1
|
Man S, Bi J, Liu F, Xie W, Ma L. Vitamin C Inhibited Pulmonary Metastasis through Activating Nrf2/HO-1 Pathway. Mol Nutr Food Res 2024; 68:e2300706. [PMID: 38419398 DOI: 10.1002/mnfr.202300706] [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: 10/03/2023] [Revised: 12/12/2023] [Indexed: 03/02/2024]
Abstract
As an important nutritional component, vitamin C (Vc) shows good antitumor activity in a variety of cancer, but there are few studies in pulmonary metastasis. In order to verify its anticancer and antimetastatic effect, the study sets up H22 pulmonary metastasis mouse model. The results show that intraperitoneal injection of Vc inhibits pulmonary metastasis through up-regulating the expression of Nrf2, HO-1, cleaved caspases 3 and 9, and causing DNA damage and apoptosis which is similar to the pro-oxidant effect of Vc in p53 null cells (H1299 cells). Meanwhile, oral administration of Vc up-regulates the expression of p53, directly activates Nrf2/HO-1 pathway, increases expression of cleaved caspases 3 and 9, and ultimately inhibits pulmonary metastasis, which is the same as the antioxidant result of Vc in p53 wild-type cells. In addition, Vc inhibits the proliferation and migration of lung cancer cells in a concentration-dependent manner and has little cytotoxic effects on normal cells. Notably, the experiment further illustrates that besides intravenous Vc, oral Vc significantly inhibits the pulmonary metastasis in mice. All in all, these findings provide new clues for Vc-treated pulmonary metastasis in clinical research.
Collapse
Affiliation(s)
- Shuli Man
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, 300457, China
| | - Jingxian Bi
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, 300457, China
| | - Furui Liu
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, 300457, China
| | - Wenwen Xie
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, 300457, China
| | - Long Ma
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, 300457, China
| |
Collapse
|
2
|
Vo HVT, Nguyen YT, Kim N, Lee HJ. Vitamin A, D, E, and K as Matrix Metalloproteinase-2/9 Regulators That Affect Expression and Enzymatic Activity. Int J Mol Sci 2023; 24:17038. [PMID: 38069361 PMCID: PMC10707015 DOI: 10.3390/ijms242317038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 11/25/2023] [Accepted: 11/29/2023] [Indexed: 12/18/2023] Open
Abstract
Fat-soluble vitamins (vitamin A, D, E, and K) assume a pivotal role in maintaining human homeostasis by virtue of their enzymatic functions. The daily inclusion of these vitamins is imperative to the upkeep of various physiological processes including vision, bone health, immunity, and protection against oxidative stress. Current research highlights fat-soluble vitamins as potential therapeutics for human diseases, especially cancer. Fat-soluble vitamins exert their therapeutic effects through multiple pathways, including regulation of matrix metalloproteinases' (MMPs) expression and enzymatic activity. As MMPs have been reported to be involved in the pathology of various diseases, such as cancers, cardiovascular diseases, and neurological disorders, regulating the expression and/or activity of MMPs could be considered as a potent therapeutic strategy. Here, we summarize the properties of fat-soluble vitamins and their potential as promising candidates capable of effectively modulating MMPs through multiple pathways to treat human diseases.
Collapse
Affiliation(s)
- Ha Vy Thi Vo
- Department of Chemistry Education, Kongju National University, Gongju 32588, Republic of Korea;
| | - Yen Thi Nguyen
- Department of Chemistry, Kongju National University, Gongju 32588, Republic of Korea;
| | - Namdoo Kim
- Department of Chemistry, Kongju National University, Gongju 32588, Republic of Korea;
| | - Hyuck Jin Lee
- Department of Chemistry Education, Kongju National University, Gongju 32588, Republic of Korea;
- Kongju National University Institute of Science Education, Kongju National University, Gongju 32588, Republic of Korea
- Kongju National University’s Physical Fitness for Health Research Lab (KNUPFHR), Kongju National University, Gongju 32588, Republic of Korea
| |
Collapse
|
3
|
Gul S, Maqbool MF, Maryam A, Khan M, Shakir HA, Irfan M, Ara C, Li Y, Ma T. Vitamin K: A novel cancer chemosensitizer. Biotechnol Appl Biochem 2022; 69:2641-2657. [PMID: 34993998 DOI: 10.1002/bab.2312] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Accepted: 12/24/2021] [Indexed: 12/27/2022]
Abstract
Cancer incidences are growing rapidly and causing millions of deaths globally. Cancer treatment is one of the most exigent challenges. Drug resistance is a natural phenomenon and is considered one of the major obstacles in the successful treatment of cancer by chemotherapy. Combination therapy by the amalgamation of various anticancer drugs has suggested modulating tumor response by targeting various signaling pathways in a synergistic or additive manner. Vitamin K is an essential nutrient and has recently been investigated as a potential anticancer agent. The combination of vitamin K analogs, such as vitamins K1, K2, K3, and K5, with other chemotherapeutic drugs have demonstrated a safe, cost-effective, and most efficient way to overcome drug resistance and improved the outcomes of prevailing chemotherapy. Published reports have shown that vitamin K in combination therapy improved the efficacy of clinical drugs by promoting apoptosis and cell cycle arrest and overcoming drug resistance by inhibiting P-glycoprotein. In this review, we discuss the mechanism, cellular targets, and possible ways to develop vitamin K subtypes into effective cancer chemosensitizers. Finally, this review will provide a scientific basis for exploiting vitamin K as a potential agent to improve the efficacy of chemotherapeutic drugs.
Collapse
Affiliation(s)
- Sameena Gul
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China.,Cancer Research Lab, Institute of Zoology, University of the Punjab, Quaid-e-Azam Campus Lahore, Lahore, Pakistan
| | - Muhammad Faisal Maqbool
- Cancer Research Lab, Institute of Zoology, University of the Punjab, Quaid-e-Azam Campus Lahore, Lahore, Pakistan
| | - Amara Maryam
- Cancer Research Lab, Institute of Zoology, University of the Punjab, Quaid-e-Azam Campus Lahore, Lahore, Pakistan
| | - Muhammad Khan
- Cancer Research Lab, Institute of Zoology, University of the Punjab, Quaid-e-Azam Campus Lahore, Lahore, Pakistan
| | - Hafiz Abdullah Shakir
- Cancer Research Lab, Institute of Zoology, University of the Punjab, Quaid-e-Azam Campus Lahore, Lahore, Pakistan
| | - Muhammad Irfan
- Department of Biotechnology, University of Sargodha, Sargodha, Pakistan
| | - Chaman Ara
- Cancer Research Lab, Institute of Zoology, University of the Punjab, Quaid-e-Azam Campus Lahore, Lahore, Pakistan
| | - Yongming Li
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Tonghui Ma
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
4
|
Pala EE, Pala HG, Ekmekci S, Erbas O. Vitamin C (Ascorbic acid) protects from neuropathy caused by cisplatin, through enhanced heat shock protein-70 and reduced oxidant effect. Rev Assoc Med Bras (1992) 2022; 68:1017-1022. [PMID: 36134830 PMCID: PMC9574981 DOI: 10.1590/1806-9282.20220032] [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: 04/12/2022] [Accepted: 05/06/2022] [Indexed: 11/22/2022] Open
Abstract
OBJECTIVE: We aimed to determine whether vitamin C has a protective effect on cisplatin-induced neuropathy in rats. METHODS: In total, 24 rats were included in the study of which 8 rats (no drug administered) were categorized as the control group. The remaining 16 rats were given a total dose of 20 mg/kg cisplatin to induce neuropathy. These drug-administered rats (16 rats) were randomly divided into two groups, namely, group-1 (n=8): cisplatin+saline and group-2 (n=8): cisplatin+vitamin C (500 mg/kg/day). All rats were tested for motor function and electromyographic activity 3 days after cisplatin. Motor performance was evaluated by an inclined-plane test. Compound muscle action potential was evaluated. Plasma malondialdehyde, glutathione, tumor necrosis factor-α, interleukin 6, and sciatic nerve HSP 70 levels were measured. Axon diameter and nerve growth factor expression levels were analyzed. RESULTS: Plasma malondialdehyde, tumor necrosis factor-α, and interleukin 6 levels were higher in the cisplatin+saline group than control group (p<0.001). But vitamin C significantly reduced malondialdehyde and inflammatory cytokine levels when compared with the cisplatin+saline group (p<0.001). Glutathione levels were lower in both cisplatin+saline and cisplatin+vitamin C groups than control group, but vitamin C significantly ameliorated the glutathione levels (p<0.05). Sciatic heat shock protein-70 levels were significantly higher in the cisplatin+vitamin C group than cisplatin+saline group. Compound muscle action potential amplitude and inclined plane test scores were significantly improved in the vitamin C group (p<0.05). Axon diameter and nerve growth factor expression ameliorated with vitamin C (p<0.05). CONCLUSIONS: We demonstrated the ameliorated effects of vitamin C on cisplatin-induced neuropathy through increased heat shock protein-70, nerve growth factor levels, and reduced inflammatory and oxidant effects. The results are promising to improve the neurotoxic effects of cisplatin in cancer patients.
Collapse
Affiliation(s)
- Emel Ebru Pala
- University of Health Sciences, Tepecik Health Practice & Research Center, Department of Pathology - Izmir, Turkey
| | - Halil Gursoy Pala
- University of Health Sciences, Tepecik Health Practice & Research Center, Department of Obstetrics and Gynecology - Izmir, Turkey
| | - Sumeyye Ekmekci
- University of Health Sciences, Tepecik Health Practice & Research Center, Department of Pathology - Izmir, Turkey
| | - Oytun Erbas
- Bilim University Medical Faculty, Department of Physiology - Istanbul, Turkey
| |
Collapse
|
5
|
Targeting Glioblastoma via Selective Alteration of Mitochondrial Redox State. Cancers (Basel) 2022; 14:cancers14030485. [PMID: 35158753 PMCID: PMC8833725 DOI: 10.3390/cancers14030485] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 01/06/2022] [Accepted: 01/11/2022] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Glioblastoma is characterized by a pronounced redox imbalance due to elevated glycolytic and mitochondrial oxidative metabolism. New therapeutic strategies have been developed to modulate glioblastoma redox signaling to effectively suppress growth and prolong survival. However, drug selectivity and therapeutic relapse prove to be the major challenges. We describe a pharmacological strategy for the selective targeting and treatment of glioblastoma using the redox active combination drug menadione/ascorbate, which is characterized by tolerance to normal cells and tissues. Menadione/ascorbate treatment of glioblastoma mice suppressed tumor growth and significantly increased survival without adverse side effects. This is accompanied by increased oxidative stress, decreased reducing capacity and decreased cellular density in the tumor alone, as well as increased brain perfusion and decreased regulation of several oncoproteins and oncometabolites, which implies modulation of the immune response and reduced drug resistance. We believe that this therapeutic strategy is feasible and promising and deserves the attention of clinicians. Abstract Glioblastoma is one of the most aggressive brain tumors, characterized by a pronounced redox imbalance, expressed in a high oxidative capacity of cancer cells due to their elevated glycolytic and mitochondrial oxidative metabolism. The assessment and modulation of the redox state of glioblastoma are crucial factors that can provide highly specific targeting and treatment. Our study describes a pharmacological strategy for targeting glioblastoma using a redox-active combination drug. The experiments were conducted in vivo on glioblastoma mice (intracranial model) and in vitro on cell lines (cancer and normal) treated with the redox cycling pair menadione/ascorbate (M/A). The following parameters were analyzed in vivo using MRI or ex vivo on tissue and blood specimens: tumor growth, survival, cerebral perfusion, cellular density, tissue redox state, expression of tumor-associated NADH oxidase (tNOX) and transforming growth factor-beta 1 (TGF-β1). Dose-dependent effects of M/A on cell viability, mitochondrial functionality, and redox homeostasis were evaluated in vitro. M/A treatment suppressed tumor growth and significantly increased survival without adverse side effects. This was accompanied by increased oxidative stress, decreased reducing capacity, and decreased cellular density in the tumor only, as well as increased cerebral perfusion and down-regulation of tNOX and TGF-β1. M/A induced selective cytotoxicity and overproduction of mitochondrial superoxide in isolated glioblastoma cells, but not in normal microglial cells. This was accompanied by a significant decrease in the over-reduced state of cancer cells and impairment of their “pro-oncogenic” functionality, assessed by dose-dependent decreases in: NADH, NAD+, succinate, glutathione, cellular reducing capacity, mitochondrial potential, steady-state ATP, and tNOX expression. The safety of M/A on normal cells was compromised by treatment with cerivastatin, a non-specific prenyltransferase inhibitor. In conclusion, M/A differentiates glioblastoma cells and tissues from normal cells and tissues by redox targeting, causing severe oxidative stress only in the tumor. The mechanism is complex and most likely involves prenylation of menadione in normal cells, but not in cancer cells, modulation of the immune response, a decrease in drug resistance, and a potential role in sensitizing glioblastoma to conventional chemotherapy.
Collapse
|
6
|
Li Y, Wang Y, Wang X, Jin L, Yang L, Zhu J, Wang H, Zheng F, Cui H, Li X, Jia Y. Evodiamine suppresses the progression of non-small cell lung carcinoma via endoplasmic reticulum stress-mediated apoptosis pathway in vivo and in vitro. Int J Immunopathol Pharmacol 2022; 36:3946320221086079. [PMID: 35388733 PMCID: PMC9003648 DOI: 10.1177/03946320221086079] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Evodiamine (EVO) is one of the major components isolated from Evodia rutaecarpa (Juss.). Recent studies have shown that EVO has an anti-cancer effect. However, the pharmacological mechanism by which EVO impacts cancer is still poorly understood. OBJECTIVES This study focused on asking the anti-cancer effect of EVO in human non-small cell lung carcinoma (NSCLC), and in particular to investigate whether EVO acts via modulating the endoplasmic reticulum stress (ERS)-mediated apoptosis pathway. MATERIALS AND METHODS A Lewis lung carcinoma (LLC) tumor-bearing mouse model was treated with low-dose EVO (5 mg/kg) and high-dose EVO (10 mg/kg) intraperitoneally for 14 d. The effects of EVO on tumor growth, apoptosis, and ERS were assessed. In addition, NSCLC A549 and LLC cells were treated with EVO in vitro. The effects of EVO on cell proliferation, apoptosis, and ERS were investigated. Finally, 4-phenylbutyric acid (4-PBA), an ERS inhibitor, was used to validate whether EVO induced apoptosis of NSCLC cells by modulating ERS. RESULTS EVO treatment significantly inhibited tumor growth in LLC tumor-bearing mice. H&E staining indicated that EVO treatment reduced the number of tumor cells and the nucleo-plasmic ratio. Immunostaining showed that EVO treatment significantly decreased the expression of Ki-67. TUNEL staining revealed that EVO induced apoptosis in the tumor. Likewise, EVO treatment up-regulated the expression of apoptosis-related genes and proteins and increased activation of the ERS pathway in the tumor. Additionally, EVO inhibited cell proliferation and increased cell apoptotic rates in A549 and LLC cells. EVO also increased the expression levels of genes and proteins associated with ERS-mediated apoptosis pathway in vitro. The effects of EVO on apoptosis were abolished by 4-PBA treatment. CONCLUSIONS Our study demonstrated that EVO suppresses the progression of NSCLC by modulating the ERS-mediated apoptosis pathway.
Collapse
Affiliation(s)
- Yuting Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.,74770National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China.,Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuming Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiaoqun Wang
- 74770National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China.,Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lulu Jin
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lu Yang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jinli Zhu
- 74770National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China.,Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Hongwu Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Fang Zheng
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Huantian Cui
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, 12589Shandong University, Qingdao, China
| | - Xiaojiang Li
- 74770National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China.,Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yingjie Jia
- 74770National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China.,Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
7
|
Böttger F, Vallés-Martí A, Cahn L, Jimenez CR. High-dose intravenous vitamin C, a promising multi-targeting agent in the treatment of cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:343. [PMID: 34717701 PMCID: PMC8557029 DOI: 10.1186/s13046-021-02134-y] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 10/07/2021] [Indexed: 12/21/2022]
Abstract
Mounting evidence indicates that vitamin C has the potential to be a potent anti-cancer agent when administered intravenously and in high doses (high-dose IVC). Early phase clinical trials have confirmed safety and indicated efficacy of IVC in eradicating tumour cells of various cancer types. In recent years, the multi-targeting effects of vitamin C were unravelled, demonstrating a role as cancer-specific, pro-oxidative cytotoxic agent, anti-cancer epigenetic regulator and immune modulator, reversing epithelial-to-mesenchymal transition, inhibiting hypoxia and oncogenic kinase signalling and boosting immune response. Moreover, high-dose IVC is powerful as an adjuvant treatment for cancer, acting synergistically with many standard (chemo-) therapies, as well as a method for mitigating the toxic side-effects of chemotherapy. Despite the rationale and ample evidence, strong clinical data and phase III studies are lacking. Therefore, there is a need for more extensive awareness of the use of this highly promising, non-toxic cancer treatment in the clinical setting. In this review, we provide an elaborate overview of pre-clinical and clinical studies using high-dose IVC as anti-cancer agent, as well as a detailed evaluation of the main known molecular mechanisms involved. A special focus is put on global molecular profiling studies in this respect. In addition, an outlook on future implications of high-dose vitamin C in cancer treatment is presented and recommendations for further research are discussed.
Collapse
Affiliation(s)
- Franziska Böttger
- Department of Medical Oncology, Cancer Center Amsterdam, OncoProteomics Laboratory, Amsterdam UMC, Location VU University Medical Center, 1081 HV, Amsterdam, the Netherlands
| | - Andrea Vallés-Martí
- Department of Medical Oncology, Cancer Center Amsterdam, OncoProteomics Laboratory, Amsterdam UMC, Location VU University Medical Center, 1081 HV, Amsterdam, the Netherlands
| | - Loraine Cahn
- Department of Medical Oncology, Cancer Center Amsterdam, OncoProteomics Laboratory, Amsterdam UMC, Location VU University Medical Center, 1081 HV, Amsterdam, the Netherlands
| | - Connie R Jimenez
- Department of Medical Oncology, Cancer Center Amsterdam, OncoProteomics Laboratory, Amsterdam UMC, Location VU University Medical Center, 1081 HV, Amsterdam, the Netherlands.
| |
Collapse
|
8
|
Selective Targeting of Cancerous Mitochondria and Suppression of Tumor Growth Using Redox-Active Treatment Adjuvant. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:6212935. [PMID: 33204397 PMCID: PMC7652615 DOI: 10.1155/2020/6212935] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 06/13/2020] [Accepted: 07/13/2020] [Indexed: 12/18/2022]
Abstract
Redox-active substances and their combinations, such as of quinone/ascorbate and in particular menadione/ascorbate (M/A; also named Apatone®), attract attention with their unusual ability to kill cancer cells without affecting the viability of normal cells as well as with the synergistic anticancer effect of both molecules. So far, the primary mechanism of M/A-mediated anticancer effects has not been linked to the mitochondria. The aim of our study was to clarify whether this “combination drug” affects mitochondrial functionality specifically in cancer cells. Studies were conducted on cancer cells (Jurkat, Colon26, and MCF7) and normal cells (normal lymphocytes, FHC, and MCF10A), treated with different concentrations of menadione, ascorbate, and/or their combination (2/200, 3/300, 5/500, 10/1000, and 20/2000 μM/μM of M/A). M/A exhibited highly specific and synergistic suppression on cancer cell growth but without adversely affecting the viability of normal cells at pharmacologically attainable concentrations. In M/A-treated cancer cells, the cytostatic/cytotoxic effect is accompanied by (i) extremely high production of mitochondrial superoxide (up to 15-fold over the control level), (ii) a significant decrease of mitochondrial membrane potential, (iii) a decrease of the steady-state levels of ATP, succinate, NADH, and NAD+, and (iv) a decreased expression of programed cell death ligand 1 (PD-L1)—one of the major immune checkpoints. These effects were dose dependent. The inhibition of NQO1 by dicoumarol increased mitochondrial superoxide and sensitized cancer cells to M/A. In normal cells, M/A induced relatively low and dose-independent increase of mitochondrial superoxide and mild oxidative stress, which seems to be well tolerated. These data suggest that all anticancer effects of M/A result from a specific mechanism, tightly connected to the mitochondria of cancer cells. At low/tolerable doses of M/A (1/100-3/300 μM/μM) attainable in cancer by oral and parenteral administration, M/A sensitized cancer cells to conventional anticancer drugs, exhibiting synergistic or additive cytotoxicity accompanied by impressive induction of apoptosis. Combinations of M/A with 13 anticancer drugs were investigated (ABT-737, barasertib, bleomycin, BEZ-235, bortezomib, cisplatin, everolimus, lomustine, lonafarnib, MG-132, MLN-2238, palbociclib, and PI-103). Low/tolerable doses of M/A did not induce irreversible cytotoxicity in cancer cells but did cause irreversible metabolic changes, including: (i) a decrease of succinate and NADH, (ii) depolarization of the mitochondrial membrane, and (iii) overproduction of superoxide in the mitochondria of cancer cells only. In addition, M/A suppressed tumor growth in vivo after oral administration in mice with melanoma and the drug downregulated PD-L1 in melanoma cells. Experimental data suggest a great potential for beneficial anticancer effects of M/A through increasing the sensitivity of cancer cells to conventional anticancer therapy, as well as to the immune system, while sparing normal cells. We hypothesize that M/A-mediated anticancer effects are triggered by redox cycling of both substances, specifically within dysfunctional mitochondria. M/A may also have a beneficial effect on the immune system, making cancer cells “visible” and more vulnerable to the native immune response.
Collapse
|
9
|
Belanova A, Beseda D, Chmykhalo V, Stepanova A, Belousova M, Khrenkova V, Gavalas N, Zolotukhin P. Berberine Effects on NFκB, HIF1A and NFE2L2/AP-1 Pathways in HeLa Cells. Anticancer Agents Med Chem 2019; 19:487-501. [DOI: 10.2174/1871520619666181211121405] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 11/19/2018] [Accepted: 11/28/2018] [Indexed: 12/17/2022]
Abstract
Background:
Berberine has multitudinous anti-cancer stem cells effects making it a highly promising
candidate substance for the next-generation cancer therapy. However, berberine modes of action predispose it to
significant side-effects that probably limit its clinical testing and application.
Materials and Methods:
HeLa cells were treated with two concentrations of berberine (30 and 100 µM) for 24
hours to assess the functioning of the NFE2L2/AP-1, NFκB and HIF1A pathways using 22 RNAs expression
qPCR-based analysis.
Results:
Berberine effects appeared to be highly dose-dependent, with the lower concentration being capable of
suppressing the NFκB functioning and the higher concentration causing severe signaling side-effects seen in the
HIF1A pathway and the NFE2L2 sub-pathways, and especially and more importantly in the AP-1 sub-pathway.
Conclusion:
The results of the study suggest that berberine has clinically valuable anti-NFκB effects however
jeopardized by its side effects on the HIF1A and especially NFE2L2/AP-1 pathways, its therapeutic window
phenomenon and its cancer type-specificity. These, however, may be ameliorated using the cocktail approach,
provided there is enough data on signaling effects of berberine.
Collapse
Affiliation(s)
- Anna Belanova
- Biomedical Innovations LLC, 112 Mechnikova st., 344013, Rostov-on-Don, Russian Federation
| | - Darya Beseda
- Biomedical Innovations LLC, 112 Mechnikova st., 344013, Rostov-on-Don, Russian Federation
| | - Victor Chmykhalo
- Biomedical Innovations LLC, 112 Mechnikova st., 344013, Rostov-on-Don, Russian Federation
| | - Alisa Stepanova
- Biomedical Innovations LLC, 112 Mechnikova st., 344013, Rostov-on-Don, Russian Federation
| | - Mariya Belousova
- English Language Department for Natural Sciences Faculties, Southern Federal University, 5 Sorge st., 344090, Rostov-on-Don, Russian Federation
| | - Vera Khrenkova
- Rostov State Medical University, 119 Suvorova st., 344022, Rostov-on-Don, Russian Federation
| | - Nikolaos Gavalas
- Division of Clinical Therapeutics, National and Kapodistrian University of Athens, 80 Vas. Sofias Av., 11521, Athens, Greece
| | - Peter Zolotukhin
- Biomedical Innovations LLC, 112 Mechnikova st., 344013, Rostov-on-Don, Russian Federation
| |
Collapse
|
10
|
Chao W, Deng JS, Li PY, Kuo YH, Huang GJ. Inotilone from Inonotus linteus suppresses lung cancer metastasis in vitro and in vivo through ROS-mediated PI3K/AKT/MAPK signaling pathways. Sci Rep 2019; 9:2344. [PMID: 30787353 PMCID: PMC6382761 DOI: 10.1038/s41598-019-38959-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 01/07/2019] [Indexed: 12/11/2022] Open
Abstract
Metastasis is one of the main causes of mortality in cancer patients. Inotilone, a major component of Inonotus linteus, is a traditional Chinese medical herb. In this study, MTT results showed that inotilone had no obvious cytotoxicity. Animal model results revealed that inotilone suppressed cancer metastatic efficacy. Serum results showed that inotilone reduced the activity of matrix metalloproteinase (MMP)-2 and -9 and tumor necrosis factor alpha (TNF-α) activity as well as NO content. Additionally, inotilone affected MMP-9 and tissue inhibitor of metalloproteinase (TIMP)-2 protein expression and improved the activity of the antioxidant enzymes in the lung tissues of LLC-bearing mice. In addition, cell experimental results showed that inotilone reduced the activity of MMP-2/-9 and inhibited the ability for cellular migration and invasion. Inotilone decreased interleukin (IL)-8 expression in A549 cells. Western blot results revealed that inotilone affected the protein expression of MMPs, nitric oxide synthase (iNOS), cyclooxygenase (COX)-2, anti-oxidant enzymes, mitogen activated protein kinase (MAPK), focal adhesion kinase (FAK), phosphoinositide-3 kinase (PI3K)-AKT, and nuclear factor (NF)κB. Therefore, we propose that inotilone is a potential therapeutic candidate against metastatic lung cancer cells.
Collapse
Affiliation(s)
- Wei Chao
- School of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, College of Chinese Medicine, China Medical University, Taichung, 404, Taiwan.,Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Jeng-Shyan Deng
- Department of Health and Nutrition Biotechnology, College of Medical and Health Science, Asia University, Taichung, 413, Taiwan
| | - Pei-Ying Li
- School of Pharmacy, College of Pharmacy, China Medical University, Taichung, 404, Taiwan
| | - Yueh-Hsiung Kuo
- School of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, College of Chinese Medicine, China Medical University, Taichung, 404, Taiwan
| | - Guan-Jhong Huang
- School of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, College of Chinese Medicine, China Medical University, Taichung, 404, Taiwan.
| |
Collapse
|
11
|
Chao W, Deng JS, Li PY, Liang YC, Huang GJ. 3,4-Dihydroxybenzalactone Suppresses Human Non-Small Cell Lung Carcinoma Cells Metastasis via Suppression of Epithelial to Mesenchymal Transition, ROS-Mediated PI3K/AKT/MAPK/MMP and NFκB Signaling Pathways. Molecules 2017; 22:molecules22040537. [PMID: 28350337 PMCID: PMC6154291 DOI: 10.3390/molecules22040537] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 03/17/2017] [Accepted: 03/20/2017] [Indexed: 12/19/2022] Open
Abstract
3,4-Dihydroxybenzalactone (DBL) was isolated from Phellinus linteus (PL), which is a folk medicine possessing various physiological effects. In this study, we used highly metastatic A549 cells to investigate efficacy of DBL inhibition of cancer metastasis and possible mechanisms. The results revealed DBL inhibited migratory and invasive abilities of cancer cells at noncytotoxic concentrations. We found DBL suppressed enzymatic activities, protein expression, and RNA levels of matrix metalloproteinase (MMP)-2 and MMP-9. Western blot results showed DBL decreased phosphoinositide 3-kinase (PI3K)/AKT, phosphorylation status of mitogen-activated protein kinases (MAPKs), and focal adhesion kinase (FAK)/paxillin, which correlated with cell migratory ability. DBL also affected epithelial to mesenchymal transition (EMT)-related biomarkers. In addition, DBL enhanced cytoprotective effects through elevated antioxidant enzymes including heme oxygenase 1 (HO-1), catalase, glutathione peroxidase (GPx), and superoxide dismutase (SOD). Moreover, DBL influenced the nuclear translocation of nuclear factor κB (NFκB), nuclear factor erythroid 2-related factor 2 (Nrf2), Snail, and Slug in A549 cells. Taken together, these results suggested that treatment with DBL may act as a potential candidate to inhibit lung cancer metastasis by inhibiting MMP-2 and -9 via affecting PI3K/AKT, MAPKs, FAK/paxillin, EMT/Snail and Slug, Nrf2/antioxidant enzymes, and NFκB signaling pathways.
Collapse
Affiliation(s)
- Wei Chao
- School of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, College of Chinese Medicine, China Medical University, Taichung 404, Taiwan.
| | - Jeng-Shyan Deng
- Department of Health and Nutrition Biotechnology, Asia University, Taichung 404, Taiwan.
| | - Pei-Ying Li
- School of Pharmacy, College of Pharmacy, China Medical University, Taichung 404, Taiwan.
| | - Yu-Chia Liang
- School of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, College of Chinese Medicine, China Medical University, Taichung 404, Taiwan.
| | - Guan-Jhong Huang
- School of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, College of Chinese Medicine, China Medical University, Taichung 404, Taiwan.
| |
Collapse
|
12
|
Ding F, Wang M, Du Y, Du S, Zhu Z, Yan Z. BHX Inhibits the Wnt Signaling Pathway by Suppressing β-catenin Transcription in the Nucleus. Sci Rep 2016; 6:38331. [PMID: 27910912 PMCID: PMC5133598 DOI: 10.1038/srep38331] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 11/08/2016] [Indexed: 11/25/2022] Open
Abstract
BHX (N-(4-hydroxybenzyl)-1,3,4-triphenyl-4,5-dihydro-1H-pyrazole-5-carboxamide), a Wnt signaling pathway inhibitor, effectively inhibits tumor cell growth, but the underlying mechanism is unclear. Thus, we aim to investigate the effects and associated mechanism of BHX action on A549 and MCF-7 cell lines. In our study, MTT(3-[4,5-dimethyl-2-thiazolyl]-2,5-diphenyl-2H-tetrazolium bromide) and xenograft model assay indicated that cell growth was inhibited by BHX at a range of concentrations in vitro and in vivo. The expression of β-catenin and Wnt signaling pathway downstream target genes were decreased evidently under BHX treatment. Flow cytometry also revealed that BHX treatment significantly induced G1 arrest. Further analysis showed that BHX lowered the transcriptional level of β-catenin. In conclusion, BHX inhibited the nuclear synthesis of β-catenin, thereby suppressing the Wnt signaling pathway and further inhibiting tumor growth and proliferation.
Collapse
Affiliation(s)
- Fengxia Ding
- Department of Clinical Pharmacology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, P. R. China
| | - Meisa Wang
- Department of Clinical Pharmacology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, P. R. China
| | - Yibo Du
- Department of Clinical Pharmacology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, P. R. China
| | - Shuangshuang Du
- Department of Clinical Pharmacology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, P. R. China
| | - Zhongling Zhu
- Department of Clinical Pharmacology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, P. R. China
| | - Zhao Yan
- Department of Clinical Pharmacology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, P. R. China
| |
Collapse
|
13
|
Ding C, Fan X, Wu G. Peroxiredoxin 1 - an antioxidant enzyme in cancer. J Cell Mol Med 2016; 21:193-202. [PMID: 27653015 PMCID: PMC5192802 DOI: 10.1111/jcmm.12955] [Citation(s) in RCA: 143] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 07/17/2016] [Indexed: 12/11/2022] Open
Abstract
Peroxiredoxins (PRDXs), a ubiquitous family of redox‐regulating proteins, are reported of potential to eliminate various reactive oxygen species (ROS). As a major member of the antioxidant enzymes, PRDX1 can become easily over‐oxidized on its catalytically active cysteine induced by a variety of stimuli in vitro and in vivo. In nucleus, oligomeric PRDX1 directly associates with p53 or transcription factors such as c‐Myc, NF‐κB and AR, and thus affects their bioactivities upon gene regulation, which in turn induces or suppresses cell death. Additionally, PRDX1 in cytoplasm has anti‐apoptotic potential through direct or indirect interactions with several ROS‐dependent (redox regulation) effectors, including ASK1, p66Shc, GSTpi/JNK and c‐Abl kinase. PRDX1 is proven to be a versatile molecule regulating cell growth, differentiation and apoptosis. Recent studies have found that PRDX1 and/or PRDX1‐regulated ROS‐dependent signalling pathways play an important role in the progression and metastasis of human tumours, particularly in breast, oesophageal and lung cancers. In this paper, we review the structure, effector functions of PRDX1, its role in cancer and the pivotal role of ROS in anticancer treatment.
Collapse
Affiliation(s)
- Chenbo Ding
- Medical School of Southeast University, Nanjing, China
| | - Xiaobo Fan
- Medical School of Southeast University, Nanjing, China
| | - Guoqiu Wu
- Medical School of Southeast University, Nanjing, China.,Center of Clinical Laboratory Medicine, Zhongda Hospital, Southeast University, Nanjing, China
| |
Collapse
|
14
|
Gröber U, Holzhauer P, Kisters K, Holick MF, Adamietz IA. Micronutrients in Oncological Intervention. Nutrients 2016; 8:163. [PMID: 26985904 PMCID: PMC4808891 DOI: 10.3390/nu8030163] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 02/16/2016] [Accepted: 02/24/2016] [Indexed: 12/14/2022] Open
Abstract
Nutritional supplements are widely used among patients with cancer who perceive them to be anticancer and antitoxicity agents. Depending on the type of malignancy and the gender 30%-90% of the cancer patients supplement their diets with antioxidant and immuno-stabilizing micronutrients, such as selenium, vitamin C, and vitamin D, often without the knowledge of the treating physician. From the oncological viewpoint, there are justifiable concerns that dietary supplements decrease the effectiveness of chemotherapy and radiotherapy. Recent studies, however, have provided increasing evidence that treatment is tolerated better-with an increase in patient compliance and a lower rate of treatment discontinuations-when micronutrients, such as selenium, are added as appropriate to the patient's medication. Nutritional supplementation tailored to an individual's background diet, genetics, tumor histology, and treatments may yield benefits in subsets of patients. Clinicians should have an open dialogue with patients about nutritional supplements. Supplement advice needs to be individualized and come from a credible source, and it is best communicated by the physician.
Collapse
Affiliation(s)
- Uwe Gröber
- Akademie für Mikronährstoffmedizin, Essen, Zweigertstrasse 55, 45130 Essen, Germany.
| | - Peter Holzhauer
- Akademie für Mikronährstoffmedizin, Essen, Zweigertstrasse 55, 45130 Essen, Germany.
- Interdisziplinäres onkologisches Zentrum (IOZ), München, Nußbaumstrasse 12, München 80336, Germany.
- Klinik Bad Trissl, Innere Medizin II-Onkologie und Komplementärmedizin, Oberaudorf 83080, Germany.
| | - Klaus Kisters
- Akademie für Mikronährstoffmedizin, Essen, Zweigertstrasse 55, 45130 Essen, Germany.
- St. Anna Hospital, Medizinische Klinik I, Herne, Hospitalstrasse 19, Herne 44649, Germany.
| | - Michael F Holick
- Boston University Medical Center, 85 East Newton Street M-1033, Boston, MA 02118, USA.
| | - Irenäus A Adamietz
- Klinik für Strahlentherapie und Radio-Onkologie, Ruhr Universität Bochum (RUB), Hölkeskampring 40, Herne 44625, Germany.
| |
Collapse
|
15
|
Kumar A, Chelvam V, Sakkarapalayam M, Li G, Sanchez-Cruz P, Piñero NS, Low PS, Alegria AE. Synthesis and Evaluation of Folate-Conjugated Phenanthraquinones for Tumor-Targeted Oxidative Chemotherapy. ACTA ACUST UNITED AC 2016; 6:1-17. [PMID: 27066312 PMCID: PMC4825697 DOI: 10.4236/ojmc.2016.61001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Almost all cells are easily killed by exposure to potent oxidants. Indeed, major pathogen defense mechanisms in both animal and plant kingdoms involve production of an oxidative burst, where host defense cells show an invading pathogen with reactive oxygen species (ROS). Although cancer cells can be similarly killed by ROS, development of oxidant-producing chemotherapies has been limited by their inherent nonspecificity and potential toxicity to healthy cells. In this paper, we describe the targeting of an ROS-generating molecule selectively to tumor cells using folate as the tumor-targeting ligand. For this purpose, we exploit the ability of 9,10-phenanthraquinone (PHQ) to enhance the continuous generation of H2O2 in the presence of ascorbic acid to establish a constitutive source of ROS within the tumor mass. We report here that incubation of folate receptor-expressing KB cells in culture with folate-PHQ plus ascorbate results in the death of the cancer cells with an IC50 of ~10 nM (folate-PHQ). We also demonstrate that a cleavable spacer linking folate to PHQ is significantly inferior to a noncleavable spacer, in contrast to most other folate-targeted therapeutic agents. Unfortunately, no evidence for folate-PHQ mediated tumor regression in murine tumor models is obtained, suggesting that unanticipated impediments to generation of cytotoxic quantities of ROS in vivo are encountered. Possible mechanisms and potential solutions to these unanticipated results are offered.
Collapse
Affiliation(s)
- Ajay Kumar
- International Center for Trans-disciplinary Research, School of Environmental Affairs, Universidad Metropolitana, San Juan, Puerto Rico ; Department of Chemistry, University of Puerto Rico, Humacao, Puerto Rico
| | - Venkatesh Chelvam
- Department of Chemistry, Purdue University, West Lafayette, Indiana ; Department of Chemistry, Centre for Biosciences and Biomedical Engineering, Indian Institute of Technology, Indore, Simrol Campus, Madhya Pradesh, India
| | | | - Guo Li
- Department of Chemistry, Purdue University, West Lafayette, Indiana
| | - Pedro Sanchez-Cruz
- Department of Chemistry, University of Puerto Rico, Humacao, Puerto Rico
| | - Natasha S Piñero
- Department of Chemistry, University of Puerto Rico, Humacao, Puerto Rico
| | - Philip S Low
- Department of Chemistry, Purdue University, West Lafayette, Indiana
| | - Antonio E Alegria
- Department of Chemistry, University of Puerto Rico, Humacao, Puerto Rico
| |
Collapse
|
16
|
Zhang ZW, Xu XC, Liu T, Yuan S. Mitochondrion-Permeable Antioxidants to Treat ROS-Burst-Mediated Acute Diseases. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2016:6859523. [PMID: 26649144 PMCID: PMC4663357 DOI: 10.1155/2016/6859523] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Revised: 07/09/2015] [Accepted: 07/14/2015] [Indexed: 02/03/2023]
Abstract
Reactive oxygen species (ROS) play a crucial role in the inflammatory response and cytokine outbreak, such as during virus infections, diabetes, cancer, cardiovascular diseases, and neurodegenerative diseases. Therefore, antioxidant is an important medicine to ROS-related diseases. For example, ascorbic acid (vitamin C, VC) was suggested as the candidate antioxidant to treat multiple diseases. However, long-term use of high-dose VC causes many side effects. In this review, we compare and analyze all kinds of mitochondrion-permeable antioxidants, including edaravone, idebenone, α-Lipoic acid, carotenoids, vitamin E, and coenzyme Q10, and mitochondria-targeted antioxidants MitoQ and SkQ and propose astaxanthin (a special carotenoid) to be the best antioxidant for ROS-burst-mediated acute diseases, like avian influenza infection and ischemia-reperfusion. Nevertheless, astaxanthins are so unstable that most of them are inactivated after oral administration. Therefore, astaxanthin injection is suggested hypothetically. The drawbacks of the antioxidants are also reviewed, which limit the use of antioxidants as coadjuvants in the treatment of ROS-associated disorders.
Collapse
Affiliation(s)
- Zhong-Wei Zhang
- College of Resources Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Xiao-Chao Xu
- College of Bioindustry, Chengdu University, Chengdu 610106, China
| | - Ting Liu
- Sichuan Kelun Pharmaceutical Co. Ltd., Chengdu 610071, China
| | - Shu Yuan
- College of Resources Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| |
Collapse
|
17
|
Campbell EJ, Dachs GU. Current limitations of murine models in oncology for ascorbate research. Front Oncol 2014; 4:282. [PMID: 25353008 PMCID: PMC4196513 DOI: 10.3389/fonc.2014.00282] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Accepted: 09/29/2014] [Indexed: 12/15/2022] Open
Abstract
The role of vitamin C (ascorbate) in cancer prevention, tumor growth, and treatment is of intense public interest. Clinical trial data have been sparse, contradictory, and highly controversial, and robust pre-clinical data are required for progress. This paper reviews pre-clinical models and their limitations with respect to ascorbate research. Most studies have utilized animals able to synthesize ascorbate and thus are not ideal models of the human condition. More recently, genetically modified mouse models have become available; yet, all studies compared healthy and scorbutic mice. The majority of investigations to date concluded that increased ascorbate led to decreased tumor growth, but data on mechanisms and doses are inconclusive. Clinically relevant animal studies are still required to convince a generally sceptical medical audience of the potential worth of ascorbate as an adjunct to therapy.
Collapse
Affiliation(s)
- Elizabeth J Campbell
- Mackenzie Cancer Research Group, Department of Pathology, University of Otago , Christchurch , New Zealand
| | - Gabi U Dachs
- Mackenzie Cancer Research Group, Department of Pathology, University of Otago , Christchurch , New Zealand
| |
Collapse
|
18
|
Xu F, Vostal JG. Inactivation of bacteria via photosensitization of vitamin K3 by UV-A light. FEMS Microbiol Lett 2014; 358:98-105. [DOI: 10.1111/1574-6968.12544] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Revised: 07/17/2014] [Accepted: 07/17/2014] [Indexed: 11/28/2022] Open
Affiliation(s)
- Fei Xu
- Laboratory of Cellular Hematology; CBER; FDA; Bethesda MD USA
| | | |
Collapse
|
19
|
Chen MF, Yang CM, Su CM, Hu ML. Vitamin C Protects Against Cisplatin-Induced Nephrotoxicity and Damage Without Reducing Its Effectiveness in C57BL/6 Mice Xenografted With Lewis Lung Carcinoma. Nutr Cancer 2014; 66:1085-91. [DOI: 10.1080/01635581.2014.948211] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
20
|
Menadione induces the formation of reactive oxygen species and depletion of GSH-mediated apoptosis and inhibits the FAK-mediated cell invasion. Naunyn Schmiedebergs Arch Pharmacol 2014; 387:799-809. [DOI: 10.1007/s00210-014-0997-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Accepted: 05/22/2014] [Indexed: 12/27/2022]
|
21
|
Parrow NL, Leshin JA, Levine M. Parenteral ascorbate as a cancer therapeutic: a reassessment based on pharmacokinetics. Antioxid Redox Signal 2013; 19:2141-56. [PMID: 23621620 PMCID: PMC3869468 DOI: 10.1089/ars.2013.5372] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
SIGNIFICANCE Ewan Cameron reported that ascorbate, given orally and intravenously at doses of up to 10 g/day, was effective in the treatment of cancer. Double-blind placebo-controlled clinical trials showed no survival advantage when the same doses of ascorbate were given orally, leading the medical and scientific communities to dismiss the use of ascorbate as a potential cancer treatment. However, the route of administration results in major differences in ascorbate bioavailability. Tissue and plasma concentrations are tightly controlled in response to oral administration, but this can be bypassed by intravenous administration. These data provide a plausible scientific rationale for the absence of a response to orally administered ascorbate in the Mayo clinic trials and indicate the need to reassess ascorbate as a cancer therapeutic. RECENT ADVANCES High dose ascorbate is selectively cytotoxic to cancer cell lines through the generation of extracellular hydrogen peroxide (H2O2). Murine xenograft models confirm a growth inhibitory effect of pharmacological concentrations. The safety of intravenous ascorbate has been verified in encouraging pilot clinical studies. CRITICAL ISSUES Neither the selective toxicity of pharmacologic ascorbate against cancer cells nor the mechanism of H2O2-mediated cytotoxicity is fully understood. Despite promising preclinical data, the question of clinical efficacy remains. FUTURE DIRECTIONS A full delineation of mechanism is of interest because it may indicate susceptible cancer types. Effects of pharmacologic ascorbate used in combination with standard treatments need to be defined. Most importantly, the clinical efficacy of ascorbate needs to be reassessed using proper dosing, route of administration, and controls.
Collapse
Affiliation(s)
- Nermi L Parrow
- Molecular and Clinical Nutrition Section, Digestive Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health , Bethesda, Maryland
| | | | | |
Collapse
|