1
|
Wang N, Zhou X, Zhang T, Jian W, Sun Z, Qi P, Feng Y, Liu H, Liu L, Yang S. Capsaicin from chili peppers and its analogues and their valued applications: An updated literature review. Food Res Int 2025; 208:116034. [PMID: 40263816 DOI: 10.1016/j.foodres.2025.116034] [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: 06/27/2024] [Revised: 10/24/2024] [Accepted: 02/21/2025] [Indexed: 04/24/2025]
Abstract
Chili peppers are widely sought after by consumers for not only their color, flavor, and nutritional properties but also their main component (capsaicin) various biological activities in diverse fields. Capsaicin (trans-8-methyl-N-vanillyl-6-nonenamide), the compound primarily responsible for the spicy flavor of peppers, remains a hot topic in the scientific community and shows the vast potential in various applications. Although many reviews focus comprehensively on capsaicin, most articles are limited to the medical field of capsaicin. This review provides an overview briefly of the capsaicin and its analogues in the fields of food, medicine and with a particular emphasis on their applications in agriculture and livestock farming. Overall, we aims is to expand the broad spectrum of applications for capsaicin and its analogues and explore their potential biological mechanisms. Finally, the challenges of capsaicin and future development prospects were discussed and proposed.
Collapse
Affiliation(s)
- Na Wang
- State Key Laboratory of Green Pesticides, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang 550025, China.; Innovation Center of Pesticide Research, Department of Applied Chemistry, College of Science, China Agricultural University, Beijing 100193, China
| | - Xiang Zhou
- State Key Laboratory of Green Pesticides, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang 550025, China..
| | - Taihong Zhang
- State Key Laboratory of Green Pesticides, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang 550025, China
| | - Wujun Jian
- State Key Laboratory of Green Pesticides, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang 550025, China
| | - Zhaoju Sun
- State Key Laboratory of Green Pesticides, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang 550025, China
| | - Puying Qi
- State Key Laboratory of Green Pesticides, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang 550025, China
| | - Yumei Feng
- State Key Laboratory of Green Pesticides, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang 550025, China
| | - Hongwu Liu
- State Key Laboratory of Green Pesticides, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang 550025, China
| | - Liwei Liu
- State Key Laboratory of Green Pesticides, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang 550025, China
| | - Song Yang
- State Key Laboratory of Green Pesticides, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang 550025, China..
| |
Collapse
|
2
|
Yang Y, Huangfu L, Li H, Yang D. Research progress of hyperthermia in tumor therapy by influencing metabolic reprogramming of tumor cells. Int J Hyperthermia 2023; 40:2270654. [PMID: 37871910 DOI: 10.1080/02656736.2023.2270654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 10/09/2023] [Indexed: 10/25/2023] Open
Abstract
Cellular metabolic reprogramming is an important feature of malignant tumors. Metabolic reprogramming causes changes in the levels or types of specific metabolites inside and outside the cell, which affects tumorigenesis and progression by influencing gene expression, the cellular state, and the tumor microenvironment. During tumorigenesis, a series of changes in the glucose metabolism, fatty acid metabolism, amino acid metabolism, and cholesterol metabolism of tumor cells occur, which are involved in the process of cellular carcinogenesis and constitute part of the underlying mechanisms of tumor formation. Hyperthermia, as one of the main therapeutic tools for malignant tumors, has obvious effects on tumor cell metabolism. In this paper, we will combine the latest research progress in the field of cellular metabolic reprogramming and focus on the current experimental research and clinical treatment of hyperthermia in cellular metabolic reprogramming to discuss the feasibility of cellular metabolic reprogramming-related mechanisms guiding hyperthermia in malignant tumor treatment, so as to provide more ideas for hyperthermia to treat malignant tumors through the direction of cellular metabolic reprogramming.
Collapse
Affiliation(s)
- Yuchuan Yang
- Department of Radiotherapy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, P.R. China
| | - Linkuan Huangfu
- Department of Radiotherapy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, P.R. China
| | - Huizhen Li
- Department of Radiotherapy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, P.R. China
| | - Daoke Yang
- Department of Radiotherapy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, P.R. China
| |
Collapse
|
3
|
Wikan N, Tocharus J, Oka C, Sivasinprasasn S, Chaichompoo W, Denlumpai P, Suksamrarn A, Tocharus C. Pelargonic acid vanillylamide alleviates hepatic autophagy and ER stress in hepatic steatosis model. Food Chem Toxicol 2023; 180:113987. [PMID: 37611858 DOI: 10.1016/j.fct.2023.113987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 07/24/2023] [Accepted: 08/11/2023] [Indexed: 08/25/2023]
Abstract
Pelargonic acid vanillylamide (PAVA) has been shown to reduce hepatic lipid accumulation in an obese rat model, however the underlying mechanism responsible for regulating lipid metabolism remains unclear. This study investigated the molecular mechanisms invoked by PAVA in regulating lipogenesis, autophagy, and endoplasmic reticulum (ER) stress in obese rats. Male Sprague-Dawley rats were fed on a diet consisting of 65.26% fat (16 weeks) and HepG2 cells were incubated with 200 μM oleic acid (OA) plus 100 μM palmitic acid (PA) for 48 h. These treatments resulted in a steatosis model. PAVA was shown to reduce fat deposition in hepatocytes in HepG2 by reducing lipotoxicity, the triglyceride content, the expression of sterol regulatory element binding protein 1c (SREBP-1c) and fatty acid synthase (FASN). PAVA also significantly reduced the calcium level and the expression of calpain 2 and upregulated the expression of Atg7 in comparison to the HFD group. In addition, PAVA was shown to significantly decrease the expression of autophagy pathway-related proteins including LC3 and p62. Treatment with PAVA (1 mg/day) reduced the expressions of ER stress markers Bip, ATF6 (p50), p-IRE1/IRE1, p-eIF2α/eIF2α, pJNK, CHOP and cleaved CASP12. In conclusion, PAVA ameliorated obesity induced hepatic steatosis by attenuating defective autophagy and ER stress pathways.
Collapse
Affiliation(s)
- Naruemon Wikan
- Department of Anatomy, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Jiraporn Tocharus
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Chio Oka
- Laboratory of Functional Genomics and Medicine, Division of Biological Science, Nara Institute of Science and Technology, Nara, Japan
| | | | - Waraluck Chaichompoo
- Department of Chemistry and Center of Excellence for Innovation in Chemistry, Faculty of Science, Ramkhamhaeng University, Bangkok, Thailand
| | - Panida Denlumpai
- Department of Chemistry and Center of Excellence for Innovation in Chemistry, Faculty of Science, Ramkhamhaeng University, Bangkok, Thailand
| | - Apichart Suksamrarn
- Department of Chemistry and Center of Excellence for Innovation in Chemistry, Faculty of Science, Ramkhamhaeng University, Bangkok, Thailand
| | - Chainarong Tocharus
- Department of Anatomy, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Radiation Research and Medical Imaging, Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand.
| |
Collapse
|
4
|
Vijayan S, Loganathan C, Sakayanathan P, Thayumanavan P. In silico and in vitro investigation of anticancer effect of newly synthesized nonivamide-s-allyl cysteine ester. J Biomol Struct Dyn 2022; 40:11511-11525. [PMID: 34344261 DOI: 10.1080/07391102.2021.1959404] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Nonivamide (NOV), less pungent analogue of capsaicin present in various Capsicum species is known for various biological properties. S-allyl cysteine (SAC) abundantly present in aged garlic extract is gaining importance for anticancer property. NOV was esterified with SAC to increase the biological activity. In silico ADME analysis revealed the drug-likeness of NOV-SAC. Molecular docking and dynamics simulation analysis were done to understand the interaction of NOV-SAC with therapeutic target proteins (human estrogen receptor α, tumo protein negative regulator mouse double minute 2, B-cell lymphoma 2 and cyclin-dependent kinase 2) to treat cancer. NOV-SAC interacted with these proteins stably with favorable binding energy which was calculated through MMGBSA method. In line with in silico results, NOV-SAC showed antiproliferative activity against breast cancer cell line (MCF-7). NOV-SAC treatment increased ROS generation, decreased the antioxidant level, arrested cells at G1/S phase, disrupted mitochondrial membrane potential and initiated DNA fragmentation. The expression of p53 is increased by NOV-SAC treatment, in concordance the ratio of Bcl-2/Bax was decreased. Altogether, NOV-SAC was synthesized for the first time and it induced apoptosis in MCF-7 cells through triggering ROS generation and increasing the expression of p53. The in silico results has been mirrored in in vitro analysis of NOV-SAC against cancer cell line.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Sudha Vijayan
- Department of Biochemistry, Periyar University, Salem, Tamil Nadu, India
| | - Chitra Loganathan
- Department of Biochemistry, Periyar University, Salem, Tamil Nadu, India
| | | | | |
Collapse
|
5
|
Sun J, Zhang X, Wang X, Peng J, Song G, Di Y, Feng F, Wang S. Dithiol-Activated Bioorthogonal Chemistry for Endoplasmic Reticulum-Targeted Synergistic Chemophototherapy. Angew Chem Int Ed Engl 2022; 61:e202213765. [PMID: 36342403 DOI: 10.1002/anie.202213765] [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: 09/19/2022] [Indexed: 11/09/2022]
Abstract
The controlled intracellular release of nitrite is still an unmet challenge due to the lack of bio-friendly donors, and the antitumor effect of nitrite is limited by its physiologically inert activity. Herein, we designed benzothiadiazole-based organic nitrite donors that are stable against bio-relevant species but selectively respond to dithiol species through SN Ar/intramolecular cyclization tandem reactions in the aqueous media. The bioorthogonal system was established to target the endoplasmic reticulum (ER) of liver cancer HepG2 cells. The nitrite and nonivamide were coupled to induce elevation of intracellular levels of calcium ions as well as reactive oxygen/nitrogen species, which resulted in ER stress and mitochondrial dysfunction. We demonstrated that a combination of photoactivation and "click to release" strategy could enhance antitumor effect in cellular level and show good potential for cancer precision therapy.
Collapse
Affiliation(s)
- Jian Sun
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, 100190, Beijing, P. R. China.,Department of Polymer Science & Engineering, School of Chemistry and Chemical Engineering, Nanjing University, 210023, Jiangsu, Nanjing, P. R. China
| | - Xiaoran Zhang
- Department of Polymer Science & Engineering, School of Chemistry and Chemical Engineering, Nanjing University, 210023, Jiangsu, Nanjing, P. R. China
| | - Xia Wang
- Department of Polymer Science & Engineering, School of Chemistry and Chemical Engineering, Nanjing University, 210023, Jiangsu, Nanjing, P. R. China
| | - Jinlei Peng
- Department of Polymer Science & Engineering, School of Chemistry and Chemical Engineering, Nanjing University, 210023, Jiangsu, Nanjing, P. R. China
| | - Gang Song
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, 100190, Beijing, P. R. China.,College of Chemistry, University of Chinese Academy of Sciences, 100190, Beijing, P. R. China
| | - Yufei Di
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, 100190, Beijing, P. R. China.,College of Chemistry, University of Chinese Academy of Sciences, 100190, Beijing, P. R. China
| | - Fude Feng
- Department of Polymer Science & Engineering, School of Chemistry and Chemical Engineering, Nanjing University, 210023, Jiangsu, Nanjing, P. R. China
| | - Shu Wang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, 100190, Beijing, P. R. China.,College of Chemistry, University of Chinese Academy of Sciences, 100190, Beijing, P. R. China
| |
Collapse
|
6
|
Nonivamide induces brown fat-like characteristics in porcine subcutaneous adipocytes. Biochem Biophys Res Commun 2022; 619:68-75. [PMID: 35738067 DOI: 10.1016/j.bbrc.2022.06.047] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 06/13/2022] [Accepted: 06/14/2022] [Indexed: 11/24/2022]
Abstract
Obesity, which is associated with type 2 diabetes, is a threat to human health. There are studies, which suggest that some compounds can induce browning of white adipocytes to combat obesity. In this study, we selected nonivamide, an analog of capsaicin, to detect whether it influenced the browning of porcine white adipocytes. First, we found 25 μM nonivamide promoted apoptosis of porcine subcutaneous pre-adipocytes. After pre-adipocytes differentiation, nonivamide inhibited adipogenesis by reducing the expressions of Pparγ, Cebpα, while it promoted lipolysis by up-regulating Hsl, Atgl. Nonivamide also induced browning of porcine subcutaneous adipocytes by up-regulating the expression of brown and beige adipocyte gene markers, such as Prdm16, Cidea, and Slc27a1. Additionally, thermogenesis gene markers Cpt1a and Cpt1b were significantly up-regulated by nonivamide. Furthermore, nonivamide promoted mitochondrial biogenesis by up-regulating the expression of Tfam, Nrf1, Nrf2, and Tomm20. In conclusion, nonivamide is a potent compound to induce porcine adipocyte browning for treating obesity.
Collapse
|
7
|
Hyperthermia Treatment as a Promising Anti-Cancer Strategy: Therapeutic Targets, Perspective Mechanisms and Synergistic Combinations in Experimental Approaches. Antioxidants (Basel) 2022; 11:antiox11040625. [PMID: 35453310 PMCID: PMC9030926 DOI: 10.3390/antiox11040625] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/18/2022] [Accepted: 03/19/2022] [Indexed: 02/04/2023] Open
Abstract
Despite recent developments in diagnosis and treatment options, cancer remains one of the most critical threats to health. Several anti-cancer therapies have been identified, but further research is needed to provide more treatment options that are safe and effective for cancer. Hyperthermia (HT) is a promising treatment strategy for cancer because of its safety and cost-effectiveness. This review summarizes studies on the anti-cancer effects of HT and the detailed mechanisms. In addition, combination therapies with anti-cancer drugs or natural products that can effectively overcome the limitations of HT are reviewed because HT may trigger protective events, such as an increase of heat shock proteins (HSPs). In the 115 reports included, the mechanisms related to apoptosis, cell cycle, reactive oxygen species, mitochondrial membrane potential, DNA damage, transcription factors and HSPs were considered important. This review shows that HT is an effective inducer of apoptosis. Moreover, the limitations of HT may be overcome using combined therapy with anti-cancer drugs or natural products. Therefore, appropriate combinations of such agents with HT will exert maximal effects to treat cancer.
Collapse
|
8
|
Pazouki N, Irani S, Olov N, Atyabi SM, Bagheri-Khoulenjani S. Fe 3O 4 nanoparticles coated with carboxymethyl chitosan containing curcumin in combination with hyperthermia induced apoptosis in breast cancer cells. Prog Biomater 2022; 11:43-54. [PMID: 35025086 PMCID: PMC8927563 DOI: 10.1007/s40204-021-00178-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 12/26/2021] [Indexed: 12/17/2022] Open
Abstract
Many studies have demonstrated that curcumin has potential anticancer properties. This research aims to study the effect of iron (II, III) oxide (Fe3O4) nanoparticles coated with carboxymethyl chitosan containing curcumin combination with hyperthermia on breast cancer cells. Magnetic nanoparticles coated with carboxymethyl chitosan containing curcumin (MNP-CMC-CUR) were prepared and specified. MCF-7, MDA-MB-231, and human fibroblast cells were treated with free curcumin and MNP-CMC-CUR at concentrations of 0-60 µM and at different time points. A combined therapy of MNP-CMC-CUR and hyperthermia was performed on MCF-7 cells. The cytotoxicity of curcumin and MNP-CMC-CUR combined with hyperthermia was assessed by MTT. The changes in TP53 and CASPASE3 gene expression were evaluated using real-time PCR. Both cell apoptosis and cell cycle were studied by Annexin/PI staining. The results of MTT showed that the IC50 amount of MNP-CMC-CUR has significantly decreased compared to free curcumin (p < 0.05) and MNP-CMC-CUR in combination with the hyperthermia, and significantly reducing the metabolic activity of the cells (p < 0.05). Real-time PCR results revealed the up-regulation of TP53 and CASPASE3 (p < 0.05). The combinational therapy-induced cell apoptosis (64.51%) and sub-G1 cell cycle were arrested in MCF-7 cells. Based on these observations, a combination of MNP-CMC-CUR with hyperthermia could inhibit the proliferation of MCF-7 cells.
Collapse
Affiliation(s)
- Negin Pazouki
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Shiva Irani
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran.
| | - Nafiseh Olov
- Department of Polymer and Color Engineering, Amirkabir University of Technology, Tehran, Iran
| | | | | |
Collapse
|
9
|
Enhancement of Radio-Thermo-Sensitivity of 5-Iodo-2-Deoxyuridine-Loaded Polymeric-Coated Magnetic Nanoparticles Triggers Apoptosis in U87MG Human Glioblastoma Cancer Cell Line. Cell Mol Bioeng 2021; 14:365-377. [PMID: 34295445 DOI: 10.1007/s12195-021-00675-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 05/05/2021] [Indexed: 01/22/2023] Open
Abstract
Introduction With an emphasis on the radioresistant nature of glioblastoma cells, the aim of the present study was to evaluate the radio-thermo-sensitizing effects of PCL-PEG-coated Superparamagnetic iron oxide nanoparticles (SPIONs) as a carrier of 5-iodo-2-deoxyuridine (IUdR) in monolayer culture of U87MG human glioma cell line. Methods Following monolayer culture of U87MG cells, nanoparticle uptake was assessed using Prussian blue staining and ICP-OES method. The U87MG cells were treated with an appropriate concentration of free IUdR and PCL-PEG-coated SPIONs (MNPs) loaded with IUdR (IUdR/MNPs) for 24 h, subjected to hyperthermia (water bath and alternating magnetic field (AMF)) at 43 °C, and exposed to X-ray (2 Gy, 6 MV). The combined effects of hyperthermia with or without magnetic nanoparticles on radiosensitivity of the U87MG cells were evaluated using colony formation assay (CFA) and Flowcytometry. Results Prussian blue staining and ICP-OES showed that the nanoparticles were able to enter the cells. The results also indicated that IUdR/MNPs combined with X-ray radiation and hyperthermia significantly decreased the colony formation ability of monolayer cells (1.11, 1.41 fold) and increased the percentage of apoptotic (2.47, 4.1 fold) and necrotic cells (12.28, 29.34 fold), when compared to IUdR combined with X-ray and hyperthermia or IUdR/MNPs + X-ray. MTT results revealed that the presence of IUdR/MNPs significantly increased the toxicity of AMF hyperthermia compared to the water bath method. Conclusions Our study showed that SPIONs/PCL-PEG, as a carrier of IUdR, can enhance the cytotoxic effects of radiotherapy and hyperthermia and act as a radio-thermo-sensitizing agent. Graphic Abstract Supplementary Information The online version contains supplementary material available at 10.1007/s12195-021-00675-y.
Collapse
|
10
|
Quintana M, Saavedra E, del Rosario H, González I, Hernández I, Estévez F, Quintana J. Ethanol Enhances Hyperthermia-Induced Cell Death in Human Leukemia Cells. Int J Mol Sci 2021; 22:ijms22094948. [PMID: 34066632 PMCID: PMC8125413 DOI: 10.3390/ijms22094948] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/04/2021] [Accepted: 05/04/2021] [Indexed: 12/23/2022] Open
Abstract
Ethanol has been shown to exhibit therapeutic properties as an ablative agent alone and in combination with thermal ablation. Ethanol may also increase sensitivity of cancer cells to certain physical and chemical antitumoral agents. The aim of our study was to assess the potential influence of nontoxic concentrations of ethanol on hyperthermia therapy, an antitumoral modality that is continuously growing and that can be combined with classical chemotherapy and radiotherapy to improve their efficiency. Human leukemia cells were included as a model in the study. The results indicated that ethanol augments the cytotoxicity of hyperthermia against U937 and HL60 cells. The therapeutic benefit of the hyperthermia/ethanol combination was associated with an increase in the percentage of apoptotic cells and activation of caspases-3, -8 and -9. Apoptosis triggered either by hyperthermia or hyperthermia/ethanol was almost completely abolished by a caspase-8 specific inhibitor, indicating that this caspase plays a main role in both conditions. The role of caspase-9 in hyperthermia treated cells acquired significance whether ethanol was present during hyperthermia since the alcohol enhanced Bid cleavage, translocation of Bax from cytosol to mitochondria, release of mitochondrial apoptogenic factors, and decreased of the levels of the anti-apoptotic factor myeloid cell leukemia-1 (Mcl-1). The enhancement effect of ethanol on hyperthermia-activated cell death was associated with a reduction in the expression of HSP70, a protein known to interfere in the activation of apoptosis at different stages. Collectively, our findings suggest that ethanol could be useful as an adjuvant in hyperthermia therapy for cancer.
Collapse
|
11
|
Li YL, Zhou DJ, Cui ZG, Sun L, Feng QW, Zakki SA, Hiraku Y, Wu CA, Inadera H. The molecular mechanism of a novel derivative of BTO-956 induced apoptosis in human myelomonocytic lymphoma cells. Apoptosis 2021; 26:219-231. [PMID: 33738673 DOI: 10.1007/s10495-021-01664-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/01/2021] [Indexed: 11/29/2022]
Abstract
Acute myeloid leukemia (AML) is a malignant cancer of the hematopoietic system. Although the effectiveness of arsenic compounds has been recognized and applied clinically, some patients are still found resistant to this chemotherapy. In this study, we investigated that a synthetic thyroid hormone analog (TA), 2-iodo-4-nitro-1-(o-tolyloxy) benzene, had a strong apoptosis effect on U937 cells. U937 cells were treated with TA, and examinted the generation of reactive oxygen species (ROS), dysfunction of mitochondria, expression of pro-apoptosis and anti-apoptosis, and cleavage of caspase-3 and Poly (ADP-ribose) polymerase (PARP). Further, it is also evaluated that insight molecular mechanism and signaling pathways involved in the study. It is found that TA significantly induced apoptosis in U937 cells through production of ROS, dysfunction of mitochondria, and activation of caspase cascade. It was also observed that MAPK signaling pathway including ERK, JNK, and P38 signals are involved in the induction of apoptosis. Moreover, marked activation of autophagy and ER stress markers such as LC3, P62, Beclin1 and GRP78, CHOP were observed, respectively. Pretreatment with ER stress inhibitor tauroursodeoxycholic acid (TUDCA) and autophagy inhibitor 3-Methyladenine (3-MA) have successfully attenuated and aggravated TA-induced apoptosis, respectively. We further confirmed the active involvement of ER stress and autophagy signals. In conclusion, TA induced apoptosis through ER stress and activation of autophagy, and the latter is not conducive to TA-induced cell death. Our results may provide a new insight into the strategic development of novel therapy for the treatment of AML.
Collapse
Affiliation(s)
- Yu-Lin Li
- Department of Public Health, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - De-Jun Zhou
- Graduate School of Medicine, Henan Polytechnic University, Jiaozuo, 454000, China
| | - Zheng-Guo Cui
- Department of Environmental Health, University of Fukui School of Medical Sciences, 23-3 Matsuoka Shimoaizuki Eiheiji, Fukui, 910-1193, Japan
| | - Lu Sun
- Department of Pediatric Cardiology, Heart Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Qian-Wen Feng
- Department of Public Health, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Shahbaz Ahmad Zakki
- Department of Public Health and Nutrition, The University of Haripur, Hattar Road, Haripur, KP, Pakistan
| | - Yusuke Hiraku
- Department of Environmental Health, University of Fukui School of Medical Sciences, 23-3 Matsuoka Shimoaizuki Eiheiji, Fukui, 910-1193, Japan
| | - Cheng-Ai Wu
- Department of Molecular Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Xicheng District Xinjiekou East Street on the 31st, Beijing, 100035, China.
| | - Hidekuni Inadera
- Department of Public Health, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan.
| |
Collapse
|
12
|
Zakki SA, Muhammad JS, Li JL, Sun L, Li ML, Feng QW, Li YL, Cui ZG, Inadera H. Melatonin triggers the anticancer potential of phenylarsine oxide via induction of apoptosis through ROS generation and JNK activation. Metallomics 2020; 12:396-407. [PMID: 31959998 DOI: 10.1039/c9mt00238c] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Melatonin, a safe endogenous hormone and a natural supplement, has recently been recognized to have antiproliferative effects and the ability to sensitize cells to other anticancer therapies. Phenylarsine oxide (PAO) has anticancer potential but it is considered as a toxic agent. In this study we combined melatonin to reduce the toxicity while securing the anti-cancer effects of PAO. Cell viability was determined by MTT assay, whereas cytotoxic assays were performed using an LDH cytotoxicity assay kit. Cell cycle analysis, Annexin V/PI staining, the mitochondrial membrane potential (MMP), mitochondrial calcium and reactive oxygen species (ROS) generation were analyzed using flow cytometry. Sytox stained cells were visualized by fluorescence microscopy and the expression of proteins was detected by western blotting. Melatonin increased the anticancer potential of PAO by decreasing the cell viability and increasing LDH release in various cancer cells. The mode of cell death was determined to be typical apoptosis, as evidenced by Annexin V/PI-stained cells, PARP cleavage, and caspase-3 activation, and with significant modulations in the expression of proapoptotic, antiapoptotic and cell cycle-related proteins. ROS generation played a critical role in induction of cell death by this combined treatment, which is validated by reversal of cytotoxicity upon cotreatment with NAC. Furthermore, the activation of MAPKs, especially JNK, contributed to the induction of cell death, accompanied by endoplasmic reticulum stress and autophagy, affirmed by the abrogation of cytotoxicity after JNK-IN-8 and TUDCA application. Melatonin showed promising potential as a chemotherapeutic agent in combination with PAO to achieve a better anticancer response.
Collapse
Affiliation(s)
- Shahbaz Ahmad Zakki
- Department of Public Health, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Grumezescu V, Gherasim O, Negut I, Banita S, Holban AM, Florian P, Icriverzi M, Socol G. Nanomagnetite-embedded PLGA Spheres for Multipurpose Medical Applications. MATERIALS 2019; 12:ma12162521. [PMID: 31398805 PMCID: PMC6719237 DOI: 10.3390/ma12162521] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Revised: 08/03/2019] [Accepted: 08/05/2019] [Indexed: 12/25/2022]
Abstract
We report on the synthesis and evaluation of biopolymeric spheres of poly(lactide-co-glycolide) containing different amounts of magnetite nanoparticles and Ibuprofen (PLGA-Fe3O4-IBUP), but also chitosan (PLGA-CS-Fe3O4-IBUP), to be considered as drug delivery systems. Besides morphological, structural, and compositional characterizations, the PLGA-Fe3O4-IBUP composite microspheres were subjected to drug release studies, performed both under biomimetically-simulated dynamic conditions and under external radiofrequency magnetic fields. The experimental data resulted by performing the drug release studies evidenced that PLGA-Fe3O4-IBUP microspheres with the lowest contents of Fe3O4 nanoparticles are optimal candidates for triggered drug release under external stimulation related to hyperthermia effect. The as-selected microspheres and their chitosan-containing counterparts were biologically assessed on macrophage cultures, being evaluated as biocompatible and bioactive materials that are able to promote cellular adhesion and proliferation. The composite biopolymeric spheres resulted in inhibited microbial growth and biofilm formation, as assessed against Staphylococcus aureus, Pseudomonas aeruginosa, and Candida albicans microbial strains. Significantly improved antimicrobial effects were reported in the case of chitosan-containing biomaterials, regardless of the microorganisms' type. The nanostructured composite biopolymeric spheres evidenced proper characteristics as prolonged and controlled drug release platforms for multipurpose biomedical applications.
Collapse
Affiliation(s)
- Valentina Grumezescu
- Lasers Department, National Institute for Lasers, Plasma, and Radiation Physics, 077125 Magurele, Romania.
| | - Oana Gherasim
- Lasers Department, National Institute for Lasers, Plasma, and Radiation Physics, 077125 Magurele, Romania
- Department of Science and Engineering of Oxide Materials and Nanomaterials, Faculty of Applied Chemistry and Materials Science, Politehnica University of Bucharest, 011061 Bucharest, Romania
| | - Irina Negut
- Lasers Department, National Institute for Lasers, Plasma, and Radiation Physics, 077125 Magurele, Romania
| | - Stefan Banita
- Lasers Department, National Institute for Lasers, Plasma, and Radiation Physics, 077125 Magurele, Romania
| | - Alina Maria Holban
- Microbiology & Immunology Department, Faculty of Biology, University of Bucharest, 77206 Bucharest, Romania
| | - Paula Florian
- Ligand-Receptor Interactions Department, Institute of Biochemistry, Romanian Academy, 060031 Bucharest, Romania
| | - Madalina Icriverzi
- Ligand-Receptor Interactions Department, Institute of Biochemistry, Romanian Academy, 060031 Bucharest, Romania
| | - Gabriel Socol
- Lasers Department, National Institute for Lasers, Plasma, and Radiation Physics, 077125 Magurele, Romania.
| |
Collapse
|