1
|
Cunha N, Andrade V, Macedo A, Ruivo P, Lima G. Methods of Protein Extraction from House Crickets ( Acheta domesticus) for Food Purposes. Foods 2025; 14:1164. [PMID: 40238308 PMCID: PMC11989044 DOI: 10.3390/foods14071164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Revised: 03/24/2025] [Accepted: 03/25/2025] [Indexed: 04/18/2025] Open
Abstract
Global population is projected to reach 9.1 billion by 2050, emphasizing the need for increased food production. Edible insects, such as house crickets (Acheta domesticus), emerged as promising due to higher nutritional value and efficient feed conversion rates compared to conventional protein sources. Incorporating insect powders into new food products can improve consumer acceptance but often leads to poor technological food processing functionality and/or undesirable organoleptic characteristics. Protein isolates have proven to be effective in enhancing this functionality and consumer acceptance, but existent protein extraction methods still lack improvements concerning the optimization of protein extraction rates. This study aimed to address this gap by developing and comparing the yield of three different protein extraction methods using sodium hydroxide, ascorbic acid or alcalase from house crickets (Acheta domesticus) for food applications. Protein extraction was performed on cricket powder with a mean protein content of 46.35 g/100 g, and the results were evaluated. The enzymatic method shows the highest protein extraction rate at 69.91% with a mean protein content of 60.19 g/100 g, while extraction with NaOH or ascorbic acid resulted in rates of 60.44 and 46.34%, respectively. Further studies on technological food processing functionality and sensorial evaluation of products developed with this protein extract are recommended.
Collapse
Affiliation(s)
- Nair Cunha
- School of Agriculture, Santarem Polytechnic University, Quinta do Galinheiro-S. Pedro, 2001-904 Santarém, Portugal; (N.C.); (P.R.); (G.L.)
- Life Quality Research Centre (CIEQV), Santarem Polytechnic University, Complexo Andaluz, Apartado 279, 2001-904 Santarém, Portugal;
| | - Vanda Andrade
- School of Agriculture, Santarem Polytechnic University, Quinta do Galinheiro-S. Pedro, 2001-904 Santarém, Portugal; (N.C.); (P.R.); (G.L.)
- Life Quality Research Centre (CIEQV), Santarem Polytechnic University, Complexo Andaluz, Apartado 279, 2001-904 Santarém, Portugal;
- Research Centre for Natural Resources, Environment and Society (CERNAS), Santarem Polytechnic University, Quinta do Galinheiro-S. Pedro, 2001-904 Santarém, Portugal
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisboa, Portugal
| | - Antónia Macedo
- Life Quality Research Centre (CIEQV), Santarem Polytechnic University, Complexo Andaluz, Apartado 279, 2001-904 Santarém, Portugal;
- Polytechnic Institute of Beja—Escola Superior Agrária, Rua Pedro Soares, 7800-309 Beja, Portugal
- Mediterranean Institute for Agriculture, Environment and Development (MED), Universidade de Évora, Pólo da Mitra, Apartado 94, 7006-554 Évora, Portugal
- LEAF—Linking Landscape, Environment, Agriculture and Food, Higher Institute of Agronomy, Universidade de Lisboa, Tapada da Ajuda, 1349-017 Lisboa, Portugal
| | - Paula Ruivo
- School of Agriculture, Santarem Polytechnic University, Quinta do Galinheiro-S. Pedro, 2001-904 Santarém, Portugal; (N.C.); (P.R.); (G.L.)
- Life Quality Research Centre (CIEQV), Santarem Polytechnic University, Complexo Andaluz, Apartado 279, 2001-904 Santarém, Portugal;
- Research Centre for Natural Resources, Environment and Society (CERNAS), Santarem Polytechnic University, Quinta do Galinheiro-S. Pedro, 2001-904 Santarém, Portugal
| | - Gabriela Lima
- School of Agriculture, Santarem Polytechnic University, Quinta do Galinheiro-S. Pedro, 2001-904 Santarém, Portugal; (N.C.); (P.R.); (G.L.)
- Life Quality Research Centre (CIEQV), Santarem Polytechnic University, Complexo Andaluz, Apartado 279, 2001-904 Santarém, Portugal;
| |
Collapse
|
2
|
Buljubašić A, Huremović J, Karadža A, Selović A, Gojak-Salimović S. Lemon Juice and Ascorbic Acid Effect on the Metals Extraction from Chamomile, Human Risk Assessment. Biol Trace Elem Res 2025:10.1007/s12011-025-04526-z. [PMID: 39833556 DOI: 10.1007/s12011-025-04526-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 01/15/2025] [Indexed: 01/22/2025]
Abstract
The total content of heavy metals (Cd, Co, Cr, Cu, Fe, Mn, Ni, Pb and Zn) in chamomile sample, metals content in water chamomile infusions and water infusions with additions of ascorbic acid and lemon juice at different temperatures (70, 80, and 100 °C) and steeping times (3, 5, and 7 min), were determined. The content of heavy metals was determined by flame atomic absorption spectrometry (FAAS). Mean total concentrations of Cd, Co, Cr, Cu, Fe, Mn, Ni, Pb and Zn were 0.563, 0.624, 0.254, 8.277, 32.17, 113.5, 5.102, 3.470, 30.34 μg/g, respectively. Mn, Ni and Zn were mostly extracted in pure water, up to 100%, while lemon juice showed a significant role in the extraction of Cd, Fe and Pb. Fe was relatively poor extracted in all infusions, the highest Fe extraction percentages were obtained with the addition of lemon juice (4.07-25.7%), at all applied temperatures. For most metals, the lowest extraction percentages were obtained with the addition of ascorbic acid. Non-carcinogenic hazard quotient (HQ) was less than 1 for analyzed metals in all infusions, for adults and children.
Collapse
Affiliation(s)
- Ajla Buljubašić
- Faculty of Science, Department of Chemistry, University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Jasna Huremović
- Faculty of Science, Department of Chemistry, University of Sarajevo, Sarajevo, Bosnia and Herzegovina.
| | - Amar Karadža
- Faculty of Science, Department of Chemistry, University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Alisa Selović
- Faculty of Science, Department of Chemistry, University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Sabina Gojak-Salimović
- Faculty of Science, Department of Chemistry, University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| |
Collapse
|
3
|
Kontoghiorghes GJ. New Insights into Aspirin's Anticancer Activity: The Predominant Role of Its Iron-Chelating Antioxidant Metabolites. Antioxidants (Basel) 2024; 14:29. [PMID: 39857363 PMCID: PMC11763074 DOI: 10.3390/antiox14010029] [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: 11/12/2024] [Revised: 12/06/2024] [Accepted: 12/24/2024] [Indexed: 01/27/2025] Open
Abstract
Epidemiological studies have suggested that following long-term, low-dose daily aspirin (LTLDA) administration for more than 5 years at 75-100 mg/day, 20-30% of patients (50-80 years old) had a lower risk of developing colorectal cancer (CRC) and about the same proportion in developing iron deficiency anemia (IDA). In cases of IDA, an increase in iron excretion is suspected, which is caused by aspirin chelating metabolites (ACMs): salicylic acid, salicyluric acid, 2,5-dihydroxybenzoic acid, and 2,3-dihydroxybenzoic acid. The ACMs constitute 70% of the administered aspirin dose and have much longer half-lives than aspirin in blood and tissues. The mechanisms of cancer risk reduction in LTLDA users is likely due to the ACM's targeting of iron involved in free radical damage, iron-containing toxins, iron proteins, and associated metabolic pathways such as ferroptosis. The ACMs from non-absorbed aspirin (about 30%) may also mitigate the toxicity of heme and nitroso-heme and other iron toxins from food, which are responsible for the cause of colorectal cancer. The mode of action of aspirin as a chelating antioxidant pro-drug of the ACMs, with continuous presence in LTLDA users, increases the prospect for prophylaxis in cancer and other diseases. It is suggested that the anticancer effects of aspirin depend primarily on the iron-chelating antioxidant activity of the ACMs. The role of aspirin in cancer and other diseases is incomplete without considering its rapid biotransformation and the longer half-life of the ACMs.
Collapse
Affiliation(s)
- George J Kontoghiorghes
- Postgraduate Research Institute of Science, Technology, Environment and Medicine, Limassol 3021, Cyprus
| |
Collapse
|
4
|
Selyutina OY, Ul’yanova MA, Chinak OA, Timoshnikov VA, Fedenok LG, Stepanov AA, Yanshole VV, Kulik LV, Vasilevsky SF, Polyakov NE, Kontoghiorghes GJ. Novel Anthraquinone Derivatives and Their Complexes with Metal Ions with Anticancer Activity: Structure/Redox and Chelation Activity Correlations. Pharmaceuticals (Basel) 2024; 17:1717. [PMID: 39770559 PMCID: PMC11678833 DOI: 10.3390/ph17121717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 12/13/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
Background/Objectives: Some specific anthraquinone derivatives (AQs) are known to be used widely as effective chemotherapeutic agents in the treatment of cancer. However, their fundamental shortcoming is the high rate of cardiotoxicity observed in treated patients, which is thought to be caused by the increase in production of reactive oxygen species (ROS) catalyzed by iron and copper. The development of improved AQs and other anticancer drugs with enhanced efficacy but reduced toxicity remains a high priority. The aim of this study was to evaluate the cytotoxic and ROS production effects of chelate iron and copper complexes of two novel AQs, namely 4-hydroxynaphto[2,3-h]cinnoline-7,12-dione (Q2) and 3-(hydroxymethyl)naphto[2,3-h]cinnoline-4,7,12(1H)-trione (Q3). Methods: The chelation ability of Q2 and Q3 was studied using NMR and UV-Vis spectroscopy. Cytotoxicity studies were carried out using the MTT assay. The influence of chelation on ROS production was studied using NMR spectroscopy in linoleic acid micelles. Results: It was found that only Q3 forms complexes with Fe(III) and Cu(II) ions, whereas Q2 does not demonstrate chelating properties. A cytotoxicity study revealed that Fe[Q3]3 significantly decreased the viability of lung cancer A549 cells, while Q3 and Cu[Q3]2 did not demonstrate cytotoxic properties in this cell line. Furthermore, the presence of Q3 lowered the rate of iron-induced lipid peroxidation in linoleic acid micelles. By contrast, Q2 did not influence the rate of lipid peroxidation, probably due to the absence of effective metal chelating ability. Conclusions: The high cytotoxic effects observed with the iron complex of Q3 against cancer cells in combination with a reduced rate of iron induced lipid peroxidation in the presence of Q3, make Q3 and its iron complex promising for further evaluation and use as chemotherapeutic agents in cancer.
Collapse
Affiliation(s)
- Olga Yu. Selyutina
- Institute of Chemical Kinetics and Combustion, Institutskaya Str. 3, Novosibirsk 630090, Russia; (O.Y.S.); (M.A.U.); (O.A.C.); (V.A.T.); (L.G.F.); (A.A.S.); (L.V.K.); (N.E.P.)
| | - Maya A. Ul’yanova
- Institute of Chemical Kinetics and Combustion, Institutskaya Str. 3, Novosibirsk 630090, Russia; (O.Y.S.); (M.A.U.); (O.A.C.); (V.A.T.); (L.G.F.); (A.A.S.); (L.V.K.); (N.E.P.)
| | - Olga A. Chinak
- Institute of Chemical Kinetics and Combustion, Institutskaya Str. 3, Novosibirsk 630090, Russia; (O.Y.S.); (M.A.U.); (O.A.C.); (V.A.T.); (L.G.F.); (A.A.S.); (L.V.K.); (N.E.P.)
- Institute of Chemical Biology and Fundamental Medicine SB RAS, Lavrentyev Ave. 8, Novosibirsk 630090, Russia
| | - Viktor A. Timoshnikov
- Institute of Chemical Kinetics and Combustion, Institutskaya Str. 3, Novosibirsk 630090, Russia; (O.Y.S.); (M.A.U.); (O.A.C.); (V.A.T.); (L.G.F.); (A.A.S.); (L.V.K.); (N.E.P.)
| | - Lidiya G. Fedenok
- Institute of Chemical Kinetics and Combustion, Institutskaya Str. 3, Novosibirsk 630090, Russia; (O.Y.S.); (M.A.U.); (O.A.C.); (V.A.T.); (L.G.F.); (A.A.S.); (L.V.K.); (N.E.P.)
| | - Alexander A. Stepanov
- Institute of Chemical Kinetics and Combustion, Institutskaya Str. 3, Novosibirsk 630090, Russia; (O.Y.S.); (M.A.U.); (O.A.C.); (V.A.T.); (L.G.F.); (A.A.S.); (L.V.K.); (N.E.P.)
| | - Vadim V. Yanshole
- International Tomography Center SB RAS, Institutskaya Str. 3a, Novosibirsk 630090, Russia;
| | - Leonid V. Kulik
- Institute of Chemical Kinetics and Combustion, Institutskaya Str. 3, Novosibirsk 630090, Russia; (O.Y.S.); (M.A.U.); (O.A.C.); (V.A.T.); (L.G.F.); (A.A.S.); (L.V.K.); (N.E.P.)
| | - Sergey F. Vasilevsky
- Institute of Chemical Kinetics and Combustion, Institutskaya Str. 3, Novosibirsk 630090, Russia; (O.Y.S.); (M.A.U.); (O.A.C.); (V.A.T.); (L.G.F.); (A.A.S.); (L.V.K.); (N.E.P.)
| | - Nikolay E. Polyakov
- Institute of Chemical Kinetics and Combustion, Institutskaya Str. 3, Novosibirsk 630090, Russia; (O.Y.S.); (M.A.U.); (O.A.C.); (V.A.T.); (L.G.F.); (A.A.S.); (L.V.K.); (N.E.P.)
| | - George J. Kontoghiorghes
- Postgraduate Research Institute of Science, Technology, Environment and Medicine, Limassol CY-3021, Cyprus
| |
Collapse
|
5
|
Wu X, Koch M, Martínez FPP, Schirhagl R, Włodarczyk‐Biegun MK. Quantum Sensing Unravels Antioxidant Efficacy Within PCL/Matrigel Skin Equivalents. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2403729. [PMID: 39246220 PMCID: PMC11618742 DOI: 10.1002/smll.202403729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 07/27/2024] [Indexed: 09/10/2024]
Abstract
Skin equivalents (SE) that recapitulate biological and mechanical characteristics of the native tissue are promising platforms for assessing cosmetics and studying fundamental biological processes. Methods to achieve SEs with well-organized structure, and ideal biological and mechanical properties are limited. Here, the combination of melt electrowritten PCL scaffolds and cell-laden Matrigel to fabricate SE is described. The PCL scaffold provides ideal structural and mechanical properties, preventing deformation of the model. The model consists of a top layer for seeding keratinocytes to mimic the epidermis, and a bottom layer of Matrigel-based dermal compartment with fibroblasts. The compressive modulus and the biological properties after 3-day coculture indicate a close resemblance with the native skin. Using the SE, a testing system to study the damage caused by UVA irradiation and evaluate antioxidant efficacy is established. The effectiveness of Tea polyphenols (TPs) and L-ascorbic acid (Laa) is compared based on free radical generation. TPs are demonstrated to be more effective in downregulating free radical generation. Further, T1 relaxometry is used to detect the generation of free radicals at a single-cell level, which allows tracking of the same cell before and after UVA treatment.
Collapse
Affiliation(s)
- Xixi Wu
- Department of Biomedical EngineeringUniversity Medical Centre Groningen and University of GroningenAnt. Deusinglaan 1Groningen9713 AVThe Netherlands
| | - Marcus Koch
- INM – Leibniz Institute for New MaterialsCampus D2 266123SaarbrueckenGermany
| | - Felipe P. Perona Martínez
- Department of Biomedical EngineeringUniversity Medical Centre Groningen and University of GroningenAnt. Deusinglaan 1Groningen9713 AVThe Netherlands
| | - Romana Schirhagl
- Department of Biomedical EngineeringUniversity Medical Centre Groningen and University of GroningenAnt. Deusinglaan 1Groningen9713 AVThe Netherlands
| | - Małgorzata K. Włodarczyk‐Biegun
- Polymer ScienceZernike Institute for Advanced MaterialsUniversity of GroningenNijenborgh 4Groningen9747 AGThe Netherlands
- Biotechnology CentreThe Silesian University of TechnologyKrzywoustego 8Gliwice44‐100Poland
| |
Collapse
|
6
|
Seddon AR, MacArthur CP, Hampton MB, Stevens AJ. Inflammation and DNA methylation in Alzheimer's disease: mechanisms of epigenetic remodelling by immune cell oxidants in the ageing brain. Redox Rep 2024; 29:2428152. [PMID: 39579010 PMCID: PMC11587723 DOI: 10.1080/13510002.2024.2428152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2024] Open
Abstract
Alzheimer's disease is a neurodegenerative disease involving memory impairment, confusion, and behavioural changes. The disease is characterised by the accumulation of amyloid beta plaques and neurofibrillary tangles in the brain, which disrupt normal neuronal function. There is no known cure for Alzheimer's disease and due to increasing life expectancy, occurrence is projected to rise over the coming decades. The causes of Alzheimer's disease are multifactorial with inflammation, oxidative stress, genetic and epigenetic variation, and cerebrovascular abnormalities among the strongest contributors. We review the current literature surrounding inflammation and epigenetics in Alzheimer's disease, with a focus on how oxidants from infiltrating immune cells have the potential to alter DNA methylation profiles in the ageing brain.
Collapse
Affiliation(s)
- A. R. Seddon
- Mātai Hāora – Centre for Redox Biology and Medicine, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
- Department of Pathology and Molecular Medicine, University of Otago, Wellington, New Zealand
| | - C. P. MacArthur
- Department of Pathology and Molecular Medicine, University of Otago, Wellington, New Zealand
| | - M. B. Hampton
- Mātai Hāora – Centre for Redox Biology and Medicine, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - A. J. Stevens
- Department of Pathology and Molecular Medicine, University of Otago, Wellington, New Zealand
| |
Collapse
|
7
|
Galkin F, Pulous FE, Fu Y, Zhang M, Pun FW, Ren F, Zhavoronkov A. Roles of hypoxia-inducible factor-prolyl hydroxylases in aging and disease. Ageing Res Rev 2024; 102:102551. [PMID: 39447706 DOI: 10.1016/j.arr.2024.102551] [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: 05/08/2024] [Revised: 10/07/2024] [Accepted: 10/15/2024] [Indexed: 10/26/2024]
Abstract
The prolyl hydroxylase domain-containing (PHD or EGL9-homologs) enzyme family is mainly known for its role in the cellular response to hypoxia. HIF-PH inhibitors can stabilize hypoxia-inducible factors (HIFs), activating transcriptional programs that promote processes such as angiogenesis and erythropoiesis to adapt to changes in oxygen levels. HIF-PH inhibitors have been clinically approved for treating several types of anaemia. While most discussions of the HIF-PH signalling axis focus on hypoxia, there is a growing recognition of its importance under normoxic conditions. Recent advances in PHD biology have highlighted the potential of targeting this pathway therapeutically for a range of aging-related diseases. In this article, we review these recent discoveries, situate them within the broader context of aging and disease, and explore current therapeutic strategies that target PHD enzymes for these indications.
Collapse
Affiliation(s)
- Fedor Galkin
- Insilico Medicine AI Ltd., Level 6, Unit 08, Block A, IRENA HQ Building, Masdar City, Abu Dhabi, UAE
| | - Fadi E Pulous
- Insilico Medicine US Inc., 1000 Massachusetts Avenue, Suite 126, Cambridge, MA 02138, United States
| | - Yanyun Fu
- Insilico Medicine Shanghai Ltd., Suite 902, Tower C, Changtai Plaza, 2889 Jinke Road, Pudong, Shanghai 201203, China
| | - Man Zhang
- Insilico Medicine Shanghai Ltd., Suite 902, Tower C, Changtai Plaza, 2889 Jinke Road, Pudong, Shanghai 201203, China
| | - Frank W Pun
- Insilico Medicine Hong Kong Ltd., Unit 310, 3/F, Building 8W, Hong Kong Science and Technology Park, Hong Kong SAR
| | - Feng Ren
- Insilico Medicine AI Ltd., Level 6, Unit 08, Block A, IRENA HQ Building, Masdar City, Abu Dhabi, UAE; Insilico Medicine Shanghai Ltd., Suite 902, Tower C, Changtai Plaza, 2889 Jinke Road, Pudong, Shanghai 201203, China; Insilico Medicine Hong Kong Ltd., Unit 310, 3/F, Building 8W, Hong Kong Science and Technology Park, Hong Kong SAR
| | - Alex Zhavoronkov
- Insilico Medicine AI Ltd., Level 6, Unit 08, Block A, IRENA HQ Building, Masdar City, Abu Dhabi, UAE; Insilico Medicine US Inc., 1000 Massachusetts Avenue, Suite 126, Cambridge, MA 02138, United States; Insilico Medicine Hong Kong Ltd., Unit 310, 3/F, Building 8W, Hong Kong Science and Technology Park, Hong Kong SAR; Insilico Medicine Canada Inc., 1250 René-Lévesque Ouest, Suite 3710, Montréal, Québec H3B 4W8, Canada; Buck Institute for Research on Aging, Novato, CA, United States.
| |
Collapse
|
8
|
Javed S, Shahzadi Z, Yousaf Z, Anjum I, Aftab A, Hanif S, Maqbool Z, Ullah R, Raza MA, Iqbal Z. Anti-anemic potential of Eruca sativa L. in iron-deficient rat model; network pharmacology profiling. Food Sci Nutr 2024; 12:7331-7346. [PMID: 39479620 PMCID: PMC11521654 DOI: 10.1002/fsn3.4314] [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: 01/08/2024] [Revised: 05/21/2024] [Accepted: 06/21/2024] [Indexed: 11/02/2024] Open
Abstract
Iron deficiency anemia is a global health concern, affecting around 2 billion people. Oral iron therapy often causes severe gastro-intestinal issues. Eruca sativa, member of the Brassicaceae family, is valued in traditional medicine and renowned for its rich iron and vitamin C content. This study aims to evaluate the anti-anemic properties of E. sativa extract in vivo and identify its compounds targeting anemia mechanisms using network pharmacology. Thirty-two Sprague-Dawley rats (200 ± 250 g) were split into two distinct groups, iron-deficient and iron-sufficient. Three different doses (200, 400, and 800 mg/kg) of aqueous extract of E. sativa were checked against anemia by studying hematological, oxidative stress, and histopathological parameters. GC-MS analysis of E. sativa revealed its phytochemical profile, followed by ADME screening. Network pharmacology explored targets related to iron deficiency anemia, with oral bioavailability and drug likeness assessment for compounds. The administration of extracts significantly improved various blood parameters, including osmotic fragility, Hb, RBCs, MCV, PCV, and alkaline phosphatase; catalase activity; and histopathological parameters such as liver in both iron-deficient and iron-sufficient rats (p < .001). Seventy-nine compounds were identified in E. sativa aqueous extract, with only six of them found to be bioavailable and drug-like against multiple targets. Gene ontology and pathway analysis revealed their diverse molecular, biological, and cellular functions. One gene EGFR was found to have functional association with ID anemia, suggesting potential for using E. sativa extracts. The study concludes that E. sativa extract has potential for iron deficiency anemia treatment, offering hope for future pharmaceutical interventions.
Collapse
Affiliation(s)
- Sana Javed
- Department of BotanyLahore College for Women UniversityLahorePakistan
| | - Zainab Shahzadi
- Department of BotanyLahore College for Women UniversityLahorePakistan
| | - Zubaida Yousaf
- Department of BotanyLahore College for Women UniversityLahorePakistan
| | - Irfan Anjum
- Department of Basic Medical SciencesShifa College of Pharmaceutical Sciences, Shifa Tameer‐e‐Millat UniversityIslamabadPakistan
| | - Arusa Aftab
- Department of BotanyLahore College for Women UniversityLahorePakistan
| | - Samina Hanif
- Department of BotanyLahore College for Women UniversityLahorePakistan
| | - Zainab Maqbool
- Department of BotanyLahore College for Women UniversityLahorePakistan
| | - Riaz Ullah
- Department of PharmacognosyCollege of Pharmacy King Saud UniversityRiyadhSaudi Arabia
| | - Muhammad Ahmer Raza
- Department of Social and Clinical Pharmacy, Faculty of Pharmacy in Hradec KrálovéCharles University in PraguePragueCzech Republic
| | - Zafar Iqbal
- Department of SurgeryCollege of Medicine, King Saud UniversityRiyadhKingdom of Saudi Arabia
| |
Collapse
|
9
|
Hlukhaniuk A, Świętek M, Patsula V, Hodan J, Janoušková O, Bystrianský L, Brož A, Malić M, Zasońska B, Tokarz W, Bačáková L, Horák D. Poly(ε-Caprolactone)-Based Composites Modified With Polymer-Grafted Magnetic Nanoparticles and L-Ascorbic Acid for Bone Tissue Engineering. J Biomed Mater Res B Appl Biomater 2024; 112:e35480. [PMID: 39223717 DOI: 10.1002/jbm.b.35480] [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: 02/08/2024] [Revised: 07/03/2024] [Accepted: 08/09/2024] [Indexed: 09/04/2024]
Abstract
The aim of this study was to develop multifunctional magnetic poly(ε-caprolactone) (PCL) mats with antibacterial properties for bone tissue engineering and osteosarcoma prevention. To provide good dispersion of magnetic iron oxide nanoparticles (IONs), they were first grafted with PCL using a novel three-step approach. Then, a series of PCL-based mats containing a fixed amount of ION@PCL particles and an increasing content of ascorbic acid (AA) was prepared by electrospinning. AA is known for increasing osteoblast activity and suppressing osteosarcoma cells. Composites were characterized in terms of morphology, mechanical properties, hydrolytic stability, antibacterial performance, and biocompatibility. AA affected both the fiber diameter and the mechanical properties of the nanocomposites. All produced mats were nontoxic to rat bone marrow-derived mesenchymal cells; however, a composite with 5 wt.% of AA suppressed the initial proliferation of SAOS-2 osteoblast-like cells. Moreover, AA improved antibacterial properties against Staphylococcus aureus and Escherichia coli compared to PCL. Overall, these magnetic composites, reported for the very first time, can be used as scaffolds for both tissue regeneration and osteosarcoma prevention.
Collapse
Affiliation(s)
- Anna Hlukhaniuk
- Institute of Macromolecular Chemistry of the Czech Academy of Sciences, Prague, Czech Republic
- Faculty of Science, Charles University, Prague, Czech Republic
| | - Małgorzata Świętek
- Institute of Macromolecular Chemistry of the Czech Academy of Sciences, Prague, Czech Republic
| | - Vitalii Patsula
- Institute of Macromolecular Chemistry of the Czech Academy of Sciences, Prague, Czech Republic
| | - Jiří Hodan
- Institute of Macromolecular Chemistry of the Czech Academy of Sciences, Prague, Czech Republic
| | - Olga Janoušková
- Faculty of Science, Jan Evangelista Purkyně University in Ústí nad Labem, Ústí nad Labem, Czech Republic
| | - Lukáš Bystrianský
- Faculty of Science, Jan Evangelista Purkyně University in Ústí nad Labem, Ústí nad Labem, Czech Republic
| | - Antonín Brož
- Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Marina Malić
- Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Beata Zasońska
- Institute of Macromolecular Chemistry of the Czech Academy of Sciences, Prague, Czech Republic
| | - Waldemar Tokarz
- Faculty of Physics and Applied Computer Science, AGH University of Krakow, Krakow, Poland
| | - Lucie Bačáková
- Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Daniel Horák
- Institute of Macromolecular Chemistry of the Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
10
|
Gvozdenko A, Blinov A, Golik A, Rekhman Z, Nagdalian A, Filippov D, Askerova A, Bocharov N, Kastarnova E, Hassan FA, AL-Farga A, Shariati MA. Harnessing the Power of a Novel Triple Chelate Complex in Fermented Probiotic Dairy Products: A Promising Solution for Combating Iron Deficiency Anemia. ACS OMEGA 2024; 9:28594-28610. [PMID: 38973905 PMCID: PMC11223220 DOI: 10.1021/acsomega.4c02664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 06/02/2024] [Accepted: 06/07/2024] [Indexed: 07/09/2024]
Abstract
This study discovered and examined novel triple chelate complexes involving iron, ascorbic acid, and essential amino acids (AsA-Fe-AmA triple chelate complexes) for the first time. The mechanism of complex formation was studied using FTIR spectroscopy and quantum chemical modeling. The produced complexes were shown to be suitable for fortifying food items with a pH of 3-7 that have not been exposed to heat treatment at temperatures over 75 °C for more than 15 min. Thus, it can be said that the concentration for milk fortification should be 0.005 mol/L or less. In vivo experiments in rats models revealed that the synthesized complexes increased serum iron levels after a single application to reference values within 24 h of oral administration. The iron level increased by 14.0 mmol/L at 2 mL dose of the complex. This fact makes it possible to consider the use of developed complexes and developed fermented dairy products for the prevention of iron deficiency and iron deficiency anemia. Research on the effect of discovered compounds on the physicochemical and organoleptic qualities of milk was conducted. Furthermore, iron ascorbate threoninate, iron ascorbate methioninate, iron ascorbate lysinate, and iron ascorbate tryptophanate all had a beneficial effect on Lacticaseibacillus rhamnosus at concentrations as low as 0.0005 mol/L, which is significant for milk fermentation. A study of fermented milk products revealed that the most effective AsA-Fe-AmA triple chelate complex is iron ascorbate lysinate, which might be further investigated as a viable molecule for dietary fortification in iron deficiency anemia. It was found that fortified fermented milk products had a titratable acidity of 67 ± 1°T, pH of 4.38 ± 0.05, and a viscosity of 2018 ± 142 Pa·s.
Collapse
Affiliation(s)
- Alexey Gvozdenko
- Physical
and Technical Faculty, North-Caucasus Federal
University, Stavropol 355017, Russia
| | - Andrey Blinov
- Physical
and Technical Faculty, North-Caucasus Federal
University, Stavropol 355017, Russia
| | - Alexey Golik
- Physical
and Technical Faculty, North-Caucasus Federal
University, Stavropol 355017, Russia
| | - Zafar Rekhman
- Physical
and Technical Faculty, North-Caucasus Federal
University, Stavropol 355017, Russia
| | - Andrey Nagdalian
- Laboratory
of Food and Industrial Biotechnology, North-Caucasus
Federal University, Stavropol 355017, Russia
| | - Dionis Filippov
- Physical
and Technical Faculty, North-Caucasus Federal
University, Stavropol 355017, Russia
| | - Alina Askerova
- Laboratory
of Food and Industrial Biotechnology, North-Caucasus
Federal University, Stavropol 355017, Russia
| | - Nikita Bocharov
- Physical
and Technical Faculty, North-Caucasus Federal
University, Stavropol 355017, Russia
| | - Elena Kastarnova
- Eterinary
Faculty, Stavropol Sate Agrarian University, Zootechnicheskiy Street 9, Stavropol 355017, Russia
| | - Faten Abdo Hassan
- Faculty
of Science, Department of Microbiology, Taiz University, Taiz 9674, Yemen
| | - Ammar AL-Farga
- Department
of Biochemistry, College of Science, University
of Jeddah, Jeddah 21577, Saudi Arabia
| | - Mohammad Ali Shariati
- Scientific
Department, Semey Branch of the Kazakh Research
Institute of Processing and Food Industry, Gagarin Avenue 238G, Almaty 050060, Kazakhstan
| |
Collapse
|
11
|
Roy N, Paira P. Glutathione Depletion and Stalwart Anticancer Activity of Metallotherapeutics Inducing Programmed Cell Death: Opening a New Window for Cancer Therapy. ACS OMEGA 2024; 9:20670-20701. [PMID: 38764686 PMCID: PMC11097382 DOI: 10.1021/acsomega.3c08890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 03/22/2024] [Accepted: 04/05/2024] [Indexed: 05/21/2024]
Abstract
The cellular defense system against exogenous substances makes therapeutics inefficient as intracellular glutathione (GSH) exhibits an astounding antioxidant activity in scavenging reactive oxygen species (ROS) or reactive nitrogen species (RNS) or other free radicals produced by the therapeutics. In the cancer cell microenvironment, the intracellular GSH level becomes exceptionally high to fight against oxidative stress created by the production of ROS/RNS or any free radicals, which are the byproducts of intracellular redox reactions or cellular respiration processes. Thus, in order to maintain redox homeostasis for survival of cancer cells and their rapid proliferation, the GSH level starts to escalate. In this circumstance, the administration of anticancer therapeutics is in vain, as the elevated GSH level reduces their potential by reduction or by scavenging the ROS/RNS they produce. Therefore, in order to augment the therapeutic potential of anticancer agents against elevated GSH condition, the GSH level must be depleted by hook or by crook. Hence, this Review aims to compile precisely the role of GSH in cancer cells, the importance of its depletion for cancer therapy and examples of anticancer activity of a few selected metal complexes which are able to trigger cancer cell death by depleting the GSH level.
Collapse
Affiliation(s)
- Nilmadhab Roy
- Department of Chemistry, School of
Advanced Sciences, Vellore Institute of
Technology, Vellore-632014, Tamilnadu, India
| | - Priyankar Paira
- Department of Chemistry, School of
Advanced Sciences, Vellore Institute of
Technology, Vellore-632014, Tamilnadu, India
| |
Collapse
|
12
|
Kontoghiorghes GJ. The Importance and Essentiality of Natural and Synthetic Chelators in Medicine: Increased Prospects for the Effective Treatment of Iron Overload and Iron Deficiency. Int J Mol Sci 2024; 25:4654. [PMID: 38731873 PMCID: PMC11083551 DOI: 10.3390/ijms25094654] [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: 04/02/2024] [Revised: 04/19/2024] [Accepted: 04/22/2024] [Indexed: 05/13/2024] Open
Abstract
The supply and control of iron is essential for all cells and vital for many physiological processes. All functions and activities of iron are expressed in conjunction with iron-binding molecules. For example, natural chelators such as transferrin and chelator-iron complexes such as haem play major roles in iron metabolism and human physiology. Similarly, the mainstay treatments of the most common diseases of iron metabolism, namely iron deficiency anaemia and iron overload, involve many iron-chelator complexes and the iron-chelating drugs deferiprone (L1), deferoxamine (DF) and deferasirox. Endogenous chelators such as citric acid and glutathione and exogenous chelators such as ascorbic acid also play important roles in iron metabolism and iron homeostasis. Recent advances in the treatment of iron deficiency anaemia with effective iron complexes such as the ferric iron tri-maltol complex (feraccru or accrufer) and the effective treatment of transfusional iron overload using L1 and L1/DF combinations have decreased associated mortality and morbidity and also improved the quality of life of millions of patients. Many other chelating drugs such as ciclopirox, dexrazoxane and EDTA are used daily by millions of patients in other diseases. Similarly, many other drugs or their metabolites with iron-chelation capacity such as hydroxyurea, tetracyclines, anthracyclines and aspirin, as well as dietary molecules such as gallic acid, caffeic acid, quercetin, ellagic acid, maltol and many other phytochelators, are known to interact with iron and affect iron metabolism and related diseases. Different interactions are also observed in the presence of essential, xenobiotic, diagnostic and theranostic metal ions competing with iron. Clinical trials using L1 in Parkinson's, Alzheimer's and other neurodegenerative diseases, as well as HIV and other infections, cancer, diabetic nephropathy and anaemia of inflammation, highlight the importance of chelation therapy in many other clinical conditions. The proposed use of iron chelators for modulating ferroptosis signifies a new era in the design of new therapeutic chelation strategies in many other diseases. The introduction of artificial intelligence guidance for optimal chelation therapeutic outcomes in personalised medicine is expected to increase further the impact of chelation in medicine, as well as the survival and quality of life of millions of patients with iron metabolic disorders and also other diseases.
Collapse
Affiliation(s)
- George J Kontoghiorghes
- Postgraduate Research Institute of Science, Technology, Environment and Medicine, Limassol 3021, Cyprus
| |
Collapse
|
13
|
Zhang L, Gao J, Liu Y, Zhou Z, Sheng X, Li D, Chen Y, Lyu S. Ascorbic acid enhanced the circulation between Fe(II) and Fe(III) in peroxymonosulfate system for fluoranthene degradation. WATER SCIENCE AND TECHNOLOGY : A JOURNAL OF THE INTERNATIONAL ASSOCIATION ON WATER POLLUTION RESEARCH 2024; 89:1682-1700. [PMID: 38619897 DOI: 10.2166/wst.2024.098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 03/11/2024] [Indexed: 04/17/2024]
Abstract
In this research, ascorbic acid (AA) was used to enhance Fe(II)/Fe(III)-activated permonosulfate (PMS) systems for the degradation of fluoranthene (FLT). AA enhanced the production of ROS in both PMS/Fe(II) and PMS/Fe(III) systems through chelation and reduction and thus improved the degradation performance of FLT. The optimal molar ratio in PMS/Fe(II)/AA/FLT and PMS/Fe(III)/AA/FLT processes were 2/2/4/1 and 5/10/5/1, respectively. In addition, the experimental results on the effect of FLT degradation under different groundwater matrixes indicated that PMS/Fe(III)/AA system was more adaptable to different water quality conditions than the PMS/Fe(II)/AA system. SO4·- was the major reactive oxygen species (ROS) responsible for FLT removal through the probe and scavenging tests in both systems. Furthermore, the degradation intermediates of FLT were analyzed using gas chromatograph-mass spectrometry (GC-MS), and the probable degradation pathways of FLT degradation were proposed. In addition, the removal of FLT was also tested in actual groundwater and the results showed that by increasing the dose and pre-adjusting the solution pH, 88.8 and 100% of the FLT was removed for PMS/Fe(II)/AA and PMS/Fe(III)/AA systems. The above experimental results demonstrated that PMS/Fe(II)/AA and PMS/Fe(III)/AA processes have a great perspective in practice for the rehabilitation of FLT-polluted groundwater.
Collapse
Affiliation(s)
- Longbin Zhang
- State Environmental Protection Key Laboratory of Environmental Risk Assessment and Control on Chemical Process, East China University of Science and Technology, Shanghai 200237, China
| | - Jianxiong Gao
- State Environmental Protection Key Laboratory of Environmental Risk Assessment and Control on Chemical Process, East China University of Science and Technology, Shanghai 200237, China
| | - Yulong Liu
- State Environmental Protection Key Laboratory of Environmental Risk Assessment and Control on Chemical Process, East China University of Science and Technology, Shanghai 200237, China
| | - Zhengyuan Zhou
- State Environmental Protection Key Laboratory of Environmental Risk Assessment and Control on Chemical Process, East China University of Science and Technology, Shanghai 200237, China
| | - Xianxian Sheng
- State Environmental Protection Key Laboratory of Environmental Risk Assessment and Control on Chemical Process, East China University of Science and Technology, Shanghai 200237, China
| | - Dexiao Li
- State Environmental Protection Key Laboratory of Environmental Risk Assessment and Control on Chemical Process, East China University of Science and Technology, Shanghai 200237, China
| | - Yuantian Chen
- State Environmental Protection Key Laboratory of Environmental Risk Assessment and Control on Chemical Process, East China University of Science and Technology, Shanghai 200237, China
| | - Shuguang Lyu
- State Environmental Protection Key Laboratory of Environmental Risk Assessment and Control on Chemical Process, East China University of Science and Technology, Shanghai 200237, China E-mail:
| |
Collapse
|
14
|
Sun B, Tan B, Zhang P, Zhu L, Wei H, Huang T, Li C, Yang W. Iron deficiency anemia: a critical review on iron absorption, supplementation and its influence on gut microbiota. Food Funct 2024; 15:1144-1157. [PMID: 38235788 DOI: 10.1039/d3fo04644c] [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: 01/19/2024]
Abstract
Iron deficiency anemia (IDA) caused by micronutrient iron deficiency has attracted global attention due to its adverse health effects. The regulation of iron uptake and metabolism is finely controlled by various transporters and hormones in the body. Dietary iron intake and regulation are essential in maintaining human health and iron requirements. The review aims to investigate literature concerning dietary iron intake and systemic regulation. Besides, recent IDA treatment and dietary iron supplementation are discussed. Considering the importance of the gut microbiome, the interaction between bacteria and micronutrient iron in the gut is also a focus of this review. The iron absorption efficiency varies considerably according to iron type and dietary factors. Iron fortification remains the cost-effective strategy, although challenges exist in developing suitable iron fortificants and food vehicles regarding bioavailability and acceptability. Iron deficiency may alter the microbiome structure and promote the growth of pathogenic bacteria in the gut, affecting immune balance and human health.
Collapse
Affiliation(s)
- Bolun Sun
- College of Food and Pharmaceutical Sciences, Key Laboratory of Animal Protein Food Deep Processing Technology of Zhejiang Province, Ningbo University, Ningbo 315211, China.
- School of Nursing, Wenzhou Medical University, Wenzhou 325035, China
| | - Beibei Tan
- School of Agriculture and Food, Faculty of Science, University of Melbourne, Australia
| | - Panxue Zhang
- College of Food and Pharmaceutical Sciences, Key Laboratory of Animal Protein Food Deep Processing Technology of Zhejiang Province, Ningbo University, Ningbo 315211, China.
| | - Lianlian Zhu
- School of Nursing, Wenzhou Medical University, Wenzhou 325035, China
| | - Huamao Wei
- College of Food and Pharmaceutical Sciences, Key Laboratory of Animal Protein Food Deep Processing Technology of Zhejiang Province, Ningbo University, Ningbo 315211, China.
| | - Tao Huang
- College of Food and Pharmaceutical Sciences, Key Laboratory of Animal Protein Food Deep Processing Technology of Zhejiang Province, Ningbo University, Ningbo 315211, China.
| | - Chao Li
- College of Food and Pharmaceutical Sciences, Key Laboratory of Animal Protein Food Deep Processing Technology of Zhejiang Province, Ningbo University, Ningbo 315211, China.
| | - Wenge Yang
- College of Food and Pharmaceutical Sciences, Key Laboratory of Animal Protein Food Deep Processing Technology of Zhejiang Province, Ningbo University, Ningbo 315211, China.
| |
Collapse
|
15
|
Albreht A, Martelanc M, Žiberna L. Simultaneous determination of free biliverdin and free bilirubin in serum: A comprehensive LC-MS approach. Anal Chim Acta 2024; 1287:342073. [PMID: 38182377 DOI: 10.1016/j.aca.2023.342073] [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: 09/27/2023] [Revised: 11/21/2023] [Accepted: 11/25/2023] [Indexed: 01/07/2024]
Abstract
BACKGROUND Prognosis, diagnosis, and treatment of several diseases strongly rely on the sensitive, selective, and accurate determination of specific biomarkers in relevant biological samples. Free biliverdin and free bilirubin represent important new biomarkers of oxidative stress, however, the lack of suitable analytical methods for their determination has hindered progress in biomedical and clinical research. RESULTS Here, we introduce a first comprehensive approach for robust and simultaneous determination of these bilins in serum using liquid chromatography - mass spectrometry (LC-MS). The developed analytical method exhibits linearity for both analytes within the concentration range of 0.5-100 nM, with limits of detection and quantitation determined at 0.1 nM and 0.5 nM, respectively. Moreover, several analytical pitfalls related to the intrinsic molecular structures of free bilirubin and free biliverdin and their trace concentration levels in biological samples are discussed here in detail for the first time. We have shown that the solubility, chemical stability, and affinity of these bilins to various materials strongly depend on the solvent, pH, and addition of stabilizing and chelating agents. Finally, the validated LC-MS method was successfully applied to the analysis of both bilins in fetus bovine serums, yielding higher free bilirubin/biliverdin ratios compared with previously reported values for human serum. SIGNIFICANCE Failure to recognize and address the challenges presented here often leads to substantial analytical errors and consequently biased interpretation of the obtained results. This pertains not only to LC-MS, but also to many other analytical platforms due to the compound-derived sources of error.
Collapse
Affiliation(s)
- Alen Albreht
- Laboratory for Food Chemistry, Department of Analytical Chemistry, National Institute of Chemistry, Hajdrihova 19, Ljubljana, SI-1000, Slovenia.
| | - Mitja Martelanc
- University of Nova Gorica, Wine Research Centre, Glavni trg 8, Vipava, SI-5271, Slovenia; University of Nova Gorica, School for Viticulture and Enology, Glavni trg 8, Vipava, SI-5271, Slovenia
| | - Lovro Žiberna
- University of Ljubljana, Faculty of Medicine, Institute of Pharmacology and Experimental Toxicology, Korytkova 2, Ljubljana, SI-1000, Slovenia; University of Ljubljana, Faculty of Pharmacy, Department of Biopharmaceutics and Pharmacokinetics, Aškerčeva 7, Ljubljana, SI-1000, Slovenia
| |
Collapse
|
16
|
Fowler AA. Vitamin C: Rationale for Its Use in Sepsis-Induced Acute Respiratory Distress Syndrome (ARDS). Antioxidants (Basel) 2024; 13:95. [PMID: 38247519 PMCID: PMC10812524 DOI: 10.3390/antiox13010095] [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: 11/27/2023] [Revised: 01/08/2024] [Accepted: 01/09/2024] [Indexed: 01/23/2024] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a life-threatening event that occurs in patients suffering from bacterial, fungal, or viral sepsis. Research performed over the last five decades showed that ARDS is a consequence of severe unrestrained systemic inflammation, which leads to injury of the lung's microvasculature and alveolar epithelium. ARDS leads to acute hypoxic/hypercapnic respiratory failure and death in a significant number of patients hospitalized in intensive care units worldwide. Basic and clinical research performed during the time since ARDS was first described has been unable to construct a pharmacological agent that will combat the inflammatory fire leading to ARDS. In-depth studies of the molecular pharmacology of vitamin C indicate that it can serve as a potent anti-inflammatory agent capable of attenuating the pathobiological events that lead to acute injury of the lungs and other body organs. This analysis of vitamin C's role in the treatment of ARDS includes a focused systematic review of the literature relevant to the molecular physiology of vitamin C and to the past performance of clinical trials using the agent.
Collapse
Affiliation(s)
- Alpha A Fowler
- Division of Pulmonary Disease and Critical Care Medicine, Department of Internal Medicine, School of Medicine, Virginia Commonwealth University, Richmond, VA 23219, USA
| |
Collapse
|
17
|
Kontoghiorghes GJ. Drug Selection and Posology, Optimal Therapies and Risk/Benefit Assessment in Medicine: The Paradigm of Iron-Chelating Drugs. Int J Mol Sci 2023; 24:16749. [PMID: 38069073 PMCID: PMC10706143 DOI: 10.3390/ijms242316749] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 11/19/2023] [Accepted: 11/22/2023] [Indexed: 12/18/2023] Open
Abstract
The design of clinical protocols and the selection of drugs with appropriate posology are critical parameters for therapeutic outcomes. Optimal therapeutic protocols could ideally be designed in all diseases including for millions of patients affected by excess iron deposition (EID) toxicity based on personalised medicine parameters, as well as many variations and limitations. EID is an adverse prognostic factor for all diseases and especially for millions of chronically red-blood-cell-transfused patients. Differences in iron chelation therapy posology cause disappointing results in neurodegenerative diseases at low doses, but lifesaving outcomes in thalassemia major (TM) when using higher doses. In particular, the transformation of TM from a fatal to a chronic disease has been achieved using effective doses of oral deferiprone (L1), which improved compliance and cleared excess toxic iron from the heart associated with increased mortality in TM. Furthermore, effective L1 and L1/deferoxamine combination posology resulted in the complete elimination of EID and the maintenance of normal iron store levels in TM. The selection of effective chelation protocols has been monitored by MRI T2* diagnosis for EID levels in different organs. Millions of other iron-loaded patients with sickle cell anemia, myelodysplasia and haemopoietic stem cell transplantation, or non-iron-loaded categories with EID in different organs could also benefit from such chelation therapy advances. Drawbacks of chelation therapy include drug toxicity in some patients and also the wide use of suboptimal chelation protocols, resulting in ineffective therapies. Drug metabolic effects, and interactions with other metals, drugs and dietary molecules also affected iron chelation therapy. Drug selection and the identification of effective or optimal dose protocols are essential for positive therapeutic outcomes in the use of chelating drugs in TM and other iron-loaded and non-iron-loaded conditions, as well as general iron toxicity.
Collapse
Affiliation(s)
- George J Kontoghiorghes
- Postgraduate Research Institute of Science, Technology, Environment and Medicine, Limassol 3021, Cyprus
| |
Collapse
|
18
|
Wang J, Fu J, Zhao Y, Liu Q, Yan X, Su J. Iron and Targeted Iron Therapy in Alzheimer's Disease. Int J Mol Sci 2023; 24:16353. [PMID: 38003544 PMCID: PMC10671546 DOI: 10.3390/ijms242216353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 10/31/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease worldwide. β-amyloid plaque (Aβ) deposition and hyperphosphorylated tau, as well as dysregulated energy metabolism in the brain, are key factors in the progression of AD. Many studies have observed abnormal iron accumulation in different regions of the AD brain, which is closely correlated with the clinical symptoms of AD; therefore, understanding the role of brain iron accumulation in the major pathological aspects of AD is critical for its treatment. This review discusses the main mechanisms and recent advances in the involvement of iron in the above pathological processes, including in iron-induced oxidative stress-dependent and non-dependent directions, summarizes the hypothesis that the iron-induced dysregulation of energy metabolism may be an initiating factor for AD, based on the available evidence, and further discusses the therapeutic perspectives of targeting iron.
Collapse
Affiliation(s)
| | | | | | | | | | - Jing Su
- Key Laboratory of Pathobiology, Department of Pathophysiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Street, Changchun 130012, China; (J.W.); (J.F.); (Y.Z.); (Q.L.); (X.Y.)
| |
Collapse
|
19
|
Feng J, Zheng Y, Guo M, Ares I, Martínez M, Lopez-Torres B, Martínez-Larrañaga MR, Wang X, Anadón A, Martínez MA. Oxidative stress, the blood-brain barrier and neurodegenerative diseases: The critical beneficial role of dietary antioxidants. Acta Pharm Sin B 2023; 13:3988-4024. [PMID: 37799389 PMCID: PMC10547923 DOI: 10.1016/j.apsb.2023.07.010] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 03/16/2023] [Accepted: 06/13/2023] [Indexed: 10/07/2023] Open
Abstract
In recent years, growing awareness of the role of oxidative stress in brain health has prompted antioxidants, especially dietary antioxidants, to receive growing attention as possible treatments strategies for patients with neurodegenerative diseases (NDs). The most widely studied dietary antioxidants include active substances such as vitamins, carotenoids, flavonoids and polyphenols. Dietary antioxidants are found in usually consumed foods such as fresh fruits, vegetables, nuts and oils and are gaining popularity due to recently growing awareness of their potential for preventive and protective agents against NDs, as well as their abundant natural sources, generally non-toxic nature, and ease of long-term consumption. This review article examines the role of oxidative stress in the development of NDs, explores the 'two-sidedness' of the blood-brain barrier (BBB) as a protective barrier to the nervous system and an impeding barrier to the use of antioxidants as drug medicinal products and/or dietary antioxidants supplements for prevention and therapy and reviews the BBB permeability of common dietary antioxidant suplements and their potential efficacy in the prevention and treatment of NDs. Finally, current challenges and future directions for the prevention and treatment of NDs using dietary antioxidants are discussed, and useful information on the prevention and treatment of NDs is provided.
Collapse
Affiliation(s)
- Jin Feng
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan 430070, China
| | - Youle Zheng
- MAO Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan 430070, China
| | - Mingyue Guo
- MAO Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan 430070, China
| | - Irma Ares
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Universidad Complutense de Madrid (UCM), And Research Institute Hospital 12 de Octubre (i+12), Madrid 28040, Spain
| | - Marta Martínez
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Universidad Complutense de Madrid (UCM), And Research Institute Hospital 12 de Octubre (i+12), Madrid 28040, Spain
| | - Bernardo Lopez-Torres
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Universidad Complutense de Madrid (UCM), And Research Institute Hospital 12 de Octubre (i+12), Madrid 28040, Spain
| | - María-Rosa Martínez-Larrañaga
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Universidad Complutense de Madrid (UCM), And Research Institute Hospital 12 de Octubre (i+12), Madrid 28040, Spain
| | - Xu Wang
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan 430070, China
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Universidad Complutense de Madrid (UCM), And Research Institute Hospital 12 de Octubre (i+12), Madrid 28040, Spain
| | - Arturo Anadón
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Universidad Complutense de Madrid (UCM), And Research Institute Hospital 12 de Octubre (i+12), Madrid 28040, Spain
| | - María-Aránzazu Martínez
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Universidad Complutense de Madrid (UCM), And Research Institute Hospital 12 de Octubre (i+12), Madrid 28040, Spain
| |
Collapse
|
20
|
von Siebenthal HK, Moretti D, Zimmermann MB, Stoffel NU. Effect of dietary factors and time of day on iron absorption from oral iron supplements in iron deficient women. Am J Hematol 2023; 98:1356-1363. [PMID: 37357807 DOI: 10.1002/ajh.26987] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 05/02/2023] [Accepted: 05/23/2023] [Indexed: 06/27/2023]
Abstract
Guidelines generally recommend taking iron supplements in the morning away from meals and with ascorbic acid (AA) to increase iron absorption. However, there is little direct evidence on the effects of dietary factors and time of day on absorption from iron supplements. In iron-depleted women (n = 34; median serum ferritin 19.4 μg/L), we administered 100 mg iron doses labeled with 54 Fe, 57 Fe, or 58 Fe in each of six different conditions with: (1) water (reference) in the morning; (2) 80 mg AA; (3) 500 mg AA; (4) coffee; (5) breakfast including coffee and orange juice (containing ~90 mg AA); and (6) water in the afternoon. Fractional iron absorption (FIA) from these n = 204 doses was calculated based on erythrocyte incorporation of multiple isotopic labels. Compared to the reference: 80 mg AA increased FIA by 30% (p < .001) but 500 mg AA did not further increase FIA (p = .226); coffee decreased FIA by 54% (p = .004); coffee with breakfast decreased FIA by 66% (p < .001) despite the presence of ~90 mg of AA. Serum hepcidin was higher (p < .001) and FIA was 37% lower (p = .059) in the afternoon compared to the morning. Our data suggest that to maximize efficacy, ferrous iron supplements should be consumed in the morning, away from meals or coffee, and with an AA-rich food or beverage. Compared to consuming a 100 mg iron dose in the morning with coffee or breakfast, consuming it with orange juice alone results in a ~ 4-fold increase in iron absorption, and provides ~20 more mg of absorbed iron per dose. The trial was registered at Clinicaltrials.gov(NCT04074707).
Collapse
Affiliation(s)
- Hanna K von Siebenthal
- Laboratory of Human Nutrition, Department of Health Sciences and Technology, ETH Zurich, Zürich, Switzerland
| | - Diego Moretti
- Department of Health, Swiss Distance University of Applied Sciences, Zürich, Switzerland
| | - Michael B Zimmermann
- Laboratory of Human Nutrition, Department of Health Sciences and Technology, ETH Zurich, Zürich, Switzerland
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Nicole U Stoffel
- Laboratory of Human Nutrition, Department of Health Sciences and Technology, ETH Zurich, Zürich, Switzerland
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| |
Collapse
|
21
|
Kontoghiorghes GJ. Iron Load Toxicity in Medicine: From Molecular and Cellular Aspects to Clinical Implications. Int J Mol Sci 2023; 24:12928. [PMID: 37629109 PMCID: PMC10454416 DOI: 10.3390/ijms241612928] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 08/12/2023] [Accepted: 08/15/2023] [Indexed: 08/27/2023] Open
Abstract
Iron is essential for all organisms and cells. Diseases of iron imbalance affect billions of patients, including those with iron overload and other forms of iron toxicity. Excess iron load is an adverse prognostic factor for all diseases and can cause serious organ damage and fatalities following chronic red blood cell transfusions in patients of many conditions, including hemoglobinopathies, myelodyspasia, and hematopoietic stem cell transplantation. Similar toxicity of excess body iron load but at a slower rate of disease progression is found in idiopathic haemochromatosis patients. Excess iron deposition in different regions of the brain with suspected toxicity has been identified by MRI T2* and similar methods in many neurodegenerative diseases, including Alzheimer's disease and Parkinson's disease. Based on its role as the major biological catalyst of free radical reactions and the Fenton reaction, iron has also been implicated in all diseases associated with free radical pathology and tissue damage. Furthermore, the recent discovery of ferroptosis, which is a cell death program based on free radical generation by iron and cell membrane lipid oxidation, sparked thousands of investigations and the association of iron with cardiac, kidney, liver, and many other diseases, including cancer and infections. The toxicity implications of iron in a labile, non-protein bound form and its complexes with dietary molecules such as vitamin C and drugs such as doxorubicin and other xenobiotic molecules in relation to carcinogenesis and other forms of toxicity are also discussed. In each case and form of iron toxicity, the mechanistic insights, diagnostic criteria, and molecular interactions are essential for the design of new and effective therapeutic interventions and of future targeted therapeutic strategies. In particular, this approach has been successful for the treatment of most iron loading conditions and especially for the transition of thalassemia from a fatal to a chronic disease due to new therapeutic protocols resulting in the complete elimination of iron overload and of iron toxicity.
Collapse
Affiliation(s)
- George J Kontoghiorghes
- Postgraduate Research Institute of Science, Technology, Environment and Medicine, 3, Ammochostou Street, Limassol 3021, Cyprus
| |
Collapse
|
22
|
Schreiner OD, Schreiner TG. Iron chelators as a therapeutic option for Alzheimer's disease-A mini-review. FRONTIERS IN AGING 2023; 4:1234958. [PMID: 37602277 PMCID: PMC10433644 DOI: 10.3389/fragi.2023.1234958] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 07/17/2023] [Indexed: 08/22/2023]
Abstract
Neurodegenerative disorders, particularly Alzheimer's disease (AD), remain a great challenge regarding the finding of effective treatment, one main reason being the incomplete understanding of their etiology. With many intensely debated hypotheses, a newer approach based on the impact of iron imbalance in sustaining neurodegeneration in the central nervous system becomes increasingly popular. Altered iron homeostasis leads to increased iron accumulation in specific brain areas, explaining the clinical picture of AD patients. Moreover, growing evidence sustains the significant impact of iron metabolism in relationship to other pathological processes encountered in the AD-affected brain, such as the amyloidogenic pathway, chronic inflammation, or oxidative stress. In this context, this mini-review aims to summarize the novel data from the continuously expanding literature on this topic in a didactic manner. Thus, in the first part, the authors briefly highlight the most relevant aspects related to iron absorption, transport, regulation, and elimination at the cerebral level, focusing on the role of the blood-brain barrier and the newer concept of ferroptosis. Subsequently, currently available iron chelation therapies are discussed, including an overview of the most relevant clinical trials on this topic. In the final part, based on the latest results from in vitro and in vivo studies, new research directions are suggested to enhance the development of effective antidementia therapies.
Collapse
Affiliation(s)
- Oliver Daniel Schreiner
- Grigore T. Popa University of Medicine and Pharmacy, Iasi, Romania
- Medical Oncology Department, Regional Institute of Oncology, Iasi, Romania
| | - Thomas Gabriel Schreiner
- Grigore T. Popa University of Medicine and Pharmacy, Iasi, Romania
- Faculty of Electrical Engineering and Information Technology, Gheorghe Asachi Technical University of Iasi, Iasi, Romania
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, Bucharest, Romania
| |
Collapse
|
23
|
Kontoghiorghes GJ. The Vital Role Played by Deferiprone in the Transition of Thalassaemia from a Fatal to a Chronic Disease and Challenges in Its Repurposing for Use in Non-Iron-Loaded Diseases. Pharmaceuticals (Basel) 2023; 16:1016. [PMID: 37513928 PMCID: PMC10384919 DOI: 10.3390/ph16071016] [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] [Received: 06/26/2023] [Revised: 07/13/2023] [Accepted: 07/15/2023] [Indexed: 07/30/2023] Open
Abstract
The iron chelating orphan drug deferiprone (L1), discovered over 40 years ago, has been used daily by patients across the world at high doses (75-100 mg/kg) for more than 30 years with no serious toxicity. The level of safety and the simple, inexpensive synthesis are some of the many unique properties of L1, which played a major role in the contribution of the drug in the transition of thalassaemia from a fatal to a chronic disease. Other unique and valuable clinical properties of L1 in relation to pharmacology and metabolism include: oral effectiveness, which improved compliance compared to the prototype therapy with subcutaneous deferoxamine; highly effective iron removal from all iron-loaded organs, particularly the heart, which is the major target organ of iron toxicity and the cause of mortality in thalassaemic patients; an ability to achieve negative iron balance, completely remove all excess iron, and maintain normal iron stores in thalassaemic patients; rapid absorption from the stomach and rapid clearance from the body, allowing a greater frequency of repeated administration and overall increased efficacy of iron excretion, which is dependent on the dose used and also the concentration achieved at the site of drug action; and its ability to cross the blood-brain barrier and treat malignant, neurological, and microbial diseases affecting the brain. Some differential pharmacological activity by L1 among patients has been generally shown in relation to the absorption, distribution, metabolism, elimination, and toxicity (ADMET) of the drug. Unique properties exhibited by L1 in comparison to other drugs include specific protein interactions and antioxidant effects, such as iron removal from transferrin and lactoferrin; inhibition of iron and copper catalytic production of free radicals, ferroptosis, and cuproptosis; and inhibition of iron-containing proteins associated with different pathological conditions. The unique properties of L1 have attracted the interest of many investigators for drug repurposing and use in many pathological conditions, including cancer, neurodegenerative conditions, microbial conditions, renal conditions, free radical pathology, metal intoxication in relation to Fe, Cu, Al, Zn, Ga, In, U, and Pu, and other diseases. Similarly, the properties of L1 increase the prospects of its wider use in optimizing therapeutic efforts in many other fields of medicine, including synergies with other drugs.
Collapse
Affiliation(s)
- George J Kontoghiorghes
- Postgraduate Research Institute of Science, Technology, Environment and Medicine, Limassol 3021, Cyprus
| |
Collapse
|
24
|
Elhassan S, Dong F, Buckner T, Johnson RK, Seifert JA, Carry PM, Vanderlinden L, Waugh K, Rewers M, Norris JM. Investigating iron intake in risk of progression from islet autoimmunity to type 1 diabetes: The diabetes autoimmunity study in the young. Front Immunol 2023; 14:1124370. [PMID: 37056761 PMCID: PMC10086157 DOI: 10.3389/fimmu.2023.1124370] [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: 12/15/2022] [Accepted: 03/15/2023] [Indexed: 03/30/2023] Open
Abstract
Background Studies of the role of iron in the risk of type 1 diabetes (T1D) have been inconsistent. Given that iron generates reactive oxygen radicals, which can lead to oxidative damage and apoptosis in the beta cells of the pancreas, we examined whether iron intake was associated with the risk of progressing to T1D in individuals with islet autoimmunity (IA), the pre-clinical phase of T1D. Methods DAISY is a prospective cohort following 2,547 children at increased risk for IA and progression to T1D. IA is defined as at least two consecutive serum samples positive for at least one autoantibody (insulin, GAD, IA-2, or ZnT8). We measured dietary intake at the time of IA seroconversion in 175 children with IA, and of these, 64 progressed to T1D. We used Cox regression to examine the association between energy-adjusted iron intake and progression to T1D, adjusting for HLA-DR3/4 genotype, race/ethnicity, age at seroconversion, presence of multiple autoantibodies at seroconversion, and multiple vitamin use. In addition, we tested whether this association was modified by vitamin C or calcium intake. Results In children with IA, high iron intake (as defined as above the 75th percentile, > 20.3 mg/day) was associated with decreased risk of progression to T1D compared to moderate iron intake (as defined by the middle 25-75th percentiles, 12.7-20.3 mg/day) (adjusted hazard ratio (HR): 0.35; 95% confidence interval (CI): 0.15, 0.79). The association between iron intake and T1D was not modified by vitamin C nor calcium intake. In a sensitivity analysis, the removal of six children who had been diagnosed with celiac disease prior to IA seroconversion did not affect this association. Conclusion Higher iron intake at the time of IA seroconversion is associated with a lower risk of progression to T1D, independent of multivitamin supplement use. Further research that includes plasma biomarkers of iron status is needed to investigate the relationship between iron and the risk of T1D.
Collapse
Affiliation(s)
- Sulafa Elhassan
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Fran Dong
- Barbara Davis Center for Diabetes, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Teresa Buckner
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Department of Kinesiology, Nutrition, and Dietetics, University of Northern Colorado, Greeley, CO, United States
| | - Randi K. Johnson
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Department of Biomedical Informatics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Jennifer A. Seifert
- Department of Medicine, Division of Rheumatology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Patrick M. Carry
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Colorado Program for Musculoskeletal Research, Department of Orthopedics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Lauren Vanderlinden
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Kathleen Waugh
- Barbara Davis Center for Diabetes, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Marian Rewers
- Barbara Davis Center for Diabetes, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Jill M. Norris
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
25
|
Shuai C, Chen X, He C, Chen M, Peng S, Yang W. Fe-doped mesoporous silica catalyzes ascorbic acid oxidation for tumor-specific therapy in scaffold. Colloids Surf B Biointerfaces 2023; 225:113251. [PMID: 36931045 DOI: 10.1016/j.colsurfb.2023.113251] [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] [Received: 10/11/2022] [Revised: 02/15/2023] [Accepted: 03/04/2023] [Indexed: 03/09/2023]
Abstract
Ascorbic acid (AA) is a promising antitumor agent, yet its autooxidation is too slow which constrains the further application. Fortunately, the autoxidation process can be accelerated by transition metal catalysts, especially Fe3+ ions. In this study, AA was loaded to Fe-doped mesoporous silica (designated as AA@Fe-SiO2), which was introduced into poly-L-lactic acid (PLLA) and then prepared into a scaffold. Mechanistically, AA@Fe-SiO2 degraded in acidic tumor microenvironment because excessive H+ substituted Fe atoms in the iron silicate framework, releasing Fe3+ and AA. The Fe3+ boosted the pro-oxidation reaction of AA, generating numerous hydrogen peroxide (H2O2) and Fe2+. Then, Fe2+ reacted with H2O2 to initiate Fenton reactions favoring hydroxyl radical generation, triggering oxidative damage on tumor cells to implement tumor-specific therapy. Results showed that the release amount of AA in acidic solution was about 3 times higher than that in neutral solution, which was attributed to the pH-dependency of the degradation of AA@Fe-SiO2 in scaffold. Furthermore, the scaffold generated numerous ascorbate radical intermediate and increased the H2O2 concentration by 120.2%, demonstrating that Fe3+ remarkably accelerated the oxidation rate of AA. Cell experimental results showed that the scaffold caused massive apoptosis of tumor cells, while no obvious cytotoxicity to normal cells, confirming the antitumor specificity of scaffold. This work paves a promising way to construct a biodegradable and catalytic scaffold, featuring effective tumor-specific therapy.
Collapse
Affiliation(s)
- Cijun Shuai
- Institute of Additive Manufacturing, Jiangxi University of Science and Technology, Nanchang 330013, China; State Key Laboratory of High Performance Complex Manufacturing, Central South University, Changsha 410083, China; Shenzhen Institute of Information Technology, School of Sino-German Robotics, Shenzhen 518115, China
| | - Xuan Chen
- Institute of Additive Manufacturing, Jiangxi University of Science and Technology, Nanchang 330013, China
| | - Chongxian He
- State Key Laboratory of High Performance Complex Manufacturing, Central South University, Changsha 410083, China
| | - Min Chen
- Institute of Additive Manufacturing, Jiangxi University of Science and Technology, Nanchang 330013, China
| | - Shuping Peng
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha 410078, China; NHC Key Laboratory of Carcinogenesis of Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha 410013, China; School of Energy and Machinery Engineering, Jiangxi University of Science and Technology, Nanchang 330013, China.
| | - Wenjing Yang
- Institute of Additive Manufacturing, Jiangxi University of Science and Technology, Nanchang 330013, China.
| |
Collapse
|
26
|
Kontoghiorghes GJ. Deferiprone and Iron-Maltol: Forty Years since Their Discovery and Insights into Their Drug Design, Development, Clinical Use and Future Prospects. Int J Mol Sci 2023; 24:ijms24054970. [PMID: 36902402 PMCID: PMC10002863 DOI: 10.3390/ijms24054970] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 02/24/2023] [Accepted: 03/02/2023] [Indexed: 03/08/2023] Open
Abstract
The historical insights and background of the discovery, development and clinical use of deferiprone (L1) and the maltol-iron complex, which were discovered over 40 years ago, highlight the difficulties, complexities and efforts in general orphan drug development programs originating from academic centers. Deferiprone is widely used for the removal of excess iron in the treatment of iron overload diseases, but also in many other diseases associated with iron toxicity, as well as the modulation of iron metabolism pathways. The maltol-iron complex is a recently approved drug used for increasing iron intake in the treatment of iron deficiency anemia, a condition affecting one-third to one-quarter of the world's population. Detailed insights into different aspects of drug development associated with L1 and the maltol-iron complex are revealed, including theoretical concepts of invention; drug discovery; new chemical synthesis; in vitro, in vivo and clinical screening; toxicology; pharmacology; and the optimization of dose protocols. The prospects of the application of these two drugs in many other diseases are discussed under the light of competing drugs from other academic and commercial centers and also different regulatory authorities. The underlying scientific and other strategies, as well as the many limitations in the present global scene of pharmaceuticals, are also highlighted, with an emphasis on the priorities for orphan drug and emergency medicine development, including the roles of the academic scientific community, pharmaceutical companies and patient organizations.
Collapse
Affiliation(s)
- George J Kontoghiorghes
- Postgraduate Research Institute of Science, Technology, Environment and Medicine, Limassol 3021, Cyprus
| |
Collapse
|
27
|
Osetrov K, Uspenskaya M, Olekhnovich R. The model pH-controlled delivery system based on gelatin-tannin hydrogels containing ferrous ascorbate: iron release in vitro. Biomed Phys Eng Express 2023; 9. [PMID: 36758228 DOI: 10.1088/2057-1976/acbaa1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 02/09/2023] [Indexed: 02/11/2023]
Abstract
Hydrogels have become an essential class among all biomaterials. The specialized biomaterials are highly valued in the field of biomedical applications. One of the problems in wound management is local microelement deficiency associated with extensive wound lesions. The significant lack of elemental iron in the human body leads to serious consequences and prolongs treatment. The synthesis of gelatin-tannin hydrogels with ion delivery function is proposed in this study. The ability to release ions in low acid solution is a sphere of great interest. The pH drop in the wound cavity is usually associated with the contamination of some bacterial cultures. pH-controlled delivery of iron in buffer solutions (рН = 5.5/6.4/7.4) was considered for these hydrogels. The kinetics of iron release was determined by visible spectroscopy. Theoretical models were applied to describe the process of ion delivery. The structure of materials was examined by IR-spectroscopy and demonstrated the incorporation of ferrous ascorbate into hydrogel matrix. Thermal analysis was used to point out the key differences in thermal behavior by isoconversional methods (Flynn-Wall-Ozawa/Kissinger-Akahira-Sunose). The mechanical properties of the materials have been studied. The effect of iron ascorbate on polymer network parameters was discussed. The current study demonstrated the possibility of obtaining gelatin-tannin hydrogels for pH-dependent iron delivery. That provides future perspectives to expand the set of releasing microelements for biomedical applications.
Collapse
Affiliation(s)
- Konstantin Osetrov
- Center for Chemical Engineering, ITMO University, 197101, Saint-Petersburg, Russia
| | - Mayya Uspenskaya
- Center for Chemical Engineering, ITMO University, 197101, Saint-Petersburg, Russia
| | - Roman Olekhnovich
- Center for Chemical Engineering, ITMO University, 197101, Saint-Petersburg, Russia
| |
Collapse
|
28
|
The Combined Administration of Vitamin C and Copper Induces a Systemic Oxidative Stress and Kidney Injury. Biomolecules 2023; 13:biom13010143. [PMID: 36671529 PMCID: PMC9856059 DOI: 10.3390/biom13010143] [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: 01/05/2023] [Accepted: 01/07/2023] [Indexed: 01/12/2023] Open
Abstract
Vitamin C (ascorbic acid; AA) and copper (Cu2+) are well used supplements with many health-promoting actions. However, when they are used in combination, the Fenton reaction occurs, leading to the formation of highly reactive hydroxyl radicals. Given that kidney is vulnerable to many toxicants including free radicals, we speculated that the in vivo administration of AA plus Cu2+ may cause oxidative kidney injury. The purpose of this study was to address this possibility. Mice were administered with AA and Cu2+, alone or in combination, via oral gavage once a day for various periods. Changes in the systemic oxidative status, as well renal structure and functions, were examined. The administration of AA plus Cu2+ elevated protein oxidation in serum, intestine, bladder, and kidney, as evidenced by the increased sulfenic acid formation and decreased level of free sulfhydryl groups (-SH). The systemic oxidative stress induced by AA plus Cu2+ was associated with a significant loss of renal function and structure, as indicated by the increased blood urea nitrogen (BUN), creatinine and urinary proteins, as well as glomerular and tubular cell injury. These effects of AA and Cu2+ were only observed when used in combination, and could be entirely prevented by thiol antioxidant NAC. Further analysis using cultured renal tubular epithelial cells revealed that AA plus Cu2+ caused cellular protein oxidation and cell death, which could be abolished by NAC and catalase. Moreover, coincubation of AA and Cu2+ led to H2O2 production. Collectively, our study revealed that a combined administration of AA and Cu2+ resulted in systemic oxidative stress and renal cell injury. As health-promoting supplements, AA and Cu2+ should not be used together.
Collapse
|
29
|
Correlation between Perturbation of Redox Homeostasis and Antibiofilm Capacity of Phytochemicals at Non-Lethal Concentrations. Antioxidants (Basel) 2022; 11:antiox11122451. [PMID: 36552659 PMCID: PMC9774353 DOI: 10.3390/antiox11122451] [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: 10/26/2022] [Revised: 12/05/2022] [Accepted: 12/08/2022] [Indexed: 12/15/2022] Open
Abstract
Biofilms are the multicellular lifestyle of microorganisms and are present on potentially every type of biotic or abiotic surface. Detrimental biofilms are generally targeted with antimicrobial compounds. Phytochemicals at sub-lethal concentrations seem to be an exciting alternative strategy to control biofilms, as they are less likely to impose selective pressure leading to resistance. This overview gathers the literature on individual phytocompounds rather than on extracts of which the use is difficult to reproduce. To the best of our knowledge, this is the first review to target only individual phytochemicals below inhibitory concentrations against biofilm formation. We explored whether there is an overall mechanism that can explain the effects of individual phytochemicals at sub-lethal concentrations. Interestingly, in all experiments reported here in which oxidative stress was investigated, a modest increase in intracellular reactive oxygen species was reported in treated cells compared to untreated specimens. At sub-lethal concentrations, polyphenolic substances likely act as pro-oxidants by disturbing the healthy redox cycle and causing an accumulation of reactive oxygen species.
Collapse
|
30
|
Comparison of bioavailability and transporters gene expression of four iron fortificants added to infant cereals. FOOD BIOSCI 2022. [DOI: 10.1016/j.fbio.2022.102023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
31
|
New Iron Metabolic Pathways and Chelation Targeting Strategies Affecting the Treatment of All Types and Stages of Cancer. Int J Mol Sci 2022; 23:ijms232213990. [PMID: 36430469 PMCID: PMC9696688 DOI: 10.3390/ijms232213990] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/02/2022] [Accepted: 11/09/2022] [Indexed: 11/16/2022] Open
Abstract
There is new and increasing evidence from in vitro, in vivo and clinical studies implicating the pivotal role of iron and associated metabolic pathways in the initiation, progression and development of cancer and in cancer metastasis. New metabolic and toxicity mechanisms and pathways, as well as genomic, transcription and other factors, have been linked to cancer and many are related to iron. Accordingly, a number of new targets for iron chelators have been identified and characterized in new anticancer strategies, in addition to the classical restriction of/reduction in iron supply, the inhibition of transferrin iron delivery, the inhibition of ribonucleotide reductase in DNA synthesis and high antioxidant potential. The new targets include the removal of excess iron from iron-laden macrophages, which affects anticancer activity; the modulation of ferroptosis; ferritin iron removal and the control of hyperferritinemia; the inhibition of hypoxia related to the role of hypoxia-inducible factor (HIF); modulation of the function of new molecular species such as STEAP4 metalloreductase and the metastasis suppressor N-MYC downstream-regulated gene-1 (NDRG1); modulation of the metabolic pathways of oxidative stress damage affecting mitochondrial function, etc. Many of these new, but also previously known associated iron metabolic pathways appear to affect all stages of cancer, as well as metastasis and drug resistance. Iron-chelating drugs and especially deferiprone (L1), has been shown in many recent studies to fulfill the role of multi-target anticancer drug linked to the above and also other iron targets, and has been proposed for phase II trials in cancer patients. In contrast, lipophilic chelators and their iron complexes are proposed for the induction of ferroptosis in some refractory or recurring tumors in drug resistance and metastasis where effective treatments are absent. There is a need to readdress cancer therapy and include therapeutic strategies targeting multifactorial processes, including the application of multi-targeting drugs involving iron chelators and iron-chelator complexes. New therapeutic protocols including drug combinations with L1 and other chelating drugs could increase anticancer activity, decrease drug resistance and metastasis, improve treatments, reduce toxicity and increase overall survival in cancer patients.
Collapse
|
32
|
Engelhardt L, Pöhnl T, Neugart S. Edible Wild Vegetables Urtica dioica L. and Aegopodium podagraria L.-Antioxidants Affected by Processing. PLANTS (BASEL, SWITZERLAND) 2022; 11:plants11202710. [PMID: 36297734 PMCID: PMC9610176 DOI: 10.3390/plants11202710] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/14/2022] [Accepted: 09/28/2022] [Indexed: 05/26/2023]
Abstract
Urtica dioica L. and Aegopodium podagraria L., also known as stinging nettle and ground elder, are edible wild green vegetables rich in bioactive and antioxidant polyphenols, vitamins, and minerals. Antioxidant activity assays (TEAC-, DPPH-, and TPC-assay) in combination with HPLC measurements, to qualify and quantify their chemical compositions, were used. Firstly, the drying methods affected the antioxidant activity of further processing stages, and outcomes were dependent on the species. Secondly, cooking increased the antioxidant activity due to higher concentrations of bioactive compounds, and released bound compounds through the rupture of cell structures. Furthermore, fridge storage (3 days at 7 °C) resulted in the lowest antioxidant activity, compared to freezer storage (30 days at -20 °C). Added 5-caffeoylquinic acid (0.3 mM) led to an increased antioxidant activity, most noticeably in freeze-dried samples. Synergistic effects of 5-caffeoylquinic acid were primary found in freeze-dried samples, analyzed fresh or after storage in the fridge. Metal-chelates can lower the antioxidant activity in plant matrices. Edible wild green vegetables are rich in polyphenols and processing can even increase their concentrations to boost the potential health effects. In general, selected quantified phenolics are not solely responsible for the antioxidant activity; minerals, processing, and interactions in plant matrices also contribute decisively.
Collapse
|
33
|
Murillo MI, Gaiddon C, Le Lagadec R. Targeting of the intracellular redox balance by metal complexes towards anticancer therapy. Front Chem 2022; 10:967337. [PMID: 36034648 PMCID: PMC9405673 DOI: 10.3389/fchem.2022.967337] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Accepted: 06/29/2022] [Indexed: 11/13/2022] Open
Abstract
The development of cancers is often linked to the alteration of essential redox processes, and therefore, oxidoreductases involved in such mechanisms can be considered as attractive molecular targets for the development of new therapeutic strategies. On the other hand, for more than two decades, transition metals derivatives have been leading the research on drugs as alternatives to platinum-based treatments. The success of such compounds is particularly due to their attractive redox kinetics properties, favorable oxidation states, as well as routes of action different to interactions with DNA, in which redox interactions are crucial. For instance, the activity of oxidoreductases such as PHD2 (prolyl hydroxylase domain-containing protein) which can regulate angiogenesis in tumors, LDH (lactate dehydrogenase) related to glycolysis, and enzymes, such as catalases, SOD (superoxide dismutase), TRX (thioredoxin) or GSH (glutathione) involved in controlling oxidative stress, can be altered by metal effectors. In this review, we wish to discuss recent results on how transition metal complexes have been rationally designed to impact on redox processes, in search for effective and more specific cancer treatments.
Collapse
Affiliation(s)
- María Isabel Murillo
- Instituto de Química, Universidad Nacional Autónoma de México, Ciudad Universitaria, Ciudad de México, Mexico
| | - Christian Gaiddon
- Strasbourg Université, Inserm UMR_S U1113, IRFAC, Strasbourg, France
| | - Ronan Le Lagadec
- Instituto de Química, Universidad Nacional Autónoma de México, Ciudad Universitaria, Ciudad de México, Mexico
- *Correspondence: Ronan Le Lagadec,
| |
Collapse
|
34
|
San-Martín-Martínez D, Serrano-Lemus D, Cornejo V, Gajardo AIJ, Rodrigo R. Pharmacological Basis for Abrogating Myocardial Reperfusion Injury Through a Multi-Target Combined Antioxidant Therapy. Clin Pharmacokinet 2022; 61:1203-1218. [DOI: 10.1007/s40262-022-01151-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/29/2022] [Indexed: 11/29/2022]
|
35
|
Kontoghiorghes GJ. Deferiprone: A Forty-Year-Old Multi-Targeting Drug with Possible Activity against COVID-19 and Diseases of Similar Symptomatology. Int J Mol Sci 2022; 23:ijms23126735. [PMID: 35743183 PMCID: PMC9223898 DOI: 10.3390/ijms23126735] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/13/2022] [Accepted: 06/14/2022] [Indexed: 02/01/2023] Open
Abstract
The need for preparing new strategies for the design of emergency drug therapies against COVID-19 and similar diseases in the future is rather urgent, considering the high rate of morbidity and especially mortality associated with COVID-19, which so far has exceeded 18 million lives. Such strategies could be conceived by targeting the causes and also the serious toxic side effects of the diseases, as well as associated biochemical and physiological pathways. Deferiprone (L1) is an EMA- and FDA-approved drug used worldwide for the treatment of iron overload and also other conditions where there are no effective treatments. The multi-potent effects and high safety record of L1 in iron loaded and non-iron loaded categories of patients suggests that L1 could be developed as a “magic bullet” drug against COVID-19 and diseases of similar symptomatology. The mode of action of L1 includes antiviral, antimicrobial, antioxidant, anti-hypoxic and anti-ferroptotic effects, iron buffering interactions with transferrin, iron mobilizing effects from ferritin, macrophages and other cells involved in the immune response and hyperinflammation, as well as many other therapeutic interventions. Similarly, several pharmacological and other characteristics of L1, including extensive tissue distribution and low cost of production, increase the prospect of worldwide availability, as well as many other therapeutic approach strategies involving drug combinations, adjuvant therapies and disease prevention.
Collapse
Affiliation(s)
- George J Kontoghiorghes
- Postgraduate Research Institute of Science, Technology, Environment and Medicine, Limassol 3021, Cyprus
| |
Collapse
|
36
|
Selyutina OY, Kononova PA, Koshman VE, Fedenok LG, Polyakov NE. The Interplay of Ascorbic Acid with Quinones-Chelators—Influence on Lipid Peroxidation: Insight into Anticancer Activity. Antioxidants (Basel) 2022; 11:antiox11020376. [PMID: 35204258 PMCID: PMC8869476 DOI: 10.3390/antiox11020376] [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: 12/14/2021] [Revised: 01/24/2022] [Accepted: 02/11/2022] [Indexed: 12/11/2022] Open
Abstract
Ascorbic acid is a multifaceted compound that can perform both antioxidant and pro-oxidant activities in the redox reactions induced by transition metal ions, so its role in nature and especially in the human body is still the subject of debate. In the present study, we have examined the influence of ascorbic acid on lipid peroxidation in a model system that mimics the cell membrane, namely micelles of linoleic acid (LA), induced by chelate complexes of iron and copper ions with quinone-chelator 2-phenyl-4-(butylamino)-naphtholquinoline-7,12-dione (Q1). This quinone effectively generates reactive oxygen species and semiquinone radicals inside cancer cells via a cycling redox reaction. Here it was demonstrated that in the absence of quinone-chelator ascorbic acid significantly accelerates the lipid peroxidation induced by both Fe(II) and Cu(II) ions. It has been shown also that Q1 chelate complexes with Fe(II) and Cu(II) ions are redox active in the LA micelles oxidation. No effect of ascorbate was detected on the reactivity of chelate complex with Fe(II) ions. On the other hand, ascorbate performs pro-oxidant activity in Q1-Cu(II) complex induced reaction. We can conclude that ascorbate-driven redox cycling of Q1 may promote its anti-tumor activity.
Collapse
|
37
|
Mechanistic Insights of Chelator Complexes with Essential Transition Metals: Antioxidant/Pro-Oxidant Activity and Applications in Medicine. Int J Mol Sci 2022; 23:ijms23031247. [PMID: 35163169 PMCID: PMC8835618 DOI: 10.3390/ijms23031247] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/13/2022] [Accepted: 01/20/2022] [Indexed: 12/24/2022] Open
Abstract
The antioxidant/pro-oxidant activity of drugs and dietary molecules and their role in the maintenance of redox homeostasis, as well as the implications in health and different diseases, have not yet been fully evaluated. In particular, the redox activity and other interactions of drugs with essential redox metal ions, such as iron and copper, need further investigation. These metal ions are ubiquitous in human nutrition but also widely found in dietary supplements and appear to exert major effects on redox homeostasis in health, but also on many diseases of free radical pathology. In this context, the redox mechanistic insights of mainly three prototype groups of drugs, namely alpha-ketohydroxypyridines (alpha-hydroxypyridones), e.g., deferiprone, anthraquinones, e.g., doxorubicin and thiosemicarbazones, e.g., triapine and their metal complexes were examined; details of the mechanisms of their redox activity were reviewed, with emphasis on the biological implications and potential clinical applications, including anticancer activity. Furthermore, the redox properties of these three classes of chelators were compared to those of the iron chelating drugs and also to vitamin C, with an emphasis on their potential clinical interactions and future clinical application prospects in cancer, neurodegenerative and other diseases.
Collapse
|
38
|
Selyutina OY, Kononova PA, Koshman VE, Shelepova EA, Azad MG, Afroz R, Dharmasivam M, Bernhardt PV, Polyakov NE, Richardson DR. Ascorbate-and iron-driven redox activity of Dp44mT and emodin facilitates peroxidation of micelles and bicelles. Biochim Biophys Acta Gen Subj 2021; 1866:130078. [PMID: 34974127 DOI: 10.1016/j.bbagen.2021.130078] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/10/2021] [Accepted: 12/20/2021] [Indexed: 12/28/2022]
Abstract
BACKGROUND Iron (Fe)-induced oxidative stress leads to reactive oxygen species that damage biomembranes, with this mechanism being involved in the activity of some anti-cancer chemotherapeutics. METHODS Herein, we compared the effect of Fe complexes of the ligand, di-2-pyridylketone 4,4-dimethyl-3-thiosemicarbazone (Dp44mT), or the potential ligand, Emodin, on lipid peroxidation in cell membrane models (micelles and bicelles). These studies were performed in the presence of hydrogen peroxide (H2O2) and the absence or presence of ascorbate. RESULTS In the absence of ascorbate, Fe(II)/Emodin mixtures incubated with H2O2 demonstrated slight pro-oxidant properties on micelles versus Fe(II) alone, while the Fe(III)-Dp44mT complex exhibited marked antioxidant properties. Examining more physiologically relevant phospholipid-containing bicelles, the Fe(II)- and Fe(III)-Dp44mT complexes demonstrated antioxidant activity without ascorbate. Upon adding ascorbate, there was a significant increase in the peroxidation of micelles and bicelles in the presence of unchelated Fe(II) and H2O2. The addition of ascorbate to Fe(III)-Dp44mT substantially increased the peroxidation of micelles and bicelles, with the Fe(III)-Dp44mT complex being reduced by ascorbate to the Fe(II) state, explaining the increased reactivity. Electron paramagnetic resonance spectroscopy demonstrated ascorbyl radical anion generation after mixing ascorbate and Emodin, with signal intensity being enhanced by H2O2. This finding suggested Emodin semiquinone radical formation that could play a role in its reactivity via ascorbate-driven redox cycling. Examining cultured melanoma cells in vitro, ascorbate at pharmacological levels enhanced the anti-proliferative activity of Dp44mT and Emodin. CONCLUSIONS AND GENERAL SIGNIFICANCE Ascorbate-driven redox cycling of Dp44mT and Emodin promotes their anti-proliferative activity.
Collapse
Affiliation(s)
- O Yu Selyutina
- Institute of Chemical Kinetics and Combustion, Institutskaya St., 3, 630090 Novosibirsk, Russia; Institute of Solid State Chemistry and Mechanochemistry, Kutateladze St., 18, 630128 Novosibirsk, Russia.
| | - P A Kononova
- Institute of Chemical Kinetics and Combustion, Institutskaya St., 3, 630090 Novosibirsk, Russia
| | - V E Koshman
- Institute of Chemical Kinetics and Combustion, Institutskaya St., 3, 630090 Novosibirsk, Russia
| | - E A Shelepova
- Institute of Chemical Kinetics and Combustion, Institutskaya St., 3, 630090 Novosibirsk, Russia
| | - M Gholam Azad
- Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland 4111, Australia
| | - R Afroz
- Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland 4111, Australia
| | - M Dharmasivam
- Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland 4111, Australia
| | - P V Bernhardt
- Department of Chemistry, University of Queensland, St. Lucia, Brisbane, Queensland 4072, Australia
| | - N E Polyakov
- Institute of Chemical Kinetics and Combustion, Institutskaya St., 3, 630090 Novosibirsk, Russia; Institute of Solid State Chemistry and Mechanochemistry, Kutateladze St., 18, 630128 Novosibirsk, Russia
| | - D R Richardson
- Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland 4111, Australia; Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan.
| |
Collapse
|
39
|
Interactions of Ascorbic Acid, 5-Caffeoylquinic Acid, and Quercetin-3-Rutinoside in the Presence and Absence of Iron during Thermal Processing and the Influence on Antioxidant Activity. Molecules 2021; 26:molecules26247698. [PMID: 34946775 PMCID: PMC8706688 DOI: 10.3390/molecules26247698] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/10/2021] [Accepted: 12/12/2021] [Indexed: 01/07/2023] Open
Abstract
Bioactive compounds in fruit and vegetables influence each other’s antioxidant activity. Pure standards, and mixtures of the common plant compounds, namely ascorbic acid, 5-caffeoylquinic acid, and quercetin-3-rutinoside (sum 0.3 mM), in the presence and absence of iron, were analyzed pre- and post-thermal processing in an aqueous solution. Antioxidant activity was measured by total phenolic content (TPC), 1,1-diphenyl-2-picrylhydrazyl (DPPH), and 2,2′-azino-bis (3-ethylbenzothiazoline-6-sulfonic acid) (TEAC) radical-scavenging assays. Ionic ferrous iron (Fe2+) and ferric iron (Fe3+) were measured photometrically. For qualification and quantification of reaction products, HPLC was used. Results showed that thermal processing does not necessarily lead to a decreased antioxidant activity, even if the compound concentrations decreased, as then degradation products themselves have an antioxidant activity. In all used antioxidant assays the 2:1 ratio of ascorbic acid and 5-caffeoylquinic acid in the presence of iron had strong synergistic effects, while the 1:2 ratio had strong antagonistic effects. The pro-oxidant iron positively influenced the antioxidant activity in combination with the used antioxidants, while ferrous iron itself interacted with common in vitro assays for total antioxidant activity. These results indicate that the antioxidant activity of compounds is influenced by factors such as interaction with other molecules, temperature, and the minerals present.
Collapse
|
40
|
Abstract
Significance: Vitamin C (ascorbate), in regard to its effectiveness against malignancies, has had a controversial history in cancer treatment. It has been shown that in vitro and in vivo anticancer efficacy of ascorbate relies on its pro-oxidant effect mainly from an increased generation of reactive oxygen species (ROS). A growing understanding of its anticancer activities and pharmacokinetic properties has prompted scientists to re-evaluate the significance of ascorbate in cancer treatment. Recent Advances: A recent resurge in ascorbate research emerged after discovering that, at high doses, ascorbate preferentially kills Kirsten-Ras (K-ras)- and B-raf oncogene (BRAF)-mutant cancer cells. In addition, some of the main hallmarks of cancer cells, such as redox homeostasis and oxygen-sensing regulation (through inhibition of hypoxia-inducible factor-1 alpha [HIF-1α] activity), are affected by vitamin C. Critical Issues: Currently, there is no clear consensus from the literature in regard to the beneficial effects of antioxidants. Results from both human and animal studies provide no clear evidence about the benefit of antioxidant treatment in preventing or suppressing cancer development. Since pro-oxidants may affect both normal and tumor cells, the extremely low toxicity of ascorbate represents a main advantage. This guarantees the safe inclusion of ascorbate in clinical protocols to treat cancer patients. Future Directions: Current research could focus on elucidating the wide array of reactions between ascorbate and reactive species, namely ROS, reactive nitrogen species as well as reactive sulfide species, and their intracellular molecular targets. Unraveling these mechanisms could allow researchers to assess what could be the optimal combination of ascorbate with standard treatments.
Collapse
Affiliation(s)
- Christophe Glorieux
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, P. R. China
| | - Pedro Buc Calderon
- Química y Farmacia, Facultad de Ciencias de la Salud, Universidad Arturo Prat, Iquique, Chile.,Research Group in Metabolism and Nutrition, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| |
Collapse
|
41
|
Timoshnikov VA, Kichigina LA, Selyutina OY, Polyakov NE, Kontoghiorghes GJ. Antioxidant Activity of Deferasirox and Its Metal Complexes in Model Systems of Oxidative Damage: Comparison with Deferiprone. Molecules 2021; 26:molecules26165064. [PMID: 34443652 PMCID: PMC8401497 DOI: 10.3390/molecules26165064] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 08/17/2021] [Accepted: 08/18/2021] [Indexed: 12/28/2022] Open
Abstract
Deferasirox is an orally active, lipophilic iron chelating drug used on thousands of patients worldwide for the treatment of transfusional iron overload. The essential transition metals iron and copper are the primary catalysts of reactive oxygen species and oxidative damage in biological systems. The redox effects of deferasirox and its metal complexes with iron, copper and other metals are of pharmacological, toxicological, biological and physiological importance. Several molecular model systems of oxidative damage caused by iron and copper catalysis including the oxidation of ascorbic acid, the peroxidation of linoleic acid micelles and the oxidation of dihydropyridine have been investigated in the presence of deferasirox using UV-visible and NMR spectroscopy. Deferasirox has shown antioxidant activity in all three model systems, causing substantial reduction in the rate of oxidation and oxidative damage. Deferasirox showed the greatest antioxidant activity in the oxidation of ascorbic acid with the participation of iron ions and reduced the reaction rate by about a 100 times. Overall, deferasirox appears to have lower affinity for copper in comparison to iron. Comparative studies of the antioxidant activity of deferasirox and the hydrophilic oral iron chelating drug deferiprone in the peroxidation of linoleic acid micelles showed lower efficiency of deferasirox in comparison to deferiprone.
Collapse
Affiliation(s)
- Viktor A. Timoshnikov
- Institute of Chemical Kinetics & Combustion, 630090 Novosibirsk, Russia; (V.A.T.); (L.A.K.); (O.Y.S.); (N.E.P.)
| | - Lilia A. Kichigina
- Institute of Chemical Kinetics & Combustion, 630090 Novosibirsk, Russia; (V.A.T.); (L.A.K.); (O.Y.S.); (N.E.P.)
| | - Olga Yu. Selyutina
- Institute of Chemical Kinetics & Combustion, 630090 Novosibirsk, Russia; (V.A.T.); (L.A.K.); (O.Y.S.); (N.E.P.)
| | - Nikolay E. Polyakov
- Institute of Chemical Kinetics & Combustion, 630090 Novosibirsk, Russia; (V.A.T.); (L.A.K.); (O.Y.S.); (N.E.P.)
| | - George J. Kontoghiorghes
- Postgraduate Research Institute of Science, Technology, Environment and Medicine, Limassol CY-3021, Cyprus
- Correspondence:
| |
Collapse
|
42
|
Rochette L, Ghibu S. Mechanics Insights of Alpha-Lipoic Acid against Cardiovascular Diseases during COVID-19 Infection. Int J Mol Sci 2021; 22:7979. [PMID: 34360751 PMCID: PMC8348748 DOI: 10.3390/ijms22157979] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 07/19/2021] [Accepted: 07/20/2021] [Indexed: 02/06/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) was first reported in Wuhan, China, in late December 2019. Since then, COVID-19 has spread rapidly worldwide and was declared a global pandemic on 20 March 2020. Cardiovascular complications are rapidly emerging as a major peril in COVID-19 in addition to respiratory disease. The mechanisms underlying the excessive effect of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection on patients with cardiovascular comorbidities remain only partly understood. SARS-CoV-2 infection is caused by binding of the viral surface spike (S) protein to the human angiotensin-converting enzyme 2 (ACE2), followed by the activation of the S protein by transmembrane protease serine 2 (TMPRSS2). ACE2 is expressed in the lung (mainly in type II alveolar cells), heart, blood vessels, small intestine, etc., and appears to be the predominant portal to the cellular entry of the virus. Based on current information, most people infected with SARS-CoV-2 virus have a good prognosis, while a few patients reach critical condition, especially the elderly and those with chronic underlying diseases. The "cytokine storm" observed in patients with severe COVID-19 contributes to the destruction of the endothelium, leading to "acute respiratory distress syndrome" (ARDS), multiorgan failure, and death. At the origin of the general proinflammatory state may be the SARS-CoV-2-mediated redox status in endothelial cells via the upregulation of ACE/Ang II/AT1 receptors pathway or the increased mitochondrial reactive oxygen species (mtROS) production. Furthermore, this vicious circle between oxidative stress (OS) and inflammation induces endothelial dysfunction, endothelial senescence, high risk of thrombosis and coagulopathy. The microvascular dysfunction and the formation of microthrombi in a way differentiate the SARS-CoV-2 infection from the other respiratory diseases and bring it closer to cardiovascular diseases like myocardial infarction and stroke. Due the role played by OS in the evolution of viral infection and in the development of COVID-19 complications, the use of antioxidants as adjuvant therapy seems appropriate in this new pathology. Alpha-lipoic acid (ALA) could be a promising candidate that, through its wide tissue distribution and versatile antioxidant properties, interferes with several signaling pathways. Thus, ALA improves endothelial function by restoring the endothelial nitric oxide synthase activity and presents an anti-inflammatory effect dependent or independent of its antioxidant properties. By improving mitochondrial function, it can sustain the tissues' homeostasis in critical situation and by enhancing the reduced glutathione it could indirectly strengthen the immune system. This complex analysis could open a new therapeutic perspective for ALA in COVID-19 infection.
Collapse
Affiliation(s)
- Luc Rochette
- Equipe d’Accueil (EA 7460), Physiopathologie et Epidémiologie Cérébro-Cardiovasculaires (PEC2), Faculté des Sciences de Santé, Université de Bourgogne-Franche Comté, 21000 Dijon, France;
| | - Steliana Ghibu
- Department of Pharmacology, Physiology and Pathophysiology, Faculty of Pharmacy, “Iuliu Haţieganu” University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| |
Collapse
|
43
|
Conventional and Unconventional Approaches for Innovative Drug Treatments in COVID-19: Looking Outside of Plato's Cave. Int J Mol Sci 2021; 22:ijms22137208. [PMID: 34281262 PMCID: PMC8268874 DOI: 10.3390/ijms22137208] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 06/29/2021] [Indexed: 11/17/2022] Open
|
44
|
DE FLORA SILVIO, BALANSKY ROUMEN, LA MAESTRA SEBASTIANO. Antioxidants and COVID-19. JOURNAL OF PREVENTIVE MEDICINE AND HYGIENE 2021; 62:E34-E45. [PMID: 34622082 PMCID: PMC8452284 DOI: 10.15167/2421-4248/jpmh2021.62.1s3.1895] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 02/03/2021] [Indexed: 12/24/2022]
Abstract
Oxidative mechanisms are not only involved in chronic degenerative diseases but also in infectious diseases, among which viral respiratory diseases. Antioxidants have the capability to counteract the action of oxidants by scavenging reactive oxygen species (ROS) and by inhibiting oxidant generating enzymes. Overproduction of ROS and deprivation of antioxidant systems play a major role in COVID-19 occurrence, progression, and severity. Interconnected pathways account for the relationships between oxidative damage and inflammation resulting from an interplay between transcription factors having opposite effects. For instance, Nrf2 downregulates inflammation by inhibiting endogenous antioxidant enzymes such as NQO-1 and HO-1. On the other hand, NF-κB upregulates pro-inflammatory cytokines and chemokines, such as IL-1β, IL-6, IL-8, PGE-2, COX-2, TNF-α, MMP-3, and MMP-4. A central protective role against oxidants is played by reduced glutathione (GSH), which is depleted in SARS-CoV-2 infection. N-acetylcysteine (NAC), a precursor of GSH, is of particular interest as an anti-COVID-19 agent. GSH and NAC hamper binding of the S1 subunit of SARS-CoV-2 spike proteins to the angiotensin-converting enzyme 2 (ACE2) receptor. In addition, NAC and its derivatives possess a broad array of antioxidant and antiinflammatory mechanisms that could be exploited for COVID-19 prevention and adjuvant therapy. In particular, as demonstrated in a previous clinical trial evaluating influenza and influenza-like illnesses, the oral administration of NAC may be expected to decrease the risk of developing COVID-19. Furthermore, at the very high doses used worldwide as an antidote against paracetamol intoxication, intravenous NAC is likely to attenuate the pulmonary and systemic symptoms of COVID-19.
Collapse
Affiliation(s)
| | | | - SEBASTIANO LA MAESTRA
- Department of Health Sciences, University of Genoa, Italy
- Correspondence: Sebastiano La Maestra, Department of Health Sciences, University of Genoa, via A. Pastore 1, 16132 Genoa, Italy – E-mail:
| |
Collapse
|
45
|
Deferoxamine B: A Natural, Excellent and Versatile Metal Chelator. Molecules 2021; 26:molecules26113255. [PMID: 34071479 PMCID: PMC8198152 DOI: 10.3390/molecules26113255] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 05/25/2021] [Accepted: 05/27/2021] [Indexed: 02/06/2023] Open
Abstract
Deferoxamine B is an outstanding molecule which has been widely studied in the past decade for its ability to bind iron and many other metal ions. The versatility of this metal chelator makes it suitable for a number of medicinal and analytical applications, from the well-known iron chelation therapy to the most recent use in sensor devices. The three bidentate hydroxamic functional groups of deferoxamine B are the centerpiece of its metal binding ability, which allows the formation of stable complexes with many transition, lanthanoid and actinoid metal ions. In addition to the ferric ion, in fact, more than 20 different metal complexes of deferoxamine b have been characterized in terms of their chemical speciation in solution. In addition, the availability of a terminal amino group, most often not involved in complexation, opens the way to deferoxamine B modification and functionalization. This review aims to collect and summarize the available data concerning the complex-formation equilibria in solutions of deferoxamine B with different metal ions. A general overview of the progress of its applications over the past decade is also discussed, including the treatment of iron overload-associated diseases, its clinical use against cancer and neurodegenerative disorders and its role as a diagnostic tool.
Collapse
|
46
|
Kontoghiorghes GJ, Kolnagou A, Demetriou T, Neocleous M, Kontoghiorghe CN. New Era in the Treatment of Iron Deficiency Anaemia Using Trimaltol Iron and Other Lipophilic Iron Chelator Complexes: Historical Perspectives of Discovery and Future Applications. Int J Mol Sci 2021; 22:ijms22115546. [PMID: 34074010 PMCID: PMC8197347 DOI: 10.3390/ijms22115546] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/08/2021] [Accepted: 05/18/2021] [Indexed: 12/11/2022] Open
Abstract
The trimaltol iron complex (International Non-proprietary Name: ferric maltol) was originally designed, synthesised, and screened in vitro and in vivo in 1980–1981 by Kontoghiorghes G.J. following his discovery of the novel alpha-ketohydroxyheteroaromatic (KHP) class of iron chelators (1978–1981), which were intended for clinical use, including the treatment of iron deficiency anaemia (IDA). Iron deficiency anaemia is a global health problem affecting about one-third of the world’s population. Many (and different) ferrous and ferric iron complex formulations are widely available and sold worldwide over the counter for the treatment of IDA. Almost all such complexes suffer from instability in the acidic environment of the stomach and competition from other dietary molecules or drugs. Natural and synthetic lipophilic KHP chelators, including maltol, have been shown in in vitro and in vivo studies to form stable iron complexes, to transfer iron across cell membranes, and to increase iron absorption in animals. Trimaltol iron, sold as Feraccru or Accrufer, was recently approved for clinical use in IDA patients in many countries, including the USA and in EU countries, and was shown to be effective and safe, with a better therapeutic index in comparison to other iron formulations. Similar properties of increased iron absorption were also shown by lipophilic iron complexes of 8-hydroxyquinoline, tropolone, 2-hydroxy-4-methoxypyridine-1-oxide, and related analogues. The interactions of the KHP iron complexes with natural chelators, drugs, metal ions, proteins, and other molecules appear to affect the pharmacological and metabolic effects of both iron and the KHP chelators. A new era in the treatment of IDA and other possible clinical applications, such as theranostic and anticancer formulations and metal radiotracers in diagnostic medicine, are envisaged from the introduction of maltol, KHP, and similar lipophilic chelators.
Collapse
|
47
|
Nowak M, Tryniszewski W, Sarniak A, Wlodarczyk A, Nowak PJ, Nowak D. Effect of Physiological Concentrations of Vitamin C on the Inhibitation of Hydroxyl Radical Induced Light Emission from Fe 2+-EGTA-H 2O 2 and Fe 3+-EGTA-H 2O 2 Systems In Vitro. Molecules 2021; 26:molecules26071993. [PMID: 33915907 PMCID: PMC8037725 DOI: 10.3390/molecules26071993] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/18/2021] [Accepted: 03/22/2021] [Indexed: 12/20/2022] Open
Abstract
Ascorbic acid (AA) has antioxidant properties. However, in the presence of Fe2+/Fe3+ ions and H2O2, it may behave as a pro-oxidant by accelerating and enhancing the formation of hydroxyl radicals (•OH). Therefore, in this study we evaluated the effect of AA at concentrations of 1 to 200 µmol/L on •OH-induced light emission (at a pH of 7.4 and temperature of 37 °C) from 92.6 µmol/L Fe2+—185.2 µmol/L EGTA (ethylene glycol-bis (β-aminoethyl ether)-N,N,N′,N′-tetraacetic acid)—2.6 mmol/L H2O2, and 92.6 µmol/L Fe3+—185.2 µmol/L EGTA—2.6 mmol/L H2O2 systems. Dehydroascorbic acid (DHAA) at the same range of concentrations served as the reference compound. Light emission was measured with multitube luminometer (AutoLumat Plus LB 953) for 120 s after automatic injection of H2O2. AA at concentrations of 1 to 50 µmol/L and of 1 to 75 µmol/L completely inhibited light emission from Fe2+-EGTA-H2O2 and Fe3+-EGTA-H2O2, respectively. Concentrations of 100 and 200 µmol/L did not affect chemiluminescence of Fe3+-EGTA-H2O2 but tended to increase light emission from Fe2+-EGTA-H2O2. DHAA at concentrations of 1 to 100 µmol/L had no effect on chemiluminescence of both systems. These results indicate that AA at physiological concentrations exhibits strong antioxidant activity in the presence of chelated iron and H2O2.
Collapse
Affiliation(s)
- Michal Nowak
- Radiation Protection, University Hospital No. 2, Medical University of Lodz, Zeromskiego 113, 90-549 Lodz, Poland;
| | - Wieslaw Tryniszewski
- Department of Radiological and Isotopic Diagnostics and Therapy, Medical University of Lodz, Zeromskiego 113, 90-549 Lodz, Poland;
| | - Agata Sarniak
- Department of General Physiology, Medical University of Lodz, Mazowiecka 6/8, 92-215 Lodz, Poland;
| | - Anna Wlodarczyk
- Department of Sleep Medicine and Metabolic Disorders, Medical University of Lodz, Mazowiecka 6/8, 92-215 Lodz, Poland;
| | - Piotr J. Nowak
- Department of Nephrology, Hypertension, and Kidney Transplantation, Medical University of Lodz, Pomorska 251, 92-213 Lodz, Poland;
| | - Dariusz Nowak
- Department of Clinical Physiology, Medical University of Lodz, Mazowiecka 6/8, 92-215 Lodz, Poland
- Correspondence: ; Tel.: +48-422-725-656; Fax: +48-422-725-652
| |
Collapse
|