51
|
Liu X, Ren Y, Qin S, Yang Z. Exploring the mechanism of 6-Methoxydihydrosanguinarine in the treatment of lung adenocarcinoma based on network pharmacology, molecular docking and experimental investigation. BMC Complement Med Ther 2024; 24:202. [PMID: 38783288 PMCID: PMC11119275 DOI: 10.1186/s12906-024-04497-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 05/10/2024] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND 6-Methoxydihydrosanguinarine (6-MDS) has shown promising potential in fighting against a variety of malignancies. Yet, its anti‑lung adenocarcinoma (LUAD) effect and the underlying mechanism remain largely unexplored. This study sought to explore the targets and the probable mechanism of 6-MDS in LUAD through network pharmacology and experimental validation. METHODS The proliferative activity of human LUAD cell line A549 was evaluated by Cell Counting Kit-8 (CCK8) assay. LUAD related targets, potential targets of 6-MDS were obtained from databases. Venn plot analysis were performed on 6-MDS target genes and LUAD related genes to obtain potential target genes for 6-MDS treatment of LUAD. The Search Tool for the Retrieval of Interacting Genes/Proteins (STRING) database was utilized to perform a protein-protein interaction (PPI) analysis, which was then visualized by Cytoscape. The hub genes in the network were singled out by CytoHubba. Metascape was employed for GO and KEGG enrichment analyses. molecular docking was carried out using AutoDock Vina 4.2 software. Gene expression levels, overall survival of hub genes were validated by the GEPIA database. Protein expression levels, promotor methylation levels of hub genes were confirmed by the UALCAN database. Timer database was used for evaluating the association between the expression of hub genes and the abundance of infiltrating immune cells. Furthermore, correlation analysis of hub genes expression with immune subtypes of LUAD were performed by using the TISIDB database. Finally, the results of network pharmacology analysis were validated by qPCR. RESULTS Experiments in vitro revealed that 6-MDS significantly reduced tumor growth. A total of 33 potential targets of 6-MDS in LUAD were obtained by crossing the LUAD related targets with 6-MDS targets. Utilizing CytoHubba, a network analysis tool, the top 10 genes with the highest centrality measures were pinpointed, including MMP9, CDK1, TYMS, CCNA2, ERBB2, CHEK1, KIF11, AURKB, PLK1 and TTK. Analysis of KEGG enrichment hinted that these 10 hub genes were located in the cell cycle signaling pathway, suggesting that 6-MDS may mainly inhibit the occurrence of LUAD by affecting the cell cycle. Molecular docking analysis revealed that the binding energies between 6-MDS and the hub proteins were all higher than - 6 kcal/Mol with the exception of AURKB, indicating that the 9 targets had strong binding ability with 6-MDS.These results were corroborated through assessments of mRNA expression levels, protein expression levels, overall survival analysis, promotor methylation level, immune subtypes andimmune infiltration. Furthermore, qPCR results indicated that 6-MDS can significantly decreased the mRNA levels of CDK1, CHEK1, KIF11, PLK1 and TTK. CONCLUSIONS According to our findings, it appears that 6-MDS could possibly serve as a promising option for the treatment of LUAD. Further investigations in live animal models are necessary to confirm its potential in fighting cancer and to delve into the mechanisms at play.
Collapse
Affiliation(s)
- Xingyun Liu
- The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, 421000, China
| | - Yanling Ren
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510000, China
- NMPA Key Laboratory for Technology Research and Evaluation of Pharmacovigilance, Guangdong Pharmaceutical University, Guangzhou, 510086, China
| | - Shuanglin Qin
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, 437000, China.
| | - Zerui Yang
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510000, China.
- NMPA Key Laboratory for Technology Research and Evaluation of Pharmacovigilance, Guangdong Pharmaceutical University, Guangzhou, 510086, China.
| |
Collapse
|
52
|
Li SB, Zou J, Zhao TSY, Liang B, Wang LS, Huang LZ, Liang CQ, Zhou XL. Antitumor effects of Cypaliuruside F from Cyclocarya paliurus on HepG2 cells. Nat Prod Res 2024:1-7. [PMID: 38771014 DOI: 10.1080/14786419.2024.2355590] [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: 01/13/2024] [Accepted: 05/09/2024] [Indexed: 05/22/2024]
Abstract
An undescribed dammarane triterpenoid saponin Cypaliuruside F was isolated from the leaves of Cyclocarya paliurus in our preliminary study. The MTT assay, flow cytometry, cell scratch, and DAPI staining were used to detect the antitumor effects of Cypaliuruside F on HepG2 cells. Subsequently, network pharmacology and molecular docking analysis were used to analyse the key targets of Cypaliuruside F against HCC. In addition, a Western blot was performed to determine the effects of Cypaliuruside F on the expression of key proteins in HepG2 cells. The experimental results indicated that the damarane triterpenoid saponin Cypaliuruside F from Cyclocarya paliurus inhibits the proliferation of HepG2 cells by inducing apoptosis and cell cycle arrest. These changes may promote the apoptosis of HepG2 cells by inhibiting the expression of mTOR, STAT3, and Bcl-2 while activating Bax.
Collapse
Affiliation(s)
- Shan-Bin Li
- Education Department of Guangxi Zhuang Autonomous Region, Key Laboratory of Biochemistry and Molecular Biology (Guilin Medical University), Guilin, P. R. China
| | - Jian Zou
- Education Department of Guangxi Zhuang Autonomous Region, Key Laboratory of Biochemistry and Molecular Biology (Guilin Medical University), Guilin, P. R. China
| | - Tong-Shi-Yao Zhao
- Education Department of Guangxi Zhuang Autonomous Region, Key Laboratory of Biochemistry and Molecular Biology (Guilin Medical University), Guilin, P. R. China
| | - Bin Liang
- Education Department of Guangxi Zhuang Autonomous Region, Key Laboratory of Biochemistry and Molecular Biology (Guilin Medical University), Guilin, P. R. China
| | - Li-Sheng Wang
- College of Pharmacy, Guilin Medical University, Guilin, P. R. China
| | - Lan-Zhen Huang
- Science Experiment Center, Guilin Medical University, Guilin, P. R. China
| | - Cheng-Qin Liang
- College of Pharmacy, Guilin Medical University, Guilin, P. R. China
| | - Xian-Li Zhou
- Education Department of Guangxi Zhuang Autonomous Region, Key Laboratory of Biochemistry and Molecular Biology (Guilin Medical University), Guilin, P. R. China
| |
Collapse
|
53
|
Xie L, Liang S, Jiwa H, Zhang L, Lu Q, Wang X, Luo L, Xia H, Li Z, Wang J, Luo X, Luo J. Securinine inhibits the tumor growth of human bladder cancer cells by suppressing Wnt/β-catenin signaling pathway and activating p38 and JNK signaling pathways. Biochem Pharmacol 2024; 223:116125. [PMID: 38484850 DOI: 10.1016/j.bcp.2024.116125] [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: 11/05/2023] [Revised: 02/28/2024] [Accepted: 03/11/2024] [Indexed: 03/18/2024]
Abstract
Bladder cancer (BC) is the most common malignant tumor in urinary system. Although chemotherapy is one of the most important adjuvant treatments for BC, drug resistance, non-specific toxicity and severe side effects are the major obstacles to BC chemotherapy. Natural products have always been a leading resource of antitumor drug discovery, with the advantages of excellent effectiveness, low toxicity, multi-targeting potency and easy availability. In this study, we evaluated the potential anti-tumor effect of securinine (SEC), a natural alkaloid from Securinega suffruticosa, on BC cells in vitro and in vivo, and delineated the underlying mechanism. We found that SEC inhibited the proliferation, migration and invasion, induced the apoptosis of BC cells in vitro, and retarded the xenograft tumor growth of BC cell in vivo. Notably, SEC had a promising safety profile because it presented no or low toxicity on normal cells and mice. Mechanistically, SEC inactivated Wnt/β-catenin signaling pathway while activated p38 and JNK signaling pathway. Moreover, β-catenin overexpression, the p38 inhibitor SB203580 and the JNK inhibitor SP600125 both mitigated the inhibitory effect of SEC on BC cells. Furthermore, we demonstrated a synergistic inhibitory effect of SEC and gemcitabine (GEM) on BC cells in vitro and in vivo. Taken together, our findings suggest that SEC may exert anti-BC cell effect at least through the activation of p38 and JNK signaling pathways, and the inhibition of Wnt/β-catenin signaling pathway. More meaningfully, the findings indicate that GEM-induced BC cell killing can be enhanced by combining with SEC.
Collapse
Affiliation(s)
- Liping Xie
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Shiqiong Liang
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Habu Jiwa
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, China
| | - Lulu Zhang
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Qiuping Lu
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Xiaoxuan Wang
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Lijuan Luo
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Haichao Xia
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Ziyun Li
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Jiayu Wang
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Xiaoji Luo
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, China
| | - Jinyong Luo
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
54
|
Yang K, Ma Y, Chen W, Liu L, Yang Z, He C, Zheng N, Liu X, Cheng X, Song J, Chen Y, Qiao H, Zhang R. CCDC58 is a potential biomarker for diagnosis, prognosis, immunity, and genomic heterogeneity in pan-cancer. Sci Rep 2024; 14:8575. [PMID: 38609450 PMCID: PMC11014850 DOI: 10.1038/s41598-024-59154-9] [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: 07/09/2023] [Accepted: 04/08/2024] [Indexed: 04/14/2024] Open
Abstract
Coiled-coil domain-containing 58 (CCDC58) is a member of the CCDC protein family. Similar to other members, CCDC58 exhibits potential tumorigenic roles in a variety of malignancies. However, there is no systematic and comprehensive pan-cancer analysis to investigate the diagnosis, prognosis, immune infiltration, and other related functions of CCDC58. We used several online websites and databases, such as TCGA, GTEx, UALCAN, HPA, CancerSEA, BioGRID, GEPIA 2.0, TIMER 2.0, and TISIDB, to extract CCDC58 expression data and clinical data of patients in pan-cancer. Then, the relationship between CCDC58 expression and diagnosis, prognosis, genetic alterations, DNA methylation, genomic heterogeneity, and immune infiltration level were determined. In addition, the biological function of CCDC58 in liver hepatocellular carcinoma (LIHC) was investigated. Pan-cancer analysis results showed that CCDC58 was differentially expressed in most tumors and showed excellent performance in diagnosis and prediction of prognosis. The expression of CCDC58 was highly correlated with genetic alterations, DNA methylation, and genomic heterogeneity in some tumors. In addition, the correlation analysis of CCDC58 with the level of immune infiltration and immune checkpoint marker genes indicated that CCDC58 might affect the composition of the tumor immune microenvironment. Enrichment analysis showed that CCDC58-related genes were mainly linked to mitosis, chromosome, and cell cycle. Finally, biological function experiments demonstrated that CCDC58 plays an important role in tumor cell proliferation and migration. CCDC58 was first identified as a pan-cancer biomarker. It may be used as a potential therapeutic target to improve the prognosis of patients in the future.
Collapse
Affiliation(s)
- Kai Yang
- Department of Hepatobiliary Surgery, Xi Jing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Yan Ma
- Department of Gynecology and Obstetrics, Xi Jing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Weigang Chen
- Department of Hepatobiliary Surgery, Xi Jing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Lu Liu
- College of Life Sciences, Northwest University, Xi'an, 710000, China
| | - Zelong Yang
- Department of Hepatobiliary Surgery, Xi Jing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Chaokui He
- Department of Oncology, The First Affiliated Hospital of Shanxi Medical University, Taiyuan, 030000, China
| | - Nanbei Zheng
- Department of General Surgery, The First Affiliated Hospital of Jiamusi University, Jiamusi, 154002, China
| | - Xinyu Liu
- Department of Hepatobiliary Surgery, Xi Jing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Xin Cheng
- Department of Hepatobiliary Surgery, Xi Jing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Junbo Song
- Department of Hepatobiliary Surgery, Xi Jing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Yong Chen
- Department of Hepatobiliary Surgery, Xi Jing Hospital, Air Force Medical University, Xi'an, 710032, China.
| | - Hongyu Qiao
- Department of Pediatrics, Xi Jing Hospital, Air Force Medical University, Xi'an, 710032, China.
| | - Ruohan Zhang
- Department of Hepatobiliary Surgery, Xi Jing Hospital, Air Force Medical University, Xi'an, 710032, China.
| |
Collapse
|
55
|
Xu W, Tang Y, Yang Y, Wang C, Liu C, Zhang J, Zhao L, Wang G. Depletion of CPNE7 sensitizes colorectal cancer to 5-fluorouracil by downregulating ATG9B expression. J Cell Mol Med 2024; 28:e18261. [PMID: 38526029 PMCID: PMC10962129 DOI: 10.1111/jcmm.18261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 02/10/2024] [Accepted: 02/20/2024] [Indexed: 03/26/2024] Open
Abstract
We aimed to explore the biological function of CPNE7 and determine the impact of CPNE7 on chemotherapy resistance in colorectal cancer (CRC) patients. According to the Gene Expression Profiling Interactive Analysis database and previously published data, CPNE7 was identified as a potential oncogene in CRC. RT-qPCR and Western blotting were performed to verify the expression of CPNE7. Chi-square test was used to evaluate the associations between CPNE7 and clinical features. Cell proliferation, colony formation, cell migration and invasion, cell cycle and apoptosis were assessed to determine the effects of CPNE7. Transcriptome sequencing was used to identify potential downstream regulatory genes, and gene set enrichment analysis was performed to investigate downstream pathways. The effect of CPNE7 on 5-fluorouracil chemosensitivity was verified by half maximal inhibitory concentration (IC50). Subcutaneous tumorigenesis assay was used to examine the role of CPNE7 in sensitivity of CRC to chemotherapy in vivo. Transmission electron microscopy was used to detect autophagosomes. CPNE7 was highly expressed in CRC tissues, and its expression was correlated with T stage and tumour site. Knockdown of CPNE7 inhibited the proliferation and colony formation of CRC cells and promoted apoptosis. Knockdown of CPNE7 suppressed the expression of ATG9B and enhanced the sensitivity of CRC cells to 5-fluorouracil in vitro and in vivo. Knockdown of CPNE7 reversed the induction of the autophagy pathway by rapamycin and reduced the number of autophagosomes. Depletion of CPNE7 attenuated the malignant proliferation of CRC cells and enhanced the chemosensitivity of CRC cells to 5-fluorouracil.
Collapse
Affiliation(s)
- Weile Xu
- The Department of General surgeryThe Second Hospital of Hebei Medical UniversityShijiazhuangHebeiChina
- The Department of General surgeryHebei Chest HospitalShijiazhuangHebeiChina
- The Second Department of SurgeryThe Fourth Hospital of Hebei Medical UniversityShijiazhuangHebeiChina
| | - Yujie Tang
- The Department of Gastrointestinal surgeryThe Third Hospital of Hebei Medical UniversityShijiazhuangHebeiChina
| | - Yang Yang
- The Department of Gastrointestinal surgeryThe Third Hospital of Hebei Medical UniversityShijiazhuangHebeiChina
| | - Changjing Wang
- The Department of Gastrointestinal surgeryThe Third Hospital of Hebei Medical UniversityShijiazhuangHebeiChina
| | - Chen Liu
- The Department of Gastrointestinal surgeryThe Third Hospital of Hebei Medical UniversityShijiazhuangHebeiChina
| | - Jianqing Zhang
- The Department of Gastrointestinal surgeryThe Third Hospital of Hebei Medical UniversityShijiazhuangHebeiChina
| | - Lianmei Zhao
- Scientific Research CenterThe Fourth Hospital of Hebei Medical UniversityShijiazhuangHebeiChina
| | - Guiying Wang
- The Department of General surgeryThe Second Hospital of Hebei Medical UniversityShijiazhuangHebeiChina
- The Second Department of SurgeryThe Fourth Hospital of Hebei Medical UniversityShijiazhuangHebeiChina
| |
Collapse
|
56
|
Franco Machado J, Cordeiro S, Duarte JN, Costa PJ, Mendes PJ, Garcia MH, Baptista PV, Fernandes AR, Morais TS. Exploiting Co(III)-Cyclopentadienyl Complexes To Develop Anticancer Agents. Inorg Chem 2024; 63:5783-5804. [PMID: 38502532 PMCID: PMC10988555 DOI: 10.1021/acs.inorgchem.3c03696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 02/29/2024] [Accepted: 03/06/2024] [Indexed: 03/21/2024]
Abstract
In recent years, organometallic complexes have attracted much attention as anticancer therapeutics aiming at overcoming the limitations of platinum drugs that are currently marketed. Still, the development of half-sandwich organometallic cobalt complexes remains scarcely explored. Four new cobalt(III)-cyclopentadienyl complexes containing N,N-heteroaromatic bidentate, and phosphane ligands were synthesized and fully characterized by elemental analysis, spectroscopic techniques, and DFT methods. The cytotoxicity of all complexes was determined in vitro by the MTS assay in colorectal (HCT116), ovarian (A2780), and breast (MDA-MB-231 and MCF-7) human cancer cell lines and in a healthy human cell line (fibroblasts). The complexes showed high cytotoxicity in cancer cell lines, mostly due to ROS production, apoptosis, autophagy induction, and disruption of the mitochondrial membrane. Also, these complexes were shown to be nontoxic in vivo in an ex ovo chick embryo yolk sac membrane (YSM) assay.
Collapse
Affiliation(s)
- João Franco Machado
- Centro
de Química Estrutural, Institute of Molecular Sciences, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
| | - Sandra Cordeiro
- Associate
Laboratory i4HB − Institute for Health and Bioeconomy, NOVA
School of Science and Technology, NOVA University
Lisbon, 2819-516 Caparica, Portugal
- UCIBIO,
Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, 2819-516 Caparica, Portugal
| | - Joana N. Duarte
- Centro
de Química Estrutural, Institute of Molecular Sciences, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
| | - Paulo J. Costa
- BioISI
− Instituto de Biosistemas e Ciências Integrativas, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisboa, Portugal
| | - Paulo J. Mendes
- LAQV-REQUIMTE
(Polo de Évora), Escola de Ciências e Tecnologia, Universidade de Évora, R. Romão Ramalho 59, 7000-671 Évora, Portugal
| | - Maria Helena Garcia
- Centro
de Química Estrutural, Institute of Molecular Sciences, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
| | - Pedro V. Baptista
- Associate
Laboratory i4HB − Institute for Health and Bioeconomy, NOVA
School of Science and Technology, NOVA University
Lisbon, 2819-516 Caparica, Portugal
- UCIBIO,
Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, 2819-516 Caparica, Portugal
| | - Alexandra R. Fernandes
- Associate
Laboratory i4HB − Institute for Health and Bioeconomy, NOVA
School of Science and Technology, NOVA University
Lisbon, 2819-516 Caparica, Portugal
- UCIBIO,
Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, 2819-516 Caparica, Portugal
| | - Tânia S. Morais
- Centro
de Química Estrutural, Institute of Molecular Sciences, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
| |
Collapse
|
57
|
Rong Z, Zheng K, Chen J, Jin X. The cross talk of ubiquitination and chemotherapy tolerance in colorectal cancer. J Cancer Res Clin Oncol 2024; 150:154. [PMID: 38521878 PMCID: PMC10960765 DOI: 10.1007/s00432-024-05659-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 02/20/2024] [Indexed: 03/25/2024]
Abstract
Ubiquitination, a highly adaptable post-translational modification, plays a pivotal role in maintaining cellular protein homeostasis, encompassing cancer chemoresistance-associated proteins. Recent findings have indicated a potential correlation between perturbations in the ubiquitination process and the emergence of drug resistance in CRC cancer. Consequently, numerous studies have spurred the advancement of compounds specifically designed to target ubiquitinates, offering promising prospects for cancer therapy. In this review, we highlight the role of ubiquitination enzymes associated with chemoresistance to chemotherapy via the Wnt/β-catenin signaling pathway, epithelial-mesenchymal transition (EMT), and cell cycle perturbation. In addition, we summarize the application and role of small compounds that target ubiquitination enzymes for CRC treatment, along with the significance of targeting ubiquitination enzymes as potential cancer therapies.
Collapse
Affiliation(s)
- Ze Rong
- Department of Chemoradiotherapy, the Affiliated People's Hospital of Ningbo University, Ningbo, 315040, China.
| | - Kaifeng Zheng
- Department of Chemoradiotherapy, the Affiliated People's Hospital of Ningbo University, Ningbo, 315040, China
| | - Jun Chen
- Department of Chemoradiotherapy, the Affiliated People's Hospital of Ningbo University, Ningbo, 315040, China.
| | - Xiaofeng Jin
- Department of Chemoradiotherapy, the Affiliated People's Hospital of Ningbo University, Ningbo, 315040, China.
- Department of Biochemistry and Molecular Biology, Health Science Center, Ningbo, 315211, China.
| |
Collapse
|
58
|
Yang Q, Meng D, Zhang Q, Wang J. Advances in research on the anti-tumor mechanism of Astragalus polysaccharides. Front Oncol 2024; 14:1334915. [PMID: 38515577 PMCID: PMC10955345 DOI: 10.3389/fonc.2024.1334915] [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: 11/09/2023] [Accepted: 02/22/2024] [Indexed: 03/23/2024] Open
Abstract
The dry root of the soybean plant Astragalus membranaceus (Fisch) Bge. var. mongholicus (Bge) Hsiao or A. membranaceus (Fisch) Bge, Astragali Radix (AR) has a long medicinal history. Astragalus polysaccharide (APS), the natural macromolecule that exhibits immune regulatory, anti-inflammatory, anti-tumor, and other pharmacological activities, is an important active ingredient extracted from AR. Recently, APS has been increasingly used in cancer therapy owing to its anti-tumor ability as it prevents the progression of prostate, liver, cervical, ovarian, and non-small-cell lung cancer by suppressing tumor cell growth and invasion and enhancing apoptosis. In addition, APS enhances the sensitivity of tumors to antineoplastic agents and improves the body's immunity. This macromolecule has prospects for broad application in tumor therapy through various pathways. In this article, we present the latest progress in the research on the anti-tumor effects of APS and its underlying mechanisms, aiming to provide novel theoretical support and reference for its use in cancer therapy.
Collapse
Affiliation(s)
| | | | - Qinyuan Zhang
- School of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jin Wang
- School of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
59
|
Sedky NK, Mahdy NK, Abdel-Kader NM, Abdelhady MMM, Maged M, Allam AL, Alfaifi MY, Shamma SN, Hassan HAFM, Fahmy SA. Facile sonochemically-assisted bioengineering of titanium dioxide nanoparticles and deciphering their potential in treating breast and lung cancers: biological, molecular, and computational-based investigations. RSC Adv 2024; 14:8583-8601. [PMID: 38487521 PMCID: PMC10938292 DOI: 10.1039/d3ra08908h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 03/07/2024] [Indexed: 03/17/2024] Open
Abstract
Combining sonochemistry with phytochemistry is a modern trend in the biosynthesis of metallic nanoparticles (NPs), which contributes to the sustainability of chemical processes and minimizes hazardous effects. Herein, titanium dioxide (TiO2) NPs were bioengineered using a novel and facile ultrasound-assisted approach utilizing the greenly extracted essential oil of Ocimum basilicum. FTIR and UV-Vis spectrophotometry were used to confirm the formation of TiO2 NPs. The X-ray diffraction (XRD) analysis showed the crystalline nature of TiO2 NPs. TEM analysis revealed the spherical morphology of the NPs with sizes ranging from 5.55 to 13.89 nm. Energy-dispersive X-ray (EDX) confirmed the purity of the greenly synthesized NPs. TiO2 NPs demonstrated outstanding antitumor activity against breast (MCF-7) and lung (A-549) cancer cells with estimated IC50 values of 1.73 and 4.79 μg mL-1. The TiO2 NPs were cytocompatible to normal cells (MCF-10A) with a selectivity index (SI) of 8.77 for breast and 3.17 for lung cancer. Biological assays revealed a promising potential for TiO2 NPs to induce apoptosis and arrest cells at the sub-G1 phase of the cell cycle phase in both cancer cell lines. Molecular investigations showed the ability of TiO2 NPs to increase apoptotic genes' expression (Bak and Bax) and their profound ability to elevate the expression of apoptotic proteins (caspases 3 and 7). Molecular docking demonstrated strong binding interactions for TiO2 NPs with caspase 3 and EGFR-TK targets. In conclusion, the greenly synthesized TiO2 NPs exhibited potent antitumor activity and mitochondrion-based cell death against breast and lung cancer cell lines while maintaining cytocompatibility against normal cells.
Collapse
Affiliation(s)
- Nada K Sedky
- Department of Biochemistry, School of Life and Medical Sciences, University of Hertfordshire Hosted by Global Academic Foundation R5 New Garden City, New Administrative Capital Cairo 11835 Egypt
| | - Noha Khalil Mahdy
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University Kasr El-Aini Street Cairo 11562 Egypt
| | - Nour M Abdel-Kader
- Department of Biochemistry, School of Life and Medical Sciences, University of Hertfordshire Hosted by Global Academic Foundation R5 New Garden City, New Administrative Capital Cairo 11835 Egypt
- Department of Biochemistry, Faculty of Science, Ain Shams University Cairo 11566 Egypt
| | - Manal M M Abdelhady
- Clinical Pharmacy Department, Faculty of Pharmacy, Badr University Cairo 11829 Egypt
| | - Mohamad Maged
- Faculty of Biotechnology, Nile University Giza Egypt
| | - Aya L Allam
- School of Life and Medical Sciences, University of Hertfordshire Hosted by Global Academic Foundation New Administrative Capital Egypt
| | - Mohammad Y Alfaifi
- King Khalid University, Faculty of Science, Biology Department Abha 9004 Saudi Arabia
| | - Samir N Shamma
- Institute of Global Health and Human Ecology, School of Sciences & Engineering, The American University in Cairo AUC Avenue, P.O. Box 74 New Cairo 11835 Egypt
| | - Hatem A F M Hassan
- School of Life and Medical Sciences, University of Hertfordshire Hosted by Global Academic Foundation New Administrative Capital Egypt
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University 11562 Cairo Egypt
| | - Sherif Ashraf Fahmy
- Department of Chemistry, School of Life and Medical Sciences, University of Hertfordshire Hosted by Global Academic Foundation R5 New Garden City, New Capital Cairo 11835 Egypt +20-1222613344
| |
Collapse
|
60
|
Li Y, Zhu L, Mao J, Zheng H, Hu Z, Yang S, Mao T, Zhou T, Cao P, Wu H, Wang X, Wang J, Lin F, Shen H. Genome-scale CRISPR-Cas9 screen identifies PAICS as a therapeutic target for EGFR wild-type non-small cell lung cancer. MedComm (Beijing) 2024; 5:e483. [PMID: 38463398 PMCID: PMC10924642 DOI: 10.1002/mco2.483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 01/11/2024] [Accepted: 01/14/2024] [Indexed: 03/12/2024] Open
Abstract
Epidermal growth factor receptor-targeted (EGFR-targeted) therapies show promise for non-small cell lung cancer (NSCLC), but they are ineffective in a third of patients who lack EGFR mutations. This underlines the need for personalized treatments for patients with EGFR wild-type NSCLC. A genome-wide CRISPR/Cas9 screen has identified the enzyme phosphoribosylaminoimidazole carboxylase/phosphoribosylaminoimidazole succinocarboxamide synthetase (PAICS), which is vital in de novo purine biosynthesis and tumor development, as a potential drug target for EGFR wild-type NSCLC. We have further confirmed that PAICS expression is significantly increased in NSCLC tissues and correlates with poor patient prognosis. Knockdown of PAICS resulted in a marked reduction in both in vitro and in vivo proliferation of EGFR wild-type NSCLC cells. Additionally, PAICS silencing led to cell-cycle arrest in these cells, with genes involved in the cell cycle pathway being differentially expressed. Consistently, an increase in cell proliferation ability and colony number was observed in cells with upregulated PAICS in EGFR wild-type NSCLC. PAICS silencing also caused DNA damage and cell-cycle arrest by interacting with DNA repair genes. Moreover, decreased IMPDH2 activity and activated PI3K-AKT signaling were observed in NSCLC cells with EGFR mutations, which may compromise the effectiveness of PAICS knockdown. Therefore, PAICS plays an oncogenic role in EGFR wild-type NSCLC and represents a potential therapeutic target for this disease.
Collapse
Affiliation(s)
- Yufeng Li
- Department of Medical OncologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsuChina
- Department of Medical OncologyThe Affiliated Sir Run Run Hospital of Nanjing Medical UniversityNanjingJiangsuChina
| | - Lingyun Zhu
- Department of Cell BiologySchool of Basic Medical Sciences, Nanjing Medical UniversityNanjingJiangsuChina
| | - Jiaqi Mao
- Department of Cell BiologySchool of Basic Medical Sciences, Nanjing Medical UniversityNanjingJiangsuChina
| | - Hongrui Zheng
- Department of OrthopedicsTaizhou Hospital of Zhejiang ProvinceAffiliated to Wenzhou Medical UniversityZhejiangChina
| | - Ziyi Hu
- Department of Cell BiologySchool of Basic Medical Sciences, Nanjing Medical UniversityNanjingJiangsuChina
| | - Suisui Yang
- Department of Cell BiologySchool of Basic Medical Sciences, Nanjing Medical UniversityNanjingJiangsuChina
| | - Tianyu Mao
- Department of Medical OncologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsuChina
| | - Tingting Zhou
- Department of Cell BiologySchool of Basic Medical Sciences, Nanjing Medical UniversityNanjingJiangsuChina
| | - Pingping Cao
- Department of Cell BiologySchool of Basic Medical Sciences, Nanjing Medical UniversityNanjingJiangsuChina
| | - Hongshuai Wu
- Department of Cell BiologySchool of Basic Medical Sciences, Nanjing Medical UniversityNanjingJiangsuChina
- Department of PharmacologyNanjing Medical UniversityNanjingJiangsuChina
| | - Xuerong Wang
- Department of PharmacologyNanjing Medical UniversityNanjingJiangsuChina
| | - Jing Wang
- Department of Cell BiologySchool of Basic Medical Sciences, Nanjing Medical UniversityNanjingJiangsuChina
| | - Fan Lin
- Department of Cell BiologySchool of Basic Medical Sciences, Nanjing Medical UniversityNanjingJiangsuChina
- Institute for Brain Tumors & Key Laboratory of Rare Metabolic Diseases, Nanjing Medical UniversityNanjingJiangsuChina
- Department of GastroenterologyThe First Affiliated Hospitaland College of Clinical Medicine of Henan University of Science and TechnologyLuoyangHenanChina
| | - Hua Shen
- Department of Medical OncologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsuChina
- Department of Medical OncologyThe Affiliated Sir Run Run Hospital of Nanjing Medical UniversityNanjingJiangsuChina
| |
Collapse
|
61
|
Arafat M, Sakkal M, Beiram R, AbuRuz S. Nanomedicines: Emerging Platforms in Smart Chemotherapy Treatment-A Recent Review. Pharmaceuticals (Basel) 2024; 17:315. [PMID: 38543101 PMCID: PMC10974155 DOI: 10.3390/ph17030315] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 02/21/2024] [Accepted: 02/24/2024] [Indexed: 01/06/2025] Open
Abstract
Cancer continues to pose one of the most critical challenges in global healthcare. Despite the wide array of existing cancer drugs, the primary obstacle remains in selectively targeting and eliminating cancer cells while minimizing damage to healthy ones, thereby reducing treatment side effects. The revolutionary approach of utilizing nanomaterials for delivering cancer therapeutic agents has significantly enhanced the efficacy and safety of chemotherapeutic drugs. This crucial shift is attributed to the unique properties of nanomaterials, enabling nanocarriers to transport therapeutic agents to tumor sites in both passive and active modes, while minimizing drug elimination from delivery systems. Furthermore, these nanocarriers can be designed to respond to internal or external stimuli, thus facilitating controlled drug release. However, the production of nanomedications for cancer therapy encounters various challenges that can impede progress in this field. This review aims to provide a comprehensive overview of the current state of nanomedication in cancer treatment. It explores a variety of nanomaterials, focusing on their unique properties that are crucial for overcoming the limitations of conventional chemotherapy. Additionally, the review delves into the properties and functionalities of nanocarriers, highlighting their significant impact on the evolution of nanomedicine. It also critically assesses recent advancements in drug delivery systems, covering a range of innovative delivery methodologies. Finally, the review succinctly addresses the challenges encountered in developing nanomedications, offering insightful perspectives to guide future research in this field.
Collapse
Affiliation(s)
- Mosab Arafat
- College of Pharmacy, Al Ain University, Al Ain P.O. Box 64141, United Arab Emirates; (M.A.)
| | - Molham Sakkal
- College of Pharmacy, Al Ain University, Al Ain P.O. Box 64141, United Arab Emirates; (M.A.)
| | - Rami Beiram
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 17666, United Arab Emirates
| | - Salahdein AbuRuz
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 17666, United Arab Emirates
- Department of Biopharmaceutics and Clinical Pharmacy, School of Pharmacy, The University of Jordan, Amman 11942, Jordan
| |
Collapse
|
62
|
Jiang W, Yuan L, Liu Q, Li X, Yang Y, Li J, Jiao T, Niu Y, Zhang L, Dou H, Nan Y. The mechanism of action and experimental verification of Gan-song Yin on renal clear cell carcinoma based on network pharmacology and bioinformatics. Discov Oncol 2024; 15:52. [PMID: 38416262 PMCID: PMC10902223 DOI: 10.1007/s12672-024-00909-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 02/25/2024] [Indexed: 02/29/2024] Open
Abstract
BACKGROUND Gan-song Yin (GSY) is originated from the scripture "Gan-song Pills", a medical work of the Ningxia ethnic minorities, and its treatment of kidney diseases has good results. Its method of treating Renal clear cell carcinoma (KIRC) is still unknown, nevertheless. METHODS Firstly, utilizing a network pharmacology strategy to screen GSY for active components and targets and looking up KIRC-related targets in GeneCards and GEO databases. Secondly, protein interaction networks were constructed and analyzed for GO and KEGG enrichment. Molecular docking was then performed and clinical and other correlations of the network pharmacology results were analyzed using bioinformatic analysis methods. Finally, we performed in vitro cellular experiments with 786-O cells and ACHN cells to validate the results of network pharmacology and bioinformatic analysis. RESULTS With the help of network pharmacological analysis, six hub targets were eliminated. Bioinformatics study revealed that the hub targets has clinically significant clinical guiding importance. The results showed that GSY inhibited the proliferation of 786-O cells and ACHN cells, induced cell apoptosis, blocked cell cycle, and reduced cell colony formation ability. qRT-PCR results showed that GSY promoted the expression of ALB and CASP3 genes, and inhibited the expression of EGFR, JUN, MYC and VEGFA genes. Western blot results showed that GSY could promote the expression of ALB and CASP3 protein, and inhibit the expression of EGFR, JUN, MYC and VEGFA protein. CONCLUSIONS Network pharmacology and bioinformatics analysis showed that GSY could act on multiple targets through a variety of components to achieve the effect of treating KIRC. In this study, we confirmed that GSY inhibits KIRC by regulating the expression of core targets through in vitro cellular experiments, thus providing a reference for subsequent related studies.
Collapse
Affiliation(s)
- Wenjie Jiang
- Key Laboratory of Ningxia Minority Medicine Modernization Ministry of Education, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
- School of Traditional Chinese Medicine, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Ling Yuan
- College of Pharmacy, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Qian Liu
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Xiangyang Li
- School of Traditional Chinese Medicine, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Yifan Yang
- Key Laboratory of Ningxia Minority Medicine Modernization Ministry of Education, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
- School of Traditional Chinese Medicine, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Jiaqing Li
- Key Laboratory of Ningxia Minority Medicine Modernization Ministry of Education, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
- School of Traditional Chinese Medicine, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Taiqiang Jiao
- School of Traditional Chinese Medicine, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Yang Niu
- Key Laboratory of Ningxia Minority Medicine Modernization Ministry of Education, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
- School of Traditional Chinese Medicine, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Lei Zhang
- School of Traditional Chinese Medicine, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Hongli Dou
- Key Laboratory of Ningxia Minority Medicine Modernization Ministry of Education, Ningxia Medical University, Yinchuan, 750004, Ningxia, China.
- School of Traditional Chinese Medicine, Ningxia Medical University, Yinchuan, 750004, Ningxia, China.
- Shaanxi University of Traditional Chinese Medicine, Xianyang, 712046, Shaanxi, China.
| | - Yi Nan
- Key Laboratory of Ningxia Minority Medicine Modernization Ministry of Education, Ningxia Medical University, Yinchuan, 750004, Ningxia, China.
| |
Collapse
|
63
|
Correa WA, das Neves SC, Oliveira RJ, Kassuya CA, Navarro SD, Martins ACF, Saroja B, Mitsuyasu B, da Silveira IOMF, Vitor N, Coelho HRS, Vilela MLB, do Nascimento VA, de Lima DP, Beatriz A, da Silva Gomes R. Chemotherapeutic Mechanism of Action of the Synthetic Resorcinolic Methyl 3,5-dimethoxy-2-octanoylbenzoate. Chem Res Toxicol 2024; 37:259-273. [PMID: 38183658 PMCID: PMC11987490 DOI: 10.1021/acs.chemrestox.3c00269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2024]
Abstract
Resorcinolic lipids are described as potential examples of selective chemotherapeutic adjuvants that can enhance the effects of cyclophosphamide (CYC) while promoting cell death without causing DNA damage. Therefore, the current study attempted to describe how the resorcinolic lipid methyl 3,5-dimethoxy-2-octanoylbenzoate (AMS35BB) interacted with DNA (DNA docking) and how this compound affected genetic toxicology models and other biological characteristics when combined with CYC. We observed that AMS35BB, used alone (7.5 and 10 mg/kg), increases the frequency of genomic damage (comet assay) but not chromosomal damage (micronuclei assay), lowers phagocytosis, and promotes cell death in Swiss male mice. When used in association with CYC, AMS35BB can reduce the risk of genomic damage by up to 33.8% as well as chromosomal damage, splenic phagocytosis, cell death, and lymphocyte frequency. Molecular docking showed that AMS35BB had a higher affinity than the active metabolite of CYC for binding to the DNA double helix major groove. As a result, AMS35BB has the potential to be both an adjuvant when used in association with CYC and a therapeutic candidate for the development of a selective chemotherapeutic drug.
Collapse
Affiliation(s)
- Willian Ayala Correa
- Institute of Chemistry, Federal University of Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul 79070-900, Brazil
| | - Silvia Cordeiro das Neves
- Stem Cell, Cell Therapy and Toxicological Genetics Research Centre (CeTroGen), Medical School, Federal University of Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul 79080-190, Brazil; Graduate Program in Health and Development in the Midwest Region, Medical School, Federal University of Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul 79070-900, Brazil
| | - Rodrigo Juliano Oliveira
- Stem Cell, Cell Therapy and Toxicological Genetics Research Centre (CeTroGen), Medical School, Federal University of Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul 79080-190, Brazil; Graduate Program in Health and Development in the Midwest Region, Medical School, Federal University of Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul 79070-900, Brazil
| | - Cândida A. Kassuya
- School of Health Sciences, Federal University of Grande Dourados, Dourados, Mato Grosso do Sul 79804-970, Brazil
| | - Stephanie D. Navarro
- Institute of Chemistry, Federal University of Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul 79070-900, Brazil; Stem Cell, Cell Therapy and Toxicological Genetics Research Centre (CeTroGen), Medical School, Federal University of Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul 79080-190, Brazil
| | | | - Baby Saroja
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota 58102, United States
| | - Barbara Mitsuyasu
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota 58102, United States; Department of Chemical and Biological Sciences, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo 18618-689, Brazil
| | | | - Neimar Vitor
- Institute of Chemistry, Federal University of Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul 79070-900, Brazil
| | - Henrique Rodrigues Scherer Coelho
- Stem Cell, Cell Therapy and Toxicological Genetics Research Centre (CeTroGen), Medical School, Federal University of Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul 79080-190, Brazil
| | - Marcelo L. B. Vilela
- Institute of Chemistry, Federal University of Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul 79070-900, Brazil
| | - Valter A. do Nascimento
- Graduate Program in Health and Development in the Midwest Region, Medical School, Federal University of Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul 79070-900, Brazil
| | - Dênis P. de Lima
- Institute of Chemistry, Federal University of Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul 79070-900, Brazil
| | - Adilson Beatriz
- Institute of Chemistry, Federal University of Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul 79070-900, Brazil
| | - Roberto da Silva Gomes
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota 58102, United States
| |
Collapse
|
64
|
Chen J, Li W, Li G, Liu X, Huang C, Nie H, Liang L, Wang Y, Liu Y. Targeted liposomes encapsulated iridium(III) compound greatly enhance anticancer efficacy and induce cell death via ferroptosis on HepG2 cells. Eur J Med Chem 2024; 265:116078. [PMID: 38141286 DOI: 10.1016/j.ejmech.2023.116078] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 12/01/2023] [Accepted: 12/18/2023] [Indexed: 12/25/2023]
Abstract
In this study, ligands 2-phenyl-1H-imidazo[4,5-f][1,10]phenanthroline (PIP), 2-(2-nitrophenyl)-1H-imidazo[4,5-f][1,10]phenanthroline (NPIP), 2-(2-nitronaphthalen-1-yl)-1H-imidazo[4,5-f][1,10]phenanthroline (NNIP) and their iridium(III) metal compounds [Ir(ppy)2(PIP)](PF6) (ppy = 2-phenylpyridine, 1a), [Ir(ppy)2(NPIP)](PF6) (1b), [Ir(ppy)2(NNIP)](PF6) (1c) were designed and synthesized. The anti-cancer activities of 1a, 1b and 1c on BEL-7402, HepG2, SK-Hep1 and non-cancer LO2 were detected using MTT method. 1a shows moderate, 1b and 1c display low or no anti-cancer activities. To elevate the anti-cancer effectiveness, encapsulating the compounds 1a, 1b and 1c into the ordinary or targeted liposomes to produce 1alip, 1blip, 1clip, or targeted 1aTlip, 1bTlip and 1cTlip. The IC50 values of 1alip, 1blip, 1clip, 1aTlip, 1bTlip and 1cTlip against HepG2 cells are 7.9 ± 0.1, 8.6 ± 0.2, 16.9 ± 0.5, 5.9 ± 0.2, 7.3 ± 0.1 and 9.7 ± 0.7 μM, respectively. Specifically, the anti-tumor activity assays in vivo found that the inhibitory rates are 23.24 % for 1a, 61.27 % for 1alip, 76.06 % for 1aTlip. It is obvious that the targeted liposomes entrapped iridium(III) compound greatly enhance anti-cancer efficacy. Additionally, 1alip, 1blip and 1clip or targeted 1aTlip, 1bTlip and 1cTlip can effectively restrain the cell colony and proliferation in the G0/G1 period. 1alip, 1blip, 1clip, 1aTlip, 1bTlip and 1cTlip can increase reactive oxygen species (ROS) concentration, arouse a decline in the mitochondrial membrane potential and promote Ca2+ release. RNA-sequence was applied to examine the signaling pathways. Taken together, the liposomes or targeted liposomes encapsulated compounds trigger cell death by way of apoptosis, autophagy, ferroptosis, disruption of mitochondrial function and PI3K/AKT/mTOR signaling pathways.
Collapse
Affiliation(s)
- Jing Chen
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Wenlong Li
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Gechang Li
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | | | - Chunxia Huang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Hua Nie
- Jiaying University, Meizhou, 514031, PR China.
| | - Lijuan Liang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Yi Wang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Yunjun Liu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China.
| |
Collapse
|
65
|
Falcone N, Ermis M, Gangrade A, Choroomi A, Young P, Mathes TG, Monirizad M, Zehtabi F, Mecwan M, Rodriguez M, Zhu Y, Byun Y, Khademhosseini A, de Barros NR, Kim H. Drug‐Eluting Shear‐Thinning Hydrogel for the Delivery of Chemo‐ and Immunotherapeutic Agents for the Treatment of Hepatocellular Carcinoma. ADVANCED FUNCTIONAL MATERIALS 2024; 34. [DOI: 10.1002/adfm.202309069] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Indexed: 01/06/2025]
Abstract
AbstractHepatocellular carcinoma (HCC) is a malignant and deadly form of liver cancer with limited treatment options. Transcatheter arterial chemoembolization, a procedure that delivers embolic and chemotherapeutic agents through blood vessels, is a promising cancer treatment strategy. However, it still faces limitations, such as inefficient agent delivery and the inability to address tumor‐induced immunosuppression. Here, a drug‐eluting shear‐thinning hydrogel (DESTH) loaded with chemotherapeutic and immunotherapeutic agents in nanocomposite hydrogels composed of gelatin and nanoclays is presented as a therapeutic strategy for a catheter‐based endovascular anticancer approach. DESTH is manually deliverable using a conventional needle and catheter. In addition, drug release studies show a sustained and pH‐dependent co‐delivery of the chemotherapy doxorubicin (acidic pH) and the immune‐checkpoint inhibitor aPD‐1 (neutral pH). In a mouse liver tumor model, the DESTH‐based chemo/immunotherapy combination has the highest survival rate and smallest residual tumor size. Finally, immunofluorescence analysis confirms that DESTH application enhances cell death and increases intratumoral infiltration of cytotoxic T‐cells. In conclusion, the results show that DESTH, which enables efficient ischemic tumor cell death and effective co‐delivery of chemo‐ and immunotherapeutic agents, may have the potential to be an effective therapeutic modality in the treatment of HCC.
Collapse
Affiliation(s)
- Natashya Falcone
- Terasaki Institute for Biomedical Innovation (TIBI) Los Angeles CA 90024 USA
| | - Menekse Ermis
- Terasaki Institute for Biomedical Innovation (TIBI) Los Angeles CA 90024 USA
| | - Ankit Gangrade
- Terasaki Institute for Biomedical Innovation (TIBI) Los Angeles CA 90024 USA
| | - Auveen Choroomi
- Terasaki Institute for Biomedical Innovation (TIBI) Los Angeles CA 90024 USA
| | - Patric Young
- Terasaki Institute for Biomedical Innovation (TIBI) Los Angeles CA 90024 USA
| | - Tess G. Mathes
- Terasaki Institute for Biomedical Innovation (TIBI) Los Angeles CA 90024 USA
| | - Mahsa Monirizad
- Terasaki Institute for Biomedical Innovation (TIBI) Los Angeles CA 90024 USA
| | - Fatemeh Zehtabi
- Terasaki Institute for Biomedical Innovation (TIBI) Los Angeles CA 90024 USA
| | - Marvin Mecwan
- Terasaki Institute for Biomedical Innovation (TIBI) Los Angeles CA 90024 USA
| | - Marco Rodriguez
- Terasaki Institute for Biomedical Innovation (TIBI) Los Angeles CA 90024 USA
| | - Yangzhi Zhu
- Terasaki Institute for Biomedical Innovation (TIBI) Los Angeles CA 90024 USA
| | - Youngjoo Byun
- Department of Pathophysiology and Preclinical Science College of Pharmacy Korea University 30019 Sejong Republic of Korea
| | - Ali Khademhosseini
- Terasaki Institute for Biomedical Innovation (TIBI) Los Angeles CA 90024 USA
| | | | - Han‐Jun Kim
- Terasaki Institute for Biomedical Innovation (TIBI) Los Angeles CA 90024 USA
- Department of Pathophysiology and Preclinical Science College of Pharmacy Korea University 30019 Sejong Republic of Korea
- Vellore Institute of Technology (VIT) Vellore 632014 India
| |
Collapse
|
66
|
Diehl FF, Sapp KM, Vander Heiden MG. The bidirectional relationship between metabolism and cell cycle control. Trends Cell Biol 2024; 34:136-149. [PMID: 37385879 DOI: 10.1016/j.tcb.2023.05.012] [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: 02/03/2023] [Revised: 05/30/2023] [Accepted: 05/31/2023] [Indexed: 07/01/2023]
Abstract
The relationship between metabolism and cell cycle progression is complex and bidirectional. Cells must rewire metabolism to meet changing biosynthetic demands across cell cycle phases. In turn, metabolism can influence cell cycle progression through direct regulation of cell cycle proteins, through nutrient-sensing signaling pathways, and through its impact on cell growth, which is linked to cell division. Furthermore, metabolism is a key player in mediating quiescence-proliferation transitions in physiologically important cell types, such as stem cells. How metabolism impacts cell cycle progression, exit, and re-entry, as well as how these processes impact metabolism, is not fully understood. Recent advances uncovering mechanistic links between cell cycle regulators and metabolic processes demonstrate a complex relationship between metabolism and cell cycle control, with many questions remaining.
Collapse
Affiliation(s)
- Frances F Diehl
- Department of Molecular Biology and Genetics, Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Koch Institute for Integrative Cancer Research and the Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Kiera M Sapp
- Koch Institute for Integrative Cancer Research and the Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Matthew G Vander Heiden
- Koch Institute for Integrative Cancer Research and the Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA; Dana-Farber Cancer Institute, Boston, MA, USA.
| |
Collapse
|
67
|
Eissa IH, Elkady H, Rashed M, Elwan A, Hagras M, Dahab MA, Taghour MS, Ibrahim IM, Husein DZ, Elkaeed EB, Al-ghulikah HA, Metwaly AM, Mahdy HA. Discovery of new thiazolidine-2,4-dione derivatives as potential VEGFR-2 inhibitors: In vitro and in silico studies. Heliyon 2024; 10:e24005. [PMID: 38298627 PMCID: PMC10828660 DOI: 10.1016/j.heliyon.2024.e24005] [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: 10/06/2023] [Revised: 12/12/2023] [Accepted: 01/02/2024] [Indexed: 02/02/2024] Open
Abstract
In this study, a series of seven novel 2,4-dioxothiazolidine derivatives with potential anticancer and VEGFR-2 inhibiting abilities were designed and synthesized as VEGFR-2 inhibitors. The synthesized compounds were tested in vitro for their potential to inhibit VEGFR-2 and the growth of HepG2 and MCF-7 cancer cell lines. Among the compounds tested, compound 22 (IC50 = 0.079 μM) demonstrated the highest anti-VEGFR-2 efficacy. Furthermore, it demonstrated significant anti-proliferative activities against HepG2 (IC50 = 2.04 ± 0.06 μM) and MCF-7 (IC50 = 1.21 ± 0.04 M). Additionally, compound 22 also increased the total apoptotic rate of the MCF-7 cancer cell lines with cell cycle arrest at S phase. As well, computational methods were applied to study the VEGFR-2-22 complex at the molecular level. Molecular docking and molecular dynamics (MD) simulations were used to investigate the complex's structural and kinetic characteristics. The DFT calculations further revealed the structural and electronic properties of compound 22. Finally, computational ADMET and toxicity tests were performed indicating the likeness of the proposed compounds to be drugs. The results suggest that compound 22 displays promise as an effective anticancer treatment and can serve as a model for future structural modifications and biological investigations in this field.
Collapse
Affiliation(s)
- Ibrahim H. Eissa
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, 11884, Egypt
| | - Hazem Elkady
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, 11884, Egypt
| | - Mahmoud Rashed
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, 11884, Egypt
| | - Alaa Elwan
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, 11884, Egypt
| | - Mohamed Hagras
- Department of Pharmaceutical Organic Chemistry, College of Pharmacy, Al-Azhar University, Cairo, 11884, Egypt
| | - Mohammed A. Dahab
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, 11884, Egypt
| | - Mohammed S. Taghour
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, 11884, Egypt
| | - Ibrahim M. Ibrahim
- Biophysics Department, Faculty of Science, Cairo University, Giza, 12613, Egypt
| | - Dalal Z. Husein
- Chemistry Department, Faculty of Science, New Valley University, El-Kharja, 72511, Egypt
| | - Eslam B. Elkaeed
- Department of Pharmaceutical Sciences, College of Pharmacy, AlMaarefa University, Riyadh, 13713, Saudi Arabia
| | - Hanan A. Al-ghulikah
- Department of Chemistry, College of Science, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Ahmed M. Metwaly
- Pharmacognosy and Medicinal Plants Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, 11884, Egypt
| | - Hazem A. Mahdy
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, 11884, Egypt
| |
Collapse
|
68
|
Wang J, Wei W, Xing C, Wang H, Liu M, Xu J, He X, Liu Y, Guo X, Jiang R. Transcriptome and Weighted Gene Co-Expression Network Analysis for Feather Follicle Density in a Chinese Indigenous Breed. Animals (Basel) 2024; 14:173. [PMID: 38200904 PMCID: PMC10778273 DOI: 10.3390/ani14010173] [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: 12/04/2023] [Revised: 12/27/2023] [Accepted: 01/03/2024] [Indexed: 01/12/2024] Open
Abstract
Feather follicle density plays an important role in appealing to consumers' first impressions when making purchasing decisions. However, the molecular network that contributes to this trait remains largely unknown. The aim of this study was to perform transcriptome and weighted gene co-expression network analyses to determine the candidate genes relating to feather follicle density in Wannan male chickens. In total, five hundred one-day-old Wannan male chickens were kept in a conventional cage system. Feather follicle density was recorded for each bird at 12 weeks of age. At 12 weeks, fifteen skin tissue samples were selected for weighted gene co-expression network analysis, of which six skin tissue samples (three birds in the H group and three birds in the L group) were selected for transcriptome analysis. The results showed that, in total, 95 DEGs were identified, and 56 genes were upregulated and 39 genes were downregulated in the high-feather-follicle-density group when compared with the low-feather-follicle-density group. Thirteen co-expression gene modules were identified. The red module was highly significantly negatively correlated with feather follicle density (p < 0.01), with a significant negative correlation coefficient of -0.72. In total, 103 hub genes from the red module were screened. Upon comparing the 103 hub genes with differentially expressed genes (DEGs), it was observed that 13 genes were common to both sets, including MELK, GTSE1, CDK1, HMMR, and CENPE. From the red module, FOXM1, GTSE1, MELK, CDK1, ECT2, and NEK2 were selected as the most important genes. These genes were enriched in the DNA binding pathway, the heterocyclic compound binding pathway, the cell cycle pathway, and the oocyte meiosis pathway. This study suggests that FOXM1, GTSE1, MELK, CDK1, ECT2, and NEK2 may be involved in regulating the development of feather follicle density in Wannan male chickens. The results of this study reveal the genetic structure and molecular regulatory network of feather follicle density in Wannan male chickens, and provide a basis for further elucidating the genetic regulatory mechanism and identifying molecular markers with breeding value.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Runshen Jiang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China; (J.W.); (W.W.); (C.X.); (H.W.); (M.L.); (J.X.); (X.H.); (Y.L.); (X.G.)
| |
Collapse
|
69
|
Maleki EH, Bahrami AR, Matin MM. Cancer cell cycle heterogeneity as a critical determinant of therapeutic resistance. Genes Dis 2024; 11:189-204. [PMID: 37588236 PMCID: PMC10425754 DOI: 10.1016/j.gendis.2022.11.025] [Citation(s) in RCA: 37] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 10/20/2022] [Accepted: 11/16/2022] [Indexed: 01/15/2023] Open
Abstract
Intra-tumor heterogeneity is now arguably one of the most-studied topics in tumor biology, as it represents a major obstacle to effective cancer treatment. Since tumor cells are highly diverse at genetic, epigenetic, and phenotypic levels, intra-tumor heterogeneity can be assumed as an important contributing factor to the nullification of chemotherapeutic effects, and recurrence of the tumor. Based on the role of heterogeneous subpopulations of cancer cells with varying cell-cycle dynamics and behavior during cancer progression and treatment; herein, we aim to establish a comprehensive definition for adaptation of neoplastic cells against therapy. We discuss two parallel and yet distinct subpopulations of tumor cells that play pivotal roles in reducing the effects of chemotherapy: "resistant" and "tolerant" populations. Furthermore, this review also highlights the impact of the quiescent phase of the cell cycle as a survival mechanism for cancer cells. Beyond understanding the mechanisms underlying the quiescence, it provides an insightful perspective on cancer stem cells (CSCs) and their dual and intertwined functions based on their cell cycle state in response to treatment. Moreover, CSCs, epithelial-mesenchymal transformed cells, circulating tumor cells (CTCs), and disseminated tumor cells (DTCs), which are mostly in a quiescent state of the cell cycle are proved to have multiple biological links and can be implicated in our viewpoint of cell cycle heterogeneity in tumors. Overall, increasing our knowledge of cell cycle heterogeneity is a key to identifying new therapeutic solutions, and this emerging concept may provide us with new opportunities to prevent the dreadful cancer recurrence.
Collapse
Affiliation(s)
- Ebrahim H. Maleki
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, 9177948974 Mashhad, Iran
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 31-007 Krakow, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, 30-348 Krakow, Poland
| | - Ahmad Reza Bahrami
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, 9177948974 Mashhad, Iran
- Industrial Biotechnology Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, 9177948974 Mashhad, Iran
| | - Maryam M. Matin
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, 9177948974 Mashhad, Iran
- Novel Diagnostics and Therapeutics Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, 9177948974 Mashhad, Iran
- Stem Cell and Regenerative Medicine Research Group, Iranian Academic Center for Education, Culture and Research (ACECR), Khorasan Razavi Branch, 917751376 Mashhad, Iran
| |
Collapse
|
70
|
Namazinia M, Mazlum SR, Mohajer S, Lim Abdullah K, Salehian M. A structured laughter yoga therapy program on patients with chemotherapy-induced nausea and vomiting: A randomized clinical trial. Asia Pac J Oncol Nurs 2024; 11:100337. [PMID: 38222968 PMCID: PMC10784676 DOI: 10.1016/j.apjon.2023.100337] [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: 08/28/2023] [Accepted: 11/08/2023] [Indexed: 01/16/2024] Open
Abstract
Objetive Chemotherapy is a prevalent cancer treatment, often accompanied by debilitating side effects such as nausea and vomiting. This study explores the potential effectiveness of laughter yoga, a combination of exercise and voluntary laughter, in alleviating chemotherapy-induced nausea and vomiting. Methods This two-group randomized clinical trial was conducted on 69 cancer patients undergoing chemotherapy at the Reza Chemotherapy and Oncology Center, Mashhad, Iran, in 2018. Patients were randomly divided into intervention and control groups. Both groups received routine self-care training, with the addition of four 20-min to 30-min laughter yoga sessions held immediately before one of their chemotherapy appointments for the intervention group only. Nausea and vomiting were assessed using the Morrow Assessment of Nausea and Emesis questionnaire at two stages, before and after the intervention. Data were analyzed with Chi-square, Independent-t, Mann-Whitney, Wilcoxon, and McNemar tests using Statistical Package for the Social Sciences (SPSS). Results The mean age of patients in the intervention group was 49.0 ± 9.6 years, while in the control group, it was 45.2 ± 12.6 years. The intragroup comparison showed a statistically significant decrease in the severity and duration of nausea in the intervention group and a statistically significant increase in the severity and duration of nausea in the control group from pre-test to post-test (P < 0.05). The intergroup comparison showed no statistically significant difference between the two groups in terms of vomiting conditions. Conclusions Laughter yoga demonstrates promise in improving chemotherapy-induced nausea, suggesting its potential recommendation for managing this distressing side effect. Further research is warranted to explore its broader application in cancer care. Trial registration This study (No. IRCT20180429039463N1) was registered in the Iranian Registry of Clinical Trials on 21/08/2018.
Collapse
Affiliation(s)
- Mohammad Namazinia
- Department of Nursing, School of Nursing and Midwifery, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
- Department of Medical Surgical Nursing, School of Nursing and Midwifery, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyyed Reza Mazlum
- Department of Medical Surgical Nursing, School of Nursing and Midwifery, Mashhad University of Medical Sciences, Mashhad, Iran
- Nursing and Midwifery Care Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Samira Mohajer
- Nursing and Midwifery Care Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Khatijah Lim Abdullah
- Nursing and Midwifery Care Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Nursing, School of Medical and Life Science, Sunway University, Bandar Sunway, Malaysia
| | - Maryam Salehian
- Department of Medical Surgical Nursing, School of Nursing and Midwifery, Mashhad University of Medical Sciences, Mashhad, Iran
- Nursing and Midwifery Care Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
71
|
Liu Z, Tan X, Peng L, Gao W, Zeng P. Hederagenin Induces Apoptosis of Human Hepatoma HepG2 Cells via the Mitochondrial Pathway. Comb Chem High Throughput Screen 2024; 27:1495-1503. [PMID: 37817515 PMCID: PMC11327765 DOI: 10.2174/0113862073254353230925074944] [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: 04/04/2023] [Revised: 07/23/2023] [Accepted: 08/18/2023] [Indexed: 10/12/2023]
Abstract
OBJECTIVE The objective of this study is to assess the antitumor effects of hederagenin (HDG) in liver cancer (LC) cells and explore the related mechanisms. MATERIALS AND METHODS HepG2 cells were treated with HDG and cisplatin, respectively. The CCK8 assay was used to detect cell activity, DAPI staining was used to detect the proportion of living cells, TUNEL assay to detect the proportion of apoptotic cells, flow cytometry to detect the membrane potential, fluoroscopic electron microscopy to detect microstructural changes to the mitochondrial, and western blot analysis and high-content screening to detect apoptosisrelated proteins. RESULTS Treatment with HDG inhibited the growth of HepG2 cells, decreased the proportion of viable cells, increased the proportion of apoptotic cells, and significantly increased the proportion of cells in the G1 phase. Fluorescence staining showed that HDG damaged the mitochondria of HepG2 cells and significantly decreased the number of mitochondria. Flow cytometry showed that HDG decreased the mitochondrial membrane potential of HepG2 cells. Observations by electron microscopy showed that HDG caused swelling and vacuole formation of the mitochondria of HepG2 cells. HDG significantly reduced the average fluorescence intensity of Bcl-2 in HepG2 cells and significantly increased that of the pro-apoptosis proteins Bax, Cytochrome-c, and Caspase-3. CONCLUSION HDG induced apoptosis of HepG2 cells via the mitochondrial pathway.
Collapse
Affiliation(s)
- Zhuo Liu
- Affiliated Hospital, Hunan Academy of Traditional Chinese Medicine, Changsha 410006, P.R. China
- College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208, P.R. China
| | - Xiaoning Tan
- Affiliated Hospital, Hunan Academy of Traditional Chinese Medicine, Changsha 410006, P.R. China
| | - Lian Peng
- College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208, P.R. China
| | - Wenhui Gao
- College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208, P.R. China
| | - Puhua Zeng
- Affiliated Hospital, Hunan Academy of Traditional Chinese Medicine, Changsha 410006, P.R. China
| |
Collapse
|
72
|
Wang J, Li L, Xu ZP. Enhancing Cancer Chemo-Immunotherapy: Innovative Approaches for Overcoming Immunosuppression by Functional Nanomaterials. SMALL METHODS 2024; 8:e2301005. [PMID: 37743260 DOI: 10.1002/smtd.202301005] [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: 08/04/2023] [Revised: 09/05/2023] [Indexed: 09/26/2023]
Abstract
Chemotherapy is a critical modality in cancer therapy to combat malignant cell proliferation by directly attacking cancer cells and inducing immunogenic cell death, serving as a vital component of multi-modal treatment strategies for enhanced therapeutic outcomes. However, chemotherapy may inadvertently contribute to the immunosuppression of the tumor microenvironment (TME), inducing the suppression of antitumor immune responses, which can ultimately affect therapeutic efficacy. Chemo-immunotherapy, combining chemotherapy and immunotherapy in cancer treatment, has emerged as a ground-breaking approach to target and eliminate malignant tumors and revolutionize the treatment landscape, offering promising, durable responses for various malignancies. Notably, functional nanomaterials have substantially contributed to chemo-immunotherapy by co-delivering chemo-immunotherapeutic agents and modulating TME. In this review, recent advancements in chemo-immunotherapy are thus summarized to enhance treatment effectiveness, achieved by reversing the immunosuppressive TME (ITME) through the exploitation of immunotherapeutic drugs, or immunoregulatory nanomaterials. The effects of two-way immunomodulation and the causes of immunoaugmentation and suppression during chemotherapy are illustrated. The current strategies of chemo-immunotherapy to surmount the ITME and the functional materials to target and regulate the ITME are discussed and compared. The perspective on tumor immunosuppression reversal strategy is finally proposed.
Collapse
Affiliation(s)
- Jingjing Wang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Li Li
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Zhi Ping Xu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, 4072, Australia
| |
Collapse
|
73
|
Kwon M, Jung HJ. Hovenia dulcis Suppresses the Growth of Huh7-Derived Liver Cancer Stem Cells by Inducing Necroptosis and Apoptosis and Blocking c-MET Signaling. Cells 2023; 13:22. [PMID: 38201226 PMCID: PMC10778038 DOI: 10.3390/cells13010022] [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: 10/24/2023] [Revised: 12/09/2023] [Accepted: 12/19/2023] [Indexed: 01/12/2024] Open
Abstract
Liver cancer stem cells (LCSCs) contribute to the initiation, metastasis, treatment resistance, and recurrence of hepatocellular carcinoma (HCC). Therefore, exploring potential anticancer agents targeting LCSCs may offer new therapeutic options to overcome HCC treatment failure. Hovenia dulcis Thunberg (HDT), a tree from the buckthorn family found in Asia, exhibits various biological activities, including antifatigue, antidiabetic, neuroprotective, hepatoprotective, and antitumor activities. However, the therapeutic effect of HDT in eliminating LCSCs remains to be confirmed. In this study, we evaluated the inhibitory activity of ethanol, chloroform, and ethyl acetate extracts from HDT branches on the growth of Huh7-derived LCSCs. The ethyl acetate extract of HDT (EAHDT) exhibited the most potent inhibitory activity against the growth of Huh7 LCSCs among the three HDT extracts. EAHDT suppressed the in vitro self-renewal ability of Huh7 LCSCs and reduced tumor growth in vivo using the Huh7 LCSC-transplanted chick embryo chorioallantoic membrane model. Furthermore, EAHDT not only arrested the cell cycle in the G0/G1 phase but also induced receptor-interacting protein kinase 3/mixed-lineage kinase domain-like protein-mediated necroptosis and caspase-dependent apoptosis in Huh7 LCSCs in a concentration-dependent manner. Furthermore, the growth inhibitory effect of EAHDT on Huh7 LCSCs was associated with the downregulation of c-MET-mediated downstream signaling pathways and key cancer stemness markers. Based on these findings, we propose that EAHDT can be used as a new natural drug candidate to prevent and treat HCC by eradicating LCSCs.
Collapse
Affiliation(s)
- Mikyoung Kwon
- Department of Life Science and Biochemical Engineering, Graduate School, Sun Moon University, Asan 31460, Republic of Korea;
| | - Hye Jin Jung
- Department of Life Science and Biochemical Engineering, Graduate School, Sun Moon University, Asan 31460, Republic of Korea;
- Department of Pharmaceutical Engineering and Biotechnology, Sun Moon University, Asan 31460, Republic of Korea
- Genome-Based BioIT Convergence Institute, Sun Moon University, Asan 31460, Republic of Korea
| |
Collapse
|
74
|
Dong Z, Hou X, Wang X, Shen Z, Pang H, Chen L, Yin Z, Ren F, Li W, Ge Y, Ning H, Hu D. Proteomic Analysis of the Mitochondrial Responses in P19 Embryonic Stem Cells Exposed to Florfenicol. TOXICS 2023; 11:992. [PMID: 38133393 PMCID: PMC10747307 DOI: 10.3390/toxics11120992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 11/29/2023] [Accepted: 12/04/2023] [Indexed: 12/23/2023]
Abstract
Florfenicol (FLO) has been shown to elicit diverse toxic effects in plants, insects, and mammals. Previously, our investigations revealed that FLO induced abnormal cardiac development and early embryonic mortality in chicken embryos. However, the effect of FLO on mitochondrial responses in stem cells remains unclear. In this study, we show that FLO significantly diminishes proliferation viability and obstructs the directed differentiation of P19 stem cells (P19SCs) into cardiomyocytes. Proteomic analysis revealed 148 differentially expressed proteins in response to FLO. Functional analysis has pinpointed FLO interference with biological processes associated with oxidative phosphorylation within the mitochondria. In alignment with the results of proteomic analysis, we confirmed that FLO inhibits the expression of both nuclear DNA-encoded and mitochondrial DNA-encoded subunits of the electron transport chain. Subsequent experiments demonstrated that FLO disrupts mitochondrial dynamics and induces the mitochondrial unfolded protein response to maintain mitochondrial homeostasis. These findings collectively highlight the significance of mitochondrial dynamics and the mitochondrial unfolded protein response to mediate the decreased proliferation viability and directed differentiation potential in P19SCs treated with FLO. In conclusion, this study provides a comprehensive overview of mitochondrial responses to FLO-induced cytotoxicity and enhances our understandings of the molecular mechanisms underlying FLO-induced embryonic toxicity.
Collapse
Affiliation(s)
- Zhihua Dong
- College of Animal Science and Technology, Henan Institute of Science and Technology, Xinxiang 453003, China; (Z.D.); (X.H.); (X.W.); (Z.S.); (H.P.); (L.C.); (Z.Y.); (F.R.); (Y.G.)
| | - Xueke Hou
- College of Animal Science and Technology, Henan Institute of Science and Technology, Xinxiang 453003, China; (Z.D.); (X.H.); (X.W.); (Z.S.); (H.P.); (L.C.); (Z.Y.); (F.R.); (Y.G.)
| | - Xueying Wang
- College of Animal Science and Technology, Henan Institute of Science and Technology, Xinxiang 453003, China; (Z.D.); (X.H.); (X.W.); (Z.S.); (H.P.); (L.C.); (Z.Y.); (F.R.); (Y.G.)
| | - Zihui Shen
- College of Animal Science and Technology, Henan Institute of Science and Technology, Xinxiang 453003, China; (Z.D.); (X.H.); (X.W.); (Z.S.); (H.P.); (L.C.); (Z.Y.); (F.R.); (Y.G.)
| | - Huiqing Pang
- College of Animal Science and Technology, Henan Institute of Science and Technology, Xinxiang 453003, China; (Z.D.); (X.H.); (X.W.); (Z.S.); (H.P.); (L.C.); (Z.Y.); (F.R.); (Y.G.)
| | - Lingli Chen
- College of Animal Science and Technology, Henan Institute of Science and Technology, Xinxiang 453003, China; (Z.D.); (X.H.); (X.W.); (Z.S.); (H.P.); (L.C.); (Z.Y.); (F.R.); (Y.G.)
| | - Zhihong Yin
- College of Animal Science and Technology, Henan Institute of Science and Technology, Xinxiang 453003, China; (Z.D.); (X.H.); (X.W.); (Z.S.); (H.P.); (L.C.); (Z.Y.); (F.R.); (Y.G.)
| | - Fei Ren
- College of Animal Science and Technology, Henan Institute of Science and Technology, Xinxiang 453003, China; (Z.D.); (X.H.); (X.W.); (Z.S.); (H.P.); (L.C.); (Z.Y.); (F.R.); (Y.G.)
| | - Weiguo Li
- Postdoctoral Research Station in Biological Sciences, Henan Normal University, Xinxiang 453003, China;
| | - Yaming Ge
- College of Animal Science and Technology, Henan Institute of Science and Technology, Xinxiang 453003, China; (Z.D.); (X.H.); (X.W.); (Z.S.); (H.P.); (L.C.); (Z.Y.); (F.R.); (Y.G.)
| | - Hongmei Ning
- College of Animal Science and Technology, Henan Institute of Science and Technology, Xinxiang 453003, China; (Z.D.); (X.H.); (X.W.); (Z.S.); (H.P.); (L.C.); (Z.Y.); (F.R.); (Y.G.)
| | - Dongfang Hu
- College of Animal Science and Technology, Henan Institute of Science and Technology, Xinxiang 453003, China; (Z.D.); (X.H.); (X.W.); (Z.S.); (H.P.); (L.C.); (Z.Y.); (F.R.); (Y.G.)
- Postdoctoral Research Station in Biological Sciences, Henan Normal University, Xinxiang 453003, China;
- Postdoctoral Research and Development Base, Henan Institute of Science and Technology, Xinxiang 453003, China
| |
Collapse
|
75
|
Feng Q, Hu K, Hu H, Lu Y, Zhang H, Wang G, Zhang Q, Xu Z, Gao X, Jia X, Zhu H, Song D, Yi H, Peng Y, Wu X, Li B, Zhu W, Shi J. Berberine derivative DCZ0358 induce oxidative damage by ROS-mediated JNK signaling in DLBCL cells. Int Immunopharmacol 2023; 125:111139. [PMID: 37913572 DOI: 10.1016/j.intimp.2023.111139] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 10/12/2023] [Accepted: 10/24/2023] [Indexed: 11/03/2023]
Abstract
The most common neoplasm among adult lymphomas is diffuse large B-cell lymphoma (DLBCL), typically characterized by pain-free and progressive lymph node enlargement. Due to high heterogeneity of DLBCL, 30-40 % of patients are resistant to R-CHOP standard chemoimmunotherapy. DCZ0358 is a new compound designed and synthesized from berberine by our group and the molecular mechanism by which it inhibited DLBCL growth has attracted our widespread attention. In this study, we employed the CCK8 assay to reveal that DCZ0358 inhibited proliferation in a dependent manner of time and dosage of DLBCL cells. Moreover, flowcytometry and western blot results showed that DCZ0358 downregulated the expression of CDK4, CDK6 and CyclinD1 to block cell cycle progression in G0/G1 phase. Furthermore, DCZ0358 enhanced mitochondrial membrane potential depolarization, promoted mitochondrial permeability transport pore openness, increased cytoplastic Ca2+ levels and decreased intracellular adenosine triphosphate production, which led to mitochondrial dysfunction. In particular, DCZ0358 treatment triggered cell apoptosis and elevated intracellular reactive oxygen species (ROS) levels, which subsequently mediated JNK pathway activation. Further research indicated the pre-treatment with ROS scavenger N-acetylcysteine (NAC) and JNK inhibitor SP600125 could partially attenuate apoptosis and DNA damage triggered by DCZ0358. Most importantly, DCZ0358 exhibited synergistic anti-tumor effects when combined with etoposide, a common clinical anti-DLBCL drug, both in vitro and certainly in vivo. Above results demonstrated anti-tumor molecular mechanism of DCZ0358 in DLBCL cells and highlighted the ROS/JNK/DNA damage pathway as a potential target in therapies, which have implications for the development of more effective clinical treatments for DLBCL.
Collapse
Affiliation(s)
- Qilin Feng
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Ke Hu
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Huifang Hu
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Yumeng Lu
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Hui Zhang
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Guanli Wang
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Qikai Zhang
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Zhijian Xu
- State Key Laboratory of Drug Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xuejie Gao
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Xinyan Jia
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Huabin Zhu
- Department of Hematology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Dongliang Song
- Department of Hematology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Hongfei Yi
- Department of Hematology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Yu Peng
- Department of Hematology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Xiaosong Wu
- Department of Hematology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Bo Li
- State Key Laboratory of Drug Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.
| | - Weiliang Zhu
- State Key Laboratory of Drug Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.
| | - Jumei Shi
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China.
| |
Collapse
|
76
|
Du Y, Liu L, Yan W, Li Y, Li Y, Cui K, Yu P, Gu Z, Zhang W, Feng J, Li Z, Tang H, Du Y, Zhao H. The anticancer mechanisms of exopolysaccharide from Weissella cibaria D-2 on colorectal cancer via apoptosis induction. Sci Rep 2023; 13:21117. [PMID: 38036594 PMCID: PMC10689803 DOI: 10.1038/s41598-023-47943-7] [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: 09/11/2023] [Accepted: 11/20/2023] [Indexed: 12/02/2023] Open
Abstract
Exopolysaccharide (EPS) from Weissella cibaria has been devoted to the study of food industry. However, the anticancer activity of W. cibaria derived EPS has not yet been investigated. In this study, we obtained the EPS from W. cibaria D-2 isolated from the feces of healthy infants and found that D-2-EPS, a homopolysaccharide with porous web like structure, could effectively inhibit the proliferation, migration, invasion and induce cell cycle arrest in G0/G1 phase of colorectal cancer (CRC) cells. In HT-29 tumor xenografts, D-2-EPS significantly retarded tumor growth without obvious cytotoxicity to normal organs. Furthermore, we revealed that D-2-EPS promoted the apoptosis of CRC cells by increasing the levels of Fas, FasL and activating Caspase-8/Caspase-3, indicating that D-2-EPS might induce apoptosis through the extrinsic Fas/FasL pathway. Taken together, the D-2-EPS has the potential to be developed as a nutraceutical or drug to prevent and treat colorectal cancer.
Collapse
Affiliation(s)
- Yurong Du
- Oncology Department, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Zhengzhou, 450052, Henan, China
| | - Lei Liu
- Oncology Department, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Zhengzhou, 450052, Henan, China
| | - Weiliang Yan
- Oncology Department, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Zhengzhou, 450052, Henan, China
| | - Yang Li
- International Institutes of Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, 322000, Zhejiang, China
| | - Yuanzhe Li
- Department of Pediatrics, Children's Hospital Affiliated of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Kang Cui
- Oncology Department, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Zhengzhou, 450052, Henan, China
| | - Pu Yu
- Oncology Department, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Zhengzhou, 450052, Henan, China
| | - Zhuoyu Gu
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - WanCun Zhang
- Department of Pediatrics, Children's Hospital Affiliated of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Jianguo Feng
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Zhen Li
- National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Heart Center of Henan Provincial People's Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, 451464, Henan, China
| | - Hao Tang
- National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Heart Center of Henan Provincial People's Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, 451464, Henan, China
| | - Yabing Du
- Oncology Department, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Zhengzhou, 450052, Henan, China.
| | - Huan Zhao
- Oncology Department, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Zhengzhou, 450052, Henan, China.
| |
Collapse
|
77
|
Wang J, Mi Y, Sun X, Xue X, Zhao H, Zhang M, Hu B, Bukhari I, Zheng P. Lnc-PTCHD4-AS inhibits gastric cancer through MSH2-MSH6 dimerization and ATM-p53-p21 activation. Aging (Albany NY) 2023; 15:13558-13578. [PMID: 38016120 DOI: 10.18632/aging.205329] [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: 08/25/2023] [Accepted: 11/06/2023] [Indexed: 11/30/2023]
Abstract
Conserved long non-coding RNAs (lncRNAs) have not thoroughly been studied in many cancers, including gastric cancer (GC). We have identified a novel lncRNA PTCHD4-AS which was highly conserved between humans and mice and naturally downregulated in GC cell lines and tissues. Notably, PTCHD4-AS was found to be transcriptionally induced by DNA damage agents and its upregulation led to cell cycle arrest at the G2/M phase, in parallel, it facilitated the cell apoptosis induced by cisplatin (CDDP) in GC. Mechanistically, PTCHD4-AS directly bound to the DNA mismatch repair protein MSH2-MSH6 dimer, and facilitated the binding of dimer to ATM, thereby promoting the expression of phosphorylated ATM, p53 and p21. Here we conclude that the upregulation of PTCHD4-AS inhibits proliferation and increases CDDP sensitivity of GC cells via binding with MSH2-MSH6 dimer, activating the ATM-p53-p21 pathway.
Collapse
Affiliation(s)
- Jingyun Wang
- Henan Key Laboratory for Helicobacter pylori and Microbiota and GI Cancer, Marshall Medical Research Center, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
- Academy of Medical Science, Zhengzhou University, Zhengzhou 450000, China
| | - Yang Mi
- Henan Key Laboratory for Helicobacter pylori and Microbiota and GI Cancer, Marshall Medical Research Center, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
- Academy of Medical Science, Zhengzhou University, Zhengzhou 450000, China
- Department of Gastroenterology, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
| | - Xiangdong Sun
- Henan Key Laboratory for Helicobacter pylori and Microbiota and GI Cancer, Marshall Medical Research Center, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
- Academy of Medical Science, Zhengzhou University, Zhengzhou 450000, China
| | - Xia Xue
- Henan Key Laboratory for Helicobacter pylori and Microbiota and GI Cancer, Marshall Medical Research Center, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
| | - Huanjie Zhao
- Henan Key Laboratory for Helicobacter pylori and Microbiota and GI Cancer, Marshall Medical Research Center, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
- Academy of Medical Science, Zhengzhou University, Zhengzhou 450000, China
| | - Mengfei Zhang
- Henan Key Laboratory for Helicobacter pylori and Microbiota and GI Cancer, Marshall Medical Research Center, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
- Department of Gastroenterology, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
| | - Baitong Hu
- Henan Key Laboratory for Helicobacter pylori and Microbiota and GI Cancer, Marshall Medical Research Center, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
- Academy of Medical Science, Zhengzhou University, Zhengzhou 450000, China
| | - Ihtisham Bukhari
- Henan Key Laboratory for Helicobacter pylori and Microbiota and GI Cancer, Marshall Medical Research Center, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
- Department of Gastroenterology, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
| | - Pengyuan Zheng
- Henan Key Laboratory for Helicobacter pylori and Microbiota and GI Cancer, Marshall Medical Research Center, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
- Academy of Medical Science, Zhengzhou University, Zhengzhou 450000, China
- Department of Gastroenterology, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
| |
Collapse
|
78
|
Barghi Lish A, Foroumadi A, Kolvari E, Safari F. Synthesis and Biological Evaluation of 12-Aryl-11-hydroxy-5,6-dihydropyrrolo[2″,1″:3',4']pyrazino[1',2':1,5]pyrrolo[2,3- d]pyridazine-8(9 H)-one Derivatives as Potential Cytotoxic Agents. ACS OMEGA 2023; 8:42212-42224. [PMID: 38024677 PMCID: PMC10653054 DOI: 10.1021/acsomega.3c04167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 10/11/2023] [Accepted: 10/16/2023] [Indexed: 12/01/2023]
Abstract
In the present paper, a facile and efficient synthetic procedure has been applied to obtain dihydrodipyrrolo[1,2-a:2',1'-c]pyrazine-2,3-dicarboxylates (5a-s), which have subsequently gone through the cyclization in the presence of hydrazine hydrate to afford 12-aryl-11-hydroxy-5,6-dihydropyrrolo[2″,1″:3',4']pyrazino[1',2':1,5]pyrrolo[2,3-d]pyridazine-8(9H)-ones (7a-q). The molecular structures of these novel compounds were extensively examined through the analysis of spectroscopic data in combination with X-ray crystallography techniques. Following that, the in vitro cytotoxic activities of all derivatives against three human cancer cell lines (Panc-1, PC3, and MDA-MB-231) were comprehensively evaluated alongside the assessment on normal human dermal fibroblast (HDF) cells using the MTT assay. Among the compounds, the 3-nitrophenyl derivative (7m) from the second series showed the best antiproliferative activity against all tested cell lines, particularly against Panc-1 cell line, (IC50 = 12.54 μM), being nearly twice as potent as the standard drug etoposide. The induction of apoptosis and sub-G1 cell cycle arrest in Panc-1 cancer cells by compound 7m was confirmed through further assessment. Moreover, the inhibition of kinases and the induction of cellular apoptosis by compound 7m in Panc-1 cancer cells were validated using the Western blotting assay.
Collapse
Affiliation(s)
- Azam Barghi Lish
- Department
of Chemistry, Semnan University, Semnan 35351-19111, Iran
| | - Alireza Foroumadi
- Department
of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran1417614411, Iran
- Drug
Design and Development Research Center, The Institute of Pharmaceutical
Sciences (TIPS), Tehran University of Medical
Sciences, Tehran 1417614411, Iran
| | - Eskandar Kolvari
- Department
of Chemistry, Semnan University, Semnan 35351-19111, Iran
| | - Fatemeh Safari
- Department
of Biology, Faculty of Science, University
of Guilan, Rasht 4193833697, Iran
| |
Collapse
|
79
|
Lee HM, Wright WC, Pan M, Low J, Currier D, Fang J, Singh S, Nance S, Delahunty I, Kim Y, Chapple RH, Zhang Y, Liu X, Steele JA, Qi J, Pruett-Miller SM, Easton J, Chen T, Yang J, Durbin AD, Geeleher P. A CRISPR-drug perturbational map for identifying compounds to combine with commonly used chemotherapeutics. Nat Commun 2023; 14:7332. [PMID: 37957169 PMCID: PMC10643606 DOI: 10.1038/s41467-023-43134-0] [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: 07/10/2023] [Accepted: 11/01/2023] [Indexed: 11/15/2023] Open
Abstract
Combination chemotherapy is crucial for successfully treating cancer. However, the enormous number of possible drug combinations means discovering safe and effective combinations remains a significant challenge. To improve this process, we conduct large-scale targeted CRISPR knockout screens in drug-treated cells, creating a genetic map of druggable genes that sensitize cells to commonly used chemotherapeutics. We prioritize neuroblastoma, the most common extracranial pediatric solid tumor, where ~50% of high-risk patients do not survive. Our screen examines all druggable gene knockouts in 18 cell lines (10 neuroblastoma, 8 others) treated with 8 widely used drugs, resulting in 94,320 unique combination-cell line perturbations, which is comparable to the largest existing drug combination screens. Using dense drug-drug rescreening, we find that the top CRISPR-nominated drug combinations are more synergistic than standard-of-care combinations, suggesting existing combinations could be improved. As proof of principle, we discover that inhibition of PRKDC, a component of the non-homologous end-joining pathway, sensitizes high-risk neuroblastoma cells to the standard-of-care drug doxorubicin in vitro and in vivo using patient-derived xenograft (PDX) models. Our findings provide a valuable resource and demonstrate the feasibility of using targeted CRISPR knockout to discover combinations with common chemotherapeutics, a methodology with application across all cancers.
Collapse
Affiliation(s)
- Hyeong-Min Lee
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - William C Wright
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Min Pan
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Jonathan Low
- Department of Chemical Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Duane Currier
- Department of Chemical Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Jie Fang
- Department of Surgery, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Shivendra Singh
- Department of Surgery, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Stephanie Nance
- Division of Molecular Oncology, Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Ian Delahunty
- Division of Molecular Oncology, Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Yuna Kim
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Richard H Chapple
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Yinwen Zhang
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Xueying Liu
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Jacob A Steele
- Center for Advanced Genome Engineering, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Jun Qi
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Shondra M Pruett-Miller
- Center for Advanced Genome Engineering, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - John Easton
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Taosheng Chen
- Department of Chemical Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Jun Yang
- Department of Surgery, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA.
- Department of Pathology and Laboratory Medicine, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, 38163, USA.
| | - Adam D Durbin
- Division of Molecular Oncology, Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA.
| | - Paul Geeleher
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA.
| |
Collapse
|
80
|
Ning J, Ding J, Wang S, Jiang Y, Wang D, Jiang S. GPC3 Promotes Lung Squamous Cell Carcinoma Progression and HLA-A2-Restricted GPC3 Antigenic Peptide-Modified Dendritic Cell-Induced Cytotoxic T Lymphocytes to Kill Lung Squamous Cell Carcinoma Cells. J Immunol Res 2023; 2023:5532617. [PMID: 37965271 PMCID: PMC10643027 DOI: 10.1155/2023/5532617] [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: 05/26/2023] [Revised: 09/01/2023] [Accepted: 09/29/2023] [Indexed: 11/16/2023] Open
Abstract
Lung squamous cell carcinoma (LUSC) is associated with poor clinical prognosis and lacks available targeted agents. GPC3 is upregulated in LUSC. Our study aimed to explore the roles of GPC3 in LUSC and the antitumor effects of HLA-A2-restricted GPC3 antigenic peptide-sensitized dendritic cell (DC)-induced cytotoxic T lymphocytes (CTLs) on LUSC. LUSC cells with GPC3 knockdown and overexpression were built using lentivirus packaging, and cell viability, clone formation, apoptosis, cycle, migration, and invasion were determined. Western blotting was used to detect the expression of cell cycle-related proteins and PI3K-AKT pathway-associated proteins. Subsequently, HLA-A2-restricted GPC3 antigenic peptides were predicted and synthesized by bioinformatic databases, and DCs were induced and cultured in vitro. Finally, HLA-A2-restricted GPC3 antigenic peptide-modified DCs were co-cultured with T cells to generate specific CTLs, and the killing effects of different CTLs on LUSC cells were studied. A series of cell function experiments showed that GPC3 overexpression promoted the proliferation, migration, and invasion of LUSC cells, inhibited their apoptosis, increased the number of cells in S phase, and reduced the cells in G2/M phase. GPC3 knockdown downregulated cyclin A, c-Myc, and PI3K, upregulated E2F1, and decreased the pAKT/AKT level. Three HLA-A2-restricted GPC3 antigenic peptides were synthesized, with GPC3522-530 FLAELAYDL and GPC3102-110 FLIIQNAAV antigenic peptide-modified DCs inducing CTL production, and exhibiting strong targeted killing ability in LUSC cells at 80 : 1 multiplicity of infection. GPC3 may advance the onset and progression of LUSC, and GPC3522-530 FLAELAYDL and GPC3102-110 FLIIQNAAV antigenic peptide-loaded DC-induced CTLs have a superior killing ability against LUSC cells.
Collapse
Affiliation(s)
- Jing Ning
- Department of General Medicine, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang 110042, China
- Molecular Oncology Department of Cancer Research Institution, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
| | - Jianqiao Ding
- Department of Thoracic Surgery (2), Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang 110042, China
| | - Shu Wang
- Department of Radiotherapy, The Second Hospital of Jilin University, Changchun 130000, China
| | - Youhong Jiang
- Molecular Oncology Department of Cancer Research Institution, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
| | - Daqing Wang
- Hope Plaza Children's Hospital District of Dalian Municipal Women and Children's Medical Center, Dalian 116000, China
| | - Shenyi Jiang
- Department of General Practice, The First Affiliated Hospital of China Medical University, Shengyang 110001, China
| |
Collapse
|
81
|
Łopuszyński W, Brzana A, Szczubiał M, Bulak K, Śmiech A. Topoisomerase IIα immunoexpression in feline mammary carcinomas: A correlation with Ki67 immunoexpression and the mitotic count. Res Vet Sci 2023; 164:104992. [PMID: 37657395 DOI: 10.1016/j.rvsc.2023.104992] [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: 05/25/2023] [Revised: 08/07/2023] [Accepted: 08/23/2023] [Indexed: 09/03/2023]
Abstract
The aim of the study was to compare the immunohistochemical expression of topoisomerase IIα protein (Topo IIα) with Ki67 expression and mitotic count in feline mammary carcinomas (FMCs). Topo IIα is considered as a proliferation indicator as well as a molecular target of anthracycline chemotherapy. The studied material included 70 FMCs from female cats treated with mastectomy. Primary mouse monoclonal antibodies directed against Topo IIα and Ki67 were used in immunohistochemical reactions. The number of mitotic figures was counted at 400× magnification in a field of 2.37 mm2. Immunohistochemical reaction for Topo IIα occurred in cell nuclei. The Topo IIα index ranged from 6.12% to 54.60% and was positively correlated with the values of the Ki67 index (r = 0.7193) and the mitotic count (r = 0. 2858). This indicates the potential possibility of use of the immunohistochemical expression of Topo IIα to assess the rate of proliferation in FMCs. The wide range of expression of Topo IIα in individual tumorus found in the conducted studies allows us to hypothesize that its assessment could be used as a predictive marker in chemotherapy of FMCs with the use of anthracyclines. However, this requires confirmation in clinical trials.
Collapse
Affiliation(s)
- Wojciech Łopuszyński
- Department of Pathomorphology and Forensic Veterinary Medicine, University of Life Sciences, Lublin, Poland.
| | - Adam Brzana
- Regional Veterinary Inspectorate in Opole, Regional Veterinary Laboratory, Opole, Poland
| | - Marek Szczubiał
- Department and Clinic of Animal Reproduction, University of Life Sciences, Lublin, Poland
| | - Kamila Bulak
- Department of Pathomorphology and Forensic Veterinary Medicine, University of Life Sciences, Lublin, Poland
| | - Anna Śmiech
- Department of Pathomorphology and Forensic Veterinary Medicine, University of Life Sciences, Lublin, Poland
| |
Collapse
|
82
|
Le TTH, Lei M, Hoang PH, Hoang VH, Pham VK, Nguyen PH. Anti-cancer activity of Marsdenialongise A, a new C21 steroidal glycoside isolated from Marsdenia longipes W.T. Wang (Apocynaceae). Steroids 2023; 199:109310. [PMID: 37714250 DOI: 10.1016/j.steroids.2023.109310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 09/09/2023] [Accepted: 09/12/2023] [Indexed: 09/17/2023]
Abstract
C21 steroidal glycosides are a group of natural compounds with biological activities such as anti-cancer, anti-microbial, and anti-viral properties. In this study, we isolated and determined the structure of a new C21 steroidal glycoside, named Marsdenialongise A from Marsdenia longipes W.T. Wang, using nuclear magnetic resonance spectroscopy and mass spectra data. Marsdenialongise A is a derivative of tenacigenin B and was isolated for the first time from a plant. The inhibitory effect of Marsdenialongise A on cancer cells was evaluated through MTT and cell migration assays, cell cycle, and apoptosis analyses. The results of the MTT assay showed that Marsdenialongise A reduces the cell viability of cancer cells, with the AGS cell line being more sensitive than other cell lines, with an IC50 value of 5.69 µM (for 48 h of treatment). Marsdenialongise A also exhibited an ability to prevent the migration of cancer cells in AGS cells. Further analysis using flow cytometry has revealed that Marsdenialongise A is capable of inducing cell cycle arrest and apoptosis. The overexpression of reactive oxygen species (ROS) production induced by Marsdenialongise A can be considered a cause that leads to the influence on the cell cycle and apoptosis of cancer cells. Thus, Marsdenialongise A can be considered a potential anti-cancer agent.
Collapse
Affiliation(s)
- Thi Thanh Huong Le
- Thai Nguyen University of Sciences (TNUS), Thai Nguyen University, Thai Nguyen City, Viet Nam
| | - Ma Lei
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, PR China
| | - Phu Hiep Hoang
- Thai Nguyen University of Education (TNUE), Thai Nguyen University, Thai Nguyen City, Viet Nam
| | - Van Hung Hoang
- Center for Interdisciplinary Science and Education, Thai Nguyen University, Thai Nguyen City, Viet Nam
| | - Van Khang Pham
- Thai Nguyen University of Education (TNUE), Thai Nguyen University, Thai Nguyen City, Viet Nam.
| | - Phu Hung Nguyen
- Thai Nguyen University of Sciences (TNUS), Thai Nguyen University, Thai Nguyen City, Viet Nam; Center for Interdisciplinary Science and Education, Thai Nguyen University, Thai Nguyen City, Viet Nam.
| |
Collapse
|
83
|
García-Reyes B, Kuzmanov I, Schneider R, Schneiker B, Efferz P, Kalff JC, Wehner S. Glial cell-derived soluble factors increase the metastatic potential of pancreatic adenocarcinoma cells and induce epithelial-to-mesenchymal transition. J Cancer Res Clin Oncol 2023; 149:14315-14327. [PMID: 37572121 PMCID: PMC10590291 DOI: 10.1007/s00432-023-05133-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: 06/26/2023] [Accepted: 07/04/2023] [Indexed: 08/14/2023]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive types of cancer, characterized by the spreading of highly metastatic cancer cells, including invasion into surrounding nerves and perineural spaces. Nerves, in turn, can invade the tumor tissue and, through the secretion of neurotrophic factors, chemokines, and cytokines, contribute to PDAC progression. However, the contribution of the nerve-associated glial cells to PDAC progression is not well characterized. METHODS Two murine PDAC cell lines were cultured with the conditioned media (CM) of primary enteric glial cells or IMS32 Schwann cells (SCs). Different properties of PDAC cells, such as invasiveness, migratory capacity, and resistance to gemcitabine, were measured by RT-qPCR, microscopy, and MTT assays. Using a neuronal cell line, the observed effects were confirmed to be specific to the glial lineage. RESULTS Compared to the control medium, PDAC cells in the glial cell-conditioned medium showed increased invasiveness and migratory capacity. These cells showed reduced E-cadherin and increased N-cadherin and Vimentin levels, all markers of epithelial-mesenchymal transition (EMT). Primary enteric glial cell CM inhibited the proliferation of PDAC cells but preserved their viability, upregulated transcription factor Snail, and increased their resistance to gemcitabine. The conditioned medium generated from the IMS32 SCs produced comparable results. CONCLUSION Our data suggest that glial cells can increase the metastatic potential of PDAC cells by increasing their migratory capacity and inducing epithelial-to-mesenchymal transition, a re-programming that many solid tumors use to undergo metastasis. Glial cell-conditioned medium also increased the chemoresistance of PDAC cells. These findings may have implications for future therapeutic strategies, such as targeting glial cell-derived factor signaling in PDAC.
Collapse
Affiliation(s)
- Balbina García-Reyes
- Department of Surgery, Medical Faculty, University Hospital Bonn, Bonn, Germany
- Mildred Scheel School of Oncology, Aachen Bonn Cologne Düsseldorf (MSSO ABCD), Medical Faculty, University Hospital Bonn, Bonn, Germany
| | - Ivan Kuzmanov
- Department of Surgery, Medical Faculty, University Hospital Bonn, Bonn, Germany
| | - Reiner Schneider
- Department of Surgery, Medical Faculty, University Hospital Bonn, Bonn, Germany
| | - Bianca Schneiker
- Department of Surgery, Medical Faculty, University Hospital Bonn, Bonn, Germany
| | - Patrik Efferz
- Department of Surgery, Medical Faculty, University Hospital Bonn, Bonn, Germany
| | - Jörg C Kalff
- Department of Surgery, Medical Faculty, University Hospital Bonn, Bonn, Germany
| | - Sven Wehner
- Department of Surgery, Medical Faculty, University Hospital Bonn, Bonn, Germany.
| |
Collapse
|
84
|
Kim MJ, Kim HS, Kang HW, Lee DE, Hong WC, Kim JH, Kim M, Cheong JH, Kim HJ, Park JS. SLC38A5 Modulates Ferroptosis to Overcome Gemcitabine Resistance in Pancreatic Cancer. Cells 2023; 12:2509. [PMID: 37887353 PMCID: PMC10605569 DOI: 10.3390/cells12202509] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 10/20/2023] [Accepted: 10/22/2023] [Indexed: 10/28/2023] Open
Abstract
Pancreatic cancer is characterized by a poor prognosis, with its five-year survival rate lower than that of any other cancer type. Gemcitabine, a standard treatment for pancreatic cancer, often has poor outcomes for patients as a result of chemoresistance. Therefore, novel therapeutic targets must be identified to overcome gemcitabine resistance. Here, we found that SLC38A5, a glutamine transporter, is more highly overexpressed in gemcitabine-resistant patients than in gemcitabine-sensitive patients. Furthermore, the deletion of SLC38A5 decreased the proliferation and migration of gemcitabine-resistant PDAC cells. We also found that the inhibition of SLC38A5 triggered the ferroptosis signaling pathway via RNA sequencing. Also, silencing SLC38A5 induced mitochondrial dysfunction and reduced glutamine uptake and glutathione (GSH) levels, and downregulated the expressions of GSH-related genes NRF2 and GPX4. The blockade of glutamine uptake negatively modulated the mTOR-SREBP1-SCD1 signaling pathway. Therefore, suppression of SLC38A5 triggers ferroptosis via two pathways that regulate lipid ROS levels. Similarly, we observed that knockdown of SLC38A5 restored gemcitabine sensitivity by hindering tumor growth and metastasis in the orthotopic mouse model. Altogether, our results demonstrate that SLC38A5 could be a novel target to overcome gemcitabine resistance in PDAC therapy.
Collapse
Affiliation(s)
- Myeong Jin Kim
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06229, Republic of Korea; (M.J.K.); (H.S.K.); (H.W.K.); (D.E.L.); (W.C.H.); (J.H.K.); (M.K.)
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul 03722, Republic of Korea;
| | - Hyung Sun Kim
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06229, Republic of Korea; (M.J.K.); (H.S.K.); (H.W.K.); (D.E.L.); (W.C.H.); (J.H.K.); (M.K.)
| | - Hyeon Woong Kang
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06229, Republic of Korea; (M.J.K.); (H.S.K.); (H.W.K.); (D.E.L.); (W.C.H.); (J.H.K.); (M.K.)
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul 03722, Republic of Korea;
| | - Da Eun Lee
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06229, Republic of Korea; (M.J.K.); (H.S.K.); (H.W.K.); (D.E.L.); (W.C.H.); (J.H.K.); (M.K.)
| | - Woosol Chris Hong
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06229, Republic of Korea; (M.J.K.); (H.S.K.); (H.W.K.); (D.E.L.); (W.C.H.); (J.H.K.); (M.K.)
- Yonsei University College of Medicine, Seoul 06229, Republic of Korea
| | - Ju Hyun Kim
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06229, Republic of Korea; (M.J.K.); (H.S.K.); (H.W.K.); (D.E.L.); (W.C.H.); (J.H.K.); (M.K.)
- Yonsei University College of Medicine, Seoul 06229, Republic of Korea
| | - Minsoo Kim
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06229, Republic of Korea; (M.J.K.); (H.S.K.); (H.W.K.); (D.E.L.); (W.C.H.); (J.H.K.); (M.K.)
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul 03722, Republic of Korea;
| | - Jae-Ho Cheong
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul 03722, Republic of Korea;
- Department of Surgery, Yonsei University College of Medicine, Seoul 06229, Republic of Korea
| | - Hyo Jung Kim
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06229, Republic of Korea; (M.J.K.); (H.S.K.); (H.W.K.); (D.E.L.); (W.C.H.); (J.H.K.); (M.K.)
| | - Joon Seong Park
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06229, Republic of Korea; (M.J.K.); (H.S.K.); (H.W.K.); (D.E.L.); (W.C.H.); (J.H.K.); (M.K.)
| |
Collapse
|
85
|
Islam F, Nath N, Zehravi M, Khan J, Jashim SBT, Charde MS, Chakole RD, Kumar KP, Babu AK, Nainu F, Khan SL, Rab SO, Emran TB, Wilairatana P. Exploring the role of natural bioactive molecules in genitourinary cancers: how far has research progressed? NATURAL PRODUCTS AND BIOPROSPECTING 2023; 13:39. [PMID: 37843642 PMCID: PMC10579213 DOI: 10.1007/s13659-023-00400-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Accepted: 09/17/2023] [Indexed: 10/17/2023]
Abstract
The primary approaches to treat cancerous diseases include drug treatment, surgical procedures, biotherapy, and radiation therapy. Chemotherapy has been the primary treatment for cancer for a long time, but its main drawback is that it kills cancerous cells along with healthy ones, leading to deadly adverse health effects. However, genitourinary cancer has become a concern in recent years as it is more common in middle-aged people. So, researchers are trying to find possible therapeutic options from natural small molecules due to the many drawbacks associated with chemotherapy and other radiation-based therapies. Plenty of research was conducted regarding genitourinary cancer to determine the promising role of natural small molecules. So, this review focused on natural small molecules along with their potential therapeutic targets in the case of genitourinary cancers such as prostate cancer, renal cancer, bladder cancer, testicular cancer, and so on. Also, this review states some ongoing or completed clinical evidence in this regard.
Collapse
Affiliation(s)
- Fahadul Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, 1207, Bangladesh
| | - Nikhil Nath
- Department of Pharmacy, International Islamic University Chittagong, Kumira, Chittagong, 4318, Bangladesh
| | - Mehrukh Zehravi
- Department of Clinical Pharmacy, College of Dentistry & Pharmacy, Buraydah Private Colleges, Buraydah, 51418, Kingdom of Saudi Arabia.
| | - Jishan Khan
- Department of Pharmacy, International Islamic University Chittagong, Kumira, Chittagong, 4318, Bangladesh
| | - Sumiya Ben-Ta Jashim
- Department of Pharmacy, International Islamic University Chittagong, Kumira, Chittagong, 4318, Bangladesh
| | - Manoj Shrawan Charde
- Government College of Pharmacy, Vidyanagar, Karad, Satara, 415124, Maharashtra, India
| | - Rita Dadarao Chakole
- Government College of Pharmacy, Vidyanagar, Karad, Satara, 415124, Maharashtra, India
| | - K Praveen Kumar
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Govt. of NCT of Delhi, Delhi Pharmaceutical Sciences and Research University (DPSRU), Mehrauli-Badarpur Road, PushpVihar, Sector 3, New Delhi, 110017, India
| | - A Kishore Babu
- Ratnadeep College of Pharmacy, Ratnapur, Jamkhed, Ahmednagar, 413206, Maharashtra, India
| | - Firzan Nainu
- Department of Pharmacy, Faculty of Pharmacy, Hasanuddin University, Makassar, 90245, Indonesia
| | - Sharuk L Khan
- Department of Pharmaceutical Chemistry, N.B.S. Institute of Pharmacy, Ausa, 413520, Maharashtra, India
| | - Safia Obaidur Rab
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Talha Bin Emran
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, 1207, Bangladesh.
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School & Legorreta Cancer Center, Brown University, Providence, RI, 02912, USA.
| | - Polrat Wilairatana
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, 10400, Thailand.
| |
Collapse
|
86
|
Tulk A, Watson R, Erdrich J. The Influence of Statin Use on Chemotherapeutic Efficacy in Studies of Mouse Models: A Systematic Review. Anticancer Res 2023; 43:4263-4275. [PMID: 37772570 PMCID: PMC10637576 DOI: 10.21873/anticanres.16621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 08/08/2023] [Accepted: 08/10/2023] [Indexed: 09/30/2023]
Abstract
BACKGROUND/AIM Using statins as antitumor agents is an approach to cancer therapy that has been explored extensively in specific cancer types. Reframing the query to how a statin interacts with the treatment regimen instead might provide new insight. Given that cell-cycle regulation influences tumorigenesis, it is possible that the cell-cycle phase which a given chemotherapy acts on influences the synergistic effects with adjuvant statin use. In this review, we outline the effect of statins in combination with chemotherapeutic drugs in in vivo animal model studies based on the class of chemotherapy and its relation to the cell cycle. MATERIALS AND METHODS This systematic review was conducted using the Preferred Reporting Items for Systematic reviews and Meta-Analyses for Protocols 2015 with 23 articles deemed eligible to be included. RESULTS Our review suggests that statins influence the success of chemotherapy treatments. Furthermore, enhanced efficacy was demonstrated with chemotherapeutic drugs that act at every phase of the cell cycle. CONCLUSION This type of compilation departs from the norm of describing statin influence on named cancer subtypes and instead catalogs how statins interact with categorical chemotherapy agents which might be beneficial for broader therapeutic decision-making across cancer subtypes, possibly contributing to pharmaceutical development, and thereby helping to maximize patient outcomes.
Collapse
Affiliation(s)
- Angela Tulk
- Department of Surgery, University of Arizona College of Medicine, Tucson, AZ, U.S.A.;
| | - Raj Watson
- A.T. Still University-Kirksville College of Osteopathic Medicine, Kirksville, MO, U.S.A
| | - Jennifer Erdrich
- Department of Surgery, University of Arizona College of Medicine, Tucson, AZ, U.S.A
| |
Collapse
|
87
|
Miller AI, Diaz D, Lin B, Krzesaj PK, Ustoyev S, Shim A, Fine EJ, Sarafraz-Yazdi E, Pincus MR, Feinman RD. Ketone Bodies Induce Unique Inhibition of Tumor Cell Proliferation and Enhance the Efficacy of Anti-Cancer Agents. Biomedicines 2023; 11:2515. [PMID: 37760956 PMCID: PMC10526402 DOI: 10.3390/biomedicines11092515] [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: 06/25/2023] [Revised: 08/30/2023] [Accepted: 09/06/2023] [Indexed: 09/29/2023] Open
Abstract
The ketone bodies, sodium and lithium salts of acetoacetate (AcAc) and sodium 3-hydroxybutyrate (3-HB; commonly called beta-hydroxybutyrate) have been found to inhibit the proliferation of cancer cells. Previous studies have suggested that lithium itself may be an inhibiting agent but may be additive or synergistic with the effect of AcAc. We previously found that sodium acetoacetate (NaAcAc) inhibits the growth of human colon cancer cell line SW480. We report here similar results for several other cancer cell lines including ovarian, cervical and breast cancers. We found that NaAcAc does not kill cancer cells but rather blocks their proliferation. Similar inhibition of growth was seen in the effect of lithium ion alone (as LiCl). The effect of LiAcAc appears to be due to the combined effects of acetoacetate and the lithium ion. The ketone bodies, when given together with chemotherapeutic agents, rapamycin, methotrexate and the new peptide anti-cancer agent, PNC-27, substantially lowers their IC50 values for cancer cell, killing suggesting that ketone bodies and ketogenic diets may be powerful adjunct agents in treating human cancers.
Collapse
Affiliation(s)
- Anna I. Miller
- Department of Cell Biology, SUNY Downstate Health Sciences University, 450 Clarkson Avenue, Brooklyn, NY 11203, USA
| | - David Diaz
- Department of Cell Biology, SUNY Downstate Health Sciences University, 450 Clarkson Avenue, Brooklyn, NY 11203, USA
- Department of Pathology, SUNY Downstate Health Sciences University, 450 Clarkson Avenue, Brooklyn, NY 11203, USA
| | - Bo Lin
- Department of Pathology, AdventHealth, 301 Memorial Medical Pkwy, Daytona Beach, FL 32117, USA
| | - Patryk K. Krzesaj
- Department of Cell Biology, SUNY Downstate Health Sciences University, 450 Clarkson Avenue, Brooklyn, NY 11203, USA
- Department of Pathology, SUNY Downstate Health Sciences University, 450 Clarkson Avenue, Brooklyn, NY 11203, USA
| | - Sarah Ustoyev
- Department of Cell Biology, SUNY Downstate Health Sciences University, 450 Clarkson Avenue, Brooklyn, NY 11203, USA
- Department of Pathology, SUNY Downstate Health Sciences University, 450 Clarkson Avenue, Brooklyn, NY 11203, USA
| | - Alfred Shim
- Department of Cell Biology, SUNY Downstate Health Sciences University, 450 Clarkson Avenue, Brooklyn, NY 11203, USA
- Department of Pathology, SUNY Downstate Health Sciences University, 450 Clarkson Avenue, Brooklyn, NY 11203, USA
| | - Eugene J. Fine
- Department of Radiology (Nuclear Medicine), Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Ehsan Sarafraz-Yazdi
- NomoCan Pharmaceuticals LLC, New York Blood Center, 310 East 67th Street, New York, NY 10065, USA
| | - Matthew R. Pincus
- Department of Pathology, SUNY Downstate Health Sciences University, 450 Clarkson Avenue, Brooklyn, NY 11203, USA
| | - Richard D. Feinman
- Department of Cell Biology, SUNY Downstate Health Sciences University, 450 Clarkson Avenue, Brooklyn, NY 11203, USA
| |
Collapse
|
88
|
Choi HK, Park SH, Lee J, Hwang JT. Review of Patents for Anticancer Agents Targeting Adenosine Monophosphate-Activated Protein Kinase. J Med Food 2023; 26:605-615. [PMID: 37590001 DOI: 10.1089/jmf.2023.k.0026] [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: 08/18/2023] Open
Abstract
Cancer, caused by abnormal and excessive cellular proliferation, can invade and destroy surrounding tissues and organs through the spreading of cancer cells. A general strategy for developing anticancer agents is to identify biomarkers that, if targeted, can produce a robust cytotoxic effect with minimal side effects. Cell-cycle regulators, checkpoint regulatory genes, and apoptosis-related genes are well-known biomarkers that inhibit cancer cell proliferation. Several compounds that target such biomarkers have been patented and more are being developed as novel therapies. Recent additions to this list include anticancer drugs that target signaling pathway proteins, such as 5' adenosine monophosphate-activated protein kinase (AMPK), which plays a vital role in cancer and normal cell metabolism. Herein, we have reviewed recent patents related to AMPK-targeting anticancer drugs and discussed the mechanisms of action of these drugs. We conclude that these recently published patents include several attractive compounds and methods for targeting AMPK. Further research and clinical trials are required to elucidate the comprehensive role of AMPK in cancer cell metabolism, identify its associated signal transduction systems, and develop novel activators that may find applications in cancer therapy. Clinical Trial Registration number: NCT01904123.
Collapse
Affiliation(s)
- Hyo-Kyoung Choi
- Food Functionality Research Division, Korea Food Research Institute, Jeollabuk-do, Republic of Korea
| | - Soo-Hyun Park
- Food Functionality Research Division, Korea Food Research Institute, Jeollabuk-do, Republic of Korea
| | - Jangho Lee
- Food Functionality Research Division, Korea Food Research Institute, Jeollabuk-do, Republic of Korea
| | - Jin-Taek Hwang
- Food Functionality Research Division, Korea Food Research Institute, Jeollabuk-do, Republic of Korea
| |
Collapse
|
89
|
He M, He Y, Xu J, Zhang Y, Cao X, Wang L, Luo F. Upregulated FADD is associated with poor prognosis, immune exhaustion, tumor malignancy, and immunotherapy resistance in patients with lung adenocarcinoma. Front Oncol 2023; 13:1228889. [PMID: 37671047 PMCID: PMC10476093 DOI: 10.3389/fonc.2023.1228889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 07/24/2023] [Indexed: 09/07/2023] Open
Abstract
Background FAS-associated death structural domain (FADD) proteins are important proteins that regulate apoptosis and are also involved in many nonapoptotic pathways in tumors. However, how dysregulated FADD affects the development of lung adenocarcinoma (LUAD) remains unknown. Method Transcriptome profiles and corresponding clinical information of LUAD patients were convened from different databases, and the results were validated by qRT-PCR and cell counting kit-8 using LUAD cell lines. Potential associations between FADD and tumor malignancy, the immune microenvironment, genomic stability, and treatment sensitivity in LUAD patients were revealed by systematic bioinformatics analysis. Results FADD was significantly overexpressed in LUAD, and patients with higher expression levels of FADD had a worse prognosis and more advanced tumor stage. Functional analysis revealed that elevated expression of FADD was associated with cell cycle dysregulation, angiogenesis, and metabolic disturbances. In addition, overexpression of FADD was associated with a higher infiltration of suppressive immune cells. From a single-cell perspective, cells with lower FADD expression are more active in immune-related pathways. FADD was associated with more genomic mutations, especially TP53. Patients with high FADD expression are more likely to benefit from conventional chemotherapy, while those with low FADD expression are more suitable for immunotherapy. Conclusions Upregulated FADD is associated with worse prognosis, immune exhaustion, and tumor malignancy in LUAD patients. In addition, FADD can be an efficient indicator for assessing sensitivity to chemotherapy and immunotherapy. Therefore, FADD has the potential to serve as a new target for precision medicine and targeted therapy for LUAD.
Collapse
Affiliation(s)
- Miao He
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Department of Oncology, Deyang People’s Hospital, Chengdu, China
| | - Yingying He
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Department of Oncology, Deyang People’s Hospital, Chengdu, China
| | - Jian Xu
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Department of Oncology, Deyang People’s Hospital, Chengdu, China
| | - Youcai Zhang
- Department of Oncology, Deyang People’s Hospital, Chengdu, China
| | - Xiaoyu Cao
- Department of Oncology, Deyang People’s Hospital, Chengdu, China
| | - Li Wang
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Feng Luo
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
90
|
Salmani-Javan E, Jafari-Gharabaghlou D, Bonabi E, Zarghami N. Fabricating niosomal-PEG nanoparticles co-loaded with metformin and silibinin for effective treatment of human lung cancer cells. Front Oncol 2023; 13:1193708. [PMID: 37664043 PMCID: PMC10471189 DOI: 10.3389/fonc.2023.1193708] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 07/13/2023] [Indexed: 09/05/2023] Open
Abstract
Background Despite current therapies, lung cancer remains a global issue and requires the creation of novel treatment methods. Recent research has shown that biguanides such as metformin (MET) and silibinin (SIL) have a potential anticancer effect. As a consequence, the effectiveness of MET and SIL in combination against lung cancer cells was investigated in this study to develop an effective and novel treatment method. Methods Niosomal nanoparticles were synthesized via the thin-film hydration method, and field emission scanning electron microscopy (FE-SEM), Fourier transform infrared (FTIR), atomic force microscopy (AFM), and dynamic light scattering (DLS) techniques were used to evaluate their physico-chemical characteristics. The cytotoxic effects of free and drug-loaded nanoparticles (NPs), as well as their combination, on A549 cells were assessed using the MTT assay. An apoptosis test was used while under the influence of medication to identify the molecular mechanisms behind programmed cell death. With the use of a cell cycle test, it was determined whether pharmaceutical effects caused the cell cycle to stop progressing. Additionally, the qRT-PCR technique was used to evaluate the levels of hTERT, BAX, and BCL-2 gene expression after 48-h medication treatment. Results In the cytotoxicity assay, the growth of A549 lung cancer cells was inhibited by both MET and SIL. Compared to the individual therapies, the combination of MET and SIL dramatically and synergistically decreased the IC50 values of MET and SIL in lung cancer cells. Furthermore, the combination of MET and SIL produced lower IC50 values and a better anti-proliferative effect on A549 lung cancer cells. Real-time PCR results showed that the expression levels of hTERT and BCL-2 were significantly reduced in lung cancer cell lines treated with MET and SIL compared to single treatments (p< 0.001). Conclusion It is anticipated that the use of nano-niosomal-formed MET and SIL would improve lung cancer treatment outcomes and improve the therapeutic efficiency of lung cancer cells.
Collapse
Affiliation(s)
- Elnaz Salmani-Javan
- Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Davoud Jafari-Gharabaghlou
- Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Esat Bonabi
- Department of Medical Microbiology, Faculty of Medicine, Istanbul Aydin University, Istanbul, Türkiye
| | - Nosratollah Zarghami
- Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Medical Biochemistry, Faculty of Medicine, Istanbul Aydin University, Istanbul, Türkiye
| |
Collapse
|
91
|
Li Z, Bao X, Liu X, Wang W, Yang J, Zhu X, Wang S. Transcriptome Profiling Based at Different Time Points after Hatching Deepened Our Understanding on Larval Growth and Development of Amphioctopus fangsiao. Metabolites 2023; 13:927. [PMID: 37623871 PMCID: PMC10456336 DOI: 10.3390/metabo13080927] [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: 07/09/2023] [Revised: 07/22/2023] [Accepted: 08/04/2023] [Indexed: 08/26/2023] Open
Abstract
As the quality of life improves, there is an increasing demand for nutrition-rich marine organisms like fish, shellfish, and cephalopods. To address this, artificial cultivation of these organisms is being explored along with ongoing research on their growth and development. A case in point is Amphioctopus fangsiao, a highly valued cephalopod known for its tasty meat, nutrient richness, and rapid growth rate. Despite its significance, there is a dearth of studies on the A. fangsiao growth mechanism, particularly of its larvae. In this study, we collected A. fangsiao larvae at 0, 4, 12, and 24 h post-hatching and conducted transcriptome profiling. Our analysis identified 4467, 5099, and 4181 differentially expressed genes (DEGs) at respective intervals, compared to the 0 h sample. We further analyzed the expression trends of these DEGs, noting a predominant trend of continuous upregulation. Functional exploration of this trend entailed GO and KEGG functional enrichment along with protein-protein interaction network analyses. We identified GLDC, DUSP14, DPF2, GNAI1, and ZNF271 as core genes, based on their high upregulation rate, implicated in larval growth and development. Similarly, CLTC, MEF2A, PPP1CB, PPP1R12A, and TJP1, marked by high protein interaction numbers, were identified as hub genes and the gene expression levels identified via RNA-seq analysis were validated through qRT-PCR. By analyzing the functions of key and core genes, we found that the ability of A. fangsiao larvae to metabolize carbohydrates, lipids, and other energy substances during early growth may significantly improve with the growth of the larvae. At the same time, muscle related cells in A. fangsiao larvae may develop rapidly, promoting the growth and development of larvae. Our findings provide preliminary insights into the growth and developmental mechanism of A. fangsiao, setting the stage for more comprehensive understanding and broader research into cephalopod growth and development mechanisms.
Collapse
Affiliation(s)
- Zan Li
- School of Agriculture, Ludong University, Yantai 264025, China
| | - Xiaokai Bao
- School of Agriculture, Ludong University, Yantai 264025, China
| | - Xiumei Liu
- College of Life Sciences, Yantai University, Yantai 264005, China
| | - Weijun Wang
- School of Agriculture, Ludong University, Yantai 264025, China
| | - Jianmin Yang
- School of Agriculture, Ludong University, Yantai 264025, China
| | - Xibo Zhu
- Fishery Technology Service Center of Lanshan District, Rizhao 276800, China
| | - Shuhai Wang
- Ocean and Aquatic Research Center of Hekou District, Dongying 257200, China
| |
Collapse
|
92
|
Abdel-Sattar OE, Allam RM, Al-Abd AM, El-Halawany AM, EL-Desoky AM, Mohamed SO, Sweilam SH, Khalid M, Abdel-Sattar E, Meselhy MR. Hypophyllanthin and Phyllanthin from Phyllanthus niruri Synergize Doxorubicin Anticancer Properties against Resistant Breast Cancer Cells. ACS OMEGA 2023; 8:28563-28576. [PMID: 37576627 PMCID: PMC10413485 DOI: 10.1021/acsomega.3c02953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Accepted: 07/21/2023] [Indexed: 08/15/2023]
Abstract
Doxorubicin (DOX) is a cornerstone chemotherapeutic agent for the treatment of several malignancies such as breast cancer; however, its activity is ameliorated by the development of a resistant phenotype. Phyllanthus species have been studied previously for their potential anticancer properties. The current work is aimed to study the potential cytotoxicity and chemomodulatory effects of hypophyllanthin (PN4) and phyllanthin (PN5) isolated from Phyllanthus niruri to DOX against the adriamycin multidrug-resistant breast cancer cells (MCF-7ADR) and elucidate their mechanism of action. The major compounds of the active methylene chloride fraction were isolated and assessed for their potential cytotoxicity and chemomodulatory effects on DOX against naïve (MCF-7) and resistant breast (MCF-7ADR) cancer cells. The mechanism of action of both compounds in terms of their impacts on programmed/non-programmed cell death (apoptosis and autophagy/necrosis), cell cycle progression/arrest, and tumor cell migration/invasion was investigated. Both compounds PN4 and PN5 showed a moderate but similar potency against MCF-7 as well as MCF-7ADR and significantly synergized DOX-induced anticancer properties against MCF-7ADR. The chemomodulatory effect of both compounds to DOX was found to be via potentiating DOX-induced cell cycle interference and apoptosis induction. It was found that PN4 and PN5 blocked the apoptosis-escape autophagy pathway in MCF-7ADR. On the molecular level, both compounds interfered with SIRT1 expression and consequently suppressed Akt phosphorylation, and PN5 blocked apoptosis escape. Furthermore, PN4 and PN5 showed promising antimigratory and anti-invasive effects against MCF-7ADR, as confirmed by suppression of N-cadherin/β-catenin expression. In conclusion, for the first time, hypophyllanthin and phyllanthin isolated from P. niruri showed promising chemomodulatory effects to the DOX-induced chemotherapeutic activity against MCF-7ADR. Both compounds significantly synergized DOX-induced anticancer properties against MCF-7ADR. This enhanced activity was explained by further promoting DOX-induced apoptosis and suppressing the apoptosis-escape autophagy feature of the resistant breast cancer cells. Both compounds (hypophyllanthin and phyllanthin) interfered with the SIRT1/Akt pathway and suppressed the N-cadherin/β-catenin axis, confirming the observed antiproliferative, cytotoxic, and anti-invasive effects of hypophyllanthin and phyllanthin.
Collapse
Affiliation(s)
- Ola E. Abdel-Sattar
- Pharmacognosy Department,
Faculty of Pharmacy, Cairo University, Kasr El Aini St., Cairo 11562, Egypt
| | - Rasha M. Allam
- Pharmacology
Department, Medical Research Institute, National Research Centre, Dokki, Cairo 12622, Egypt
| | - Ahmed M. Al-Abd
- Pharmacology
Department, Medical Research Institute, National Research Centre, Dokki, Cairo 12622, Egypt
| | - Ali M. El-Halawany
- Pharmacognosy Department,
Faculty of Pharmacy, Cairo University, Kasr El Aini St., Cairo 11562, Egypt
| | - Ahmed M. EL-Desoky
- Department of Molecular Biology,
Genetic Engineering and Biotechnology Research Institute (GEBRI), University of Sadat City (USC), Sadat City 32958, Egypt
| | - Shanaz O. Mohamed
- School of Pharmaceutical
Sciences, Universiti Sains Malaysia, Gelugor, Penang 11700, Malaysia
| | - Sherouk Hussein Sweilam
- Department of Pharmacognosy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
- Department of Pharmacognosy, Faculty of
Pharmacy, Egyptian Russian University, Cairo-Suez Road, Badr City, Cairo 11829, Egypt
| | - Mohammad Khalid
- Department of Pharmacognosy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Essam Abdel-Sattar
- Pharmacognosy Department,
Faculty of Pharmacy, Cairo University, Kasr El Aini St., Cairo 11562, Egypt
| | - Meselhy R. Meselhy
- Pharmacognosy Department,
Faculty of Pharmacy, Cairo University, Kasr El Aini St., Cairo 11562, Egypt
| |
Collapse
|
93
|
Cao J, Tao X, Shi B, Wang J, Ma R, Zhao J, Tian J, Huang Q, Yu J, Wang L. NKD1 targeting PCM1 regulates the therapeutic effects of homoharringtonine on colorectal cancer. Mol Biol Rep 2023; 50:6543-6556. [PMID: 37338734 DOI: 10.1007/s11033-023-08572-5] [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: 03/29/2023] [Accepted: 05/31/2023] [Indexed: 06/21/2023]
Abstract
BACKGROUND Colorectal cancer (CRC) is the most common primary malignancy. Recently, antineoplastic attributes of homoharringtonine (HHT) have attracted lots of attention. This study investigated the molecular target and underlying mechanism of HHT in the CRC process by using a cellular and animal models. METHODS This study first detected the effects of HHT on the proliferation, cell cycle and apoptosis ability of CRC cells using CCK-8, Edu staining, flow cytometry and Western blotting assay. In vitro recovery experiment and in vivo tumorigenesis experiment were used to detect the targeted interaction between HHT and NKD1. After that, the downstream target and mechanism of action of HHT targeting NKD1 was determined using quantitative proteomics combined with co-immunoprecipitation/immunofluorescence assay. RESULTS HHT suppressed CRC cells proliferation by inducing cell cycle arrest and apoptosis in vitro and vivo. HHT inhibited NKD1 expression in a concentration and time dependent manner. NKD1 was overexpressed in CRC and its depletion enhanced the therapeutic sensitivity of HHT on CRC, which indicating that NKD1 plays an important role in the development of CRC as the drug delivery target of HHT. Furthermore, proteomic analysis revealed that PCM1 participated the process of NKD1-regulated cell proliferation and cell cycle. NKD1 interacted with PCM1 and promoted PCM1 degradation through the ubiquitin-proteasome pathway. The overexpression of PCM1 effectively reversed the inhibition of siNKD1 on cell cycle. CONCLUSIONS The present findings revealed that HHT blocked NKD1 expression to participate in inhibiting cell proliferation and inducing cell apoptosis, ultimately leading to obstruction of CRC development through NKD1/PCM1 dependent mechanism. Our research provide evidence for clinical application of NKD1-targeted therapy in improving HHT sensitivity for CRC treatment.
Collapse
Affiliation(s)
- Jia Cao
- Department of Beijing National Biochip Research Center Sub-Center in Ningxia, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
- Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Xiang Tao
- College of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Bin Shi
- Department of Emergency, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Jia Wang
- Department of Beijing National Biochip Research Center Sub-Center in Ningxia, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
- Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Rong Ma
- Department of Beijing National Biochip Research Center Sub-Center in Ningxia, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
- Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Jufen Zhao
- College of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Jinhai Tian
- Department of Beijing National Biochip Research Center Sub-Center in Ningxia, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Qi Huang
- Department of Beijing National Biochip Research Center Sub-Center in Ningxia, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
- Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Jingjing Yu
- Department of Beijing National Biochip Research Center Sub-Center in Ningxia, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
- Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Libin Wang
- Department of Beijing National Biochip Research Center Sub-Center in Ningxia, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China.
- College of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia, China.
| |
Collapse
|
94
|
Abedi H, Nemati M, Ebrahimi B, Dehghani M, Mikaeiliagah E, Abdollahzadeh P, Ghanaatpishe A, Shafiee Jahromi N, Kargar Jahromi H. The protective effect of hydroalcoholic extract of Ephedra pachyclada leaves on ovarian damage induced by cyclophosphamide in rat: An experimental study. Int J Reprod Biomed 2023; 21:629-638. [PMID: 37885971 PMCID: PMC10598471 DOI: 10.18502/ijrm.v21i8.14018] [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: 03/16/2023] [Revised: 06/12/2023] [Accepted: 07/19/2023] [Indexed: 10/28/2023] Open
Abstract
Background Cyclophosphamide (CP) is an anticancer drug that acts as an alkylation agent after metabolism in the liver. CP has toxic effects on the body's cells, especially the reproductive system's function, and causes infertility. Moreover, medicinal plants have few side effects and are psychologically acceptable to patients. Objective This study aimed to investigate the impact of Ephedra pachyclada hydroalcoholic extract (EPHE) on ovarian tissue and hypothalamic-pituitary-gonadal axis in rats treated with CP. Materials and Methods In this experimental study, 48 adult female Wistar rats (180-200 gr, 9-10 wk) were randomly assigned to 6 experimental groups (n = 8/each): (a) control; (b) sham; (c) CP; (d) CP+250 mg/kg EPHE; (e) CP+500 mg/kg EPHE; (f) CP+1000 mg/kg EPHE. On the 29th day of the experiment, serum was collected; serum concentration of the luteinizing hormone, follicle-stimulating hormone, estrogen, progesterone, and antioxidant activity were measured. The number of ovarian follicles were also counted. Results In the CP groups, serum concentrations of follicle-stimulating hormone and luteinizing hormone significantly increased, and estrogen and progesterone significantly decreased (p ≤ 0.05). EPHE significantly compensated for the complications caused by CP and 1000 mg/kg had the greatest effect. Antioxidant reduction by CP was significantly enhanced by EPHE, especially at higher doses (p ≤ 0.05). The number of primordial, primary, secondary, and Graafian follicles showed a significant decrease in CP groups and EPHE groups showed a significant increase compared to the CP. EPHE showed that the concentration of 1000 mg/kg was more effective than other doses (p ≤ 0.05). Conclusion In addition to proving the effect of EPHE on the hypothalamic-pituitary-gonadal axis, our investigation showed antioxidant properties, which can be an effective factor in CP-treated rats.
Collapse
Affiliation(s)
- Hassanali Abedi
- Research Center for Noncommunicable Diseases, Jahrom University of Medical Sciences, Jahrom, Iran
| | - Mahnaz Nemati
- Amir Oncology Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Bahare Ebrahimi
- Shiraz Geriatric Research Center, Shiraz University of Medical Science, Shiraz, Iran
| | - Maryam Dehghani
- Research Center for Noncommunicable Diseases, Jahrom University of Medical Sciences, Jahrom, Iran
| | - Elmira Mikaeiliagah
- Department of Biology, Ardabil Branch, Islamic Azad University, Ardabil, Iran
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, USA
| | - Pegah Abdollahzadeh
- Research Center for Noncommunicable Diseases, Jahrom University of Medical Sciences, Jahrom, Iran
| | - Aref Ghanaatpishe
- Student Research Committee, Jahrom University of Medical Sciences, Jahrom, Iran
| | | | - Hossein Kargar Jahromi
- Research Center for Noncommunicable Diseases, Jahrom University of Medical Sciences, Jahrom, Iran
| |
Collapse
|
95
|
Scimeca M, Rovella V, Palumbo V, Scioli MP, Bonfiglio R, Tor Centre, Melino G, Piacentini M, Frati L, Agostini M, Candi E, Mauriello A. Programmed Cell Death Pathways in Cholangiocarcinoma: Opportunities for Targeted Therapy. Cancers (Basel) 2023; 15:3638. [PMID: 37509299 PMCID: PMC10377326 DOI: 10.3390/cancers15143638] [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: 05/16/2023] [Revised: 07/06/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
Cholangiocarcinoma is a highly aggressive cancer arising from the bile ducts. The limited effectiveness of conventional therapies has prompted the search for new approaches to target this disease. Recent evidence suggests that distinct programmed cell death mechanisms, namely, apoptosis, ferroptosis, pyroptosis and necroptosis, play a critical role in the development and progression of cholangiocarcinoma. This review aims to summarize the current knowledge on the role of programmed cell death in cholangiocarcinoma and its potential implications for the development of novel therapies. Several studies have shown that the dysregulation of apoptotic signaling pathways contributes to cholangiocarcinoma tumorigenesis and resistance to treatment. Similarly, ferroptosis, pyroptosis and necroptosis, which are pro-inflammatory forms of cell death, have been implicated in promoting immune cell recruitment and activation, thus enhancing the antitumor immune response. Moreover, recent studies have suggested that targeting cell death pathways could sensitize cholangiocarcinoma cells to chemotherapy and immunotherapy. In conclusion, programmed cell death represents a relevant molecular mechanism of pathogenesis in cholangiocarcinoma, and further research is needed to fully elucidate the underlying details and possibly identify therapeutic strategies.
Collapse
Affiliation(s)
- Manuel Scimeca
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Valentina Rovella
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Valeria Palumbo
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Maria Paola Scioli
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Rita Bonfiglio
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133 Rome, Italy
| | | | - Gerry Melino
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Mauro Piacentini
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Luigi Frati
- Institute Pasteur Italy-Cenci Bolognetti Foundation, Via Regina Elena 291, 00161 Rome, Italy
- IRCCS Neuromed S.p.A., Via Atinense 18, 86077 Pozzilli, Italy
| | - Massimiliano Agostini
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Eleonora Candi
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Alessandro Mauriello
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133 Rome, Italy
| |
Collapse
|
96
|
Pellot Ortiz KI, Rechberger JS, Nonnenbroich LF, Daniels DJ, Sarkaria JN. MDM2 Inhibition in the Treatment of Glioblastoma: From Concept to Clinical Investigation. Biomedicines 2023; 11:1879. [PMID: 37509518 PMCID: PMC10377337 DOI: 10.3390/biomedicines11071879] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 06/28/2023] [Accepted: 06/30/2023] [Indexed: 07/30/2023] Open
Abstract
Inhibition of the interaction between MDM2 and p53 has emerged as a promising strategy for combating cancer, including the treatment of glioblastoma (GBM). Numerous MDM2 inhibitors have been developed and are currently undergoing rigorous testing for their potential in GBM therapy. Encouraging results from studies conducted in cell culture and animal models suggest that MDM2 inhibitors could effectively treat a specific subset of GBM patients with wild-type TP53 or functional p53. Combination therapy with clinically established treatment modalities such as radiation and chemotherapy offers the potential to achieve a more profound therapeutic response. Furthermore, an increasing array of other molecularly targeted therapies are being explored in combination with MDM2 inhibitors to increase the effects of individual treatments. While some MDM2 inhibitors have progressed to early phase clinical trials in GBM, their efficacy, alone and in combination, is yet to be confirmed. In this article, we present an overview of MDM2 inhibitors currently under preclinical and clinical investigation, with a specific focus on the drugs being assessed in ongoing clinical trials for GBM patients.
Collapse
Affiliation(s)
| | - Julian S Rechberger
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905, USA
| | - Leo F Nonnenbroich
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA
- Hopp Children's Cancer Center Heidelberg (KiTZ), 69120 Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), 69120 Heidelberg, Germany
| | - David J Daniels
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905, USA
| | - Jann N Sarkaria
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
97
|
Mao X, Cai Y, Long S, Perez-Losada J, Mao JH, Chang H. Pan-cancer evaluation of clinical value of mitotic network activity index (MNAI) and its predictive value for immunotherapy. Front Oncol 2023; 13:1178568. [PMID: 37456231 PMCID: PMC10349373 DOI: 10.3389/fonc.2023.1178568] [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: 03/03/2023] [Accepted: 06/12/2023] [Indexed: 07/18/2023] Open
Abstract
Increased mitotic activity is associated with the genesis and aggressiveness of many cancers. To assess the clinical value of mitotic activity as prognostic biomarker, we performed a pan-cancer study on the mitotic network activity index (MNAI) constructed based on 54-gene mitotic apparatus network. Our pan-cancer assessment on TCGA (33 tumor types, 10,061 patients) and validation on other publicly available cohorts (23 tumor types, 9,209 patients) confirmed the significant association of MNAI with overall survival, progression-free survival, and other prognostic endpoints in multiple cancer types, including lower-grade gliomas (LGG), breast invasive carcinoma (BRCA), as well as many others. We also showed significant association between MNAI and genetic instability, which provides a biological explanation of its prognostic impact at pan-cancer landscape. Our association analysis revealed that patients with high MNAI benefitted more from anti-PD-1 and Anti-CTLA-4 treatment. In addition, we demonstrated that multimodal integration of MNAI and the AI-empowered Cellular Morphometric Subtypes (CMS) significantly improved the predictive power of prognosis compared to using MNAI and CMS alone. Our results suggest that MNAI can be used as a potential prognostic biomarker for different tumor types toward different clinical endpoints, and multimodal integration of MNAI and CMS exceeds individual biomarker for precision prognosis.
Collapse
Affiliation(s)
- Xuanyu Mao
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA, United States
- Berkeley Biomedical Data Science Center, Lawrence Berkeley National Laboratory, Berkeley, CA, United States
| | - Yimeng Cai
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA, United States
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, United States
| | - Sarah Long
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA, United States
| | - Jesus Perez-Losada
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca/CSIC, Salamanca, Spain
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), Salamanca, Spain
| | - Jian-Hua Mao
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA, United States
- Berkeley Biomedical Data Science Center, Lawrence Berkeley National Laboratory, Berkeley, CA, United States
| | - Hang Chang
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA, United States
- Berkeley Biomedical Data Science Center, Lawrence Berkeley National Laboratory, Berkeley, CA, United States
| |
Collapse
|
98
|
Zhao J, Zhang N, Ma X, Li M, Feng H. The dual role of ferroptosis in anthracycline-based chemotherapy includes reducing resistance and increasing toxicity. Cell Death Discov 2023; 9:184. [PMID: 37344500 DOI: 10.1038/s41420-023-01483-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 06/02/2023] [Accepted: 06/14/2023] [Indexed: 06/23/2023] Open
Abstract
In conjunction with previous studies, we have noted that ferroptosis, as an emerging mode of regulated cell death (RCD), is intimately related to anthracycline pharmacotherapy. Not only does ferroptosis significantly modulate tumour resistance and drug toxicity, which are core links of the relevant chemotherapeutic process, but it also appears to play a conflicting role that has yet to be appreciated. By targeting the dual role of ferroptosis in anthracycline-based chemotherapy, this review aims to focus on the latest findings at this stage, identify the potential associations and provide novel perspectives for subsequent research directions and therapeutic strategies.
Collapse
Affiliation(s)
- Jiazheng Zhao
- Department of Orthopedics, The Fourth Hospital of Hebei Medical University, 12 Health Road, Shijiazhuang, Hebei, 050011, China
| | - Ning Zhang
- Department of Cardiology, The Fourth Hospital of Hebei Medical University, 12 Health Road, Shijiazhuang, Hebei, 050011, China
| | - Xiaowei Ma
- Departments of Orthopedics, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Nanli, Panjiayuan, Chaoyang District, Beijing, 100021, China
| | - Ming Li
- Department of Orthopedics, The Second Hospital of Hebei Medical University, 215 Heping Road, Shijia-zhuang, Hebei, China
| | - Helin Feng
- Departments of Orthopedics, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Nanli, Panjiayuan, Chaoyang District, Beijing, 100021, China.
| |
Collapse
|
99
|
Ciardulli MC, Mariconda A, Sirignano M, Lamparelli EP, Longo R, Scala P, D'Auria R, Santoro A, Guadagno L, Della Porta G, Longo P. Activity and Selectivity of Novel Chemical Metallic Complexes with Potential Anticancer Effects on Melanoma Cells. Molecules 2023; 28:4851. [PMID: 37375406 DOI: 10.3390/molecules28124851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 06/07/2023] [Accepted: 06/10/2023] [Indexed: 06/29/2023] Open
Abstract
Human malignant melanoma cells from lymph node metastatic site (MeWo) were selected for testing several synthesized and purified silver(I) and gold(I) complexes stabilized by unsymmetrically substituted N-heterocyclic carbene (NHC) ligands, called L20 (N-methyl, N'-[2-hydroxy ethylphenyl]imidazol-2-ylide) and M1 (4,5-dichloro, N-methyl, N'-[2-hydroxy ethylphenyl]imidazol-2-ylide), having halogenide (Cl- or I-) or aminoacyl (Gly=N-(tert-Butoxycarbonyl)glycinate or Phe=(S)-N-(tert-Butoxycarbonyl)phenylalaninate) counterion. For AgL20, AuL20, AgM1 and AuM1, the Half-Maximal Inhibitory Concentration (IC50) values were measured, and all complexes seemed to reduce cell viability more effectively than Cisplatin, selected as control. The complex named AuM1 was the most active just after 8 h of treatment at 5 μM, identified as effective growth inhibition concentration. AuM1 also showed a linear dose and time-dependent effect. Moreover, AuM1 and AgM1 modified the phosphorylation levels of proteins associated with DNA lesions (H2AX) and cell cycle progression (ERK). Further screening of complex aminoacyl derivatives indicated that the most powerful were those indicated with the acronyms: GlyAg, PheAg, AgL20Gly, AgM1Gly, AuM1Gly, AgL20Phe, AgM1Phe, AuM1Phe. Indeed, the presence of Boc-Glycine (Gly) and Boc-L-Phenylalanine (Phe) showed an improved efficacy of Ag main complexes, as well as that of AuM1 derivatives. Selectivity was further checked on a non-cancerous cell line, a spontaneously transformed aneuploid immortal keratinocyte from adult human skin (HaCaT). In such a case, AuM1 and PheAg complexes resulted as the most selective allowing HaCaT viability at 70 and 40%, respectively, after 48 h of treatment at 5 μM. The same complexes tested on 3D MeWo static culture induced partial spheroid disaggregation after 24 h of culture, with almost half of the cells dead.
Collapse
Affiliation(s)
- Maria Camilla Ciardulli
- Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 84081 Baronissi, Italy
| | - Annaluisa Mariconda
- Department of Science, University of Basilicata, Viale dell'Ateneo Lucano 10, 85100 Potenza, Italy
| | - Marco Sirignano
- Department of Chemistry and Biology "Adolfo Zambelli", University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy
| | - Erwin Pavel Lamparelli
- Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 84081 Baronissi, Italy
| | - Raffaele Longo
- Department of Industrial Engineering, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy
| | - Pasqualina Scala
- Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 84081 Baronissi, Italy
| | - Raffaella D'Auria
- Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 84081 Baronissi, Italy
| | - Antonietta Santoro
- Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 84081 Baronissi, Italy
- Interdepartment Centre BIONAM, University of Salerno, Via Giovanni Paolo II, 84084 Fisciano, Italy
| | - Liberata Guadagno
- Department of Industrial Engineering, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy
| | - Giovanna Della Porta
- Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 84081 Baronissi, Italy
- Interdepartment Centre BIONAM, University of Salerno, Via Giovanni Paolo II, 84084 Fisciano, Italy
| | - Pasquale Longo
- Department of Chemistry and Biology "Adolfo Zambelli", University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy
| |
Collapse
|
100
|
Choroba K, Filipe B, Świtlicka A, Penkala M, Machura B, Bieńko A, Cordeiro S, Baptista PV, Fernandes AR. In Vitro and In Vivo Biological Activities of Dipicolinate Oxovanadium(IV) Complexes. J Med Chem 2023. [PMID: 37311060 DOI: 10.1021/acs.jmedchem.3c00255] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The work is focused on anticancer properties of dipicolinate (dipic)-based vanadium(IV) complexes [VO(dipic)(N∩N)] bearing different diimines (2-(1H-imidazol-2-yl)pyridine, 2-(2-pyridyl)benzimidazole, 1,10-phenanthroline-5,6-dione, 1,10-phenanthroline, and 2,2'-bipyridine), as well as differently 4,7-substituted 1,10-phenanthrolines. The antiproliferative effect of V(IV) systems was analyzed in different tumors (A2780, HCT116, and HCT116-DoxR) and normal (primary human dermal fibroblasts) cell lines, revealing a high cytotoxic effect of [VO(dipic)(N∩N)] with 4,7-dimethoxy-phen (5), 4,7-diphenyl-phen (6), and 1,10-phenanthroline (8) against HCT116-DoxR cells. The cytotoxicity differences between these complexes can be correlated with their different internalization by HCT116-DoxR cells. Worthy of note, these three complexes were found to (i) induce cell death through apoptosis and autophagy pathways, namely, through ROS production; (ii) not to be cytostatic; (iii) to interact with the BSA protein; (iv) do not promote tumor cell migration or a pro-angiogenic capability; (v) show a slight in vivo anti-angiogenic capability, and (vi) do not show in vivo toxicity in a chicken embryo.
Collapse
Affiliation(s)
- Katarzyna Choroba
- University of Silesia, Institute of Chemistry, Szkolna 9, 40-006 Katowice, Poland
| | - Beatriz Filipe
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal
- UCIBIO - Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal
| | - Anna Świtlicka
- University of Silesia, Institute of Chemistry, Szkolna 9, 40-006 Katowice, Poland
| | - Mateusz Penkala
- University of Silesia, Institute of Chemistry, Szkolna 9, 40-006 Katowice, Poland
| | - Barbara Machura
- University of Silesia, Institute of Chemistry, Szkolna 9, 40-006 Katowice, Poland
| | - Alina Bieńko
- Faculty of Chemistry, University of Wroclaw, F. Joliot-Curie 14, 50-383 Wroclaw, Poland
| | - Sandra Cordeiro
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal
- UCIBIO - Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal
| | - Pedro V Baptista
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal
- UCIBIO - Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal
| | - Alexandra R Fernandes
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal
- UCIBIO - Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal
| |
Collapse
|