1
|
Bhuia MS, Chowdhury R, Afroz M, Akbor MS, Al Hasan MS, Ferdous J, Hasan R, de Alencar MVOB, Mubarak MS, Islam MT. Therapeutic Efficacy Studies on the Monoterpenoid Hinokitiol in the Treatment of Different Types of Cancer. Chem Biodivers 2025:e202401904. [PMID: 39776341 DOI: 10.1002/cbdv.202401904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 12/10/2024] [Accepted: 12/15/2024] [Indexed: 01/11/2025]
Abstract
Hinokitiol (HK), a monoterpenoid that naturally occurs in plants belonging to the Cupressaceae family, possesses important biological activities, including an anticancer effect. This review summarizes its anticancer potential and draws possible molecular interventions. In addition, it evaluates the biopharmaceutical, toxicological properties, and clinical application of HK to establish its viability for future advancement as a dependable anticancer medication. The assessment is based on the most recent information available from various databases. Findings demonstrate that HK possesses substantial therapeutic advantages against diverse types of cancer (colon, cervical, breast, bone, endometrial, liver, prostate, oral, and skin) through various molecular mechanisms. HK induces oxidative stress, cytotoxicity, apoptosis, cell-cycle arrest at the G and S phases, and autophagy through modulation of phosphatidylinositol 3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR), p38/ERK/MAPK, nuclear factor kappa B, and c-Jun N-terminal kinase signaling pathways. Furthermore, this compound exhibits good oral bioavailability with excellent plasma clearance. Clinical uses of HK demonstrate therapeutic advantages without any significant negative effects. A thorough study of the pertinent data suggests that HK may serve as a viable candidate for developing novel cancer therapies. Consequently, more extensive studies are necessary to evaluate its cancer treatment efficacy, safety, and possible long-term hazards.
Collapse
Affiliation(s)
- Md Shimul Bhuia
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Dhaka, Bangladesh
- Phytochemistry and Biodiversity Research Laboratory, BioLuster Research Center Ltd, Gopalganj, Dhaka, Bangladesh
| | - Raihan Chowdhury
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Dhaka, Bangladesh
- Phytochemistry and Biodiversity Research Laboratory, BioLuster Research Center Ltd, Gopalganj, Dhaka, Bangladesh
| | - Meher Afroz
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Dhaka, Bangladesh
| | - Md Showkot Akbor
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Dhaka, Bangladesh
| | - Md Sakib Al Hasan
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Dhaka, Bangladesh
- Phytochemistry and Biodiversity Research Laboratory, BioLuster Research Center Ltd, Gopalganj, Dhaka, Bangladesh
| | - Jannatul Ferdous
- Department of Biotechnology and Genetic Engineering, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Dhaka, Bangladesh
| | - Rubel Hasan
- Phytochemistry and Biodiversity Research Laboratory, BioLuster Research Center Ltd, Gopalganj, Dhaka, Bangladesh
| | | | | | - Muhammad Torequl Islam
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Dhaka, Bangladesh
- Phytochemistry and Biodiversity Research Laboratory, BioLuster Research Center Ltd, Gopalganj, Dhaka, Bangladesh
- Pharmacy Discipline, Khulna University, Khulna, Dhaka, Bangladesh
| |
Collapse
|
2
|
Zafar S, Bai Y, Muhammad SA, Guo J, Khurram H, Zafar S, Muqaddas I, Shaikh RS, Bai B. Molecular dynamics simulation based prediction of T-cell epitopes for the production of effector molecules for liver cancer immunotherapy. PLoS One 2025; 20:e0309049. [PMID: 39752339 PMCID: PMC11698456 DOI: 10.1371/journal.pone.0309049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 08/05/2024] [Indexed: 01/06/2025] Open
Abstract
Liver cancer is the sixth most frequent malignancy and the fourth major cause of deaths worldwide. The current treatments are only effective in early stages of cancer. To overcome the therapeutic challenges and exploration of immunotherapeutic options, broad spectral therapeutic vaccines could have significant impact. Based on immunoinformatic and integrated machine learning tools, we predicted the potential therapeutic vaccine candidates of liver cancer. In this study, machine learning and MD simulation-based approach are effectively used to design T-cell epitopes that aid the immune system against liver cancer. Antigenicity, molecular weight, subcellular localization and expression site predictions were used to shortlist liver cancer associated proteins including AMBP, CFB, CDHR5, VTN, APOBR, AFP, SERPINA1 and APOE. We predicted CD8+ T-cell epitopes of these proteins containing LGEGATEAE, LLYIGKDRK, EDIGTEADV, QVDAAMAGR, HLEARKKSK, HLCIRHEMT, LKLSKAVHK, EQGRVRAAT and CD4+ T-cell epitopes of VLGEGATEA, WVTKQLNEI, VEEDTKVNS, FTRINCQGK, WGILGREEA, LQDGEKIMS, VKFNKPFVF, VRAATVGSL. We observed the substantial physicochemical properties of these epitopes with a significant binding affinity with MHC molecules. A polyvalent construct of these epitopes was designed using suitable linkers and adjuvant indicated significant binding energy (>-10.5 kcal/mol) with MHC class-I and II molecule. Based on in silico cloning, we found the considerable compatibility of this polyvalent construct with the E. coli expression system and the efficiency of its translation in host. The system-level and machine learning based cross validations showed the possible effect of these T-cell epitopes as potential vaccine candidates for the treatment of liver cancer.
Collapse
Affiliation(s)
- Sidra Zafar
- Institute of Molecular Biology and Biotechnology, Bahauddin Zakariya University Multan, Multan, Punjab, Pakistan
| | - Yuhe Bai
- Department of Computer Science, Sorbonne University, Paris, France
| | - Syed Aun Muhammad
- Institute of Molecular Biology and Biotechnology, Bahauddin Zakariya University Multan, Multan, Punjab, Pakistan
| | - Jinlei Guo
- School of Intelligent Medical Engineering, Sanquan College of Xinxiang Medical University, Xinxiang, Henan, China
| | - Haris Khurram
- Department of Mathematics and Computer Science, Faculty of Science and Technology, Prince of Songkla University, Pattani Campus, Pattani, Thailand
- Department of Sciences and Humanities, National University of Computer and Emerging Sciences, Chiniot, Punjab, Pakistan
| | - Saba Zafar
- Department of Biochemistry and Biotechnology, The Women University Multan, Multan, Punjab, Pakistan
| | - Iraj Muqaddas
- Institute of Molecular Biology and Biotechnology, Bahauddin Zakariya University Multan, Multan, Punjab, Pakistan
| | - Rehan Sadiq Shaikh
- Center for Applied Molecular Biology (CAMB), University of the Punjab, Lahore, Punjab, Pakistan
| | - Baogang Bai
- School of Information and Technology, Wenzhou Business College, Wenzhou, Zhejiang, China
- Zhejiang Province Engineering Research Center of Intelligent Medicine, Wenzhou, China
- The 1 School of Medical, School of Information and Engineering, The 1st Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
3
|
Lin FX, Gu HY, He W. MAPK signaling pathway in spinal cord injury: Mechanisms and therapeutic potential. Exp Neurol 2025; 383:115043. [PMID: 39522804 DOI: 10.1016/j.expneurol.2024.115043] [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: 10/06/2024] [Revised: 10/30/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
Spinal cord injury (SCI) is a severe disabling injury of the central nervous system that can lead to motor, sensory, and autonomic dysfunction below the level of the injury. According to its pathophysiological process, SCI can be divided into primary injury and secondary injury. Currently, multiple therapeutic strategies have been proposed to alleviate secondary injury and overcome the occurrence of neurodegenerative events. Although current treatment modalities have achieved varying degrees of success, they cannot effectively intervene or treat its pathological processes, which may be due to the complex treatment and protection mechanisms involved. Research has confirmed that signaling pathways play a crucial role in the pathological processes of SCI and the mechanisms of neuronal recovery. Mitogen-activated protein kinase (MAPK) signaling pathway plays a crucial role in neuronal differentiation, growth, survival and axon regeneration after central nervous system injury. Meanwhile, the MAPK signaling pathway is an important pathway closely related to the pathological processes of SCI. The MAPK signaling pathway is abnormally activated after SCI, and inhibiting the activity of MAPK pathway can effectively inhibit inflammation, oxidative stress, pain and apoptosis to promote the recovery of nerve function after SCI. Based on the role of the MAPK pathway in SCI, it may be a potential therapeutic target. This article summarizes the role and mechanism of MAPK pathway in SCI, and discusses the shortcomings and shortcomings of MAPK pathway in SCI field, as well as the potential challenges of targeting MAPK pathway in SCI treatment strategies. This article aims to elucidate the mechanism of the MAPK pathway in SCI to emphasize the role of targeting the MAPK pathway in the treatment of SCI, providing a theoretical basis for the MAPK pathway as a potential therapeutic target for SCI treatment.
Collapse
Affiliation(s)
- Fei-Xiang Lin
- Department of Spine Surgery, Ganzhou People's Hospital, 16 Meiguan Avenue, Ganzhou 341000, Jiangxi Province, PR China; Department of Spine Surgery, The Affiliated Ganzhou Hospital of Nanchang University (Ganzhou Hospital-Nanfang Hospital, Southern Medical University), 16 Meiguan Avenue, Ganzhou 341000, Jiangxi Province, PR China
| | - Hou-Yun Gu
- Department of Spine Surgery, Ganzhou People's Hospital, 16 Meiguan Avenue, Ganzhou 341000, Jiangxi Province, PR China; Department of Spine Surgery, The Affiliated Ganzhou Hospital of Nanchang University (Ganzhou Hospital-Nanfang Hospital, Southern Medical University), 16 Meiguan Avenue, Ganzhou 341000, Jiangxi Province, PR China
| | - Wei He
- Department of Spine Surgery, Ganzhou People's Hospital, 16 Meiguan Avenue, Ganzhou 341000, Jiangxi Province, PR China; Department of Spine Surgery, The Affiliated Ganzhou Hospital of Nanchang University (Ganzhou Hospital-Nanfang Hospital, Southern Medical University), 16 Meiguan Avenue, Ganzhou 341000, Jiangxi Province, PR China.
| |
Collapse
|
4
|
Chen YS, Lee CH, Hsieh YH, Chiou HL, Hung MC, Lee HL. Sorafenib, a Tyrosine Kinase Inhibitor, Synergistically Enhances the Ferroptosis Effects of Asiatic Acid in Hepatocellular Carcinoma Cells. ENVIRONMENTAL TOXICOLOGY 2025; 40:79-87. [PMID: 39264136 DOI: 10.1002/tox.24415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 07/03/2024] [Accepted: 07/23/2024] [Indexed: 09/13/2024]
Abstract
Hepatocellular carcinoma (HCC) remains one of the most common cancers worldwide. Asiatic acid (AA) is a natural triterpene, which is recognized as effect of antioxidant and antitumor. Sorafenib (Sor), an orally target drug, has been applicate for the HCC therapy. However, the synergistic effect of AA and Sor on human HCC is still unclear. Here, we explore the effect of combined treatment with AA and Sor in the HCC cell line SK-HEP-1 and HepG2. Compared with treating alone, our results demonstrated that AA combined with Sor synergistically inhibited proliferative rates in MTT assay and colony formation assay. We also found that AA combined with Sor in HCC cells strongly caused cell cycle arrest in G0/G1 phase and affected the protein level of cyclin D1 and SKP2. Furthermore, combination treatment strongly enhanced ferroptosis through cellular accumulation of iron ions, lipid peroxidation, and ferroptosis-related proteins (GPX4 and FTH1) in HCC cells. In addition, the combined treatment resulted in higher phosphorylation of JNK1/2 in the promotion of ferroptosis than drug treatment alone. These results indicate that AA combined with Sor synergistically improved ferroptosis in HCC cells through the regulation of JNK1/2 signaling. Taken together, the combinatorial strategy may serve as the potential treatment in HCC.
Collapse
Affiliation(s)
- Yong-Syuan Chen
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Chien-Hsing Lee
- Division of Pediatric Surgery, Children's Hospital of China Medical University, Taichung, Taiwan
- Department of Surgery, Children's Hospital of China Medical University, Taichung, Taiwan
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Yi-Hsien Hsieh
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Hui-Ling Chiou
- School of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung, Taiwan
| | - Ming-Chun Hung
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Hsiang-Lin Lee
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Surgery, Chung Shan Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
5
|
Yang S, Liu Y, Zhang B, Li J, Xu F, Yu M, Chen Y, Li C, Liu T, Zhao Y, Zhao Q, Zhang J. GRHPR, Targeted by miR-138-5p, Inhibits the Proliferation and Metastasis of Hepatocellular Carcinoma Through PI3K/AKT Signaling Pathway. Cancer Biother Radiopharm 2024; 39:733-744. [PMID: 38934120 DOI: 10.1089/cbr.2023.0018] [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: 06/28/2024] Open
Abstract
Background: Hepatocellular carcinoma (HCC) is a highly aggressive cancer. This study elucidates the role of Glyoxylate reductase/hydroxypyruvate reductase (GRHPR) in HCC proliferation and metastasis, along with its molecular mechanism, and identifies miRNAs targeting GRHPR. Materials and Methods: Expression levels of GRHPR and miR-138-5p were assessed using real-time fluorescent quantitative polymerase chain reaction and Western blot techniques. Bioinformatic analysis was employed to identify miRNAs targeting GRHPR, and the results were confirmed via dual-luciferase reporter assays. HCC cell lines overexpressing GRHPR were established to investigate its roles in cell proliferation, migration, and invasion. The biological function of miR-138-5p targeting GRHPR in HCC cells was also evaluated. Furthermore, a xenograft mouse model was utilized to examine the in vivo functions of GRHPR. Results: GRHPR expression was downregulated in HCC, whereas miR-138-5p was upregulated. Overexpression of GRHPR suppressed HCC cell proliferation, migration, and invasion. Conversely, inhibition of GRHPR by miR-138-5p promoted HCC cell proliferation and invasive properties. MiR-138-5p was found to regulate Phosphoinositide 3-kinase (PI3K) and protein kinase B (AKT) phosphorylation levels by inhibiting GRHPR expression. Conclusion: This study highlights GRHPR's role as a tumor suppressor in HCC, with its function being regulated by miR-138-5p.
Collapse
Affiliation(s)
- Shuangshuang Yang
- BGI College & Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yixian Liu
- BGI College & Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
- Henan Key Medical Laboratory of Tumor Molecular Biomarkers, Zhengzhou University, Zhengzhou, China
| | - Bushi Zhang
- BGI College & Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Jinxia Li
- School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Fang Xu
- BGI College & Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
- Henan Key Medical Laboratory of Tumor Molecular Biomarkers, Zhengzhou University, Zhengzhou, China
| | - Mengdan Yu
- School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Ying Chen
- Zhengzhou Railway Vocational and Technical College, Zhengzhou, China
| | - Chenglong Li
- School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Ting Liu
- BGI College & Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Ying Zhao
- BGI College & Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Qianwei Zhao
- BGI College & Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
- Henan Key Medical Laboratory of Tumor Molecular Biomarkers, Zhengzhou University, Zhengzhou, China
| | - Jintao Zhang
- BGI College & Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
- Henan Key Medical Laboratory of Tumor Molecular Biomarkers, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Tumor Epidemiology and State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
6
|
Peng C, Wang Y, Guo Y, Li J, Liu F, Fu Y, Yu Y, Zhang C, Fu J, Han F. A literature review on signaling pathways of cervical cancer cell death-apoptosis induced by Traditional Chinese Medicine. JOURNAL OF ETHNOPHARMACOLOGY 2024; 334:118491. [PMID: 38936644 DOI: 10.1016/j.jep.2024.118491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 06/16/2024] [Accepted: 06/22/2024] [Indexed: 06/29/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Cervical cancer (CC) is a potentially lethal disorder that can have serious consequences for a woman's health. Because early symptoms are typically only present in the middle to late stages of the disease, clinical diagnosis and treatment can be challenging. Traditional Chinese medicine (TCM) has been shown to have unique benefits in terms of alleviating cancer clinical symptoms, lowering the risk of recurrence after surgery, and reducing toxic side effects and medication resistance after radiation therapy. It has also been shown to improve the quality of life for patients. Because of its improved anti-tumor effectiveness and biosafety, it could be considered an alternative therapy option. This study examines how TCM causes apoptosis in CC cells via signal transduction, including the active components and medicinal tonics. It also intends to provide a reliable clinical basis and protocol selection for the TCM therapy of CC. METHODS The following search terms were employed in PubMed, Web of Science, Embase, CNKI, Wanfang, VIP, SinoMed, and other scientific databases to retrieve pertinent literature on "cervical cancer," "apoptosis," "signaling pathway," "traditional Chinese medicine," "herbal monomers," "herbal components," "herbal extracts," and "herbal formulas." RESULTS It has been demonstrated that herbal medicines can induce apoptosis in cells of the cervix, a type of cancer, by influencing the signaling pathways involved. CONCLUSION A comprehensive literature search was conducted, and 148 papers from the period between January 2017 and December 2023 were identified as eligible for inclusion. After a meticulous process of screening, elimination and summary, generalization, and analysis, it was found that TCM can regulate multiple intracellular signaling pathways and related molecular targets, such as STAT3, PI3K/AKT, Wnt/β-catenin, MAPK, NF-κB, p53, HIF-1α, Fas/FasL and so forth. This regulatory capacity was observed to induce apoptosis in cervical cancer cells. The study of the mechanism of TCM against cervical cancer and the screening of new drug targets is of great significance for future research in this field. The results of this study will provide ideas and references for the future development of Chinese medicine in the diagnosis and treatment of cervical cancer.
Collapse
Affiliation(s)
- Cheng Peng
- Department of Obstetrics and Gynecology, Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Yu Wang
- Department of Obstetrics and Gynecology, Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Ying Guo
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Jia Li
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Fangyuan Liu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Yang Fu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Yang Yu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Chengxin Zhang
- Department of Obstetrics and Gynecology, Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Jiangmei Fu
- Department of Obstetrics and Gynecology, Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Fengjuan Han
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, 150040, China.
| |
Collapse
|
7
|
Saber S, Abdelhady R, Elhemely MA, Elmorsy EA, Hamad RS, Abdel-Reheim MA, El-Kott AF, AlShehri MA, Morsy K, AlSheri AS, Youssef ME. PU-H71 (NSC 750424): a molecular masterpiece that targets HSP90 in cancer and beyond. Front Pharmacol 2024; 15:1475998. [PMID: 39564119 PMCID: PMC11573589 DOI: 10.3389/fphar.2024.1475998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Accepted: 10/22/2024] [Indexed: 11/21/2024] Open
Abstract
Heat shock protein 90 (HSP90) is a pivotal molecular chaperone with multifaceted roles in cellular health and disease. Herein, we explore how HSP90 orchestrates cellular stress responses, particularly through its partnership with heat shock factor 1 (HSF-1). PU-H71, a selective inhibitor of HSP90, demonstrates significant potential in cancer therapy by targeting a wide array of oncogenic pathways. By inducing the degradation of multiple client proteins, PU-H71 disrupts critical signaling pathways such as MAPK, PI3K/Akt, JAK/STAT, EGFR, and mTOR, which are essential for cancer cell survival, proliferation, and metastasis. We examined its impact on combating triple-negative breast cancer and enhancing the effectiveness of carbon-ion beam therapy, offering new avenues for cancer treatment. Furthermore, the dual inhibition of HSP90A and HSP90B1 by PU-H71 proves highly effective in the context of myeloma, providing fresh hope for patients with this challenging malignancy. We delve into its potential to induce apoptosis in B-cell lymphomas that rely on Bcl6 for survival, highlighting its relevance in the realm of hematologic cancers. Shifting our focus to hepatocellular carcinoma, we explore innovative approaches to chemotherapy. Moreover, the current review elucidates the potential capacity of PU-H71 to suppress glial cell activation paving the way for developing novel therapeutic strategies for neuroinflammatory disorders. Additionally, the present report also suggests the promising role of PU-H71 in JAK2-dependent myeloproliferative neoplasms. Eventually, our report sheds more light on the multiple functions of HSP90 protein as well as the potential therapeutic benefit of its selective inhibitor PU-H71 in the context of an array of diseases, laying the foundations for the development of novel therapeutic approaches that could achieve better treatment outcomes.
Collapse
Affiliation(s)
- Sameh Saber
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, Egypt
| | - Rasha Abdelhady
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Fayoum University, Fayoum, Egypt
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Egyptian Chinese University, Cairo, Egypt
| | - Mai A Elhemely
- School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Elsayed A Elmorsy
- Department of Pharmacology and Therapeutics, College of Medicine, Qassim University, Buraidah, Saudi Arabia
- Department of Clinical Pharmacology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Rabab S Hamad
- Biological Sciences Department, College of Science, King Faisal University, Al Ahsa, Saudi Arabia
- Central Laboratory, Theodor Bilharz Research Institute, Giza, Egypt
| | - Mustafa Ahmed Abdel-Reheim
- Department of Pharmaceutical Sciences, College of Pharmacy, Shaqra University, Shaqra, Saudi Arabia
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni Suef, Egypt
| | - Attalla F El-Kott
- Department of Biology, College of Science, King Khalid University, Abha, Saudi Arabia
- Department of Zoology, Faculty of Science, Damanhour University, Damanhour, Egypt
| | - Mohammed A AlShehri
- Department of Biology, College of Science, King Khalid University, Abha, Saudi Arabia
| | - Kareem Morsy
- Department of Biology, College of Science, King Khalid University, Abha, Saudi Arabia
- Department of Zoology, Faculty of Science, Cairo University, Cairo, Egypt
| | - Ali S AlSheri
- Department of Biology, College of Science, King Khalid University, Abha, Saudi Arabia
| | - Mahmoud E Youssef
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, Egypt
| |
Collapse
|
8
|
Hatawsh A, Al-Haddad RH, Okafor UG, Diab LM, Dekanoidze N, Abdulwahab AA, Mohammed OA, Doghish AS, Moussa R, Elimam H. Mitoepigenetics pathways and natural compounds: a dual approach to combatting hepatocellular carcinoma. Med Oncol 2024; 41:302. [PMID: 39465473 DOI: 10.1007/s12032-024-02538-8] [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: 08/27/2024] [Accepted: 10/07/2024] [Indexed: 10/29/2024]
Abstract
Hepatocellular carcinoma (HCC) is a leading liver cancer that significantly impacts global life expectancy and remains challenging to treat due to often late diagnoses. Despite advances in treatment, the prognosis is still poor, especially in advanced stages. Studies have pointed out that investigations into the molecular mechanisms underlying HCC, including mitochondrial dysfunction and epigenetic regulators, are potentially important targets for diagnosis and therapy. Mitoepigenetics, or the epigenetic modifications of mitochondrial DNA, have drawn wide attention for their role in HCC progression. Besides, molecular biomarkers such as mitochondrial DNA alterations and non-coding RNAs showed early diagnosis and prognosis potential. Additionally, natural compounds like alkaloids, resveratrol, curcumin, and flavonoids show promise in HCC show promise in modulating mitochondrial and epigenetic pathways involved in cancer-related processes. This review discusses how mitochondrial dysfunction and epigenetic modifications, especially mitoepigenetics, influence HCC and delves into the potential of natural products as new adjuvant treatments against HCC.
Collapse
Affiliation(s)
- Abdulrahman Hatawsh
- Biotechnology School, Nile University, 26th of July Corridor, Sheikh Zayed City, Giza, 12588, Egypt
| | - Roya Hadi Al-Haddad
- Research and Technology Center of Environment, Water and Renewable Energy, Scientific Research Commission, Baghdad, Iraq
| | | | - Lamis M Diab
- Department of Medical Biochemistry, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | | | | | - Osama A Mohammed
- Department of Pharmacology, College of Medicine, University of Bisha, 61922, Bisha, Saudi Arabia
| | - Ahmed S Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo, 11829, Egypt.
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, Cairo, 11231, Egypt.
| | - Rewan Moussa
- Faculty of Medicine, Helwan University, Helwan, Cairo, 11795, Egypt
| | - Hanan Elimam
- Department of Biochemistry, Faculty of Pharmacy, University of Sadat City, Sādāt, 32897, Egypt.
| |
Collapse
|
9
|
Chu X, Wu Q, Kong L, Peng Q, Shen J. Multiomics Analysis Identifies Prognostic Signatures for Sepsis-Associated Hepatocellular Carcinoma in Emergency Medicine. Emerg Med Int 2024; 2024:1999820. [PMID: 39421149 PMCID: PMC11486536 DOI: 10.1155/2024/1999820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 08/06/2024] [Accepted: 08/30/2024] [Indexed: 10/19/2024] Open
Abstract
Objectives Sepsis, caused by the body's response to infection, poses a life-threatening condition and represents a significant global health challenge. Characterized by dysregulated immune response to infection, sepsis may lead to organ dysfunction and failure, ultimately resulting in high mortality rates. The liver plays a crucial role in sepsis, yet the role of differentially expressed genes in septic patients remains unclear in hepatocellular carcinoma (HCC). In this study, we aim to investigate the significance of differentially expressed genes related to sepsis in the occurrence and prognosis of tumors in HCC. Methods We conducted analyses by obtaining gene transcriptome data and clinical data of HCC cases from The Cancer Genome Atlas (TCGA). Furthermore, we obtained transcriptomic sequencing results of septic patients from the Gene Expression Omnibus (GEO) database, identified intersecting differentially expressed genes between the two, and performed survival analysis on the samples using LASSO and Cox regression analysis. Combining analyses of tumor mutation burden (TMB) and immune function, we further elucidated the mechanisms of sepsis-related genes in the prognosis and treatment of HCC. Results We established a prognostic model consisting of four sepsis-related genes: KRT20, PAEP, CCR3, and ANLN. Both the training and validation sets showed excellent outcomes in the prognosis of tumor patients, with significantly longer survival times observed in the low-risk group based on this model compared to the high-risk group. Furthermore, analyses, such as differential analysis of tumor mutation burden, immune function analysis, GO/KEGG pathway enrichment analysis, and drug sensitivity analysis, also demonstrated the potential mechanisms of action of sepsis-related genes. Conclusions Models constructed based on sepsis-related genes have shown excellent predictive ability in prognosis and differential analysis of drug sensitivity among tumor patients. These predictive models can enhance patient prognosis and inform the creation of early treatment protocols for sepsis, consequently aiding in the prevention of sepsis-induced HCC development through the modulation of the overall immune status. This may play a crucial role in patient management and immunotherapy, providing valuable reference for subsequent research.
Collapse
Affiliation(s)
- Xin Chu
- Department of Emergency, The Second Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, China
| | - Qi Wu
- Department of Emergency, The Second Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, China
| | - Linglin Kong
- Department of Infectious Disease, The Second Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, China
| | - Qiang Peng
- Department of Emergency, The Second Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, China
| | - Junhua Shen
- Department of Emergency, The Second Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, China
| |
Collapse
|
10
|
Kale SR, Karande G, Gudur A, Garud A, Patil MS, Patil S. Recent Trends in Liver Cancer: Epidemiology, Risk Factors, and Diagnostic Techniques. Cureus 2024; 16:e72239. [PMID: 39583507 PMCID: PMC11584332 DOI: 10.7759/cureus.72239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 10/23/2024] [Indexed: 11/26/2024] Open
Abstract
Liver cancer, particularly hepatocellular carcinoma (HCC), poses a significant global health challenge due to its high mortality rate. Hepatocellular carcinoma and intrahepatic cholangiocarcinoma (ICC) are the two main types of primary liver cancer (PLC), each with its own set of complexities. Secondary or metastatic liver cancer is more common than PLC. It is frequently observed in malignancies such as colorectal, pancreatic, melanoma, lung, and breast cancer. Liver cancer is often diagnosed at an advanced stage, making it difficult to treat. This highlights the need for focused research on early detection and effective treatment strategies. This review explores the epidemiology, risk factors, and diagnostic techniques for HCC. The development of HCC involves various risk factors, including chronic liver diseases, hepatitis B and C infections, alcohol consumption, obesity, smoking, and genetic predispositions. Various invasive and non-invasive diagnostic techniques, such as biopsy, liquid biopsy, and imaging modalities like ultrasonography, computed tomography scans (CT scans), magnetic resonance imaging (MRI), and positron emission tomography (PET) scans, are utilized for HCC detection and monitoring. Advances in imaging technology and biomarker research have led to more accurate and sensitive methods for early HCC detection. We also reviewed advanced research on emerging techniques, including next-generation sequencing, metabolomics, epigenetic biomarkers, and microbiome analysis, which show great potential for advancing early diagnosis and personalized treatment strategies. This literature review provides insights into the current state of liver cancer diagnosis and promising future advancements. Ongoing research and innovation in these areas are essential for improving early diagnosis and reducing the global burden of liver cancer.
Collapse
Affiliation(s)
- Shivani R Kale
- Molecular Biology and Genetics, Krishna Institute of Medical Sciences, Krishna Vishwa Vidyapeeth (Deemed to be University), Karad, IND
| | - Geeta Karande
- Microbiology, Krishna Institute of Medical Sciences, Krishna Vishwa Vidyapeeth (Deemed to be University), Karad, IND
| | - Anand Gudur
- Oncology, Krishna Institute of Medical Sciences, Krishna Vishwa Vidyapeeth (Deemed to be University), Karad, IND
| | - Aishwarya Garud
- Molecular Biology and Genetics, Krishna Institute of Medical Sciences, Krishna Vishwa Vidyapeeth (Deemed to be University), Karad, IND
| | - Monika S Patil
- Molecular Biology and Genetics, Krishna Institute of Medical Sciences, Krishna Vishwa Vidyapeeth (Deemed to be University), Karad, IND
| | - Satish Patil
- Microbiology, Krishna Institute of Medical Sciences, Krishna Vishwa Vidyapeeth (Deemed to be University), Karad, IND
| |
Collapse
|
11
|
Jin Q, Jiao W, Lian Y, Chitrakar B, Sang Y, Wang X. Study on antihepatocellular carcinoma effect of 6-shogaol and curcumin through network-based pharmacological and cellular assay. Front Pharmacol 2024; 15:1367417. [PMID: 39224778 PMCID: PMC11368042 DOI: 10.3389/fphar.2024.1367417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 07/26/2024] [Indexed: 09/04/2024] Open
Abstract
Background Hepatocellular carcinoma currently has the third highest mortality rate in the world. Patients with hepatocellular carcinoma are on the rise and at a younger age, but research into the pharmacological effects of cancer is mostly single-component, and natural plant products can have additive or synergistic effects that can better amplify the effects of intervention in cancer. Aim To evaluate the synergistic therapeutic effects of 6-shogaol and curcumin against hepatocellular carcinoma line HepG2 cells. Methods In this study, a network pharmacology approach was used to predict and validate the mol ecular targets and pathways of the hepatocellular carcinoma (HCC) of 6-shogaol and curcumin in combination and to investigate their mechanism of action. The results were also validated by cellular assays. HepG2 cells were treated with 6-shogaol and curcumin as well as the combination of the two. The combination index of 6-shogaol and curcumin in HepG2 cells was calculated using Compusyn software according to the Chou-Talalay equation. The synergistic anti-cancer effect was next investigated by MTT assay, apoptosis assay and cell cycle assay. The combined anti-hepatocellular carcinoma effect of the Ras-mediated PI3K/AKT and MAPK signalling pathways was analysed using protein blotting assays. Results A network pharmacology-based screening identified 72 core targets of 6-curcumin and curcumin in hepatocellular carcinoma, and predicted that the main signalling pathway is the Ras signalling pathway. The anti-cancer effects of 6-shogaol and curcumin were validated in cell-based assays and the optimal synergistic concentrations of 5 μmoL/L for 6-shogaol and 30 μmoL/L for curcumin were determined. 6-shogaol and curcumin synergistically blocked the cell cycle in the G2/M phase and promoted apoptosis. Immunoblot analysis confirmed for the first time the combined action of both in down-regulating the Ras-mediated PI3K/AKT and MAPK signaling pathways. In addition, 6-shogaol and curcumin acting together downregulated Cyclin-B, CDK-1, Bcl-2, and upregulated BAX. Conclusion 6-shogaol and curcumin act synergistically to alter the morphology of hepatocellular carcinoma cells, block the cell cycle in the G2/M phase, inhibit proliferation and division, and effectively promote late apoptosis. The combined action of these two components provides a theoretical basis for the further development of novel anti-liver cancer products.
Collapse
Affiliation(s)
- Qiuxia Jin
- College of Food Science and Technology, Hebei Agricultural University, Baoding, China
| | - Wenya Jiao
- College of Food Science and Technology, Hebei Agricultural University, Baoding, China
| | - Yunhe Lian
- Chenguang Biotechnology Group Co., Ltd., Handan, China
| | - Bimal Chitrakar
- College of Food Science and Technology, Hebei Agricultural University, Baoding, China
| | - Yaxin Sang
- College of Food Science and Technology, Hebei Agricultural University, Baoding, China
| | - Xianghong Wang
- College of Food Science and Technology, Hebei Agricultural University, Baoding, China
| |
Collapse
|
12
|
Liu T, Meng G, Ma S, You J, Yu L, He R, Zhao X, Cui Y. Progress of immune checkpoint inhibitors in the treatment of advanced hepatocellular carcinoma. Front Immunol 2024; 15:1455716. [PMID: 39185414 PMCID: PMC11341420 DOI: 10.3389/fimmu.2024.1455716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 07/30/2024] [Indexed: 08/27/2024] Open
Abstract
Among primary liver cancers, hepatocellular carcinoma is the most common pathological type. Its onset is insidious, and most patients have no obvious discomfort in the early stage, so it is found late, and the opportunity for surgical radical treatment is lost, resulting in a poor prognosis. With the introduction of molecular-targeted drugs represented by sorafenib, patients with middle- and late-stage liver cancer have regained the light of day. However, their therapeutic efficacy is relatively low due to the limited target of drug action, toxic side effects, and other reasons. At this time, the emergence of immunotherapy represented by immune checkpoint inhibitors (ICIs) well breaks this embarrassing situation, which mainly achieves the anti-tumor purpose by improving the tumor immune microenvironment. Currently, ICI monotherapy, as well as combination therapy, has been widely used in the clinic, further prolonging the survival of patients with advanced hepatocellular carcinoma. This article reviews the development of monotherapy and combination therapy for ICIs in advanced hepatocellular carcinoma and the latest research progress.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Yunfu Cui
- Department of Hepatopancreatobiliary Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
13
|
Kotulkar M, Paine-Cabrera D, Apte U. Role of Hepatocyte Nuclear Factor 4 Alpha in Liver Cancer. Semin Liver Dis 2024; 44:383-393. [PMID: 38901435 DOI: 10.1055/a-2349-7236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/22/2024]
Abstract
Liver cancer is the sixth most common cancer and the fourth leading cause of cancer-related deaths worldwide. Hepatocellular carcinoma (HCC) is the most prevalent primary liver cancer and the incidence of HCC is on the rise. Liver cancers in general and HCC in particular do not respond to chemotherapy. Radiological ablation, surgical resection, and liver transplantation are the only medical therapies currently available. Hepatocyte nuclear factor 4 α (HNF4α) is an orphan nuclear receptor expressed only in hepatocytes in the liver. HNF4α is considered the master regulator of hepatic differentiation because it regulates a significant number of genes involved in various liver-specific functions. In addition to maintaining hepatic differentiation, HNF4α also acts as a tumor suppressor by inhibiting hepatocyte proliferation by suppressing the expression of promitogenic genes and inhibiting epithelial to mesenchymal transition in hepatocytes. Loss of HNF4α expression and function is associated with rapid progression of chronic liver diseases that ultimately lead to liver cirrhosis and HCC, including metabolism-associated steatohepatitis, alcohol-associated liver disease, and hepatitis virus infection. This review summarizes the role of HNF4α in liver cancer pathogenesis and highlights its potential as a potential therapeutic target for HCC.
Collapse
Affiliation(s)
- Manasi Kotulkar
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas
| | - Diego Paine-Cabrera
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas
| | - Udayan Apte
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas
| |
Collapse
|
14
|
Sundi PRIO, Thipe VC, Omar MA, Adelusi TI, Gedefa J, Olaoba OT. Preclinical human and murine models of hepatocellular carcinoma (HCC). Clin Res Hepatol Gastroenterol 2024; 48:102418. [PMID: 39004339 DOI: 10.1016/j.clinre.2024.102418] [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: 12/09/2023] [Revised: 05/17/2024] [Accepted: 07/04/2024] [Indexed: 07/16/2024]
Abstract
Hepatocellular carcinoma (HCC) is the most frequent liver cancer, which account for more than 90 % of all liver cancer cases. It is the fifth leading cause of cancer globally and the second leading cause of cancer-related mortality in men. The availability of competent HCC preclinical models is fundamental to the success of mechanistic studies, molecular target identification, and drug testing. However, there are challenges associated with the use of these models. In this review, we provided updates on various cell lines, animals, and human HCC models, their specific preclinic use and associated potential challenges. Overall, the understanding of the merits and demerits of a particular HCC model will improve model selection for various preclinical studies.
Collapse
Affiliation(s)
- Pharidah Rajan Ibrahim Omar Sundi
- Lusaka Apex Medical University, Off Mumbwa Road, Lusaka 10101, Zambia; Pan African Organization for Health, Education and Research (POHER), United States
| | - Velaphi C Thipe
- Department of Radiology, Institute of Green Nanotechnology and Cancer Nanotechnology, University of Missouri, Columbia, MO 65211, USA
| | | | | | - Jalene Gedefa
- Collage of Health Sciences, Addis Ababa University, Ethiopia
| | - Olamide T Olaoba
- Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, MO 65211, USA.
| |
Collapse
|
15
|
Li C, Zhang ED, Ye Y, Xiao Z, Huang H, Zeng Z. Association of mitochondrial phosphoenolpyruvate carboxykinase with prognosis and immune regulation in hepatocellular carcinoma. Sci Rep 2024; 14:14051. [PMID: 38890507 PMCID: PMC11189538 DOI: 10.1038/s41598-024-64907-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 06/14/2024] [Indexed: 06/20/2024] Open
Abstract
Mitochondrial phosphoenolpyruvate carboxykinase (PCK2), a mitochondrial isoenzyme, supports the growth of cancer cells under glucose deficiency conditions in vitro. This study investigated the role and potential mechanism of PCK2 in the occurrence and development of Hepatocellular carcinoma (HCC). The Cancer Genome Atlas (TCGA), Gene Expression Omnibus (GEO), and other databases distinguish the expression of PCK2 and verified by qRT-PCR and Western blotting. Kaplan-Meier was conducted to assess PCK2 survival in HCC. The potential biological function of PCK2 was verified by enrichment analysis and gene set enrichment analysis (GSEA). The correlation between PCK2 expression and immune invasion and checkpoint was found by utilizing Tumor Immune Estimation Resource (TIMER). Lastly, the effects of PCK2 on the proliferation and metastasis of hepatocellular carcinoma cells were evaluated by cell tests, and the expressions of Epithelial mesenchymal transformation (EMT) and apoptosis related proteins were detected. PCK2 is down-regulated in HCC, indicating a poor prognosis. PCK2 gene mutation accounted for 1.3% of HCC. Functional enrichment analysis indicated the potential of PCK2 as a metabolism-related therapeutic target. Subsequently, we identified several signaling pathways related to the biological function of PCK2. The involvement of PCK2 in immune regulation was verified and key immune checkpoints were predicted. Ultimately, after PCK2 knockdown, cell proliferation and migration were significantly increased, and N-cadherin and vimentin expression were increased. PCK2 has been implicated in immune regulation, proliferation, and metastasis of hepatocellular carcinoma, and is emerging as a novel predictive biomarker and metabolic-related clinical target.
Collapse
Affiliation(s)
| | | | - Youzhi Ye
- Kunming Medical University, Kunming, China
| | | | - Hanfei Huang
- The First Affiliated Hospital of Kunming Medical University, Kunming Medical University, Kunming, China.
| | - Zhong Zeng
- The First Affiliated Hospital of Kunming Medical University, Kunming Medical University, Kunming, China.
| |
Collapse
|
16
|
Liu BJ, Guan YY, Qiao LX, Zhang JM, Li AJ, Yang PX, Gao YX, Chen DX, Wang CX, Wu J. The mechanism and experimental verification of Ixeris sonchifolia promoting apoptosis of hepatocellular carcinoma based on network pharmacology: Ixeris sonchifolia Induces Hepatocellular Carcinoma Apoptosis via the PI3K/AKT Pathway. JOURNAL OF ETHNOPHARMACOLOGY 2024; 327:117994. [PMID: 38437889 DOI: 10.1016/j.jep.2024.117994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 02/22/2024] [Accepted: 02/26/2024] [Indexed: 03/06/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ixeris sonchifolia alias Kudiezi, it was named Ixeris sonchifolia (Bunge) Hance, a synonym for Crepidiastrum sonchifolium (Bunge) Pak & Kawano in the https://www.iplant.cn/. And it was first published in J. Linn. Soc., Bot. 13: 108 (1873), which was named Ixeris sonchifolia (Maxim.) Hance in the MPNS (http://mpns.kew.org). As a widely distributed medicinal and edible wild plant, it possesses unique bitter-cold characteristics and constituents with various pharmacological activities. Its main antitumor substances, same as artemisinin and paclitaxel, are classified as terpenoids and have become research foci in recent years. However, its specific biological activity and role in antitumor treatment remain largely unclear. AIM OF THE STUDY This study aimed to elucidate the molecular targets and potential mechanisms of hepatocellular carcinoma apoptosis induced by Ixeris sonchifolia. MATERIALS AND METHODS We used network pharmacology methods to analyze and screen the active ingredients and possible underlying mechanisms of Ixeris sonchifolia in treating liver cancer and employed integrative time- and dose-dependent toxicity, transcriptomics, and molecular biology approaches to comprehensively verify the function of Ixeris sonchifolia extract (IsE) in human hepatoblastoma cell (HepG2) apoptosis and its potential mechanism. RESULTS A total of 169 common targets were screened by network pharmacology, and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis showed that IsE inhibited HepG2 cell activity in a time- and dose-dependent manner. Western blot analysis confirmed that IsE promoted HepG2 cell apoptosis by inhibiting the PI3K/AKT signaling pathway and that the PI3K/AKT inhibitor LY294002 also substantially enhanced IsE-induced apoptosis. The PI3K/AKT signaling pathway exhibited significant differences compared to that in the control group. CONCLUSION Combining network pharmacology with experimental verification, IsE inhibited mitochondrial function and the PI3K/AKT pathway while inducing hepatoma cell apoptosis. IsE may have promising potential for liver cancer treatment and chemoprevention.
Collapse
Affiliation(s)
- Bao-Jun Liu
- Department of Head, Neck and Genitourinary Oncology, Harbin Medical University Cancer Hospital, Heilongjiang, 150081, China; Department of Oncology, The Second Affiliated Hospital of Shandong First Medical University, Shandong, 271000, China
| | - Yuan-Yue Guan
- Department of Beijing Institute of Hepatology, Beijing You an Hospital, Capital Medical University, Beijing, 100069, China
| | - Lu-Xin Qiao
- Department of Beijing Institute of Hepatology, Beijing You an Hospital, Capital Medical University, Beijing, 100069, China
| | - Ji-Mei Zhang
- School of Pharmacy, Shandong First Medical University, Shandong, 271000, China
| | - Ai-Ju Li
- Department of Oncology, The Second Affiliated Hospital of Shandong First Medical University, Shandong, 271000, China
| | - Peng-Xiang Yang
- Department of Beijing Institute of Hepatology, Beijing You an Hospital, Capital Medical University, Beijing, 100069, China
| | - Yu-Xue Gao
- Department of Beijing Institute of Hepatology, Beijing You an Hospital, Capital Medical University, Beijing, 100069, China
| | - De-Xi Chen
- Department of Beijing Institute of Hepatology, Beijing You an Hospital, Capital Medical University, Beijing, 100069, China.
| | - Chun-Xiao Wang
- Department of Pharmacy, The Second Affiliated Hospital of Shandong First Medical University, Shandong, 271000, China.
| | - Jin Wu
- Department of Head, Neck and Genitourinary Oncology, Harbin Medical University Cancer Hospital, Heilongjiang, 150081, China.
| |
Collapse
|
17
|
Lin M, Zheng X, Yan J, Huang F, Chen Y, Ding R, Wan J, Zhang L, Wang C, Pan J, Cao X, Fu K, Lou Y, Feng XH, Ji J, Zhao B, Lan F, Shen L, He X, Qiu Y, Jin J. The RNF214-TEAD-YAP signaling axis promotes hepatocellular carcinoma progression via TEAD ubiquitylation. Nat Commun 2024; 15:4995. [PMID: 38862474 PMCID: PMC11167002 DOI: 10.1038/s41467-024-49045-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Accepted: 05/22/2024] [Indexed: 06/13/2024] Open
Abstract
RNF214 is an understudied ubiquitin ligase with little knowledge of its biological functions or protein substrates. Here we show that the TEAD transcription factors in the Hippo pathway are substrates of RNF214. RNF214 induces non-proteolytic ubiquitylation at a conserved lysine residue of TEADs, enhances interactions between TEADs and YAP, and promotes transactivation of the downstream genes of the Hippo signaling. Moreover, YAP and TAZ could bind polyubiquitin chains, implying the underlying mechanisms by which RNF214 regulates the Hippo pathway. Furthermore, RNF214 is overexpressed in hepatocellular carcinoma (HCC) and inversely correlates with differentiation status and patient survival. Consistently, RNF214 promotes tumor cell proliferation, migration, and invasion, and HCC tumorigenesis in mice. Collectively, our data reveal RNF214 as a critical component in the Hippo pathway by forming a signaling axis of RNF214-TEAD-YAP and suggest that RNF214 is an oncogene of HCC and could be a potential drug target of HCC therapy.
Collapse
Affiliation(s)
- Mengjia Lin
- State Key Laboratory for Diagnosis and Treatment of Infectious Disease, and National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310000, Zhejiang, China
- Life Sciences Institute, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Xiaoyun Zheng
- Life Sciences Institute, Zhejiang University, Hangzhou, 310058, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Jianing Yan
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, Zhejiang, China
| | - Fei Huang
- Life Sciences Institute, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Yilin Chen
- Life Sciences Institute, Zhejiang University, Hangzhou, 310058, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Ran Ding
- Life Sciences Institute, Zhejiang University, Hangzhou, 310058, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Jinkai Wan
- International Co-laboratory of Medical Epigenetics and Metabolism of Ministry of Science and Technology, and Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, and Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Lei Zhang
- International Co-laboratory of Medical Epigenetics and Metabolism of Ministry of Science and Technology, and Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Chenliang Wang
- Life Sciences Institute, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Jinchang Pan
- Life Sciences Institute, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Xiaolei Cao
- Life Sciences Institute, Zhejiang University, Hangzhou, 310058, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Kaiyi Fu
- Life Sciences Institute, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Yan Lou
- Zhejiang Provincial Key Laboratory for Drug Clinical Research and Evaluation, Department of Clinical Pharmacy, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310000, Zhejiang, China
| | - Xin-Hua Feng
- Life Sciences Institute, Zhejiang University, Hangzhou, 310058, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, 310058, Zhejiang, China
- Center for Life Sciences, Shaoxing Institute, Zhejiang University, Shaoxing, 321000, China
| | - Junfang Ji
- Life Sciences Institute, Zhejiang University, Hangzhou, 310058, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, 310058, Zhejiang, China
- Center for Life Sciences, Shaoxing Institute, Zhejiang University, Shaoxing, 321000, China
| | - Bin Zhao
- Life Sciences Institute, Zhejiang University, Hangzhou, 310058, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, 310058, Zhejiang, China
- Center for Life Sciences, Shaoxing Institute, Zhejiang University, Shaoxing, 321000, China
| | - Fei Lan
- International Co-laboratory of Medical Epigenetics and Metabolism of Ministry of Science and Technology, and Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, and Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Li Shen
- Life Sciences Institute, Zhejiang University, Hangzhou, 310058, Zhejiang, China
- Department of Orthopedics Surgery, School of Medicine, The Second Affiliated Hospital, Zhejiang University, Hangzhou, 310009, Zhejiang, China
| | - Xianglei He
- Department of Pathology, Zhejiang Provincial People's Hospital, Hangzhou, 3100014, Zhejiang, China
| | - Yunqing Qiu
- State Key Laboratory for Diagnosis and Treatment of Infectious Disease, and National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310000, Zhejiang, China.
- Zhejiang Provincial Key Laboratory for Drug Clinical Research and Evaluation, Department of Clinical Pharmacy, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310000, Zhejiang, China.
| | - Jianping Jin
- Life Sciences Institute, Zhejiang University, Hangzhou, 310058, Zhejiang, China.
- Cancer Center, Zhejiang University, Hangzhou, 310058, Zhejiang, China.
- Zhejiang Provincial Key Laboratory for Drug Clinical Research and Evaluation, Department of Clinical Pharmacy, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310000, Zhejiang, China.
- Center for Life Sciences, Shaoxing Institute, Zhejiang University, Shaoxing, 321000, China.
| |
Collapse
|
18
|
Szilveszter RM, Muntean M, Florea A. Molecular Mechanisms in Tumorigenesis of Hepatocellular Carcinoma and in Target Treatments-An Overview. Biomolecules 2024; 14:656. [PMID: 38927059 PMCID: PMC11201617 DOI: 10.3390/biom14060656] [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: 04/30/2024] [Revised: 05/29/2024] [Accepted: 05/30/2024] [Indexed: 06/28/2024] Open
Abstract
Hepatocellular carcinoma is the most common primary malignancy of the liver, with hepatocellular differentiation. It is ranked sixth among the most common cancers worldwide and is the third leading cause of cancer-related deaths. The most important etiological factors discussed here are viral infection (HBV, HCV), exposure to aflatoxin B1, metabolic syndrome, and obesity (as an independent factor). Directly or indirectly, they induce chromosomal aberrations, mutations, and epigenetic changes in specific genes involved in intracellular signaling pathways, responsible for synthesis of growth factors, cell proliferation, differentiation, survival, the metastasis process (including the epithelial-mesenchymal transition and the expression of adhesion molecules), and angiogenesis. All these disrupted molecular mechanisms contribute to hepatocarcinogenesis. Furthermore, equally important is the interaction between tumor cells and the components of the tumor microenvironment: inflammatory cells and macrophages-predominantly with a pro-tumoral role-hepatic stellate cells, tumor-associated fibroblasts, cancer stem cells, extracellular vesicles, and the extracellular matrix. In this paper, we reviewed the molecular biology of hepatocellular carcinoma and the intricate mechanisms involved in hepatocarcinogenesis, and we highlighted how certain signaling pathways can be pharmacologically influenced at various levels with specific molecules. Additionally, we mentioned several examples of recent clinical trials and briefly described the current treatment protocol according to the NCCN guidelines.
Collapse
Affiliation(s)
- Raluca-Margit Szilveszter
- Department of Pathology, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400340 Cluj-Napoca, Romania
- Department of Cell and Molecular Biology, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania; (M.M.); (A.F.)
- Cluj County Emergency Clinical Hospital, 400340 Cluj-Napoca, Romania
| | - Mara Muntean
- Department of Cell and Molecular Biology, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania; (M.M.); (A.F.)
| | - Adrian Florea
- Department of Cell and Molecular Biology, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania; (M.M.); (A.F.)
| |
Collapse
|
19
|
Shourideh A, Maddah R, Amiri BS, Basharat Z, Shadpirouz M. A Bioinformatics-Based Approach to Discover Novel Biomarkers in Hepatocellular Carcinoma. IRANIAN JOURNAL OF PUBLIC HEALTH 2024; 53:1332-1342. [PMID: 39430152 PMCID: PMC11488550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 08/15/2023] [Indexed: 10/22/2024]
Abstract
Background Liver hepatocellular carcinoma (LIHC) is a common cancer with a poor prognosis and high recurrence rate. We aimed to identify potential biomarkers for LIHC by investigating the involvement of hub genes, microRNAs (miRNAs), transcription factors (TFs), and protein kinases (PKs) in its occurrence. Methods we conducted a bioinformatics analysis using microarray datasets, the TCGA-LIHC dataset, and text mining to identify differentially expressed genes (DEGs) associated with LIHC. They then performed functional enrichment analysis and gene-disease association analysis. The protein-protein interaction network of the genes was established, and hub genes were identified. The expression levels and survival analysis of these hub genes were evaluated, and their association with miRNAs, TFs, and PKs was assessed. Results The analysis identified 122 common genes involved in LIHC pathogenesis. Ten hub genes were filtered out, including CDK1, CCNB1, CCNB2, CCNA2, ASPM, NCAPG, BIRC5, RRM2, KIF20A, and CENPF. The expression level of all hub genes was confirmed, and high expression levels of all hub genes were correlated with poor overall survival of LIHC patients. Conclusion Identifying potential biomarkers for LIHC can aid in the design of targeted treatments and improve the survival of LIHC patients. The findings of this study provide a basis for further research in the field of LIHC and contribute to the understanding of its molecular pathogenesis.
Collapse
Affiliation(s)
- Amir Shourideh
- Faculty of Pharmacy, Eastern Mediterranean University, Famagusta, Cyprus
| | - Reza Maddah
- Department of Bioprocess Engineering, Institute of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Bahareh Shateri Amiri
- Department of Internal Medicine, School of Medicine Hazrat-e Rasool General Hospital Iran University of Medical Sciences, Tehran, Iran
| | | | - Marzieh Shadpirouz
- Department of Applied Mathematics, Faculty of Mathematical Sciences, Shahrood University of Technology, Semnan, Iran
| |
Collapse
|
20
|
Xue Y, Ruan Y, Wang Y, Xiao P, Xu J. Signaling pathways in liver cancer: pathogenesis and targeted therapy. MOLECULAR BIOMEDICINE 2024; 5:20. [PMID: 38816668 PMCID: PMC11139849 DOI: 10.1186/s43556-024-00184-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 04/23/2024] [Indexed: 06/01/2024] Open
Abstract
Liver cancer remains one of the most prevalent malignancies worldwide with high incidence and mortality rates. Due to its subtle onset, liver cancer is commonly diagnosed at a late stage when surgical interventions are no longer feasible. This situation highlights the critical role of systemic treatments, including targeted therapies, in bettering patient outcomes. Despite numerous studies on the mechanisms underlying liver cancer, tyrosine kinase inhibitors (TKIs) are the only widely used clinical inhibitors, represented by sorafenib, whose clinical application is greatly limited by the phenomenon of drug resistance. Here we show an in-depth discussion of the signaling pathways frequently implicated in liver cancer pathogenesis and the inhibitors targeting these pathways under investigation or already in use in the management of advanced liver cancer. We elucidate the oncogenic roles of these pathways in liver cancer especially hepatocellular carcinoma (HCC), as well as the current state of research on inhibitors respectively. Given that TKIs represent the sole class of targeted therapeutics for liver cancer employed in clinical practice, we have particularly focused on TKIs and the mechanisms of the commonly encountered phenomena of its resistance during HCC treatment. This necessitates the imperative development of innovative targeted strategies and the urgency of overcoming the existing limitations. This review endeavors to shed light on the utilization of targeted therapy in advanced liver cancer, with a vision to improve the unsatisfactory prognostic outlook for those patients.
Collapse
Affiliation(s)
- Yangtao Xue
- Key Laboratory of Laparoscopic Technology of Zhejiang Province, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
- National Engineering Research Center of Innovation and Application of Minimally Invasive Instruments, Hangzhou, 310016, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, Hangzhou, 310016, China
- Zhejiang University Cancer Center, Hangzhou, 310058, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, 311121, China
| | - Yeling Ruan
- Key Laboratory of Laparoscopic Technology of Zhejiang Province, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
- National Engineering Research Center of Innovation and Application of Minimally Invasive Instruments, Hangzhou, 310016, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, Hangzhou, 310016, China
- Zhejiang University Cancer Center, Hangzhou, 310058, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, 311121, China
| | - Yali Wang
- Key Laboratory of Laparoscopic Technology of Zhejiang Province, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
- National Engineering Research Center of Innovation and Application of Minimally Invasive Instruments, Hangzhou, 310016, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, Hangzhou, 310016, China
- Zhejiang University Cancer Center, Hangzhou, 310058, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, 311121, China
| | - Peng Xiao
- Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China.
| | - Junjie Xu
- Key Laboratory of Laparoscopic Technology of Zhejiang Province, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China.
- National Engineering Research Center of Innovation and Application of Minimally Invasive Instruments, Hangzhou, 310016, China.
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, Hangzhou, 310016, China.
- Zhejiang University Cancer Center, Hangzhou, 310058, China.
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, 311121, China.
| |
Collapse
|
21
|
Wang L, Wang L, Wang R, Xu T, Wang J, Cui Z, Cheng F, Wang W, Yang X. Endometrial stem cell-derived exosomes repair cisplatin-induced premature ovarian failure via Hippo signaling pathway. Heliyon 2024; 10:e31639. [PMID: 38831834 PMCID: PMC11145543 DOI: 10.1016/j.heliyon.2024.e31639] [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: 12/16/2023] [Revised: 05/15/2024] [Accepted: 05/20/2024] [Indexed: 06/05/2024] Open
Abstract
Stem cells have been documented as a new therapeutic method for ovarian injuries such as premature ovarian failure (POF). However, effects of exosomes (Exos) derived from human endometrial stem cells (EnSCs) on diminished ovarian failure remain to be carefully elucidated. Our study aims to investigate the mechanisms of EnSC-Exos in the recovery of the cisplatin-induced granulosa cell injury model in vitro or POF mouses model in vivo and whether the Hippo signaling pathway is involved in the regulation. In this study, we established successful construction of the cisplatin-induced granulosa cell injury model and evaluated Hippo signaling pathway activation in cisplatin-damaged granulosa cells (GCs). Furthermore, laser scanning confocal microscope and immunofluorescence demonstrated that EnSC-Exos can be transferred to cisplatin-damaged GCs to decrease apoptosis. In addition, the enhanced expression of YAP at the protein level as well as YAP/TEAD target genes, such as CTGF, ANKRD1, and the increase of YAP into the nucleus in immunofluorescence staining after the addition of EnSC-Exos to cisplatin-damaged GCs confirmed the suppression of Hippo signaling pathway. While in vivo, EnSC-Exos successfully remedied POF in a mouse model. Collectively, our findings suggest that chemotherapy-induced POF was associated with the activating of Hippo signaling pathway. Human EnSC-Exos significantly elevated the proliferation of ovarian GCs and the ovarian function by regulating Hippo signaling pathway. These findings provide new insights for further understanding of EnSC-Exos in the recovery of ovary function.
Collapse
Affiliation(s)
- Lijun Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, 710061, China
- Department of Obstetrics and Gynecology, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, 250014, China
- Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, 250014, China
| | - Lihui Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, 710061, China
| | - Rongli Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, 710061, China
| | - Ting Xu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, 710061, China
| | - Jingyuan Wang
- Department of Clinical Laboratory, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, 710061, China
| | - Zhiwei Cui
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, 710061, China
| | - Feiyan Cheng
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, 710061, China
| | - Wei Wang
- Department of Anesthesiology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, 710061, China
| | - Xinyuan Yang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, 710061, China
| |
Collapse
|
22
|
Yang J, Ren Z, Wang F, Zheng J, Zhuang Z, Zeng Y. γ-Synuclein promotes proliferation and inhibits apoptosis of oral squamous cell carcinoma via JAK2/STAT5b signaling pathway. Am J Cancer Res 2024; 14:2408-2423. [PMID: 38859859 PMCID: PMC11162669 DOI: 10.62347/flim3367] [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: 11/26/2023] [Accepted: 05/03/2024] [Indexed: 06/12/2024] Open
Abstract
γ-Synuclein (SNCG) has various biological functions associated with tumorigenesis. However, the role of SNCG in oral squamous cell carcinoma (OSCC) remains unknown. In this study, we found that SNCG expression is associated with the malignancy of OSCC. We showed that SNCG promotes cell proliferation and inhibits apoptosis in OSCC. Mechanistically, we demonstrated for the first time, that SNCG interacts with ERK1/2 and promotes its phosphorylation leading to activation of the JAK2/STAT5b signaling pathway. Subsequent experiments with STAT5b interference and ERK1/2 inhibitor treatment reversed the effects of SNCG on OSCC cell proliferation, apoptosis and cell cycle progression. Our findings suggest that SNCG functions as an oncogene in OSCC by targeting the JAK2/STAT5b axis and thus may be a potential new prognostic marker and therapeutic target in OSCC.
Collapse
Affiliation(s)
- Jie Yang
- Precision Clinical Laboratory, Zhanjiang Central Hospital, Guangdong Medical UniversityZhanjiang 524037, Guangdong, China
- Department of Laboratory, The First Affiliated Hospital of Shihezi UniversityShihezi 832008, Xinjiang, China
| | - Zuodong Ren
- Precision Clinical Laboratory, Zhanjiang Central Hospital, Guangdong Medical UniversityZhanjiang 524037, Guangdong, China
| | - Fazhan Wang
- Shaanxi Energy InstituteXianyang 712000, Shaanxi, China
| | - Jun Zheng
- Department of Stomatology, Zhanjiang Central Hospital, Guangdong Medical UniversityZhanjiang 524037, Guangdong, China
| | - Zhaowei Zhuang
- Precision Clinical Laboratory, Zhanjiang Central Hospital, Guangdong Medical UniversityZhanjiang 524037, Guangdong, China
| | - Yan Zeng
- Precision Clinical Laboratory, Zhanjiang Central Hospital, Guangdong Medical UniversityZhanjiang 524037, Guangdong, China
| |
Collapse
|
23
|
Younis MA, Harashima H. Understanding Gene Involvement in Hepatocellular Carcinoma: Implications for Gene Therapy and Personalized Medicine. Pharmgenomics Pers Med 2024; 17:193-213. [PMID: 38737776 PMCID: PMC11088404 DOI: 10.2147/pgpm.s431346] [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: 12/29/2023] [Accepted: 04/09/2024] [Indexed: 05/14/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is the dominant type of liver cancers and is one of the deadliest health threats globally. The conventional therapeutic options for HCC are hampered by low efficiency and intolerable side effects. Gene therapy, however, now offers hope for the treatment of many disorders previously considered incurable, and gene therapy is beginning to address many of the shortcomings of conventional therapies. Herein, we summarize the involvement of genes in the pathogenesis and prognosis of HCC, with a special focus on dysregulated signaling pathways, genes involved in immune evasion, and non-coding RNAs as novel two-edged players, which collectively offer potential targets for the gene therapy of HCC. Herein, the opportunities and challenges of HCC gene therapy are discussed. These include innovative therapies such as genome editing and cell therapies. Moreover, advanced gene delivery technologies that recruit nanomedicines for use in gene therapy for HCC are highlighted. Finally, suggestions are offered for improved clinical translation and future directions in this area of endeavor.
Collapse
Affiliation(s)
- Mahmoud A Younis
- Laboratory of Innovative Nanomedicine, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, 060-0812, Japan
- Department of Industrial Pharmacy, Faculty of Pharmacy, Assiut University, Assiut, 71526, Egypt
| | - Hideyoshi Harashima
- Laboratory of Innovative Nanomedicine, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, 060-0812, Japan
| |
Collapse
|
24
|
Üremis N, Türköz Y, Üremiş MM, Çiğremiş Y, Şalva E. RETRACTED ARTICLE: Investigating EGFR-VEGF-mediated apoptotic effect of cucurbitacin D and I combination with sorafenib via Ras/Raf/MEK/ERK and PI3K/Akt signaling pathways. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:3247. [PMID: 37917368 DOI: 10.1007/s00210-023-02811-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 10/20/2023] [Indexed: 11/04/2023]
Affiliation(s)
- Nuray Üremis
- Department of Medical Biochemistry, Medical Faculty, Inonu University, Malatya, Turkey.
| | - Yusuf Türköz
- Department of Medical Biochemistry, Medical Faculty, Inonu University, Malatya, Turkey
| | - Muhammed Mehdi Üremiş
- Department of Medical Biochemistry, Medical Faculty, Inonu University, Malatya, Turkey
| | - Yılmaz Çiğremiş
- Department of Medical Biology and Genetics, Medical Faculty, Inonu University, Malatya, Turkey
| | - Emine Şalva
- Department of Pharmacy Technology, Pharmacy Faculty, Inonu University, Malatya, Turkey
| |
Collapse
|
25
|
Ramirez CFA, Taranto D, Ando-Kuri M, de Groot MHP, Tsouri E, Huang Z, de Groot D, Kluin RJC, Kloosterman DJ, Verheij J, Xu J, Vegna S, Akkari L. Cancer cell genetics shaping of the tumor microenvironment reveals myeloid cell-centric exploitable vulnerabilities in hepatocellular carcinoma. Nat Commun 2024; 15:2581. [PMID: 38519484 PMCID: PMC10959959 DOI: 10.1038/s41467-024-46835-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 03/12/2024] [Indexed: 03/25/2024] Open
Abstract
Myeloid cells are abundant and plastic immune cell subsets in the liver, to which pro-tumorigenic, inflammatory and immunosuppressive roles have been assigned in the course of tumorigenesis. Yet several aspects underlying their dynamic alterations in hepatocellular carcinoma (HCC) progression remain elusive, including the impact of distinct genetic mutations in shaping a cancer-permissive tumor microenvironment (TME). Here, in newly generated, clinically-relevant somatic female HCC mouse models, we identify cancer genetics' specific and stage-dependent alterations of the liver TME associated with distinct histopathological and malignant HCC features. Mitogen-activated protein kinase (MAPK)-activated, NrasG12D-driven tumors exhibit a mixed phenotype of prominent inflammation and immunosuppression in a T cell-excluded TME. Mechanistically, we report a NrasG12D cancer cell-driven, MEK-ERK1/2-SP1-dependent GM-CSF secretion enabling the accumulation of immunosuppressive and proinflammatory monocyte-derived Ly6Clow cells. GM-CSF blockade curbs the accumulation of these cells, reduces inflammation, induces cancer cell death and prolongs animal survival. Furthermore, GM-CSF neutralization synergizes with a vascular endothelial growth factor (VEGF) inhibitor to restrain HCC outgrowth. These findings underscore the profound alterations of the myeloid TME consequential to MAPK pathway activation intensity and the potential of GM-CSF inhibition as a myeloid-centric therapy tailored to subsets of HCC patients.
Collapse
Affiliation(s)
- Christel F A Ramirez
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Daniel Taranto
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Masami Ando-Kuri
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Marnix H P de Groot
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Efi Tsouri
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Zhijie Huang
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China
| | - Daniel de Groot
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Roelof J C Kluin
- Genomics Core facility, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Daan J Kloosterman
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Joanne Verheij
- Department of Pathology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Jing Xu
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China
| | - Serena Vegna
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands.
- Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands.
| | - Leila Akkari
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands.
- Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands.
| |
Collapse
|
26
|
Jiang D, Zhu XL, An Y, Li YR. Clinical significance of small nuclear ribonucleoprotein U1 subunit 70 in patients with hepatocellular carcinoma. PeerJ 2024; 12:e16876. [PMID: 38500533 PMCID: PMC10946392 DOI: 10.7717/peerj.16876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 01/11/2024] [Indexed: 03/20/2024] Open
Abstract
Background & Aims Small nuclear ribonucleoprotein U1 subunit 70 (SNRNP70) as one of the components of the U1 small nuclear ribonucleoprotein (snRNP) is rarely reported in cancers. This study aims to estimate the application potential of SNRNP70 in hepatocellular carcinoma (HCC) clinical practice. Methods Based on the TCGA database and cohort of HCC patients, we investigated the expression patterns and prognostic value of SNRNP70 in HCC. Then, the combination of SNRNP70 and alpha-fetoprotein (AFP) in 278 HCC cases was analyzed. Next, western blotting and immunohistochemistry were used to detect the expression of SNRNP70 in nucleus and cytoplasm. Finally, Cell Counting Kit-8 (CCK-8) and scratch wound healing assays were used to detect the effect of SNRNP70 on the proliferation and migration of HCC cells. Results SNRNP70 was highly expressed in HCC. Its expression was increasingly high during the progression of HCC and was positively related to immune infiltration cells. Higher SNRNP70 expression indicated a poor outcome of HCC patients. In addition, nuclear SNRNP70/AFP combination could be a prognostic biomarker for overall survival and recurrence. Cell experiments confirmed that knockdown of SNRNP70 inhibited the proliferation and migration of HCC cells. Conclusion SNRNP70 may be a new biomarker for HCC progression and HCC diagnosis as well as prognosis. SNRNP70 combined with serum AFP may indicate the prognosis and recurrence status of HCC patients after operation.
Collapse
Affiliation(s)
- Dong Jiang
- Department of Ultrasound, Eastern Hepatobiliary Surgery Hospital, The Third Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Xia-Ling Zhu
- Department of Ultrasound, Eastern Hepatobiliary Surgery Hospital, The Third Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Yan An
- Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi-ran Li
- Department of Intensive Care Medicine, Eastern Hepatobiliary Surgery Hospital, The Third Affiliated Hospital of Naval Medical University, Shanghai, China
| |
Collapse
|
27
|
Gajos-Michniewicz A, Czyz M. WNT/β-catenin signaling in hepatocellular carcinoma: The aberrant activation, pathogenic roles, and therapeutic opportunities. Genes Dis 2024; 11:727-746. [PMID: 37692481 PMCID: PMC10491942 DOI: 10.1016/j.gendis.2023.02.050] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 12/28/2022] [Accepted: 02/14/2023] [Indexed: 09/12/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a liver cancer, highly heterogeneous both at the histopathological and molecular levels. It arises from hepatocytes as the result of the accumulation of numerous genomic alterations in various signaling pathways, including canonical WNT/β-catenin, AKT/mTOR, MAPK pathways as well as signaling associated with telomere maintenance, p53/cell cycle regulation, epigenetic modifiers, and oxidative stress. The role of WNT/β-catenin signaling in liver homeostasis and regeneration is well established, whereas in development and progression of HCC is extensively studied. Herein, we review recent advances in our understanding of how WNT/β-catenin signaling facilitates the HCC development, acquisition of stemness features, metastasis, and resistance to treatment. We outline genetic and epigenetic alterations that lead to activated WNT/β-catenin signaling in HCC. We discuss the pivotal roles of CTNNB1 mutations, aberrantly expressed non-coding RNAs and complexity of crosstalk between WNT/β-catenin signaling and other signaling pathways as challenging or advantageous aspects of therapy development and molecular stratification of HCC patients for treatment.
Collapse
Affiliation(s)
- Anna Gajos-Michniewicz
- Department of Molecular Biology of Cancer, Medical University of Lodz, Lodz 92-215, Poland
| | - Malgorzata Czyz
- Department of Molecular Biology of Cancer, Medical University of Lodz, Lodz 92-215, Poland
| |
Collapse
|
28
|
Lu X, Jin J, Wu Y, Liu X, Liang X, Lin J, Sun Q, Qin J, Zhang W, Luan X. Progress in RAS-targeted therapeutic strategies: From small molecule inhibitors to proteolysis targeting chimeras. Med Res Rev 2024; 44:812-832. [PMID: 38009264 DOI: 10.1002/med.21993] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 04/14/2023] [Accepted: 10/29/2023] [Indexed: 11/28/2023]
Abstract
As a widely considerable target in chemical biology and pharmacological research, rat sarcoma (RAS) gene mutations play a critical driving factor in several fatal cancers. Despite the great progress of RAS subtype-specific inhibitors, rapid acquired drug resistance could limit their further clinical applications. Proteolysis targeting chimera (PROTAC) has emerged as a powerful tool to handle "undruggable" targets and exhibited significant therapeutic benefit for the combat of drug resistance. Owing to unique molecular mechanism and binding kinetics, PROTAC is expected to become a feasible strategy to break the bottleneck of classical RAS inhibitors. This review aims to discuss the current advances of RAS inhibitors and especially focus on PROTAC strategy targeting RAS mutations and their downstream effectors for relevant cancer treatment.
Collapse
Affiliation(s)
- Xinchen Lu
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
- State Key Laboratory of New Drug and Pharmaceutical Process, Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, Shanghai, China
| | - Jinmei Jin
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ye Wu
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaoxia Liu
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaohui Liang
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jiayi Lin
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qingyan Sun
- State Key Laboratory of New Drug and Pharmaceutical Process, Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, Shanghai, China
| | - Jiangjiang Qin
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Weidong Zhang
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- State Key Laboratory of New Drug and Pharmaceutical Process, Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, Shanghai, China
| | - Xin Luan
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
29
|
Pessino G, Scotti C, Maggi M, Immuno-Hub Consortium. Hepatocellular Carcinoma: Old and Emerging Therapeutic Targets. Cancers (Basel) 2024; 16:901. [PMID: 38473265 DOI: 10.3390/cancers16050901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 02/16/2024] [Accepted: 02/20/2024] [Indexed: 03/14/2024] Open
Abstract
Liver cancer, predominantly hepatocellular carcinoma (HCC), globally ranks sixth in incidence and third in cancer-related deaths. HCC risk factors include non-viral hepatitis, alcohol abuse, environmental exposures, and genetic factors. No specific genetic alterations are unequivocally linked to HCC tumorigenesis. Current standard therapies include surgical options, systemic chemotherapy, and kinase inhibitors, like sorafenib and regorafenib. Immunotherapy, targeting immune checkpoints, represents a promising avenue. FDA-approved checkpoint inhibitors, such as atezolizumab and pembrolizumab, show efficacy, and combination therapies enhance clinical responses. Despite this, the treatment of hepatocellular carcinoma (HCC) remains a challenge, as the complex tumor ecosystem and the immunosuppressive microenvironment associated with it hamper the efficacy of the available therapeutic approaches. This review explores current and advanced approaches to treat HCC, considering both known and new potential targets, especially derived from proteomic analysis, which is today considered as the most promising approach. Exploring novel strategies, this review discusses antibody drug conjugates (ADCs), chimeric antigen receptor T-cell therapy (CAR-T), and engineered antibodies. It then reports a systematic analysis of the main ligand/receptor pairs and molecular pathways reported to be overexpressed in tumor cells, highlighting their potential and limitations. Finally, it discusses TGFβ, one of the most promising targets of the HCC microenvironment.
Collapse
Affiliation(s)
- Greta Pessino
- Unit of Immunology and General Pathology, Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
| | - Claudia Scotti
- Unit of Immunology and General Pathology, Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
| | - Maristella Maggi
- Unit of Immunology and General Pathology, Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
| | - Immuno-Hub Consortium
- Unit of Immunology and General Pathology, Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
| |
Collapse
|
30
|
Leng G, Gong H, Liu G, Kong Y, Guo L, Zhang Y. Alpha-fetoprotein upregulates hepatocellular carcinoma cell-intrinsic PD-1 expression through the LATS2/YAP/TEAD1 pathway. Biochim Biophys Acta Gen Subj 2024; 1868:130592. [PMID: 38395204 DOI: 10.1016/j.bbagen.2024.130592] [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: 10/24/2023] [Revised: 02/11/2024] [Accepted: 02/19/2024] [Indexed: 02/25/2024]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) cell-intrinsic programmed death 1 (PD-1) promotes tumor progression. However, the mechanisms that regulate its expression are unclear. This study investigated the impact of alpha-fetoprotein (AFP) on HCC cell-intrinsic PD-1 expression. METHODS The expression of PD-1 and AFP at the gene and protein levels was detected using real-time fluorescence quantitative polymerase chain reaction (RT-qPCR) and western blotting (WB). Proteins interacting with AFP were examined by co-immunoprecipitation (CO-IP). Chromatin immunoprecipitation (ChIP) and dual luciferase reporter assays were used to identify transcription-enhanced association domain 1 (TEAD1) binding to the promoter of PD-1. RESULTS The expression of HCC cell-intrinsic PD-1 was positively correlated with AFP. Mechanistically, AFP inhibited the phosphorylation of large tumor suppressor 2 (LATS2) and yes-associated protein (YAP). As a result, YAP is transferred to the nucleus and forms a transcriptional complex with TEAD1, promoting PD-1 transcription by binding to its promoter. CONCLUSION AFP is an upstream regulator of the HCC cell-intrinsic PD-1 and increases PD-1 expression via the LATS2/YAP/TEAD1 axis. GENERAL Our findings provide insight into the mechanisms of HCC development and offer new ideas for further in-depth studies of HCC.
Collapse
Affiliation(s)
- Guangxian Leng
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou 730030, Gansu Province, China
| | - Hongxia Gong
- Provincial-Level Key Laboratory of Molecular Medicine of Major Diseases and Study on Prevention and Treatment of Traditional Chinese Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou 730000, Gansu Province, China
| | - Guiyuan Liu
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou 730030, Gansu Province, China; People's Hospital affiliated with Chongqing Three Gorges Medical Higher Specialized School, Chongqing 404100, China
| | - Yin Kong
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou 730030, Gansu Province, China; Department of Hepatology, Lanzhou University Second Hospital, Lanzhou 730030, Gansu Province, China
| | - Liuqing Guo
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou 730030, Gansu Province, China
| | - Youcheng Zhang
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou 730030, Gansu Province, China.
| |
Collapse
|
31
|
Kinsey E, Lee HM. Management of Hepatocellular Carcinoma in 2024: The Multidisciplinary Paradigm in an Evolving Treatment Landscape. Cancers (Basel) 2024; 16:666. [PMID: 38339417 PMCID: PMC10854554 DOI: 10.3390/cancers16030666] [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: 12/22/2023] [Revised: 01/26/2024] [Accepted: 01/31/2024] [Indexed: 02/12/2024] Open
Abstract
Liver cancer is the third most common cause of cancer-related deaths worldwide, and hepatocellular carcinoma (HCC) makes up the majority of liver cancer cases. Despite the stabilization of incidence rates in recent years due to effective viral hepatitis treatments, as well as improved outcomes from early detection and treatment advances, the burden of HCC is anticipated to rise again due to increasing rates of metabolic dysfunction-associated steatotic liver disease and alcohol-related liver disease. The treatment landscape is evolving and requires a multidisciplinary approach, often involving multi-modal treatments that include surgical resection, transplantation, local regional therapies, and systemic treatments. The optimal approach to the care of the HCC patient requires a multidisciplinary team involving hepatology, medical oncology, diagnostic and interventional radiology, radiation oncology, and surgery. In order to determine which approach is best, an individualized treatment plan should consider the patient's liver function, functional status, comorbidities, cancer stage, and preferences. In this review, we provide an overview of the current treatment options and key trials that have revolutionized the management of HCC. We also discuss evolving treatment paradigms for the future.
Collapse
Affiliation(s)
- Emily Kinsey
- Division of Hematology, Oncology, and Palliative Care, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA
- Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, VA 23219, USA
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA
- Stravitz-Sanyal Institute for Liver Disease and Metabolic Health, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Hannah M. Lee
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA
- Stravitz-Sanyal Institute for Liver Disease and Metabolic Health, Virginia Commonwealth University, Richmond, VA 23298, USA
- Hume-Lee Transplant Center, Virginia Commonwealth University, Richmond, VA 23298, USA
| |
Collapse
|
32
|
Chen Y, Yang Y, Wang N, Liu R, Wu Q, Pei H, Li W. β-Sitosterol suppresses hepatocellular carcinoma growth and metastasis via FOXM1-regulated Wnt/β-catenin pathway. J Cell Mol Med 2024; 28:e18072. [PMID: 38063438 PMCID: PMC10844700 DOI: 10.1111/jcmm.18072] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 11/20/2023] [Accepted: 11/24/2023] [Indexed: 02/08/2024] Open
Abstract
β-Sitosterol is a natural compound with demonstrated anti-cancer properties against various cancers. However, its effects on hepatocellular carcinoma (HCC) and the underlying mechanisms are not well understood. This study aims to investigate the impact of β-sitosterol on HCC. In this study, we investigated the effects of β-sitosterol on HCC tumour growth and metastasis using a xenograft mouse model and a range of molecular analyses, including bioinformatics, real-time PCR, western blotting, lentivirus transfection, CCK8, scratch and transwell assays. The results found that β-sitosterol significantly inhibits HepG2 cell proliferation, migration and invasion both in vitro and in vivo. Bioinformatics analysis identifies forkhead box M1 (FOXM1) as a potential target for β-sitosterol in HCC treatment. FOXM1 is upregulated in HCC tissues and cell lines, correlating with poor prognosis in patients. β-Sitosterol downregulates FOXM1 expression in vitro and in vivo. FOXM1 overexpression mitigates β-sitosterol's inhibitory effects on HepG2 cells. Additionally, β-sitosterol suppresses epithelial-mesenchymal transition (EMT) in HepG2 cells, while FOXM1 overexpression promotes EMT. Mechanistically, β-sitosterol inhibits Wnt/β-catenin signalling by downregulating FOXM1, regulating target gene transcription related to HepG2 cell proliferation and metastasis. β-Sitosterol shows promising potential as a therapeutic candidate for inhibiting HCC growth and metastasis through FOXM1 downregulation and Wnt/β-catenin signalling inhibition.
Collapse
Affiliation(s)
- Yuankun Chen
- Department of Infectious and Tropical DiseasesThe Second Affiliated Hospital of Hainan Medical UniversityHaikouHainanChina
- Key Laboratory of Tropical Translational Medicine of Ministry of HealthHainan Medical UniversityHaikouHainanChina
- Department of Clinical LaboratoryThe Second Affiliated Hospital of Hainan Medical UniversityHaikouHainanChina
| | - Yijun Yang
- Department of Infectious and Tropical DiseasesThe Second Affiliated Hospital of Hainan Medical UniversityHaikouHainanChina
- Key Laboratory of Tropical Translational Medicine of Ministry of HealthHainan Medical UniversityHaikouHainanChina
| | - Nengyi Wang
- Department of Infectious and Tropical DiseasesThe Second Affiliated Hospital of Hainan Medical UniversityHaikouHainanChina
- Key Laboratory of Tropical Translational Medicine of Ministry of HealthHainan Medical UniversityHaikouHainanChina
| | - Rui Liu
- Department of Infectious and Tropical DiseasesThe Second Affiliated Hospital of Hainan Medical UniversityHaikouHainanChina
- Key Laboratory of Tropical Translational Medicine of Ministry of HealthHainan Medical UniversityHaikouHainanChina
| | - Qiuping Wu
- Department of Infectious and Tropical DiseasesThe Second Affiliated Hospital of Hainan Medical UniversityHaikouHainanChina
- Key Laboratory of Tropical Translational Medicine of Ministry of HealthHainan Medical UniversityHaikouHainanChina
| | - Hua Pei
- Department of Infectious and Tropical DiseasesThe Second Affiliated Hospital of Hainan Medical UniversityHaikouHainanChina
- Key Laboratory of Tropical Translational Medicine of Ministry of HealthHainan Medical UniversityHaikouHainanChina
- Department of Clinical LaboratoryThe Second Affiliated Hospital of Hainan Medical UniversityHaikouHainanChina
| | - Wenting Li
- Department of Infectious and Tropical DiseasesThe Second Affiliated Hospital of Hainan Medical UniversityHaikouHainanChina
- Key Laboratory of Tropical Translational Medicine of Ministry of HealthHainan Medical UniversityHaikouHainanChina
- Department of Infectious DiseasesThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhuiChina
| |
Collapse
|
33
|
Dubey R, Sharma A, Gupta S, Gupta GD, Asati V. A comprehensive review of small molecules targeting PI3K pathway: Exploring the structural development for the treatment of breast cancer. Bioorg Chem 2024; 143:107077. [PMID: 38176377 DOI: 10.1016/j.bioorg.2023.107077] [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: 10/04/2023] [Revised: 11/28/2023] [Accepted: 12/27/2023] [Indexed: 01/06/2024]
Abstract
Cancer stands as one of the deadliest diseases, ranking second in terms of its global impact. Despite the presence of numerous compelling theories concerning its origins, none have succeeded in fully elucidating the intricate nature of this ailment. Among the prevailing concerns in today's world, breast cancer proliferation remains a significant issue, particularly affecting females. The abnormal proliferation of the PI3K pathway emerges as a prominent driver of breast cancer, underscoring its role in cellular survival and proliferation. Consequently, targeting this pathway has emerged as a leading strategy in breast cancer therapeutics. Within this context, the present article explores the current landscape of anti-tumour drug development, focusing on structural activity relationships (SAR) in PI3K targeting breast cancer treatment. Notably, certain moieties like triazines, pyrimidine, quinazoline, quinoline, and pyridoxine have been explored as potential PI3K inhibitors for combating breast cancer. Various heterocyclic small molecules are undergoing clinical trials, such as Alpelisib, the first orally available FDA-approved drug targeting PI3K; others include buparlisib, pictilisib, and taselisib, which inhibit class I PI3K. These drugs are used for the treatment of breast cancer but still have various side effects with their high cost. Therefore, the primary goal of this review is to include all current advances in the development of anticancer medicines that target PI3K over-activation in the treatment of breast cancer.
Collapse
Affiliation(s)
- Rahul Dubey
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga, Punjab, India
| | - Anushka Sharma
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga, Punjab, India
| | - Shankar Gupta
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga, Punjab, India
| | - G D Gupta
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, Punjab, India
| | - Vivek Asati
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga, Punjab, India.
| |
Collapse
|
34
|
Wang Y, Xie L, Liu F, Ding D, Wei W, Han F. Research progress on traditional Chinese medicine-induced apoptosis signaling pathways in ovarian cancer cells. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117299. [PMID: 37816474 DOI: 10.1016/j.jep.2023.117299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 10/05/2023] [Accepted: 10/07/2023] [Indexed: 10/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE As a "silent killer" that threatens women's lives and health, ovarian cancer (OC) has the clinical characteristics of being difficult to detect, difficult to treat, and high recurrence. Traditional Chinese medicine (TCM) can be utilized as a long-term complementary and alternative therapy since it has shown benefits in alleviating clinical symptoms of OC, decreasing toxic side effects of radiation and chemotherapy, as well as enhancing patients' quality of life. AIM OF THE REVIEW This paper reviews how TCM contributes to the apoptosis of OC cells through signaling pathways, including active constituents, extracts, and herbal formulas, with the aim of providing a basis for the development and clinical application of therapeutic strategies for TCM in OC. METHODS The search was conducted from scientific databases PubMed, Embase, Web of Science, CNKI, Wanfang, VIP, and SinoMed databases aiming to elucidate the apoptosis signaling pathways in OC cells by TCM. The articles were searched by the keywords "ovarian cancer", "apoptosis", "signaling pathway", "traditional Chinese medicine", "Chinese herbal monomer", "Chinese herbal extract", and "herbal formula". The search was conducted from January 2013 to June 2023. A total of 97 potentially relevant articles were included, including 93 articles on Chinese medicine active constituents or extracts and 4 articles on Chinese herbal compound prescriptions. RESULTS TCM can induce apoptosis in OC cells by regulating signaling pathways with obvious advantages, including STAT3, PI3K/AKT, Wnt/β-catenin, MAPK, NF-κB, Nrf2, HIF-1α, Fas/Fas L signaling pathway, etc. CONCLUSION: Chinese medicine can induce apoptosis in OC cells through multiple pathways, targets, and routes. TCM has special advantages for treating OC, providing more reasonable evidence for the research and development of new apoptosis inducers.
Collapse
Affiliation(s)
- Yu Wang
- Department of Obstetrics and Gynecology, Heilongjiang University of Chinese Medicine, Harbin, 150040, China.
| | - Liangzhen Xie
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, 150040, China.
| | - Fangyuan Liu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, 150040, China.
| | - Danni Ding
- Department of Obstetrics and Gynecology, Heilongjiang University of Chinese Medicine, Harbin, 150040, China.
| | - Wei Wei
- Department of Obstetrics and Gynecology, Heilongjiang University of Chinese Medicine, Harbin, 150040, China.
| | - Fengjuan Han
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, 150040, China.
| |
Collapse
|
35
|
Yüregir Y, Kaçaroğlu D, Yaylacı S. Regulation of Hepatocellular Carcinoma Epithelial-Mesenchymal Transition Mechanism and Targeted Therapeutic Approaches. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1450:93-102. [PMID: 37452258 DOI: 10.1007/5584_2023_781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/18/2023]
Abstract
Hepatocellular carcinoma (HCC) is a primary liver malignancy that accounts for the majority of liver cancer cases, with multiple risk factors including chronic hepatitis B and C infections, alcohol abuse, and non-alcoholic fatty liver disease (NAFLD). Despite advancements in diagnosis and treatment, the survival rate of patients with advanced HCC remains low, creating an urgent need for new therapeutic targets and strategies.One biological process crucial to HCC progression is the epithelial-mesenchymal transition (EMT). EMT is a process that enables epithelial cells to acquire mesenchymal properties, including motility and invasiveness, by losing their cell-cell adhesion. Various signaling pathways, including TGF-β, Wnt/β-catenin, and Notch, have been implicated in regulating EMT in HCC.To inhibit EMT, targeted therapeutic approaches have been developed, and preclinical studies suggest that the inhibition of the TGF-β, Wnt/β-catenin, and Notch signaling pathways is promising. TGF-β receptor inhibitors, Wnt/β-catenin pathway inhibitors, and gamma-secretase inhibitors have shown efficacy in preclinical studies by inhibiting EMT and reducing tumor growth in HCC models. However, further clinical studies are necessary to determine their effectiveness in human patients.In addition to these approaches, further research is needed to identify other novel therapeutic targets and develop new treatment strategies for HCC. This review emphasizes the critical role of EMT in HCC progression and highlights the potential of targeting the TGF-β, Wnt/β-catenin, and Notch signaling pathways to inhibit EMT and reduce tumor growth in HCC. Future studies and clinical trials are necessary to validate these therapeutic strategies and develop effective treatments for HCC.
Collapse
Affiliation(s)
- Yelda Yüregir
- Molecular Biology and Genetics Department, İhsan Doğramacı Bilkent University, Ankara, Turkey
| | - Demet Kaçaroğlu
- Faculty of Medicine, Medical Biology Department, Lokman Hekim University, Ankara, Turkey
| | - Seher Yaylacı
- Faculty of Medicine, Medical Biology Department, Lokman Hekim University, Ankara, Turkey.
| |
Collapse
|
36
|
Noor A, Shafi S, Sehar N, Qadir I, Bilquees, Rashid S, Arafah A, Rasool S, Dar NJ, Masoodi MH, Rehman MU. Curcuminoids as Cell Signaling Pathway Modulators: A Potential Strategy for Cancer Prevention. Curr Med Chem 2024; 31:3093-3117. [PMID: 37559247 DOI: 10.2174/0929867331666230809100335] [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: 09/13/2022] [Revised: 02/23/2023] [Accepted: 03/03/2023] [Indexed: 08/11/2023]
Abstract
Despite substantial advancements in curative modern medicine in the last few decades, cancer risk and casualty rates have continued to mount globally. The exact reason for cancer's onset and progression is still unknown. However, skeletal and functional abnormalities in the genetic code are assumed to be the primary cause of cancer. Many lines of evidence reported that some medicinal plants can be utilized to curb cancer cell proliferation with a safe, fruitful, and cost-efficient perspective. Curcuminoid, isolated from Curcuma longa, have gotten a lot of focus due to their anticancer potential as they reduce tumor progression, invasion, and dissemination. Further, they modulated signal transduction routes like MAPK, PI3K/Akt/mTOR, JAK/STAT, and Wnt/β-catenin, etc., and triggered apoptosis as well as actuated autophagy in malignant cells without altering the normal cells, thus preventing cancer progression. Besides, Curcuminoid also regulate the function and expression of anti-tumor and carcinogenic miRNAs. Clinical studies also reported the therapeutic effect of Curcuminoid against various cancer through decreasing specific biomarkers like TNF-α, Bcl-2, COX-2, PGE2, VEGF, IκKβ, and various cytokines like IL-12p70, IL-10, IL-2, IFN-γ levels and increasing in p53 and Bax levels. Thus, in the present review, we abridged the modulation of several signal transduction routes by Curcuminoids in various malignancies, and its modulatory role in the initiation of tumor-suppressive miRNAs and suppression of the oncogenic miRNAs are explored. Additionally, various pharmacokinetic approaches have been projected to address the Curcuminoids bioavailability like the use of piperine as an adjuvant; nanotechnology- based Curcuminoids preparations utilizing Curcuminoids analogues are also discussed.
Collapse
Affiliation(s)
- Aneeza Noor
- Natural Products Research Laboratory, Department of Pharmaceutical Sciences, School of Applied Sciences & Technology, University of Kashmir, Hazratbal Srinagar, J&K, India
| | - Saimeena Shafi
- Natural Products Research Laboratory, Department of Pharmaceutical Sciences, School of Applied Sciences & Technology, University of Kashmir, Hazratbal Srinagar, J&K, India
| | - Nouroz Sehar
- Centre for Translational and Clinical Research, School of Chemical & Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Insha Qadir
- Natural Products Research Laboratory, Department of Pharmaceutical Sciences, School of Applied Sciences & Technology, University of Kashmir, Hazratbal Srinagar, J&K, India
| | - Bilquees
- Natural Products Research Laboratory, Department of Pharmaceutical Sciences, School of Applied Sciences & Technology, University of Kashmir, Hazratbal Srinagar, J&K, India
| | - Summya Rashid
- Department of Pharmacology & Toxicology, College of Pharmacy, Prince Sattam Bin Abdul Aziz University, Al Kharj, 11942, Saudi Arabia
| | - Azher Arafah
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Saiema Rasool
- Department of School Education, Govt. of Jammu & Kashmir, Srinagar, J&K 190001, India
| | - Nawab John Dar
- Cellular Neurobiology Laboratory (CNB-P), Salk Institute, 10010 N. Torrey Pines Rd., La Jolla, CA92037, USA
| | - Mubashir Hussain Masoodi
- Natural Products Research Laboratory, Department of Pharmaceutical Sciences, School of Applied Sciences & Technology, University of Kashmir, Hazratbal Srinagar, J&K, India
| | - Muneeb U Rehman
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| |
Collapse
|
37
|
Yang Z, Shen X, Wang Z, Li R, Hou W, Liu Z, Gao Y, Zhu C, Qin X. The Biological Function of POLA2 in Hepatocellular Carcinoma. Comb Chem High Throughput Screen 2024; 27:1758-1775. [PMID: 37861044 DOI: 10.2174/0113862073254083231002052550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 08/28/2023] [Accepted: 08/31/2023] [Indexed: 10/21/2023]
Abstract
INTRODUCTION The role and prognostic value of POLA2 in liver cancer were comprehensively analyzed through TCGA, GEO, and ICGC databases, and the role of POLA2 in liver cancer cells and the regulatory mechanism involved were further verified through cell experiments. Hepatocellular carcinoma (HCC) is the most prevalent malignancy with high morbidity and mortality. Consequently, it is critical to identify robust and reliable predictive biomarkers and therapeutic targets for HCC patients. POLA2 is involved in the regulation of various tumors, but the specific role of POLA2 in HCC has not been reported. The regulatory role and prognostic value of POLA2 in HCC were determined by bioinformatics techniques and cell experiments. METHODS The specific role and prognostic value of POLA2 in HCC were comprehensively analyzed by combining the expression data of POLA2 in TCGA, GEO, and ICGC databases and clinical data. In clinical samples, the expression of POLA2 in liver cancer was verified by QPCR. Further, the regulatory role of POLA2 in HCC was explored through cell experiments such as CCK-8, clonal formation experiment, EDU cell proliferation experiment, and flow cytometry. In terms of mechanism exploration, western blot was used to verify the specific regulatory mechanism that POLA2 participated in. Finally, the relationship between POLA2 and immune invasion of HCC was analyzed by using the TIMER database. RESULTS A POLA2 expression and prognosis analysis of HCC patients was conducted using the TCGA, GEO, and ICGC databases. We hypothesized that POLA2 might be one of the key factors contributing to the HCC progression. According to a combined analysis of TCGA, ICGC, and GEO databases, POLA2 was highly expressed in HCC. This was further confirmed in clinical samples using the qPCR. POLA2 knockdown was also performed in vitro on HCC cell lines to study the changes in their biological behavior. We confirmed that POLA2 was associated with HCC proliferation by CCK-8, Colony Formation, and EDU assay. We verified the POLA2's involvement in cell cycle regulation using flow techniques. The relationship between POLA2 and PI3K/AKT/mTOR pathway was explored using Western Blotting experiments regarding its mechanism. Further analysis revealed that the POLA2 expression was significantly associated with HCC immune infiltration. CONCLUSION Our study demonstrated POLA2's importance in HCC development and progression and its potential role as a biomarker for disease progression on multiple levels. POLA2 has an important role in regulating the cell cycle and cell proliferation. By interfering with the cell cycle and proliferation, HCC cell growth is inhibited. Furthermore, POLA2 expression was significantly associated with immune infiltration. POLA2 may play a role in HCC immunotherapy based on its correlation with several immune cell types' genetic markers. The findings of this study are expected to lead to new anticancer strategies for HCC.
Collapse
Affiliation(s)
- Zhen Yang
- Dalian Medical University, Dalian, Liaoning, 116000, China
- Department of General Surgery, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, 213000, China
| | - Xingyuan Shen
- Dalian Medical University, Dalian, Liaoning, 116000, China
- Department of General Surgery, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, 213000, China
| | - Zhihuai Wang
- Dalian Medical University, Dalian, Liaoning, 116000, China
| | - Renzhi Li
- Dalian Medical University, Dalian, Liaoning, 116000, China
| | - Wenqiang Hou
- Dalian Medical University, Dalian, Liaoning, 116000, China
| | - Zengyuan Liu
- Dalian Medical University, Dalian, Liaoning, 116000, China
| | - Yuan Gao
- Department of General Surgery, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, 213000, China
| | - Chunfu Zhu
- Dalian Medical University, Dalian, Liaoning, 116000, China
- Department of General Surgery, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, 213000, China
| | - Xihu Qin
- Dalian Medical University, Dalian, Liaoning, 116000, China
- Department of General Surgery, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, 213000, China
| |
Collapse
|
38
|
Prasad M, Tamil Selvan S, Shanmugam R. Influence of Pterostilbene on Gene Expression in Liver Cancer: An In Silico Analysis. Cureus 2024; 16:e53098. [PMID: 38414698 PMCID: PMC10898499 DOI: 10.7759/cureus.53098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 01/27/2024] [Indexed: 02/29/2024] Open
Abstract
Background Liver cancer, in particular, is a serious threat to global health and has few viable treatments. One natural molecule that shows potential in cancer therapy is pterostilbene, especially for hepatocellular carcinoma (HCC). The molecular details of pterostilbene's interactions with liver cancer are uncovered in this study using an in silico method. Methodology This study determines the differentially expressed genes (DEGs) in HCC and the way pterostilbene affects them using data from Gene Expression Omnibus (GEO) datasets. To identify the intricate linkages and possible treatment targets, network pharmacology, protein-protein interaction (PPI) analysis, and pathway enrichment investigations were performed. Results The study revealed complex relationships between pterostilbene and liver cancer, identified important DEGs in HCC, and showed enriched pathways. Pterostilbene shows promise as a target for therapeutic approaches in HCC due to its modulation of important signaling pathways. Conclusions This work offers an extensive knowledge of pterostilbene's potential in liver cancer, despite intrinsic computational limitations. In addition to the importance of experimental validation, the pathways and DEGs that have been found provide insightful information for future investigation, highlighting the ongoing research that is necessary to create targeted therapeutics for HCC.
Collapse
Affiliation(s)
- Monisha Prasad
- Centre for Global Health Research, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Chennai, IND
| | - Silambarasan Tamil Selvan
- Centre for Global Health Research, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Chennai, IND
| | - Rajeshkumar Shanmugam
- Nanobiomedicine Lab, Centre for Global Health Research, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Chennai, IND
| |
Collapse
|
39
|
Liu Z, Wang X, Zhang C, Lin K, Yang J, Zhang Y, Hao J, Tian F. Folic acid-coupled bovine serum albumin-modified magnetic nanocomposites from quantum-sized Fe 3O 4 and layered double hydroxide for actively targeted delivery of 5-fluorouracil. Int J Biol Macromol 2024; 256:128385. [PMID: 38000576 DOI: 10.1016/j.ijbiomac.2023.128385] [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: 08/16/2023] [Revised: 11/15/2023] [Accepted: 11/21/2023] [Indexed: 11/26/2023]
Abstract
The development of multifunctional magnetic nanocomposites as a drug delivery system for cancer therapy is highly desirable in current nanomedicine. Herein, folic acid-bovine serum albumin conjugate (FA-BSA) was modified on nanocomposites by combining quantum-sized Fe3O4 and layered double hydroxide (LDH) to obtain a novel FA-BSA/Fe3O4@LDH for the delivery of the anticancer drug 5-Fluorouracil (5-Fu). The prepared nanocomposites showed good dispersibility, colloidal stability, magnetic property and erythrocyte compatibility. FA-BSA/Fe3O4@LDH/5-Fu showed pH responsiveness, with both the amount and duration of release of FA-BSA/Fe3O4@LDH/5-Fu being significantly higher in pH 5.0 release medium than in pH 7.4 release medium. The cellular experiments implied that no significant cytotoxicity of FA-BSA/Fe3O4@LDH, particularly due to the presence of FA-BSA, which further enhanced the biocompatibility of the nanocomposite. Furthermore, FA-BSA/Fe3O4@LDH/5-Fu could specifically target the 2D HepG2 cells model and 3D hepatoma cell microspheres model in vitro, and efficient internalization through folate receptor-mediated endocytosis, showing excellent anti-cancer cell activity in a concentration-dependent manner. Therefore, the constructed FA-BSA/Fe3O4@LDH was able to provide a potential novel multifunctional nanocomposite for magnetic-targeting drug delivery and pH-responsive release of drugs to enhance the efficiency of cancer therapy.
Collapse
Affiliation(s)
- Zhaoyun Liu
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, PR China
| | - Xi Wang
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, PR China
| | - Chen Zhang
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, PR China
| | - Kui Lin
- Analytical Instrumentation Centre, Tianjin University, Tianjin 300072, PR China
| | - Jian Yang
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, PR China
| | - Yi Zhang
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Jia Hao
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China.
| | - Fei Tian
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, PR China.
| |
Collapse
|
40
|
Liu WF, Zhang QW, Quan B, Zhang F, Li M, Lu SX, Dong L, Yin X, Liu BB. Gas7 attenuates hepatocellular carcinoma progression and chemoresistance through the PI3K/Akt signaling pathway. Cell Signal 2023; 112:110908. [PMID: 37769891 DOI: 10.1016/j.cellsig.2023.110908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 09/01/2023] [Accepted: 09/25/2023] [Indexed: 10/03/2023]
Abstract
Growth arrest-specific gene 7 (Gas7) was involved in various cellular functions, although its specific roles and molecular mechanisms in hepatocellular carcinoma (HCC) remained unclear. So the current study was to investigate the role of Gas7 in HCC. Our findings revealed that Gas7 was downregulated in various HCC cell lines and low Gas7 expression was associated with decreased overall survival in patients with HCC. Additionally, our functional assays showed that Gas7 inhibited cell proliferation and migration, induced cell cycle arrest, apoptosis, and autophagy, and enhanced oxaliplatin sensitivity by inhibiting the PI3K/Akt signaling pathway. We also observed that transcription factorSp1 was responsible for inhibiting Gas7. These findings provide insights into the role and elucidated a potential mechanism of Gas7 in HCC progression and metastasis. It was also observed that the Sp1/Gas7/PI3K/Akt axis was critical for malignant phenotype and oxaliplatin sensitivity in HCC. Therefore, Gas7 can be considered as a prognostic predictor and therapeutic target for HCC.
Collapse
Affiliation(s)
- Wen-Feng Liu
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai 200032, China; Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Qi-Wei Zhang
- Department of Interventional Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Bing Quan
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Feng Zhang
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai 200032, China; Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Miao Li
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Shen-Xin Lu
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Ling Dong
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| | - Xin Yin
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai 200032, China.
| | - Bin-Bin Liu
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai 200032, China.
| |
Collapse
|
41
|
Deng H, Shu X, Wang Y, Zhang J, Yin Y, Wu F, He J. Matrix Stiffness Regulated Endoplasmic Reticulum Stress-mediated Apoptosis of Osteosarcoma Cell through Ras Signal Cascades. Cell Biochem Biophys 2023; 81:839-850. [PMID: 37789235 DOI: 10.1007/s12013-023-01184-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/20/2023] [Indexed: 10/05/2023]
Abstract
The modulating effects of matrix stiffness on spreading and apoptosis of tumor cells have been well recognized. Nevertheless, the detail road map leading to the apoptosis and the underlying mechanisms governing the cell apoptosis have remained to be elucidated. To this aim, we provided a tunable elastic hydrogel matrix that promoted cell adhesion by modifying the surface of polyacrylamide with polydopamine, with stiffness value of 1, 10, 30, and 250 kPa, respectively. While the cell spreading increased and the apoptosis decreased with the matrix stiffness, such modulating effect of matrix on cell spreading exhibited different time evolvement behaviors as a function of stiffness, which likely led to surprisingly similar apoptosis rates for the 30 kPa and 250 kPa samples. Matrix stiffness mediated the spreading and apoptosis of MG-63 cells by regulating cell adhesion to matrix and in particular cytoskeletal organization, which was dependent on Ras, Rap1 and PI3K-Akt signaling pathways and finally led to the apoptosis of cancer cells dominated by endoplasmic reticulum stress pathway. Our results provided an insight into the regulation of tumor cell fate by the mechanical clues of ECM, which would have implication for future cancer research and the design of novel anticancer materials.
Collapse
Affiliation(s)
- Huan Deng
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, PR China
| | - Xuedong Shu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, PR China
| | - Yao Wang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, PR China
| | - Junwei Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, PR China
| | - Yue Yin
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, PR China
| | - Fang Wu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, PR China
| | - Jing He
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, PR China.
| |
Collapse
|
42
|
Jadhav PA, Thomas AB, Nanda RK, Chitlange SS. Unveiling the role of gut dysbiosis in non-alcoholic fatty liver disease. Eur J Gastroenterol Hepatol 2023; 35:1324-1333. [PMID: 37823422 DOI: 10.1097/meg.0000000000002654] [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] [Indexed: 10/13/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a multifactorial complicated condition, reflected by the accumulation of extra fat in the liver. A detailed study of literature throws light on the fascinating connection between gut dysbiosis and NAFLD. The term 'gut dysbiosis' describes an imbalance in the harmony and operation of the gut microflora, which can upshoot a number of metabolic disorders. To recognize the underlying mechanisms and determine treatment options, it is essential to comprehend the connection between gut dysbiosis and NAFLD. This in-depth review discusses the normal gut microflora composition and its role in health, alterations in the gut microflora composition that leads to disease state focusing on NAFLD. The potential mechanisms influencing the advent and aggravation of NAFLD suggested disturbance of microbial metabolites, changes in gut barrier integrity, and imbalances in the composition of the gut microflora. Furthermore, it was discovered that gut dysbiosis affected immune responses, liver inflammation, and metabolic pathways, aggravating NAFLD.
Collapse
Affiliation(s)
- Pranali A Jadhav
- Department of Pharmaceutical Chemistry, Dr. D. Y. Patil Institute of Pharmaceutical Sciences and Research, Pimpri, Pune, Maharashtra, India
| | | | | | | |
Collapse
|
43
|
Li Z, Zhang Z, Fang L, Zhao J, Niu Z, Chen H, Cao G. Tumor Microenvironment Composition and Related Therapy in Hepatocellular Carcinoma. J Hepatocell Carcinoma 2023; 10:2083-2099. [PMID: 38022729 PMCID: PMC10676104 DOI: 10.2147/jhc.s436962] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 11/10/2023] [Indexed: 12/01/2023] Open
Abstract
Globally, primary liver cancer is the third leading cause of cancer death, and hepatocellular carcinoma (HCC) accounts for 75%-95%. The tumor microenvironment (TME), composed of the extracellular matrix, helper cells, immune cells, cytokines, chemokines, and growth factors, promotes the immune escape, invasion, and metastasis of HCC. Tumor metastasis and postoperative recurrence are the main threats to the long-term prognosis of HCC. TME-related therapies are increasingly recognized as effective treatments. Molecular-targeted therapy, immunotherapy, and their combined therapy are the main approaches. Immunotherapy, represented by immune checkpoint inhibitors (ICIs), and targeted therapy, highlighted by tyrosine kinase inhibitors (TKIs), have greatly improved the prognosis of HCC. This review focuses on the TME compositions and emerging therapeutic approaches to TME in HCC.
Collapse
Affiliation(s)
- Zishuai Li
- Key Laboratory of Biological Defense, Ministry of Education, Second Military Medical University, Shanghai, 200433, People’s Republic of China
- Shanghai Key Laboratory of Medical Bioprotection, Second Military Medical University, Shanghai, 200433, People’s Republic of China
- Department of Epidemiology, Second Military Medical University, Shanghai, 200433, People’s Republic of China
| | - Zihan Zhang
- Department of Epidemiology, Tongji University School of Medicine Tongji University, Shanghai, 200120, People’s Republic of China
| | - Letian Fang
- Key Laboratory of Biological Defense, Ministry of Education, Second Military Medical University, Shanghai, 200433, People’s Republic of China
- Shanghai Key Laboratory of Medical Bioprotection, Second Military Medical University, Shanghai, 200433, People’s Republic of China
- Department of Epidemiology, Second Military Medical University, Shanghai, 200433, People’s Republic of China
| | - Jiayi Zhao
- Key Laboratory of Biological Defense, Ministry of Education, Second Military Medical University, Shanghai, 200433, People’s Republic of China
- Shanghai Key Laboratory of Medical Bioprotection, Second Military Medical University, Shanghai, 200433, People’s Republic of China
- Department of Epidemiology, Second Military Medical University, Shanghai, 200433, People’s Republic of China
| | - Zheyun Niu
- Department of Epidemiology, Tongji University School of Medicine Tongji University, Shanghai, 200120, People’s Republic of China
| | - Hongsen Chen
- Key Laboratory of Biological Defense, Ministry of Education, Second Military Medical University, Shanghai, 200433, People’s Republic of China
- Shanghai Key Laboratory of Medical Bioprotection, Second Military Medical University, Shanghai, 200433, People’s Republic of China
- Department of Epidemiology, Second Military Medical University, Shanghai, 200433, People’s Republic of China
| | - Guangwen Cao
- Key Laboratory of Biological Defense, Ministry of Education, Second Military Medical University, Shanghai, 200433, People’s Republic of China
- Shanghai Key Laboratory of Medical Bioprotection, Second Military Medical University, Shanghai, 200433, People’s Republic of China
- Department of Epidemiology, Second Military Medical University, Shanghai, 200433, People’s Republic of China
| |
Collapse
|
44
|
Matera I, Miglionico R, Abruzzese V, Marchese G, Ventola GM, Castiglione Morelli MA, Bisaccia F, Ostuni A. A Regulator Role for the ATP-Binding Cassette Subfamily C Member 6 Transporter in HepG2 Cells: Effect on the Dynamics of Cell-Cell and Cell-Matrix Interactions. Int J Mol Sci 2023; 24:16391. [PMID: 38003580 PMCID: PMC10670978 DOI: 10.3390/ijms242216391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 11/12/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
There is growing evidence that various ATP-binding cassette (ABC) transporters contribute to the growth and development of tumors, but relatively little is known about how the ABC transporter family behaves in hepatocellular carcinoma (HCC), one of the most common cancers worldwide. Cellular model studies have shown that ABCC6, which belongs to the ABC subfamily C (ABCC), plays a role in the cytoskeleton rearrangement and migration of HepG2 hepatocarcinoma cells, thus highlighting its role in cancer biology. Deep knowledge on the molecular mechanisms underlying the observed results could provide therapeutic insights into the tumors in which ABCC6 is modulated. In this study, differential expression levels of mRNA transcripts between ABCC6-silenced HepG2 and control groups were measured, and subsequently, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were performed. Real-Time PCR and Western blot analyses confirmed bioinformatics; functional studies support the molecular mechanisms underlying the observed effects. The results provide valuable information on the dysregulation of fundamental cellular processes, such as the focal adhesion pathway, which allowed us to obtain detailed information on the active role that the down-regulation of ABCC6 could play in the biology of liver tumors, as it is involved not only in cell migration but also in cell adhesion and invasion.
Collapse
Affiliation(s)
- Ilenia Matera
- Department of Sciences, University of Basilicata, 85100 Potenza, Italy; (I.M.); (R.M.); (V.A.); (M.A.C.M.)
| | - Rocchina Miglionico
- Department of Sciences, University of Basilicata, 85100 Potenza, Italy; (I.M.); (R.M.); (V.A.); (M.A.C.M.)
| | - Vittorio Abruzzese
- Department of Sciences, University of Basilicata, 85100 Potenza, Italy; (I.M.); (R.M.); (V.A.); (M.A.C.M.)
| | - Giovanna Marchese
- Genomix4Life Srl, 84081 Baronissi, Italy; (G.M.); (G.M.V.)
- Genome Research Center for Health—CRGS, 84081 Baronissi, Italy
| | | | | | - Faustino Bisaccia
- Department of Sciences, University of Basilicata, 85100 Potenza, Italy; (I.M.); (R.M.); (V.A.); (M.A.C.M.)
| | - Angela Ostuni
- Department of Sciences, University of Basilicata, 85100 Potenza, Italy; (I.M.); (R.M.); (V.A.); (M.A.C.M.)
| |
Collapse
|
45
|
Weidle UH, Nopora A. Hepatocellular Carcinoma: Up-regulated Circular RNAs Which Mediate Efficacy in Preclinical In Vivo Models. Cancer Genomics Proteomics 2023; 20:500-521. [PMID: 37889063 PMCID: PMC10614070 DOI: 10.21873/cgp.20401] [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: 07/03/2023] [Revised: 08/22/2023] [Accepted: 08/30/2023] [Indexed: 10/28/2023] Open
Abstract
Hepatocellular carcinoma (HCC) ranges as number two with respect to the incidence of tumors and is associated with a dismal prognosis. The therapeutic efficacy of approved multi-tyrosine kinase inhibitors and checkpoint inhibitors is modest. Therefore, the identification of new therapeutic targets and entities is of paramount importance. We searched the literature for up-regulated circular RNAs (circRNAs) which mediate efficacy in preclinical in vivo models of HCC. Our search resulted in 14 circRNAs which up-regulate plasma membrane transmembrane receptors, while 5 circRNAs induced secreted proteins. Two circRNAs facilitated replication of Hepatitis B or C viruses. Three circRNAs up-regulated high mobility group proteins. Six circRNAs regulated components of the ubiquitin system. Seven circRNAs induced GTPases of the family of ras-associated binding proteins (RABs). Three circRNAs induced redox-related proteins, eight of them up-regulated metabolic enzymes and nine circRNAs induced signaling-related proteins. The identified circRNAs up-regulate the corresponding targets by sponging microRNAs. Identified circRNAs and their targets have to be validated by standard criteria of preclinical drug development. Identified targets can potentially be inhibited by small molecules or antibody-based moieties and circRNAs can be inhibited by small-interfering RNAs (siRNAs) or short hairpin RNAs (shRNAs) for therapeutic purposes.
Collapse
Affiliation(s)
- Ulrich H Weidle
- Roche Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany
| | - Adam Nopora
- Roche Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany
| |
Collapse
|
46
|
Hu M, Xia X, Chen L, Jin Y, Hu Z, Xia S, Yao X. Emerging biomolecules for practical theranostics of liver hepatocellular carcinoma. Ann Hepatol 2023; 28:101137. [PMID: 37451515 DOI: 10.1016/j.aohep.2023.101137] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 06/17/2023] [Accepted: 06/28/2023] [Indexed: 07/18/2023]
Abstract
Most cases of hepatocellular carcinoma (HCC) are able to be diagnosed through regular surveillance in an identifiable patient population with chronic hepatitis B or cirrhosis. Nevertheless, 50% of global cases might present incidentally owing to symptomatic advanced-stage HCC after worsening of liver dysfunction. A systematic search based on PUBMED was performed to identify relevant outcomes, covering newer surveillance modalities including secretory proteins, DNA methylation, miRNAs, and genome sequencing analysis which proposed molecular expression signatures as ideal tools in the early-stage HCC detection. In the face of low accuracy without harmonization on the analytical approaches and data interpretation for liquid biopsy, a more accurate incidence of HCC will be unveiled by using deep machine learning system and multiplex immunohistochemistry analysis. A combination of molecular-secretory biomarkers, high-definition imaging and bedside clinical indexes in a surveillance setting offers a comprehensive range of HCC potential indicators. In addition, the sequential use of numerous lines of systemic anti-HCC therapies will simultaneously benefit more patients in survival. This review provides an overview on the most recent developments in HCC theranostic platform.
Collapse
Affiliation(s)
- Miner Hu
- International Institutes of Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, China
| | - Xiaojun Xia
- International Institutes of Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, China
| | - Lichao Chen
- International Institutes of Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, China
| | - Yunpeng Jin
- International Institutes of Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, China
| | - Zhenhua Hu
- International Institutes of Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, China; Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Fourth Affiliated Hospital, School of Medicine, Zhejiang University, Yiwu, Zhejiang, China.
| | - Shudong Xia
- International Institutes of Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, China.
| | - Xudong Yao
- International Institutes of Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, China.
| |
Collapse
|
47
|
Sharma D, Xu W. ReGeNNe: genetic pathway-based deep neural network using canonical correlation regularizer for disease prediction. Bioinformatics 2023; 39:btad679. [PMID: 37963055 PMCID: PMC10666205 DOI: 10.1093/bioinformatics/btad679] [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: 07/14/2023] [Revised: 10/06/2023] [Accepted: 11/13/2023] [Indexed: 11/16/2023] Open
Abstract
MOTIVATION Common human diseases result from the interplay of genes and their biologically associated pathways. Genetic pathway analyses provide more biological insight as compared to conventional gene-based analysis. In this article, we propose a framework combining genetic data into pathway structure and using an ensemble of convolutional neural networks (CNNs) along with a Canonical Correlation Regularizer layer for comprehensive prediction of disease risk. The novelty of our approach lies in our two-step framework: (i) utilizing the CNN's effectiveness to extract the complex gene associations within individual genetic pathways and (ii) fusing features from ensemble of CNNs through Canonical Correlation Regularization layer to incorporate the interactions between pathways which share common genes. During prediction, we also address the important issues of interpretability of neural network models, and identifying the pathways and genes playing an important role in prediction. RESULTS Implementation of our methodology into three real cancer genetic datasets for different prediction tasks validates our model's generalizability and robustness. Comparing with conventional models, our methodology provides consistently better performance with AUC improvement of 11% on predicting early/late-stage kidney cancer, 10% on predicting kidney versus liver cancer type and 7% on predicting survival status in ovarian cancer as compared to the next best conventional machine learning model. The robust performance of our deep learning algorithm indicates that disease prediction using neural networks in multiple functionally related genes across different pathways improves genetic data-based prediction and understanding molecular mechanisms of diseases. AVAILABILITY AND IMPLEMENTATION https://github.com/divya031090/ReGeNNe.
Collapse
Affiliation(s)
- Divya Sharma
- Biostatistics Department, Princess Margaret Cancer Center, University Health Network, Toronto, ON M5G2C4, Canada
- Division of Biostatistics, Dalla Lana School of Public Health, University of Toronto, Toronto, ON M5T 3M7, Canada
| | - Wei Xu
- Biostatistics Department, Princess Margaret Cancer Center, University Health Network, Toronto, ON M5G2C4, Canada
- Division of Biostatistics, Dalla Lana School of Public Health, University of Toronto, Toronto, ON M5T 3M7, Canada
| |
Collapse
|
48
|
Wu X, Xu Z, Li W, Lu Y, Pu J. HIF‑1α and RACGAP1 promote the progression of hepatocellular carcinoma in a mutually regulatory way. Mol Med Rep 2023; 28:218. [PMID: 37772389 PMCID: PMC10568255 DOI: 10.3892/mmr.2023.13105] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 09/12/2023] [Indexed: 09/30/2023] Open
Abstract
Hypoxia, a condition characterized by low oxygen levels, serves an important role in the progression of hepatocellular carcinoma (HCC). However, the precise molecular mechanisms underlying hypoxia‑induced HCC progression are yet to be fully elucidated. The present study assessed the involvement of two key factors, hypoxia‑inducible factor‑1α (HIF‑1α) and Rac GTPase activating protein 1 (RACGAP1), in HCC development under hypoxic conditions. HIF‑1α and RACGAP1 genes were overexpressed and knocked down in Hep3B and Huh7 cells using lentiviral transduction and the levels of HIF‑1α and RACGAP1 in the cells were assessed using quantitative PCR, western blotting and immunofluorescence. Co‑immunoprecipitation experiments were performed to evaluate the interaction between HIF‑1α and RACGAP1. Subsequently, the proliferation, apoptosis, migration and invasion of Hep3B and Huh7 cells were assessed using the Cell Counting Kit‑8 assay, flow cytometry, Transwell assay and migration experiments. The expression levels of HIF‑1α and RACGAP1 in normal and HCC tumor samples were analyzed utilizing the Gene Expression Profiling Interactive Analysis database. Furthermore, correlations between HIF‑1α/RACGAP1 gene expression levels and patient survival outcomes were evaluated using the Kaplan‑Meier plotter. Knockdown of HIF‑1α resulted in a significant decrease in RACGAP1 expression, whilst overexpression of HIF‑1α resulted in a significant increase in RACGAP1 expression. Moreover, overexpression and knockdown of RACGAP1 had the same effect on HIF‑1α expression. Additionally, it was demonstrated that HIF‑1α and RACGAP1 interacted directly within a complex. Overexpression of HIF‑1α or RACGAP1 significantly increased proliferation, invasion and migration, and significantly decreased the proportion of apoptotic Hep3B and Huh7 cells. Conversely, knockdown of HIF‑1α or RACGAP1 significantly decreased proliferation, invasion and migration, and significantly increased the proportion of apoptotic Hep3B and Huh7 cells. In addition, the combined knockdown or overexpression of HIF‑1α and RACGAP1 had a more pronounced effect on HCC cell migration compared with knockdown of HIF‑1α alone. Furthermore, there was a significant positive correlation between the expression levels of HIF‑1α and RACGAP1 in HCC tissues and patients with HCC and upregulation of both HIF‑1α and RACGAP1 demonstrated a lower overall survival probability. In conclusion, HIF‑1α and RACGAP1 may synergistically contribute to the development of HCC, highlighting their potential as valuable targets for HCC therapy.
Collapse
Affiliation(s)
- Xianjian Wu
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi 533000, P.R. China
| | - Zuoming Xu
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi 533000, P.R. China
| | - Wenchuan Li
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi 533000, P.R. China
| | - Yuan Lu
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi 533000, P.R. China
| | - Jian Pu
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi 533000, P.R. China
| |
Collapse
|
49
|
Escutia-Gutiérrez R, Sandoval-Rodríguez A, Zamudio-Ojeda A, Guevara-Martínez SJ, Armendáriz-Borunda J. Advances of Nanotechnology in the Diagnosis and Treatment of Hepatocellular Carcinoma. J Clin Med 2023; 12:6867. [PMID: 37959332 PMCID: PMC10647688 DOI: 10.3390/jcm12216867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 10/20/2023] [Accepted: 10/22/2023] [Indexed: 11/15/2023] Open
Abstract
Nanotechnology has emerged as a promising technology in the field of hepatocellular carcinoma (HCC), specifically in the implementation of diagnosis and treatment strategies. Nanotechnology-based approaches, such as nanoparticle-based contrast agents and nanoscale imaging techniques, have shown great potential for enhancing the sensitivity and specificity of HCC detection. These approaches provide high-resolution imaging and allow for the detection of molecular markers and alterations in cellular morphology associated with HCC. In terms of treatment, nanotechnology has revolutionized HCC therapy by enabling targeted drug delivery, enhancing therapeutic efficacy, and minimizing off-target effects. Nanoparticle-based drug carriers can be functionalized with ligands specific to HCC cells, allowing for selective accumulation of therapeutic agents at the tumor site. Furthermore, nanotechnology can facilitate combination therapy by co-encapsulating multiple drugs within a single nanoparticle, allowing for synergistic effects and overcoming drug resistance. This review aims to provide an overview of recent advances in nanotechnology-based approaches for the diagnosis and treatment of HCC. Further research is needed to optimize the design and functionality of nanoparticles, improve their biocompatibility and stability, and evaluate their long-term safety and efficacy. Nonetheless, the integration of nanotechnology in HCC management holds great promise and may lead to improved patient outcomes in the future.
Collapse
Affiliation(s)
- Rebeca Escutia-Gutiérrez
- Department of Molecular Biology and Genomics, Institute for Molecular Biology in Medicine and Gene Therapy, Health Sciences University Center, University of Guadalajara, Guadalajara 44340, Mexico; (R.E.-G.); (A.S.-R.)
| | - Ana Sandoval-Rodríguez
- Department of Molecular Biology and Genomics, Institute for Molecular Biology in Medicine and Gene Therapy, Health Sciences University Center, University of Guadalajara, Guadalajara 44340, Mexico; (R.E.-G.); (A.S.-R.)
| | - Adalberto Zamudio-Ojeda
- Department of Physics, Exact Sciences and Engineering University Center, University of Guadalajara, Guadalajara 44340, Mexico;
| | - Santiago José Guevara-Martínez
- Department of Physics, Exact Sciences and Engineering University Center, University of Guadalajara, Guadalajara 44340, Mexico;
| | - Juan Armendáriz-Borunda
- Department of Molecular Biology and Genomics, Institute for Molecular Biology in Medicine and Gene Therapy, Health Sciences University Center, University of Guadalajara, Guadalajara 44340, Mexico; (R.E.-G.); (A.S.-R.)
- Tecnologico de Monterrey, School of Medicine and Health Sciences, Zapopan 45201, Mexico
| |
Collapse
|
50
|
Abd El-Lateef HM, Abdel Ghany LMA, Saleem RM, Ahmed Maghrabi AH, Yahya Alahdal MA, Khalifa Ali EH, Beshay BY, Zaki I, Masoud RE. Design, synthesis and antiproliferative screening of newly synthesized coumarin-acrylamide hybrids as potential cytotoxic and apoptosis inducing agents. RSC Adv 2023; 13:32547-32557. [PMID: 37936638 PMCID: PMC10626558 DOI: 10.1039/d3ra06644d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 10/23/2023] [Indexed: 11/09/2023] Open
Abstract
On the basis of the observed biological activity of coumarin and acrylamide derivatives, a new set of coumarin-acrylamide-CA-4 hybrids was designed and synthesized. These compounds were investigated for their cytotoxic activity against cancerous human liver cell line HepG2 cells using 5-fluorouracil (5-FU) as a reference drug. Compound 6e had promising antiproliferative activity with an IC50 value of 1.88 μM against HepG2 cells compared to 5-FU (IC50 = 7.18 μM). The results of β-tubulin polymerization inhibition indicated that coumarin-acrylamide derivative 6e was the most active, with a percentage inhibition value of 84.34% compared to podophyllotoxin (88.19% β-tubulin inhibition). Moreover, the active coumarin-acrylamide molecule 6e exerted cell cycle cession at the G2/M phase stage of HepG2 cells. In addition, this compound produced a 15.24-fold increase in apoptotic cell induction compared to no-treatment control. These observations were supported by histopathological studies of liver sections. The conducted docking studies illustrated that 6e is perfectly positioned within the tubulin colchicine binding site, indicating a significant interaction that may underlie its potent tubulin inhibitory activity. The main objective of the study was to develop new potent anticancer compounds that might be further optimized to prevent the progression of cancer disease.
Collapse
Affiliation(s)
- Hany M Abd El-Lateef
- Department of Chemistry, College of Science, King Faisal University Al-Ahsa 31982 Saudi Arabia
- Department of Chemistry, Faculty of Science, Sohag University Sohag 82524 Egypt
| | - Lina M A Abdel Ghany
- Pharmaceutical Chemistry Department, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology 6th of October City Egypt
| | - Rasha Mohammed Saleem
- Department of Laboratory Medicine, Faculty of Applied Medical Sciences, Al-Baha University Al-Baha 65431 Saudi Arabia
| | | | | | - Eman Hussain Khalifa Ali
- Department of Laboratory Medicine, Faculty of Applied Medical Sciences, Albaha University Saudi Arabia
| | - Botros Y Beshay
- Pharmaceutical Sciences (Pharmaceutical Chemistry) Department, College of Pharmacy, Arab Academy for Science, Technology and Maritime Transport Alexandria Egypt
| | - Islam Zaki
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Port Said University Port Said Egypt
| | - Reham E Masoud
- Clinical Pharmacology Department, Faculty of Medicine, Port Said University Port Said 42526 Egypt
| |
Collapse
|