Hepatocellular Carcinoma. ACS APPLIED MATERIALS & INTERFACES 2025; 17:5893-5908. [PMID: 39812132 DOI: 10.1021/acsami.4c17563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
Hepatocellular carcinoma (HCC) is a common malignancy and generally develops from liver cirrhosis (LC), which is primarily caused by the chronic hepatitis B (CHB) virus. Reliable liquid biopsy methods for HCC screening in high-risk populations are urgently needed. Here, we establish a porous silicon-assisted laser desorption ionization mass spectrometry (PSALDI-MS) technology to profile metabolite information hidden in human serum in a high throughput manner. Serum metabolites can be captured in the pore channel of APTES-modified porous silicon (pSi) particles and well-preserved during storage or transportation. Furthermore, serum metabolites captured in the APTES-pSi particles can be directly detected on the LDI-MS without the addition of an organic matrix, thus greatly accelerating the acquisition of metabolic fingerprints of serum samples. The PSALDI-MS displays the capability of high throughput (5 min per 96 samples), high reproducibility (coefficient of variation <15%), high sensitivity (LOD ∼ 1 pmol), and high tolerance to background salt and proteins. In a multicenter cohort study, 1433 subjects including healthy controls (HC), CHB, LC, and HCC volunteers were enrolled and nontargeted serum metabolomic analysis was performed on the PSALDI-MS platform. After the selection of feature metabolites, a stepwise diagnostic model for the classification of different liver disease stages was constructed by the machine learning algorithm. In external testing, the accuracy of 91.2% for HC, 71.4% for CHB, 70.0% for LC, and 95.3% for HCC was achieved by chemometrics. Preliminary studies indicated that the diagnostic model constructed from serum metabolic fingerprint also displays good predictive performance in a prospective observation. We believe that the combination of PSALDI-MS technology and machine learning may serve as an efficient tool in clinical practice.
Collapse
Affiliation(s)
- Xinrong Jiang
- Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China
- Institution of Analytical Chemistry, Department of Chemistry, Zhejiang University, Hangzhou, Zhejiang 310058, China
- Biomedical Research Center, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China
| | - Liye Tao
- Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China
- Zhejiang Key Laboratory of Multi-omics Precision Diagnosis and Treatment of Liver Diseases, Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China
| | - Shuo Cao
- Institution of Analytical Chemistry, Department of Chemistry, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Zhengao Xu
- Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China
- Zhejiang Key Laboratory of Multi-omics Precision Diagnosis and Treatment of Liver Diseases, Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China
| | - Shuang Zheng
- Taizhou First People's Hospital, Taizhou, Zhejiang 318020, China
| | - Huafang Zhang
- Wuyi First People's Hospital, Jinhua, Zhejiang 321200, China
| | - Xinran Xu
- Institution of Analytical Chemistry, Department of Chemistry, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Xuetong Qu
- Institution of Analytical Chemistry, Department of Chemistry, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Xingyue Liu
- Institution of Analytical Chemistry, Department of Chemistry, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Jiekai Yu
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, China
| | - Xiaoming Chen
- Zhejiang Key Laboratory of Multi-omics Precision Diagnosis and Treatment of Liver Diseases, Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China
- Well-healthcare Technologies Co., Hangzhou, Zhejiang 310051, China
| | - Jianmin Wu
- Zhejiang Key Laboratory of Multi-omics Precision Diagnosis and Treatment of Liver Diseases, Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China
- Institution of Analytical Chemistry, Department of Chemistry, Zhejiang University, Hangzhou, Zhejiang 310058, China
- Children's Hospital, School of Medicine, Zhejiang University, Hangzhou 310052, China
| | - Xiao Liang
- Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China
- Zhejiang Key Laboratory of Multi-omics Precision Diagnosis and Treatment of Liver Diseases, Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China
- School of medicine, Shaoxing University, Shaoxing, Zhejiang 312000, China
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang 310000, China
| |
Collapse
|
6
|
Rahmani D, Taheri RA, Moosazadeh Moghaddam M. Targeted delivery of curcumin and CM11 peptide against hepatocellular carcinoma cells based on binding affinity of PreS1-coated chitosan nanoparticles to SB3 protein. Amino Acids 2025; 57:12. [PMID: 39862295 PMCID: PMC11762422 DOI: 10.1007/s00726-024-03438-x] [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] [Accepted: 12/18/2024] [Indexed: 01/27/2025]
Abstract
In recent years, the use of cationic peptides as alternative drugs with anticancer activity has received attention. In this study, the targeted release of curcumin (Cur) and CM11 peptide alone and together against hepatocellular carcinoma (HCC) was evaluated using chitosan nanoparticles (CS NPs) coated with Pres1 that target the SB3 antigen of HCC cells (PreS1-Cur-CM11-CS NPs). SB3 protein is the specific antigen of HCC and the PreS1 peptide is a part of the hepatitis B antigen, which can specifically bind to the SB3 protein. Chitosan was used to prepare NPs. To Cur and CM11 loading, drugs were added to the CS solution in appropriate concentrations. Pres1 was coupled to the surface of the NPs using EDC catalyst to target NPs against HepG2 cells. SEM and DLS analysis confirmed that the PreS1-Cur-CM11-CS NPs had a size of about 132 nm, the ideal size for penetrating the cell membrane. The loading of Cur and CM11 was equal to 87% and 65%, respectively, which had a sustained and better release in the acidic environment than in the physiological environment. The MTT assay showed that PreS1-Cur-CM11-CS NPs act in a targeted and specific manner with the highest toxicity on the HepG2 cells compared to the control by a decrease in viability of about 26% after 48 h based on cell apoptosis. The results showed that PreS1-Cur-CM11-CS NPs are capable of targeted and specific drug release against HepG2 cancer cells and have significant potential to fight this cancer.
Collapse
Affiliation(s)
- Danial Rahmani
- Tissue Engineering and Regenerative Medicine Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Ramezan Ali Taheri
- Nanobiotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mehrdad Moosazadeh Moghaddam
- Tissue Engineering and Regenerative Medicine Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
7
|
Yang S, Zou Y, Zhong C, Zhou Z, Peng X, Tang C. Dual role of pyroptosis in liver diseases: mechanisms, implications, and therapeutic perspectives. Front Cell Dev Biol 2025; 13:1522206. [PMID: 39917567 PMCID: PMC11798966 DOI: 10.3389/fcell.2025.1522206] [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: 11/04/2024] [Accepted: 01/10/2025] [Indexed: 02/09/2025] Open
Abstract
Pyroptosis, a form of programmed cell death induced by inflammasome with a mechanism distinct from that of apoptosis, occurs via one of the three pathway types: classical, non-classical, and granzyme A/B-dependent pyroptosis pathways. Pyroptosis is implicated in various diseases, notably exhibiting a dual role in liver diseases. It facilitates the clearance of damaged hepatocytes, preventing secondary injury, and triggers immune responses to eliminate pathogens and damaged cells. Conversely, excessive pyroptosis intensifies inflammatory responses, exacerbates hepatocyte damage and promotes the activation and proliferation of hepatic stellate cells, accelerating liver fibrosis. Furthermore, by sustaining an inflammatory state, impacts the survival and proliferation of cancer cells. This review comprehensively summarizes the dual role of pyroptosis in liver diseases and its therapeutic strategies, offering new theoretical foundations and practical guidance for preventing and treating of liver diseases.
Collapse
Affiliation(s)
| | | | | | - Zuoqiong Zhou
- State Key Laboratory of Developmental Biology of Freshwater Fish, Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, College of Physical Education, Hunan Normal University, Changsha, China
| | - Xiyang Peng
- State Key Laboratory of Developmental Biology of Freshwater Fish, Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, College of Physical Education, Hunan Normal University, Changsha, China
| | - Changfa Tang
- State Key Laboratory of Developmental Biology of Freshwater Fish, Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, College of Physical Education, Hunan Normal University, Changsha, China
| |
Collapse
|
8
|
Hakoda H, Ichida A, Hasegawa K. Advances in systemic therapy leading to conversion surgery for advanced hepatocellular carcinoma. Biosci Trends 2025; 18:525-534. [PMID: 39647858 DOI: 10.5582/bst.2024.01372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/10/2024]
Abstract
Recently, a systemic therapy for advanced hepatocellular carcinoma (HCC) has been developed. The regimen for unresectable HCC varies and includes single or multi-tyrosine kinase inhibitors, monoclonal antibodies, immune checkpoint inhibitors, or their combinations. Treatment with these agents begins with sorafenib as the first-line drug for unresectable HCC. Subsequently, several systemic therapies, including lenvatinib, ramucirumab, cabozantinib, and regorafenib have been investigated and established. With advances in systemic therapy for unresectable HCC, the prognosis of patients with unresectable HCC has improved significantly than previously. Conversion surgery, consisting of systemic therapy and surgery, showed the possibility of improving the prognosis than systemic therapy alone. Although a combination of atezolizumab and bevacizumab is mostly used for initially unresectable HCC to conduct conversion surgery because of the high response rate and fewer adverse events compared to others, many trials are being conducted to assess their efficacy for initially unresectable HCC. However, the appropriate timing of surgery and interval between systemic therapy and surgery remain controversial. To address these issues, a multidisciplinary team can play a vital role in determining the strategies for treating unresectable HCC. This review describes previous and current trends in the treatment of HCC, with a particular focus on conversion surgery for initially unresectable HCC.
Collapse
Affiliation(s)
- Hiroyuki Hakoda
- Hepato-Biliary-Pancreatic Surgery Division, Department of Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Akihiko Ichida
- Hepato-Biliary-Pancreatic Surgery Division, Department of Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kiyoshi Hasegawa
- Hepato-Biliary-Pancreatic Surgery Division, Department of Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
9
|
Saraswat I, Goel A, Gupta J. An In-depth Review on Argemone mexicana in the Management of Liver Health and Liver Cancer. Anticancer Agents Med Chem 2025; 25:24-34. [PMID: 39225208 DOI: 10.2174/0118715206307964240821051756] [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: 03/11/2024] [Revised: 05/25/2024] [Accepted: 07/23/2024] [Indexed: 09/04/2024]
Abstract
INTRODUCTION Argemone mexicana, commonly known as the Mexican prickly poppy, has been historically employed in traditional medicine for various ailments, including liver disorders. Given the rising prevalence of liver diseases, including cancer, investigating the potential efficacy of Argemone mexicana in promoting liver health is of paramount importance. This review aims to provide a comprehensive analysis of the existing literature on the hepatoprotective and anticancer properties of Argemone mexicana. METHODOLOGY A systematic literature search was conducted across PubMed, Google Scholar, and relevant botanical and pharmacological databases. Studies from various sources, including in vitro experiments, animal models, and clinical trials, were included in the review. The search focused on articles published up to 2010-2023, encompassing research that explored the botanical characteristics, chemical composition, traditional uses, and pharmacological properties of Argemone mexicana, specifically emphasizing its impact on liver health and cancer. RESULTS The review revealed a wealth of studies highlighting the diverse pharmacological properties of Argemone mexicana. The botanical composition includes compounds with antioxidant and anti-inflammatory potential, suggesting hepatoprotective effects. Studies using in vitro and in vivo models demonstrated promising outcomes regarding liver function improvement and inhibition of liver cancer cell proliferation. While some clinical studies supported the traditional uses of Argemone mexicana, further well-designed trials are warranted to establish its clinical efficacy. CONCLUSION In conclusion, Argemone mexicana shows promise as a natural agent for promoting liver health and combating liver cancer. Bioactive compounds with antioxidant and anti-inflammatory properties suggest potential hepatoprotective effects. However, translating these findings into clinical practice requires further rigorous investigation, including well-designed clinical trials. This review provides a foundation for future research efforts aimed at elucidating the full therapeutic potential of Argemone mexicana in liver health and cancer management.
Collapse
Affiliation(s)
- Istuti Saraswat
- Department of Biotechnology, GLA University, 17km Stone, NH-2 Mathura-Delhi Road Mathura, Chaumuhan, Mathura, Uttar Pradesh, India
| | - Anjana Goel
- Department of Biotechnology, GLA University, 17km Stone, NH-2 Mathura-Delhi Road Mathura, Chaumuhan, Mathura, Uttar Pradesh, India
| | - Jyoti Gupta
- Department of Biotechnology, GLA University, 17km Stone, NH-2 Mathura-Delhi Road Mathura, Chaumuhan, Mathura, Uttar Pradesh, India
| |
Collapse
|
10
|
Wu Q, Chen Q, Yang J, Zhang J, Yang A. Material basis revelation of anti-hepatoma effect of Huachansu (Cinobufacini) through down-regulation of thymidylate synthase. CHINESE HERBAL MEDICINES 2025; 17:127-138. [PMID: 39949800 PMCID: PMC11814253 DOI: 10.1016/j.chmed.2024.04.002] [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: 05/06/2023] [Revised: 01/11/2024] [Accepted: 04/30/2024] [Indexed: 02/16/2025] Open
Abstract
Objective Hepatocellular carcinoma (HCC) is a leading cause of mortality worldwide. Huachansu (Cinobufacini) is active extract isolated from the dry skin of Bufo Bufo gargarizans. It has now been widely used in clinical treatment of cancer, this study is to clarify the material basis of down-regulation of thymidylate synthase (TYMS) induced by Huachansu. Methods Our study utilized UPLC-MS/MS to identify major bioactive components from Huachansu. Cell Counting Kit 8 (CCK-8) assay and clone formation assay were used to examine the cell viability of tumor cells. TYMS and γ-H2AX level were detected by using quantitative real-time RT-PCR and/or western blotting. Small interfering RNA (siRNA) transfection was used to explore whether inhibition of TYMS could enhance the suppressive effect of Huachansu on cell growth of HCC cells. Results In our study, firstly, we identify 21 major bioactive components from Huachansu. CCK-8 assay results showed that Huachansu and its bioactive bufadienolides (Bufalin, Bufotalin, Cinobufotalin, Desacetylcinobufagin, Arenobufagin, Telocinobufagin, and Resibufogenin) significantly inhibited the proliferation of HepG2 and SK-HEP-1 cells in a dose- and time-dependent manner. Further molecular mechanistic investigation demonstrates that Huachansu significantly suppresses thymidylate synthase (TYMS), the enzyme which provides the sole de novo source of thymidylate for DNA synthesis. The inhibition of TYMS could lead to cell-cycle block and DNA damage of HCC cells. Furthermore, we identified that Huachansu markedly increased γ-H2AX expression, which indicated the presence of DNA damage. Moreover, we confirmed that transfection of cells with small interfering RNA specific to TYMS could increase the suppressive effects of Huachansu on the HCC cells proliferation. Quantitative RT-PCR analysis showed that Huachansu treatment had no effect on the transcription level of TYMS. Furthermore, proteasomal inhibitor MG132 could block TYMS inhibition induced by Huachansu, and concomitant administration of protein synthesis inhibitor cycloheximide (CHX) with Huachansu could further suppress the protein level of TYMS, indicating that Huachansu promotes proteasome-dependent degradation of TYMS in liver cancer cells. More importantly, the bioactive bufadienolides of Huachansu such as Bufalin, Bufotalin, Cinobufotalin, Desacetylcinobufagin, Arenobufagin, Telocinobufagin, and Resibufogenin could also significantly restrain the protein level of TYMS, revealing the material basis of inhibition of TYMS exposed to Huachansu. 5-Fluorouracil (5-FU) is a TYMS inhibitor, we also evaluate the effects of the combined treatment of Huachansu with 5-FU, the results show that interactions between Huachansu and 5-FU are synergistic or antagonistic. Thus, in clinical, attention should be paid to the dosage of Huachansu in combination with 5-FU. Conclusion Huachansu inhibits the growth and induces DNA damage of human HCC cells through proteasome-dependent degradation of TYMS, bioactive bufadienolides are the material basis of down-regulation of TYMS induced by Huachansu.
Collapse
Affiliation(s)
- Qi Wu
- School of Pharmacy, Binzhou Medical University, Yantai 264003, China
| | - Qimei Chen
- School of Pharmacy, Binzhou Medical University, Yantai 264003, China
| | - Jingyi Yang
- School of Pharmacy, Binzhou Medical University, Yantai 264003, China
| | - Jiayu Zhang
- School of Pharmacy, Binzhou Medical University, Yantai 264003, China
| | - Ailin Yang
- School of Pharmacy, Binzhou Medical University, Yantai 264003, China
| |
Collapse
|
11
|
Suh JY, Sim DY, Ahn CH, Park SY, Shim BS, Kim B, Lee DY, Jeong HB, Lee HE, Kim SH. Crucial Role of c-Myc/Monocarboxylate Transporter 4 Signaling in Capsaicin Induced Apoptotic and Anti-Warburg Effects in Hepatocellular Carcinoma. Phytother Res 2025; 39:536-547. [PMID: 39655472 DOI: 10.1002/ptr.8388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 10/10/2024] [Accepted: 10/29/2024] [Indexed: 01/30/2025]
Abstract
Though Capsaicin from chili peppers was known to have antitumor effects in several cancers, the underlying antitumor pathogenesis of Capsaicin is not clear to date. Thus, the antitumor mechanism of Capsaicin was explored in Hep3B and Huh7 hepatocellular carcinoma (HCC) cells in relation to c-Myc/monocarboxylate transporter 4 (MCT4) signaling. To elucidate the antitumor mechanism of capsaicin, cytotoxicity assay, cell cycle analysis, Western blotting, RT-qPCR, RNA interference, ELISA, immunoprecipitation, and mouse xenograft model were used in this work. Capsaicin increased the cytotoxicity, subG1 population, and the number of TUNEL-positive bodies in Huh7 and Hep3B cells. Consistently, Capsaicin diminished the expression of pro-PARP, HK2, PKM2, LDHA, glucose transporter type 1 (Glut1), c-Myc, and monocarboxylate transporter 4 (MCT4) in Huh7 and Hep3B cells, along with decreased production of glucose, lactate, and ATP. However, a glycolysis end product pyruvate treatment reversed the capacity of Capsaicin to attenuate the expression of pro-PARP, HK2, c-Myc, and MCT4 in Hep3B cells. Furthermore, Capsaicin reduced c-Myc stability in the presence of cycloheximide and induced c-Myc ubiquitination in Hep3B cells, while c-Myc directly binds to MCT4 as a lactate transporter and downstream of c-Myc in Hep3B cells by immunoprecipitation and correlation factor (Spearman efficient = 0.0027). Furthermore, a preliminary analysis of an animal study reveals that Capsaicin significantly suppressed the growth of Hep3B cells inoculated in BALB/c nude mice without hurting body weight, liver, and spleen. Our findings provide novel evidence that Capsaicin exerts apoptotic and anti-Warburg effect via c-Myc/MCT4 signaling axis as a potent anticancer candidate for liver cancer therapy.
Collapse
Affiliation(s)
- Jin Young Suh
- College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Deok Yong Sim
- College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Chi-Hoon Ahn
- College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Su-Yeon Park
- College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Bum-Sang Shim
- College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Bonglee Kim
- College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Dae Young Lee
- BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Hyo Bong Jeong
- Department of Horticultural Crop Research, National Institute of Horticultural and Herbal Science, Rural Development Administration, Wanju, Republic of Korea
| | - Hye Eun Lee
- Department of Horticultural Crop Research, National Institute of Horticultural and Herbal Science, Rural Development Administration, Wanju, Republic of Korea
| | - Sung-Hoon Kim
- College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| |
Collapse
|
12
|
Ozbay MF, Harputluoglu H, Karaca M, Tekin O, Şendur MAN, Kaplan MA, Sahin B, Geredeli C, Teker F, Tural D, Saglam S, Çil T, Bilici A, Erol C, Kalkan Z, Bayram E, Selvi O, Gültürk İ, Göksu SS, Tatlı AM. Sequential Use of Sorafenib and Regorafenib in Hepatocellular Cancer Recurrence After Liver Transplantation: Treatment Strategies and Outcomes. Cancers (Basel) 2024; 16:3880. [PMID: 39594835 PMCID: PMC11592833 DOI: 10.3390/cancers16223880] [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: 10/24/2024] [Revised: 11/14/2024] [Accepted: 11/16/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND AND AIMS During liver transplantation, hepatocellular carcinoma (HCC) recurrence remains a critical challenge for patient survival. Targeted therapies, such as sorafenib and regorafenib, have been utilized to manage relapsed HCC in this unique setting. This study aimed to assess the efficacy of Sorafenib and Regorafenib in patients with HCC who experienced recurrence after liver transplantation. We focused on survival outcomes, treatment responses, and the management of side effects in this patient group. METHODS We conducted a retrospective analysis of 73 patients who experienced HCC recurrence post-liver transplantation between 2012 and 2022 across 11 oncology centers in Turkey. Patients were categorized according to Child-Pugh classification and treated with sorafenib as first-line therapy and Regorafenib in case of progression. Survival rates were analyzed using the Kaplan-Meier method, and risk factors were evaluated using Cox regression analysis. RESULTS Of the 73 patients included in the study, 62 were male (84.9%), and 11 were female (15.1%), with a mean age of 61.5 ± 10.9 years. All patients received sorafenib as first-line treatment. Among patients who experienced progression with sorafenib or discontinued treatment due to toxicity, 45.2% (n = 33) continued treatment with regorafenib. The median progression-free survival (PFS1) time with sorafenib was 5.6 months, and the one-year survival rate was 24.3%. The median progression-free survival (PFS2) time with regorafenib, which was administered as second-line treatment, was also calculated as 5.9 months. Overall survival (OS) duration was determined as 35.9 months. The most common side effects associated with both drugs included fatigue, hand and foot syndrome, and hypertension. Significantly better survival outcomes were shown in the Child-Pugh A group compared to other patients. CONCLUSIONS These results suggest that Sorafenib and Regorafenib treatments offer a survival advantage in patients with relapsed HCC post-transplantation. However, individualized treatment strategies and close follow-up are crucial for optimizing outcomes. Further studies are needed to refine therapeutic protocols and enhance the care of this specific patient group.
Collapse
Affiliation(s)
- Mehmet Fatih Ozbay
- Department of Medical Oncology, Kırsehir Training and Research Hospital, Kirsehir 40200, Turkey;
| | - Hakan Harputluoglu
- Department of Medical Oncology, Faculty of Medicine, Inonu University, Malatya 44000, Turkey
| | - Mustafa Karaca
- Department of Medical Oncology, Faculty of Medicine, Akdeniz University, Antalya 07070, Turkey; (S.S.G.); (A.M.T.)
| | - Omer Tekin
- Department of Medical Oncology, Faculty of Medicine, Inonu University, Malatya 44000, Turkey
| | - Mehmet Ali Nahit Şendur
- Department of Medical Oncology, Republic of Turkey Ministry of Health Ankara Bilkent City Hospital, Ankara 06800, Turkey
| | - Muhammed Ali Kaplan
- Department of Medical Oncology, Faculty of Medicine, Dicle University, Diyarbakır 21280, Turkey
| | - Berksoy Sahin
- Department of Medical Oncology, Faculty of Medicine, Çukurova University, Adana 01330, Turkey
| | - Caglayan Geredeli
- Department of Medical Oncology, Health Sciences University Okmeydanı Training and Research Hospital, Istanbul 34098, Turkey
| | - Fatih Teker
- Department of Medical Oncology, Faculty of Medicine, Gaziantep University, Gaziantep 27410, Turkey
| | - Deniz Tural
- Department of Medical Oncology, Bakırköy Sadi Konuk Training and Research Hospital, Istanbul 34147, Turkey
| | - Sezer Saglam
- Department of Medical Oncology, Demiroglu Bilim University Gayrettepe Florence Nightingale Hospital, Istanbul 34394, Turkey
| | - Timuçin Çil
- Department of Medical Oncology, Adana City Training and Research Hospital, Adana Faculty of Medicine, University of Health Sciences, Adana 01230, Turkey
| | - Ahmet Bilici
- Department of Medical Oncology, Istanbul Medipol University, Istanbul 34815, Turkey
| | - Cihan Erol
- Department of Medical Oncology, Republic of Turkey Ministry of Health Ankara Bilkent City Hospital, Ankara 06800, Turkey
| | - Ziya Kalkan
- Department of Medical Oncology, Faculty of Medicine, Dicle University, Diyarbakır 21280, Turkey
| | - Ertugrul Bayram
- Department of Medical Oncology, Faculty of Medicine, Çukurova University, Adana 01330, Turkey
| | - Oguzhan Selvi
- Department of Medical Oncology, Health Sciences University Okmeydanı Training and Research Hospital, Istanbul 34098, Turkey
| | - İlkay Gültürk
- Department of Medical Oncology, Bakırköy Sadi Konuk Training and Research Hospital, Istanbul 34147, Turkey
| | - Sema Sezgin Göksu
- Department of Medical Oncology, Faculty of Medicine, Akdeniz University, Antalya 07070, Turkey; (S.S.G.); (A.M.T.)
| | - Ali Murat Tatlı
- Department of Medical Oncology, Faculty of Medicine, Akdeniz University, Antalya 07070, Turkey; (S.S.G.); (A.M.T.)
| |
Collapse
|
13
|
Anwar J, Arslan HM, Sarfraz Z, Shuroog J, Abdelhakeem A, Saeed A, Saeed A. Immunotherapy Responses in Viral Hepatitis-Induced HCC: A Systematic Review and Meta-Analysis. Curr Oncol 2024; 31:7204-7225. [PMID: 39590162 PMCID: PMC11592516 DOI: 10.3390/curroncol31110532] [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: 09/02/2024] [Revised: 11/03/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024] Open
Abstract
Background: Hepatocellular carcinoma (HCC) is a prevalent liver cancer with poor prognosis, often linked to hepatitis B (HBV) and C (HCV) infections. This meta-analysis evaluates the efficacy of immunotherapy in HCC, particularly in cases arising from viral hepatitis. Methods: In adherence to PRISMA Statement 2020 guidelines, the immunotherapeutic outcomes comprised objective response rate (ORR), progression-free survival (PFS), and overall survival (OS). Data were analyzed from randomized controlled trials up to April 2024 using the fixed-effects models in R (V.4.3.3.) and RevMan (Cochrane). Results: This study included 9 trials with 5316 patients. The ORR was slightly higher in the viral group at 27.93% compared to 24.07% in the non-viral group, though this difference was not significant (p = 0.15). Viral HCC patients exhibited a median PFS of 7.3 months (IQR: 6.2-8.4) compared to 5.8 months (IQR: 5.48-6.13) in non-viral patients, a significant improvement (p = 0.005). Similarly, median OS was longer in the viral group at 16.8 months (IQR: 12.99-20.61) versus 15.2 months (IQR: 13.25-17.15) for non-viral HCC, which was also significant (p < 0.0001). The median OS for viral HCC was 16.8 months (IQR: 14.11-19.49 months), with HBV patients experiencing slightly higher survival at 17.15 months (IQR: 14.3-20 months) compared to 16.8 months (IQR: 12.99-20.61 months) for HCV patients; this difference was not statistically significant (p = 0.89). Conclusions: Immunotherapy shows potential in treating HCC, with significantly better outcomes in viral HCC, particularly HBV-associated cases. The heterogeneity highlights the need for personalized treatment approaches based on the viral background of HCC patients. Further research should aim to optimize these therapies to improve survival rates.
Collapse
Affiliation(s)
- Junaid Anwar
- Department of Medicine, Baptist Hospitals of Southeast Texas, Beaumont, TX 77701, USA;
| | - Hafiz Muhammad Arslan
- Department of Medicine, Lincoln Medical and Mental Health Center, Bronx, NY 10451, USA;
| | - Zouina Sarfraz
- Department of Medicine, Fatima Jinnah Medical University, Lahore 54000, Pakistan;
| | - Juwairiya Shuroog
- Department of Medicine, TidalHealth Peninsula Regional, Salisbury, MD 21801, USA;
| | - Ahmed Abdelhakeem
- Department of Medicine, Division of Hematology & Oncology, Mayo Clinic College of Medicine and Science, Jacksonville, FL 32224, USA;
| | - Ali Saeed
- Department of Medicine, Ochsner Lafayette General Medical Center, Lafayette, LA 70503, USA;
| | - Anwaar Saeed
- Department of Medicine, Division of Hematology & Oncology, University of Pittsburgh Medical Center, Pittsburgh, PA 15232, USA
| |
Collapse
|
14
|
Wang Y, Wang Q, Tao S, Li H, Zhang X, Xia Y, Wang Y, Yang C, Sui C. Identification of SPP1 + macrophages in promoting cancer stemness via vitronectin and CCL15 signals crosstalk in liver cancer. Cancer Lett 2024; 604:217199. [PMID: 39216547 DOI: 10.1016/j.canlet.2024.217199] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 08/14/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024]
Abstract
Macrophages play a multifaceted role in cancer biology, with both pro-tumorigenic and anti-tumorigenic functions. Understanding the mechanisms underlying macrophage involvement in cancer progression is essential for the development of therapeutic strategies. Our study analyzed single-cell RNA sequencing data from 12 patients with liver cancer and identified a subpopulation of macrophages characterized by elevated expression of SPP1, which correlates with poor prognosis in liver cancer patients. These SPP1+ macrophages induce upregulation of tumor stemness through a vitronectin (VTN)-dependent paracrine mechanism. Mechanistically, VTN derived from SPP1+ macrophages promote integrin αvβ5/adenosine 5'-monophosphate-activated protein kinase (AMPK)/Yes-associated protein 1 (YAP1)/SYR-box transcription factor 4 (SOX4) signaling, mediating liver tumor stemness and progression. Conversely, CCL15 produced by liver cancer cells drives polarization of M0 macrophages toward an SPP1+ macrophage phenotype, establishing a positive feedback loop of macrophage-tumor stemness. Furthermore, the presence of SPP1+ macrophages confers chemoresistance in liver cancer, and inhibition of the macrophage-tumor feedback loop through targeting integrin αvβ5/YAP1 signaling sensitizes liver cancer cells to chemotherapy. Our study highlights the crucial role of SPP1+ macrophages in liver cancer progression, providing novel insights for clinical liver cancer therapy.
Collapse
Affiliation(s)
- Yizhou Wang
- Department of Hepatic Surgery IV, The Eastern Hepatobiliary Surgery Hospital, Third Affiliated Hospital of Naval Medical University, Shanghai, 200438, PR China; Eastern Hepatobiliary Clinical Research Institute, Third Affiliated Hospital of Navy Medical University, Shanghai, 200438, PR China.
| | - Qing Wang
- Department of Oncology, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, PR China.
| | - Shuangfen Tao
- Department of Oncology, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, PR China.
| | - Haoyu Li
- Department of Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, PR China.
| | - Xiaofeng Zhang
- Department of Hepatic Surgery IV, The Eastern Hepatobiliary Surgery Hospital, Third Affiliated Hospital of Naval Medical University, Shanghai, 200438, PR China; Eastern Hepatobiliary Clinical Research Institute, Third Affiliated Hospital of Navy Medical University, Shanghai, 200438, PR China.
| | - Yong Xia
- Department of Hepatic Surgery IV, The Eastern Hepatobiliary Surgery Hospital, Third Affiliated Hospital of Naval Medical University, Shanghai, 200438, PR China; Eastern Hepatobiliary Clinical Research Institute, Third Affiliated Hospital of Navy Medical University, Shanghai, 200438, PR China.
| | - Yue Wang
- Department of Stem Cell and Regeneration Medicine, Translational Medicine Research Center, Naval Medical University, Shanghai, 200433, PR China; Department of Histology and Embryology, Basic Medicine Collage, Naval Medical University, Shanghai, 200433, PR China; Shanghai Key Laboratory of Cell Engineering, Shanghai, 200062, PR China; Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, 200092, PR China.
| | - Cheng Yang
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, PR China; Shanghai GoBroad Cancer Hospital, China Pharmaceutical University, Shanghai, 200131, PR China.
| | - Chengjun Sui
- Department of Special Treatment, Eastern Hepatobiliary Surgery Hospital, Third Affiliated Hospital of Naval Medical University, Shanghai, 200438, PR China.
| |
Collapse
|
15
|
Shetty M, Shenoy S, Amuthan A, Devi V, Kumar N, Kiran A, Shenoy G, Chinta DR, Prasada K S, Shetty A, Rao K G M. Kadukkai maathirai (Indian herbal drug) prevents hepatocellular cancer progression by enhancing GSTM1 expression and modulating β catenin transcription: in-silico and in-vivo study. F1000Res 2024; 13:639. [PMID: 39916986 PMCID: PMC11800331 DOI: 10.12688/f1000research.145961.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/09/2024] [Indexed: 02/09/2025] Open
Abstract
Background Hepatocellular carcinoma (HCC) is an aggressive malignancy with poor clinical outcomes. Hence cost-effective drugs with fewer side effects as a standard supportive therapy might yield substantial advantages in efficacy and safety. Kadukkai maathirai (KM) is being used as a supplement in hepatocellular carcinoma. We evaluated whether KM has any preventive action on cancer progression in diethyl nitrosamine (DEN) - induced HCC in rats. Methods DEN was injected to produce HCC in rats, which was confirmed after 16 weeks. All the rats were orally administered KM for 4 weeks. Hepatoprotective potential (serum AST, ALT, ALP, Bilirubin) and anticancer efficacy (body weight, nodule count, tumor progression by histopathology, expression of GSTM1 by Liquid chromatography-mass spectrometry (LC-MS), and In-silico analysis of phytoconstituents against β catenin and LRP analysis were evaluated. Results KM prevented cancer progression against DEN-induced HCC by an increase in GSTM1, a phase II detoxifying enzyme. It significantly reversed altered nodule count, relative liver weight, body weight, and histopathological features of HCC. In silico analysis of phytoconstituents of KM showed that they modulate the intracellular transcription process by inhibiting the armadillo repeat region of β catenin. Conclusions Our results elucidate the potential of KM as a supplement in HCC by reducing nodule count, protecting the liver from further damage, GSTM1 expression, and inhibiting armadillo repeat region of β catenin.
Collapse
Affiliation(s)
- Manjunath Shetty
- Centre Of Excellence, Ocular Nanoscience, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
- Division of Pharmacology, Department of Basic Medical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Smita Shenoy
- Department of Pharmacology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Arul Amuthan
- Division of Pharmacology, Department of Basic Medical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Vasudha Devi
- Department of Pharmacology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Nitesh Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Vaishali, Bihar, 844102, India
| | - Amruth Kiran
- Division of Pharmacology, Department of Basic Medical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Ganesh Shenoy
- Division of Pharmacology, Department of Basic Medical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Diya Rajasekhar Chinta
- Department of Pharmacology, Manipal University College Malaysia, Bukit Baru, Melaka, 75150, Malaysia
| | - Shama Prasada K
- Department of Cell and Molecular Biology, School of Life Sciences, Manipal Academy of Higher Education, Manipal, Manipal, Karnataka, 576104, India
| | - Akshatha Shetty
- Department of Research and Development, Muniyal Institute of Ayurveda and Medical Sciences, Manipal, Manipal, Karnataka, 576104, India
| | - Mohandas Rao K G
- Division of Anatomy, Department of Basic Medical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| |
Collapse
|
16
|
Zhang Y, Tang X, Liu L, Cai D, Gou S, Hao S, Li Y, Shen J, Chen Y, Zhao Y, Wu X, Li M, Chen M, Li X, Sun Y, Gu L, Li W, Wang F, Zhang Z, Wang X, Deng S, Xiao Z, Yao L, Du F. GLO1 regulates hepatocellular carcinoma proliferation and migration through the cell cycle pathway. BMC Cancer 2024; 24:1297. [PMID: 39434012 PMCID: PMC11492659 DOI: 10.1186/s12885-024-12927-x] [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: 07/06/2023] [Accepted: 09/10/2024] [Indexed: 10/23/2024] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a malignant tumor characterized by a high mortality rate. The occurrence and progression of HCC are linked to oxidative stress. Glyoxalase-1 (GLO1) plays an important role in regulating oxidative stress, yet the underlying mechanism remains unclear. GLO1 may serve as a prognostic biomarker and therapeutic target for HCC. METHODS Based on TCGA database hepatocellular carcinoma samples, we conducted a bioinformatics analysis to explore the correlation between GLO1 expression and HCC cell proliferation and viability. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis revealed that differentially expressed genes (DEGs) were mainly enriched in the cell cycle pathway. We analyzed the relationships between GLO1 and 24 genes enriched in the cell cycle pathway using a protein-protein interaction (PPI) network. Finally, experimental validation was performed to assess GLO1's impact on the distribution of cells at different cell cycle stages and on the proliferation and migration of HCC cells. RESULTS Our study demonstrated that GLO1 was overexpressed in HCC tissues and was associated with a poor prognosis. Data analysis indicated that overexpression of GLO1 activated the cell cycle pathway and positively correlated with expression of the majority of key cell cycle genes. Experimental validation showed that GLO1 expression affects the number of HCC cells in G2 and S phases and regulates HCC cell proliferation and migration. CONCLUSIONS GLO1 represents a promising therapeutic target for HCC, providing valuable insights into its role in the viability and proliferation of HCC cells.
Collapse
Affiliation(s)
- Yao Zhang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, 646000, China
| | - Xiaolong Tang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, 646000, China
| | - Lin Liu
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, 646000, China
| | - Dan Cai
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, 646000, China
| | - Shuang Gou
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, 646000, China
| | - Siyu Hao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, 646000, China
| | - Yan Li
- Public Center of Experimental Technology, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Jing Shen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, 646000, China
| | - Yu Chen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, 646000, China
| | - Yueshui Zhao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, 646000, China
| | - Xu Wu
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, 646000, China
| | - Mingxing Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, 646000, China
| | - Meijuan Chen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Xiaobing Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Yuhong Sun
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Li Gu
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Wanping Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Fang Wang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Zhuo Zhang
- Key Laboratory of Luzhou City for Aging Medicine, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Xiaodong Wang
- Department of Hepatobiliary Disease, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Shuai Deng
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, 646000, China
| | - Zhangang Xiao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China.
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, 646000, China.
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, 646000, China.
- Department of Oncology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China.
| | - Lei Yao
- The Key Laboratory for Human Disease Gene Study of Sichuan Province and Department of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
- Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences, Chengdu, 610072, China.
| | - Fukuan Du
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China.
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, 646000, China.
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, 646000, China.
| |
Collapse
|
17
|
Fan M, Hu J, Xu X, Chen J, Zhang W, Zheng X, Pan J, Xu W, Feng S. Mass spectrometry-based multi-omics analysis reveals distinct molecular features in early and advanced stages of hepatocellular carcinoma. Heliyon 2024; 10:e38182. [PMID: 39381095 PMCID: PMC11456867 DOI: 10.1016/j.heliyon.2024.e38182] [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: 02/04/2024] [Revised: 09/18/2024] [Accepted: 09/19/2024] [Indexed: 10/10/2024] Open
Abstract
Hepatocellular Carcinoma (HCC) is a serious primary solid tumor that is prevalent worldwide. Due to its high mortality rate, it is crucial to explore both early diagnosis and advanced treatment for HCC. In recent years, multi-omics approaches have emerged as promising tools to identify biomarkers and investigate molecular mechanisms of biological processes and diseases. In this study, we performed proteomics, phosphoproteomics, metabolomics, and lipidomics to reveal the molecular features of early- and advanced-stage HCC. The data obtained from these omics were analyzed separately and then integrated to provide a comprehensive understanding of the disease. The multi-omics results unveiled intricate biological pathways and interaction networks underlying the initiation and progression of HCC. Moreover, we proposed specific potential biomarker panels for both early- and advanced-stage HCC by overlapping our data with CPTAC database for HCC diagnosis, and deduced novel insights and mechanisms related to HCC origination and development, such as glucose depletion during tumor progression, ROCK1 deactivation and GSK3A activation.
Collapse
Affiliation(s)
- Mingzhu Fan
- Key Laboratory of Structural Biology of Zhejiang Province, Westlake University, Hangzhou, 310024, Zhejiang, China
- Mass Spectrometry & Metabolomics Core Facility, The Biomedical Research Core Facility, Westlake University, Hangzhou, 310024, Zhejiang, China
| | - Jin Hu
- Key Laboratory of Structural Biology of Zhejiang Province, Westlake University, Hangzhou, 310024, Zhejiang, China
- Mass Spectrometry & Metabolomics Core Facility, The Biomedical Research Core Facility, Westlake University, Hangzhou, 310024, Zhejiang, China
| | - Xiaoyan Xu
- Mass Spectrometry & Metabolomics Core Facility, The Biomedical Research Core Facility, Westlake University, Hangzhou, 310024, Zhejiang, China
| | - Jia Chen
- Mass Spectrometry & Metabolomics Core Facility, The Biomedical Research Core Facility, Westlake University, Hangzhou, 310024, Zhejiang, China
| | - Wenwen Zhang
- Mass Spectrometry & Metabolomics Core Facility, The Biomedical Research Core Facility, Westlake University, Hangzhou, 310024, Zhejiang, China
| | - Xiaoping Zheng
- Pathology Department, Shulan (Hangzhou) Hospital, Hangzhou, 311112, Zhejiang, China
| | - Jinheng Pan
- Key Laboratory of Structural Biology of Zhejiang Province, Westlake University, Hangzhou, 310024, Zhejiang, China
| | - Wei Xu
- College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
- Hangzhou Tongchuang Medical Laboratory, Shulan Health Group, Hangzhou, 310015, Zhejiang, China
| | - Shan Feng
- Key Laboratory of Structural Biology of Zhejiang Province, Westlake University, Hangzhou, 310024, Zhejiang, China
- Mass Spectrometry & Metabolomics Core Facility, The Biomedical Research Core Facility, Westlake University, Hangzhou, 310024, Zhejiang, China
| |
Collapse
|
18
|
Devan AR, Nair B, Pradeep GK, Alexander R, Vinod BS, Nath LR, Calina D, Sharifi-Rad J. The role of glypican-3 in hepatocellular carcinoma: Insights into diagnosis and therapeutic potential. Eur J Med Res 2024; 29:490. [PMID: 39369212 PMCID: PMC11453014 DOI: 10.1186/s40001-024-02073-2] [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: 05/10/2024] [Accepted: 09/22/2024] [Indexed: 10/07/2024] Open
Abstract
Glypican-3 (GPC-3) is predominantly found in the placenta and fetal liver, with limited expression in adult tissues. Its re-expression in hepatocellular carcinoma (HCC) and secretion into the serum highlights its potential as a diagnostic marker. GPC-3 is involved in important cellular processes such as proliferation, metastasis, apoptosis, and epithelial-mesenchymal transition through various signaling pathways including Wnt, IGF, YAP, and Hedgehog. To review the structure, biosynthesis, and post-translational modifications of GPC-3, and to elucidate its signaling mechanisms and role as a pro-proliferative protein in HCC, emphasizing its diagnostic and therapeutic potential. A comprehensive literature review was conducted, focusing on the expression of GPC-3 in various tumors, with a special emphasis on HCC. The review synthesized findings from experimental studies and clinical trials, analyzing the overexpression of GPC-3 in HCC, its differentiation from other liver diseases, and its potential as a diagnostic and therapeutic target. GPC-3 overexpression in HCC is linked to aggressive tumor behavior and poor prognosis, including shorter overall and disease-free survival. Additionally, GPC-3 has emerged as a promising therapeutic target. Ongoing investigations, including immunotherapies such as monoclonal antibodies and CAR-T cell therapies, demonstrate potential in inhibiting tumor growth and improving clinical outcomes. The review details the multifaceted roles of GPC-3 in tumorigenesis, including its impact on tumor-associated macrophages, glucose metabolism, and epithelial-mesenchymal transition, all contributing to HCC progression. GPC-3's re-expression in HCC and its involvement in key tumorigenic processes underscore its value as a biomarker for early diagnosis and a target for therapeutic intervention. Further research is warranted to fully exploit GPC-3's diagnostic and therapeutic potential in HCC management.
Collapse
Affiliation(s)
- Aswathy R Devan
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Ponekkara P.O., Kochi, Kerala, 682041, India
| | - Bhagyalakshmi Nair
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Ponekkara P.O., Kochi, Kerala, 682041, India
| | - Govind K Pradeep
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Ponekkara P.O., Kochi, Kerala, 682041, India
| | - Roshini Alexander
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Ponekkara P.O., Kochi, Kerala, 682041, India
| | - Balachandran S Vinod
- Department of Biochemistry, Sree Narayana College, Kollam, Kerala, 691001, India
| | - Lekshmi R Nath
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Ponekkara P.O., Kochi, Kerala, 682041, India.
| | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, 200349, Craiova, Romania.
| | - Javad Sharifi-Rad
- Universidad Espíritu Santo, Samborondón, 092301, Ecuador.
- Department of Medicine, College of Medicine, Korea University, Seoul, 02841, Republic of Korea.
| |
Collapse
|
19
|
Afyouni S, Zandieh G, Nia IY, Pawlik TM, Kamel IR. State-of-the-art imaging of hepatocellular carcinoma. J Gastrointest Surg 2024; 28:1717-1725. [PMID: 39117267 DOI: 10.1016/j.gassur.2024.08.001] [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: 05/07/2024] [Revised: 07/20/2024] [Accepted: 08/01/2024] [Indexed: 08/10/2024]
Abstract
Hepatocellular carcinoma (HCC) is the third most fatal and fifth most common cancer worldwide, with rising incidence due to obesity and nonalcoholic fatty liver disease. Imaging modalities, including ultrasound (US), multidetector computed tomography (MDCT), and magnetic resonance imaging (MRI) play a vital role in detecting HCC characteristics, aiding in early detection, detailed visualization, and accurate differentiation of liver lesions. Liver-specific contrast agents, the Liver Imaging Reporting and Data System, and advanced techniques, including diffusion-weighted imaging and artificial intelligence, further enhance diagnostic accuracy. This review emphasizes the significant role of imaging in managing HCC, from diagnosis to treatment assessment, without the need for invasive biopsies.
Collapse
Affiliation(s)
- Shadi Afyouni
- Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Ghazal Zandieh
- Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Iman Yazdani Nia
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, United States
| | - Timothy M Pawlik
- Department of Surgery, The Ohio State University, Wexner Medical Center, The James Comprehensive Cancer Center, Columbus, OH, United States
| | - Ihab R Kamel
- Department of Radiology, University of Colorado School of Medicine, Aurora, CO, United States.
| |
Collapse
|
20
|
Zhang Q, Yu M, Yang L, Sun D. MiR-875-5p suppresses Gli1 to alter the hedgehog signaling pathway, which in turn has hepatocellular cancer-related tumor suppressing properties. Heliyon 2024; 10:e37771. [PMID: 39381215 PMCID: PMC11459020 DOI: 10.1016/j.heliyon.2024.e37771] [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: 12/03/2023] [Revised: 09/08/2024] [Accepted: 09/09/2024] [Indexed: 10/10/2024] Open
Abstract
Background One of the most prevalent cancers worldwide is HCC, which has put patient health at risk. Increasing evidence indicated that messenger RNAs (mRNAs) played significant roles in modulating tumorigenesis. It has been established that Gli1 acts as an oncogene in a number of malignancies. However, more research was necessary to understand the Gli1 regulation mechanism in HCC. Methods Microarray technology was used to evaluate the expression of mRNAs. RT-qPCR was utilized to evaluate Gli1 and miR-875-5p expression. To investigate the role of Gli1, tests using CCK-8, EdU, transwell, immunofluorescence, and Western blot analysis was performed. RIP, RNA pull down, and luciferase reporter assays were employed to verify the interaction between Gli1 and miR-875-5p. Results In tissues and cells of HCC, Gli1 expression appeared to be upregulated, especially in metastatic samples and advanced stages of the disease. A worse outcome was predicted by elevated Gli1 expression. Additionally, in HCC, Gli1 inhibition impeded the growth, migration, and development of the EMT. Since miR-875-5p was shown to have a molecular target in Gli1, miR-875-5p mediated the negative regulation of Gli1. In HCC tissues, its expression pattern was less prominent. In HCC tissues, there was an inverse relationship between Gli1 expression and miR-875-5p expression. Overexpressing Gli1 helped to partially counteract the suppression of HCC migration, proliferation, and EMT formation by miR-875-5p overexpression. Conclusions MiR-875-5p in HCC suppresses tumors by downregulating Gli1, which supplies a novel treatment for HCC patients.
Collapse
Affiliation(s)
- Qi Zhang
- Department of Pathology, Central Hospital Affiliated to Jiangnan University, Wuxi Clinical College of Nantong University, Wuxi, 214002, Jiangsu Province, China
| | - Miao Yu
- Department of Clinical Laboratory, Central Hospital Affiliated to Jiangnan University, Wuxi Clinical College of Nantong University, Wuxi, 214002, Jiangsu Province, China
| | - Leilei Yang
- Department of Hepatobiliary Surgery, Central Hospital Affiliated to Jiangnan University, Wuxi Clinical College of Nantong University, Wuxi, 214002, Jiangsu Province, China
| | - Defeng Sun
- Department of Hepatobiliary Surgery, Central Hospital Affiliated to Jiangnan University, Wuxi Clinical College of Nantong University, Wuxi, 214002, Jiangsu Province, China
| |
Collapse
|
21
|
Xiong Y, Zheng Y, Long W, Wang Y, Wang Q, You Y, Zhou Y, Zhong J, Ge Y, Li Y, Huang Y, Zhou Z. Study on microwave ablation temperature prediction model based on grayscale ultrasound texture and machine learning. PLoS One 2024; 19:e0308968. [PMID: 39321182 PMCID: PMC11423965 DOI: 10.1371/journal.pone.0308968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Accepted: 08/03/2024] [Indexed: 09/27/2024] Open
Abstract
BACKGROUND Temperature prediction is crucial in the clinical ablation treatment of liver cancer, as it can be used to estimate the coagulation zone of microwave ablation. METHODS Experiments were conducted on 83 fresh ex vivo porcine liver tissues at two ablation powers of 15 W and 20 W. Ultrasound grayscale images and temperature data from multiple sampling points were collected. The machine learning method of random forests was used to train the selected texture features, obtaining temperature prediction models for sampling points and the entire ultrasound imaging area. The accuracy of the algorithm was assessed by measuring the area of the hyperechoic area in the porcine liver tissue cross-section and ultrasound grayscale images. RESULTS The model exhibited a high degree of accuracy in temperature prediction and the identification of coagulation zone. Within the test sets for the 15 W and 20 W power groups, the average absolute error for temperature prediction was 1.14°C and 4.73°C, respectively. Notably, the model's accuracy in measuring the area of coagulation was higher than that of traditional ultrasonic grey-scale imaging, with error ratios of 0.402 and 0.182 for the respective power groups. Additionally, the model can filter out texture features with a high correlation to temperature, providing a certain degree of interpretability. CONCLUSION The temperature prediction model proposed in this study can be applied to temperature monitoring and coagulation zone range assessment in microwave ablation.
Collapse
Affiliation(s)
- Yan Xiong
- Nanjing University of Chinese Medicine, Nanjing, China
| | - Yi Zheng
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Science, Suzhou, China
| | - Wei Long
- Nanjing University of Chinese Medicine, Nanjing, China
| | - Yuxin Wang
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Science, Suzhou, China
| | - Qin Wang
- Nanjing University of Chinese Medicine, Nanjing, China
| | - Yi You
- Nanjing University of Chinese Medicine, Nanjing, China
| | - Yuheng Zhou
- Nanjing University of Chinese Medicine, Nanjing, China
| | - Jiang Zhong
- Department of Ultrasound, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Yunxi Ge
- Department of Ultrasound, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Youchen Li
- Department of Ultrasound, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Yan Huang
- Department of Ultrasound, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhiyong Zhou
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Science, Suzhou, China
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Suzhou, China
| |
Collapse
|
22
|
Cao LQ, Xie Y, Fleishman JS, Liu X, Chen ZS. Hepatocellular carcinoma and lipid metabolism: Novel targets and therapeutic strategies. Cancer Lett 2024; 597:217061. [PMID: 38876384 DOI: 10.1016/j.canlet.2024.217061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 05/10/2024] [Accepted: 06/11/2024] [Indexed: 06/16/2024]
Abstract
Hepatocellular carcinoma (HCC) is an increasingly prevalent disease that is associated with high and continually rising mortality rates. Lipid metabolism holds a crucial role in the pathogenesis of HCC, in which abnormalities pertaining to the delicate balance of lipid synthesis, breakdown, and storage, predispose for the pathogenesis of the nonalcoholic fatty liver disease (NAFLD), a disease precursor to HCC. If caught early enough, HCC treatment may be curative. In later stages, treatment is only halting the inevitable outcome of death, boldly prompting for novel drug discovery to provide a fighting chance for this patient population. In this review, we begin by providing a summary of current local and systemic treatments against HCC. From such we discuss hepatic lipid metabolism and highlight novel targets that are ripe for anti-cancer drug discovery. Lastly, we provide a targeted summary of current known risk factors for HCC pathogenesis, providing key insights that will be essential for rationalizing future development of anti-HCC therapeutics.
Collapse
Affiliation(s)
- Lu-Qi Cao
- Institute for Biotechnology, St. John's University, New York, NY, 11439, USA; College of Pharmacy and Health Sciences, St. John's University, New York, NY, 11439, USA
| | - Yuhao Xie
- College of Pharmacy and Health Sciences, St. John's University, New York, NY, 11439, USA
| | - Joshua S Fleishman
- College of Pharmacy and Health Sciences, St. John's University, New York, NY, 11439, USA
| | - Xuan Liu
- Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, 518034, China.
| | - Zhe-Sheng Chen
- Institute for Biotechnology, St. John's University, New York, NY, 11439, USA; College of Pharmacy and Health Sciences, St. John's University, New York, NY, 11439, USA.
| |
Collapse
|
23
|
Mao T, Zhang M, Peng Z, Tang M, Li T, Liang C. Integrative analysis of ferroptosis-related genes reveals that ABHD12 is a novel prognostic biomarker and facilitates hepatocellular carcinoma tumorigenesis. Discov Oncol 2024; 15:330. [PMID: 39093379 PMCID: PMC11297018 DOI: 10.1007/s12672-024-01211-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 07/31/2024] [Indexed: 08/04/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is a highly heterogeneous disease, making the prognostic prediction challenging. Ferroptosis, an iron-dependent form of cell death, is a key regulator in the initiation, progression, and metastasis of HCC. However, the expression and function of ferroptosis-related genes (FRGs) in HCC remained largely unclear. In this study, we analyzed TCGA datasets and identified 58 survival-related deferentially expressed FRGs (DE-FRGs). Then, based on the results of LASSO analysis, we developed a novel prognostic model based on 12 survival-related DE-FRGs. Survival assays indicated a strong prognostic ability of this new model in predicting clinical prognosis of HCC patients. In addition, we conducted an exploration of molecular subtypes related to HCC and delved into the associated immune characteristics and gene expression patterns. Among the 12 survival-related DE-FRGs, our attention focused on ABHD12 (abhydrolase domain containing 12) which was highly expressed in HCC and associated with advanced clinical stages. Multivariate assays confirmed that ABHD12 was a significant prognostic factor for HCC patients. Immune analysis revealed that ABHD12 may play an important role in tumor microenvironment. Finally, we performed RT-PCR and confirmed that ABHD12 was highly expressed in HCC cells. Functional experiments revealed that ABHD12 knockdown may suppress the proliferation and migration of HCC cells. These findings emphasized the significance of ABHD12 as a potential prognostic marker for HCC and its crucial role in the field of tumor biology. Additionally, the study introduces a novel survival model that holds promise for enhancing prognostic predictions in HCC patients. Overall, this research provided valuable insights for a deeper comprehension of the complexity of HCC and the development of personalized treatment strategies.
Collapse
Affiliation(s)
- Tiantao Mao
- Department of Oncology, Wuxi County People's Hospital, No. 100 Wantong Road, Baiyang Street, Chongqing, 405899, China
| | - Maosong Zhang
- Department of Oncology, Wuxi County People's Hospital, No. 100 Wantong Road, Baiyang Street, Chongqing, 405899, China
| | - Zupei Peng
- Department of Oncology, Wuxi County People's Hospital, No. 100 Wantong Road, Baiyang Street, Chongqing, 405899, China
| | - Min Tang
- Department of Oncology, Wuxi County People's Hospital, No. 100 Wantong Road, Baiyang Street, Chongqing, 405899, China.
| | - Tianyu Li
- Department of Oncology, Wuxi County People's Hospital, No. 100 Wantong Road, Baiyang Street, Chongqing, 405899, China.
| | - Chengshu Liang
- Department of Oncology, Wuxi County People's Hospital, No. 100 Wantong Road, Baiyang Street, Chongqing, 405899, China.
| |
Collapse
|
24
|
Kan NN, Yu CY, Cheng YF, Hsu CC, Chen CL, Hsu HW, Weng CC, Tsang LLC, Chuang YH, Huang PH, Lim WX, Chen CP, Liao CC, Ou HY. Combined Hounsfield units of hepatocellular carcinoma on computed tomography and PET as a noninvasive predictor of early recurrence after living donor liver transplantation: Time-to-recurrence survival analysis. Eur J Radiol 2024; 177:111551. [PMID: 38875747 DOI: 10.1016/j.ejrad.2024.111551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 04/26/2024] [Accepted: 06/02/2024] [Indexed: 06/16/2024]
Abstract
BACKGROUND Liver transplantation is an effective treatment for preventing hepatocellular carcinoma (HCC) recurrence. This retrospective study aimed to quantitatively evaluate the attenuation in Hounsfield units (HU) on contrast-enhanced computed tomography (CECT) as a prognostic factor for hepatocellular carcinoma (HCC) following liver transplantation as a treatment. Our goal is to optimize its predictive ability for early tumor recurrence and compare it with the other imaging modality-positron emission tomography (PET). METHODS In 618 cases of LDLT for HCC, only 131 patients with measurable viable HCC on preoperative CECT and preoperative positron emission tomography (PET) evaluations were included, with a minimum follow-up period of 6 years. Cox regression models were developed to identify predictors of postoperative recurrence. Performance metrics for both CT and PET were assessed. The correlation between these two imaging modalities was also evaluated. Survival analyses were conducted using time-dependent receiver operating characteristic (ROC) curve analysis and area under the curve (AUC) to assess accuracy and determine optimized cut-off points. RESULTS Univariate and multivariate analyses revealed that both arterial-phase preoperative tumor attenuation (HU) and PET were independent prognostic factors for recurrence-free survival. Both lower arterial tumor enhancement (Cut-off value = 59.2, AUC 0.88) on CT and PET positive (AUC 0.89) increased risk of early tumor recurrence 0.5-year time-dependent ROC. Composites with HU < 59.2 and a positive PET result exhibited significantly higher diagnostic accuracy in detecting early tumor recurrence (AUC = 0.96). CONCLUSION Relatively low arterial tumor enhancement values on CECT effectively predict early HCC recurrence after LDLT. The integration of CT and PET imaging may serve as imaging markers of early tumor recurrence in HCC patients after LDLT.
Collapse
Affiliation(s)
- Na-Ning Kan
- Liver Transplantation Program and Departments of Diagnostic Radiology and Surgery, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan; Department of Diagnostic Radiology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Chun-Yen Yu
- Liver Transplantation Program and Departments of Diagnostic Radiology and Surgery, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan; Department of Diagnostic Radiology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Yu-Fan Cheng
- Liver Transplantation Program and Departments of Diagnostic Radiology and Surgery, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan; Department of Diagnostic Radiology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Chien-Chin Hsu
- Liver Transplantation Program and Departments of Diagnostic Radiology and Surgery, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan; Department of Nuclear Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Chao-Long Chen
- Liver Transplantation Program and Departments of Diagnostic Radiology and Surgery, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan; Department of Surgery, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Hsien-Wen Hsu
- Liver Transplantation Program and Departments of Diagnostic Radiology and Surgery, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan; Department of Diagnostic Radiology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Ching-Chun Weng
- Liver Transplantation Program and Departments of Diagnostic Radiology and Surgery, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan; Department of Diagnostic Radiology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Leo Leung-Chit Tsang
- Liver Transplantation Program and Departments of Diagnostic Radiology and Surgery, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan; Department of Diagnostic Radiology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Yi-Hsuan Chuang
- Liver Transplantation Program and Departments of Diagnostic Radiology and Surgery, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan; Department of Diagnostic Radiology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Po-Hsun Huang
- Liver Transplantation Program and Departments of Diagnostic Radiology and Surgery, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan; Department of Diagnostic Radiology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Wei-Xiong Lim
- Liver Transplantation Program and Departments of Diagnostic Radiology and Surgery, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan; Department of Diagnostic Radiology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Chen-Pei Chen
- Liver Transplantation Program and Departments of Diagnostic Radiology and Surgery, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan; Department of Diagnostic Radiology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Chien-Chang Liao
- Liver Transplantation Program and Departments of Diagnostic Radiology and Surgery, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan; Department of Diagnostic Radiology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Hsin-You Ou
- Liver Transplantation Program and Departments of Diagnostic Radiology and Surgery, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan; Department of Diagnostic Radiology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan.
| |
Collapse
|
25
|
Manzella A, Ecker BL, Eskander MF, Grandhi MS, In H, Kravchenko T, Langan RC, Kennedy T, Alexander HR, Beninato T, Pitt HA. Operative trends for pancreatic and hepatic malignancies during the COVID-19 pandemic. Surgery 2024; 176:364-370. [PMID: 38582733 DOI: 10.1016/j.surg.2024.02.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 02/25/2024] [Indexed: 04/08/2024]
Abstract
BACKGROUND The COVID-19 pandemic disrupted routine health care, including many elective and non-cancer operations in the United States. Most hepato-pancreato-biliary malignancy patients require outpatient imaging, tissue sampling, and staging, and many undergo neoadjuvant therapy before operative intervention. The aims of this study were to evaluate the effect of the COVID-19 pandemic on hepato-pancreato-biliary oncologic operations and to determine whether trends in neoadjuvant therapy were altered by the pandemic. METHODS Adult patients in the United States undergoing oncologic operations for pancreatic, primary and secondary hepatic malignancies, with or without neoadjuvant therapy, were extracted from the Vizient Clinical Data Base. Control chart analysis was used to plot trends over time and to determine whether changes were statistically significant. Wilcoxon rank-sum tests also compared monthly operative volume from pre-pandemic (12 month) and pandemic (28 months) periods. RESULTS A total of 36,553 patients were identified over 40 months. Mean monthly pancreatic oncologic operations were unaffected by the pandemic (P = .257). Operations for pancreatic oncologic operations with prior neoadjuvant therapy increased throughout the pandemic (P = .002). Oncologic operations for primary and secondary hepatic malignancies were significantly reduced for 4 and 2 months, respectively, at the beginning of the pandemic but returned to their pre-pandemic baseline within 4 months (P = .169 and P = .598). CONCLUSION Pancreatic operation volumes for cancer did not change, but pancreatic operations after neoadjuvant therapy continued to increase during the pandemic. Operations for hepatic malignancy were transiently disrupted but quickly normalized. These observations suggest that surgery for hepato-pancreato-biliary malignancies was prioritized during the pandemic.
Collapse
Affiliation(s)
- Alexander Manzella
- Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ; Rutgers Cancer Institute of New Jersey, New Brunswick, NJ
| | - Brett L Ecker
- Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ; Rutgers Cancer Institute of New Jersey, New Brunswick, NJ
| | - Mariam F Eskander
- Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ; Rutgers Cancer Institute of New Jersey, New Brunswick, NJ
| | - Miral S Grandhi
- Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ; Rutgers Cancer Institute of New Jersey, New Brunswick, NJ
| | - Haejin In
- Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ; Rutgers Cancer Institute of New Jersey, New Brunswick, NJ
| | - Timothy Kravchenko
- Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ; Rutgers Cancer Institute of New Jersey, New Brunswick, NJ
| | - Russell C Langan
- Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ; Rutgers Cancer Institute of New Jersey, New Brunswick, NJ
| | - Timothy Kennedy
- Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ; Rutgers Cancer Institute of New Jersey, New Brunswick, NJ
| | - H Richard Alexander
- Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ; Rutgers Cancer Institute of New Jersey, New Brunswick, NJ
| | - Toni Beninato
- Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ; Rutgers Cancer Institute of New Jersey, New Brunswick, NJ
| | - Henry A Pitt
- Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ; Rutgers Cancer Institute of New Jersey, New Brunswick, NJ.
| |
Collapse
|
26
|
Li W, Zhao B, Wang Q, Lu J, Wu X, Chen X. M2 macrophage exosomes promote resistance to sorafenib in hepatocellular carcinoma cells via miR-200c-3p. Int Immunopharmacol 2024; 139:112807. [PMID: 39068757 DOI: 10.1016/j.intimp.2024.112807] [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: 04/09/2024] [Revised: 07/08/2024] [Accepted: 07/23/2024] [Indexed: 07/30/2024]
Abstract
OBJECTIVE Sorafenib is a chemotherapeutic agent used to treat hepatocellular carcinoma (HCC). However, its clinical response rates are often low. Tumour-associated macrophages (TAMs) have been implicated in tumour resistance. The relationship between TAMs-derived exosomes and primary resistance to sorafenib in hepatocellular carcinoma is unclear. METHODS The study analysed RNA-SEQ data from TCGA-LIHC to explore the relationship between TAMs and sorafenib IC50. THP-1-induced M2 macrophages were used as a model to investigate the relationship between M2 macrophage exosomes and primary resistance to sorafenib in hepatocellular carcinoma cells using apoptosis, colony generation, cell viability and dual luciferase. RESULTS M2 macrophage score and sorafenib IC50 were positively correlated in hepatocellular carcinoma patients, M2 macrophage exosomes promoted sorafenib resistance in hepatocellular carcinoma cells, and M2-exo-miR-200c-3p facilitated the development of sorafenib resistance in hepatocellular carcinoma cells by mediating the activation of PI3K/AKT. CONCLUSION We propose and demonstrate for the first time that M2 macrophage exosomes promote sorafenib resistance in hepatocellular carcinoma, providing a new perspective for the clinical treatment of hepatocellular carcinoma patients.
Collapse
Affiliation(s)
- Wenhua Li
- Shihezi University School of Medicine, Shihezi 832000, China; Key Laboratory for Prevention and Treatment of High Morbidity in Central Asia, National Health and Health Commission, Shihezi 832000, China
| | - Bin Zhao
- Shihezi University School of Medicine, Shihezi 832000, China; Key Laboratory for Prevention and Treatment of High Morbidity in Central Asia, National Health and Health Commission, Shihezi 832000, China
| | - Qianwen Wang
- Shihezi University School of Medicine, Shihezi 832000, China; Key Laboratory for Prevention and Treatment of High Morbidity in Central Asia, National Health and Health Commission, Shihezi 832000, China
| | - Junxia Lu
- Shihezi University School of Medicine, Shihezi 832000, China; Key Laboratory for Prevention and Treatment of High Morbidity in Central Asia, National Health and Health Commission, Shihezi 832000, China
| | - Xiangwei Wu
- Shihezi University School of Medicine, Shihezi 832000, China; The First Affiliated Hospital, Shihezi University School of Medicine, Shihezi 832000, China; Key Laboratory for Prevention and Treatment of High Morbidity in Central Asia, National Health and Health Commission, Shihezi 832000, China.
| | - Xueling Chen
- Shihezi University School of Medicine, Shihezi 832000, China; Key Laboratory for Prevention and Treatment of High Morbidity in Central Asia, National Health and Health Commission, Shihezi 832000, China.
| |
Collapse
|
27
|
Kotsifa E, Machairas N, Angelis A, Nikiteas NI, Dimitroulis D, Sotiropoulos GC. Decoding the Prognostic Significance and Therapeutic Implications of Inflammation-Based Scores in Hepatocellular Carcinoma: A Comprehensive Review. Cancers (Basel) 2024; 16:2549. [PMID: 39061188 PMCID: PMC11274930 DOI: 10.3390/cancers16142549] [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: 05/23/2024] [Revised: 07/11/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common primary liver cancer, posing a significant global health challenge with an increasing incidence. In recent years, multiple staging systems and scores have been proposed, emphasising the necessity for the development of precise prognostic tools. The well-documented etiological relationship between chronic inflammation and carcinogenesis has prompted researchers to explore novel prognostic markers associated with the inflammatory status of HCC patients. This review summarises the current data about inflammation-based scores in the context of HCC. We discuss established scores like the Glasgow Prognostic Score (GPS), modified GPS (mGPS) and the neutrophil-to-lymphocyte ratio (NLR) and others not as extensively studied, examining their utility in predicting survival outcomes and treatment response in HCC patients. Furthermore, we explore emerging scores, including the prognostic nutritional index (PNI) and other lymphocyte-based scores, assessing their potential in refining risk stratification and guiding therapeutic decisions in the era of precision medicine. As research progresses and these scores undergo further refinement and integration into the evolving landscape of HCC management, they carry significant potential for improving patient outcomes.
Collapse
Affiliation(s)
- Evgenia Kotsifa
- 2nd Propaedeutic Department of Surgery, General Hospital of Athens “Laiko”, National and Kapodistrian University of Athens, Agiou Thoma 17, 11527 Athens, Greece
| | | | | | | | | | | |
Collapse
|
28
|
Kraglund F, Skou N, Villadsen GE, Jepsen P. Landmark analysis of the risk of recurrence after resection or ablation for HCC: A nationwide study. Hepatol Commun 2024; 8:e0472. [PMID: 38896083 PMCID: PMC11186808 DOI: 10.1097/hc9.0000000000000472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 04/24/2024] [Indexed: 06/21/2024] Open
Abstract
BACKGROUND The risk of HCC recurrence at particular landmarks since the initial treatment is unknown. With this registry-based study, we aimed to provide a nuanced description of the prognosis following resection or ablation for HCC, including landmark analyses. METHODS Using the Danish nationwide health care registries, we identified all patients who received resection or ablation in 2000-2018 as the first HCC treatment. HCC recurrence was defined as a new HCC treatment > 90 days after the first treatment. We conducted competing risk landmark analyses of the cumulative risk of recurrence and death. RESULTS Among 4801 patients with HCC, we identified 426 patients who received resection and 544 who received ablation. The 2 treatment cohorts differed in cirrhosis prevalence and tumor stage. The 5-year recurrence risk was 40.7% (95% CI 35.5%-45.8%) following resection and 60.7% (95% CI: 55.9%-65.1%) following ablation. The 1-year recurrence risk decreased over the landmarks from 20.4% (95% CI: 16.6%-24.6%) at the time of resection to 4.7% (95% CI: 0.9%-13.9%) at the 5-year landmark. For ablation, the risk decreased from 36.1% (95% CI: 31.9%-40.4%) at the time of treatment to 5.3% (95% CI: 0.4%-21.4%) at the 5-year landmark. The risk of death without recurrence was stable over the landmarks following both resection and ablation. CONCLUSIONS In conclusion, the risk of recurrence or death following resection or ablation for HCC is high from the treatment date, but the risk of recurrence decreases greatly over the survival landmarks. This information is valuable for clinicians and their patients.
Collapse
Affiliation(s)
- Frederik Kraglund
- Department of Hepatology and Gastroenterology, Aarhus University Hospital, Aarhus N, Denmark
| | - Nikolaj Skou
- Department of Radiology, Aarhus University Hospital, Aarhus N, Denmark
| | | | - Peter Jepsen
- Department of Hepatology and Gastroenterology, Aarhus University Hospital, Aarhus N, Denmark
- Department of Clinical Epidemiology, Aarhus University Hospital, Aarhus N, Denmark
| |
Collapse
|
29
|
He H, Zhang Q, Gu Q, Yang H, Yue C. CircGNAO1 strengthens its host gene GNAO1 expression for suppression of hepatocarcinogenesis. Heliyon 2024; 10:e32848. [PMID: 38988568 PMCID: PMC11233958 DOI: 10.1016/j.heliyon.2024.e32848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 06/08/2024] [Accepted: 06/10/2024] [Indexed: 07/12/2024] Open
Abstract
Background Hepatocellular carcinoma (HCC) is one of the most prevalent primary liver carcinoma. Guanine nucleotide-binding protein, α-activating activity polypeptide O (GNAO1) was reported to be under-expressed in HCC tissues. This study aimed to investigate the GNAO1-derived circular RNA (circRNA) and its molecular mechanisms in HCC. Methods Real-time quantitative polymerase chain reaction (RT-qPCR) and Western blot were applied to examine RNA and protein levels. Functional experiments were performed to study HCC cell proliferation, cell cycle and cellular senescence. The interactions among circGNAO1, GNAO1 and DNA methyltransferase 1 (DNMT1) were examined by mechanism assays. The methylation level was analyzed by bisulfite sequencing PCR (BSP). Results CircGNAO1 is down-regulated and positively associated with GNAO1 in HCC tissues. Overexpression of circGNAO1 inhibits cell proliferation, induces cell cycle arrest and facilitates cell senescence in HCC cells. CircGNAO1 facilitates the progression of HCC via modulating GNAO1. Mechanistically, circGNAO1 enhances the transcription of GNAO1 by sequestering DNMT1, thereby up-regulating GNAO1 expression in HCC cells. Conclusions CircGNAO1 up-regulates its host gene GNAO1 expression for suppression of hepatocarcinogenesis.
Collapse
Affiliation(s)
- Hongwei He
- Department of Hepatobiliary Surgery, Jinshan Branch of Shanghai Sixth People's Hospital, Shanghai Jiaotong University, Shanghai, 201599, China
| | - Qing Zhang
- Trade Union of Shandong Second Provincial General Hospital, Jinan, 250022, Shandong Province, China
| | - Qiyun Gu
- Department of Hepatobiliary Surgery, Jinshan Branch of Shanghai Sixth People's Hospital, Shanghai Jiaotong University, Shanghai, 201599, China
| | - Hui Yang
- Department of Neurology Medicine, The Second Hospital, Shandong University, Jinan, 250033, Shandong Province, China
| | - Caibin Yue
- General Medicine Department, The Second Hospital, Shandong University, Jinan, 250033, Shandong Province, China
| |
Collapse
|
30
|
Wang W, Fu R, Gao R, Luo L, Wang Z, Xue Y, Sun J, Pan M, Hong M, Qiao L, Qiao W, Mei Q, Wu J, Wang Y, Zhong Y, Liu J, Tong F. H 2S-Powered Nanomotors for Active Therapy of Tumors by Inducing Ferroptosis and Lactate-Pyruvate Axis Disorders. ACS Biomater Sci Eng 2024; 10:3994-4008. [PMID: 38736179 DOI: 10.1021/acsbiomaterials.3c01665] [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: 05/14/2024]
Abstract
Disruption of the symbiosis of extra/intratumoral metabolism is a good strategy for treating tumors that shuttle resources from the tumor microenvironment. Here, we report a precision treatment strategy for enhancing pyruvic acid and intratumoral acidosis to destroy tumoral metabolic symbiosis to eliminate tumors; this approach is based on PEGylated gold and lactate oxidase-modified aminated dendritic mesoporous silica with lonidamine and ferrous sulfide loading (PEG-Au@DMSNs/FeS/LND@LOX). In the tumor microenvironment, LOX oxidizes lactic acid to produce pyruvate, which represses tumor cell proliferation by inhibiting histone gene expression and induces ferroptosis by partial histone monoubiquitination. In acidic tumor conditions, the nanoparticles release H2S gas and Fe2+ ions, which can inhibit catalase activity to promote the Fenton reaction of Fe2+, resulting in massive ·OH production and ferroptosis via Fe3+. More interestingly, the combination of H2S and LND (a monocarboxylic acid transporter inhibitor) can cause intracellular acidosis by lactate, and protons overaccumulate in cells. Multiple intracellular acidosis is caused by lactate-pyruvate axis disorders. Moreover, H2S provides motive power to intensify the shuttling of nanoparticles in the tumor region. The findings confirm that this nanomedicine system can enable precise antitumor effects by disrupting extra/intratumoral metabolic symbiosis and inducing ferroptosis and represents a promising active drug delivery system candidate for tumor treatment.
Collapse
Affiliation(s)
- Weixin Wang
- Department of Pharmacology, School of Pharmacy, Binzhou Medical University, Yantai, 264003, PR China
| | - Renquan Fu
- Key Laboratory of Tropical Biological Resources of Ministry of Education and One Health Institute, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China
| | - Rui Gao
- Department of Pharmacology, School of Pharmacy, Binzhou Medical University, Yantai, 264003, PR China
| | - Lei Luo
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Zhongchao Wang
- Institute of Cardiovascular Disease, Shanxi Medical University, Taiyuan, 030001, PR China
| | - Yingli Xue
- Xi'an Medical University, Xi'an, 710000, PR China
| | - Jiahui Sun
- Department of Pharmacology, School of Pharmacy, Binzhou Medical University, Yantai, 264003, PR China
| | - Min Pan
- Department of Pharmacology, School of Pharmacy, Binzhou Medical University, Yantai, 264003, PR China
| | - Miaofang Hong
- Department of Pharmacology, School of Pharmacy, Binzhou Medical University, Yantai, 264003, PR China
| | - Lingyan Qiao
- Clinical Medical College, Binzhou Medical University, Yantai, 264003, PR China
| | - Weiwei Qiao
- Clinical Medical College, Binzhou Medical University, Yantai, 264003, PR China
| | - Qibing Mei
- Education Ministry Key Laboratory of Medical Electrophysiology, Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou; Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Jianming Wu
- Education Ministry Key Laboratory of Medical Electrophysiology, Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou; Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Yini Wang
- Clinical Medical College, Binzhou Medical University, Yantai, 264003, PR China
| | - Yali Zhong
- Southwest University of Science and Technology, 621000 Mianyang, China
| | - Jin Liu
- Key Laboratory of Tropical Biological Resources of Ministry of Education and One Health Institute, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China
| | - Fei Tong
- Department of Pharmacology, School of Pharmacy, Binzhou Medical University, Yantai, 264003, PR China
- Education Ministry Key Laboratory of Medical Electrophysiology, Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou; Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
- School of Medicine, Zhejiang University, 310000 Hangzhou, China
| |
Collapse
|
31
|
Pan Y, You B, Zhao X, Zhang S, Li W. CHOP regulated by METTL14-m6A affects cell cycle arrest and regorafenib sensitivity in HCC cells. BMC Cancer 2024; 24:525. [PMID: 38664644 PMCID: PMC11046807 DOI: 10.1186/s12885-024-12275-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND Regorafenib, a multi-targeted kinase inhibitor, has been used in the treatment of Hepatocellular carcinoma (HCC). The purpose of this study is to investigate the mechanism of Regorafenib in HCC. METHODS Regorafenib's impact on the sensitivity of HCC cells was assessed using CCK8. Differential gene expression analysis was performed by conducting mRNA sequencing after treatment with Regorafenib. The m6A methylation status of CHOP and differential expression of m6A methylation-related proteins were assessed by RIP and Western Blot. To explore the molecular mechanisms involved in the therapeutic effects of Regorafenib in HCC and the impact of METTL14 and CHOP on Regorafenib treatment, we employed shRNA/overexpression approaches to transfect METTL14 and CHOP genes, as well as conducted in vivo experiments. RESULTS Treatment with Regorafenib led to a notable decrease in viability and proliferation of SK-Hep-1 and HCC-LM3 cells. The expression level of CHOP was upregulated after Regorafenib intervention, and CHOP underwent m6A methylation. Among the m6A methylation-related proteins, METTL14 exhibited the most significant downregulation. Mechanistic studies revealed that Regorafenib regulated the cell cycle arrest in HCC through METTL14-mediated modulation of CHOP, and the METTL14/CHOP axis affected the sensitivity of HCC to Regorafenib. In vivo, CHOP enhanced the anticancer effect of Regorafenib. CONCLUSION The inhibition of HCC development by Regorafenib is attributed to its modulation of m6A expression of CHOP, mediated by METTL14, and the METTL14/CHOP axis enhances the sensitivity of HCC to Regorafenib. These findings provide insights into the treatment of HCC and the issue of drug resistance to Regorafenib.
Collapse
Affiliation(s)
- Yipeng Pan
- Department of Transplantation, The Second Affiliated Hospital of Hainan Medical University, Haikou, 570100, China
| | - Bo You
- Department of Transplantation, The Hainan General Hospital, Haikou, 570100, China
| | - Xue Zhao
- Department of Transplantation, The Second Affiliated Hospital of Hainan Medical University, Haikou, 570100, China
| | - Shanxin Zhang
- Department of Ultrasound Medicine, Zhejiang Provincial People's Hospital, Zhejiang, 311100, China.
| | - Wei Li
- Department of Transplantation, The Second Affiliated Hospital of Hainan Medical University, Haikou, 570100, China.
| |
Collapse
|
32
|
Shu J, Zhang J, Jee K, Liu L, Hu M, Huo W, Cui X, Wang H, Lu HM. Impact of iodinated oil in proton therapy on relative stopping power of liver post-cTACE. Phys Med Biol 2024; 69:09NT03. [PMID: 38537311 DOI: 10.1088/1361-6560/ad388b] [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: 09/28/2023] [Accepted: 03/27/2024] [Indexed: 04/18/2024]
Abstract
Objective. Conventional transarterial chemoembolization (cTACE) is a common treatment for hepatocellular carcinoma (HCC), often with unsatisfactory local controls. Combining cTACE with radiotherapy shows a promise for unresectable large HCC, with proton therapy preserving healthy liver tissue. However, the proton therapy benefits are subject to the accuracy of tissue relative stopping power (RSP) prediction. The RSP values are typically derived from computed tomography (CT) images using stoichiometric calibration. Lipiodol deposition significantly increases CT numbers in liver regions of post-cTACE. Hence, it is necessary to evaluate the accuracy of RSP in liver regions of post-cTACE.Approach. Liver, water, and iodinated oil samples were prepared. Some liver samples contained iodinated oil. The water equivalent path length (WEPL) of sample was measured through the pullbacks of spread-out Bragg peak (SOBP) depth-dose profiles scanned in a water tank with and without sample in the beam path. Measured RSP values were compared to estimated RSP values derived from the CT number based on the stoichiometric calibration method.Main results. The measured RSP of water was 0.991, confirming measurement system calibration. After removing the RSP contribution from container walls, the pure iodinated oil and liver samples had RSP values of 1.12 and 1.06, while the liver samples mixed with varying oil volumes (5 ml, 10 ml, 15 ml) showed RSP values of 1.05, 1.05 and 1.06. Using the stoichiometric calibration method, pure iodinated oil and liver samples had RSP values of 2.79 and 1.06. Liver samples mixed with iodinated oil (5 ml, 10 ml, 15 ml) had calculated RSP values of 1.21, 1.34, and 1.46. The RSP discrepancy reached 149.1% for pure iodinated oil.Significance.Iodinated oil notably raises CT numbers in liver tissue. However, there is almost no effect on its RSP value. Proton treatment of post-cTACE HCC patients can therefore be overshooting if no proper measures are taken against this specific effect.
Collapse
Affiliation(s)
- Jiong Shu
- Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, People's Republic of China
- University of Science and Technology of China, Hefei, Anhui 230026, People's Republic of China
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, Anhui, 230031, People's Republic of China
| | - Jianguang Zhang
- Department of oncology, zibo wanjie hospital, Zibo, Shandong, 255213, People's Republic of China
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, United States of America
| | - Kyungwook Jee
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, United States of America
| | - LingLing Liu
- Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, People's Republic of China
- University of Science and Technology of China, Hefei, Anhui 230026, People's Republic of China
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, Anhui, 230031, People's Republic of China
| | - Man Hu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, People's Republic of China
| | - Wanli Huo
- Key Laboratory of Electromagnetic Wave Information Technology and Metrology of Zhejiang Province, College of Information Engineering, China Jiliang University, Hangzhou, 310018, People's Republic of China
| | - Xiangli Cui
- Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, People's Republic of China
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, Anhui, 230031, People's Republic of China
| | - Hongzhi Wang
- Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, People's Republic of China
- University of Science and Technology of China, Hefei, Anhui 230026, People's Republic of China
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, Anhui, 230031, People's Republic of China
| | - Hsiao-Ming Lu
- Hefei Ion Medical Center, Hefei, Anhui, 230088, People's Republic of China
| |
Collapse
|
33
|
Jiang K, Ning N, Huang J, Chang Y, Wang R, Ma J. Psilostachyin C reduces malignant properties of hepatocellular carcinoma cells by blocking CREBBP-mediated transcription of GATAD2B. Funct Integr Genomics 2024; 24:75. [PMID: 38600341 DOI: 10.1007/s10142-024-01353-8] [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: 02/05/2024] [Revised: 03/16/2024] [Accepted: 03/30/2024] [Indexed: 04/12/2024]
Abstract
Hepatocellular carcinoma (HCC) is a leading cause of cancer-related mortality globally. Many herbal medicines and their bioactive compounds have shown anti-tumor properties. This study was conducted to examine the effect of psilostachyin C (PSC), a sesquiterpenoid lactone isolated from Artemisia vulgaris L., in the malignant properties of HCC cells. CCK-8, flow cytometry, wound healing, and Transwell assays revealed that 25 μM PSC treatment significantly suppressed proliferation, cell cycle progression, migration, and invasion of two HCC cell lines (Hep 3B and Huh7) while promoting cell apoptosis. Bioinformatics prediction suggests CREB binding protein (CREBBP) as a promising target of PSC. CREBBP activated transcription of GATA zinc finger domain containing 2B (GATAD2B) by binding to its promoter. CREBBP and GATAD2B were highly expressed in clinical HCC tissues and the acquired HCC cell lines, but their expression was reduced by PSC. Either upregulation of CREBBP or GATAD2B restored the malignant properties of HCC cells blocked by PSC. Collectively, this evidence demonstrates that PSC pocessess anti-tumor functions in HCC cells by blocking CREBBP-mediated transcription of GATAD2B.
Collapse
Affiliation(s)
- Kai Jiang
- Department of Clinical Pharmacy, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, 710054, P.R. China
| | - Ning Ning
- Department of Orthopedics, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, 710054, P.R. China
| | - Jing Huang
- Department of Clinical Pharmacy, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, 710054, P.R. China
| | - Yu Chang
- Department of Clinical Pharmacy, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, 710054, P.R. China
| | - Rao Wang
- Department of TCM Orthopedic Center, Honghui Hospital, Xi'an Jiaotong University, No. 555, Youyi East Road, Beilin District, Xi'an, Shaanxi, 710054, P.R. China.
| | - Jie Ma
- Department of Neurology, Honghui Hospital, Xi'an Jiaotong University, No. 555, Youyi East Road, Beilin District, Xi'an, Shaanxi, 710054, P.R. China.
| |
Collapse
|
34
|
Sarkar J, Oshi M, Satyananda V, Chida K, Yan L, Maiti A, Hait N, Endo I, Takabe K. Spinster Homologue 2 Expression Correlates With Improved Patient Survival in Hepatocellular Carcinoma Despite Association With Lymph-Angiogenesis. World J Oncol 2024; 15:181-191. [PMID: 38545475 PMCID: PMC10965268 DOI: 10.14740/wjon1732] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 01/30/2024] [Indexed: 01/04/2025] Open
Abstract
BACKGROUND Spinster homologue 2 (SPNS2) is a transporter of sphingosine-1-phosphate (S1P), a bioactive lipid linked to cancer progression. We studied the link between SPNS2 gene expression, tumor aggressiveness, and outcomes in patients with hepatocellular carcinoma (HCC). METHODS Gene expression in patients with HCC was analyzed from the Cancer Genome Atlas (TCGA) (n = 350) and GSE76427 (n = 115) as a validation cohort, as well as liver tissue cohort GSE6764 (n = 75). RESULTS High-SPNS2 HCC was significantly associated with high level of lymph-angiogenesis-related factors. SPNS2 expression was significantly higher in normal liver and early HCC versus advanced HCC (P < 0.02). High SPNS2 levels enriched immune response-related gene sets; inflammatory, interferon (IFN)-α, IFN-γ responses, and tumor necrosis factor (TNF)-α, interleukin (IL)-6/Janus kinase/signal transducer and activator of transcription (JAK/STAT3) signaling, complement and allograft rejection, but did not significantly infiltrate specific immune cells nor cytolytic activity score. High-SPNS2 HCC enriched tumor aggravating pathway gene sets such as KRAS (Kirsten rat sarcoma virus) signaling, but inversely correlated with Nottingham histological grade, MKI67 (marker of proliferation Ki-67) expression, and cell proliferation-related gene sets. Further, high-SPNS2 HCC had significantly high infiltration of stromal cells, showing that low-SPNS2 HCC is highly proliferative. Finally, high-SPNS2 HCC was associated with better disease-free, disease-specific, and overall survival (P = 0.031, 0.046, and 0.040, respectively). CONCLUSIONS Although SPNS2 expression correlated with lymph-angiogenesis and other cancer-promoting pathways, it also enriched immune response. SPNS2 levels were higher in normal liver compared to HCC, and inversely correlated with cancer cell proliferation and better survival. SPNS2 expression may be beneficial in HCC patients despite detrimental in-vitro effects.
Collapse
Affiliation(s)
- Joy Sarkar
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
- These authors contributed equally to this work
| | - Masanori Oshi
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
- Department of Gastroenterological Surgery, Yokohama, Kanagawa 236-004, Japan
- These authors contributed equally to this work
| | - Vikas Satyananda
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Kohei Chida
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Li Yan
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Aparna Maiti
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Nitai Hait
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Itaru Endo
- Department of Gastroenterological Surgery, Yokohama, Kanagawa 236-004, Japan
| | - Kazuaki Takabe
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
- Department of Gastroenterological Surgery, Yokohama, Kanagawa 236-004, Japan
- Department of Surgery, University at Buffalo Jacobs School of Medicine and Biomedical Sciences, the State University of New York, Buffalo, NY, USA
- Department of Breast Surgery and Oncology, Tokyo Medical University, Tokyo 160-8402, Japan
- Department of Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan
- Department of Breast Surgery, Fukushima Medical University, Fukushima, Japan
| |
Collapse
|
35
|
de Sousa DJM, Feitosa de Oliveira KG, Pereira IC, do Nascimento GTM, Barrense CO, Martins JA, Pereira Rêgo BDM, Oliveira da Silva TE, Carneiro da Silva FC, Torres-Leal FL. Dietary restriction and hepatic cancer: Systematic review and meta-analysis of animal studies. Crit Rev Oncol Hematol 2024; 196:104264. [PMID: 38341120 DOI: 10.1016/j.critrevonc.2024.104264] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 12/21/2023] [Accepted: 01/10/2024] [Indexed: 02/12/2024] Open
Abstract
The effect of calorie restriction, fasting, and ketogenic diets on the treatment of liver cancer remains uncertain. Therefore, we conducted a systematic review to evaluate the effect of restrictive diets on the development and progression of liver cancer in animal models. We did a meta-analysis using the Cochrane Collaboration's Review Manager software, with the random effects model and the inverse variance technique. We examined 19 studies that were conducted between 1983 and 2020. Of these, 63.2% investigated calorie restriction, 21.0% experimented with a ketogenic diet, and 15.8% investigated the effects of fasting. The intervention lasted anything from 48 h to 221 weeks. Results showed that restrictive diets may reduce tumor incidence and progression, with a significant reduction in the risk of liver cancer development. Thereby, our results suggest that putting limits on what you eat may help treat liver cancer in more ways than one.
Collapse
Affiliation(s)
- Dallyla Jennifer Morais de Sousa
- Metabolic Diseases Glauto Tuquarre Laboratory, Metabolic Diseases, Exercise and Nutrition Research Group (DOMEN), Department of Biophysics and Physiology, Center for Health Sciences, Federal University of Piauí, Teresina, Brazil
| | - Kynnara Gabriella Feitosa de Oliveira
- Metabolic Diseases Glauto Tuquarre Laboratory, Metabolic Diseases, Exercise and Nutrition Research Group (DOMEN), Department of Biophysics and Physiology, Center for Health Sciences, Federal University of Piauí, Teresina, Brazil
| | - Irislene Costa Pereira
- Metabolic Diseases Glauto Tuquarre Laboratory, Metabolic Diseases, Exercise and Nutrition Research Group (DOMEN), Department of Biophysics and Physiology, Center for Health Sciences, Federal University of Piauí, Teresina, Brazil
| | - Glauto Tuquarre Melo do Nascimento
- Metabolic Diseases Glauto Tuquarre Laboratory, Metabolic Diseases, Exercise and Nutrition Research Group (DOMEN), Department of Biophysics and Physiology, Center for Health Sciences, Federal University of Piauí, Teresina, Brazil
| | - Clenio Oliveira Barrense
- Metabolic Diseases Glauto Tuquarre Laboratory, Metabolic Diseases, Exercise and Nutrition Research Group (DOMEN), Department of Biophysics and Physiology, Center for Health Sciences, Federal University of Piauí, Teresina, Brazil
| | - Jorddam Almondes Martins
- Metabolic Diseases Glauto Tuquarre Laboratory, Metabolic Diseases, Exercise and Nutrition Research Group (DOMEN), Department of Biophysics and Physiology, Center for Health Sciences, Federal University of Piauí, Teresina, Brazil
| | - Beatriz de Mello Pereira Rêgo
- Metabolic Diseases Glauto Tuquarre Laboratory, Metabolic Diseases, Exercise and Nutrition Research Group (DOMEN), Department of Biophysics and Physiology, Center for Health Sciences, Federal University of Piauí, Teresina, Brazil
| | | | | | - Francisco Leonardo Torres-Leal
- Metabolic Diseases Glauto Tuquarre Laboratory, Metabolic Diseases, Exercise and Nutrition Research Group (DOMEN), Department of Biophysics and Physiology, Center for Health Sciences, Federal University of Piauí, Teresina, Brazil.
| |
Collapse
|
36
|
Burden of liver cancer mortality by county, race, and ethnicity in the USA, 2000-19: a systematic analysis of health disparities. Lancet Public Health 2024; 9:e186-e198. [PMID: 38429018 PMCID: PMC10986755 DOI: 10.1016/s2468-2667(24)00002-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 12/21/2023] [Accepted: 01/02/2024] [Indexed: 03/03/2024]
Abstract
BACKGROUND Understanding how specific populations are affected by liver cancer is important for identifying priorities, policies, and interventions to mitigate health risks and reduce disparities. This study aims to provide comprehensive analysis of rates and trends in liver cancer mortality for different racial and ethnic populations in the USA nationally and at the county level from 2000 to 2019. METHODS We applied small-area estimation methods to death registration data from the US National Vital Statistics System and population data from the US National Center for Health Statistics to estimate liver cancer mortality rates by county, racial and ethnic population, and year (2000-19) in the USA. Race and ethnicity were categorised as non-Latino and non-Hispanic American Indian or Alaska Native (AIAN), non-Latino and non-Hispanic Asian or Pacific Islander (Asian), non-Latino and non-Hispanic Black (Black), Latino or Hispanic (Latino), and non-Latino and non-Hispanic White (White). Estimates were adjusted using published misclassification ratios to correct for inaccuracies in race or ethnicity as recorded on death certificates, and then age-standardised. Mortality rate estimates are presented for all county and racial and ethnic population combinations with a mean annual population greater than 1000. FINDINGS Nationally, the age-standardised liver cancer mortality rate increased between the years 2000 (4·2 deaths per 100 000 population [95% uncertainty interval 4·1-4·3]) and 2016 (6·0 per 100 000 [5·9-6·1]), followed by a stabilisation in rates from 2016 to 2019 (6·1 per 100 000 [6·0-6·2]). Similar trends were observed across the AIAN, Black, Latino, and White populations, whereas the Asian population showed an overall decrease across the 20-year study period. Qualitatively similar trends were observed in most counties; however, the mortality rate and the rate of change varied substantially across counties, both within and across racial and ethnic populations. For the 2016-19 period, mortality continued to increase at a substantial rate in some counties even while it stabilised nationally. Nationally, the White population had the lowest mortality rate in all years, while the racial and ethnic population with the highest rate changed from the Asian population in 2000 to the AIAN population in 2019. Racial and ethnic disparities were substantial: in 2019, mortality was highest in the AIAN population (10·5 deaths per 100 000 [9·1-12·0]), notably lower for the Asian (7·5 per 100 000 [7·1-7·9]), Black (7·6 per 100 000 [7·3-7·8]), and Latino (7·7 per 100 000 [7·5-8·0]) populations, and lowest for the White population (5·5 [5·4-5·6]). These racial and ethnic disparities in mortality were prevalent throughout the country: in 2019, mortality was higher in minoritised racial and ethnic populations than in the White population living in the same county in 408 (87·7%) of 465 counties with unmasked estimates for the AIAN population, 604 (90·6%) of 667 counties for the Asian population, 1207 (81·2%) of 1486 counties for the Black population, and 1073 (73·0%) of 1469 counties for the Latino population. INTERPRETATION Although the plateau in liver cancer mortality rates in recent years is encouraging, mortality remains too high in many locations throughout the USA, particularly for minoritised racial and ethnic populations. Addressing population-specific risk factors and differences in access to quality health care is essential for decreasing the burden and disparities in liver cancer mortality across racial and ethnic populations and locations. FUNDING US National Institutes of Health (Intramural Research Program, National Institute on Minority Health and Health Disparities; National Heart, Lung, and Blood Institute; Intramural Research Program, National Cancer Institute; National Institute on Aging; National Institute of Arthritis and Musculoskeletal and Skin Diseases; Office of Disease Prevention; and Office of Behavioral and Social Sciences Research).
Collapse
|
37
|
Pu J, Wang J, Li W, Lu Y, Wu X, Long X, Luo C, Wei H. hsa_circ_0000092 promotes hepatocellular carcinoma progression through up-regulating HN1 expression by binding to microRNA-338-3p. J Cell Mol Med 2024; 28:e15010. [PMID: 32077624 PMCID: PMC10941524 DOI: 10.1111/jcmm.15010] [Citation(s) in RCA: 41] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 12/22/2019] [Accepted: 01/06/2020] [Indexed: 12/15/2022] Open
Abstract
Circular RNAs (circRNAs) have been identified in diverse cancers for their role in regulating multiple cellular processes by antagonizing microRNAs (miRNAs or miRs). However, the role of circRNA hsa_circ_0000092 in hepatocellular carcinoma (HCC) still remains enigmatic. Therefore, we aimed to investigate the specific mechanism of hsa_circ_0000092 in HCC. Differentially expressed circRNAs associated to HCC were initially analysed. The expression of hsa_circ_0000092, miR-338-3p and HN1 in HCC tissues and cell lines was examined. Next, the interaction among hsa_circ_0000092, miR-338-3p and HN1 was determined by dual-luciferase reporter, RNA pull-down and northern blot assays. Subsequently, a series of mimic, inhibitor or siRNA plasmids were delivered into HCC cells to validate the effects of hsa_circ_0000092, miR-338-3p and HN1 in controlling cell proliferation, migration, invasion and angiogenesis in vitro. Furthermore, the role of hsa_circ_0000092 in tumour growth of HCC in vivo was assessed with hsa_circ_0000092 depleted with siRNA. The hsa_circ_0000092/miR-338-3p/HN1 axis was predicted to participate in the development of HCC. hsa_circ_0000092 and HN1 were highly expressed while miR-338-3p was poorly expressed in HCC tissues and cell lines. hsa_circ_0000092 could competitively bind to miR-338-3p to up-regulate HN1 expression. Moreover, depleted hsa_circ_0000092 or elevated miR-338-3p was shown to suppress HCC cell proliferation, migration, invasion and angiogenesis in vitro via down-regulation of HN1. Furthermore, silencing hsa_circ_0000092 was demonstrated to suppress tumour growth in HCC in vivo. The results of this study suggested that hsa_circ_0000092 impaired miR-338-3p-mediated HN1 inhibition to aggravate the development of HCC, indicating that hsa_circ_0000092 is a potential candidate marker and therapeutic target for HCC.
Collapse
Affiliation(s)
- Jian Pu
- Department of Hepatobiliary SurgeryAffiliated Hospital of Youjiang Medical University for NationalitiesBaiseChina
| | - Jianchu Wang
- Department of Hepatobiliary SurgeryAffiliated Hospital of Youjiang Medical University for NationalitiesBaiseChina
| | - Wenchuan Li
- Department of Hepatobiliary SurgeryAffiliated Hospital of Youjiang Medical University for NationalitiesBaiseChina
| | - Yuan Lu
- Graduate College of Youjiang Medical University for NationalitiesBaiseChina
| | - Xianjian Wu
- Graduate College of Youjiang Medical University for NationalitiesBaiseChina
| | - Xidai Long
- Department of PathologyAffiliated Hospital of Youjiang Medical University for NationalitiesBaiseChina
| | - Chunying Luo
- Department of PathologyAffiliated Hospital of Youjiang Medical University for NationalitiesBaiseChina
| | - Huamei Wei
- Department of PathologyAffiliated Hospital of Youjiang Medical University for NationalitiesBaiseChina
| |
Collapse
|
38
|
Chen C, Wu H, Fu X, Li R, Cheng H, Wang M, Zhou A, Zhang M, Li Q. A UPLC-QTOF/MS-based hepatic tissue metabolomics approach deciphers the mechanism of Huachansu tablets-based intervention against hepatocellular carcinoma. J Pharm Biomed Anal 2024; 239:115875. [PMID: 38061172 DOI: 10.1016/j.jpba.2023.115875] [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: 08/02/2023] [Revised: 11/13/2023] [Accepted: 11/23/2023] [Indexed: 01/05/2024]
Abstract
Huachansu (HCS) tablets, classified as well-known traditional Chinese medicine (TCM) preparation, have been proved to be effective in the treatment of hepatocellular carcinoma (HCC) in clinical studies. However, the underlying mechanism of HCS tablets against HCC has not been comprehensively elucidated. In this study, a rat model of HCC was established with diethylnitrosamine (DEN) inducer. The efficacy of HCS tablets against HCC was assessed through liver histopathological examination and evaluation of biochemical indicators. A metabolomics method based on UPLC-Q-TOF/MS combined with multivariate data analysis was established to identify differential metabolites related to the inhibition effect of HCS tablets on HCC, and then the relevant metabolic pathway analysis was performed to investigate the anti-HCC mechanisms of HCS tablets. The results showed that compared to the control group, the HCC model group showed a significant increase in the values of HCC-related biochemical indicators and the number of tumor nodules, indicating the successful establishment of the HCC rat model. Upon treatment with HCS tablets, the values of HCC-related biochemical indicators decreased, liver fibrosis and nuclear deformation were also significantly alleviated. A total of 15 differential metabolites associated with the anti-tumor effect of HCS tablets on HCC were screened and annotated through hepatic tissue metabolomics studies. Analysis of metabolic pathways revealed that the therapeutic effects of HCS tablets on HCC mainly involved the pentose and glucuronate interconversions and arachidonic acid metabolism. Further western blotting corroborated that the alteration in arachidonic acid (AA) level after the intervention of HCS tablets was related to the inhibition of cPLA2α expression in rat liver tissues. In conclusion, HCS tablets exhibit a certain anti-tumor effect on HCC, and the metabolomics method based on UPLC-Q-TOF/MS combined with further verification at the biochemical level is a promising way to reveal its underlying mechanism.
Collapse
Affiliation(s)
- Chang Chen
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Hefei 230038, China
| | - Huan Wu
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Hefei 230038, China; Anhui Province Key Laboratory of Research and Development of Chinese Medicine, Hefei 230012, China; Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei 230012, China.
| | - Xiaojie Fu
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Hefei 230038, China
| | - Ruijuan Li
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Hefei 230038, China
| | - Hui Cheng
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Hefei 230038, China
| | - Meng Wang
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Hefei 230038, China
| | - An Zhou
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Hefei 230038, China
| | - Mei Zhang
- Oncology Department of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Qinglin Li
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Hefei 230038, China.
| |
Collapse
|
39
|
Mohamed Azar KAH, Ezhilarasan D, Shree Harini K. Coleus vettiveroides ethanolic root extract induces cytotoxicity by intrinsic apoptosis in HepG2 cells. J Appl Toxicol 2024; 44:245-259. [PMID: 37661188 DOI: 10.1002/jat.4536] [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: 04/19/2023] [Revised: 08/08/2023] [Accepted: 08/14/2023] [Indexed: 09/05/2023]
Abstract
Hepatocellular carcinoma (HCC) contributes to more than 80% of all primary cancers globally and ranks fourth in cancer-related deaths, due to the lack of an effective, definite therapeutic drug. Coleus vettiveroides (CV) has been used in Indian traditional medicine to treat diabetes, liver ailments, skin diseases, leukoderma, and leprosy. This study investigates the anticancer effect of CV ethanolic root extract in HepG2 cells. HepG2 cells were treated with CV extract, and its cytotoxicity was analyzed by MTT assay. AO/EB staining, propidium iodide staining, DCFH-DA assay, phalloidine staining, flow cytometry, and qPCR studies were performed for ROS expression, apoptosis and cell cycle analysis. The phytochemical analysis confirmed the presence of quercetin and galangin in CV root extract. The results showed that CV inhibited the proliferation of HepG2 cells, with altered cellular and nuclear morphology. CV was also found to increase intracellular ROS levels and oxidative stress markers in HepG2 cells. CV significantly altered the actin microfilament distribution in HepG2 cells and caused cell cycle arrest at the sub G0 -G1 phase. CV also induced mitochondria-mediated apoptosis, as evidenced by increased expression of p53, Bax, cytochrome C, Apaf-1, PARP, caspase-3 and caspase-9, and downregulated Bcl-2 expression. Therefore, CV exerts its anticancer effect by inducing mitochondrial dysfunction, oxidative stress, cytoskeletal disorganization, cell cycle arrest, and mitochondria-mediated apoptosis, and it could be a potent therapeutic option for HCC.
Collapse
Affiliation(s)
- Kadmad Abdul Hameed Mohamed Azar
- Department of Pharmacology, Koppal Institute of Medical Sciences, Koppal, India
- Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai, India
| | - Devaraj Ezhilarasan
- Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai, India
| | - Karthik Shree Harini
- Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai, India
| |
Collapse
|
40
|
Lin X, Zhang J, Chu Y, Nie Q, Zhang J. Berberine prevents NAFLD and HCC by modulating metabolic disorders. Pharmacol Ther 2024; 254:108593. [PMID: 38301771 DOI: 10.1016/j.pharmthera.2024.108593] [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: 10/25/2023] [Revised: 12/28/2023] [Accepted: 12/29/2023] [Indexed: 02/03/2024]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a global metabolic disease with high prevalence in both adults and children. Importantly, NAFLD is becoming the main cause of hepatocellular carcinoma (HCC). Berberine (BBR), a naturally occurring plant component, has been demonstrated to have advantageous effects on a number of metabolic pathways as well as the ability to kill liver tumor cells by causing cell death and other routes. This permits us to speculate and make assumptions about the value of BBR in the prevention and defense against NAFLD and HCC by a global modulation of metabolic disorders. Herein, we briefly describe the etiology of NAFLD and NAFLD-related HCC, with a particular emphasis on analyzing the potential mechanisms of BBR in the treatment of NAFLD from aspects including increasing insulin sensitivity, controlling the intestinal milieu, and controlling lipid metabolism. We also elucidate the mechanism of BBR in the treatment of HCC. More significantly, we provided a list of clinical studies for BBR in NAFLD. Taking into account our conclusions and perspectives, we can make further progress in the treatment of BBR in NAFLD and NAFLD-related HCC.
Collapse
Affiliation(s)
- Xinyue Lin
- School of Pharmacy, State Key Laboratory of Applied Organic Chemistry, and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Juanhong Zhang
- School of Pharmacy, State Key Laboratory of Applied Organic Chemistry, and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China; College of Life Science, Northwest Normal University, Lanzhou 730070, China
| | - Yajun Chu
- School of Pharmacy, State Key Laboratory of Applied Organic Chemistry, and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Qiuying Nie
- School of Pharmacy, State Key Laboratory of Applied Organic Chemistry, and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Junmin Zhang
- School of Pharmacy, State Key Laboratory of Applied Organic Chemistry, and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China.
| |
Collapse
|
41
|
Liu Y, Meng J, Ruan X, Wei F, Zhang F, Qin X. A disulfidptosis-related lncRNAs signature in hepatocellular carcinoma: prognostic prediction, tumor immune microenvironment and drug susceptibility. Sci Rep 2024; 14:746. [PMID: 38185671 PMCID: PMC10772085 DOI: 10.1038/s41598-024-51459-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 01/05/2024] [Indexed: 01/09/2024] Open
Abstract
Disulfidptosis, a novel type of programmed cell death, has attracted researchers' attention worldwide. However, the role of disulfidptosis-related lncRNAs (DRLs) in liver hepatocellular carcinoma (LIHC) not yet been studied. We aimed to establish and validate a prognostic signature of DRLs and analyze tumor microenvironment (TME) and drug susceptibility in LIHC patients. RNA sequencing data, mutation data, and clinical data were obtained from the Cancer Genome Atlas Database (TCGA). Lasso algorithm and cox regression analysis were performed to identify a prognostic DRLs signature. Kaplan-Meier curves, principal component analysis (PCA), nomogram and calibration curve, function enrichment, TME, immune dysfunction and exclusion (TIDE), tumor mutation burden (TMB), and drug sensitivity analyses were analyzed. External datasets were used to validate the predictive value of DRLs. qRT-PCR was also used to validate the differential expression of the target lncRNAs in tissue samples and cell lines. We established a prognostic signature for the DRLs (MKLN1-AS and TMCC1-AS1) in LIHC. The signature could divide the LIHC patients into low- and high-risk groups, with the high-risk subgroup associated with a worse prognosis. We observed discrepancies in tumor-infiltrating immune cells, immune function, function enrichment, and TIDE between two risk groups. LIHC patients in the high-risk group were more sensitive to several chemotherapeutic drugs. External datasets, clinical tissue, and cell lines confirmed the expression of MKLN1-AS and TMCC1-AS1 were upregulated in LIHC and associated with a worse prognosis. The novel signature based on the two DRLs provide new insight into LIHC prognostic prediction, TME, and potential therapeutic strategies.
Collapse
Affiliation(s)
- Yanqiong Liu
- Key Laboratory of Clinical Laboratory Medicine of Guangxi Department of Education, Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Jiyu Meng
- Key Laboratory of Clinical Laboratory Medicine of Guangxi Department of Education, Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Xuelian Ruan
- Key Laboratory of Clinical Laboratory Medicine of Guangxi Department of Education, Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Fangyi Wei
- Key Laboratory of Clinical Laboratory Medicine of Guangxi Department of Education, Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Fuyong Zhang
- Key Laboratory of Clinical Laboratory Medicine of Guangxi Department of Education, Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Xue Qin
- Key Laboratory of Clinical Laboratory Medicine of Guangxi Department of Education, Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China.
| |
Collapse
|
42
|
Li L, Qiu H. Asperulosidic Acid Restrains Hepatocellular Carcinoma Development and Enhances Chemosensitivity Through Inactivating the MEKK1/NF-κB Pathway. Appl Biochem Biotechnol 2024; 196:1-17. [PMID: 37097403 DOI: 10.1007/s12010-023-04500-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/11/2023] [Indexed: 04/26/2023]
Abstract
Asperulosidic acid (ASPA) is a plant-extracted iridoid terpenoid with tumor-suppressive and anti-inflammatory properties. At present, the antitumor function of ASPA and its related mechanisms in hepatocellular carcinoma (HCC) cells were explored. Human normal hepatocytes HL-7702 and HCC cells (Huh7 and HCCLM3) were treated with varying concentrations (0 to 200 μg/mL) of ASPA. Cell viability, proliferation, apoptosis, migration, and invasion were checked. The expression of proteins was detected by Western blot. Furthermore, the effect of ASPA (100 μg/mL) on the sensitivity of HCC cells to chemotherapeutic agents, including doxorubicin and cisplatin, was evaluated. A subcutaneous xenografted tumor model was set up in nude mice, and the antitumor effects of ASPA were evaluated. ASPA hindered HCC cells' proliferation, migration, and invasion, and amplified their apoptosis and sensitivity to chemotherapeutic agents. Additionally, ASPA inactivated the MEKK1/NF-κB pathway. Overexpression of MEKK1 increased HCC proliferation, migration, and invasion and facilitated chemoresistance. ASPA treatment alleviated the carcinogenic effect mediated by MEKK1 overexpression. MEKK1 knockdown slowed down HCC progression. However, ASPA could not exert additional antitumor effects in MEKK1 knockdown cells. In vivo results displayed that ASPA substantially curbed tumor growth and inactivated the MEKK1/NF-κB pathway in mice. All over, ASPA exerts antitumor effects in HCC by suppressing the MEKK1/NF-κB pathway.
Collapse
Affiliation(s)
- Liang Li
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jiaxing, Affiliated Hospital of Jiaxing University, No.1882 South Zhong Huan Road, Jiaxing City, Zhejiang Province, 314001, China
| | - Huiwen Qiu
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jiaxing, Affiliated Hospital of Jiaxing University, No.1882 South Zhong Huan Road, Jiaxing City, Zhejiang Province, 314001, China.
| |
Collapse
|
43
|
Jia G, Wang T, Li R, Li X, Sun G, Chen W, Peng Y, Cheng C, Yang J, Zuo C. Radioiodine-131-Labeled Theranostic Nanoparticles for Transarterial Radioembolization and Chemoembolization Combination Therapy of VX2 Liver Tumor. Adv Healthc Mater 2023; 12:e2301559. [PMID: 37807421 DOI: 10.1002/adhm.202301559] [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: 05/14/2023] [Revised: 09/16/2023] [Indexed: 10/10/2023]
Abstract
In interventional treatment, materials are administered into the blood supply artery and directly delivered to tumors, offering proper scenarios for nanomedicine potential clinical applications. Transarterial chemoembolization (TACE) and transarterial radioembolization (TARE) are effective treatment methods for hepatocellular carcinoma (HCC), but postoperative residual tumor may result in intrahepatic recurrence and distant metastasis. The combination therapy of TACE and TARE based on multifunctional nanoparticles (NPs) is expected to overcome the drug resistance in hypoxic tumors and improve the therapeutic effect. Herein, BaGdF5 NPs are synthesized and then coated with polydopamine (PDA), conjugated with the chemotherapeutic drug cis-diamminedichloride platinum (CDDP), radio-labeled with therapeutic radionuclide 131 I, yielding 131 I-BaGdF5 @PDA-CDDP NPs. The in vitro anti-cancer effects of 131 I-BaGdF5 @PDA-CDDP NPs are confirmed using CCK-8 and γ-H2AX assays in Huh7 cells. Mixed with Lipiodol, 131 I-BaGdF5 @PDA-CDDP NPs are injected into the hepatic artery via a microcatheter to realize the TACE and TARE combination therapy in a rabbit VX2 liver tumor model. The results indicate that glucose metabolism is clearly decreased based on 18 F-FDG PET imaging and the apoptosis of tumor cells is increased. Furthermore, 131 I and BaGdF5 NPs can be used for SPECT imaging and CT/MR imaging respectively, facilitating real-time monitoring of the in vivo biodistribution of 131 I-BaGdF5 @PDA-CDDP NPs.
Collapse
Affiliation(s)
- Guorong Jia
- Department of Nuclear Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, China
| | - Tao Wang
- Department of Nuclear Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, China
| | - Rou Li
- Department of Nuclear Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, China
| | - Xiao Li
- Department of Nuclear Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, China
| | - Gaofeng Sun
- Department of Nuclear Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, China
| | - Wei Chen
- Department of Radiology, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, China
| | - Ye Peng
- Department of Nuclear Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, China
| | - Chao Cheng
- Department of Nuclear Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, China
| | - Jijin Yang
- Department of Interventional Radiology, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, China
| | - Changjing Zuo
- Department of Nuclear Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, China
| |
Collapse
|
44
|
Liu J, Yan S, Zhang G, Yang L, Wei S, Yi P. A retrospective study of transarterial chemoembolization (TACE) combined with lenvatinib compared with TACE monotherapy for BCLC B2 stage hepatocellular carcinoma. Oncol Lett 2023; 26:507. [PMID: 37920437 PMCID: PMC10618929 DOI: 10.3892/ol.2023.14094] [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: 04/05/2023] [Accepted: 09/25/2023] [Indexed: 11/04/2023] Open
Abstract
The present study aimed to compare the efficacy and safety of combination therapy with lenvatinib (Len) plus transarterial chemoembolization (TACE) and TACE alone in patients with Barcelona Clinic Liver Cancer (BCLC) B2 stage hepatocellular carcinoma (HCC). A total of 66 patients with BCLC B2 stage HCC were retrospectively reviewed in the present study, of which 34 patients received Len + TACE, while 32 patients received TACE alone between May 2018 and May 2020. Survival outcome, tumor response and adverse events (AEs) were compared between the two treatment groups. The 6-month, 1- and 2-year overall survival (OS) rates were significantly higher in the Len + TACE group (97.1, 85.3 and 76.3%, respectively) compared with those in the TACE group [(93.8, 81.1 and 45.4%, respectively); hazard ratio (HR), 0.395; 95% confidence interval (CI), 0.180-0.867; P=0.023], but no significant difference in progression-free survival rate was observed between the two groups (HR, 0.815; 95% CI, 0.437-1.520; P=0.510). Patients receiving Len + TACE demonstrated a higher objective response rate compared with those receiving TACE alone (64.7 vs. 34.4%; P=0.014). Therefore, Len + TACE combination therapy was associated with increased OS and tumor response compared with that of TACE monotherapy in patients with BCLC B2 stage HCC. However, large-scale, multicenter, prospective studies are needed to further confirm these results.
Collapse
Affiliation(s)
- Junning Liu
- Department of Hepato-Biliary-Pancreas II, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Shu Yan
- Department of Hepato-Biliary-Pancreas II, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Guangnian Zhang
- Department of Hepato-Biliary-Pancreas II, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Linfeng Yang
- Department of Hepato-Biliary-Pancreas II, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Song Wei
- Department of Hepato-Biliary-Pancreas II, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Pengsheng Yi
- Department of Hepato-Biliary-Pancreas II, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| |
Collapse
|
45
|
Chung S, Gogna A, Chandramohan S, Lo R, Irani FG, Venkatanarasimha N. Review of outcomes of combination therapy using yttrium 90 radioembolization and sorafenib/nivolumab for HCC with hepatic vein or IVC invasion. PROCEEDINGS OF SINGAPORE HEALTHCARE 2023. [DOI: 10.1177/20101058231154666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Background A systematic review of the outcomes of combination therapy using Yttrium 90 radioembolization (Y90) and sorafenib/nivolumab for patients with hepatocellular carcinoma (HCC), with hepatic vein (HV) or inferior vena cava (IVC) invasion. The aim of this study is to summarise the results of different studies that used the combination therapy for HCC patients with tumor thrombosis involving the HV or IVC. Method A literature search was performed using keywords in Medline and Google Scholar limited to publications from 2010 to 2021. There were 173 articles identified during the initial literature search. During abstract screening, 81 articles were excluded. Another 83 did not contain information on hepatic vein or IVC invasion. Therefore, 9 articles met the eligibility criteria and were included in the synthesis. Results In total, 37 patients with hepatic vein or IVC invasion were identified. There were 31 patients who were given sorafenib, 7 were given nivolumab and 1 was given both sorafenib and nivolumab. Among the 37 patients, 21 had hepatic vein invasion, 22 had IVC invasion and 6 had both HV and IVC invasion. The median OS was 20.55 months and median PFS was 8.18 months. For the results, 23 patients were evaluated via modified RECIST (mRECIST) criteria and 14 were evaluated via RECIST 1.1. Conclusion The combination of local and systemic therapies demonstrated potential results for increased response rates, OS and PFS benefits. Further studies are required to determine the long-term outcomes of the combination therapy for this group of patients.
Collapse
Affiliation(s)
| | - Apoorva Gogna
- Department of Vascular and Interventional Radiology, Singapore General Hospital, Singapore
| | | | - Richard Lo
- Department of Vascular and Interventional Radiology, Singapore General Hospital, Singapore
| | - Farah Gillan Irani
- Department of Vascular and Interventional Radiology, Singapore General Hospital, Singapore
| | - Nanda Venkatanarasimha
- Department of Vascular and Interventional Radiology, Singapore General Hospital, Singapore
| |
Collapse
|
46
|
Ding J, Su Y, Liu Y, Xu Y, Yang D, Wang X, Hao S, Zhou H, Li H. The role of CSTF2 in cancer: from technology to clinical application. Cell Cycle 2023; 22:2622-2636. [PMID: 38166492 PMCID: PMC10936678 DOI: 10.1080/15384101.2023.2299624] [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: 03/17/2023] [Revised: 12/03/2023] [Accepted: 12/20/2023] [Indexed: 01/04/2024] Open
Abstract
A protein called cleavage-stimulating factor subunit 2 (CSTF2, additionally called CSTF-64) binds RNA and is needed for the cleavage and polyadenylation of mRNA. CSTF2 is an important component subunit of the cleavage stimulating factor (CSTF), which is located on the X chromosome and encodes 557 amino acids. There is compelling evidence linking elevated CSTF2 expression to the pathological advancement of cancer and on its impact on the clinical aspects of the disease. The progression of cancers, including hepatocellular carcinoma, melanoma, prostate cancer, breast cancer, and pancreatic cancer, is correlated with the upregulation of CSTF2 expression. This review provides a fresh perspective on the investigation of the associations between CSTF2 and various malignancies and highlights current studies on the regulation of CSTF2. In particular, the mechanism of action and potential clinical applications of CSTF2 in cancer suggest that CSTF2 can serve as a new biomarker and individualized treatment target for a variety of cancer types.
Collapse
Affiliation(s)
- Jiaxiang Ding
- Clinical Trial Center of the First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
- School of Public Foundation, Bengbu Medical University, Bengbu, Anhui, China
| | - Yue Su
- Clinical Trial Center of the First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
- School of Public Foundation, Bengbu Medical University, Bengbu, Anhui, China
| | - Youru Liu
- The People’s Hospital of Bozhou, Bozhou, Anhui, China
| | - Yuanyuan Xu
- Clinical Trial Center of the First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
- School of Pharmacy, Bengbu Medical University, Bengbu, Anhui, China
| | - Dashuai Yang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province; School of Pharmacy, Anhui Medical University, Hefei, Anhui, China
| | - Xuefeng Wang
- Clinical Trial Center of the First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
- School of Pharmacy, Bengbu Medical University, Bengbu, Anhui, China
| | - Shuli Hao
- The People’s Hospital of Bozhou, Bozhou, Anhui, China
| | - Huan Zhou
- Clinical Trial Center of the First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
- School of Public Foundation, Bengbu Medical University, Bengbu, Anhui, China
- School of Pharmacy, Bengbu Medical University, Bengbu, Anhui, China
| | - Hongtao Li
- Clinical Trial Center of the First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| |
Collapse
|
47
|
Li J, Zhou K, Wu M, Zhang R, Jin X, Qiao H, Li J, Cao X, Zhang S, Dong G. The Characteristics of Transcription Factors Regulating T Cell Exhaustion Were Analyzed to Predict the Prognosis and Therapeutic Effect in Patients with HCC. Int J Gen Med 2023; 16:5597-5619. [PMID: 38045905 PMCID: PMC10693252 DOI: 10.2147/ijgm.s435620] [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: 09/30/2023] [Accepted: 11/08/2023] [Indexed: 12/05/2023] Open
Abstract
Purpose Hepatocellular carcinoma (HCC) ranks as the third leading cause of cancer-related deaths, posing a significant threat to people in diverse regions. T-cell exhaustion (Tex) can hinder the efficacy of immunotherapy in patients with HCC, and the transcription factors that regulate Tex in HCC have not yet been fully elucidated. Patients and Methods We used the single sample gene set enrichment analysis (ssGSEA) method to define the transcription factor pathway that regulates Tex and employed LASSO regression analysis to establish Tex related genes (TEXRS). To predict differences in immunotherapy efficacy between the two groups, we used the immunophenotype score and submap algorithm. RT-qPCR was used to detect the expression levels of the model genes in 21 pairs of HCC tissues. Finally, we assessed the cell communication strength and identified ligand receptors using the "CellChat" R package. Results Nine Tex transcription factors were identified as regulators of the HCC immune microenvironment, with Tex scores affecting patient survival. Patients with a high Tex Risk Score (TEXRS) had significantly worse overall survival compared to patients with low TEXRS. After adjusting for confounding factors, TEXRS remained an independent prognostic factor. Importantly, TEXRS performed well in multiple independent external validation cohorts. Various algorithms have shown that patients in the low-TEXRS group might benefit more from immunotherapy. Finally, RT-qPCR analysis of 21 HCC samples showed that C7, CD5L, and SDS were significantly downregulated in HCC tissues, consistent with the bioinformatics analysis results. Conclusion TEXRS proved to be a valuable predictor of immunotherapy and transcatheter arterial chemoembolization efficacy in patients with HCC. This holds promise for enhancing the prognosis and treatment outcomes of patients with HCC.
Collapse
Affiliation(s)
- Jingbo Li
- Department of Anesthesiology Research Institute, The Second Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Kun Zhou
- Department of Clinical Laboratory, Beidahuang Industry Group General Hospital, Harbin, People’s Republic of China
- Scientific Research Center, The Second Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Meng Wu
- Scientific Research Center, The Second Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Rongzheng Zhang
- Scientific Research Center, The Second Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Xi Jin
- Scientific Research Center, The Second Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Han Qiao
- Scientific Research Center, The Second Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Jiaqi Li
- Scientific Research Center, The Second Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Xinyang Cao
- Scientific Research Center, The Second Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Shuyun Zhang
- Scientific Research Center, The Second Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Guanglu Dong
- Department of Tumor Radiotherapy, The Second Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| |
Collapse
|
48
|
Wu ZY, Wang Y, Hu H, Ai XN, Zhang Q, Qin YG. Long Noncoding RNA Cytoskeleton Regulator RNA Suppresses Apoptosis in Hepatoma Cells by Modulating the miR-125a-5p/HS1-Associated Protein X-1 Axis to Induce Caspase-9 Inactivation. Gut Liver 2023; 17:916-925. [PMID: 36700300 PMCID: PMC10651376 DOI: 10.5009/gnl210572] [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: 12/15/2021] [Revised: 04/01/2022] [Accepted: 05/13/2022] [Indexed: 01/27/2023] Open
Abstract
Background/Aims The involvement of long noncoding RNAs in the carcinogenesis of hepatocellular carcinoma (HCC) has been well documented by substantial evidence. However, whether cytoskeleton regulator RNA (CYTOR) could affect the progression of HCC remains unclear. Methods The relative expression of CYTOR, miR-125a-5p and HS1-associated protein X-1 (HAX-1) mRNA in HCC cells were determined via quantitative real-time polymerase chain reaction. The viability of treated HCC cells was measured by Cell Counting Kit-8 assay. Cell apoptosis was estimated by flow cytometry analysis, assessment of caspase-9 activity and terminal deoxynucleotidyl transferase dUTP nick-end labeling staining, and Western blot of apoptosis-related proteins. The interplay between CYTOR or HAX-1 and miR-125a-5p was validated by dual-luciferase reporter assay. Results CYTOR was upregulated and miR-125a-5p was downregulated in HCC cells. CYTOR silencing inhibited cell proliferation and promoted cell apoptosis in HepG2 and SMMC-7721 cells. miR-125a-5p was sponged and negatively regulated by CYTOR, and HAX-1 was directly targeted and negatively modulated by miR-125a-5p. Overexpression of miR-125a-5p enhanced the repressive effects of CYTOR knockdown on HCC cells, and knockdown of HAX-1 enhanced the inhibitory effects of miR-125a-5p mimics on HCC cells. Conclusions CYTOR silencing facilitates HCC cell apoptosis in vitro via the miR-125a-5p/HAX-1 axis.
Collapse
Affiliation(s)
- Zhen-Yu Wu
- Department of Hepatobiliary Surgery, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, China
| | - Yumin Wang
- Department of Respiratory and Critical Care Medicine, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, China
| | - Hao Hu
- Department of Hepatobiliary Surgery, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, China
| | - Xiang-Nan Ai
- Department of Hepatobiliary Surgery, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, China
| | - Qiang Zhang
- Department of Hepatobiliary Surgery, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, China
| | - Yu-Gang Qin
- Department of Hepatobiliary Surgery, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, China
| |
Collapse
|
49
|
Diggs LP, Fagenson AM, Putatunda V, Lau KN, Grandhi MS, Pitt HA. Intrahepatic cholangiocarcinoma: how do hepatectomy outcomes compare to liver metastases and hepatocellular carcinoma? HPB (Oxford) 2023; 25:1420-1428. [PMID: 37573232 DOI: 10.1016/j.hpb.2023.07.898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 05/25/2023] [Accepted: 07/20/2023] [Indexed: 08/14/2023]
Abstract
BACKGROUND The incidence of intrahepatic cholangiocarcinoma (ICC) continues to rise, and hepatectomy is the only cure. Perioperative outcomes following hepatectomy for colorectal liver metastases (CRLM) and hepatocellular carcinoma (HCC) are better described than for ICC. The aim was to compare post-hepatectomy outcomes for ICC to CRLM and HCC. METHODS The 2014-2020 ACS NSQIP hepatectomy PUF was utilized. Patients with ICC, CRLM, and HCC were identified and others excluded. Demographic, disease, and procedural characteristics were collected. Univariable and multivariable analyses (Chi-Square for categorical variables; Kruskal-Wallis for continuous variables) were performed for mortality, serious morbidity, bile leak, post-hepatectomy liver failure (PHLF), and 30-day readmission. RESULTS 17,789 patients underwent hepatectomy including 2377 for ICC, 10,195 for CRLM, and 5217 for HCC. Patients undergoing hepatectomy for ICC vs. HCC vs. CRLM were noted to have higher 30-day mortality (4.8% vs. 2.5% vs. 1.0%, respectively p < 0.05). ICC was associated with higher overall and serious morbidity, bile leak, severe PHLF, and readmission. Multivariable analyses confirmed higher odds ratios for mortality and morbidity (p < 0.05) in patients with ICC. CONCLUSION Hepatectomy for ICC is associated with worse short-term outcomes than for CRLM or HCC. Surgeons should be aware of these risks during surgical planning.
Collapse
Affiliation(s)
- Laurence P Diggs
- Department of Surgery, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Alexander M Fagenson
- Department of Surgery, Temple University Hospital, 3401NBroad Street, Philadelphia, PA, USA
| | - Vijay Putatunda
- Department of Surgery, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Kwan Nang Lau
- Department of Surgery, Temple University Hospital, 3401NBroad Street, Philadelphia, PA, USA
| | - Miral S Grandhi
- Department of Surgery, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA; Rutgers Cancer Institute of New Jersey, 195 Little Albany Street, New Brunswick, NJ 08901, USA
| | - Henry A Pitt
- Department of Surgery, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA; Rutgers Cancer Institute of New Jersey, 195 Little Albany Street, New Brunswick, NJ 08901, USA.
| |
Collapse
|
50
|
Cheng Q, Wang T, Zhang J, Tian L, Zeng C, Meng Z, Zhang C, Meng Q. Multifunctional gene delivery vectors containing different liver-targeting fragments for specifically transfecting hepatocellular carcinoma (HCC) cells. J Mater Chem B 2023; 11:9721-9731. [PMID: 37791430 DOI: 10.1039/d3tb01866k] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
Gene therapy is a promising strategy for HCC treatment, but it commonly faces the problem of low specificity in gene transfection. In this study, we designed and synthesized a series of peptide-based gene delivery vectors (H-01 to H-18) containing varied HCC cell-targeting fragments for specifically binding different receptors highly expressed on HCC cell membranes. The physicochemical properties of peptide vectors or peptide/DNA complexes were characterized, and the gene delivery abilities of peptide vectors were evaluated in HepG2 cell lines. The results showed that peptide vectors H-02 and H-09, which contained targeted fragments for ACE2 and c-Met receptors, respectively, could specifically transfect HCC cells in a highly -efficient manner in vitro. Furthermore, the liver-targeting function in vivo of Cy5.5 labeled H-02 (H-17) and H-09 (H-18) was investigated by fluorescence imaging.
Collapse
Affiliation(s)
- Qin Cheng
- State Key laboratory of Toxicology and Medical Countermeasures, Beijing institute of Pharmacology and Toxicology, Beijing, 100850, China.
- Key laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, 133002, China.
| | - Taoran Wang
- State Key laboratory of Toxicology and Medical Countermeasures, Beijing institute of Pharmacology and Toxicology, Beijing, 100850, China.
| | - Jing Zhang
- State Key laboratory of Toxicology and Medical Countermeasures, Beijing institute of Pharmacology and Toxicology, Beijing, 100850, China.
- Key laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, 133002, China.
| | - Long Tian
- State Key laboratory of Toxicology and Medical Countermeasures, Beijing institute of Pharmacology and Toxicology, Beijing, 100850, China.
- Key Laboratory of Structure-Based Drug Design and Discovery of the Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| | - Chunlan Zeng
- State Key laboratory of Toxicology and Medical Countermeasures, Beijing institute of Pharmacology and Toxicology, Beijing, 100850, China.
| | - Zhao Meng
- State Key laboratory of Toxicology and Medical Countermeasures, Beijing institute of Pharmacology and Toxicology, Beijing, 100850, China.
| | - Changhao Zhang
- Key laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, 133002, China.
| | - Qingbin Meng
- State Key laboratory of Toxicology and Medical Countermeasures, Beijing institute of Pharmacology and Toxicology, Beijing, 100850, China.
- Key Laboratory of Structure-Based Drug Design and Discovery of the Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| |
Collapse
|