1
|
Qiu J, Zhao Z, Suo H, Paraghamian SE, Hawkins GM, Sun W, Zhang X, Hao T, Deng B, Shen X, Zhou C, Bae-Jump V. Linoleic acid exhibits anti-proliferative and anti-invasive activities in endometrial cancer cells and a transgenic model of endometrial cancer. Cancer Biol Ther 2024; 25:2325130. [PMID: 38465855 PMCID: PMC10936646 DOI: 10.1080/15384047.2024.2325130] [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: 07/31/2023] [Accepted: 02/26/2024] [Indexed: 03/12/2024] Open
Abstract
Emerging evidence has provided considerable insights into the integral function of reprogramming fatty acid metabolism in the carcinogenesis and progression of endometrial cancer. Linoleic acid, an essential fatty acid with the highest consumption in the Western diet regimen, has shown pro-tumorigenic or anti-tumorigenic effects on tumor cell growth and invasion in multiple types of cancer. However, the biological role of linoleic acid in endometrial cancer remains unclear. In the present study, we aimed to investigate the functional impact of linoleic acid on cell proliferation, invasion, and tumor growth in endometrial cancer cells and in a transgenic mouse model of endometrial cancer. The results showed that Linoleic acid significantly inhibited the proliferation of endometrial cancer cells in a dose-dependent manner. The treatment of HEC-1A and KLE cells with linoleic acid effectively increased intracellular reactive oxygen species (ROS) production, decreased mitochondrial membrane potential, caused cell cycle G1 arrest, and induced intrinsic and extrinsic apoptosis pathways. The anti-invasive ability of linoleic acid was found to be associated with the epithelial-mesenchymal transition process in both cell lines, including the decreased expression of N-cadherin, snail, and vimentin. Furthermore, treatment of Lkb1fl/flp53fl/fl transgenic mice with linoleic acid for four weeks significantly reduced the growth of endometrial tumors and decreased the expression of VEGF, vimentin, Ki67, and cyclin D1 in tumor tissues. Our findings demonstrate that linoleic acid exhibits anti-proliferative and anti-invasive activities in endometrial cancer cell lines and the Lkb1fl/flp53fl/fl mouse model of endometrial cancer, thus providing a pre-clinical basis for future dietary interventions with linoleic acid in endometrial cancer.
Collapse
Affiliation(s)
- Jianqing Qiu
- Department of Obstetrics and Gynecology, the Second Hospital of Shandong University, Jinan, PR, China
| | - Ziyi Zhao
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital MedicalUniversity, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Hongyan Suo
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital MedicalUniversity, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Sarah E. Paraghamian
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Gabrielle M. Hawkins
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Wenchuan Sun
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Xin Zhang
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital MedicalUniversity, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Tianran Hao
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Beor Deng
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital MedicalUniversity, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Xiaochang Shen
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital MedicalUniversity, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Chunxiao Zhou
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Victoria Bae-Jump
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
2
|
Zhang S, Gao R, Ding B, Li J, Wang T, Chen J, Li C, Jiao Y, Song L. Antihepatoma activity of Marsdenia tenacissima polysaccharide-decorated selenium nanoparticles by regulating the Bax/Bcl-2/caspases and p21/Akt/cyclin A2 signaling pathways. Int J Biol Macromol 2024; 279:134981. [PMID: 39182863 DOI: 10.1016/j.ijbiomac.2024.134981] [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: 01/30/2024] [Revised: 07/31/2024] [Accepted: 08/21/2024] [Indexed: 08/27/2024]
Abstract
Combining selenium nanoparticles (SeNPs) with bioactive polysaccharides is one of the effective ways to overcome the shortcomings of SeNPs and polysaccharides and obtain novel antitumor drug candidates. In this study, a heteropolysaccharide (MTP70) with moderate antihepatoma activity was isolated from the stems of Marsdenia tenacissima (Roxb.) Wight et Arn. To further improve the antihepatoma activity of MTP70 and the application of SeNPs, a novel stable nanoparticle (MTP-SeNP) was designed and fabricated. MTP-SeNPs (Se content of 8.25 %) were characterized as monodisperse spherical nanoparticles (50 nm) with MTP70 wrapped on the surface of the SeNPs by the formation of CO⋯Se bonds and possessed high stability and good dispersion in water for almost a month. In addition, MTP-SeNPs showed higher inhibitory effect compared with MTP70. MTP-SeNPs could effectively inhibit the proliferation, invasion, and metastasis of HepG2 cells by inducing apoptosis and arresting the cell cycle at the S phase, which were closely related to the activation of the Bax/Bcl-2/Caspases and p21/Akt/Cyclin A2 signaling pathways. Our results provide a theoretical basis for further development and application of M. tenacissima polysaccharide, and show that MTP-SeNPs could be explored as a promising anti-hepatoma agent in the pharmaceutical and biomedical industries.
Collapse
Affiliation(s)
- Shaojie Zhang
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, The Affiliated Dongguan Songshan Lake Central Hospital, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China.
| | - Renjie Gao
- Taian City Central Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian 271099, China; Department of Geriatrics, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Baocong Ding
- Department of Rehabilitation, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250000, China
| | - Junhao Li
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, The Affiliated Dongguan Songshan Lake Central Hospital, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Tanggan Wang
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, The Affiliated Dongguan Songshan Lake Central Hospital, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Jiaheng Chen
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong 510530, China
| | - Chong Li
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong 510530, China
| | - Yukun Jiao
- Institute of Traditional Chinese Medicine Health Industry, China Academy of Chinese Medical Sciences, Nanchang 330000, China.
| | - Lijun Song
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, The Affiliated Dongguan Songshan Lake Central Hospital, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China.
| |
Collapse
|
3
|
Yan Z, Chen HQ. Anti-liver cancer effects and mechanisms and its application in nano DDS of polysaccharides: A review. Int J Biol Macromol 2024; 279:135181. [PMID: 39218183 DOI: 10.1016/j.ijbiomac.2024.135181] [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: 05/13/2024] [Revised: 07/23/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
Liver cancer is the third leading cause of cancer death, with high incidence and poor treatment effect. In recent years, polysaccharides have attracted more and more attention in the research field of anti-liver cancer because of their high efficiency, low toxicity, good biocompatibility, wide sources and low cost. Polysaccharides have been proven to have good anti-liver cancer activity. In this paper, the pathways and molecular mechanisms of polysaccharides against liver cancer were reviewed in detail. Polysaccharides exert anti-liver cancer activity by blocking cell cycle, inducing apoptosis, regulating immunity, inhibiting cancer cell metastasis, inhibiting tumor angiogenesis and so on. The primary structure and chain conformation of polysaccharides have an important influence on their anti-liver cancer activity. Structural modification enhanced the anti-liver cancer activity of polysaccharides. Polysaccharides have good attenuated and synergistic effects on chemotherapy drugs. Polysaccharides can be used as functional carriers to construct intelligent nano drug delivery systems (DDS) targeting liver cancer. This review can provide theoretical support for the further development and application of polysaccharides in the field of anti-liver cancer, and provide theoretical reference and clues for relevant researchers in food, nutrition, medicine and other fields.
Collapse
Affiliation(s)
- Zheng Yan
- School of Food and Biological Engineering, Hefei University of Technology, 420 Feicui Road, Hefei, Anhui 230601, PR China; Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, 420 Feicui Road, Hefei, Anhui 230601, PR China
| | - Han-Qing Chen
- School of Food and Biological Engineering, Hefei University of Technology, 420 Feicui Road, Hefei, Anhui 230601, PR China; Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, 420 Feicui Road, Hefei, Anhui 230601, PR China.
| |
Collapse
|
4
|
Zhao G, Jia M, Zhu S, Ren H, Wang G, Xin G, Sun M, Wang X, Lin Q, Jiang Q, Zhang C. Mitotic ER-mitochondria contact enhances mitochondrial Ca 2+ influx to promote cell division. Cell Rep 2024; 43:114794. [PMID: 39342616 DOI: 10.1016/j.celrep.2024.114794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 07/31/2024] [Accepted: 09/09/2024] [Indexed: 10/01/2024] Open
Abstract
Cell division is tightly regulated and requires an expanded energy supply. However, how this energy is generated remains unclear. Here, we establish a correlation between two mitochondrial Ca2+ influx events and ATP production during mitosis. While both events promote ATP production during mitosis, the second event, the Ca2+ influx surge, is substantial. To facilitate this Ca2+ influx surge, the lamin B receptor (LBR) organizes a mitosis-specific endoplasmic reticulum (ER)-mitochondrial contact site (ERMCS), creating a rapid Ca2+ transport pathway. LBR acts as a tether, connecting the ER Ca2+ release channel IP3R with the mitochondrial VDAC2. Depletion of LBR disrupts the Ca2+ influx surge, reduces ATP production, and postpones the metaphase-anaphase transition and subsequent cell division. These findings provide insight into the mechanisms underlying mitotic energy production and supply required for cell proliferation.
Collapse
Affiliation(s)
- Gan Zhao
- The Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking University, Beijing 100871, China
| | - Mingkang Jia
- The Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking University, Beijing 100871, China
| | - Shicong Zhu
- The Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking University, Beijing 100871, China
| | - He Ren
- The Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking University, Beijing 100871, China
| | - Guopeng Wang
- The Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking University, Beijing 100871, China
| | - Guangwei Xin
- The Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking University, Beijing 100871, China
| | - Mengjie Sun
- The Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking University, Beijing 100871, China
| | - Xiangyang Wang
- The Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking University, Beijing 100871, China
| | - Qiaoyu Lin
- The Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking University, Beijing 100871, China
| | - Qing Jiang
- The Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking University, Beijing 100871, China
| | - Chuanmao Zhang
- The Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking University, Beijing 100871, China; The Academy for Cell and Life Health, Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China.
| |
Collapse
|
5
|
Hu H, Chen J, Zhang F, Sheng Z, Yang Y, Xie Y, Zhou L, Liu Y. Evaluation of Efficiency of Liposome-Entrapped Iridium(III) Complexes Inhibiting Tumor Growth In Vitro and In Vivo. J Med Chem 2024; 67:16195-16208. [PMID: 39264254 DOI: 10.1021/acs.jmedchem.4c01026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/13/2024]
Abstract
In this paper, three new iridium(III) complexes: [Ir(piq)2(DFIPP)]PF6 (piq = deprotonated 1-phenylisoquinoline, DFIPP = 3,4-difluoro-2-(1H-imidazo[4,5-f][1,10]phenenthrolin-2-yl)phenol, 3a), [Ir(bzq)2(DFIPP)]PF6 (bzq = deprotonated benzo[h]quinoline, 3b), and [Ir(ppy)2(DFIPP)]PF6 (ppy = deprotonated 1-phenylpyridine, 3c), were synthesized and characterized. The complexes were found to be nontoxic to tumor cells via 3-(4,5-dimethylthiazole-2-yl)-diphenyltetrazolium bromide (MTT) assay. Surprisingly, its liposome-entrapped complexes 3alip, 3blip, and 3clip on B16 cells showed strong cytotoxicity (IC50 = 13.6 ± 2.8, 9.6 ± 1.1, and 18.9 ± 2.1 μM). Entry of 3alip, 3blip, and 3clip into B16 cells decreases mitochondrial membrane potential, regulates Bcl-2 family proteins, releases cytochrome c, triggers caspase family cascade reaction, and induces apoptosis. In addition, we also found that 3alip, 3blip, and 3clip triggered ferroptosis and autophagy. In vivo studies demonstrated that 3blip inhibited melanoma growth in C57 mice with a high inhibitory rate of 83.95%, and no organic damage was found in C57 mice.
Collapse
Affiliation(s)
- Huiyan Hu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Jing Chen
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Fan Zhang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Zhujun Sheng
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Yan Yang
- Department of Pharmacy, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, PR China
| | - Yufeng Xie
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Lin Zhou
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Yunjun Liu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| |
Collapse
|
6
|
Teotia V, Jha P, Chopra M. Discovery of Potential Inhibitors of CDK1 by Integrating Pharmacophore-Based Virtual Screening, Molecular Docking, Molecular Dynamics Simulation Studies, and Evaluation of Their Inhibitory Activity. ACS OMEGA 2024; 9:39873-39892. [PMID: 39346877 PMCID: PMC11425824 DOI: 10.1021/acsomega.4c05414] [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: 06/10/2024] [Revised: 08/26/2024] [Accepted: 08/29/2024] [Indexed: 10/01/2024]
Abstract
The ability of CDK1 to compensate for the absence of other cell cycle CDKs poses a great challenge to treat cancers that overexpress these proteins. Despite several studies focusing on the area, there are no FDA-approved drugs selectively targeting CDK1. Here, the study aimed to develop potential CDK1 selective inhibitors through drug repurposing and leveraging the structural insights provided by the hit molecules generated. Approximately 280,000 compounds from DrugBank, Selleckchem, Otava and an in-house library were screened initially based on fit values using 3D QSAR pharmacophores built for CDK1 and subsequently through Lipinski, ADMET, and TOPKAT filters. 10,310 hits were investigated for docking into the binding site of CDK1 determined using the crystal structure of human CDK1 in complex with NU6102. The best 55 hits with better docking scores were further analyzed, and 12 hits were selected for 100 ns MD simulations followed by binding energy calculations using the MM-PBSA method. Finally, 10 hit molecules were tested in an in vitro CDK1 Kinase inhibition assay. Out of these, 3 hits showed significant CDK1 inhibitory potential with IC50 < 5 μM. These results indicate these compounds can be used to develop subtype-selective CDK1 inhibitors with better efficacy and reduced toxicities in the future.
Collapse
Affiliation(s)
- Vineeta Teotia
- Laboratory
of Molecular Modeling and Anti-Cancer Drug Development, Dr. B. R.
Ambedkar Center for Biomedical Research, University of Delhi, Delhi 110007, India
| | - Prakash Jha
- Laboratory
of Molecular Modeling and Anti-Cancer Drug Development, Dr. B. R.
Ambedkar Center for Biomedical Research, University of Delhi, Delhi 110007, India
| | - Madhu Chopra
- Laboratory
of Molecular Modeling and Anti-Cancer Drug Development, Dr. B. R.
Ambedkar Center for Biomedical Research, University of Delhi, Delhi 110007, India
| |
Collapse
|
7
|
Gambardella AR, Antonucci C, Zanetti C, Noto F, Andreone S, Vacca D, Pellerito V, Sicignano C, Parrottino G, Tirelli V, Tinari A, Falchi M, De Ninno A, Businaro L, Loffredo S, Varricchi G, Tripodo C, Afferni C, Parolini I, Mattei F, Schiavoni G. IL-33 stimulates the anticancer activities of eosinophils through extracellular vesicle-driven reprogramming of tumor cells. J Exp Clin Cancer Res 2024; 43:209. [PMID: 39061080 PMCID: PMC11282757 DOI: 10.1186/s13046-024-03129-1] [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: 05/01/2024] [Accepted: 07/14/2024] [Indexed: 07/28/2024] Open
Abstract
Immune cell-derived extracellular vesicles (EV) affect tumor progression and hold promise for therapeutic applications. Eosinophils are major effectors in Th2-related pathologies recently implied in cancer. Here, we evaluated the anti-tumor activities of eosinophil-derived EV following activation with the alarmin IL-33. We demonstrate that IL-33-activated mouse and human eosinophils produce higher quantities of EV with respect to eosinophils stimulated with IL-5. Following incorporation of EV from IL-33-activated eosinophils (Eo33-EV), but not EV from IL-5-treated eosinophils (Eo5-EV), mouse and human tumor cells increased the expression of cyclin-dependent kinase inhibitor (CDKI)-related genes resulting in cell cycle arrest in G0/G1, reduced proliferation and inhibited tumor spheroid formation. Moreover, tumor cells incorporating Eo33-EV acquired an epithelial-like phenotype characterized by E-Cadherin up-regulation, N-Cadherin downregulation, reduced cell elongation and migratory extent in vitro, and impaired capacity to metastasize to lungs when injected in syngeneic mice. RNA sequencing revealed distinct mRNA signatures in Eo33-EV and Eo5-EV with increased presence of tumor suppressor genes and enrichment in pathways related to epithelial phenotypes and negative regulation of cellular processes in Eo33-EV compared to Eo5-EV. Our studies underscore novel IL-33-stimulated anticancer activities of eosinophils through EV-mediated reprogramming of tumor cells opening perspectives on the use of eosinophil-derived EV in cancer therapy.
Collapse
Affiliation(s)
| | - Caterina Antonucci
- Department of Oncology and Molecular Medicine, Istituto Superiore Di Sanità, Rome, Italy
| | - Cristiana Zanetti
- Department of Oncology and Molecular Medicine, Istituto Superiore Di Sanità, Rome, Italy
| | - Francesco Noto
- Department of Oncology and Molecular Medicine, Istituto Superiore Di Sanità, Rome, Italy
| | - Sara Andreone
- Department of Oncology and Molecular Medicine, Istituto Superiore Di Sanità, Rome, Italy
| | - Davide Vacca
- Tumor Immunology Unit, Department of Sciences for Health Promotion and Mother-Child Care "G. D'Alessandro", University of Palermo, Palermo, 90127, Italy
| | - Valentina Pellerito
- Tumor Immunology Unit, Department of Sciences for Health Promotion and Mother-Child Care "G. D'Alessandro", University of Palermo, Palermo, 90127, Italy
| | - Chiara Sicignano
- Department of Oncology and Molecular Medicine, Istituto Superiore Di Sanità, Rome, Italy
| | - Giuseppe Parrottino
- Department of Oncology and Molecular Medicine, Istituto Superiore Di Sanità, Rome, Italy
| | | | - Antonella Tinari
- National Center for Gender Medicine, Istituto Superiore Di Sanità, Rome, Italy
| | - Mario Falchi
- National AIDS Center, Istituto Superiore Di Sanità, Rome, Italy
| | - Adele De Ninno
- CNR-IFN Institute for Photonics and Nanotechnologies, Rome, Italy
| | - Luca Businaro
- CNR-IFN Institute for Photonics and Nanotechnologies, Rome, Italy
| | - Stefania Loffredo
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
- World Allergy Organization (WAO), Center of Excellence (CoE), Naples, 80131, Italy
- Institute of Experimental Endocrinology and Oncology, National Research Council (CNR), Naples, 80131, Italy
| | - Gilda Varricchi
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
- World Allergy Organization (WAO), Center of Excellence (CoE), Naples, 80131, Italy
- Institute of Experimental Endocrinology and Oncology, National Research Council (CNR), Naples, 80131, Italy
| | - Claudio Tripodo
- Tumor Immunology Unit, Department of Sciences for Health Promotion and Mother-Child Care "G. D'Alessandro", University of Palermo, Palermo, 90127, Italy
| | - Claudia Afferni
- National Center for Drug Research and Evaluation, Istituto Superiore Di Sanità, Rome, Italy
| | - Isabella Parolini
- Department of Oncology and Molecular Medicine, Istituto Superiore Di Sanità, Rome, Italy
- Laboratory of Molecular Medicine and DNA Repair, Department of Medicine, University of Udine, Udine, Italy
| | - Fabrizio Mattei
- Department of Oncology and Molecular Medicine, Istituto Superiore Di Sanità, Rome, Italy.
| | - Giovanna Schiavoni
- Department of Oncology and Molecular Medicine, Istituto Superiore Di Sanità, Rome, Italy.
| |
Collapse
|
8
|
Ou WT, Wan QX, Wu YB, Sun X, Li YL, Tang D, Zhang J, Li SS, Wang NY, Liu ZL, Wu JJ. Long Noncoding RNA PSMB8-AS1 Mediates the Tobacco-Carcinogen-Induced Transformation of a Human Bronchial Epithelial Cell Line by Regulating Cell Cycle. Chem Res Toxicol 2024; 37:957-967. [PMID: 38771128 DOI: 10.1021/acs.chemrestox.4c00025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Lung cancer is the main cause of cancer deaths around the world. Nitrosamine 4-(methyl nitrosamine)-1-(3-pyridyl)-1-butanone (NNK) is a tobacco-specific carcinogen of lung cancer. Abundant evidence implicates long noncoding RNAs (lncRNAs) in tumorigenesis. Yet, the effects and mechanisms of lncRNAs in NNK-induced carcinogenesis are still unclear. In this study, we discovered that NNK-induced transformed Beas-2B cells (Beas-2B-NNK) showed increased cell migration and proliferation while decreasing rates of apoptosis. RNA sequencing and differentially expressed lncRNAs analyses showed that lncRNA PSMB8-AS1 was obviously upregulated. Interestingly, silencing the lncRNA PSMB8-AS1 in Beas-2B-NNK cells reduced cell proliferation and migration and produced cell cycle arrest in the G2/M phase along with a decrease in CDK1 expression. Conclusively, our results demonstrate that lncRNA PSMB8-AS1 could promote the malignant characteristics of Beas-2B-NNK cells by regulating CDK1 and affecting the cell cycle, suggesting that it may supply a new prospective epigenetic mechanism for lung cancer.
Collapse
Affiliation(s)
- Wan-Ting Ou
- School of Public Health, Guangzhou Medical University, Guangzhou 511436, P. R. China
| | - Qiu-Xian Wan
- School of Public Health, Guangzhou Medical University, Guangzhou 511436, P. R. China
| | - Yi-Bo Wu
- School of Public Health, Guangzhou Medical University, Guangzhou 511436, P. R. China
| | - Xuan Sun
- School of Public Health, Guangzhou Medical University, Guangzhou 511436, P. R. China
| | - Yan-Li Li
- School of Public Health, Guangzhou Medical University, Guangzhou 511436, P. R. China
| | - Dan Tang
- School of Public Health, Guangzhou Medical University, Guangzhou 511436, P. R. China
| | - Jian Zhang
- School of Public Health, Guangzhou Medical University, Guangzhou 511436, P. R. China
| | - Sheng-Sheng Li
- School of Public Health, Guangzhou Medical University, Guangzhou 511436, P. R. China
| | - Nuo-Yan Wang
- School of Public Health, Guangzhou Medical University, Guangzhou 511436, P. R. China
| | - Zhuo-Lin Liu
- School of Public Health, Guangzhou Medical University, Guangzhou 511436, P. R. China
| | - Jian-Jun Wu
- School of Public Health, Guangzhou Medical University, Guangzhou 511436, P. R. China
- State Key Laboratory of Respiratory Disease, Institute for Chemical Carcinogenesis, Guangzhou Medical University, Guangzhou 511436, P. R. China
| |
Collapse
|
9
|
Alharbi HM. Exploring the Frontier of Biopolymer-Assisted Drug Delivery: Advancements, Clinical Applications, and Future Perspectives in Cancer Nanomedicine. Drug Des Devel Ther 2024; 18:2063-2087. [PMID: 38882042 PMCID: PMC11178098 DOI: 10.2147/dddt.s441325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 05/21/2024] [Indexed: 06/18/2024] Open
Abstract
The burgeoning global mortality rates attributed to cancer have precipitated a critical reassessment of conventional therapeutic modalities, most notably chemotherapy, due to their pronounced adverse effects. This reassessment has instigated a paradigmatic shift towards nanomedicine, with a particular emphasis on the potentialities of biopolymer-assisted drug delivery systems. Biopolymers, distinguished by their impeccable biocompatibility, versatility, and intrinsic biomimetic properties, are rapidly ascending as formidable vectors within the cancer theragnostic arena. This review endeavors to meticulously dissect the avant-garde methodologies central to biopolymer-based nanomedicine, exploring their synthesis, functional mechanisms, and subsequent clinical ramifications. A key focus of this analysis is the pioneering roles and efficacies of lipid-based, polysaccharide, and composite nano-carriers in enhancing drug delivery, notably amplifying the enhanced permeation and retention effect. This examination is further enriched by referencing flagship nano formulations that have received FDA endorsement, thereby underscoring the transformative potential and clinical viability of biopolymer-based nanomedicines. Furthermore, this discourse illuminates groundbreaking advancements in the realm of photodynamic therapy and elucidates the implications of advanced imaging techniques in live models. Conclusively, this review not only synthesizes current research trajectories but also delineates visionary pathways for the integration of cutting-edge biomaterials in cancer treatment. It charts a course for future explorations within the dynamic domain of biopolymer-nanomedicine, thereby contributing to a deeper understanding and enhanced application of these novel therapeutic strategies.
Collapse
Affiliation(s)
- Hanan M Alharbi
- Department of Pharmaceutical Sciences, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| |
Collapse
|
10
|
Lee HJ, Choi HJ, Jeong YJ, Na YH, Hong JT, Han JM, Hoe HS, Lim KH. Developing theragnostics for Alzheimer's disease: Insights from cancer treatment. Int J Biol Macromol 2024; 269:131925. [PMID: 38685540 DOI: 10.1016/j.ijbiomac.2024.131925] [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: 01/01/2024] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 05/02/2024]
Abstract
The prevalence of Alzheimer's disease (AD) and its associated economic and societal burdens are on the rise, but there are no curative treatments for AD. Interestingly, this neurodegenerative disease shares several biological and pathophysiological features with cancer, including cell-cycle dysregulation, angiogenesis, mitochondrial dysfunction, protein misfolding, and DNA damage. However, the genetic factors contributing to the overlap in biological processes between cancer and AD have not been actively studied. In this review, we discuss the shared biological features of cancer and AD, the molecular targets of anticancer drugs, and therapeutic approaches. First, we outline the common biological features of cancer and AD. Second, we describe several anticancer drugs, their molecular targets, and their effects on AD pathology. Finally, we discuss how protein-protein interactions (PPIs), receptor inhibition, immunotherapy, and gene therapy can be exploited for the cure and management of both cancer and AD. Collectively, this review provides insights for the development of AD theragnostics based on cancer drugs and molecular targets.
Collapse
Affiliation(s)
- Hyun-Ju Lee
- Korea Brain Research Institute (KBRI), 61, Cheomdan-ro, Dong-gu, Daegu 41062, Republic of Korea
| | - Hee-Jeong Choi
- Korea Brain Research Institute (KBRI), 61, Cheomdan-ro, Dong-gu, Daegu 41062, Republic of Korea
| | - Yoo Joo Jeong
- Korea Brain Research Institute (KBRI), 61, Cheomdan-ro, Dong-gu, Daegu 41062, Republic of Korea; Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science & Technology (DGIST), 333, Techno jungang-daero, Hyeonpung-eup, Dalseong-gun, Daegu 42988, Republic of Korea
| | - Yoon-Hee Na
- College of Pharmacy, Chungbuk National University, Cheongju-si 28160, Republic of Korea
| | - Jin Tae Hong
- College of Pharmacy, Chungbuk National University, Cheongju-si 28160, Republic of Korea
| | - Ji Min Han
- College of Pharmacy, Chungbuk National University, Cheongju-si 28160, Republic of Korea.
| | - Hyang-Sook Hoe
- Korea Brain Research Institute (KBRI), 61, Cheomdan-ro, Dong-gu, Daegu 41062, Republic of Korea; Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science & Technology (DGIST), 333, Techno jungang-daero, Hyeonpung-eup, Dalseong-gun, Daegu 42988, Republic of Korea.
| | - Key-Hwan Lim
- College of Pharmacy, Chungbuk National University, Cheongju-si 28160, Republic of Korea.
| |
Collapse
|
11
|
Conway PJ, Dao J, Kovalskyy D, Mahadevan D, Dray E. Polyploidy in Cancer: Causal Mechanisms, Cancer-Specific Consequences, and Emerging Treatments. Mol Cancer Ther 2024; 23:638-647. [PMID: 38315992 PMCID: PMC11174144 DOI: 10.1158/1535-7163.mct-23-0578] [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: 08/31/2023] [Revised: 11/19/2023] [Accepted: 01/30/2024] [Indexed: 02/07/2024]
Abstract
Drug resistance is the major determinant for metastatic disease and fatalities, across all cancers. Depending on the tissue of origin and the therapeutic course, a variety of biological mechanisms can support and sustain drug resistance. Although genetic mutations and gene silencing through epigenetic mechanisms are major culprits in targeted therapy, drug efflux and polyploidization are more global mechanisms that prevail in a broad range of pathologies, in response to a variety of treatments. There is an unmet need to identify patients at risk for polyploidy, understand the mechanisms underlying polyploidization, and to develop strategies to predict, limit, and reverse polyploidy thus enhancing efficacy of standard-of-care therapy that improve better outcomes. This literature review provides an overview of polyploidy in cancer and offers perspective on patient monitoring and actionable therapy.
Collapse
Affiliation(s)
- Patrick J Conway
- Mays Cancer Center, University of Texas Health San Antonio, San Antonio, Texas
- Department of Molecular Immunology & Microbiology, University of Texas Health San Antonio, San Antonio, Texas
| | - Jonathan Dao
- Mays Cancer Center, University of Texas Health San Antonio, San Antonio, Texas
- Long School of Medicine, University of Texas Health San Antonio, San Antonio, Texas
| | - Dmytro Kovalskyy
- Greehey Children's Cancer Research Institute, University of Texas Health San Antonio, San Antonio, Texas
| | - Daruka Mahadevan
- Mays Cancer Center, University of Texas Health San Antonio, San Antonio, Texas
- Department of Molecular Immunology & Microbiology, University of Texas Health San Antonio, San Antonio, Texas
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, Texas
| | - Eloise Dray
- Long School of Medicine, University of Texas Health San Antonio, San Antonio, Texas
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, Texas
| |
Collapse
|
12
|
Bansal S, Li Y, Bansal S, Klotzbier W, Singh B, Jayatilake M, Sridharan V, Fernández JA, Griffin JH, Weiler H, Boerma M, Cheema AK. Genetic Upregulation of Activated Protein C Mitigates Delayed Effects of Acute Radiation Exposure in the Mouse Plasma. Metabolites 2024; 14:245. [PMID: 38786722 PMCID: PMC11122730 DOI: 10.3390/metabo14050245] [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: 03/27/2024] [Revised: 04/17/2024] [Accepted: 04/22/2024] [Indexed: 05/25/2024] Open
Abstract
Exposure to ionizing radiation, accidental or intentional, may lead to delayed effects of acute radiation exposure (DEARE) that manifest as injury to organ systems, including the kidney, heart, and brain. This study examines the role of activated protein C (APC), a known mitigator of radiation-induced early toxicity, in long-term plasma metabolite and lipid panels that may be associated with DEARE in APCHi mice. The APCHi mouse model used in the study was developed in a C57BL/6N background, expressing the D168F/N173K mouse analog of the hyper-activatable human D167F/D172K protein C variant. This modification enables increased circulating APC levels throughout the mouse's lifetime. Male and female cohorts of C57BL/6N wild-type and APCHi transgenic mice were exposed to 9.5 Gy γ-rays with their hind legs shielded to allow long-term survival that is necessary to monitor DEARE, and plasma was collected at 6 months for LC-MS-based metabolomics and lipidomics. We observed significant dyslipidemia, indicative of inflammatory phenotype, upon radiation exposure. Additionally, observance of several other metabolic dysregulations was suggestive of gut damage, perturbations in TriCarboxylic Acid (TCA) and urea cycles, and arginine metabolism. We also observed gender- and genotype-modulated metabolic perturbations post radiation exposure. The APCHi mice showed near-normal abundance for several lipids. Moreover, restoration of plasma levels of some metabolites, including amino acids, citric acid, and hypoxanthine, in APCHi mice is indicative of APC-mediated protection from radiation injuries. With the help of these findings, the role of APC in plasma molecular events after acute γ-radiation exposure in a gender-specific manner can be established for the first time.
Collapse
Affiliation(s)
- Shivani Bansal
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA; (S.B.); (S.B.); (W.K.); (B.S.); (M.J.)
| | - Yaoxiang Li
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA; (S.B.); (S.B.); (W.K.); (B.S.); (M.J.)
| | - Sunil Bansal
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA; (S.B.); (S.B.); (W.K.); (B.S.); (M.J.)
| | - William Klotzbier
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA; (S.B.); (S.B.); (W.K.); (B.S.); (M.J.)
| | - Baldev Singh
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA; (S.B.); (S.B.); (W.K.); (B.S.); (M.J.)
| | - Meth Jayatilake
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA; (S.B.); (S.B.); (W.K.); (B.S.); (M.J.)
| | - Vijayalakshmi Sridharan
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (V.S.); (M.B.)
| | - José A. Fernández
- Department of Molecular Medicine, Scripps Research Institute, La Jolla, CA 92037, USA; (J.A.F.); (J.H.G.)
| | - John H. Griffin
- Department of Molecular Medicine, Scripps Research Institute, La Jolla, CA 92037, USA; (J.A.F.); (J.H.G.)
| | - Hartmut Weiler
- Versiti Blood Research Institute, Medical College of Wisconsin, Milwaukee, WI 53233, USA;
| | - Marjan Boerma
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (V.S.); (M.B.)
| | - Amrita K. Cheema
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA; (S.B.); (S.B.); (W.K.); (B.S.); (M.J.)
- Departments of Biochemistry, Molecular and Cellular Biology, Georgetown University Medical Center, Washington, DC 20057, USA
| |
Collapse
|
13
|
Carvalho L, de Lima FP, Cerqueira M, Silva A, Pontes O, Oliveira-Pinto S, Guerreiro S, Costa MD, Granja S, Maciel P, Longatto-Filho A, Baltazar F, Proença F, Costa M. In vitro and in vivo evaluation of novel chromeno[2,3- d]pyrimidinones as therapeutic agents for triple negative breast cancer. RSC Med Chem 2024; 15:1362-1380. [PMID: 38665823 PMCID: PMC11042168 DOI: 10.1039/d3md00682d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 02/20/2024] [Indexed: 04/28/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer, and the limited therapeutic options show poor efficacy in patients, associated to severe side effects and development of resistance. Considering that chromene-based scaffolds proved to be attractive candidates for cancer therapy, herein we prepared new chromeno[2,3-d]pyrimidinone derivatives by a simple two step procedure, starting from the reaction of cyanoacetamide and a salicylaldehyde. A cell viability screening in several breast cancer cell lines allowed to identify two promising compounds with IC50 values in the low micromolar range for TNBC cells. These chromenes inhibited cell proliferation, induced cell cycle arrest and triggered cell death through apoptosis. In vivo studies revealed a safe profile in invertebrate and vertebrate animal models and confirmed their capacity to inhibit tumor growth in the CAM model, inducing significant tumor regression after 4 days of treatment. The two compounds identified in this study are promising drug candidates for TNBC treatment and valuable hits for future optimization, using the versatile synthetic platform that was developed.
Collapse
Affiliation(s)
- Luísa Carvalho
- Life and Health Sciences Research Institute (ICVS), University of Minho Campus of Gualtar Braga Portugal
- ICVS/3B's - PT Government Associate Laboratory Braga/Guimarães Portugal
| | | | - Mónica Cerqueira
- Life and Health Sciences Research Institute (ICVS), University of Minho Campus of Gualtar Braga Portugal
- ICVS/3B's - PT Government Associate Laboratory Braga/Guimarães Portugal
| | - Ana Silva
- Life and Health Sciences Research Institute (ICVS), University of Minho Campus of Gualtar Braga Portugal
- ICVS/3B's - PT Government Associate Laboratory Braga/Guimarães Portugal
| | - Olívia Pontes
- Life and Health Sciences Research Institute (ICVS), University of Minho Campus of Gualtar Braga Portugal
- ICVS/3B's - PT Government Associate Laboratory Braga/Guimarães Portugal
| | - Sofia Oliveira-Pinto
- Life and Health Sciences Research Institute (ICVS), University of Minho Campus of Gualtar Braga Portugal
- ICVS/3B's - PT Government Associate Laboratory Braga/Guimarães Portugal
| | - Sara Guerreiro
- Life and Health Sciences Research Institute (ICVS), University of Minho Campus of Gualtar Braga Portugal
- ICVS/3B's - PT Government Associate Laboratory Braga/Guimarães Portugal
- Department of Pathological, Cytological and Thanatological Anatomy, School of Health, Polytechnic Institute of Porto 4200-072 Porto Portugal
| | - Marta D Costa
- Life and Health Sciences Research Institute (ICVS), University of Minho Campus of Gualtar Braga Portugal
- ICVS/3B's - PT Government Associate Laboratory Braga/Guimarães Portugal
| | - Sara Granja
- Life and Health Sciences Research Institute (ICVS), University of Minho Campus of Gualtar Braga Portugal
- ICVS/3B's - PT Government Associate Laboratory Braga/Guimarães Portugal
- Department of Pathological, Cytological and Thanatological Anatomy, School of Health, Polytechnic Institute of Porto 4200-072 Porto Portugal
| | - Patrícia Maciel
- Life and Health Sciences Research Institute (ICVS), University of Minho Campus of Gualtar Braga Portugal
- ICVS/3B's - PT Government Associate Laboratory Braga/Guimarães Portugal
| | - Adhemar Longatto-Filho
- Life and Health Sciences Research Institute (ICVS), University of Minho Campus of Gualtar Braga Portugal
- ICVS/3B's - PT Government Associate Laboratory Braga/Guimarães Portugal
- Molecular Oncology Research Center, Barretos Cancer Hospital São Paulo Brazil
- Medical Laboratory of Medical Investigation (LIM) 14, Department of Pathology, Medical School, University of São Paulo São Paulo Brazil
| | - Fátima Baltazar
- Life and Health Sciences Research Institute (ICVS), University of Minho Campus of Gualtar Braga Portugal
- ICVS/3B's - PT Government Associate Laboratory Braga/Guimarães Portugal
| | - Fernanda Proença
- Department of Chemistry, University of Minho Campus of Gualtar Braga Portugal
| | - Marta Costa
- Life and Health Sciences Research Institute (ICVS), University of Minho Campus of Gualtar Braga Portugal
- ICVS/3B's - PT Government Associate Laboratory Braga/Guimarães Portugal
| |
Collapse
|
14
|
Enlund S, Sinha I, Neofytou C, Amor AR, Papadakis K, Nilsson A, Jiang Q, Hermanson O, Holm F. The CNS microenvironment promotes leukemia cell survival by disrupting tumor suppression and cell cycle regulation in pediatric T-cell acute lymphoblastic leukemia. Exp Cell Res 2024; 437:114015. [PMID: 38561062 DOI: 10.1016/j.yexcr.2024.114015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 03/21/2024] [Accepted: 03/23/2024] [Indexed: 04/04/2024]
Abstract
A major obstacle in improving survival in pediatric T-cell acute lymphoblastic leukemia is understanding how to predict and treat leukemia relapse in the CNS. Leukemia cells are capable of infiltrating and residing within the CNS, primarily the leptomeninges, where they interact with the microenvironment and remain sheltered from systemic treatment. These cells can survive in the CNS, by hijacking the microenvironment and disrupting normal functions, thus promoting malignant transformation. While the protective effects of the bone marrow niche have been widely studied, the mechanisms behind leukemia infiltration into the CNS and the role of the CNS niche in leukemia cell survival remain unknown. We identified a dysregulated gene expression profile in CNS infiltrated T-ALL and CNS relapse, promoting cell survival, chemoresistance, and disease progression. Furthermore, we discovered that interactions between leukemia cells and human meningeal cells induced epigenetic alterations, such as changes in histone modifications, including H3K36me3 levels. These findings are a step towards understanding the molecular mechanisms promoting leukemia cell survival in the CNS microenvironment. Our results highlight genetic and epigenetic alterations induced by interactions between leukemia cells and the CNS niche, which could potentially be utilized as biomarkers to predict CNS infiltration and CNS relapse.
Collapse
Affiliation(s)
- Sabina Enlund
- Deparment of Women's and Children's Health, Division of Pediatric Oncology and Surgery, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Indranil Sinha
- Deparment of Women's and Children's Health, Division of Pediatric Oncology and Surgery, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Christina Neofytou
- Department of Neuroscience, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Amanda Ramilo Amor
- Deparment of Women's and Children's Health, Division of Pediatric Oncology and Surgery, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Konstantinos Papadakis
- Deparment of Women's and Children's Health, Division of Pediatric Oncology and Surgery, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Anna Nilsson
- Deparment of Women's and Children's Health, Division of Pediatric Oncology and Surgery, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Qingfei Jiang
- Division of Regenerative Medicine, Department of Medicine, Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Ola Hermanson
- Department of Neuroscience, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Frida Holm
- Deparment of Women's and Children's Health, Division of Pediatric Oncology and Surgery, Karolinska Institutet, 171 77, Stockholm, Sweden.
| |
Collapse
|
15
|
Pham TM, Ahmed M, Lai TH, Bahar ME, Hwang JS, Maulidi RF, Ngo QN, Kim DR. Regulation of Cell Cycle Progression through RB Phosphorylation by Nilotinib and AT-9283 in Human Melanoma A375P Cells. Int J Mol Sci 2024; 25:2956. [PMID: 38474202 DOI: 10.3390/ijms25052956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 02/20/2024] [Accepted: 02/21/2024] [Indexed: 03/14/2024] Open
Abstract
BCR-ABL tyrosine kinase inhibitors are commonly employed for the treatment of chronic myeloid leukemia, yet their impact on human malignant melanoma remains uncertain. In this study, we delved into the underlying mechanisms of specific BCR-ABL tyrosine kinase inhibitors (imatinib, nilotinib, ZM-306416, and AT-9283) in human melanoma A375P cells. We first evaluated the influence of these inhibitors on cell growth using cell proliferation and wound-healing assays. Subsequently, we scrutinized cell cycle regulation in drug-treated A375P cells using flow cytometry and Western blot assays. Notably, imatinib, nilotinib, ZM-306416, and AT-9283 significantly reduced cell proliferation and migration in A375P cells. In particular, nilotinib and AT-9283 impeded the G1/S transition of the cell cycle by down-regulating cell cycle-associated proteins, including cyclin E, cyclin A, and CDK2. Moreover, these inhibitors reduced RB phosphorylation, subsequently inhibiting E2F transcriptional activity. Consequently, the expression of the E2F target genes (CCNA2, CCNE1, POLA1, and TK-1) was markedly suppressed in nilotinib and AT9283-treated A375P cells. In summary, our findings suggest that BCR-ABL tyrosine kinase inhibitors may regulate the G1-to-S transition in human melanoma A375P cells by modulating the RB-E2F complex.
Collapse
Affiliation(s)
- Trang Minh Pham
- Department of Biochemistry and Convergence Medical Science, Institute of Medical Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea
| | - Mahmoud Ahmed
- Department of Biochemistry and Convergence Medical Science, Institute of Medical Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea
| | - Trang Huyen Lai
- Department of Biochemistry and Convergence Medical Science, Institute of Medical Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea
| | - Md Entaz Bahar
- Department of Biochemistry and Convergence Medical Science, Institute of Medical Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea
| | - Jin Seok Hwang
- Department of Biochemistry and Convergence Medical Science, Institute of Medical Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea
| | - Rizi Firman Maulidi
- Department of Biochemistry and Convergence Medical Science, Institute of Medical Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea
| | - Quang Nhat Ngo
- Department of Biochemistry and Convergence Medical Science, Institute of Medical Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea
| | - Deok Ryong Kim
- Department of Biochemistry and Convergence Medical Science, Institute of Medical Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea
| |
Collapse
|
16
|
Peña-Flores JA, Muela-Campos D, Guzmán-Medrano R, Enríquez-Espinoza D, González-Alvarado K. Functional Relevance of Extracellular Vesicle-Derived Long Non-Coding and Circular RNAs in Cancer Angiogenesis. Noncoding RNA 2024; 10:12. [PMID: 38392967 PMCID: PMC10891584 DOI: 10.3390/ncrna10010012] [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: 12/19/2023] [Revised: 02/02/2024] [Accepted: 02/04/2024] [Indexed: 02/25/2024] Open
Abstract
Extracellular vesicles (EVs) are defined as subcellular structures limited by a bilayer lipid membrane that function as important intercellular communication by transporting active biomolecules, such as proteins, amino acids, metabolites, and nucleic acids, including long non-coding RNAs (lncRNAs). These cargos can effectively be delivered to target cells and induce a highly variable response. LncRNAs are functional RNAs composed of at least 200 nucleotides that do not code for proteins. Nowadays, lncRNAs and circRNAs are known to play crucial roles in many biological processes, including a plethora of diseases including cancer. Growing evidence shows an active presence of lnc- and circRNAs in EVs, generating downstream responses that ultimately affect cancer progression by many mechanisms, including angiogenesis. Moreover, many studies have revealed that some tumor cells promote angiogenesis by secreting EVs, which endothelial cells can take up to induce new vessel formation. In this review, we aim to summarize the bioactive roles of EVs with lnc- and circRNAs as cargo and their effect on cancer angiogenesis. Also, we discuss future clinical strategies for cancer treatment based on current knowledge of circ- and lncRNA-EVs.
Collapse
Affiliation(s)
- José A. Peña-Flores
- Doctoral Program in Biomedical and Stomatological Sciences, Faculty of Dentistry, Autonomous University of Chihuahua, Chihuahua 31000, Mexico; (D.M.-C.); (R.G.-M.); (D.E.-E.); (K.G.-A.)
| | | | | | | | | |
Collapse
|
17
|
Zhang S, Wang Y, Luo D, Cheng Z, Zeng Q, Wang G, Chen M, Zhang S, Luo P. Pirfenidone inhibits TGF-β1-induced metabolic reprogramming during epithelial-mesenchymal transition in non-small cell lung cancer. J Cell Mol Med 2024; 28:e18059. [PMID: 38140828 PMCID: PMC10844763 DOI: 10.1111/jcmm.18059] [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: 04/13/2023] [Revised: 11/13/2023] [Accepted: 11/16/2023] [Indexed: 12/24/2023] Open
Abstract
Metastasis is an important contributor to increased mortality rates in non-small cell lung cancer (NSCLC). The TGF-β signalling pathway plays a crucial role in facilitating tumour metastasis through epithelial-mesenchymal transition (EMT). Glycolysis, a key metabolic process, is strongly correlated with NSCLC metastasis. Pirfenidone (PFD) has been shown to safely and effectively inhibit TGF-β1 in patients with lung diseases. Furthermore, TGF-β1 and glycolysis demonstrate an interdependent relationship within the tumour microenvironment. Our previous study demonstrated that PFD effectively inhibited glycolysis in NSCLC cells, prompting further investigation into its potential antitumour effects in this context. Therefore, the present study aims to investigate the potential antitumour effect of PFD in NSCLC and explore the relationship among TGF-β1, glycolysis and EMT through further experimentation. The antitumour effects of PFD were evaluated using five different NSCLC cell lines and a xenograft tumour model. Notably, PFD demonstrated a significant antitumour effect specifically in highly glycolytic H1299 cells. To elucidate the underlying mechanism, we compared the efficacy of PFD after pretreatment with either TGF-β1 or a TGF-β receptor inhibitor (LY2109761). The energy metabolomics analysis of tumour tissue demonstrated that PFD, a chemosensitizing agent, reduced lactate and ATP production, thereby inhibiting glycolysis and exerting synergistic antineoplastic effects. Additionally, PFD combined with cisplatin targeted TGF-β1 to inhibit glycolysis during EMT and enhanced the chemosensitization of A549 and H1299 cells. The magnitude of the anticancer effect exhibited by PFD was intricately linked to its metabolic properties.
Collapse
Affiliation(s)
- Shuling Zhang
- Guizhou Medical UniversityGuiyangChina
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of EducationSchool of Public HealthGuiyangChina
- State Key Laboratory of Functions and Applications of Medicinal PlantsGuizhou Medical UniversityGuiyangChina
- Guizhou Provincial Engineering Research Center of Food Nutrition and HealthGuizhou Medical UniversityGuiyangChina
| | - Yuanmei Wang
- Guizhou Medical UniversityGuiyangChina
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of EducationSchool of Public HealthGuiyangChina
| | | | | | - Qibing Zeng
- Guizhou Medical UniversityGuiyangChina
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of EducationSchool of Public HealthGuiyangChina
- State Key Laboratory of Functions and Applications of Medicinal PlantsGuizhou Medical UniversityGuiyangChina
- Guizhou Provincial Engineering Research Center of Food Nutrition and HealthGuizhou Medical UniversityGuiyangChina
| | - Guoze Wang
- Guizhou Medical UniversityGuiyangChina
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of EducationSchool of Public HealthGuiyangChina
- State Key Laboratory of Functions and Applications of Medicinal PlantsGuizhou Medical UniversityGuiyangChina
- Guizhou Provincial Engineering Research Center of Food Nutrition and HealthGuizhou Medical UniversityGuiyangChina
| | | | - Shuai Zhang
- Guizhou Medical UniversityGuiyangChina
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of EducationSchool of Public HealthGuiyangChina
| | - Peng Luo
- Guizhou Medical UniversityGuiyangChina
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of EducationSchool of Public HealthGuiyangChina
- State Key Laboratory of Functions and Applications of Medicinal PlantsGuizhou Medical UniversityGuiyangChina
- Guizhou Provincial Engineering Research Center of Food Nutrition and HealthGuizhou Medical UniversityGuiyangChina
| |
Collapse
|
18
|
Sun Y, Wu Q, Fu Q, Cong H, Shen Y, Yu B, Hu H. Reactive oxygen species-responsive polyprodrug micelles deliver cell cycle regulators for chemosensitization. Talanta 2024; 267:125242. [PMID: 37801926 DOI: 10.1016/j.talanta.2023.125242] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 09/18/2023] [Accepted: 09/22/2023] [Indexed: 10/08/2023]
Abstract
Combination chemotherapy is a common strategy to enhance treatment efficacy and avoid multidrug resistance (MDR) in clinical practice. However, it is difficult to ensure the co-enrichment and reasonable ratio of synergistic drugs in the lesion site after intravenous administration. Integrating synergistic drugs into a nanocarrier can improve drug stability, targeting, drug loading, and importantly, ensure that synergistic drugs work at one destination. This study uses 10-hydroxycamptothecin (HCPT) to construct a polymeric prodrug micelle, and the demethylcantharidin (DMC) is proportionally encapsulated within the micelle. Triggered by reactive oxygen species (ROS), HCPT and DMC were released simultaneously from the co-delivery platform in tumor cells. DMC promotes abnormal cell division by inhibiting the synthesis of the cell cycle checkpoint kinase Protein phosphatase 2A (PP2A), leading to increased cell vulnerability to DNA damage, disordered replication, and death. The co-delivery platform exhibited satisfactory biosafety and antitumor efficacy in vivo. The proposed co-delivery platform may provide a valuable reference for the translation of clinical combination chemotherapy regimens into nano-drug delivery systems.
Collapse
Affiliation(s)
- Ying Sun
- Institute of Biomedical Materials and Engineering, College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Qingdao University, Qingdao, 266071, China
| | - Qimeng Wu
- Institute of Biomedical Materials and Engineering, College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Qingdao University, Qingdao, 266071, China
| | - Quanyou Fu
- Department of Vascular Surgery, The Affiliated Hospital of Qingdao University, Qingdao, 266071, China
| | - Hailin Cong
- Institute of Biomedical Materials and Engineering, College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Qingdao University, Qingdao, 266071, China; School of Materials Science and Engineering, Shandong University of Technology, Zibo, 255000, China
| | - Youqing Shen
- Institute of Biomedical Materials and Engineering, College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Qingdao University, Qingdao, 266071, China; Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Bing Yu
- Institute of Biomedical Materials and Engineering, College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Qingdao University, Qingdao, 266071, China; State Key Laboratory of Bio-Fibers and Eco-Textiles, Qingdao University, Qingdao, 266071, China.
| | - Hao Hu
- Institute of Biomedical Materials and Engineering, College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Qingdao University, Qingdao, 266071, China.
| |
Collapse
|
19
|
Cavalcante IL, Silva Barros CCD, Colares DF, Cruz VMS, de Andrade BAB, Nonaka CFW, Rabenhorst SHB, Cavalcante RB. BubR1 and cyclin B1 immunoexpression in pleomorphic adenoma and polymorphous adenocarcinoma of minor salivary glands. Pathol Res Pract 2024; 253:154961. [PMID: 38043194 DOI: 10.1016/j.prp.2023.154961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 11/14/2023] [Accepted: 11/16/2023] [Indexed: 12/05/2023]
Abstract
The immunoexpression of BubR1 and cyclin B1 in pleomorphic adenoma (PA) and polymorphic adenocarcinoma (PAC) in minor salivary glands is poorly studied. Thus, a retrospective and observational study was performed to provide a better understanding of the role and immunopositivity patterns of these proteins in these lesions. Sixteen cases of PA and 16 cases of PAC were selected. Parenchyma cells were submitted to quantitative immunohistochemical analysis through the labeling index. Cytoplasmic immunoexpression of BubR1 was observed in neoplastic cells from all analyzed PA and PAC cases. All PA cases and 93.7% of PAC exhibited nuclear immunoexpression of BubR1. Higher cytoplasmic and nuclear immunoexpression of BubR1 was observed in PAC (p = 0.001 and p = 0.122, respectively). Cytoplasmic immunoexpression of cyclin B1 was observed in all cases of PA and PAC, with a higher labeling index in the latter (p < 0.001). There was a significant positive correlation between nuclear and cytoplasmic BubR1 immunoexpressions (p < 0.001) in PA and a significant negative correlation between BubR1 and cyclin B1 cytoplasmic immunoexpressions (p = 0.014) in PAC. The higher cytoplasmic and nuclear immunoexpression of BubR1 in PACs suggests the continuous maintenance of neoplastic cells in the cell cycle and migration. Higher immunoexpression of cyclin B1 supports this lesion's enhanced proliferative and migration ability.
Collapse
Affiliation(s)
- Israel Leal Cavalcante
- Oral Pathology Section, Department of Dentistry, University of Fortaleza, Fortaleza, CE, Brazil; Department of Oral Diagnosis and Pathology, School of Dentistry, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| | - Caio César da Silva Barros
- Postgraduate Program in Dental Sciences, Oral Pathology and Medicine, Department of Dentistry, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Débora Frota Colares
- Postgraduate Program in Dental Sciences, Oral Pathology and Medicine, Department of Dentistry, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Vitória Maria Sousa Cruz
- Oral Pathology Section, Department of Oral Diagnosis, Piracicaba Dental School, University of Campinas, Piracicaba, SP, Brazil
| | | | | | - Silvia Helena Barem Rabenhorst
- Postgraduate Program in Biological Sciences, Molecular Genetics Laboratory, Department of Pathology and Forensic Medicine, School of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil
| | | |
Collapse
|
20
|
Vara-Ciruelos D, Malumbres M. Cross Talk Between Metabolism and the Cell Division Cycle. Methods Mol Biol 2024; 2740:141-154. [PMID: 38393474 DOI: 10.1007/978-1-0716-3557-5_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] [Indexed: 02/25/2024]
Abstract
Cell division requires a massive rewiring of cellular pathways, including molecular routes involved in providing energy for cell survival and functionality. The energetic requirements and the metabolic opportunities for generating energy change during the different phases of the cell cycle and how these processes are connected is still poorly understood. This chapter discusses basic concepts for a coordinated analysis of cell cycle progression and metabolism and provides specific protocols for studying these two connected processes in mammalian cells.
Collapse
Affiliation(s)
- Diana Vara-Ciruelos
- Cell Division and Cancer group, Spanish National Cancer Research Center (CNIO), Madrid, Spain
| | - Marcos Malumbres
- Cell Division and Cancer group, Spanish National Cancer Research Center (CNIO), Madrid, Spain.
- Cancer Cell Cycle group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain.
- ICREA, Barcelona, Spain.
| |
Collapse
|
21
|
Xu FX, Wu R, Hu K, Fu D. Measuring Drug Response with Single-Cell Growth Rate Quantification. Anal Chem 2023; 95:18114-18121. [PMID: 38016067 PMCID: PMC11016461 DOI: 10.1021/acs.analchem.3c03434] [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: 11/30/2023]
Abstract
Intratumoral heterogeneity is a substantial cause of drug resistance development during chemotherapy or other drug treatments for cancer. Therefore, monitoring and measuring cell exposure and response to drugs at the single-cell level are crucial. Previous research suggested that the single-cell growth rate can be used to investigate drug-cell interactions. However, currently established methods for quantifying single-cell growth are limited to isolated or monolayer cells. Here, we introduce a technique that accurately measures both 2D and 3D cell growth rates using label-free ratiometric stimulated Raman scattering (SRS) microscopy. We use deuterated amino acids, leucine, isoleucine, and valine, as tracers and measure the C-D SRS signal from deuterium-labeled proteins and the C-H SRS signal from unlabeled proteins simultaneously to determine the cell growth rate at the single-cell level. The technique offers single-cell level drug sensitivity measurement with a shorter turnaround time (within 12 h) than most traditional assays. The submicrometer resolution of the imaging technique allows us to examine the effects of chemotherapeutic drugs, including kinase inhibitors, mitotic inhibitors, and topoisomerase II inhibitors, on both the cell growth rate and morphology. The capability of quantifying 3D cell growth rates provides insight into a deeper understanding of the cell-drug interaction in the actual tumor environment.
Collapse
Affiliation(s)
- Fiona Xi Xu
- Department of Chemistry, University of Washington, Seattle, WA 98195, United States
| | - Ruibing Wu
- Department of Chemistry, University of Washington, Seattle, WA 98195, United States
| | - Kailun Hu
- Department of Chemistry, University of Washington, Seattle, WA 98195, United States
| | - Dan Fu
- Department of Chemistry, University of Washington, Seattle, WA 98195, United States
| |
Collapse
|
22
|
Sun R, Xu H, Liu F, Zhou B, Li M, Sun X. Unveiling the intricate causal nexus between pancreatic cancer and peripheral metabolites through a comprehensive bidirectional two-sample Mendelian randomization analysis. Front Mol Biosci 2023; 10:1279157. [PMID: 37954977 PMCID: PMC10634252 DOI: 10.3389/fmolb.2023.1279157] [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: 08/17/2023] [Accepted: 10/16/2023] [Indexed: 11/14/2023] Open
Abstract
Aim: Pancreatic cancer (PC) is a devastating malignancy characterized by its aggressive nature and poor prognosis. However, the relationship of PC with peripheral metabolites remains not fully investigated. The study aimed to explore the causal linkage between PC and peripheral metabolite profiles. Methods: Employing publicly accessible genome-wide association studies (GWAS) data, we conducted a bidirectional two-sample Mendelian randomization (MR) analysis. The primary analysis employed the inverse-variance weighted (IVW) method. To address potential concerns about horizontal pleiotropy, we also employed supplementary methods such as maximum likelihood, weighted median, MR-Egger regression, and MR pleiotropy residual sum and outlier (MR-PRESSO). Results: We ascertained 20 genetically determined peripheral metabolites with causal linkages to PC while high-density lipoprotein (HDL) and very low-density lipoprotein (VLDL) particles accounted for the vast majority. Specifically, HDL particles exhibited an elevated PC risk while VLDL particles displayed an opposing pattern. The converse MR analysis underscored a notable alteration in 17 peripheral metabolites due to PC, including branch chain amino acids and derivatives of glycerophospholipid. Cross-referencing the bidirectional MR results revealed a reciprocal causation of PC and X-02269 which might form a self-perpetuating loop in PC development. Additionally, 1-arachidonoylglycerophosphocholine indicated a reduced PC risk and an increase under PC influence, possibly serving as a negative feedback regulator. Conclusion: Our findings suggest a complex interplay between pancreatic cancer and peripheral metabolites, with potential implications for understanding the etiology of pancreatic cancer and identifying novel early diagnosis and therapeutic targets. Moreover, X-02269 may hold a pivotal role in PC onset and progression.
Collapse
Affiliation(s)
| | | | | | | | - Minli Li
- Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xiangdong Sun
- Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
23
|
Bhattacharya T, Preetam S, Ghosh B, Chakrabarti T, Chakrabarti P, Samal SK, Thorat N. Advancement in Biopolymer Assisted Cancer Theranostics. ACS APPLIED BIO MATERIALS 2023; 6:3959-3983. [PMID: 37699558 PMCID: PMC10583232 DOI: 10.1021/acsabm.3c00458] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 08/30/2023] [Indexed: 09/14/2023]
Abstract
Applications of nanotechnology have increased the importance of research and nanocarriers, which have revolutionized the method of drug delivery to treat several diseases, including cancer, in the past few years. Cancer, one of the world's fatal diseases, has drawn scientists' attention for its multidrug resistance to various chemotherapeutic drugs. To minimize the side effects of chemotherapeutic agents on healthy cells and to develop technological advancement in drug delivery systems, scientists have developed an alternative approach to delivering chemotherapeutic drugs at the targeted site by integrating it inside the nanocarriers like synthetic polymers, nanotubes, micelles, dendrimers, magnetic nanoparticles, quantum dots (QDs), lipid nanoparticles, nano-biopolymeric substances, etc., which has shown promising results in both preclinical and clinical trials of cancer management. Besides that, nanocarriers, especially biopolymeric nanoparticles, have received much attention from researchers due to their cost-effectiveness, biodegradability, treatment efficacy, and ability to target drug delivery by crossing the blood-brain barrier. This review emphasizes the fabrication processes, the therapeutic and theragnostic applications, and the importance of different biopolymeric nanocarriers in targeting cancer both in vitro and in vivo, which conclude with the challenges and opportunities of future exploration using biopolymeric nanocarriers in onco-therapy with improved availability and reduced toxicity.
Collapse
Affiliation(s)
- Tanima Bhattacharya
- Department
of Food and Nutrition, College of Human Ecology, Kyung Hee University, 26 Kyunghee-daero, Dongdaemun-gu, Seoul 02447, Republic
of Korea
- Nondestructive
Bio-Sensing Laboratory, Dept. of Biosystems Machinery Engineering,
College of Agriculture and Life Science, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Subham Preetam
- Centre
for Biotechnology, Siksha O Anusandhan (Deemed
to be University), Bhubaneswar 751024, Odisha, India
- Daegu
Gyeongbuk Institute of Science & Technology (DGIST), Daegu 42988, Republic of Korea
| | - Basab Ghosh
- KIIT
School of Biotechnology, Kalinga Institute
of Industrial Technology (KIIT-DU), Bhubaneswar 751024, Odisha, India
| | - Tulika Chakrabarti
- Department
of Chemistry, Sir Padampat Singhania University, Bhatewar, Udaipur 313601, Rajasthan, India
| | | | - Shailesh Kumar Samal
- Section of
Immunology and Chronic Disease, Institute of Environmental Medicine, Karolinska Institutet, Stockholm 171 77, Sweden
| | - Nanasaheb Thorat
- Nuffield
Department of Women’s & Reproductive Health, Medical Science
Division, John Radcliffe Hospital University
of Oxford, Oxford OX3 9DU, United Kingdom
- Department
of Physics, Bernal Institute and Limerick Digital Cancer Research
Centre (LDCRC), University of Limerick, Castletroy, Limerick V94T9PX, Ireland
| |
Collapse
|
24
|
Tang X, Tang Q, Li S, Li M, Yang T. IGF2BP2 acts as a m 6A modification regulator in laryngeal squamous cell carcinoma through facilitating CDK6 mRNA stabilization. Cell Death Discov 2023; 9:371. [PMID: 37816718 PMCID: PMC10564923 DOI: 10.1038/s41420-023-01669-7] [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: 07/28/2023] [Revised: 09/22/2023] [Accepted: 09/28/2023] [Indexed: 10/12/2023] Open
Abstract
Laryngeal squamous cell carcinoma (LSCC) is one of the most commonly seen cancers in the head and neck region with increasing morbidity and mortality globally. N6-methyladenosine (m6A) modification plays a critical role in the carcinogenesis of LSCC. In this study, two datasets from online database were analyzed for differentially expressed genes (DEGs) between LSCC and normal samples. Furthermore, we carried out a series of experiments, including hematoxylin & eosin staining, immunohistochemical (IHC) staining, CCK-8, colony formation, transwell, flow cytometry, xenograft tumor model assays, actinomycin D assay, cycloheximide (CHX) assay, methylated m6A RNA immunoprecipitation (Me-RIP), RNA immunoprecipitation (RIP) assay, to verify the relevant findings in vivo and in vitro. Insulin like growth factor 2 mRNA binding protein 2 (IGF2BP2) was identified as an up-regulated m6A regulator in LSCC samples. Lower IGF2BP2 expression was linked to higher survival probability in LSCC and other head and neck squamous cell carcinoma patients. In LSCC cells, IGF2BP2 knockdown attenuated cancer cell aggressiveness, possibly through modulating cell cycle arrest. In the xenograft tumor model derived from IGF2BP2 knocked-down LSCC cells, IGF2BP2 knockdown inhibited tumor growth. IGF2BP2 up-regulated CDK6 expression through facilitating the stability of CDK6 mRNA and protein. CDK6 knockdown caused no changes in IGF2BP2 expression, but partially eliminated the promotive effects of IGF2BP2 overexpression on LSCC cells' aggressiveness. Overexpressed IGF2BP2 in LSCC serves as an oncogenic factor, promoting LSCC cell proliferation and invasion in vitro and tumor growth in a xenograft tumor model in vivo through facilitating CDK6 mRNA stabilization.
Collapse
Affiliation(s)
- Xiaojun Tang
- Department of Otolaryngology Head and Neck Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Qinglai Tang
- Department of Otolaryngology Head and Neck Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Shisheng Li
- Department of Otolaryngology Head and Neck Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Mengmeng Li
- Department of Otolaryngology Head and Neck Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Tao Yang
- Department of Otolaryngology Head and Neck Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China.
| |
Collapse
|
25
|
Wang F, Song S, Guo B, Li Y, Wang H, Fu S, Wang L, Zhe X, Li H, Li D, Shao R, Pan Z. Increased TCP11 gene expression can inhibit the proliferation, migration and promote apoptosis of cervical cancer cells. BMC Cancer 2023; 23:853. [PMID: 37697257 PMCID: PMC10496356 DOI: 10.1186/s12885-023-11129-1] [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: 01/30/2023] [Accepted: 06/29/2023] [Indexed: 09/13/2023] Open
Abstract
BACKGROUND Cervical cancer is a common gynecological malignancy. Gene microarray found that TCP11 gene was highly expressed in cervical cancer. However, the effect of TCP11 gene on the proliferation, apoptosis and migration of cervical cancer cells and its underlying molecular mechanisms are unclear. METHODS GEPIA database, tissue microarray, western blot and qRT-PCR were used to analyze the expression of TCP11 gene in cervical cancer tissues and cells and its relationship with patients' survival rate. The cell cycle and apoptosis were detected by flow cytometry, and the expressions of cell cycle and apoptosis related molecules and EMT-related molecules were detected by Western blot and qRT-PCR. RESULTS The results showed that TCP11 gene was highly expressed in cervical cancer tissues and cells compared with normal cervical tissues and cells, and its expression was positively correlated with patients' survival rate. The results of proliferation and migration assays showed that TCP11 overexpression inhibited the proliferation and migration of HeLa and SiHa cells. The results showed that TCP11 overexpression blocked the cell cycle of HeLa and SiHa cells, decreased the expression of CDK1 and Cyclin B1, and increased the apoptosis and the expression of caspase-3, cleaved-caspase-3 and cleaved-PARP. TCP11 overexpression increased the protein and mRNA expression of EMT-related molecules ZO-1 and E-cadherin. Conversely, TCP11 knockdown promoted the proliferation of HeLa and SiHa cells and the migration of HeLa cells. CONCLUSIONS TCP11 overexpression significantly inhibited the occurrence and development of cervical cancer cells, it may be a potentially beneficial biomarker for cervical cancer.
Collapse
Affiliation(s)
- Fang Wang
- Department of Biochemistry and Molecular Biology, School of Medicine, Xinjiang Endemic and Ethnic Disease and Education Ministry Key Laboratory, Shihezi University, Shihezi, Xinjiang, 832002, China
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University, Beijing, 101149, China
| | - Shuyan Song
- Department of Biochemistry and Molecular Biology, School of Medicine, Xinjiang Endemic and Ethnic Disease and Education Ministry Key Laboratory, Shihezi University, Shihezi, Xinjiang, 832002, China
| | - Bingxuan Guo
- Department of Biochemistry and Molecular Biology, School of Medicine, Xinjiang Endemic and Ethnic Disease and Education Ministry Key Laboratory, Shihezi University, Shihezi, Xinjiang, 832002, China
| | - Yangyang Li
- Department of Biochemistry and Molecular Biology, School of Medicine, Xinjiang Endemic and Ethnic Disease and Education Ministry Key Laboratory, Shihezi University, Shihezi, Xinjiang, 832002, China
| | - Huijuan Wang
- Department of Biochemistry and Molecular Biology, School of Medicine, Xinjiang Endemic and Ethnic Disease and Education Ministry Key Laboratory, Shihezi University, Shihezi, Xinjiang, 832002, China
- Department of Clinical Laboratory, the First Affiliated Hospital of School of Medicine, Shihezi University, Shihezi, Xinjiang, 832000, China
| | - Shaowei Fu
- Department of Biochemistry and Molecular Biology, School of Medicine, Xinjiang Endemic and Ethnic Disease and Education Ministry Key Laboratory, Shihezi University, Shihezi, Xinjiang, 832002, China
| | - Luyue Wang
- Department of Biochemistry and Molecular Biology, School of Medicine, Xinjiang Endemic and Ethnic Disease and Education Ministry Key Laboratory, Shihezi University, Shihezi, Xinjiang, 832002, China
| | - Xiangyi Zhe
- Department of Biochemistry and Molecular Biology, School of Medicine, Xinjiang Endemic and Ethnic Disease and Education Ministry Key Laboratory, Shihezi University, Shihezi, Xinjiang, 832002, China.
| | - Hongtao Li
- Department of Biochemistry and Molecular Biology, School of Medicine, Xinjiang Endemic and Ethnic Disease and Education Ministry Key Laboratory, Shihezi University, Shihezi, Xinjiang, 832002, China
| | - Dongmei Li
- Department of Biochemistry and Molecular Biology, School of Medicine, Xinjiang Endemic and Ethnic Disease and Education Ministry Key Laboratory, Shihezi University, Shihezi, Xinjiang, 832002, China
| | - Renfu Shao
- Centre for Bioinnovation, School of Science, Technology and Engineering, University of the Sunshine Coast, Maroochydore, 4556, Australia
| | - Zemin Pan
- Department of Biochemistry and Molecular Biology, School of Medicine, Xinjiang Endemic and Ethnic Disease and Education Ministry Key Laboratory, Shihezi University, Shihezi, Xinjiang, 832002, China.
| |
Collapse
|
26
|
Xiao D, Gan Q, Duan X, Wang Q, Jiang Y, Han P, Zhang J. Preparation and Evaluation of [ 18F]AlF-NOTA-PBB for PET Imaging of Cyclin-dependent Kinase 4/6 in Tumors. Mol Pharm 2023; 20:4528-4536. [PMID: 37661815 DOI: 10.1021/acs.molpharmaceut.3c00216] [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: 09/05/2023]
Abstract
Cyclin-dependent kinases (CDKs), especially cyclin-dependent kinase 4/6 (CDK4/6), have been targets for the development of specific tumor imaging agents. Palbociclib is a highly selective CDK4/6 inhibitor. In this study, to develop a novel 18F-labeled palbociclib derivative for specific tumor imaging, we designed and synthesized a ligand (NOTA-PBB) consisting of palbociclib as the targeted pharmacophore and NOTA as the macrocyclic bifunctional chelator. The corresponding [18F]AlF-NOTA-PBB complex was prepared with high radiochemical purity (98.4 ± 0.15%) and yield (58.7 ± 4.5%) within 35 min without requiring HPLC purification through a simple one-step 18F-labeling strategy of NOTA-AlF chelation chemistry. The radiotracer was lipophilic (log P = 0.095 ± 0.003) and had good stability in vitro and in vivo. The cellular uptake studies performed on the MCF-7 breast cancer cell line (ER-positive and HER2-negative) showed that radioactive uptake was blocked by preincubating with a molar dose of palbociclib and it had a nanomolar binding affinity to CDK4/6 (IC50 = 16.23 ± 1.84 nM), demonstrating a CDK4/6-mediated uptake mechanism. Its ex vivo biodistribution in nude mice-bearing MCF-7 tumors showed obvious tumor uptake and a high tumor/muscle ratio of [18F]AlF-NOTA-PBB, and tumor uptake was inhibited with 100 μg of palbociclib, demonstrating specific binding to CDK4/6. Radioactivity accumulation in MCF-7 tumors was observed in PET imaging with [18F]AlF-NOTA-PBB. Based on the results of this work, [18F]AlF-NOTA-PBB has the promising capability as a CDK4/6-targeted tumor imaging agent.
Collapse
Affiliation(s)
- Di Xiao
- Key Laboratory of Radiopharmaceuticals of Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Product Administration), College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Qianqian Gan
- Key Laboratory of Radiopharmaceuticals of Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Product Administration), College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Xiaojiang Duan
- Department of Nuclear Medicine, Peking University First Hospital, Beijing 100034, China
| | - Qianna Wang
- Key Laboratory of Radiopharmaceuticals of Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Product Administration), College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Yuhao Jiang
- Key Laboratory of Radiopharmaceuticals of Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Product Administration), College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Peiwen Han
- Key Laboratory of Radiopharmaceuticals of Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Product Administration), College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Junbo Zhang
- Key Laboratory of Radiopharmaceuticals of Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Product Administration), College of Chemistry, Beijing Normal University, Beijing 100875, China
| |
Collapse
|
27
|
Li J, Guo D, Wang F, Tian M, Wang C, Guo X, Xu B, Wang Y. Functional analysis of AccCDK2-like and AccCINP-like genes in Apis cerana cerana under pesticide and heavy metal stress. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2023; 195:105540. [PMID: 37666611 DOI: 10.1016/j.pestbp.2023.105540] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 07/08/2023] [Accepted: 07/14/2023] [Indexed: 09/06/2023]
Abstract
Heavy metals and pesticides represent prominent sources of pollution in the natural habitat of Apis cerana cerana, potentially endangering their health through the induction of oxidative stress reactions. This study aimed to address this issue by isolating AccCDK2-like and AccCINP-like proteins from Apis cerana cerana and investigating their functional roles in honey bee resistance against pesticide and heavy metal stresses. Bioinformatics analysis revealed significant homology of these proteins with those found in other species. Functional studies confirmed their participation in interaction with each other, alongside demonstrating distinct patterns of expression and localization. Specifically, AccCDK2-like exhibited higher expression levels in prepupae and muscle tissues, while AccCINP-like showed maximal expression in brown pupae and abdomen. Furthermore, the expression levels of these proteins were found to be modulated in response to pesticide and heavy metal stresses. Notably, overexpression of AccCDK2-like and AccCINP-like led to a noticeable alteration in E. coli's ability to withstand external stresses. Additionally, silencing of the AccCDK2-like and AccCINP-like genes resulted in a significant reduction in antioxidant enzyme activity and the expression levels of genes related to antioxidant function. Consequently, the mortality rate of Apis cerana cerana under pesticide and heavy metal stresses conspicuously increased. Hence, our findings suggest that AccCDK2-like and AccCINP-like proteins potentially play a crucial role in the response of Apis cerana cerana to pesticide and heavy metal stress, likely by modulating the antioxidant pathway.
Collapse
Affiliation(s)
- Jing Li
- State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Taian, Shandong 271018, People's Republic of China
| | - Dezheng Guo
- State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Taian, Shandong 271018, People's Republic of China
| | - Fang Wang
- State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Taian, Shandong 271018, People's Republic of China
| | - Ming Tian
- State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Taian, Shandong 271018, People's Republic of China
| | - Chen Wang
- State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Taian, Shandong 271018, People's Republic of China
| | - Xingqi Guo
- State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Taian, Shandong 271018, People's Republic of China
| | - Baohua Xu
- College of Animal Science and Technology, Shandong Agricultural University, Taian, Shandong 271018, People's Republic of China
| | - Ying Wang
- College of Animal Science and Technology, Shandong Agricultural University, Taian, Shandong 271018, People's Republic of China.
| |
Collapse
|
28
|
Behnam B, Taghizadeh-Hesary F. Mitochondrial Metabolism: A New Dimension of Personalized Oncology. Cancers (Basel) 2023; 15:4058. [PMID: 37627086 PMCID: PMC10452105 DOI: 10.3390/cancers15164058] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 08/01/2023] [Accepted: 08/02/2023] [Indexed: 08/27/2023] Open
Abstract
Energy is needed by cancer cells to stay alive and communicate with their surroundings. The primary organelles for cellular metabolism and energy synthesis are mitochondria. Researchers recently proved that cancer cells can steal immune cells' mitochondria using nanoscale tubes. This finding demonstrates the dependence of cancer cells on normal cells for their living and function. It also denotes the importance of mitochondria in cancer cells' biology. Emerging evidence has demonstrated how mitochondria are essential for cancer cells to survive in the harsh tumor microenvironments, evade the immune system, obtain more aggressive features, and resist treatments. For instance, functional mitochondria can improve cancer resistance against radiotherapy by scavenging the released reactive oxygen species. Therefore, targeting mitochondria can potentially enhance oncological outcomes, according to this notion. The tumors' responses to anticancer treatments vary, ranging from a complete response to even cancer progression during treatment. Therefore, personalized cancer treatment is of crucial importance. So far, personalized cancer treatment has been based on genomic analysis. Evidence shows that tumors with high mitochondrial content are more resistant to treatment. This paper illustrates how mitochondrial metabolism can participate in cancer resistance to chemotherapy, immunotherapy, and radiotherapy. Pretreatment evaluation of mitochondrial metabolism can provide additional information to genomic analysis and can help to improve personalized oncological treatments. This article outlines the importance of mitochondrial metabolism in cancer biology and personalized treatments.
Collapse
Affiliation(s)
- Babak Behnam
- Department of Regulatory Affairs, Amarex Clinical Research, NSF International, Germantown, MD 20874, USA
| | - Farzad Taghizadeh-Hesary
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences, Tehran 1445613131, Iran
- Department of Radiation Oncology, Iran University of Medical Sciences, Tehran 1445613131, Iran
| |
Collapse
|
29
|
Rial SA, Shishani R, Cummings BP, Lim GE. Is 14-3-3 the Combination to Unlock New Pathways to Improve Metabolic Homeostasis and β-Cell Function? Diabetes 2023; 72:1045-1054. [PMID: 37471599 PMCID: PMC10382651 DOI: 10.2337/db23-0094] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 05/10/2023] [Indexed: 07/22/2023]
Abstract
Since their discovery nearly five decades ago, molecular scaffolds belonging to the 14-3-3 protein family have been recognized as pleiotropic regulators of diverse cellular and physiological functions. With their ability to bind to proteins harboring specific serine and threonine phosphorylation motifs, 14-3-3 proteins can interact with and influence the function of docking proteins, enzymes, transcription factors, and transporters that have essential roles in metabolism and glucose homeostasis. Here, we will discuss the regulatory functions of 14-3-3 proteins that will be of great interest to the fields of metabolism, pancreatic β-cell biology, and diabetes. We first describe how 14-3-3 proteins play a central role in glucose and lipid homeostasis by modulating key pathways of glucose uptake, glycolysis, oxidative phosphorylation, and adipogenesis. This is followed by a discussion of the contributions of 14-3-3 proteins to calcium-dependent exocytosis and how this relates to insulin secretion from β-cells. As 14-3-3 proteins are major modulators of apoptosis and cell cycle progression, we will explore if 14-3-3 proteins represent a viable target for promoting β-cell regeneration and discuss the feasibility of targeting 14-3-3 proteins to treat metabolic diseases such as diabetes. ARTICLE HIGHLIGHTS 14-3-3 proteins are ubiquitously expressed scaffolds with multiple roles in glucose homeostasis and metabolism. 14-3-3ζ regulates adipogenesis via distinct mechanisms and is required for postnatal adiposity and adipocyte function. 14-3-3ζ controls glucose-stimulated insulin secretion from pancreatic β-cells by regulating mitochondrial function and ATP synthesis as well as facilitating cross talk between β-cells and α-cells.
Collapse
Affiliation(s)
- Sabri A. Rial
- Department of Medicine, University of Montreal, Montreal, Quebec, Canada
- Cardiometabolic Axis, University of Montreal Hospital Research Center (CRCHUM), Montreal, Quebec, Canada
| | - Rahaf Shishani
- Department of Surgery, Center for Alimentary and Metabolic Sciences, School of Medicine, University of California, Davis, Sacramento, CA
| | - Bethany P. Cummings
- Department of Surgery, Center for Alimentary and Metabolic Sciences, School of Medicine, University of California, Davis, Sacramento, CA
| | - Gareth E. Lim
- Department of Medicine, University of Montreal, Montreal, Quebec, Canada
- Cardiometabolic Axis, University of Montreal Hospital Research Center (CRCHUM), Montreal, Quebec, Canada
| |
Collapse
|
30
|
Taghizadeh-Hesary F, Houshyari M, Farhadi M. Mitochondrial metabolism: a predictive biomarker of radiotherapy efficacy and toxicity. J Cancer Res Clin Oncol 2023; 149:6719-6741. [PMID: 36719474 DOI: 10.1007/s00432-023-04592-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 01/18/2023] [Indexed: 02/01/2023]
Abstract
INTRODUCTION Radiotherapy is a mainstay of cancer treatment. Clinical studies revealed a heterogenous response to radiotherapy, from a complete response to even disease progression. To that end, finding the relative prognostic factors of disease outcomes and predictive factors of treatment efficacy and toxicity is essential. It has been demonstrated that radiation response depends on DNA damage response, cell cycle phase, oxygen concentration, and growth rate. Emerging evidence suggests that altered mitochondrial metabolism is associated with radioresistance. METHODS This article provides a comprehensive evaluation of the role of mitochondria in radiotherapy efficacy and toxicity. In addition, it demonstrates how mitochondria might be involved in the famous 6Rs of radiobiology. RESULTS In terms of this idea, decreasing the mitochondrial metabolism of cancer cells may increase radiation response, and enhancing the mitochondrial metabolism of normal cells may reduce radiation toxicity. Enhancing the normal cells (including immune cells) mitochondrial metabolism can potentially improve the tumor response by enhancing immune reactivation. Future studies are invited to examine the impacts of mitochondrial metabolism on radiation efficacy and toxicity. Improving radiotherapy response with diminishing cancer cells' mitochondrial metabolism, and reducing radiotherapy toxicity with enhancing normal cells' mitochondrial metabolism.
Collapse
Affiliation(s)
- Farzad Taghizadeh-Hesary
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
- Clinical Oncology Department, Iran University of Medical Sciences, Tehran, Iran.
| | - Mohammad Houshyari
- Clinical Oncology Department, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Farhadi
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
31
|
Chandra O, Sharma M, Pandey N, Jha IP, Mishra S, Kong SL, Kumar V. Patterns of transcription factor binding and epigenome at promoters allow interpretable predictability of multiple functions of non-coding and coding genes. Comput Struct Biotechnol J 2023; 21:3590-3603. [PMID: 37520281 PMCID: PMC10371796 DOI: 10.1016/j.csbj.2023.07.014] [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: 01/25/2023] [Revised: 07/05/2023] [Accepted: 07/11/2023] [Indexed: 08/01/2023] Open
Abstract
Understanding the biological roles of all genes only through experimental methods is challenging. A computational approach with reliable interpretability is needed to infer the function of genes, particularly for non-coding RNAs. We have analyzed genomic features that are present across both coding and non-coding genes like transcription factor (TF) and cofactor ChIP-seq (823), histone modifications ChIP-seq (n = 621), cap analysis gene expression (CAGE) tags (n = 255), and DNase hypersensitivity profiles (n = 255) to predict ontology-based functions of genes. Our approach for gene function prediction was reliable (>90% balanced accuracy) for 486 gene-sets. PubMed abstract mining and CRISPR screens supported the inferred association of genes with biological functions, for which our method had high accuracy. Further analysis revealed that TF-binding patterns at promoters have high predictive strength for multiple functions. TF-binding patterns at the promoter add an unexplored dimension of explainable regulatory aspects of genes and their functions. Therefore, we performed a comprehensive analysis for the functional-specificity of TF-binding patterns at promoters and used them for clustering functions to reveal many latent groups of gene-sets involved in common major cellular processes. We also showed how our approach could be used to infer the functions of non-coding genes using the CRISPR screens of coding genes, which were validated using a long non-coding RNA CRISPR screen. Thus our results demonstrated the generality of our approach by using gene-sets from CRISPR screens. Overall, our approach opens an avenue for predicting the involvement of non-coding genes in various functions.
Collapse
Affiliation(s)
- Omkar Chandra
- Department of Computational Biology, Indraprastha Institute of Information Technology, Okhla Ph-III, New Delhi, India
| | - Madhu Sharma
- Department of Computational Biology, Indraprastha Institute of Information Technology, Okhla Ph-III, New Delhi, India
| | - Neetesh Pandey
- Department of Computational Biology, Indraprastha Institute of Information Technology, Okhla Ph-III, New Delhi, India
| | - Indra Prakash Jha
- Department of Computational Biology, Indraprastha Institute of Information Technology, Okhla Ph-III, New Delhi, India
| | - Shreya Mishra
- Department of Computational Biology, Indraprastha Institute of Information Technology, Okhla Ph-III, New Delhi, India
| | - Say Li Kong
- Genome Institute of Singapore, Agency for Science Technology and Research, Singapore, Singapore
| | - Vibhor Kumar
- Department of Computational Biology, Indraprastha Institute of Information Technology, Okhla Ph-III, New Delhi, India
| |
Collapse
|
32
|
Zhao T, Xiao X, Li L, Wu X, Yuan T. Rosline promotes p21 expression to inhibit ovarian cancer cell proliferation via p53-independent pathway. J Obstet Gynaecol Res 2023. [PMID: 37317483 DOI: 10.1111/jog.15708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 05/28/2023] [Indexed: 06/16/2023]
Abstract
AIM To investigate the effect of benzothiazole derivatives (Rosline) on ovarian cancer and the potential mechanism. METHODS Ovarian cancer tissues were collected clinically and immunohistochemistry was used to detect the expression of p53 and p21. Ovarian cancer cells were exposed to 0, 2.5, 5, 10 μmol/L Rosline for 24 h. 100 nmol/L Pifithrin-α pre-incubation was used to inhibit the transcriptional activity of p53. CCK-8 and BrdU assays were used to detect the effects of different concentrations of rosline on the proliferation and cell cycle of OVCAR420 and SKOV3 cells. Flow cytometry assay was used to detect cell cycle. The transcriptional and translational expression of p21 and p53 were detected by RT-qPCR and Western blot. RESULTS p21 was expressed in ovarian cancer tissues without p53 expression. Rosline inhibits the proliferation of ovarian cancer cells and blocks the cell cycle progression. Meanwhile, Rosline promotes p21 expression in ovarian cancer cells at both mRNA and protein levels, but with no significant effect on p53 expression. Besides, Rosline promotes p21 expression, inhibits cell proliferation, and blocks the cell cycle via the p53-independent pathway. CONCLUSION Rosline promoted p21 expression thereby inhibiting cell proliferation and blocks the cell cycle via p53-independent pathway.
Collapse
Affiliation(s)
- Ting Zhao
- Department of Gynecology, Yunnan First People's Hospital, Kunming, Yunnan, China
| | - Xiao Xiao
- Department of Gynecology, Yunnan First People's Hospital, Kunming, Yunnan, China
| | - Lingchuan Li
- Department of Gynecology, Yunnan First People's Hospital, Kunming, Yunnan, China
| | - Xiaomei Wu
- Department of Gynecology, Yunnan First People's Hospital, Kunming, Yunnan, China
| | - Tao Yuan
- Department of Gynecology, Yunnan First People's Hospital, Kunming, Yunnan, China
| |
Collapse
|
33
|
Wang X, Ding L, Jiang H, Yuan X, Xiang L, Tang C. Synthesis and biological evaluation of novel pteridin-7(8H)-one derivatives as potent CDK2 inhibitors. Bioorg Med Chem Lett 2023; 88:129284. [PMID: 37060933 DOI: 10.1016/j.bmcl.2023.129284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 04/07/2023] [Accepted: 04/09/2023] [Indexed: 04/17/2023]
Abstract
Cyclin-dependent kinase 2 (CDK2) is considered as an important target in the research of antitumor drugs. Taking the CDK2/4/6 inhibitor Ebvaciclib as the positive control and an in-house library compound (23) as the lead compound, three classes of 30 target compounds with pteridin-7(8H)-one as the core structure were designed to establish structure-activity relationships (SAR). In general, SAR of pteridin-7(8H)-one CDK2 inhibitors is systematically described in this paper, resulting in the discovery of two compounds (KII-17 and KII-21) with further research value. After the above compounds were tested for CDK2/4/6 kinase selectivity, we found that compound KII-21 was about 3 and 4 times more selective to CDK2-cyclinE2 than CDK4-cyclinD1 and CDK6-cyclinD3, respectively. This work also provides a reference basis for the subsequent research on CDK2 inhibitors.
Collapse
Affiliation(s)
- Xia Wang
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, China
| | - Lei Ding
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, China
| | - Hongyu Jiang
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, China
| | - Xin Yuan
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, China
| | - Lianghua Xiang
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, China
| | - Chunlei Tang
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, China.
| |
Collapse
|
34
|
Yang TT, Yu S, Ke CLK, Cheng ST. The Genomic Landscape of Melanoma and Its Therapeutic Implications. Genes (Basel) 2023; 14:genes14051021. [PMID: 37239381 DOI: 10.3390/genes14051021] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 03/25/2023] [Accepted: 04/28/2023] [Indexed: 05/28/2023] Open
Abstract
Melanoma is one of the most aggressive malignancies of the skin. The genetic composition of melanoma is complex and varies among different subtypes. With the aid of recent technologies such as next generation sequencing and single-cell sequencing, our understanding of the genomic landscape of melanoma and its tumor microenvironment has become increasingly clear. These advances may provide explanation to the heterogenic treatment outcomes of melanoma patients under current therapeutic guidelines and provide further insights to the development of potential new therapeutic targets. Here, we provide a comprehensive review on the genetics related to melanoma tumorigenesis, metastasis, and prognosis. We also review the genetics affecting the melanoma tumor microenvironment and its relation to tumor progression and treatment.
Collapse
Affiliation(s)
- Ting-Ting Yang
- Department of Dermatology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Dermatology, Pingtung Hospital, Ministry of Health and Welfare, Pingtung 900, Taiwan
| | - Sebastian Yu
- Department of Dermatology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Dermatology, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Neuroscience Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Chiao-Li Khale Ke
- Department of Psychiatry, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
- Department of Psychiatry, Kaohsiung Municipal SiaoGang Hospital, Kaohsiung Medical University, Kaohsiung 812, Taiwan
| | - Shih-Tsung Cheng
- Department of Dermatology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Dermatology, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| |
Collapse
|
35
|
Hislop EW, Tipping WJ, Faulds K, Graham D. Label-Free Cytometric Evaluation of Mitosis via Stimulated Raman Scattering Microscopy and Spectral Phasor Analysis. Anal Chem 2023; 95:7244-7253. [PMID: 37097612 PMCID: PMC10173251 DOI: 10.1021/acs.analchem.3c00212] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2023]
Abstract
Hyperspectral stimulated Raman scattering (SRS) microscopy is a robust imaging tool for the analysis of biological systems. Here, we present a unique perspective, a label-free spatiotemporal map of mitosis, by integrating hyperspectral SRS microscopy with advanced chemometrics to assess the intrinsic biomolecular properties of an essential process of mammalian life. The application of spectral phasor analysis to multiwavelength SRS images in the high-wavenumber (HWN) region of the Raman spectrum enabled the segmentation of subcellular organelles based on innate SRS spectra. Traditional imaging of DNA is primarily reliant on using fluorescent probes or stains which can affect the biophysical properties of the cell. Here, we demonstrate the label-free visualization of nuclear dynamics during mitosis coupled with an evaluation of its spectral profile in a rapid and reproducible manner. These results provide a snapshot of the cell division cycle and chemical variability between intracellular compartments in single-cell models, which is central to understanding the molecular foundations of these fundamental biological processes. The evaluation of HWN images by phasor analysis also facilitated the differentiation between cells in separate phases of the cell cycle based solely on their nuclear SRS spectral signal, which offers an interesting label-free approach in combination with flow cytometry. Therefore, this study demonstrates that SRS microscopy combined with spectral phasor analysis is a valuable method for detailed optical fingerprinting at the subcellular level.
Collapse
Affiliation(s)
- Ewan W Hislop
- Centre for Molecular Nanometrology, Department of Pure and Applied Chemistry, Technology and Innovation Centre, University of Strathclyde, Glasgow G1 1RD, United Kingdom
| | - William J Tipping
- Centre for Molecular Nanometrology, Department of Pure and Applied Chemistry, Technology and Innovation Centre, University of Strathclyde, Glasgow G1 1RD, United Kingdom
| | - Karen Faulds
- Centre for Molecular Nanometrology, Department of Pure and Applied Chemistry, Technology and Innovation Centre, University of Strathclyde, Glasgow G1 1RD, United Kingdom
| | - Duncan Graham
- Centre for Molecular Nanometrology, Department of Pure and Applied Chemistry, Technology and Innovation Centre, University of Strathclyde, Glasgow G1 1RD, United Kingdom
| |
Collapse
|
36
|
Maimaiti A, Liu Y, Abulaiti A, Wang X, Feng Z, Wang J, Mijiti M, Turhon M, Alimu N, Wang Y, Liang W, Jiang L, Pei Y. Genomic Profiling of Lower-Grade Gliomas Subtype with Distinct Molecular and Clinicopathologic Characteristics via Altered DNA-Damage Repair Features. J Mol Neurosci 2023; 73:269-286. [PMID: 37067735 DOI: 10.1007/s12031-023-02116-z] [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: 12/26/2022] [Accepted: 03/30/2023] [Indexed: 04/18/2023]
Abstract
Lower WHO grade II and III gliomas (LGGs) exhibit significant genetic and transcriptional heterogeneity, and the heterogeneity of DNA damage repair (DDR) and its relationship to tumor biology, transcriptome, and tumor microenvironment (TME) remains poorly understood. In this study, we conducted multi-omics data integration to investigate DDR alterations in LGG. Based on clinical parameters and molecular characteristics, LGG patients were categorized into distinct DDR subtypes, namely, DDR-activated and DDR-suppressed subtypes. We compared gene mutation, immune spectrum, and immune cell infiltration between the two subtypes. DDR scores were generated to classify LGG patients based on DDR subtype features, and the results were validated using a multi-layer data cohort. We found that DDR activation was associated with poorer overall survival and that clinicopathological features of advanced age and higher grade were more common in the DDR-activated subtype. DDR-suppressed subtypes exhibited more frequent mutations in IDH1. In addition, we observed significant upregulation of activated immune cells in the DDR-activated subgroup, which suggests that immune cell infiltration significantly influences tumor progression and immunotherapeutic responses. Furthermore, we constructed a DDR signature for LGG using six DDR genes, which allowed for the division of patients into low- and high-risk groups. Quantitative real-time PCR results showed that CDK1, CDK2, TYMS, SMC4, and WEE1 were significantly upregulated in LGG samples compared to normal brain tissue samples. Overall, our study sheds light on DDR heterogeneity in LGG and provides insight into the molecular pathways of DDR involved in LGG development.
Collapse
Affiliation(s)
- Aierpati Maimaiti
- Department of Neurosurgery, Neurosurgery Centre, The First Affiliated Hospital of Xinjiang Medical University, No. 137, South Liyushan Road, Xinshi District, 830054, Urumqi, Xinjiang, China
| | - Yanwen Liu
- Department of Medical Laboratory, Xinjiang Production and Construction Corps Hospital, 830002, Urumqi, Xinjiang, China
| | - Aimitaji Abulaiti
- Department of Neurosurgery, Neurosurgery Centre, The First Affiliated Hospital of Xinjiang Medical University, No. 137, South Liyushan Road, Xinshi District, 830054, Urumqi, Xinjiang, China
| | - Xixian Wang
- Department of Neurosurgery, Neurosurgery Centre, The First Affiliated Hospital of Xinjiang Medical University, No. 137, South Liyushan Road, Xinshi District, 830054, Urumqi, Xinjiang, China
| | - Zhaohai Feng
- Department of Neurosurgery, Neurosurgery Centre, The First Affiliated Hospital of Xinjiang Medical University, No. 137, South Liyushan Road, Xinshi District, 830054, Urumqi, Xinjiang, China
| | - Jiaming Wang
- Department of Neurosurgery, Neurosurgery Centre, The First Affiliated Hospital of Xinjiang Medical University, No. 137, South Liyushan Road, Xinshi District, 830054, Urumqi, Xinjiang, China
| | - Maimaitili Mijiti
- Department of Neurosurgery, Neurosurgery Centre, The First Affiliated Hospital of Xinjiang Medical University, No. 137, South Liyushan Road, Xinshi District, 830054, Urumqi, Xinjiang, China
| | - Mirzat Turhon
- Department of Neurointerventional Surgery, Beijing Neurosurgical Institute, Capital Medical University, 100070, Beijing, China
- Department of Neurointerventional Surgery, Beijing Tiantan Hospital, Capital Medical University, 100070, Beijing, China
| | - Nilipaer Alimu
- Department of Otorhinolaryngology, The First Affiliated Hospital of Xinjiang Medical University, No. 137, South Liyushan Road, Xinshi District, 830054, Urumqi, Xinjiang, China
| | - Yongxin Wang
- Department of Neurosurgery, Neurosurgery Centre, The First Affiliated Hospital of Xinjiang Medical University, No. 137, South Liyushan Road, Xinshi District, 830054, Urumqi, Xinjiang, China
| | - Wenbao Liang
- Department of Neurosurgery, The Fourth Affiliated Hospital of Xinjiang Medical University, No. 116, Huanghe Road, Shaibak District, 830000, Urumqi, Xinjiang, China.
| | - Lei Jiang
- Department of Neurosurgery, Neurosurgery Centre, The First Affiliated Hospital of Xinjiang Medical University, No. 137, South Liyushan Road, Xinshi District, 830054, Urumqi, Xinjiang, China.
| | - Yinan Pei
- Department of Neurosurgery, Neurosurgery Centre, The First Affiliated Hospital of Xinjiang Medical University, No. 137, South Liyushan Road, Xinshi District, 830054, Urumqi, Xinjiang, China.
| |
Collapse
|
37
|
Cheung AHK, Hui CHL, Wong KY, Liu X, Chen B, Kang W, To KF. Out of the cycle: Impact of cell cycle aberrations on cancer metabolism and metastasis. Int J Cancer 2023; 152:1510-1525. [PMID: 36093588 DOI: 10.1002/ijc.34288] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 08/17/2022] [Accepted: 09/02/2022] [Indexed: 11/11/2022]
Abstract
The use of cell cycle inhibitors has necessitated a better understanding of the cell cycle in tumor biology to optimize the therapeutic approach. Cell cycle aberrations are common in cancers, and it is increasingly acknowledged that these aberrations exert oncogenic effects beyond the cell cycle. Multiple facets such as cancer metabolism, immunity and metastasis are also affected, all of which are beyond the effect of cell proliferation alone. This review comprehensively summarized the important recent findings and advances in these interrelated processes. In cancer metabolism, cell cycle regulators can modulate various pathways in aerobic glycolysis, glucose uptake and gluconeogenesis, mainly through transcriptional regulation and kinase activities. Amino acid metabolism is also regulated through cell cycle progression. On cancer metastasis, metabolic plasticity, immune evasion, tumor microenvironment adaptation and metastatic site colonization are intricately related to the cell cycle, with distinct regulatory mechanisms at each step of invasion and dissemination. Throughout the synthesis of current understanding, knowledge gaps and limitations in the literature are also highlighted, as are new therapeutic approaches such as combinational therapy and challenges in tackling emerging targeted therapy resistance. A greater understanding of how the cell cycle modulates diverse aspects of cancer biology can hopefully shed light on identifying new molecular targets by harnessing the vast potential of the cell cycle.
Collapse
Affiliation(s)
- Alvin Ho-Kwan Cheung
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China.,Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Hong Kong, China.,State Key Laboratory of Translational Oncology, Sir Y.K. Pao Cancer Center, The Chinese University of Hong Kong, Hong Kong, China
| | - Chris Ho-Lam Hui
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China.,Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Hong Kong, China.,State Key Laboratory of Translational Oncology, Sir Y.K. Pao Cancer Center, The Chinese University of Hong Kong, Hong Kong, China
| | - Kit Yee Wong
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China.,Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Hong Kong, China.,State Key Laboratory of Translational Oncology, Sir Y.K. Pao Cancer Center, The Chinese University of Hong Kong, Hong Kong, China
| | - Xiaoli Liu
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China.,Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Hong Kong, China.,State Key Laboratory of Translational Oncology, Sir Y.K. Pao Cancer Center, The Chinese University of Hong Kong, Hong Kong, China
| | - Bonan Chen
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China.,Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Hong Kong, China.,State Key Laboratory of Translational Oncology, Sir Y.K. Pao Cancer Center, The Chinese University of Hong Kong, Hong Kong, China
| | - Wei Kang
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China.,Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Hong Kong, China.,State Key Laboratory of Translational Oncology, Sir Y.K. Pao Cancer Center, The Chinese University of Hong Kong, Hong Kong, China
| | - Ka Fai To
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China.,Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Hong Kong, China.,State Key Laboratory of Translational Oncology, Sir Y.K. Pao Cancer Center, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
38
|
Li J, Wang YF, Shen ZC, Zou Q, Lin XF, Wang XY. Recent developments on natural polysaccharides as potential anti-gastric cancer substance: Structural feature and bioactivity. Int J Biol Macromol 2023; 232:123390. [PMID: 36706878 DOI: 10.1016/j.ijbiomac.2023.123390] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 01/13/2023] [Accepted: 01/19/2023] [Indexed: 01/26/2023]
Abstract
Gastric cancer (GC) is being a serious threat to human health. Seeking safer and more effective ingredients for anti-GC is of significance. Increasing natural polysaccharides (NPs) have been demonstrated to possess anti-GC activity. However, the information on anti-GC NPs is scattered. For well-understanding the potential of NPs as anti-GC substances, the recent developments on structure, bioactivity and mechanism of anti-GC NPs were comprehensively reviewed in this article. Meanwhile, the structure-activity relationship was discussed. Recent studies indicated that anti-GC NPs could be mainly divided into glucan and heteropolysaccharide, whose structures affected by sources and protocols of extraction and purification. NPs exhibited anti-GC activities in cell and animal experiments as well as clinical trials, and the mechanisms might be anti-proliferation, inducing apoptosis, anti-metastasis and anti-invasion, inducing autophagy, boosting immunity, anti-angiogenesis, reducing drug resistance, anti-angiogenesis, improving antioxidant level and changing metabolites. Moreover, structural features included molecular weight, functional groups, uronic acid and monosaccharide composition, glycosidic linkage type, and degree of branching and conformation might influence the activities. Otherwise, modifications could enhance the anti-GC activity of NPs, and anti-GC NPs could be combinedly used with chemotherapeutic drugs. This review supports the applications of NPs in anti-GC and provides theoretical basis for future study.
Collapse
Affiliation(s)
- Jing Li
- School of Public Health and Health Management, Gannan Medical University, Ganzhou 341000, China
| | - Yi-Fei Wang
- School of Public Health and Health Management, Gannan Medical University, Ganzhou 341000, China
| | - Zi-Chun Shen
- School of Public Health and Health Management, Gannan Medical University, Ganzhou 341000, China
| | - Qi Zou
- School of Public Health and Health Management, Gannan Medical University, Ganzhou 341000, China
| | - Xiao-Fan Lin
- School of Public Health and Health Management, Gannan Medical University, Ganzhou 341000, China
| | - Xiao-Yin Wang
- School of Public Health and Health Management, Gannan Medical University, Ganzhou 341000, China.
| |
Collapse
|
39
|
Asouli A, Sadr S, Mohebalian H, Borji H. Anti-Tumor Effect of Protoscolex Hydatid Cyst Somatic Antigen on Inhibition Cell Growth of K562. Acta Parasitol 2023:10.1007/s11686-023-00680-3. [PMID: 36991291 DOI: 10.1007/s11686-023-00680-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 03/23/2023] [Indexed: 03/31/2023]
Abstract
BACKGROUND AND OBJECTIVE Today, cancer is one of the most important causes of death in the world, and so far, many treatment methods have been used in this field. Immunotherapy is considered one of the newest developments in this science, and it is still being investigated in some forms in different cancers and with a variety of antigens as well. One of the subsets of cancer immunotherapy is its treatment using parasitic antigens. The present study evaluated the effect of using somatic antigens of protoscoleces of Echinococcus granulosus on K562 cancer cells. METHODS In this study, hydatid cysts' protoscolex antigens were extracted, purified, and added to K562 cancer cells at three concentrations (0.1, 1, and 2 mg/ml) and on three times (24, 48, and 72 h). The number of apoptotic cells was compared to the control flask. The antigen concentration of 2 mg/ml was used as a control sample to investigate its cytotoxic effect on the growth of healthy HFF3 cells. Annexin V and PI tests were also performed to differentiate apoptosis from necrosis. RESULTS In flasks treated with hydatid cyst protoscolex antigen, all three concentrations significantly reduced the growth of cancer cells compared with the control flask, and concentration 2 of crude antigen significantly caused the death of cancer cells. Furthermore, more cancer cells underwent apoptosis by increasing the time of exposure to the antigen. On the other hand, flow cytometry results also showed that the amount of apoptosis has increased compared to the control group. In fact, Protoscolex hydatid cyst somatic antigens induce programmed cell death in K562 cancer cells while not having a cytotoxic effect on normal cells. CONCLUSION Therefore, it is suggested to do more research on the anti-cancer and therapeutic properties of the antigens of this parasite.
Collapse
Affiliation(s)
- Atefe Asouli
- Department of Pathobiology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, P.O. Box: 9177948974, Mashhad, Iran
| | - Soheil Sadr
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Hadi Mohebalian
- Department of Pathobiology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, P.O. Box: 9177948974, Mashhad, Iran
| | - Hassan Borji
- Department of Pathobiology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, P.O. Box: 9177948974, Mashhad, Iran.
| |
Collapse
|
40
|
Bernal-Tirapo J, Bayo Jiménez MT, Yuste-García P, Cordova I, Peñas A, García-Borda FJ, Quintela C, Prieto I, Sánchez-Ramos C, Ferrero-Herrero E, Monsalve M. Evaluation of Mitochondrial Function in Blood Samples Shows Distinct Patterns in Subjects with Thyroid Carcinoma from Those with Hyperplasia. Int J Mol Sci 2023; 24:ijms24076453. [PMID: 37047426 PMCID: PMC10094811 DOI: 10.3390/ijms24076453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/21/2023] [Accepted: 03/25/2023] [Indexed: 04/03/2023] Open
Abstract
Metabolic adaptations are a hallmark of cancer and may be exploited to develop novel diagnostic and therapeutic tools. Only about 50% of the patients who undergo thyroidectomy due to suspicion of thyroid cancer actually have the disease, highlighting the diagnostic limitations of current tools. We explored the possibility of using non-invasive blood tests to accurately diagnose thyroid cancer. We analyzed blood and thyroid tissue samples from two independent cohorts of patients undergoing thyroidectomy at the Hospital Universitario 12 de Octubre (Madrid, Spain). As expected, histological comparisons of thyroid cancer and hyperplasia revealed higher proliferation and apoptotic rates and enhanced vascular alterations in the former. Notably, they also revealed increased levels of membrane-bound phosphorylated AKT, suggestive of enhanced glycolysis, and alterations in mitochondrial sub-cellular distribution. Both characteristics are common metabolic adaptations in primary tumors. These data together with reduced mtDNA copy number and elevated levels of the mitochondrial antioxidant PRX3 in cancer tissue samples suggest the presence of mitochondrial oxidative stress. In plasma, cancer patients showed higher levels of cfDNA and mtDNA. Of note, mtDNA plasma levels inversely correlated with those in the tissue, suggesting that higher death rates were linked to lower mtDNA copy number. In PBMCs, cancer patients showed higher levels of PGC-1α, a positive regulator of mitochondrial function, but this increase was not associated with a corresponding induction of its target genes, suggesting a reduced activity in cancer patients. We also observed a significant difference in the PRDX3/PFKFB3 correlation at the gene expression level, between carcinoma and hyperplasia patients, also indicative of increased systemic metabolic stress in cancer patients. The correlation of mtDNA levels in tissue and PBMCs further stressed the interconnection between systemic and tumor metabolism. Evaluation of the mitochondrial gene ND1 in plasma, PBMCs and tissue samples, suggested that it could be a good biomarker for systemic oxidative metabolism, with ND1/mtDNA ratio positively correlating in PBMCs and tissue samples. In contrast, ND4 evaluation would be informative of tumor development, with ND4/mtDNA ratio specifically altered in the tumor context. Taken together, our data suggest that metabolic dysregulation in thyroid cancer can be monitored accurately in blood samples and might be exploited for the accurate discrimination of cancer from hyperplasia.
Collapse
|
41
|
Shaikh A, Wesner AA, Abuhattab M, Kutty RG, Premnath P. Cell cycle regulators and bone: development and regeneration. Cell Biosci 2023; 13:35. [PMID: 36810262 PMCID: PMC9942316 DOI: 10.1186/s13578-023-00988-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 02/13/2023] [Indexed: 02/24/2023] Open
Abstract
Cell cycle regulators act as inhibitors or activators to prevent cancerogenesis. It has also been established that they can play an active role in differentiation, apoptosis, senescence, and other cell processes. Emerging evidence has demonstrated a role for cell cycle regulators in bone healing/development cascade. We demonstrated that deletion of p21, a cell cycle regulator acting at the G1/S transition enhanced bone repair capacity after a burr-hole injury in the proximal tibia of mice. Similarly, another study has shown that inhibition of p27 can increase bone mineral density and bone formation. Here, we provide a concise review of cell cycle regulators that influence cells like osteoblasts, osteoclasts, and chondrocytes, during development and/or healing of bone. It is imperative to understand the regulatory processes that govern cell cycle during bone healing and development as this will pave the way to develop novel therapies to improve bone healing after injury in instances of aged or osteoporotic fractures.
Collapse
Affiliation(s)
- Alisha Shaikh
- grid.267468.90000 0001 0695 7223Department of Biomedical Engineering, University of Wisconsin-Milwaukee, College of Engineering and Applied Sciences, 3200 N Cramer St, Milwaukee, WI 53211 USA
| | - Austin A. Wesner
- grid.267468.90000 0001 0695 7223Department of Biomedical Engineering, University of Wisconsin-Milwaukee, College of Engineering and Applied Sciences, 3200 N Cramer St, Milwaukee, WI 53211 USA
| | - Mohanad Abuhattab
- grid.267468.90000 0001 0695 7223Department of Biomedical Engineering, University of Wisconsin-Milwaukee, College of Engineering and Applied Sciences, 3200 N Cramer St, Milwaukee, WI 53211 USA
| | - Raman G. Kutty
- Department of Internal Medicine, White River Health System, Batesville, AR USA
| | - Priyatha Premnath
- Department of Biomedical Engineering, University of Wisconsin-Milwaukee, College of Engineering and Applied Sciences, 3200 N Cramer St, Milwaukee, WI, 53211, USA.
| |
Collapse
|
42
|
Qiang M, Cai P, Ao M, Li X, Chen Z, Yu L. Polysaccharides from Chinese materia medica: Perspective towards cancer management. Int J Biol Macromol 2023; 224:496-509. [PMID: 36265542 DOI: 10.1016/j.ijbiomac.2022.10.139] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 10/10/2022] [Accepted: 10/15/2022] [Indexed: 11/05/2022]
Abstract
Cancer has always been a focus of global attention, and the difficulty of treatment and poor prognosis have always plagued humanity. Conventional chemotherapeutics and treatment with synthetic disciplines will cause adverse side effects and drug resistance. Therefore, searching for a safe, valid, and clinically effective drug is necessary. At present, some natural compounds have proved to have the potential to fight cancer. Polysaccharides obtained from Chinese materia medica are good anti-cancer ingredients. Polysaccharides are macromolecular compounds of equal or distinct monosaccharides with an α- or β-glycosidic bonds. The anti-cancer activity has been fully demonstrated in vivo and in vitro. However, Chinese materia medica polysaccharides are only used as adjuvant therapy for cancer-related diseases. Hence, this review mainly discusses the chemical composition, biological activity, absorption in vivo, and clinical application of Chinese materia medica polysaccharides. Also, we discussed the anti-cancer mechanism. We also discussed the current research's limitations on treating cancer with Chinese materia medica polysaccharides and insights into future research.
Collapse
Affiliation(s)
- Mengqin Qiang
- Department of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, China
| | - Pingjun Cai
- Department of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, China
| | - Mingyue Ao
- Department of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, China
| | - Xing Li
- Department of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, China
| | - Zhimin Chen
- Department of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, China.
| | - Lingying Yu
- Department of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, China.
| |
Collapse
|
43
|
D'costa M, Bothe A, Das S, Udhaya Kumar S, Gnanasambandan R, George Priya Doss C. CDK regulators—Cell cycle progression or apoptosis—Scenarios in normal cells and cancerous cells. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2023; 135:125-177. [PMID: 37061330 DOI: 10.1016/bs.apcsb.2022.11.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Serine/threonine kinases called cyclin-dependent kinases (CDKs) interact with cyclins and CDK inhibitors (CKIs) to control the catalytic activity. CDKs are essential controllers of RNA transcription and cell cycle advancement. The ubiquitous overactivity of the cell cycle CDKs is caused by a number of genetic and epigenetic processes in human cancer, and their suppression can result in both cell cycle arrest and apoptosis. This review focused on CDKs, describing their kinase activity, their role in phosphorylation inhibition, and CDK inhibitory proteins (CIP/KIP, INK 4, RPIC). We next compared the role of different CDKs, mainly p21, p27, p57, p16, p15, p18, and p19, in the cell cycle and apoptosis in cancer cells with respect to normal cells. The current work also draws attention to the use of CDKIs as therapeutics, overcoming the pharmacokinetic barriers of pan-CDK inhibitors, analyze new chemical classes that are effective at attacking the CDKs that control the cell cycle (cdk4/6 or cdk2). It also discusses CDKI's drawbacks and its combination therapy against cancer patients. These findings collectively demonstrate the complexity of cancer cell cycles and the need for targeted therapeutic intervention. In order to slow the progression of the disease or enhance clinical outcomes, new medicines may be discovered by researching the relationship between cell death and cell proliferation.
Collapse
Affiliation(s)
- Maria D'costa
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of Bio Sciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, India
| | - Anusha Bothe
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of Bio Sciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, India
| | - Soumik Das
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of Bio Sciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, India
| | - S Udhaya Kumar
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of Bio Sciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, India
| | - R Gnanasambandan
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of Bio Sciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, India.
| | - C George Priya Doss
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of Bio Sciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, India.
| |
Collapse
|
44
|
Cellular signals integrate cell cycle and metabolic control in cancer. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2023; 135:397-423. [PMID: 37061338 DOI: 10.1016/bs.apcsb.2023.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
Growth factors are the small peptides that can promote growth, differentiation, and survival of most living cells. However, aberrant activation of receptor tyrosine kinases by GFs can generate oncogenic signals, resulting in oncogenic transformation. Accumulating evidence support a link between GF/RTK signaling through the major signaling pathways, Ras/Erk and PI3K/Akt, and cell cycle progression. In response to GF signaling, the quiescent cells in the G0 stage can re-enter the cell cycle and become the proliferative stage. While in the proliferative stage, tumor cells undergo profound changes in their metabolism to support biomass production and bioenergetic requirements. Accumulating data show that the cell cycle regulators, specifically cyclin D, cyclin B, Cdk2, Cdk4, and Cdk6, and anaphase-promoting complex/cyclosome (APC/C-Cdh1) play critical roles in modulating various metabolic pathways. These cell cycle regulators can regulate metabolic enzyme activities through post-translational mechanisms or the transcriptional factors that control the expression of the metabolic genes. This fine-tune control allows only the relevant metabolic pathways to be active in a particular phase of the cell cycle, thereby providing suitable amounts of biosynthetic precursors available during the proliferative stage. The imbalance of metabolites in each cell cycle phase can induce cell cycle arrest followed by p53-induced apoptosis.
Collapse
|
45
|
Sensitization of cervical cancer cells to radiation by the cyclin-dependent kinase inhibitor dinaciclib. MEDICAL ONCOLOGY (NORTHWOOD, LONDON, ENGLAND) 2022; 40:68. [PMID: 36586018 DOI: 10.1007/s12032-022-01890-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 11/08/2022] [Indexed: 01/01/2023]
Abstract
Dinaciclib is a selective cyclin-dependent kinase inhibitor, but its radiosensitizing effect remains unclear. The aim of this study is to investigate the radiosensitizing effect of Dinaciclib on cervical cancer cells. Two cervical cancer cell lines, Hela and Siha, were selected, and the IC50 was determined by CCK8. The radiosensitizing effect of Dinaciclib was verified by plate cloning assay, and the G2/M phase arrest and apoptosis of IR cells were verified by flow cytometry. Immunofluorescence assay was used to verify the formation of γH2AX foci following DNA damage. Western blot was performed to detect cell cycle, apoptosis, autophagy, and DNA damage-related pathways. Dinaciclib increased the cell sensitivity to IR. IR induced G2/M phase arrest and apoptosis, and Dinaciclib enhanced this effect. Further, Dinaciclib delayed DNA repair, including non-homologous end joining repair and homologous recombination repair, and reduced the expression of DNA repair proteins Ku80 (SiHa cells), Ku70, and RAD51, as well as the expression of apoptotic marker Bcl-2. The expression of autophagy marker Beclin1 induced tumor cell death and increased the formation of DNA damage marker γH2AX foci. Dinaciclib improves the sensitivity of cervical cancer cells to IR by inducing cell cycle arrest, delaying DNA repair, and increasing apoptosis. However, further research is needed to unravel the complexity of DNA repair pathways.
Collapse
|
46
|
Kim CW, Lee HK, Nam MW, Choi Y, Choi KC. Overexpression of KiSS1 Induces the Proliferation of Hepatocarcinoma and Increases Metastatic Potential by Increasing Migratory Ability and Angiogenic Capacity. Mol Cells 2022; 45:935-949. [PMID: 36572562 PMCID: PMC9794555 DOI: 10.14348/molcells.2022.0105] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 09/10/2022] [Accepted: 09/30/2022] [Indexed: 12/28/2022] Open
Abstract
Liver cancer has a high prevalence, with majority of the cases presenting as hepatocellular carcinoma (HCC). The prognosis of metastatic HCC has hardly improved over the past decade, highlighting the necessity for liver cancer research. Studies have reported the ability of the KiSS1 gene to inhibit the growth or metastasis of liver cancer, but contradictory research results are also emerging. We, therefore, sought to investigate the effects of KiSS1 on growth and migration in human HCC cells. HepG2 human HCC cells were infected with lentivirus particles containing KiSS1. The overexpression of KiSS1 resulted in an increased proliferation rate of HCC cells. Quantitative polymerase chain reaction and immunoblotting revealed increased Akt activity, and downregulation of the G1/S phase cell cycle inhibitors. A significant increase in tumor spheroid formation with upregulation of β-catenin and CD133 was also observed. KiSS1 overexpression promoted the migratory, invasive ability, and metastatic capacity of the hepatocarcinoma cell line, and these effects were associated with changes in the expressions of epithelial mesenchymal transition (EMT)-related genes such as E-cadherin, N-cadherin, and slug. KiSS1 overexpression also resulted in dramatically increased tumor growth in the xenograft mouse model, and upregulation of proliferating cell nuclear antigen (PCNA) and Ki-67 in the HCC tumors. Furthermore, KiSS1 increased the angiogenic capacity by upregulation of the vascular endothelial growth factor A (VEGF-A) and CD31. Based on these observations, we infer that KiSS1 not only induces HCC proliferation, but also increases the metastatic potential by increasing the migratory ability and angiogenic capacity.
Collapse
Affiliation(s)
- Cho-Won Kim
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Korea
| | - Hong Kyu Lee
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Korea
| | - Min-Woo Nam
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Korea
| | - Youngdong Choi
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Korea
| | - Kyung-Chul Choi
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Korea
| |
Collapse
|
47
|
Shaw R, Dutta B, Ghosh D, Bandopadhyay R. Hepatitis C—Route of Asymptomatic to Symptomatic Switch in Raising Hepatocarcinogenesis: Revisiting Nobel Prize 2020 in Physiology and Medicine. NATIONAL ACADEMY SCIENCE LETTERS 2022. [DOI: 10.1007/s40009-022-01192-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
48
|
Sun SJ, Deng P, Peng CE, Ji HY, Mao LF, Peng LZ. Selenium-Modified Chitosan Induces HepG2 Cell Apoptosis and Differential Protein Analysis. Cancer Manag Res 2022; 14:3335-3345. [PMID: 36465707 PMCID: PMC9716935 DOI: 10.2147/cmar.s382546] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 11/10/2022] [Indexed: 10/29/2023] Open
Abstract
INTRODUCTION Chitosan is the product of the natural polysaccharide chitin removing part of the acetyl group, and exhibits various physiological and bioactive functions. Selenium modification has been proved to further enhance the chitosan bioactivities, and has been a hot topic recently. METHODS The present study aimed to investigate the potential inhibitory mechanism of selenium-modified chitosan (SMC) on HepG2 cells through MTT assays, morphological observation, annexin V-FITC/PI double staining, mitochondrial membrane potential determination, cell-cycle detection, Western blotting, and two-dimensional gel electrophoresis (2-DE). RESULTS The results indicated that SMC can induce HepG2 cell apoptosis with the cell cycle arrested in the S and G2/M phases and gradual disruption of mitochondrial membrane potential, reduce the expression of Bcl2, and improve the expression of Bax, cytochrome C, cleaved caspase 9, and cleaved caspase 3. Also, 2-DE results showed that tubulin α1 B chain, myosin regulatory light chain 12A, calmodulin, UPF0568 protein chromosome 14 open reading frame 166, and the cytochrome C oxidase subunit 5B of HepG2 cells were downregulated in HepG2 cells after SMC treatment. DISCUSSION These data suggested that HepG2 cells induced apoptosis after SMC treatment via blocking the cell cycle in the S and G2/M phases, which might be mediated through the mitochondrial apoptotic pathway. These results could be of benefit to future practical applications of SMC in the food and drug fields.
Collapse
Affiliation(s)
- Su-Jun Sun
- Key Laboratory of Agro-Products Processing Technology of Shandong Province, Key Laboratory of Novel Food Resources Processing Ministry of Agriculture, Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, Jinan, People’s Republic of China
| | - Peng Deng
- Key Laboratory of Agro-Products Processing Technology of Shandong Province, Key Laboratory of Novel Food Resources Processing Ministry of Agriculture, Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, Jinan, People’s Republic of China
| | - Chun-E Peng
- Key Laboratory of Agro-Products Processing Technology of Shandong Province, Key Laboratory of Novel Food Resources Processing Ministry of Agriculture, Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, Jinan, People’s Republic of China
| | - Hai-Yu Ji
- Center for Mitochondria and Healthy Aging, College of Life Sciences, Yantai University, Yantai, People’s Republic of China
| | - Long-Fei Mao
- Key Laboratory of Agro-Products Processing Technology of Shandong Province, Key Laboratory of Novel Food Resources Processing Ministry of Agriculture, Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, Jinan, People’s Republic of China
| | - Li-Zeng Peng
- Key Laboratory of Agro-Products Processing Technology of Shandong Province, Key Laboratory of Novel Food Resources Processing Ministry of Agriculture, Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, Jinan, People’s Republic of China
| |
Collapse
|
49
|
Zheng S, Pan B. Multilevel data integration and molecular docking approach to systematically elucidate the underlying pharmacological mechanisms of Er-Zhi-Wan against hepatocellular carcinoma. Aging (Albany NY) 2022; 14:8783-8804. [DOI: 10.18632/aging.204369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 10/27/2022] [Indexed: 11/09/2022]
Affiliation(s)
- Shaoyan Zheng
- Affiliated Foshan Maternity and Child Healthcare Hospital, Southern Medical University, Foshan 528000, P.R. China
- Traditional Chinese Medicine Department, Affiliated Foshan Maternity and Child Healthcare Hospital, Southern Medical University, Foshan 528000, P.R. China
| | - Botao Pan
- Affiliated Foshan Maternity and Child Healthcare Hospital, Southern Medical University, Foshan 528000, P.R. China
| |
Collapse
|
50
|
Chen M, Chen X, Huang G, Jiang Y, Gou Y, Deng J. Synthesis, anti-tumour activity, and mechanism of benzoyl hydrazine Schiff base-copper complexes. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.133730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|