1
|
Siebe P, Mark F, Alexander L, Michael SA, Susanne Z. Revealing robust antioxidant defences of a mycoparasitic Trichoderma species. Fungal Biol 2025; 129:101549. [PMID: 40222757 DOI: 10.1016/j.funbio.2025.101549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 02/07/2025] [Accepted: 02/10/2025] [Indexed: 04/15/2025]
Abstract
The fungal genus Trichoderma contains a vast array of species well known for their high opportunistic potential and adaptability to various ecological niches. The ability of many Trichoderma species to both colonize the rhizosphere and parasitize plant pathogenic fungi has led to their use in biological pathogen control for several decades. Reactive oxygen species (ROS) are linked to both the antagonism imposed by the mycoparasite Trichoderma and the elicited defence reaction by its fungal hosts during the mycoparasitic interaction. Trichoderma spp. likely tolerate higher levels of ROS compared with some of their host species, thereby giving them an advantage during the mycoparasitic interaction. In the present study, we investigated glutathione redox dynamics using the fluorescent reporter Grx1-roGFP2 stably expressed in Trichoderma asperellum following electrotransformation. Grx1-roGFP2 undergoes reversible changes in its excitation spectrum in response to variations in the cellular glutathione redox potential, providing a real-time indication of intracellular oxidative load. Considering the putative importance of ROS in mycoparasitic interactions, we performed live-cell imaging of the T. asperellum reporter strain interacting with the cereal pathogen Fusarium graminearum. Surprisingly, the glutathione redox potential did not change during this mycoparasitic interaction. We found no evidence that host-induced tip growth arrest within T. asperellum hyphae is induced by intracellular ROS accumulation. Furthermore, we show that the F. graminearum mycotoxins deoxynivalenol and zearalenone do not induce detectable changes in glutathione redox potential, even at very high concentrations. We infer that T. asperellum has a robust anti-oxidant defence system, supported by the observation that high concentrations of H2O2 are required to fully oxidize the reporter during in vivo calibration. We cannot rule out a role for ROS as a signal during mycoparasitic interactions, but, if present, this does not appear to be mediated by glutathione redox potential.
Collapse
Affiliation(s)
- Pierson Siebe
- Department of Microbiology, University of Innsbruck, Innsbruck, Austria.
| | - Fricker Mark
- Department of Biology, University of Oxford, Oxford, United Kingdom.
| | - Lichius Alexander
- Department of Microbiology, University of Innsbruck, Innsbruck, Austria; Inncellys GmbH, Mils, Austria.
| | | | - Zeilinger Susanne
- Department of Microbiology, University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
2
|
Gao Y, Chen X, Duan JA, Xiao P. A review of pharmacological mechanisms, challenges and prospects of macromolecular glycopeptides. Int J Biol Macromol 2025; 300:140294. [PMID: 39863220 DOI: 10.1016/j.ijbiomac.2025.140294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 01/13/2025] [Accepted: 01/22/2025] [Indexed: 01/27/2025]
Abstract
Macromolecular glycopeptides are natural products derived from various sources, distinguished by their structural diversity, multifaceted biological activities, and low toxicity. These compounds exhibit a wide range of biological functions, such as immunomodulation, antitumor effects, anti-inflammatory properties, antioxidant activity, and more. However, limited understanding of natural glycopeptides has hindered their development and practical application. To promote their advancement and utilization, it is crucial to thoroughly investigate the pharmacological mechanisms of glycopeptides and address the challenges in natural glycopeptide research. This review uniquely focuses on the primary biological activities and potential molecular mechanisms of glycopeptides as reported in recent literature. Moreover, we emphasize the current challenges in glycopeptide research, including extraction and isolation difficulties, purification challenges, structural analysis complexities, elucidation of structure-activity relationships, characterization of biosynthetic pathways, and ensuring bioavailability and stability. The future prospects for glycopeptide research are also explored. We argue that ongoing research into glycopeptides will significantly contribute to drug development and provide more effective therapeutic options and disease treatment alternatives for human health.
Collapse
Affiliation(s)
- Ye Gao
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Xiaoyi Chen
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Jin-Ao Duan
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, PR China.
| | - Ping Xiao
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, PR China.
| |
Collapse
|
3
|
Kim J, Gilbert JL, Lv WW, Du P, Pan H. Reduction reactions dominate the interactions between Mg alloys and cells: Understanding the mechanisms. Bioact Mater 2025; 45:363-387. [PMID: 39687558 PMCID: PMC11647666 DOI: 10.1016/j.bioactmat.2024.11.020] [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: 09/08/2024] [Revised: 11/07/2024] [Accepted: 11/15/2024] [Indexed: 12/18/2024] Open
Abstract
Magnesium (Mg) alloys are popular biodegradable metals studied for orthopedic and cardiovascular applications, mainly because Mg ions are essential trace elements known to promote angiogenesis and osteogenesis. However, Mg corrosion consists of oxidation and reduction reactions that produce by-products, such as hydrogen gas, reactive oxygen species, and hydroxides. It is still unclear how all these by-products and Mg ions concomitantly alter the microenvironment and cell behaviors spatially and temporally. This study shows that Mg corrosion can enhance cell proliferation by reducing intracellular ROS. However, Mg cannot decrease ROS and promote cell proliferation in simulated inflammatory conditions, meaning the microenvironment is critical. Furthermore, cells may respond to Mg ions differently in chronic or acute alkaline pH or oxidative stress. Depending on the corrosion rate, Mg modulates HIF1α and many signaling pathways like PI3K/AKT/mTOR, mitophagy, cell cycle, and oxidative phosphorylation. Therefore, this study provides a fundamental insight into the importance of reduction reactions in Mg alloys.
Collapse
Affiliation(s)
- Jua Kim
- Shenzhen Key Laboratory of Marine Biomaterials, CAS-HK Joint Lab of Biomaterials, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Shenzhen University Town, Shenzhen, 518055, PR China
- Syracuse Biomaterials Institute, Syracuse University, 318 Bowne Hall, Syracuse University, Syracuse, NY, 13244, USA
| | - Jeremy L. Gilbert
- Syracuse Biomaterials Institute, Syracuse University, 318 Bowne Hall, Syracuse University, Syracuse, NY, 13244, USA
- Clemson- Medical University of South Carolina Bioengineering Program, Department of Bioengineering, Clemson University, 68 Presidents St, Charleston, SC, 39425, USA
| | - William W. Lv
- Department of Orthopaedics and Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, 999077, PR China
| | - Ping Du
- Shenzhen Key Laboratory of Marine Biomaterials, CAS-HK Joint Lab of Biomaterials, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Shenzhen University Town, Shenzhen, 518055, PR China
| | - Haobo Pan
- Shenzhen Key Laboratory of Marine Biomaterials, CAS-HK Joint Lab of Biomaterials, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Shenzhen University Town, Shenzhen, 518055, PR China
- Shenzhen Healthemes Biotechnology Co. Ltd, Shenzhen, 518102, PR China
- College of Chemical Engineering, Xinjiang Normal University, Urumqi, 830054, PR China
| |
Collapse
|
4
|
Reddan B, Cummins EP. The regulation of cell metabolism by hypoxia and hypercapnia. J Biol Chem 2025; 301:108252. [PMID: 39914740 PMCID: PMC11923829 DOI: 10.1016/j.jbc.2025.108252] [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: 09/18/2024] [Revised: 01/13/2025] [Accepted: 01/25/2025] [Indexed: 03/06/2025] Open
Abstract
Every cell in the body is exposed to a certain level of CO2 and O2. Hypercapnia and hypoxia elicit stress signals to influence cellular metabolism and function. Both conditions exert profound yet distinct effects on metabolic pathways and mitochondrial dynamics, highlighting the need for cells to adapt to changes in the gaseous microenvironment. The interplay between hypercapnia and hypoxia signaling is the key for dictating cellular homeostasis as microenvironmental CO2 and O2 levels are inextricably linked. Hypercapnia, characterized by elevated pCO2, introduces metabolic adaptations within the aerobic metabolism pathways, affecting tricarboxylic acid cycle flux, lipid, and amino acid metabolism, oxidative phosphorylation and the electron transport chain. Hypoxia, defined by reduced oxygen availability, necessitates a shift from oxidative phosphorylation to anaerobic glycolysis to sustain ATP production, a process orchestrated by the stabilization of hypoxia-inducible factor-1α. Given that hypoxia and hypercapnia are present in both physiological and cancerous microenvironments, how might the coexistence of hypercapnia and hypoxia influence metabolic pathways and cellular function in physiological niches and the tumor microenvironment?
Collapse
Affiliation(s)
- Ben Reddan
- School of Medicine, University College Dublin, Dublin, Ireland; Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Eoin P Cummins
- School of Medicine, University College Dublin, Dublin, Ireland; Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland.
| |
Collapse
|
5
|
Barbosa MAG, Kruschel RD, Almeida MJ, Pereira RF, Xavier CPR, McCarthy FO, Vasconcelos MH. Isoquinolinequinone N-oxides with diverging mechanisms of action induce collateral sensitivity against multidrug resistant cancer cells. Eur J Pharmacol 2025; 988:177234. [PMID: 39725135 DOI: 10.1016/j.ejphar.2024.177234] [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: 07/22/2024] [Revised: 12/11/2024] [Accepted: 12/23/2024] [Indexed: 12/28/2024]
Abstract
Multidrug resistance (MDR) is a major challenge in cancer research. Collateral sensitizers, compounds that exploit the enhanced defense mechanisms of MDR cells as weaknesses, are a proposed strategy to overcome MDR. Our previous work reported the synthesis of two novel Isoquinolinequinone (IQQ) N-oxides that induce collateral sensitivity in MDR ABCB1-overexpressing non-small cell lung cancer (NSCLC) and colorectal cancer cells. Herein, we aimed to investigate underlying mechanisms of antitumor and collateral sensitivity activity of these compounds. We evaluated their effect on cancer cell viability, proliferation, cell cycle profile, and studied their cytotoxicity in non-tumorigenic cells. Their antitumor effect was further studied using NSCLC and colorectal cancer MDR spheroids. To understand underlying collateral sensitivity mechanisms, we assessed the effect on rhodamine-123 accumulation, ROS production, GSH/GSSG balance and expression of key proteins associated with metabolism and redox balance. Both compounds reduced the viability of MDR cells, as 2D cultures or as spheroids, without decreasing the growth of a human nontumorigenic cell line, and increased rhodamine-123 accumulation in MDR NCI-H460/R cells. Moreover, RK2 increased ROS, disrupted GSH balance, and altered expression of proteins associated with oxidative stress protection, particularly in NCI-H460/R cells. The collateral sensitivity effect of RK3 could not be attributed to redox balance disruption, but increased IDH1 expression following treatment suggests a potential metabolic shift in MDR cells. These findings highlight RK2 and RK3 as promising candidates for next stages of drug development. Their distinct mechanisms of action could lead to therapeutic solutions for MDR-related cancers, specifically linked to ABCB1 overexpression.
Collapse
Affiliation(s)
- Mélanie A G Barbosa
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135, Porto, Portugal; Cancer Drug Resistance Group, IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, 4200-135, Porto, Portugal; FFUP - Faculty of Pharmacy of the University of Porto, 4050-313, Porto, Portugal
| | - Ryan D Kruschel
- School of Chemistry, Analytical and Biological Chemistry Research Facility, University College Cork, Cork, T12 K8AF, Ireland
| | - Maria João Almeida
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135, Porto, Portugal; Cancer Drug Resistance Group, IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, 4200-135, Porto, Portugal
| | - Rúben F Pereira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135, Porto, Portugal; Biofabrication Group, INEB-Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal; ICBAS-Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua Jorge de Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - Cristina P R Xavier
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135, Porto, Portugal; Cancer Drug Resistance Group, IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, 4200-135, Porto, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Toxicologic Pathology Research Laboratory, University Institute of Health Sciences (1H-TOXRUN, IUCS-CESPU), Gandra, 4585-116, Portugal; Associate Laboratory i4HB - Institute for Health and Bioeconomy, University Institute of Health Sciences - CESPU, Gandra, 4585-116, Portugal
| | - Florence O McCarthy
- School of Chemistry, Analytical and Biological Chemistry Research Facility, University College Cork, Cork, T12 K8AF, Ireland.
| | - M Helena Vasconcelos
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135, Porto, Portugal; Cancer Drug Resistance Group, IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, 4200-135, Porto, Portugal; FFUP - Faculty of Pharmacy of the University of Porto, 4050-313, Porto, Portugal.
| |
Collapse
|
6
|
Javar N, Sadat Shandiz SA, Abbasi M. Phyto-mediated fabrication of silver nanoparticles from Scrophularia striata extract (AgNPs-SSE): a potential inducer of apoptosis in breast cancer cells. Mol Biol Rep 2025; 52:172. [PMID: 39878805 DOI: 10.1007/s11033-025-10298-5] [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: 12/16/2024] [Accepted: 01/23/2025] [Indexed: 01/31/2025]
Abstract
BACKGROUND Breast carcinoma stands out as the most widespread invasive cancer and the top contributor to cancer-related mortality in women. Nanoparticles have emerged as promising tools in cancer detection, diagnosis, and prevention. In this study, the antitumor and apoptotic capability of silver nanoparticles synthesized through Scrophularia striata extract (AgNPs-SSE) was investigated toward breast cancer cells. METHODS The produced AgNPs-SSE were identified using scanning electron micrograph (SEM), energy-dispersive X-ray (EDAX) spectroscopy, Fourier-transform infrared (FTIR) spectroscopy, and X-ray diffraction (XRD). The cell-killing effects of AgNPs-SSE on MDA-MB231 mammary carcinoma cells were evaluated in vitro using the MTT assay over 24 h. Apoptosis induction was conducted by cell cycle analysis, annexin V-FITC/PI staining, reactive oxygen species (ROS) generation, and Hoechst staining. Additionally, the gene expression of βcatenin, GSK3β, and CyclinD1 was analyzed using quantitative real-time PCR (qRT-PCR). RESULTS Microscopic analysis confirmed the successful fabrication of globular AgNPs-SSE, with a mean particle dimension of 19 ± 10 nm. The MTT assay revealed that IC50 value of AgNPs-SSE was 48.5 µg/mL for mammary carcinoma cells and 114 µg/mL for normal cells. Annexin V-FITC/PI staining specified that 85.88% of cancer cells treated with AgNPs-SSE underwent either early or late apoptosis. Treatment with AgNPs-SSE also caused a considerable rise in the subG1 cell cycle population and ROS production. Furthermore, the upregulation of βcatenin and downregulation of CyclinD1 gene expression confirmed the apoptotic mechanism. CONCLUSIONS In conclusion, the findings suggest that phyto-synthesized AgNPs-SSE can restrain the expansion of breast carcinoma cells and provoke apoptosis through oxidative stress. These results highlight the potential of AgNPs-SSE as an antitumor agent against breast cancer.
Collapse
Affiliation(s)
- Niloufar Javar
- Department of Biology, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| | | | - Maryam Abbasi
- Department of Biology, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| |
Collapse
|
7
|
Mahmoudi A, Pour VZ, Salehzadeh A. A novel cobalt oxide nanoparticle conjugated with ellagic acid arrests the cell cycle in human liver cancer cell line. Sci Rep 2025; 15:1797. [PMID: 39805967 PMCID: PMC11730305 DOI: 10.1038/s41598-025-85312-8] [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: 08/30/2024] [Accepted: 01/01/2025] [Indexed: 01/16/2025] Open
Abstract
The current chemotherapy treatments for liver cancer have shown limited effectiveness. Therefore, there is an urgent need to develop new drugs to combat this disease more effectively. This study reports synthesis of cobalt oxide nanoparticles coated with glucose, and conjugated with Ellagic acid. Physicochemical characterization of Co3O4@Glu-Ellagic acid nanoparticles was done using FT-IR, XRD, SEM, TEM, TGA, EDS-mapping, DLS, and zeta potential analyses, and the investigation of their anticancer potential on liver cancer cell lines involved the use of MTT, flow cytometry, and cell cycle analysis. The synthesized nanoparticles were somewhat spherical, arranged in a relatively cluster-shaped form, and were 33-46 nm in diameter. The zeta potential and particle hydrodynamic size were - 5.43 and 169 nm, respectively and had no elemental impurity. Also, the synthesized particles had proper thermal stability at temperatures below 100 °C. Treating cancer cells with the nanoparticles considerably increased ROS levels by 2.6 folds. Compared to normal human cells, Co3O4@Glu-Ellagic acid nanoparticles showed significantly higher toxicity for liver cancer and the 50% inhibitory concentration was 94 and 187 µg/mL for the cancer and normal cells, respectively. Co3O4@Glu-Ellagic acid increased cell apoptosis, from 0.87 to 9.24%, and the cells were mainly arrested at the G0/G1 and G2/M phases. Overall, the present work indicated that Co3O4@Glu-Ellagic acid has antiproliferative effects on liver cancer cells through an increased oxidative stress level, inhibition of cell cycle, and apoptosis induction.
Collapse
Affiliation(s)
- Ayda Mahmoudi
- Department of Biology, Damghan Branch, Islamic Azad University, Damghan, Iran
| | - Vajiheh Zarrin Pour
- Department of Biology, Damghan Branch, Islamic Azad University, Damghan, Iran.
| | - Ali Salehzadeh
- Department of Biology, Rasht Branch, Islamic Azad University, Rasht, Iran.
| |
Collapse
|
8
|
Paredes-Hernández U, Aguilar-Peña LV, Isaac-Olivé K, Ocampo-García B, Contreras I, Estrada JA, Izquierdo G, Morales-Avila E, Aranda-Lara L. Enhancing photodynamic and radionuclide therapy by small interfering RNA (siRNA)-RAD51 transfection via self-emulsifying delivery systems (SNEDDS). Cytotherapy 2025; 27:66-77. [PMID: 39186024 DOI: 10.1016/j.jcyt.2024.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 07/22/2024] [Accepted: 08/05/2024] [Indexed: 08/27/2024]
Abstract
BACKGROUND AIMS Gene-silencing by small interfering RNA (siRNA) is an attractive therapy to regulate cancer death, tumor recurrence or metastasis. Because siRNAs are easily degraded, it is necessary to develop transport and delivery systems to achieve efficient tumor targeting. Self-nanoemulsifying systems (SNEDDS) have been successfully used for pDNA transport and delivery, so they may be useful for siRNA. The aim of this work is to introduce siRNA-RAD51 into a SNEDDS prepared with Phospholipon-90G, Labrafil-M1944-CS and Cremophor-RH40 and evaluate its efficacy in preventing homologous recombination of DNA double-strand breaks caused by photodynamic therapy (PDT) and ionizing radiation (IR). METHODS The siRNA-RAD51 was loaded into SNEDDS using chitosan. Transfection capacity was estimated by comparison with Lipofectamine-2000. RESULTS SNEDDS(siRNA-RAD51) induced gene silencing effect on the therapies evaluated by cell viability and clonogenic assays using T47D breast cancer cells. CONCLUSIONS SNEDDS(siRNA-RAD51) shown to be an effective siRNA-delivery system to decrease cellular resistance in PDT or IR.
Collapse
Affiliation(s)
- Ulises Paredes-Hernández
- Laboratorio de Investigación en Teranóstica, Facultad de Medicina, Universidad Autónoma del Estado de México, Toluca, Estado de México, Mexico
| | - Leslie V Aguilar-Peña
- Laboratorio de Investigación en Teranóstica, Facultad de Medicina, Universidad Autónoma del Estado de México, Toluca, Estado de México, Mexico
| | - Keila Isaac-Olivé
- Laboratorio de Investigación en Teranóstica, Facultad de Medicina, Universidad Autónoma del Estado de México, Toluca, Estado de México, Mexico
| | - Blanca Ocampo-García
- Departamento de Materiales Radiactivos, Instituto Nacional de Investigaciones Nucleares, Ocoyoacac, Estado de México, Mexico
| | - Irazú Contreras
- Laboratorio de Neuroquímica, Facultad de Medicina, Universidad Autónoma del Estado de México, Toluca, Estado de México, Mexico
| | - José A Estrada
- Laboratorio de Neuroquímica, Facultad de Medicina, Universidad Autónoma del Estado de México, Toluca, Estado de México, Mexico
| | - Germán Izquierdo
- Facultad de Ciencias, Universidad Autónoma del Estado de México, Toluca, Estado de México, Mexico
| | - Enrique Morales-Avila
- Laboratorio de Toxicología y Farmacia, Facultad de Química, Universidad Autónoma del Estado de México, Toluca, Estado de México, Mexico.
| | - Liliana Aranda-Lara
- Laboratorio de Investigación en Teranóstica, Facultad de Medicina, Universidad Autónoma del Estado de México, Toluca, Estado de México, Mexico.
| |
Collapse
|
9
|
Wang G, Jin W, Zhang L, Dong M, Zhang X, Zhou Z, Wang X. SLC50A1 inhibits the doxorubicin sensitivity in hepatocellular carcinoma cells through regulating the tumor glycolysis. Cell Death Discov 2024; 10:495. [PMID: 39695152 DOI: 10.1038/s41420-024-02261-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 11/20/2024] [Accepted: 12/03/2024] [Indexed: 12/20/2024] Open
Abstract
Metabolic reprogramming has been found to be closely associated with the occurrence and development of hepatocellular carcinoma (HCC). The relationship between SLC50A1, a member of the SLC family involved in glucose transmembrane transport, and HCC remains unclear. This study aims to investigate the function and underlying mechanisms of SLC50A1 in the occurrence and progression of HCC. Based on bioinformatics analysis and clinical sample testing, we observed a significant upregulation of SLC50A1 in HCC, which is correlated with unfavorable prognosis in HCC patients. Additionally, there is a noticeable correlation between the expressions of SLC50A1 and METTL3. Further in vitro and in vivo experiments confirmed that SLC50A1 can regulate cellular glycolysis and the cell cycle, thereby promoting the proliferation of HCC cells while reducing apoptosis. Moreover, our findings indicate that SLC50A1 enhances resistance of HCC cells to DOX and 2-DG. Furthermore, we discovered that the m6A methyltransferase METTL3 mediates the methylation modification of SLC50A1. The recognition and binding of the modified SLC50A1 by IGF2BP2 subsequently promote its stability and translational expression. Consequently, our research identifies the METTL3/SLC50A1 axis as a novel therapeutic target in the context of HCC.
Collapse
Affiliation(s)
- Ganggang Wang
- Department of Hepatobiliary Surgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Wenzhi Jin
- Department of Hepatobiliary Surgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Lianmei Zhang
- Department of Hepatobiliary Surgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Meiyuan Dong
- Graduate School of Hebei Medical University, Shijiazhuang, People's Republic of China
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Xin Zhang
- Department of Hepatobiliary Surgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Zhijie Zhou
- Department of Hepatobiliary Surgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Xiaoliang Wang
- Department of Hepatobiliary Surgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China.
| |
Collapse
|
10
|
YIN YUAN, WANG ZHENGYIN, HU YUJIE, WANG JIA, WANG YI, LU QUN. Caffeic acid hinders the proliferation and migration through inhibition of IL-6 mediated JAK-STAT-3 signaling axis in human prostate cancer. Oncol Res 2024; 32:1881-1890. [PMID: 39574470 PMCID: PMC11576972 DOI: 10.32604/or.2024.048007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 03/26/2024] [Indexed: 11/24/2024] Open
Abstract
Background Caffeic acid (CA) is considered a promising phytochemical that has inhibited numerous cancer cell proliferation. Therefore, it is gaining increasing attention due to its safe and pharmacological applications. In this study, we investigated the role of CA in inhibiting the Interleukin-6 (IL-6)/Janus kinase (JAK)/Signal transducer and activator of transcription-3 (STAT-3) mediated suppression of the proliferation signaling in human prostate cancer cells. Materials and Methods The role of CA in proliferation and colony formation abilities was studied using 3-[4,5-dimethylthiazol-2-yl]-2,5 diphenyl tetrazolium bromide (MTT) assay and colony formation assays. Tumour cell death and cell cycle arrest were identified using flow cytometry techniques. CA treatment-associated protein expression of mitogen-activated protein kinase (MAPK) families, IL-6/JAK/STAT-3, proliferation, and apoptosis protein expressions in PC-3 and LNCaP cell lines were measured using Western blot investigation. Results We have obtained that treatment with CA inhibits prostate cancer cells (PC-3 and LNCaP) proliferation and induces reactive oxygen species (ROS), cell cycle arrest, and apoptosis cell death in a concentration-dependent manner. Moreover, CA treatment alleviates the expression phosphorylated form of MAPK families, i.e., extracellular signal-regulated kinase 1 (ERK1), c-Jun N-terminal kinase (JNK), and p38 in PC-3 cells. IL-6 mediated JAK/STAT3 expressions regulate the proliferation and antiapoptosis that leads to prostate cancer metastasis and migration. Therefore, to mitigate the expression of IL-6/JAK/STAT-3 is considered an important target for the treatment of prostate cancer. In this study, we have observed that CA inhibits the expression of IL-6, JAK1, and phosphorylated STAT-3 in both PC-3 and LNCaP cells. Due to the inhibitory effect of IL-6/JAK/STAT-3, it resulted in decreased expression of cyclin-D1, cyclin-D2, and CDK1 in both PC-3 cells. In addition, CA induces apoptosis by enhancing the expression of Bax and caspase-3; and decreased expression of Bcl-2 in prostate cancer cells. Conclusions Thus, CA might act as a therapeutical application against prostate cancer by targeting the IL-6/JAK/STAT3 signaling axis.
Collapse
Affiliation(s)
- YUAN YIN
- Department of Laboratory Medicine, Shanghai Traditional Chinese Medicine-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200082, China
| | - ZHENGYIN WANG
- Department of Laboratory Medicine, Shanghai Traditional Chinese Medicine-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200082, China
| | - YUJIE HU
- Department of Laboratory Medicine, Shanghai Traditional Chinese Medicine-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200082, China
| | - JIA WANG
- Department of Laboratory Medicine, Shanghai Traditional Chinese Medicine-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200082, China
| | - YI WANG
- Department of Acupuncture and Moxibustion, Shanghai Traditional Chinese Medicine-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200082, China
| | - QUN LU
- Department of Laboratory Medicine, Shanghai Traditional Chinese Medicine-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200082, China
| |
Collapse
|
11
|
Paliwal K, Swain A, Mishra DP, Sudhadevi Antharjanam PK, Kumar M. A novel copper(II) complex with a salicylidene carbohydrazide ligand that promotes oxidative stress and apoptosis in triple negative breast cancer cells. Dalton Trans 2024; 53:17702-17720. [PMID: 39420621 DOI: 10.1039/d4dt01914h] [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: 10/19/2024]
Abstract
We report the synthesis, characterization, anti-cancer activity and mechanism of action of a novel water-soluble Cu(II) complex with salicylidene carbohydrazide as the ligand and o-phenanthroline as the co-ligand. The synthesized complex (1) was characterized by FT-IR, EPR, and electronic spectroscopy, as well as single crystal X-ray diffraction. This compound was found to be paramagnetic from EPR spectra and X-ray crystallography revealed that the molecule crystallized in an orthorhombic crystal system. The crystal lattice was asymmetric containing two distinct binuclear copper complexes containing the Schiff base as the major ligand, o-phenanthroline as a co-ligand, two nitrate anions, and two water molecules. The Cu(II) in the first site coordinated with the enolised ligand comprising enolate O-, phenolate O-, and the imine N and N,N from o-phen. The major part of this complex exists as Cu(II) coordinated with two H2O molecules at the second site with nitrate acting as the counter anion. However, a smaller portion of the complex exists where Cu(II) is coordinated with NO3- and H2O, and the remaining water molecule acts as lattice water. It was tested for DNA binding and cleavage properties which revealed that it binds in an intercalative mode to CT-DNA with Kb value of 1.25 × 104 M-1. Furthermore, cleavage studies reveal that the complex has potential for efficient DNA cleavage under both oxidative and hydrolytic conditions. It was able to enhance the rate of cleavage by 2.8 × 108 times. The complex shows good cytotoxicity to breast cancer monolayer (2D) as well as spheroid (3D) systems. The IC50 values for MDA-MB-231 and MCF-7 monolayer culture was calculated as 1.86 ± 0.17 μM and 2.22 ± 0.08 μM, respectively, and in (3D) spheroids of MDA-MB-231 cells, the IC50 value was calculated to be 1.51 ± 0.29 μM. It was observed that the complex outperformed cisplatin in both breast cancer cell lines. The cells treated with complex 1 underwent severe DNA damage, increased oxidative stress and cell cycle arrest which finally led to programmed cell death or apoptosis in triple negative breast cancer cells through an intrinsic pathway.
Collapse
Affiliation(s)
- Kumudini Paliwal
- Department of Chemical Engineering, Birla Institute of Technology and Science-Pilani, K.K. Birla Goa Campus, Zuarinagar, Goa 403726, India.
| | - Abinash Swain
- Cell Death Research Laboratory, Endocrinology Division, CSIR-Central Drug Research Institute, B.S. 10/1, Sector-10, Jankipuram Extension, Lucknow, Uttar Pradesh 226031, India
| | - Durga Prasad Mishra
- Cell Death Research Laboratory, Endocrinology Division, CSIR-Central Drug Research Institute, B.S. 10/1, Sector-10, Jankipuram Extension, Lucknow, Uttar Pradesh 226031, India
| | - P K Sudhadevi Antharjanam
- Sophisticated Analytical Instrument Facility, Indian Institute of Technology-Madras, Chennai 600 036, India
| | - Manjuri Kumar
- Department of Chemical Engineering, Birla Institute of Technology and Science-Pilani, K.K. Birla Goa Campus, Zuarinagar, Goa 403726, India.
| |
Collapse
|
12
|
Dinarvand M, Sharifnia F, Jangravi Z. Reactive oxygen species (ROS) are a crucial factor in the anticancer activity of Oliveria decumbens extract against the A431 human skin cell line. ARCHIVES OF RAZI INSTITUTE 2024; 79:749-754. [PMID: 40256584 PMCID: PMC12004055 DOI: 10.32592/ari.2024.79.4.749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 01/12/2024] [Indexed: 04/22/2025]
Abstract
Globally, skin cancer is a main public health challenge whose incidence is continuously increasing. Given the limitations of conventional t herapies, new research and novel therapies may be promising for reducing skin cancer morbidity and mortality. Phytochemicals are attractive resources for new therapy design in cancer research due to their cost-effectiveness and lower side effects. In the present study, the anti-cancer activity of Oliveria decumbens (O. decumbens) extract was investigated on the human skin cancer A431 cell line A431. The aqueous extract of the O. decumbens plant was prepared using the traditional method. Then IC50 was determined using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide (MTT) assay under different concentrations of O. decumbens. Cell apoptosis was investigated by Annexin V-FITC/Propidium Iodide (PI) and flow cytometry. Cell cycle was investigated by PI staining and flow cytometry. Reactive oxygen species (ROS) production was analyzed by DCFH-DA (2', 7' -dichlorofluorescein-diacetate) staining and flowcytometry.IC50 for cell viability was determined 475g/ml. Cell cycle analyses showed G1 arrest in treated cells compared to control cell. Results also confirmed significant increase of apoptotic cells (8.2%1, P<0.05) under IC50 concentration of the extract in comparison to the control group (2.50.99%). A significant increase in ROS level was observed in O.ecumbens treated cells compared to control cells (738 170 vs 31655 in the control group, P<0.05.).Overall, the present results indicate that O. decumbens extract can inhibit skin cancer cell proliferation via inhibition of cell cycle and apoptosis. It seems that ROS production plays a critical role in the anticancer effect of O. decumbens extract. Therefore, its potential option for future treatment of skin cancer should be considered.
Collapse
Affiliation(s)
- M Dinarvand
- Department of biology, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - F Sharifnia
- Department of biology, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Z Jangravi
- Department of Biochemistry, Faculty of Medicine, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
13
|
Taghizadeh-Hesary F. "Reinforcement" by Tumor Microenvironment: The Seventh "R" of Radiobiology. Int J Radiat Oncol Biol Phys 2024; 119:727-733. [PMID: 38032584 DOI: 10.1016/j.ijrobp.2023.09.027] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 09/11/2023] [Accepted: 09/16/2023] [Indexed: 12/01/2023]
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.
| |
Collapse
|
14
|
Patra S, Naik PP, Mahapatra KK, Alotaibi MR, Patil S, Patro BS, Sethi G, Efferth T, Bhutia SK. Recent advancement of autophagy in polyploid giant cancer cells and its interconnection with senescence and stemness for therapeutic opportunities. Cancer Lett 2024; 590:216843. [PMID: 38579893 DOI: 10.1016/j.canlet.2024.216843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 02/11/2024] [Accepted: 04/01/2024] [Indexed: 04/07/2024]
Abstract
Recurrent chemotherapy-induced senescence and resistance are attributed to the polyploidization of cancer cells that involve genomic instability and poor prognosis due to their unique form of cellular plasticity. Autophagy, a pre-dominant cell survival mechanism, is crucial during carcinogenesis and chemotherapeutic stress, favouring polyploidization. The selective autophagic degradation of essential proteins associated with cell cycle progression checkpoints deregulate mitosis fidelity and genomic integrity, imparting polyploidization of cancer cells. In connection with cytokinesis failure and endoreduplication, autophagy promotes the formation, maintenance, and generation of the progeny of polyploid giant cancer cells. The polyploid cancer cells embark on autophagy-guarded elevation in the expression of stem cell markers, along with triggered epithelial and mesenchymal transition and senescence. The senescent polyploid escapers represent a high autophagic index than the polyploid progeny, suggesting regaining autophagy induction and subsequent autophagic degradation, which is essential for escaping from senescence/polyploidy, leading to a higher proliferative phenotypic progeny. This review documents the various causes of polyploidy and its consequences in cancer with relevance to autophagy modulation and its targeting for therapeutic intervention as a novel therapeutic strategy for personalized and precision medicine.
Collapse
Affiliation(s)
- Srimanta Patra
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology Rourkela, Rourkela, 769008, Odisha, India
| | - Prajna Paramita Naik
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology Rourkela, Rourkela, 769008, Odisha, India; Department of Zoology Panchayat College, Bargarh, 768028, Odisha, India
| | - Kewal Kumar Mahapatra
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology Rourkela, Rourkela, 769008, Odisha, India; Department of Agriculture and Allied Sciences (Zoology), C. V. Raman Global University, Bhubaneswar, 752054, Odisha, India
| | - Moureq Rashed Alotaibi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Shankargouda Patil
- College of Dental Medicine, Roseman University of Health Sciences, South Jordan, UT, 84095, USA
| | - Birija Sankar Patro
- Chemical Biology Section, Bio-Organic Division, Bhabha Atomic Research Centre, Mumbai, 400085, India
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117600, Singapore
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, 55128, Mainz, Germany
| | - Sujit Kumar Bhutia
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology Rourkela, Rourkela, 769008, Odisha, India.
| |
Collapse
|
15
|
Zafeiropoulou K, Kalampounias G, Alexis S, Anastasopoulos D, Symeonidis A, Katsoris P. Autophagy and oxidative stress modulation mediate Bortezomib resistance in prostate cancer. PLoS One 2024; 19:e0289904. [PMID: 38412186 PMCID: PMC10898778 DOI: 10.1371/journal.pone.0289904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 01/27/2024] [Indexed: 02/29/2024] Open
Abstract
Proteasome inhibitors such as Bortezomib represent an established type of targeted treatment for several types of hematological malignancies, including multiple myeloma, Waldenstrom's macroglobulinemia, and mantle cell lymphoma, based on the cancer cell's susceptibility to impairment of the proteasome-ubiquitin system. However, a major problem limiting their efficacy is the emergence of resistance. Their application to solid tumors is currently being studied, while simultaneously, a wide spectrum of hematological cancers, such as Myelodysplastic Syndromes show minimal or no response to Bortezomib treatment. In this study, we utilize the prostate cancer cell line DU-145 to establish a model of Bortezomib resistance, studying the underlying mechanisms. Evaluating the resulting resistant cell line, we observed restoration of proteasome chymotrypsin-like activity, regardless of drug presence, an induction of pro-survival pathways, and the substitution of the Ubiquitin-Proteasome System role in proteostasis by induction of autophagy. Finally, an estimation of the oxidative condition of the cells indicated that the resistant clones reduce the generation of reactive oxygen species induced by Bortezomib to levels even lower than those induced in non-resistant cells. Our findings highlight the role of autophagy and oxidative stress regulation in Bortezomib resistance and elucidate key proteins of signaling pathways as potential pharmaceutical targets, which could increase the efficiency of proteasome-targeting therapies, thus expanding the group of molecular targets for neoplastic disorders.
Collapse
Affiliation(s)
- Kalliopi Zafeiropoulou
- Division of Genetics, Cell Biology and Development, Department of Biology, University of Patras, Patras, Greece
- Hematology Division, Department of Internal Medicine, University of Patras Medical School-University Hospital, Patras, Greece
| | - Georgios Kalampounias
- Division of Genetics, Cell Biology and Development, Department of Biology, University of Patras, Patras, Greece
| | - Spyridon Alexis
- Hematology Division, Department of Internal Medicine, University of Patras Medical School-University Hospital, Patras, Greece
| | - Daniil Anastasopoulos
- Division of Genetics, Cell Biology and Development, Department of Biology, University of Patras, Patras, Greece
| | - Argiris Symeonidis
- Hematology Division, Department of Internal Medicine, University of Patras Medical School-University Hospital, Patras, Greece
| | - Panagiotis Katsoris
- Division of Genetics, Cell Biology and Development, Department of Biology, University of Patras, Patras, Greece
| |
Collapse
|
16
|
Zhang R, Zhang C, Lu Q, Liang C, Tian M, Li Z, Yang Y, Li X, Deng Y. Cancer-cell-specific Self-Reporting Photosensitizer for Precise Identification and Ablation of Cancer Cells. Anal Chem 2024; 96:1659-1667. [PMID: 38238102 DOI: 10.1021/acs.analchem.3c04578] [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: 01/31/2024]
Abstract
Cancer-cell-specific fluorescent photosensitizers (PSs) are highly desired molecular tools for cancer ablation with minimal damage to normal cells. However, such PSs that can achieve cancer specification and ablation and a self-reporting manner concurrently are rarely reported and still an extremely challenging task. Herein, we have proposed a feasible strategy and conceived a series of fluorescent PSs based on simple chemical structures for identifying and killing cancer cells as well as monitoring the photodynamic therapy (PDT) process by visualizing the change of subcellular localization. All of the constructed cationic molecules could stain mitochondria in cancer cells, identify cancer cells specifically, and monitor cancer cell viability. Among these, IVP-Br has the strongest ability to produce ROS, which serves as a potent PS for specific recognition and killing of cancer cells. IVP-Br could translocate from mitochondria to the nucleolus during PDT, self-reporting the entire therapeutic process. Mechanism study confirms that IVP-Br with light irradiation causes cancer cell ablation via inducing cell cycle arrest, cell apoptosis, and autophagy. The efficient ablation of tumor through PDT induced by IVP-Br has been confirmed in the 3D tumor spheroid chip. Particularly, IVP-Br could discriminate cancer cells from white blood cells (WBCs), exhibiting great potential to identify circulating tumor cells (CTCs).
Collapse
Affiliation(s)
- Ruoyao Zhang
- School of Medical Technology, Institute of Engineering Medicine, Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, Beijing Institute of Technology, Beijing 100081, China
| | - Chen Zhang
- School of Medical Technology, Institute of Engineering Medicine, Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, Beijing Institute of Technology, Beijing 100081, China
| | - Qing Lu
- China Fire and Rescue Institute, Changping, Beijing 102202, China
| | - Chaohui Liang
- School of Medical Technology, Institute of Engineering Medicine, Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, Beijing Institute of Technology, Beijing 100081, China
| | - Minggang Tian
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan, Shandong 250022, China
| | - Zhao Li
- School of Medical Technology, Institute of Engineering Medicine, Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, Beijing Institute of Technology, Beijing 100081, China
| | - Yuanzhan Yang
- School of Medical Technology, Institute of Engineering Medicine, Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, Beijing Institute of Technology, Beijing 100081, China
| | - Xiaoqiong Li
- School of Medical Technology, Institute of Engineering Medicine, Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, Beijing Institute of Technology, Beijing 100081, China
| | - Yulin Deng
- School of Medical Technology, Institute of Engineering Medicine, Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, Beijing Institute of Technology, Beijing 100081, China
| |
Collapse
|
17
|
Imran H, Tang Y, Wang S, Yan X, Liu C, Guo L, Wang E, Xu C. Optimized DOX Drug Deliveries via Chitosan-Mediated Nanoparticles and Stimuli Responses in Cancer Chemotherapy: A Review. Molecules 2023; 29:31. [PMID: 38202616 PMCID: PMC10780101 DOI: 10.3390/molecules29010031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 12/15/2023] [Accepted: 12/16/2023] [Indexed: 01/12/2024] Open
Abstract
Chitosan nanoparticles (NPs) serve as useful multidrug delivery carriers in cancer chemotherapy. Chitosan has considerable potential in drug delivery systems (DDSs) for targeting tumor cells. Doxorubicin (DOX) has limited application due to its resistance and lack of specificity. Chitosan NPs have been used for DOX delivery because of their biocompatibility, biodegradability, drug encapsulation efficiency, and target specificity. In this review, various types of chitosan derivatives are discussed in DDSs to enhance the effectiveness of cancer treatments. Modified chitosan-DOX NP drug deliveries with other compounds also increase the penetration and efficiency of DOX against tumor cells. We also highlight the endogenous stimuli (pH, redox, enzyme) and exogenous stimuli (light, magnetic, ultrasound), and their positive effect on DOX drug delivery via chitosan NPs. Our study sheds light on the importance of chitosan NPs for DOX drug delivery in cancer treatment and may inspire the development of more effective approaches for cancer chemotherapy.
Collapse
Affiliation(s)
- HafizMuhammad Imran
- Department of Biochemistry, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; (H.I.); (Y.T.); (S.W.); (X.Y.); (C.L.); (L.G.)
| | - Yixin Tang
- Department of Biochemistry, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; (H.I.); (Y.T.); (S.W.); (X.Y.); (C.L.); (L.G.)
| | - Siyuan Wang
- Department of Biochemistry, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; (H.I.); (Y.T.); (S.W.); (X.Y.); (C.L.); (L.G.)
| | - Xiuzhang Yan
- Department of Biochemistry, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; (H.I.); (Y.T.); (S.W.); (X.Y.); (C.L.); (L.G.)
| | - Chang Liu
- Department of Biochemistry, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; (H.I.); (Y.T.); (S.W.); (X.Y.); (C.L.); (L.G.)
| | - Lei Guo
- Department of Biochemistry, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; (H.I.); (Y.T.); (S.W.); (X.Y.); (C.L.); (L.G.)
| | - Erlei Wang
- College of Food Science and Engineering, Jilin University, Changchun 130062, China
| | - Caina Xu
- Department of Biochemistry, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; (H.I.); (Y.T.); (S.W.); (X.Y.); (C.L.); (L.G.)
| |
Collapse
|
18
|
胡 桐, 勾 文, 任 中, 刘 改, 李 祎, 左 代, 侯 文. [Icaritin increases radiosensitivity of nasopharyngeal carcinoma cells by regulating iron death]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2023; 43:1665-1673. [PMID: 37933641 PMCID: PMC10630213 DOI: 10.12122/j.issn.1673-4254.2023.10.03] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Indexed: 11/08/2023]
Abstract
OBJECTIVE To explore the radiosensitizing effect of icaritin on nasopharyngeal carcinoma (NPC) cells and the underlying mechanism. METHODS MTT assay and clonal formation assay were used to evaluate the effect of icaritin on proliferation of human NPC HONE1 and HNE1 cells. The effects of icaritin treatment, γ-ray radiation, or both on production of reactive oxygen species (ROS), cell cycle distribution and apoptosis of the NPC cells were assessed using flow cytometry. The expressions of DNA damage markers γ-H2AX, cycle-related proteins CDC25C, p-CDC25C and cyclin B1, and ferroptosis markers ACSL4 and GXP4 were detected using Western blotting. A nude mouse model bearing subcutaneous HONE1 cell xenograft was used to observe the effect of icaritin and radiation on tumor growth. RESULTS Icaritin dose-dependently inhibited the viability of the NPC cells and enhanced the inhibitory effect of radiation on cell proliferation. Flow cytometry and Western blotting showed that icaritin treatment prior to radiation significantly promoted ROS production and γ-H2AX expression in the NPC cells (P<0.001). Compared with radiation exposure alone, the combined treatment caused cell cycle arrest in G2 phase, down-regulated CDC25C and cyclin B1 expression, and up-regulated p-CDC25C expression in the cells (P<0.01), resulting also in increased cell apoptosis, enhanced expression of ferroptosis protein ACSL4 and lowered expression of GXP4 (P<0.001). In the tumor-bearing mice, icaritin treatment, compared with radiation alone, significantly reduced the tumor growth rate and decreased tumor weight (P<0.001). CONCLUSION Icaritin can enhance radiosensitivity of NPC cells both in vitro and in nude mice possibly by enhancing ROS production to promote iron death of the cells.
Collapse
Affiliation(s)
- 桐 胡
- 中国医学科学院放射医学研究所天津市放射医学与分子核医学重点实验室,天津 300192Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences, Tianjin 300192, China
- 沈阳药科大学生命科学与生物制药学院,辽宁 沈阳 110016School of Life Sciences and Biopharmaceutical, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - 文峰 勾
- 中国医学科学院放射医学研究所天津市放射医学与分子核医学重点实验室,天津 300192Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences, Tianjin 300192, China
| | - 中昊 任
- 中国医学科学院放射医学研究所天津市放射医学与分子核医学重点实验室,天津 300192Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences, Tianjin 300192, China
- 沈阳药科大学生命科学与生物制药学院,辽宁 沈阳 110016School of Life Sciences and Biopharmaceutical, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - 改廷 刘
- 中国医学科学院放射医学研究所天津市放射医学与分子核医学重点实验室,天津 300192Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences, Tianjin 300192, China
| | - 祎亮 李
- 中国医学科学院放射医学研究所天津市放射医学与分子核医学重点实验室,天津 300192Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences, Tianjin 300192, China
| | - 代英 左
- 沈阳药科大学生命科学与生物制药学院,辽宁 沈阳 110016School of Life Sciences and Biopharmaceutical, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - 文彬 侯
- 中国医学科学院放射医学研究所天津市放射医学与分子核医学重点实验室,天津 300192Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences, Tianjin 300192, China
| |
Collapse
|
19
|
Bukkuri A, Pienta KJ, Austin RH, Hammarlund EU, Amend SR, Brown JS. A mathematical investigation of polyaneuploid cancer cell memory and cross-resistance in state-structured cancer populations. Sci Rep 2023; 13:15027. [PMID: 37700000 PMCID: PMC10497555 DOI: 10.1038/s41598-023-42368-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 09/09/2023] [Indexed: 09/14/2023] Open
Abstract
The polyaneuploid cancer cell (PACC) state promotes cancer lethality by contributing to survival in extreme conditions and metastasis. Recent experimental evidence suggests that post-therapy PACC-derived recurrent populations display cross-resistance to classes of therapies with independent mechanisms of action. We hypothesize that this can occur through PACC memory, whereby cancer cells that have undergone a polyaneuploid transition (PAT) reenter the PACC state more quickly or have higher levels of innate resistance. In this paper, we build on our prior mathematical models of the eco-evolutionary dynamics of cells in the 2N+ and PACC states to investigate these two hypotheses. We show that although an increase in innate resistance is more effective at promoting cross-resistance, this trend can also be produced via PACC memory. We also find that resensitization of cells that acquire increased innate resistance through the PAT have a considerable impact on eco-evolutionary dynamics and extinction probabilities. This study, though theoretical in nature, can help inspire future experimentation to tease apart hypotheses surrounding how cross-resistance in structured cancer populations arises.
Collapse
Affiliation(s)
- Anuraag Bukkuri
- Cancer Biology and Evolution Program and Department of Integrated Mathematical Oncology, Moffitt Cancer Center, Tampa, USA.
| | - Kenneth J Pienta
- The Brady Urological Institute, Johns Hopkins School of Medicine, Baltimore, USA
| | | | - Emma U Hammarlund
- Tissue Development and Evolution Research Group, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Sarah R Amend
- The Brady Urological Institute, Johns Hopkins School of Medicine, Baltimore, USA
| | - Joel S Brown
- Cancer Biology and Evolution Program and Department of Integrated Mathematical Oncology, Moffitt Cancer Center, Tampa, USA
| |
Collapse
|
20
|
Berner J, Miebach L, Kordt M, Seebauer C, Schmidt A, Lalk M, Vollmar B, Metelmann HR, Bekeschus S. Chronic oxidative stress adaptation in head and neck cancer cells generates slow-cyclers with decreased tumour growth in vivo. Br J Cancer 2023; 129:869-883. [PMID: 37460712 PMCID: PMC10449771 DOI: 10.1038/s41416-023-02343-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 06/15/2023] [Accepted: 06/21/2023] [Indexed: 08/26/2023] Open
Abstract
BACKGROUND Reactive oxygen species (ROS) are implicated in cancer therapy and as drivers of microenvironmental tumour cell adaptations. Medical gas plasma is a multi-ROS generating technology that has been shown effective for palliative tumour control in head and neck cancer (HNC) patients before tumour cells adapted to the oxidative stress and growth regressed fatally. METHODS In a bedside-to-bench approach, we sought to explore the oxidative stress adaptation in two human squamous cell carcinoma cell lines. Gas plasma was utilised as a putative therapeutic agent and chronic oxidative stress inducer. RESULTS Cellular responses of single and multiple treated cells were compared regarding sensitivity, cellular senescence, redox state and cytokine release. Whole transcriptome analysis revealed a strong correlation of cancer cell adaption with increased interleukin 1 receptor type 2 (IL1R2) expression. Using magnetic resonance imaging, tumour growth and gas plasma treatment responses of wild-type (WT) and repeatedly exposed (RE) A431 cells were further investigated in a xenograft model in vivo. RE cells generated significantly smaller tumours with suppressed inflammatory secretion profiles and increased epidermal growth factor receptor (EGFR) activity showing significantly lower gas plasma sensitivity until day 8. CONCLUSIONS Clinically, combination treatments together with cetuximab, an EGFR inhibitor, may overcome acquired oxidative stress resistance in HNC.
Collapse
Grants
- This study was funded by the joint research project ONKOTHER-H is supported by the European Social Fund (ESF, grant numbers ESF/14-BM-A55-0003/18, ESF/14-BM-A55-0005/18, and ESF/14-BM-A55-0006/18) and the Ministry of Education, Science, and Culture of Mecklenburg-Vorpommern, Germany, as well as the German Federal Ministry of Education and Research (BMBF, grant numbers 03Z22DN11 and 03Z22Di1).
- This study was funded by the joint research project ONKOTHER-H is supported by the European Social Fund (ESF, grant numbers ESF/14-BM-A55-0005/18).
- Gerhard-Domagk-Foundation Greifswald (Germany).
- This study was funded by the joint research project ONKOTHER-H is supported by the European Social Fund (ESF, grant numbers ESF/14-BM-A55-0003/18).
Collapse
Affiliation(s)
- Julia Berner
- Department of Oral, Maxillofacial, and Plastic Surgery, Greifswald University Medical Center, Ferdinand-Sauerbruch-Str, 17475, Greifswald, Germany
- ZIK plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Felix-Hausdorff-Str. 2, 17489, Greifswald, Germany
| | - Lea Miebach
- ZIK plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Felix-Hausdorff-Str. 2, 17489, Greifswald, Germany
- Department of General, Visceral, Thoracic, and Vascular Surgery, Greifswald University Medical Center, Ferdinand-Sauerbruch-Str, 17475, Greifswald, Germany
| | - Marcel Kordt
- Rudolf-Zenker-Institute of Experimental Surgery, Rostock University Medical Center, Schillingallee 69a, 18057, Rostock, Germany
| | - Christian Seebauer
- Department of Oral, Maxillofacial, and Plastic Surgery, Greifswald University Medical Center, Ferdinand-Sauerbruch-Str, 17475, Greifswald, Germany
| | - Anke Schmidt
- ZIK plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Felix-Hausdorff-Str. 2, 17489, Greifswald, Germany
| | - Michael Lalk
- Institute for Biochemistry, University of Greifswald, Felix-Hausdorff-Str. 4, 17489, Greifswald, Germany
| | - Brigitte Vollmar
- Rudolf-Zenker-Institute of Experimental Surgery, Rostock University Medical Center, Schillingallee 69a, 18057, Rostock, Germany
| | - Hans-Robert Metelmann
- Department of Oral, Maxillofacial, and Plastic Surgery, Greifswald University Medical Center, Ferdinand-Sauerbruch-Str, 17475, Greifswald, Germany
| | - Sander Bekeschus
- ZIK plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Felix-Hausdorff-Str. 2, 17489, Greifswald, Germany.
- Clinic and Policlinic for Dermatology and Venerology, Rostock University Medical Center, Strempelstr. 13, 18057, Rostock, Germany.
| |
Collapse
|
21
|
Barhoumi T, Mansour FA, Jalouli M, Alamri HS, Ali R, Harrath AH, Aljumaa M, Boudjelal M. Angiotensin II modulates THP-1-like macrophage phenotype and inflammatory signatures via angiotensin II type 1 receptor. Front Cardiovasc Med 2023; 10:1129704. [PMID: 37692050 PMCID: PMC10485254 DOI: 10.3389/fcvm.2023.1129704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 06/30/2023] [Indexed: 09/12/2023] Open
Abstract
Angiotensin II (Ang II) is a major component of the renin-angiotensin or renin-angiotensin-aldosterone system, which is the main element found to be involved in cardiopathology. Recently, long-term metabolomics studies have linked high levels of angiotensin plasma to inflammatory conditions such as coronary heart disease, obesity, and type 2 diabetes. Monocyte/macrophage cellular function and phenotype orchestrate the inflammatory response in various pathological conditions, most notably cardiometabolic disease. An activation of the Ang II system is usually associated with inflammation and cardiovascular disease; however, the direct effect on monocyte/macrophages has still not been well elucidated. Herein, we have evaluated the cellular effects of Ang II on THP-1-derived macrophages. Ang II stimulated the expression of markers involved in monocyte/macrophage cell differentiation (e.g., CD116), as well as adhesion, cell-cell interaction, chemotaxis, and phagocytosis (CD15, CD44, CD33, and CD49F). Yet, Ang II increased the expression of proinflammatory markers (HLA-DR, TNF-α, CD64, CD11c, and CD38) and decreased CD206 (mannose receptor), an M2 marker. Moreover, Ang II induced cytosolic calcium overload, increased reactive oxygen species, and arrested cells in the G1 phase. Most of these effects were induced via the angiotensin II type 1 receptor (AT1R). Collectively, our results provide new evidence in support of the effect of Ang II in inflammation associated with cardiometabolic diseases.
Collapse
Affiliation(s)
- Tlili Barhoumi
- Medical Research Core Facility and Platforms (MRCFP), King Abdullah International Medical Research Center/King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), King Abdulaziz Medical City (KAMC), NGHA, Riyadh, Saudi Arabia
| | - Fatmah A. Mansour
- Medical Research Core Facility and Platforms (MRCFP), King Abdullah International Medical Research Center/King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), King Abdulaziz Medical City (KAMC), NGHA, Riyadh, Saudi Arabia
| | - Maroua Jalouli
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Hassan S. Alamri
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud bin Abdulaziz University for Health Sciences/King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Rizwan Ali
- Medical Research Core Facility and Platforms (MRCFP), King Abdullah International Medical Research Center/King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), King Abdulaziz Medical City (KAMC), NGHA, Riyadh, Saudi Arabia
| | - Abdel Halim Harrath
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Maha Aljumaa
- Department of Biology, College of Science, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Mohamed Boudjelal
- Medical Research Core Facility and Platforms (MRCFP), King Abdullah International Medical Research Center/King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), King Abdulaziz Medical City (KAMC), NGHA, Riyadh, Saudi Arabia
| |
Collapse
|
22
|
Novitasari D, Jenie RI, Kato JY, Meiyanto E. Chemoprevention curcumin analog 1.1 promotes metaphase arrest and enhances intracellular reactive oxygen species levels on TNBC MDA-MB-231 and HER2-positive HCC1954 cells. Res Pharm Sci 2023; 18:358-370. [PMID: 37614620 PMCID: PMC10443663 DOI: 10.4103/1735-5362.378083] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 11/12/2022] [Accepted: 05/23/2023] [Indexed: 08/25/2023] Open
Abstract
Background and purpose Previous studies highlighted that chemoprevention curcumin analog-1.1 (CCA-1.1) demonstrated an antitumor effect on breast, leukemia, and colorectal cancer cells. By utilizing immortalized MDA-MB-231 and HCC1954 cells, we evaluated the anticancer properties of CCA-1.1 and its mediated activity to promote cellular death. Experimental approach Cytotoxicity and anti-proliferation were assayed using trypan blue exclusion. The cell cycle profile after CCA-1.1 treatment was established through flow cytometry. May-Grünwald-Giemsa and Hoechst staining were performed to determine the cell cycle arrest upon CCA-1.1 treatment. The involvement of CCA-1.1 in mitotic kinases (aurora A, p-aurora A, p-PLK1, and p-cyclin B1) expression was investigated by immunoblotting. CCA-1.1-treated cells were stained with the X-gal solution to examine the effect on senescence. ROS level and mitochondrial respiration were assessed by DCFDA assay and mitochondrial oxygen consumption rate, respectively. Findings/Results CCA-1.1 exerted cytotoxic activity and inhibited cell proliferation with an irreversible effect, and the flow cytometry analysis demonstrated that CCA-1.1 significantly halted during the G2/M phase, and further assessment revealed that CCA-1.1 caused metaphase arrest. Immunoblot assays confirmed CCA-1.1 suppressed aurora A kinase in MDA-MB-231 cells. The ROS level was elevated after treatment with CCA-1.1, which might promote cellular senescence and suppress basal mitochondrial respiration in MDA-MB-231 cells. Conclusion and implications Our data suggested the in vitro proof-of-concept that supports the involvement in cell cycle regulation and ROS generation as contributors to the effectiveness of CCA-1.1 in suppressing breast cancer cell growth.
Collapse
Affiliation(s)
- Dhania Novitasari
- Cancer Chemoprevention Research Center, Faculty of Pharmacy, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Riris Istighfari Jenie
- Cancer Chemoprevention Research Center, Faculty of Pharmacy, Universitas Gadjah Mada, Yogyakarta, Indonesia
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Jun-ya Kato
- Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, Nara, Japan
| | - Edy Meiyanto
- Cancer Chemoprevention Research Center, Faculty of Pharmacy, Universitas Gadjah Mada, Yogyakarta, Indonesia
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Universitas Gadjah Mada, Yogyakarta, Indonesia
| |
Collapse
|
23
|
Mallin MM, Kim N, Choudhury MI, Lee SJ, An SS, Sun SX, Konstantopoulos K, Pienta KJ, Amend SR. Cells in the polyaneuploid cancer cell (PACC) state have increased metastatic potential. Clin Exp Metastasis 2023:10.1007/s10585-023-10216-8. [PMID: 37326720 DOI: 10.1007/s10585-023-10216-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 06/02/2023] [Indexed: 06/17/2023]
Abstract
Although metastasis is the leading cause of cancer deaths, it is quite rare at the cellular level. Only a rare subset of cancer cells (~ 1 in 1.5 billion) can complete the entire metastatic cascade: invasion, intravasation, survival in the circulation, extravasation, and colonization (i.e. are metastasis competent). We propose that cells engaging a Polyaneuploid Cancer Cell (PACC) phenotype are metastasis competent. Cells in the PACC state are enlarged, endocycling (i.e. non-dividing) cells with increased genomic content that form in response to stress. Single-cell tracking using time lapse microscopy reveals that PACC state cells have increased motility. Additionally, cells in the PACC state exhibit increased capacity for environment-sensing and directional migration in chemotactic environments, predicting successful invasion. Magnetic Twisting Cytometry and Atomic Force Microscopy reveal that cells in the PACC state display hyper-elastic properties like increased peripheral deformability and maintained peri-nuclear cortical integrity that predict successful intravasation and extravasation. Furthermore, four orthogonal methods reveal that cells in the PACC state have increased expression of vimentin, a hyper-elastic biomolecule known to modulate biomechanical properties and induce mesenchymal-like motility. Taken together, these data indicate that cells in the PACC state have increased metastatic potential and are worthy of further in vivo analysis.
Collapse
Affiliation(s)
- Mikaela M Mallin
- Cellular and Molecular Medicine Graduate Training Program, Johns Hopkins School of Medicine, Baltimore, MD, USA.
- Cancer Ecology Center, James Buchanan Brady Urological Institute, Johns Hopkins Medical Institute, Baltimore, MD, USA.
| | - Nicholas Kim
- Rutgers Institute for Translational Medicine and Science, New Brunswick, NJ, USA
| | | | - Se Jong Lee
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Steven S An
- Rutgers Institute for Translational Medicine and Science, New Brunswick, NJ, USA
| | - Sean X Sun
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | | | - Kenneth J Pienta
- Cellular and Molecular Medicine Graduate Training Program, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Cancer Ecology Center, James Buchanan Brady Urological Institute, Johns Hopkins Medical Institute, Baltimore, MD, USA
| | - Sarah R Amend
- Cellular and Molecular Medicine Graduate Training Program, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Cancer Ecology Center, James Buchanan Brady Urological Institute, Johns Hopkins Medical Institute, Baltimore, MD, USA
| |
Collapse
|
24
|
Agena R, Cortés-Sánchez ADJ, Hernández-Sánchez H, Álvarez-Salas LM, Martínez-Rodríguez OP, García VHR, Jaramillo Flores ME. Pro-Apoptotic Activity and Cell Cycle Arrest of Caulerpa sertularioides against SKLU-1 Cancer Cell in 2D and 3D Cultures. Molecules 2023; 28:molecules28114361. [PMID: 37298837 DOI: 10.3390/molecules28114361] [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: 04/27/2023] [Revised: 05/18/2023] [Accepted: 05/24/2023] [Indexed: 06/12/2023] Open
Abstract
Cancer is a disease with the highest mortality and morbidity rate worldwide. First-line drugs induce several side effects that drastically reduce the quality of life of people with this disease. Finding molecules to prevent it or generate less aggressiveness or no side effects is significant to counteract this problem. Therefore, this work searched for bioactive compounds of marine macroalgae as an alternative treatment. An 80% ethanol extract of dried Caulerpa sertularioides (CSE) was analyzed by HPLS-MS to identify the chemical components. CSE was utilized through a comparative 2D versus 3D culture model. Cisplatin (Cis) was used as a standard drug. The effects on cell viability, apoptosis, cell cycle, and tumor invasion were evaluated. The IC50 of CSE for the 2D model was 80.28 μg/mL versus 530 μg/mL for the 3D model after 24 h of treatment exposure. These results confirmed that the 3D model is more resistant to treatments and complex than the 2D model. CSE generated a loss of mitochondrial membrane potential, induced apoptosis by extrinsic and intrinsic pathways, upregulated caspases-3 and -7, and significantly decreased tumor invasion of a 3D SKLU-1 lung adenocarcinoma cell line. CSE generates biochemical and morphological changes in the plasma membrane and causes cell cycle arrest at the S and G2/M phases. These findings conclude that C. sertularioides is a potential candidate for alternative treatment against lung cancer. This work reinforced the use of complex models for drug screening and suggested using CSE's primary component, caulerpin, to determine its effect and mechanism of action on SKLU-1 in the future. A multi-approach with molecular and histological analysis and combination with first-line drugs must be included.
Collapse
Affiliation(s)
- Rosette Agena
- Ingeniería Bioquímica-Escuela Nacional de Ciencias Biológicas (ENCB)-Instituto Politécnico Nacional, Ciudad de México 07738, Mexico
| | | | - Humberto Hernández-Sánchez
- Ingeniería Bioquímica-Escuela Nacional de Ciencias Biológicas (ENCB)-Instituto Politécnico Nacional, Ciudad de México 07738, Mexico
| | - Luis Marat Álvarez-Salas
- Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV), Ciudad de México 07360, Mexico
| | - Oswaldo Pablo Martínez-Rodríguez
- Ingeniería Bioquímica-Escuela Nacional de Ciencias Biológicas (ENCB)-Instituto Politécnico Nacional, Ciudad de México 07738, Mexico
| | - Víctor Hugo Rosales García
- Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV), Ciudad de México 07360, Mexico
| | - María Eugenia Jaramillo Flores
- Ingeniería Bioquímica-Escuela Nacional de Ciencias Biológicas (ENCB)-Instituto Politécnico Nacional, Ciudad de México 07738, Mexico
| |
Collapse
|
25
|
El-Kenawi A, Berglund A, Estrella V, Zhang Y, Liu M, Putney RM, Yoder SJ, Johnson J, Brown J, Gatenby R. Elevated Methionine Flux Drives Pyroptosis Evasion in Persister Cancer Cells. Cancer Res 2023; 83:720-734. [PMID: 36480167 PMCID: PMC9978888 DOI: 10.1158/0008-5472.can-22-1002] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 09/29/2022] [Accepted: 12/02/2022] [Indexed: 12/13/2022]
Abstract
Induction of cell death represents a primary goal of most anticancer treatments. Despite the efficacy of such approaches, a small population of "persisters" develop evasion strategies to therapy-induced cell death. While previous studies have identified mechanisms of resistance to apoptosis, the mechanisms by which persisters dampen other forms of cell death, such as pyroptosis, remain to be elucidated. Pyroptosis is a form of inflammatory cell death that involves formation of membrane pores, ion gradient imbalance, water inflow, and membrane rupture. Herein, we investigate mechanisms by which cancer persisters resist pyroptosis, survive, then proliferate in the presence of tyrosine kinase inhibitors (TKI). Lung, prostate, and esophageal cancer persister cells remaining after treatments exhibited several hallmarks indicative of pyroptosis resistance. The inflammatory attributes of persisters included chronic activation of inflammasome, STING, and type I interferons. Comprehensive metabolomic characterization uncovered that TKI-induced pyroptotic persisters display high methionine consumption and excessive taurine production. Elevated methionine flux or exogenous taurine preserved plasma membrane integrity via osmolyte-mediated effects. Increased dependency on methionine flux decreased the level of one carbon metabolism intermediate S-(5'-adenosyl)-L-homocysteine, a determinant of cell methylation capacity. The consequent increase in methylation potential induced DNA hypermethylation of genes regulating metal ion balance and intrinsic immune response. This enabled thwarting TKI resistance by using the hypomethylating agent decitabine. In summary, the evolution of resistance to pyroptosis can occur via a stepwise process of physical acclimation and epigenetic changes without existing or recurrent mutations. SIGNIFICANCE Methionine enables cancer cells to persist by evading pyroptotic osmotic lysis, which leads to genome-wide hypermethylation that allows persisters to gain proliferative advantages.
Collapse
Affiliation(s)
- Asmaa El-Kenawi
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center, Tampa, Florida.,Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center, Tampa, Florida
| | - Anders Berglund
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center, Tampa, Florida
| | - Veronica Estrella
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center, Tampa, Florida.,Department of Cancer Physiology, H. Lee Moffitt Cancer Center, Tampa, Florida
| | - Yonghong Zhang
- Biostatistics and Bioinformatics Shared Resource, H. Lee Moffitt Cancer Center, Tampa, Florida
| | - Min Liu
- Proteomics and Metabolomics Core Facility, H. Lee Moffitt Cancer Center, Tampa, Florida
| | - Ryan M Putney
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center, Tampa, Florida
| | - Sean J Yoder
- Molecular Genomics Core Facility, H. Lee Moffitt Cancer Center, Tampa, Florida
| | - Joseph Johnson
- Analytic Microscopy Core Facility, H. Lee Moffitt Cancer Center, Tampa, Florida
| | - Joel Brown
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center, Tampa, Florida.,Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center, Tampa, Florida
| | - Robert Gatenby
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center, Tampa, Florida.,Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center, Tampa, Florida.,Department of Radiology, H. Lee Moffitt Cancer Center, Tampa, Florida
| |
Collapse
|
26
|
Ling JY, Wang QL, Liang HN, Liu QB, Yin DH, Lin L. Flavonoid-Rich Extract of Oldenlandia diffusa (Willd.) Roxb. Inhibits Gastric Cancer by Activation of Caspase-Dependent Mitochondrial Apoptosis. Chin J Integr Med 2023; 29:213-223. [PMID: 36044114 DOI: 10.1007/s11655-022-3679-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/25/2022] [Indexed: 11/03/2022]
Abstract
OBJECTIVE To evaluate the apoptosis and cycle arrest effects of Oldenlandia diffusa flavonoids on human gastric cancer cells, determine the action mechanisms in association with the mitochondrial dependent signal transduction pathway that controls production of reactive oxygen species (ROS), and evaluate the pharmacodynamics of a mouse xenotransplantation model to provide a reference for the use of flavonoids in prevention and treatment of gastric cancer. METHODS Flavonoids were extracted by an enzymatic-ultrasonic assisted method and purified with D-101 resin. Bioactive components were characterized by high-performance liquid chromatography. Cell lines MKN-45, AGS, and GES-1 were treated with different concentrations of flavonoids (64, 96, 128, 160 µg/mL). The effect of flavonoids on cell viability was evaluated by MTT method, and cell nuclear morphology was observed by Hoechst staining. The apoptosis rate and cell cycle phases were measured by flow cytometry, the production of ROS was detected by laser confocal microscope, the mitochondrial membrane potential (MMP) were observed by fluorescence microscope, and the expression of apoptotic proteins related to activation of mitochondrial pathway were measured by immunoblotting. MKN-45 cells were transplanted into BALB/c nude mice to establish a xenograft tumor model. Hematoxylin and eosin staining was used to reveal the subcutaneous tumor tissue. The tumor volume and tumor weight were measured, the expression levels of proliferation markers proliferating cell nuclear antigen (PCNA) and Ki-67 were detected by immunohistochemistry, and the expression levels of CA72-4 were measured by enzyme linked immunosorbent assay. RESULTS Oldenlandia diffusa flavonoids inhibited proliferation of MKN-45 and AGS human gastric cancer cells, arrested the cell cycle in G1/S phase, induced accumulation of ROS in the process of apoptosis, and altered MMP. In addition, flavonoids increased Apaf-1, Cleaved-Caspase-3, and Bax, and decreased Cyclin A, Cdk2, Bcl-2, Pro-Caspase-9, and Mitochondrial Cytochrome C (P<0.05). The MKN-45 cell mouse xenotransplantation model further clarified the growth inhibitory effect of flavonoids towards tumors. The expression levels of PCNA and Ki-67 decreased in each flavonoid dose group, the expression level of CA72-4 decreased (P<0.05). CONCLUSION Flavonoids derived from Oldenlandia diffusa can inhibit proliferation and induce apoptosis of human gastric cancer cells by activating the mitochondrial controlled signal transduction pathway.
Collapse
Affiliation(s)
- Jia-Yin Ling
- First School of Clinical Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, China.,Gansu Provincial Hospital, Lanzhou, 730000, China
| | - Qiu-Lan Wang
- First School of Clinical Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, China. .,Gansu Provincial Hospital, Lanzhou, 730000, China.
| | - Hao-Nan Liang
- First School of Clinical Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, China.,Gansu Provincial Hospital, Lanzhou, 730000, China
| | - Qing-Bo Liu
- First School of Clinical Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, China.,Gansu Provincial Hospital, Lanzhou, 730000, China
| | - Dong-Hong Yin
- First School of Clinical Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, China.,Gansu Provincial Hospital, Lanzhou, 730000, China
| | - Li Lin
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou, 730000, China.,Institute of Chinese (Tibetan) Medicine Resources, Gansu University of Chinese Medicine, Lanzhou, 730000, China
| |
Collapse
|
27
|
Roy AC, Prasad A, Priya K, Das P, Singh S, Ghosh C, Ghosh I. Anticancer effect of antioxidant-rich methanolic extract of Rauvolfia serpentina (L.) Benth. ex Kurz leaves in HepG2 and HeLa cells: A mechanistic insight. BIOCATALYSIS AND AGRICULTURAL BIOTECHNOLOGY 2023. [DOI: 10.1016/j.bcab.2023.102674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2023]
|
28
|
Kciuk M, Gielecińska A, Mujwar S, Kołat D, Kałuzińska-Kołat Ż, Celik I, Kontek R. Doxorubicin-An Agent with Multiple Mechanisms of Anticancer Activity. Cells 2023; 12:659. [PMID: 36831326 PMCID: PMC9954613 DOI: 10.3390/cells12040659] [Citation(s) in RCA: 222] [Impact Index Per Article: 111.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 02/15/2023] [Accepted: 02/17/2023] [Indexed: 02/22/2023] Open
Abstract
Doxorubicin (DOX) constitutes the major constituent of anti-cancer treatment regimens currently in clinical use. However, the precise mechanisms of DOX's action are not fully understood. Emerging evidence points to the pleiotropic anticancer activity of DOX, including its contribution to DNA damage, reactive oxygen species (ROS) production, apoptosis, senescence, autophagy, ferroptosis, and pyroptosis induction, as well as its immunomodulatory role. This review aims to collect information on the anticancer mechanisms of DOX as well as its influence on anti-tumor immune response, providing a rationale behind the importance of DOX in modern cancer therapy.
Collapse
Affiliation(s)
- Mateusz Kciuk
- Department of Molecular Biotechnology and Genetics, University of Lodz, 90-237 Lodz, Poland
- Doctoral School of Exact and Natural Sciences, University of Lodz, 90-237 Lodz, Poland
| | - Adrianna Gielecińska
- Department of Molecular Biotechnology and Genetics, University of Lodz, 90-237 Lodz, Poland
- Doctoral School of Exact and Natural Sciences, University of Lodz, 90-237 Lodz, Poland
| | - Somdutt Mujwar
- Chitkara College of Pharmacy, Chitkara University, Rajpura 140401, Punjab, India
| | - Damian Kołat
- Department of Experimental Surgery, Faculty of Medicine, Medical University of Lodz, 90-136 Lodz, Poland
| | - Żaneta Kałuzińska-Kołat
- Department of Experimental Surgery, Faculty of Medicine, Medical University of Lodz, 90-136 Lodz, Poland
| | - Ismail Celik
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Erciyes University, 38039 Kayseri, Turkey
| | - Renata Kontek
- Department of Molecular Biotechnology and Genetics, University of Lodz, 90-237 Lodz, Poland
| |
Collapse
|
29
|
Wu Z, Stangl S, Hernandez-Schnelzer A, Wang F, Hasanzadeh Kafshgari M, Bashiri Dezfouli A, Multhoff G. Functionalized Hybrid Iron Oxide-Gold Nanoparticles Targeting Membrane Hsp70 Radiosensitize Triple-Negative Breast Cancer Cells by ROS-Mediated Apoptosis. Cancers (Basel) 2023; 15:cancers15041167. [PMID: 36831510 PMCID: PMC9954378 DOI: 10.3390/cancers15041167] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 02/07/2023] [Accepted: 02/09/2023] [Indexed: 02/15/2023] Open
Abstract
Triple-negative breast cancer (TNBC) a highly aggressive tumor entity with an unfavorable prognosis, is treated by multimodal therapies, including ionizing radiation (IR). Radiation-resistant tumor cells, as well as induced normal tissue toxicity, contribute to the poor clinical outcome of the disease. In this study, we investigated the potential of novel hybrid iron oxide (Fe3O4)-gold (Au) nanoparticles (FeAuNPs) functionalized with the heat shock protein 70 (Hsp70) tumor-penetrating peptide (TPP) and coupled via a PEG4 linker (TPP-PEG4-FeAuNPs) to improve tumor targeting and uptake of NPs and to break radioresistance in TNBC cell lines 4T1 and MDA-MB-231. Hsp70 is overexpressed in the cytosol and abundantly presented on the cell membrane (mHsp70) of highly aggressive tumor cells, including TNBCs, but not on corresponding normal cells, thus providing a tumor-specific target. The Fe3O4 core of the NPs can serve as a contrast agent enabling magnetic resonance imaging (MRI) of the tumor, and the nanogold shell radiosensitizes tumor cells by the release of secondary electrons (Auger electrons) upon X-ray irradiation. We demonstrated that the accumulation of TPP-PEG4-FeAuNPs into mHsp70-positive TNBC cells was superior to that of non-conjugated FeAuNPs and FeAuNPs functionalized with a non-specific, scrambled peptide (NGL). After a 24 h co-incubation period of 4T1 and MDA-MB-231 cells with TPP-PEG4-FeAuNPs, but not with control hybrid NPs, ionizing irradiation (IR) causes a cell cycle arrest at G2/M and induces DNA double-strand breaks, thus triggering apoptotic cell death. Since the radiosensitizing effect was completely abolished in the presence of the ROS inhibitor N-acetyl-L-cysteine (NAC), we assume that the TPP-PEG4-FeAuNP-induced apoptosis is mediated via an increased production of ROS.
Collapse
Affiliation(s)
- Zhiyuan Wu
- Central Institute for Translational Cancer Research (TranslaTUM), Radiation Immuno Oncology Group, Klinikum Rechts der Isar der Technischen Universität München, 81675 Munich, Germany
| | - Stefan Stangl
- Central Institute for Translational Cancer Research (TranslaTUM), Radiation Immuno Oncology Group, Klinikum Rechts der Isar der Technischen Universität München, 81675 Munich, Germany
- Department of Nuclear Medicine, Klinikum Rechts der Isar, Technische Universität München, 81675 Munich, Germany
| | - Alicia Hernandez-Schnelzer
- Central Institute for Translational Cancer Research (TranslaTUM), Radiation Immuno Oncology Group, Klinikum Rechts der Isar der Technischen Universität München, 81675 Munich, Germany
| | - Fei Wang
- Central Institute for Translational Cancer Research (TranslaTUM), Radiation Immuno Oncology Group, Klinikum Rechts der Isar der Technischen Universität München, 81675 Munich, Germany
| | - Morteza Hasanzadeh Kafshgari
- Heinz-Nixdorf-Chair of Biomedical Electronics, TranslaTUM, Klinikum Rechts der Isar, Technische Universität München, 81675 Munich, Germany
| | - Ali Bashiri Dezfouli
- Central Institute for Translational Cancer Research (TranslaTUM), Radiation Immuno Oncology Group, Klinikum Rechts der Isar der Technischen Universität München, 81675 Munich, Germany
| | - Gabriele Multhoff
- Central Institute for Translational Cancer Research (TranslaTUM), Radiation Immuno Oncology Group, Klinikum Rechts der Isar der Technischen Universität München, 81675 Munich, Germany
- Department of Radiation Oncology, Klinikum Rechts der Isar, Technischen Universität München, 81675 Munich, Germany
- Correspondence: ; Tel.: +49-89-4140-4514; Fax: +49-89-4140-4299
| |
Collapse
|
30
|
Dukel M, Fiskin K. Combination of PAKs inhibitors IPA-3 and PF-3758309 effectively suppresses colon carcinoma cell growth by perturbing DNA damage response. Int J Radiat Biol 2023; 99:340-354. [PMID: 35939342 DOI: 10.1080/09553002.2022.2110326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
PURPOSE PAKs proteins are speculated as new promising targets for cancer therapy due to their central role in many oncogenic pathways. Because PAKs proteins are very significant during carcinogenesis, we aimed to investigate the hypothesis that inhibition of PAKs with IPA-3 and PF-3758309 treatment could synergistically reduce colon carcinoma cell growth. MATERIALS AND METHODS The cytotoxic effects of both drugs were determined by a cell viability assay. Cell cycle and apoptosis were analyzed by flow cytometry. The effects of inhibitor drugs on marker genes of apoptosis, autophagy, cell cycle, and DNA damage were tested via immunoblotting. RESULTS AND CONCLUSIONS We found out the synergistic effect of these drugs in pair on five colon cancer cell lines. Combined treatment with IPA-3+PF-3758309 in SW620 and Colo 205 cells markedly suppressed colon formation and induced apoptosis, cell cycle arrest, and autophagy compared with treatment with each drug alone. Additionally, this combination sensitized colon cancer cells to ionizing radiation that resulted in inhibition of cell growth. SIGNIFICANCE Collectively, our findings show for the first time that cotreatment of IPA-3 with PF-3758309 exhibits superior inhibitory effects on colon carcinoma cell growth via inducing DNA damage-related cell death and also enforces a cell cycle arrest.
Collapse
Affiliation(s)
- Muzaffer Dukel
- Molecular Biology and Genetics Department, Faculty of Art and Science, Mehmet Akif Ersoy University, Burdur, Turkey
| | - Kayahan Fiskin
- Biology Department, Faculty of Science, Akdeniz University, Antalya, Turkey
| |
Collapse
|
31
|
Phytochemical Analysis and Anticancer Properties of Drimia maritima Bulb Extracts on Colorectal Cancer Cells. Molecules 2023; 28:molecules28031215. [PMID: 36770882 PMCID: PMC9921424 DOI: 10.3390/molecules28031215] [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: 10/25/2022] [Revised: 01/02/2023] [Accepted: 01/04/2023] [Indexed: 01/28/2023] Open
Abstract
Cancer is a worldwide health problem and is the second leading cause of death after heart disease. Due to the high cost and severe side effects associated with chemotherapy treatments, natural products with anticancer therapeutic potential may play a promising role in anticancer therapy. The purpose of this study was to investigate the cytotoxic and apoptotic characteristics of the aqueous Drimia maritima bulb extract on Caco-2 and COLO-205 colorectal cancer cells. In order to reach such a purpose, the chemical composition was examined using the GC-MS method, and the selective antiproliferative effect was determined in colon cancer cell lines in normal gingival fibroblasts. The intracellular ROS, mitochondrial membrane potential, and gene expression changes in selected genes (CASP8, TNF-α, and IL-6 genes) were assessed to determine the molecular mechanism of the antitumor effect of the extract. GC-MS results revealed the presence of fifty-seven compounds, and Proscillaridin A was the predominant secondary metabolite in the extract. The IC50 of D. maritima bulb extract on Caco-2, COLO-205, and the normal human gingival fibroblasts were obtained at 0.9 µg/mL, 2.3 µg/mL, and 13.1 µg/mL, respectively. The apoptotic effect assay indicated that the bulb extract induced apoptosis in both colon cancer cell lines. D. maritima bulb extract was only able to induce statistically significant ROS levels in COLO-205 cells in a dose-dependent manner. The mitochondrial membrane potential (MMP) revealed a significant decrease in the MMP of Caco-2 and COLO-205 to various concentrations of the bulb extract. At the molecular level, RT-qPCR was used to assess gene expression of CASP8, TNF-α, and IL-6 genes in Caco-2 and COLO-205 cancer cells. The results showed that the expression of pro-inflammatory genes TNF-α and IL-6 were upregulated. The apoptotic initiator gene CASP8 was also upregulated in the Caco-2 cell line and did not reach significance in COLO-205 cells. These results lead to the conclusion that D. maritima extract induced cell death in both cell lines and may have the potential to be used in CRC therapy in the future.
Collapse
|
32
|
The Assessment of the Phototoxic Action of Chlortetracycline and Doxycycline as a Potential Treatment of Melanotic Melanoma-Biochemical and Molecular Studies on COLO 829 and G-361 Cell Lines. Int J Mol Sci 2023; 24:ijms24032353. [PMID: 36768675 PMCID: PMC9917077 DOI: 10.3390/ijms24032353] [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: 12/14/2022] [Revised: 01/22/2023] [Accepted: 01/22/2023] [Indexed: 01/27/2023] Open
Abstract
Melanoma is still one of the most dangerous cancers. New methods of treatment are sought due to its high aggressiveness and the relatively low effectiveness of therapies. Tetracyclines are drugs exhibiting anticancer activity. Previous studies have also shown their activity against melanoma cells. The possibility of tetracycline accumulation in pigmented tissues and the increase in their toxicity under the influence of UVA radiation creates the possibility of developing a new anti-melanoma therapy. This study aimed to analyze the phototoxic effect of doxycycline and chlortetracycline on melanotic melanoma cells COLO 829 and G-361. The results indicated that tetracycline-induced phototoxicity significantly decreased the number of live cells by cell cycle arrest as well as a decrease in cell viability. The simultaneous exposure of cells to drugs and UVA caused the depolarization of mitochondria as well as inducing oxidative stress and apoptosis. It was found that the combined treatment activated initiator and effector caspases, caused DNA fragmentation and elevated p53 level. Finally, it was concluded that doxycycline demonstrated a stronger cytotoxic and phototoxic effect. UVA irradiation of melanoma cells treated with doxycycline and chlortetracycline allows for the reduction of therapeutic drug concentrations and increases the effectiveness of tested tetracyclines.
Collapse
|
33
|
Lin L, Wu X, Jiang Y, Luo X, Cao X. Raddeanin A Improves the Therapeutic Effect of Osimertinib in NSCLC by Accelerating ROS/NLRP3-mediated Pyroptosis. Curr Pharm Des 2023; 29:2591-2600. [PMID: 37861040 DOI: 10.2174/0113816128263069231010111347] [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: 05/19/2023] [Revised: 08/29/2023] [Accepted: 09/14/2023] [Indexed: 10/21/2023]
Abstract
BACKGROUND Osimertinib (Osm) is the preferred treatment for non-small cell lung cancer (NSCLC) patients with the epidermal growth factor receptor (EGFR) T790M mutation. Nevertheless, the resistance of NSCLC cells to Osm will eventually develop, which remains the biggest obstacle to treating such diseases. Raddeanin A (RA) exhibits a potent anti-tumor effect on various types of cancer cells. In this study, we aimed to investigate whether RA suppresses NSCLC growth and increases the therapeutic effect of Osm. METHODS The effects of RA on inhibiting NSCLC cell viability and proliferation were tested using cell counting kit 8 (CCK-8) and EdU assay. The roles of RA in improving the anti-tumor effect of Osm were tested with CCK-8 and colony formation assays. The roles of RA in regulating reactive oxygen species (ROS)/NOD-, LRR-, and pyrin domain-containing protein 3 (NLRP3)-mediated pyroptosis were assessed using quantitative real- time PCR (qRT-PCR) and western blotting analysis. RESULTS RA treatment decreased A549 and H1975 cell viability in a dose- and time-dependent way. RA inhibited NSCLC cell proliferation and tumor growth in vivo. Mechanistically, RA induced ROS overgeneration and resulted in subsequent NLRP3-mediated pyroptosis. In particular, combination treatment with Osm and RA reduced cell viability and clonogenic growth capacity more efficiently than Osm mono treatment in A549 and H1975 cells. Combination treatment also promoted NLRP3-mediated pyroptosis more efficiently than Osm mono treatment. CONCLUSION RA inhibited the NSCLC growth and increased the anti-tumor role of Osm in NSCLC by facilitating ROS/NLRP3-mediated pyroptosis. These results suggested that combination therapy with RA and Osm might be an effective strategy to treat Osm-resistant NSCLC.
Collapse
Affiliation(s)
- Liping Lin
- Department of Oncology, Panyu Central Hospital, Guangzhou, China
- Cancer Institute of Panyu, Guangzhou, China
| | - Xuan Wu
- Department of Oncology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Yuanxue Jiang
- Department of Oncology, Panyu Central Hospital, Guangzhou, China
- Cancer Institute of Panyu, Guangzhou, China
| | - Xi Luo
- Department of Oncology, Panyu Central Hospital, Guangzhou, China
- Cancer Institute of Panyu, Guangzhou, China
| | - Xiaolong Cao
- Department of Oncology, Panyu Central Hospital, Guangzhou, China
- Cancer Institute of Panyu, Guangzhou, China
| |
Collapse
|
34
|
Varzandeh M, Labbaf S, Varshosaz J, Laurent S. An overview of the intracellular localization of high-Z nanoradiosensitizers. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2022; 175:14-30. [PMID: 36029849 DOI: 10.1016/j.pbiomolbio.2022.08.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 07/17/2022] [Accepted: 08/19/2022] [Indexed: 06/15/2023]
Abstract
Radiation therapy (RT) is a method commonly used for cancer treatment worldwide. Commonly, RT utilizes two routes for combating cancers: 1) high-energy radiation to generate toxic reactive oxygen species (ROS) (through the dissociation of water molecules) for damaging the deoxyribonucleic acid (DNA) inside the nucleus 2) direct degradation of the DNA. However, cancer cells have mechanisms to survive under intense RT, which can considerably decrease its therapeutic efficacy. Excessive radiation energy damages healthy tissues, and hence, low doses are applied for cancer treatment. Additionally, different radiosensitizers were used to sensitize cancer cells towards RT through individual mechanisms. Following this route, nanoparticle-based radiosensitizers (herein called nanoradiosensitizers) have recently gained attention owing to their ability to produce massive electrons which leads to the production of a huge amount of ROS. The success of the nanoradiosensitizer effect is closely correlated to its interaction with cells and its localization within the cells. In other words, tumor treatment is affected from the chain of events which is started from cell-nanoparticle interaction followed by the nanoparticles direction and homing inside the cell. Therefore, passive or active targeting of the nanoradiosensitizers in the subcellular level and the cell-nano interaction would determine the efficacy of the radiation therapy. The importance of the nanoradiosensitizer's targeting is increased while the organelles beyond nucleus are recently recognized as the mediators of the cancer cell death or resistance under RT. In this review, the principals of cell-nanomaterial interactions and which dominate nanoradiosensitizer efficiency in cancer therapy, are thoroughly discussed.
Collapse
Affiliation(s)
- Mohammad Varzandeh
- Department of Materials Engineering, Isfahan University of Technology, Isfahan, 84156-83111, Iran.
| | - Sheyda Labbaf
- Department of Materials Engineering, Isfahan University of Technology, Isfahan, 84156-83111, Iran.
| | - Jaleh Varshosaz
- Novel Drug Delivery Systems Research Center and Department of Pharmaceutics, School of Pharmacy, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Sophie Laurent
- Laboratory of NMR and Molecular Imaging, Department of General, Organic Chemistry and Biomedical, University of Mons, Mons, Belgium.
| |
Collapse
|
35
|
Cancer – A devastating disease, but also an eye-opener and window into the deep mysteries of life and its origins. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2022; 175:131-139. [DOI: 10.1016/j.pbiomolbio.2022.09.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 09/01/2022] [Accepted: 09/28/2022] [Indexed: 01/04/2023]
|
36
|
Yıldız SZ, Bilir C, Eskiler GG, Bilir F. The Anticancer Potential of Chlorine Dioxide in Small-Cell Lung Cancer Cells. Cureus 2022; 14:e29989. [PMID: 36381770 PMCID: PMC9636887 DOI: 10.7759/cureus.29989] [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] [Accepted: 10/06/2022] [Indexed: 11/06/2022] Open
Abstract
Background Chlorine dioxide (ClO2) is an effective disinfectant consisting of oxygen, chloride, and potassium. Because of its high oxidative capacity, ClO2 exerts antimicrobial, antiviral, and antifungal effects. However, its anticancer effects remain to be elucidated. Methodology The anticancer activity of CIO2 was assessed on DMS114 small-cell lung cancer (SCLC) cells and human umbilical vein endothelial cells (HUVEC) as control by WST-1, Annexin V, cell cycle analysis, and acridine orange staining. We for the first time investigated the possible therapeutic effects of long-term stabilized ClO2 solution (LTSCD). Results Our preliminary findings showed that LTSCD significantly inhibited the proliferation of SCLC cells (p < 0.01) with less toxicity in HUVEC cells. Additionally, LTSCD induced apoptotic cell death in SCLC cells through nuclear blebbing and vacuolar formation. However, LTSCD treatment did not induce cell cycle arrest in both cell lines. Conclusions LTSCD can be a therapeutic potential for the treatment of SCLC. However, further investigations are required to assess the LTSCD-induced cell death in SCLC both in vitro and in vivo.
Collapse
Affiliation(s)
- Salih Zeki Yıldız
- Department of Chemistry, Faculty of Arts and Sciences, Sakarya University, Sakarya, TUR
| | - Cemil Bilir
- Medical Oncology, Istinye University, VM Medical Park Pendik Hospital, Istanbul, TUR
| | - Gamze Guney Eskiler
- Department of Medical Biology, Faculty of Medicine, Sakarya University, Sakarya, TUR
| | - Filiz Bilir
- Department of Gynecologic Oncology, Faculty of Medicine, Kocaeli University, Kocaeli, TUR
| |
Collapse
|
37
|
Salaroli R, Andreani G, Bernardini C, Zannoni A, La Mantia D, Protti M, Forni M, Mercolini L, Isani G. Anticancer activity of an Artemisia annua L. hydroalcoholic extract on canine osteosarcoma cell lines. Res Vet Sci 2022; 152:476-484. [PMID: 36156377 DOI: 10.1016/j.rvsc.2022.09.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 09/06/2022] [Accepted: 09/12/2022] [Indexed: 11/28/2022]
Abstract
Since ancient times, Artemisia annua (A. annua) has been used as a medicinal plant in Traditional Chinese Medicine. In addition, recent studies have investigated the cytotoxic effects of A. annua extracts towards cancer cells. The leading aim of the present research is to evaluate the cytotoxic effects of an hydroalcoholic extract of A. annua on two canine osteosarcoma (OSA) cell lines, OSCA-8 and OSCA-40, focusing on the possible involvement of ferroptosis. The quantitative determination of artemisinin concentration in the extract, culture medium and OSA cells was carried out through the use of an instrumental analytical method based on liquid chromatography coupled with spectrophotometric detection and tandem mass spectrometry (LC-DAD-MS/MS). OSCA-8 and OSCA-40 were exposed to different dilutions of the extract for the EC50 calculation then the uptake of artemisinin by the cells, the effects on the cell cycle, the intracellular iron level, the cellular morphology and the lipid oxidation state were evaluated. A concentration of artemisinin of 63.8 ± 3.4 μg/mL was detected in the extract. A dose-dependent cytotoxic effect was evidenced. In OSCA-40 alterations of the cell cycle and a significantly higher intracellular iron content were observed. In both cell lines the treatment with the extract was associated with lipid peroxidation and with the appearance of a "ballooning" phenotype suggesting the activation of ferroptosis. In conclusion the A. annua idroalcoholic extract utilized in this study showed anticancer activity on canine OSA cell lines that could be useful in treating drug resistant canine OSAs.
Collapse
Affiliation(s)
- Roberta Salaroli
- Department of Veterinary Medical Sciences (DIMEVET), Alma Mater Studiorum - University of Bologna, 40064 Ozzano Emilia, Bologna, Italy.
| | - Giulia Andreani
- Department of Veterinary Medical Sciences (DIMEVET), Alma Mater Studiorum - University of Bologna, 40064 Ozzano Emilia, Bologna, Italy.
| | - Chiara Bernardini
- Department of Veterinary Medical Sciences (DIMEVET), Alma Mater Studiorum - University of Bologna, 40064 Ozzano Emilia, Bologna, Italy.
| | - Augusta Zannoni
- Department of Veterinary Medical Sciences (DIMEVET), Alma Mater Studiorum - University of Bologna, 40064 Ozzano Emilia, Bologna, Italy.
| | - Debora La Mantia
- Department of Veterinary Medical Sciences (DIMEVET), Alma Mater Studiorum - University of Bologna, 40064 Ozzano Emilia, Bologna, Italy.
| | - Michele Protti
- Research Group of Pharmaco-Toxicological Analysis (PTA Lab), Department of Pharmacy and Biotechnology (FaBiT), Alma Mater Studiorum - University of Bologna, 40126 Bologna, Italy.
| | - Monica Forni
- Department of Veterinary Medical Sciences (DIMEVET), Alma Mater Studiorum - University of Bologna, 40064 Ozzano Emilia, Bologna, Italy.
| | - Laura Mercolini
- Research Group of Pharmaco-Toxicological Analysis (PTA Lab), Department of Pharmacy and Biotechnology (FaBiT), Alma Mater Studiorum - University of Bologna, 40126 Bologna, Italy.
| | - Gloria Isani
- Department of Veterinary Medical Sciences (DIMEVET), Alma Mater Studiorum - University of Bologna, 40064 Ozzano Emilia, Bologna, Italy.
| |
Collapse
|
38
|
Li Y, Guo S, Zhao Y, Li R, Li Y, Qiu C, Xiao L, Gong K. EZH2 Regulates ANXA6 Expression via H3K27me3 and Is Involved in Angiotensin II-Induced Vascular Smooth Muscle Cell Senescence. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:4838760. [PMID: 36160712 PMCID: PMC9492406 DOI: 10.1155/2022/4838760] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/14/2022] [Accepted: 08/01/2022] [Indexed: 02/05/2023]
Abstract
Objectives Abdominal aortic aneurysm (AAA) has a high risk of rupture of the aorta and is one of the leading causes of death in older adults. This study is aimed at confirming the influence and mechanism of the abnormally expressed ANXA6 gene in AAA. Methods Clinical samples were collected for proteome sequencing to screen for differentially expressed proteins. An Ang II-induced vascular smooth muscle cell (VSMC) aging model as well as an AAA animal model was used. Using RT-qPCR to detect the mRNA levels of EZH2, ANXA6, IK-6, and IL-8 in cells and tissues were assessed. Western blotting and immunohistochemistry staining were used apply for the expression of associated proteins in cells and tissues. SA-β-gal staining, flow cytometry, and DHE staining were used to detect senescent cells and the level of ROS. The cell cycle was assessed by flow cytometry. Arterial pathology was observed by HE staining. The aging of VSMCs in arterial tissue was assessed by coimmunofluorescence for α-SMA and p53. Results There were 24 differentially expressed proteins in the AAA clinical samples, including 10 upregulated protein and 14 downregulated protein, and the differential expression of ANXA6 was associated with vascular disease. Our study found that ANXA6 was highly expressed and EZH2 was lowly expressed in an Ang II-induced VSMC aging model. Knockdown of ANXA6 or overexpression of EZH2 inhibited Ang II-induced ROS, inhibited cell senescence, decreased Ang II evoked G1 arrest, and increased cells in G2 phase, while overexpression of ANXA6 played the opposite role. Overexpression of EZH2 inhibited ANXA6 expression by increasing H3K27me3 modification at the ANXA6 promoter. Simultaneous overexpression of EZH2 and the protective effect of EZH2 on cell senescence were partially reversed by ANXA6. Similarly, ANXA6 was highly expressed and EZH2 was lowly expressed in an Ang II-induced AAA animal model. Knockdown of ANXA6 and overexpression of EZH2 alleviated Ang II-induced VSMC senescence and inhibited AAA progression, while simultaneous overexpression of EZH2 and ANXA6 partially reversed the protective effect of EZH2 on AAA. Conclusion EZH2 regulates the ANXA6 promoter H3K27me3 modification, inhibits ANXA6 expression, alleviates Ang II-induced VSMC senescence, and inhibits AAA progression.
Collapse
Affiliation(s)
- Yuejin Li
- Department of General Surgery, The First People's Hospital of Yunnan Province (The Affiliated Hospital of Kunming University of Science and Technology), Kunming, Yunnan 650032, China
| | - Shikui Guo
- Department of General Surgery, The First People's Hospital of Yunnan Province (The Affiliated Hospital of Kunming University of Science and Technology), Kunming, Yunnan 650032, China
| | - Yingpeng Zhao
- Department of Hepatic-Biliary-Pancreatic Surgery, The First Hospital of Kunming (The Calmette Hospital), Kunming, Yunnan 650224, China
| | - Rougang Li
- Department of General Surgery, The First People's Hospital of Yunnan Province (The Affiliated Hospital of Kunming University of Science and Technology), Kunming, Yunnan 650032, China
| | - Yu Li
- Department of General Surgery, The First People's Hospital of Yunnan Province (The Affiliated Hospital of Kunming University of Science and Technology), Kunming, Yunnan 650032, China
| | - Changtao Qiu
- Department of General Surgery, The First People's Hospital of Yunnan Province (The Affiliated Hospital of Kunming University of Science and Technology), Kunming, Yunnan 650032, China
| | - Le Xiao
- Department of General Surgery, The First People's Hospital of Yunnan Province (The Affiliated Hospital of Kunming University of Science and Technology), Kunming, Yunnan 650032, China
| | - Kunmei Gong
- Department of General Surgery, The First People's Hospital of Yunnan Province (The Affiliated Hospital of Kunming University of Science and Technology), Kunming, Yunnan 650032, China
| |
Collapse
|
39
|
Methanol Extract of Clavularia inflata Exerts Apoptosis and DNA Damage to Oral Cancer Cells. Antioxidants (Basel) 2022; 11:antiox11091777. [PMID: 36139851 PMCID: PMC9495492 DOI: 10.3390/antiox11091777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 09/02/2022] [Accepted: 09/06/2022] [Indexed: 11/16/2022] Open
Abstract
Antiproliferation effects of Clavularia-derived natural products against cancer cells have been reported on, but most studies have focused on identifying bioactive compounds, lacking a detailed investigation of the molecular mechanism. Crude extracts generally exhibit multiple targeting potentials for anticancer effects, but they have rarely been assessed for methanol extracts of Clavularia inflata (MECI). This investigation aims to evaluate the antiproliferation of MECI and to examine several potential mechanisms between oral cancer and normal cells. A 24 h MTS assay demonstrated that MECI decreased cell viability in several oral cancer cell lines more than in normal cells. N-acetylcysteine (NAC), an oxidative stress inhibitor, recovered these antiproliferation effects. Higher oxidative stress was stimulated by MECI in oral cancer cells than in normal cells, as proven by examining reactive oxygen species and mitochondrial superoxide. This preferential induction of oxidative stress was partly explained by downregulating more cellular antioxidants, such as glutathione, in oral cancer cells than in normal cells. Consequently, the MECI-generated high oxidative stress in oral cancer cells was preferred to trigger more subG1 population, apoptosis expression (annexin V and caspase activation), and DNA damage, reverted by NAC. In conclusion, MECI is a potent marine natural product showing preferential antiproliferation against oral cancer cells.
Collapse
|
40
|
Oh J, An H, Yeo HJ, Choi S, Oh J, Kim S, Kim JM, Choi J, Lee S. Colchicine as a novel drug for the treatment of osteosarcoma through drug repositioning based on an FDA drug library. Front Oncol 2022; 12:893951. [PMID: 36059694 PMCID: PMC9433722 DOI: 10.3389/fonc.2022.893951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 07/21/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundColchicine is a traditional medication that is currently approved to treat gout and familial Mediterranean fever (FMF). However, colchicine has a wide range of anti-inflammatory activities, and several studies have indicated that it may be useful in a variety of other conditions, such as rheumatic disease, cardiac disease, and cancer. Osteosarcoma, the most common type of bone sarcoma, is derived from primitive bone-forming mesenchymal cells. In this study, we investigated whether colchicine could be used to treat osteosarcoma through the regulation of cell cycle signaling.MethodsTwo human osteosarcoma cell lines, U2OS and Saos-2, were used. A clonogenic assay was used to determine the antiproliferative effects of colchicine on osteosarcoma cells. Reactive oxygen species (ROS) production and apoptosis were measured by flow cytometry. Migration and invasion assays were performed to investigate the inhibitory effects of colchicine. The signaling pathways related to colchicine treatment were verified by GO biological process (GOBP) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses.ResultsColchicine was selected as the lead compound based on the results of initial screening and cell viability assays conducted in Saos-2 and U2Os cells. Colchicine reduced the viability of Saos-2 and U2OS cells in a concentration-dependent manner. It also significantly inhibited colony-forming ability and induced ROS production and apoptosis. It also inhibited the migration and invasion of both Saos-2 and U2OS cells. GOBP and KEGG enrichment analyses indicated the involvement of microtubule-based processes and cancer-related pathways.ConclusionsThese findings suggest that colchicine has therapeutic potential in osteosarcoma.
Collapse
Affiliation(s)
- Jisun Oh
- Department of Orthopaedic Surgery, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam-si, South Korea
| | - Hyun−Ju An
- Department of Orthopaedic Surgery, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam-si, South Korea
| | - Hyun Jeong Yeo
- Department of Orthopaedic Surgery, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam-si, South Korea
| | - Sujin Choi
- Department of Orthopaedic Surgery, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam-si, South Korea
| | - Jisu Oh
- Division of Hemato-Oncology, Department of Internal Medicine, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin-si, South Korea
| | - Segi Kim
- Department of Orthopaedic Surgery, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam-si, South Korea
| | - Jin Man Kim
- Department of Oral Microbiology and Immunology School of Dentistry, Seoul National University, Seoul, South Korea
| | - Junwon Choi
- Department of Molecular Science and Technology, Ajou University, Suwon-si, South Korea
| | - Soonchul Lee
- Department of Orthopaedic Surgery, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam-si, South Korea
- *Correspondence: Soonchul Lee,
| |
Collapse
|
41
|
Bukkuri A, Pienta KJ, Austin RH, Hammarlund EU, Amend SR, Brown JS. A life history model of the ecological and evolutionary dynamics of polyaneuploid cancer cells. Sci Rep 2022; 12:13713. [PMID: 35962062 PMCID: PMC9374668 DOI: 10.1038/s41598-022-18137-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 08/05/2022] [Indexed: 11/09/2022] Open
Abstract
Therapeutic resistance is one of the main reasons for treatment failure in cancer patients. The polyaneuploid cancer cell (PACC) state has been shown to promote resistance by providing a refuge for cancer cells from the effects of therapy and by helping them adapt to a variety of environmental stressors. This state is the result of aneuploid cancer cells undergoing whole genome doubling and skipping mitosis, cytokinesis, or both. In this paper, we create a novel mathematical framework for modeling the eco-evolutionary dynamics of state-structured populations and use this framework to construct a model of cancer populations with an aneuploid and a PACC state. Using in silico simulations, we explore how the PACC state allows cancer cells to (1) survive extreme environmental conditions by exiting the cell cycle after S phase and protecting genomic material and (2) aid in adaptation to environmental stressors by increasing the cancer cell's ability to generate heritable variation (evolvability) through the increase in genomic content that accompanies polyploidization. In doing so, we demonstrate the ability of the PACC state to allow cancer cells to persist under therapy and evolve therapeutic resistance. By eliminating cells in the PACC state through appropriately-timed PACC-targeted therapies, we show how we can prevent the emergence of resistance and promote cancer eradication.
Collapse
Affiliation(s)
- Anuraag Bukkuri
- Cancer Biology and Evolution Program, Department of Integrated Mathematical Oncology, Moffitt Cancer Center, Tampa, USA.
| | - Kenneth J Pienta
- The Brady Urological Institute, Johns Hopkins School of Medicine, Baltimore, USA
| | | | - Emma U Hammarlund
- Nordic Center for Earth Evolution, University of Southern Denmark and Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Sarah R Amend
- The Brady Urological Institute, Johns Hopkins School of Medicine, Baltimore, USA
| | - Joel S Brown
- Cancer Biology and Evolution Program, Department of Integrated Mathematical Oncology, Moffitt Cancer Center, Tampa, USA
| |
Collapse
|
42
|
Stochastic models of Mendelian and reverse transcriptional inheritance in state-structured cancer populations. Sci Rep 2022; 12:13079. [PMID: 35906318 PMCID: PMC9338039 DOI: 10.1038/s41598-022-17456-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Accepted: 07/26/2022] [Indexed: 11/08/2022] Open
Abstract
Recent evidence suggests that a polyaneuploid cancer cell (PACC) state may play a key role in the adaptation of cancer cells to stressful environments and in promoting therapeutic resistance. The PACC state allows cancer cells to pause cell division and to avoid DNA damage and programmed cell death. Transition to the PACC state may also lead to an increase in the cancer cell’s ability to generate heritable variation (evolvability). One way this can occur is through evolutionary triage. Under this framework, cells gradually gain resistance by scaling hills on a fitness landscape through a process of mutation and selection. Another way this can happen is through self-genetic modification whereby cells in the PACC state find a viable solution to the stressor and then undergo depolyploidization, passing it on to their heritably resistant progeny. Here, we develop a stochastic model to simulate both of these evolutionary frameworks. We examine the impact of treatment dosage and extent of self-genetic modification on eco-evolutionary dynamics of cancer cells with aneuploid and PACC states. We find that under low doses of therapy, evolutionary triage performs better whereas under high doses of therapy, self-genetic modification is favored. This study generates predictions for teasing apart these biological hypotheses, examines the implications of each in the context of cancer, and provides a modeling framework to compare Mendelian and non-traditional forms of inheritance.
Collapse
|
43
|
Temiz E, Eği K, Koyuncu I, Yüksekdag O, Kurt Y, Tiken M, Akmese S. Cedrus libani tar prompts reactive oxygen species toxicity and DNA damage in colon cancer cells. Mol Biol Rep 2022; 49:7939-7952. [PMID: 35666426 DOI: 10.1007/s11033-022-07631-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 05/19/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND Many chemotherapeutic drugs used in cancer treatment have anticancer properties by inducing reactive oxygen species (ROS). However, the same effect occurs in normal cells, limiting the availability of these drugs. Therefore, studies on the detection of new herbal anticancer agents that have selective effects on cancer cells are of great importance. The aim of this study is to investigate the metabolite profile of Cedrus libani tar and its mechanism of anticancer effect on colon cancer cells. METHODS AND RESULTS Effect of cedar tar on cells (12 cancers and 5 normal cell lines) viability was determined by MTT, apoptosis induction was determined by Annexin-V, ROS and MMP determined by flow cytometry assay. Cleaved caspase-8, 9 and Ɣ-H2AX expression determined by western blot. Apoptotic and antioxidant genes expression level determined by qPCR. Metabolite profiling was performed with LC-MS/MS and GC-MS. Cedar tar showed the highest cytotoxic effect among cancer cells in colon cancer (HCT-116, IC50: 30.4 μg/mL) and its toxic effect on normal cells (HUVEC, IC50: 74.07 μg/mL) was less than cancer cell. Cedar tar increases ROS production in colon cancer cells. The metabolite profile of the cedar tar contains high amounts of metabolites such as fatty acids mainly (Duprezianene, Himachalene and Chamigrene), phenolic compounds (mostly Coumarin, p-coumaric acid, Vanillic acid and tr-Ferulic acid etc.) and organic acids (mainly 3-oh propanoic acid, 2-oh butyric acid and 3-oh isovaleric acid etc.). CONCLUSION As a result, it has been found that cedar tar has the potential to be used in the treatment of colon cancer.
Collapse
Affiliation(s)
- Ebru Temiz
- Medical Promotion and Marketing Program, Health Services Vocational School, Harran University, Şanlıurfa, Turkey
| | - Kadir Eği
- Department of Medicinal Biochemistry, Medical Faculty, Harran University, Şanlıurfa, Turkey
| | - Ismail Koyuncu
- Department of Medicinal Biochemistry, Medical Faculty, Harran University, Şanlıurfa, Turkey.
| | - Ozgür Yüksekdag
- Department of Medicinal Biochemistry, Medical Faculty, Harran University, Şanlıurfa, Turkey
| | - Yusuf Kurt
- Department of Molecular Biology and Genetic, Science Faculty, Harran University, Şanlıurfa, Turkey
| | - Murat Tiken
- Department of Medicinal Biochemistry, Medical Faculty, Harran University, Şanlıurfa, Turkey
| | - Sükrü Akmese
- Department of Medicinal Biochemistry, Medical Faculty, Harran University, Şanlıurfa, Turkey
| |
Collapse
|
44
|
Chen SY, Chao CN, Huang HY, Fang CY. Auranofin induces urothelial carcinoma cell death via reactive oxygen species production and synergy with cisplatin. Oncol Lett 2022; 23:61. [PMID: 35069870 PMCID: PMC8756563 DOI: 10.3892/ol.2021.13179] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 11/30/2021] [Indexed: 11/13/2022] Open
Abstract
Urothelial carcinoma (UC) is one of the most common cancer types of the urinary tract. UC is associated with poor 5-year survival rate, and resistance to cisplatin-based therapy remains a challenge for invasive bladder cancer treatment. Therefore, there is an urgent need to develop new drugs for advanced UC therapy. Auranofin (AF) was developed over 30 years ago for the treatment of rheumatoid arthritis and has been reported to exert an antitumor effect by increasing the level of reactive oxygen species (ROS) in cancer cells. The aim of the present study was to examine the effects of AF on cancer cell proliferation, cell cycle and apoptosis, either alone or in combination with cisplatin. AF induced cell death in two separate cell lines, HT 1376 and BFTC 909, in a concentration- and time-dependent manner by inducing cell cycle arrest. However, the distribution of cells in different phases of the cell cycle differed between the two cell lines, with G0/G1 cell cycle arrest in HT 1376 cells and S phase arrest in BFTC 909 cells. In addition, AF induced apoptosis in HT 1376, as well as redox imbalance in both HT 1376 and BFTC 909 cells. Cell viability was rescued following treatment with N-acetyl-L-cysteine, a ROS scavenger. Furthermore, AF treatment synergistically increased the cytotoxicity of HT 1376 and BFTC 909 cells when combined with cisplatin treatment. These findings suggest that AF may represent a potential candidate drug against UC and increase the therapeutic effect of cisplatin.
Collapse
Affiliation(s)
- San-Yuan Chen
- Department of Chinese Medicine, Ditmanson Medical Foundation, Chiayi Christian Hospital, Chiayi 60002, Taiwan, R.O.C
- Department of Sports Management, Chia Nan University of Pharmacy and Science, Tainan 71710, Taiwan, R.O.C
| | - Chun-Nun Chao
- Department of Pediatrics, Ditmanson Medical Foundation, Chiayi Christian Hospital, Chiayi 60002, Taiwan, R.O.C
- Department of Biotechnology, Asia University, Taichung 41354, Taiwan, R.O.C
| | - Hsin-Yi Huang
- Department of Medical Research, Ditmanson Medical Foundation, Chiayi Christian Hospital, Chiayi 60002, Taiwan, R.O.C
| | - Chiung-Yao Fang
- Department of Medical Research, Ditmanson Medical Foundation, Chiayi Christian Hospital, Chiayi 60002, Taiwan, R.O.C
| |
Collapse
|
45
|
The potential inhibitory effect of ginsenoside Rh2 on mitophagy in UV-irradiated human dermal fibroblasts. J Ginseng Res 2022; 46:646-656. [PMID: 36090683 PMCID: PMC9459079 DOI: 10.1016/j.jgr.2022.02.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 01/27/2022] [Accepted: 02/03/2022] [Indexed: 11/30/2022] Open
Abstract
Background In addition to its use as a health food, ginseng is used in cosmetics and shampoo because of its extensive health benefits. The ginsenoside, Rh2, is a component of ginseng that inhibits tumor cell proliferation and differentiation, promotes insulin secretion, improves insulin sensitivity, and shows antioxidant effects. Methods The effects of Rh2 on cell survival, extracellular matrix (ECM) protein expression, and cell differentiation were examined. The antioxidant effects of Rh2 in UV-irradiated normal human dermal fibroblast (NHDF) cells were also examined. The effects of Rh2 on mitochondrial function, morphology, and mitophagy were investigated in UV-irradiated NHDF cells. Results Rh2 treatment promoted the proliferation of NHDF cells. Additionally, Rh2 increased the expression levels of ECM proteins and growth-associated immediate-early genes in ultraviolet (UV)-irradiated NHDF cells. Rh2 also affected antioxidant protein expression and increased total antioxidant capacity. Furthermore, treatment with Rh2 ameliorated the changes in mitochondrial morphology, induced the recovery of mitochondrial function, and inhibited the initiation of mitophagy in UV-irradiated NHDF cells. Conclusion Rh2 inhibits mitophagy and reinstates mitochondrial ATP production and membrane potential in NHDF cells damaged by UV exposure, leading to the recovery of ECM, cell proliferation, and antioxidant capacity.
Collapse
|