1
|
Zhou X, An Z, Lei H, Liao H, Guo X. Role of the human cytochrome b561 family in iron metabolism and tumors (Review). Oncol Lett 2025; 29:111. [PMID: 39802312 PMCID: PMC11718626 DOI: 10.3892/ol.2024.14857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 11/25/2024] [Indexed: 01/16/2025] Open
Abstract
The human cytochrome b561 (hCytb561) family consists of electron transfer transmembrane proteins characterized by six conserved α-helical transmembrane domains and two β-type heme cofactors. These proteins contribute to the regulation of iron metabolism and numerous different physiological and pathological processes by recycling ascorbic acid and maintaining iron reductase activity. Key members of this family include cytochrome b561 (CYB561), duodenal CYB561 (Dcytb), lysosomal CYB561 (LCytb), stromal cell-derived receptor 2 (SDR2) and 101F6, which are widely expressed in human tissues and participate in the pathogenesis of several diseases and tumors. They are associated with the promotion or inhibition of tumor growth and progression in various malignancies and are potential therapeutic targets for malignant tumors. The present review summarizes the existing literature regarding the structure of the Cytb561 family, the basic functional characteristics of hCytb561 family members, and the roles of the CYB561, Dcytb, LCytb, SDR2 and 101F6 in various diseases and tumors.
Collapse
Affiliation(s)
- Xiaofeng Zhou
- Pathology Department, Qinghai University Affiliated Hospital, Xining, Qinghai 810001, P.R. China
| | - Zheng An
- Pathology Department, Qinghai Women and Children's Hospital, Xining, Qinghai 810007, P.R. China
| | - Hao Lei
- Graduate School, Qinghai University, Xining, Qinghai 810001, P.R. China
| | - Hongyuan Liao
- Graduate School, Qinghai University, Xining, Qinghai 810001, P.R. China
| | - Xinjian Guo
- Pathology Department, Qinghai University Affiliated Hospital, Xining, Qinghai 810001, P.R. China
| |
Collapse
|
2
|
Haiman ZB, Key A, D’Alessandro A, Palsson BO. RBC-GEM: A genome-scale metabolic model for systems biology of the human red blood cell. PLoS Comput Biol 2025; 21:e1012109. [PMID: 40072998 PMCID: PMC11925312 DOI: 10.1371/journal.pcbi.1012109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 03/20/2025] [Accepted: 02/04/2025] [Indexed: 03/14/2025] Open
Abstract
Advancements with cost-effective, high-throughput omics technologies have had a transformative effect on both fundamental and translational research in the medical sciences. These advancements have facilitated a departure from the traditional view of human red blood cells (RBCs) as mere carriers of hemoglobin, devoid of significant biological complexity. Over the past decade, proteomic analyses have identified a growing number of different proteins present within RBCs, enabling systems biology analysis of their physiological functions. Here, we introduce RBC-GEM, one of the most comprehensive, curated genome-scale metabolic reconstructions of a specific human cell type to-date. It was developed through meta-analysis of proteomic data from 29 studies published over the past two decades resulting in an RBC proteome composed of more than 4,600 distinct proteins. Through workflow-guided manual curation, we have compiled the metabolic reactions carried out by this proteome to form a genome-scale metabolic model (GEM) of the RBC. RBC-GEM is hosted on a version-controlled GitHub repository, ensuring adherence to the standardized protocols for metabolic reconstruction quality control and data stewardship principles. RBC-GEM represents a metabolic network is a consisting of 820 genes encoding proteins acting on 1,685 unique metabolites through 2,723 biochemical reactions: a 740% size expansion over its predecessor. We demonstrated the utility of RBC-GEM by creating context-specific proteome-constrained models derived from proteomic data of stored RBCs for 616 blood donors, and classified reactions based on their simulated abundance dependence. This reconstruction as an up-to-date curated GEM can be used for contextualization of data and for the construction of a computational whole-cell models of the human RBC.
Collapse
Affiliation(s)
- Zachary B. Haiman
- Department of Bioengineering, University of California San Diego, La Jolla, California, United States of America
| | - Alicia Key
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Angelo D’Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Bernhard O. Palsson
- Department of Bioengineering, University of California San Diego, La Jolla, California, United States of America
- Department of Pediatrics, University of California San Diego, La Jolla, California, United States of America
- Bioinformatics and Systems Biology Program, University of California, La Jolla, San Diego, California, United States of America
| |
Collapse
|
3
|
Daraghmeh DN, Karaman R. The Redox Process in Red Blood Cells: Balancing Oxidants and Antioxidants. Antioxidants (Basel) 2024; 14:36. [PMID: 39857370 PMCID: PMC11762794 DOI: 10.3390/antiox14010036] [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: 11/22/2024] [Revised: 12/21/2024] [Accepted: 12/28/2024] [Indexed: 01/27/2025] Open
Abstract
Red blood cells (RBCs) are a vital component of the body's oxygen supply system. In addition to being pro-oxidants, they are also essential components of the body's antioxidant defense mechanism. RBCs are susceptible to both endogenous and exogenous sources of oxidants. Oxyhemoglobin autoxidation is the primary source of endogenous RBC oxidant production, which produces superoxide radicals and hydrogen peroxide. Potent exogenous oxidants from other blood cells and the surrounding endothelium can also enter RBCs. Both enzymatic (like glutathione peroxidase) and non-enzymatic (like glutathione) mechanisms can neutralize oxidants. These systems are generally referred to as oxidant scavengers or antioxidants, and they work to neutralize these harmful molecules (i.e., oxidants). While their antioxidative capabilities are essential to their physiological functions and delivering oxygen to tissues, their pro-oxidant behavior plays a part in several human pathologies. The redox-related changes in RBCs can have an impact on their function and fate. The balance between pro-oxidants and antioxidants determines the oxidative status of cells, which affects signal transduction, differentiation, and proliferation. When pro-oxidant activity exceeds antioxidative capacity, oxidative stress occurs, leading to cytotoxicity. This type of stress has been linked to various pathologies, including hemolytic anemia. This review compiles the most recent literature investigating the connections between RBC redox biochemistry, antioxidants, and diverse disorders.
Collapse
Affiliation(s)
- Dala N. Daraghmeh
- Pharmaceutical Sciences Department, Faculty of Pharmacy, Al-Quds University, Jerusalem P.O. Box 20002, Palestine;
| | | |
Collapse
|
4
|
Wang Y, Lilienfeldt N, Hekimi S. Understanding coenzyme Q. Physiol Rev 2024; 104:1533-1610. [PMID: 38722242 PMCID: PMC11495197 DOI: 10.1152/physrev.00040.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 04/08/2024] [Accepted: 05/01/2024] [Indexed: 08/11/2024] Open
Abstract
Coenzyme Q (CoQ), also known as ubiquinone, comprises a benzoquinone head group and a long isoprenoid side chain. It is thus extremely hydrophobic and resides in membranes. It is best known for its complex function as an electron transporter in the mitochondrial electron transport chain (ETC) but is also required for several other crucial cellular processes. In fact, CoQ appears to be central to the entire redox balance of the cell. Remarkably, its structure and therefore its properties have not changed from bacteria to vertebrates. In metazoans, it is synthesized in all cells and is found in most, and maybe all, biological membranes. CoQ is also known as a nutritional supplement, mostly because of its involvement with antioxidant defenses. However, whether there is any health benefit from oral consumption of CoQ is not well established. Here we review the function of CoQ as a redox-active molecule in the ETC and other enzymatic systems, its role as a prooxidant in reactive oxygen species generation, and its separate involvement in antioxidant mechanisms. We also review CoQ biosynthesis, which is particularly complex because of its extreme hydrophobicity, as well as the biological consequences of primary and secondary CoQ deficiency, including in human patients. Primary CoQ deficiency is a rare inborn condition due to mutation in CoQ biosynthetic genes. Secondary CoQ deficiency is much more common, as it accompanies a variety of pathological conditions, including mitochondrial disorders as well as aging. In this context, we discuss the importance, but also the great difficulty, of alleviating CoQ deficiency by CoQ supplementation.
Collapse
Affiliation(s)
- Ying Wang
- Department of Biology, McGill University, Montreal, Quebec, Canada
| | - Noah Lilienfeldt
- Department of Biology, McGill University, Montreal, Quebec, Canada
| | - Siegfried Hekimi
- Department of Biology, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
5
|
Holst JD, Murphy LG, Gorman MJ, Ragan EJ. Comparison of insect and human cytochrome b561 proteins: Insights into candidate ferric reductases in insects. PLoS One 2023; 18:e0291564. [PMID: 38039324 PMCID: PMC10691727 DOI: 10.1371/journal.pone.0291564] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 11/19/2023] [Indexed: 12/03/2023] Open
Abstract
Cytochrome b561 (cytb561) proteins comprise a family of transmembrane oxidoreductases that transfer single electrons across a membrane. Most eukaryotic species, including insects, possess multiple cytb561 homologs. To learn more about this protein family in insects, we carried out a bioinformatics-based investigation of cytb561 family members from nine species representing eight insect orders. We performed a phylogenetic analysis to classify insect cytb561 ortholog groups. We then conducted sequence analyses and analyzed protein models to predict structural elements that may impact the biological functions and localization of these proteins, with a focus on possible ferric reductase activity. Our study revealed three ortholog groups, designated CG1275, Nemy, and CG8399, and a fourth group of less-conserved genes. We found that CG1275 and Nemy proteins are similar to a human ferric reductase, duodenal cytochrome b561 (Dcytb), and have many conserved amino acid residues that function in substrate binding in Dcytb. Notably, CG1275 and Nemy proteins contain a conserved histidine and other residues that play a role in ferric ion reduction by Dcytb. Nemy proteins were distinguished by a novel cysteine-rich cytoplasmic loop sequence. CG8399 orthologs are similar to a putative ferric reductase in humans, stromal cell-derived receptor 2. Like other members of the CYBDOM class of cytb561 proteins, these proteins contain reeler, DOMON, and cytb561 domains. Drosophila melanogaster CG8399 is the only insect cytb561 with known ferric reductase activity. Our investigation of the DOMON domain in CG8399 proteins revealed a probable heme-binding site and a possible site for ferric reduction. The fourth group includes a subgroup of proteins with a conserved "KXXXXKXH" non-cytoplasmic loop motif that may be a substrate binding site and is present in a potential ferric reductase, human tumor suppressor cytochrome b561. This study provides a foundation for future investigations of the biological functions of cytb561 genes in insects.
Collapse
Affiliation(s)
- Jessica D. Holst
- Department of Chemistry and Biochemistry, Metropolitan State University of Denver, Denver, Colorado, United States of America
| | - Laura G. Murphy
- Department of Chemistry and Biochemistry, Metropolitan State University of Denver, Denver, Colorado, United States of America
| | - Maureen J. Gorman
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, Kansas, United States of America
| | - Emily J. Ragan
- Department of Chemistry and Biochemistry, Metropolitan State University of Denver, Denver, Colorado, United States of America
| |
Collapse
|
6
|
Möller M, Orrico F, Villar S, López AC, Silva N, Donzé M, Thomson L, Denicola A. Oxidants and Antioxidants in the Redox Biochemistry of Human Red Blood Cells. ACS OMEGA 2023; 8:147-168. [PMID: 36643550 PMCID: PMC9835686 DOI: 10.1021/acsomega.2c06768] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 12/09/2022] [Indexed: 06/01/2023]
Abstract
Red blood cells (RBCs) are exposed to both external and internal sources of oxidants that challenge their integrity and compromise their physiological function and supply of oxygen to tissues. Autoxidation of oxyhemoglobin is the main source of endogenous RBC oxidant production, yielding superoxide radical and then hydrogen peroxide. In addition, potent oxidants from other blood cells and the surrounding endothelium can reach the RBCs. Abundant and efficient enzymatic systems and low molecular weight antioxidants prevent most of the damage to the RBCs and also position the RBCs as a sink of vascular oxidants that allow the body to maintain a healthy circulatory system. Among the antioxidant enzymes, the thiol-dependent peroxidase peroxiredoxin 2, highly abundant in RBCs, is essential to keep the redox balance. A great part of the RBC antioxidant activity is supported by an active glucose metabolism that provides reducing power in the form of NADPH via the pentose phosphate pathway. There are several RBC defects and situations that generate oxidative stress conditions where the defense mechanisms are overwhelmed, and these include glucose-6-phosphate dehydrogenase deficiencies (favism), hemoglobinopathies like sickle cell disease and thalassemia, as well as packed RBCs for transfusion that suffer from storage lesions. These oxidative stress-associated pathologies of the RBCs underline the relevance of redox balance in these anucleated cells that lack a mechanism of DNA-inducible antioxidant response and rely on a complex and robust network of antioxidant systems.
Collapse
Affiliation(s)
- Matias
N. Möller
- Laboratorio
de Fisicoquímica Biológica, Instituto de Química
Biológica, Facultad de Ciencias,
Universidad de la República, Montevideo 11400, Uruguay
- Centro
de Investigaciones Biomédicas (CEINBIO), Universidad de la República, Montevideo 11800, Uruguay
| | - Florencia Orrico
- Laboratorio
de Fisicoquímica Biológica, Instituto de Química
Biológica, Facultad de Ciencias,
Universidad de la República, Montevideo 11400, Uruguay
- Centro
de Investigaciones Biomédicas (CEINBIO), Universidad de la República, Montevideo 11800, Uruguay
- Laboratorio
de Enzimología, Instituto de Química Biológica,
Facultad de Ciencias, Universidad de la
República, Montevideo 11400, Uruguay
| | - Sebastián
F. Villar
- Laboratorio
de Fisicoquímica Biológica, Instituto de Química
Biológica, Facultad de Ciencias,
Universidad de la República, Montevideo 11400, Uruguay
- Centro
de Investigaciones Biomédicas (CEINBIO), Universidad de la República, Montevideo 11800, Uruguay
| | - Ana C. López
- Laboratorio
de Fisicoquímica Biológica, Instituto de Química
Biológica, Facultad de Ciencias,
Universidad de la República, Montevideo 11400, Uruguay
- Centro
de Investigaciones Biomédicas (CEINBIO), Universidad de la República, Montevideo 11800, Uruguay
- Laboratorio
de Enzimología, Instituto de Química Biológica,
Facultad de Ciencias, Universidad de la
República, Montevideo 11400, Uruguay
| | - Nicolás Silva
- Laboratorio
de Fisicoquímica Biológica, Instituto de Química
Biológica, Facultad de Ciencias,
Universidad de la República, Montevideo 11400, Uruguay
- Centro
de Investigaciones Biomédicas (CEINBIO), Universidad de la República, Montevideo 11800, Uruguay
- Laboratorio
de Enzimología, Instituto de Química Biológica,
Facultad de Ciencias, Universidad de la
República, Montevideo 11400, Uruguay
- Departamento
de Medicina Transfusional, Hospital de Clínicas, Facultad de
Medicina, Universidad de la República, Montevideo 11600, Uruguay
| | - Marcel Donzé
- Laboratorio
de Fisicoquímica Biológica, Instituto de Química
Biológica, Facultad de Ciencias,
Universidad de la República, Montevideo 11400, Uruguay
- Centro
de Investigaciones Biomédicas (CEINBIO), Universidad de la República, Montevideo 11800, Uruguay
| | - Leonor Thomson
- Centro
de Investigaciones Biomédicas (CEINBIO), Universidad de la República, Montevideo 11800, Uruguay
- Laboratorio
de Enzimología, Instituto de Química Biológica,
Facultad de Ciencias, Universidad de la
República, Montevideo 11400, Uruguay
| | - Ana Denicola
- Laboratorio
de Fisicoquímica Biológica, Instituto de Química
Biológica, Facultad de Ciencias,
Universidad de la República, Montevideo 11400, Uruguay
- Centro
de Investigaciones Biomédicas (CEINBIO), Universidad de la República, Montevideo 11800, Uruguay
| |
Collapse
|
7
|
Tzounakas VL, Anastasiadi AT, Arvaniti VZ, Lelli V, Fanelli G, Paronis EC, Apostolidou AC, Balafas EG, Kostomitsopoulos NG, Papageorgiou EG, Papassideri IS, Stamoulis K, Kriebardis AG, Rinalducci S, Antonelou MH. Supplementation with uric and ascorbic acid protects stored red blood cells through enhancement of non-enzymatic antioxidant activity and metabolic rewiring. Redox Biol 2022; 57:102477. [PMID: 36155342 PMCID: PMC9513173 DOI: 10.1016/j.redox.2022.102477] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/29/2022] [Accepted: 09/12/2022] [Indexed: 11/27/2022] Open
Abstract
Redox imbalance and oxidative stress have emerged as generative causes of the structural and functional degradation of red blood cells (RBC) that happens during their hypothermic storage at blood banks. The aim of the present study was to examine whether the antioxidant enhancement of stored RBC units following uric (UA) and/or ascorbic acid (AA) supplementation can improve their storability as well as post-transfusion phenotypes and recovery by using in vitro and animal models, respectively. For this purpose, 34 leukoreduced CPD/SAGM RBC units were aseptically split in 4 satellite units each. UA, AA or their mixture were added in the three of them, while the fourth was used as control. Hemolysis as well as redox and metabolic parameters were studied in RBC units throughout storage. The addition of antioxidants maintained the quality parameters of stored RBCs, (e.g., hemolysis, calcium homeostasis) and furthermore, shielded them against oxidative defects by boosting extracellular and intracellular (e.g., reduced glutathione; GSH) antioxidant powers. Higher levels of GSH seemed to be obtained through distinct metabolic rewiring in the modified units: methionine-cysteine metabolism in UA samples and glutamine production in the other two groups. Oxidatively-induced hemolysis, reactive oxygen species accumulation and membrane lipid peroxidation were lower in all modifications compared to controls. Moreover, denatured/oxidized Hb binding to the membrane was minor, especially in the AA and mix treatments during middle storage. The treated RBC were able to cope against pro-oxidant triggers when found in a recipient mimicking environment in vitro, and retain control levels of 24h recovery in mice circulation. The currently presented study provides (a) a detailed picture of the effect of UA/AA administration upon stored RBCs and (b) insight into the differential metabolic rewiring when distinct antioxidant "enhancers" are used.
Collapse
Affiliation(s)
- Vassilis L Tzounakas
- Department of Biology, School of Science, National and Kapodistrian University of Athens (NKUA), Athens, Greece
| | - Alkmini T Anastasiadi
- Department of Biology, School of Science, National and Kapodistrian University of Athens (NKUA), Athens, Greece
| | - Vasiliki-Zoi Arvaniti
- Department of Biology, School of Science, National and Kapodistrian University of Athens (NKUA), Athens, Greece
| | - Veronica Lelli
- Department of Ecological and Biological Sciences, University of Tuscia, Viterbo, Italy
| | - Giuseppina Fanelli
- Department of Ecological and Biological Sciences, University of Tuscia, Viterbo, Italy
| | - Efthymios C Paronis
- Center of Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation, Academy of Athens (BRFAA), Athens, Greece
| | - Anastasia C Apostolidou
- Center of Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation, Academy of Athens (BRFAA), Athens, Greece
| | - Evangelos G Balafas
- Center of Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation, Academy of Athens (BRFAA), Athens, Greece
| | - Nikolaos G Kostomitsopoulos
- Center of Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation, Academy of Athens (BRFAA), Athens, Greece
| | - Effie G Papageorgiou
- Laboratory of Reliability and Quality Control in Laboratory Hematology (HemQcR), Department of Biomedical Sciences, School of Health & Welfare Sciences, University of West Attica (UniWA), Egaleo, Greece
| | - Issidora S Papassideri
- Department of Biology, School of Science, National and Kapodistrian University of Athens (NKUA), Athens, Greece
| | | | - Anastasios G Kriebardis
- Laboratory of Reliability and Quality Control in Laboratory Hematology (HemQcR), Department of Biomedical Sciences, School of Health & Welfare Sciences, University of West Attica (UniWA), Egaleo, Greece
| | - Sara Rinalducci
- Department of Ecological and Biological Sciences, University of Tuscia, Viterbo, Italy.
| | - Marianna H Antonelou
- Department of Biology, School of Science, National and Kapodistrian University of Athens (NKUA), Athens, Greece.
| |
Collapse
|
8
|
Phadke I, Pouzolles M, Machado A, Moraly J, Gonzalez-Menendez P, Zimmermann VS, Kinet S, Levine M, Violet PC, Taylor N. Vitamin C deficiency reveals developmental differences between neonatal and adult hematopoiesis. Front Immunol 2022; 13:898827. [PMID: 36248829 PMCID: PMC9562198 DOI: 10.3389/fimmu.2022.898827] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 09/02/2022] [Indexed: 11/25/2022] Open
Abstract
Hematopoiesis, a process that results in the differentiation of all blood lineages, is essential throughout life. The production of 1x1012 blood cells per day, including 200x109 erythrocytes, is highly dependent on nutrient consumption. Notably though, the relative requirements for micronutrients during the perinatal period, a critical developmental window for immune cell and erythrocyte differentiation, have not been extensively studied. More specifically, the impact of the vitamin C/ascorbate micronutrient on perinatal as compared to adult hematopoiesis has been difficult to assess in animal models. Even though humans cannot synthesize ascorbate, due to a pseudogenization of the L-gulono-γ-lactone oxidase (GULO) gene, its generation from glucose is an ancestral mammalian trait. Taking advantage of a Gulo-/- mouse model, we show that ascorbic acid deficiency profoundly impacts perinatal hematopoiesis, resulting in a hypocellular bone marrow (BM) with a significant reduction in hematopoietic stem cells, multipotent progenitors, and hematopoietic progenitors. Furthermore, myeloid progenitors exhibited differential sensitivity to vitamin C levels; common myeloid progenitors and megakaryocyte-erythrocyte progenitors were markedly reduced in Gulo-/- pups following vitamin C depletion in the dams, whereas granulocyte-myeloid progenitors were spared, and their frequency was even augmented. Notably, hematopoietic cell subsets were rescued by vitamin C repletion. Consistent with these data, peripheral myeloid cells were maintained in ascorbate-deficient Gulo-/- pups while other lineage-committed hematopoietic cells were decreased. A reduction in B cell numbers was associated with a significantly reduced humoral immune response in ascorbate-depleted Gulo-/- pups but not adult mice. Erythropoiesis was particularly sensitive to vitamin C deprivation during both the perinatal and adult periods, with ascorbate-deficient Gulo-/- pups as well as adult mice exhibiting compensatory splenic differentiation. Furthermore, in the pathological context of hemolytic anemia, vitamin C-deficient adult Gulo-/- mice were not able to sufficiently increase their erythropoietic activity, resulting in a sustained anemia. Thus, vitamin C plays a pivotal role in the maintenance and differentiation of hematopoietic progenitors during the neonatal period and is required throughout life to sustain erythroid differentiation under stress conditions.
Collapse
Affiliation(s)
- Ira Phadke
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, United States
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, Centre National de la Recherche Scientifique (CNRS), Montpellier, France
- Laboratory of Excellence GR-Ex, Paris, France
| | - Marie Pouzolles
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Alice Machado
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, United States
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, Centre National de la Recherche Scientifique (CNRS), Montpellier, France
- Laboratory of Excellence GR-Ex, Paris, France
| | - Josquin Moraly
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Pedro Gonzalez-Menendez
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, Centre National de la Recherche Scientifique (CNRS), Montpellier, France
- Laboratory of Excellence GR-Ex, Paris, France
| | - Valérie S. Zimmermann
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, United States
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, Centre National de la Recherche Scientifique (CNRS), Montpellier, France
- Laboratory of Excellence GR-Ex, Paris, France
| | - Sandrina Kinet
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, Centre National de la Recherche Scientifique (CNRS), Montpellier, France
- Laboratory of Excellence GR-Ex, Paris, France
| | - Mark Levine
- Molecular and Clinical Nutrition Section, Intramural Research Program, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
- *Correspondence: Mark Levine, ; Pierre-Christian Violet, ; Naomi Taylor,
| | - Pierre-Christian Violet
- Molecular and Clinical Nutrition Section, Intramural Research Program, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
- *Correspondence: Mark Levine, ; Pierre-Christian Violet, ; Naomi Taylor,
| | - Naomi Taylor
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, United States
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, Centre National de la Recherche Scientifique (CNRS), Montpellier, France
- *Correspondence: Mark Levine, ; Pierre-Christian Violet, ; Naomi Taylor,
| |
Collapse
|
9
|
Eigenschink M, Savran D, Zitterer CP, Granitzer S, Fritz M, Baron DM, Müllner EW, Salzer U. Redox Properties of Human Erythrocytes Are Adapted for Vitamin C Recycling. Front Physiol 2021; 12:767439. [PMID: 34938201 PMCID: PMC8685503 DOI: 10.3389/fphys.2021.767439] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 11/16/2021] [Indexed: 01/22/2023] Open
Abstract
Ascorbic acid (AA; or vitamin C) is an important physiological antioxidant and radical scavenger. Some mammalian species, including homo sapiens, have lost the ability to synthetize AA and depend on its nutritional uptake. Erythrocytes from AA-auxotroph mammals express high amounts of the glucose transporter GLUT1. This isoform enables rapid uptake of glucose as well as dehydroascorbate (DHA), the fully oxidized form of AA. Here, we explored the effects of DHA uptake on the redox metabolism of human erythrocytes. DHA uptake enhanced plasma membrane electron transport (PMET) activity. This process is mediated by DCytb, a membrane bound cytochrome catalyzing extracellular reduction of Fe3+ and ascorbate free radical (AFR), the first oxidized form of AA. DHA uptake also decreased cellular radical oxygen species (ROS) levels. Both effects were massively enhanced in the presence of physiological glucose concentrations. Reduction of DHA to AA largely depleted intracellular glutathione (GSH) and induced the efflux of its oxidized form, GSSG. GSSG efflux could be inhibited by MK-571 (IC50 = 5 μM), indicating involvement of multidrug resistance associated protein (MRP1/4). DHA-dependent GSH depletion and GSSG efflux were completely rescued in the presence of 5 mM glucose and, partially, by 2-deoxy-glucose (2-DG), respectively. These findings indicate that human erythrocytes are physiologically adapted to recycle AA both intracellularly via GLUT1-mediated DHA uptake and reduction and extracellularly via DCytb-mediated AFR reduction. We discuss the possibility that this improved erythrocyte-mediated AA recycling was a prerequisite for the emergence of AA auxotrophy which independently occurred at least twice during mammalian evolution.
Collapse
Affiliation(s)
- Michael Eigenschink
- Center for Medical Biochemistry, Max Perutz Labs Vienna, Medical University of Vienna, Vienna, Austria
| | - Danylo Savran
- Center for Medical Biochemistry, Max Perutz Labs Vienna, Medical University of Vienna, Vienna, Austria
| | - Christoph P Zitterer
- Center for Medical Biochemistry, Max Perutz Labs Vienna, Medical University of Vienna, Vienna, Austria
| | - Sebastian Granitzer
- Center for Medical Biochemistry, Max Perutz Labs Vienna, Medical University of Vienna, Vienna, Austria.,Institute of Medical Genetics, Medical University of Vienna, Vienna, Austria
| | - Magdalena Fritz
- Center for Medical Biochemistry, Max Perutz Labs Vienna, Medical University of Vienna, Vienna, Austria
| | - David M Baron
- Department of Anaesthesia, Intensive Care Medicine and Pain Medicine, Medical University of Vienna, Vienna, Austria
| | - Ernst W Müllner
- Center for Medical Biochemistry, Max Perutz Labs Vienna, Medical University of Vienna, Vienna, Austria
| | - Ulrich Salzer
- Center for Medical Biochemistry, Max Perutz Labs Vienna, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
10
|
Highly Expressed CYBRD1 Associated with Glioma Recurrence Regulates the Immune Response of Glioma Cells to Interferon. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:2793222. [PMID: 34326882 PMCID: PMC8302377 DOI: 10.1155/2021/2793222] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 06/30/2021] [Indexed: 11/18/2022]
Abstract
Invasiveness, resistance to treatment, and recurrence of gliomas are significant hurdles to successful treatment regimens. Data sets from Gene Expression Omnibus (GEO), CGGA-RNAseq, and The Cancer Genome Atlas Glioblastoma Multiforme (TCGA-GBM) were analyzed, and an increased expression of Cytochrome B Reductase 1 (CYBRD1) was identified and could be associated with aggravated clinical outcomes. Gene ontology (GO) enrichment analysis indicated that CYBRD1 co-expressed genes are enriched during an immune response. CYBRD1 overexpression in glioma cell lines is enhanced, whereas CYBRD1 silencing attenuated the aggressiveness of glioma cells. In IFN-α-treated glioma cells, IFN-α suppressed the viability and migratory ability and invasive ability of glioma cells, whereas CYBRD1 overexpression attenuated the antitumor effects of IFN-α. CYBRD1 could potentially serve as a biomarker for glioma recurrence. CYBRD1 overexpression enhances glioma cell aggressiveness and attenuates glioma cell response to IFN-α.
Collapse
|
11
|
Pearson AG, Pullar JM, Cook J, Spencer ES, Vissers MC, Carr AC, Hampton MB. Peroxiredoxin 2 oxidation reveals hydrogen peroxide generation within erythrocytes during high-dose vitamin C administration. Redox Biol 2021; 43:101980. [PMID: 33905956 PMCID: PMC8099772 DOI: 10.1016/j.redox.2021.101980] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 03/23/2021] [Accepted: 04/13/2021] [Indexed: 12/14/2022] Open
Abstract
Intravenous infusion of high dose (>10 g) vitamin C (IVC) is a common alternative cancer therapy. IVC results in millimolar levels of circulating ascorbate, which is proposed to generate cytotoxic quantities of H2O2 in the presence of transition metal ions. In this study we report on the in vitro and in vivo effects of millimolar ascorbate on erythrocytes. Addition of ascorbate to whole blood increased erythrocyte intracellular ascorbate approximately 35-fold. Within 10 min of ascorbate addition, we detected increased oxidation of erythrocyte peroxiredoxin 2 (Prx2), a major thiol antioxidant protein and a sensitive marker of H2O2 production. Up to 50% of Prx2 was present in the oxidised form after 60 min. The presence of extracellular catalase, removal of plasma or the addition of a metal chelator did not prevent ascorbate-induced Prx2 oxidation, suggesting that the H2O2 responsible for Prx2 oxidation was generated within the erythrocyte. Ascorbate is known to increase the rate of haemoglobin autoxidation and H2O2 production. Through spectral monitoring of oxidised haemoglobin we estimated a generation rate of 15 μM H2O2/min inside erythrocytes. We also investigated changes in erythrocyte ascorbate concentration and Prx2 oxidation following IVC infusion in a cohort of patients with cancer. Plasma ascorbate levels ranged from 7.8 to 35 mM immediately post infusion, while erythrocyte ascorbate levels reached 1.5–3.4 mM 4 h after beginning infusion. Transient oxidation of erythrocyte Prx2 was observed. We conclude that erythrocytes accumulate ascorbate during IVC infusion, providing a significant reservoir of ascorbate, and this ascorbate increases H2O2 generation within the cells. The consequence of increased erythrocyte Prx2 oxidation warrants further investigation.
Collapse
Affiliation(s)
- Andree G Pearson
- Centre for Free Radical Research, Department of Pathology & Biomedical Science, University of Otago, Christchurch, New Zealand.
| | - Juliet M Pullar
- Centre for Free Radical Research, Department of Pathology & Biomedical Science, University of Otago, Christchurch, New Zealand
| | - John Cook
- New Brighton Health Care, Christchurch, New Zealand
| | - Emma S Spencer
- Nutrition in Medicine Research Group, Department of Pathology & Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Margreet Cm Vissers
- Centre for Free Radical Research, Department of Pathology & Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Anitra C Carr
- Nutrition in Medicine Research Group, Department of Pathology & Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Mark B Hampton
- Centre for Free Radical Research, Department of Pathology & Biomedical Science, University of Otago, Christchurch, New Zealand.
| |
Collapse
|
12
|
Bae DH, Gholam Azad M, Kalinowski DS, Lane DJR, Jansson PJ, Richardson DR. Ascorbate and Tumor Cell Iron Metabolism: The Evolving Story and Its Link to Pathology. Antioxid Redox Signal 2020; 33:816-838. [PMID: 31672021 DOI: 10.1089/ars.2019.7903] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Significance: Vitamin C or ascorbate (Asc) is a water-soluble vitamin and an antioxidant that is involved in many crucial biological functions. Asc's ability to reduce metals makes it an essential enzyme cofactor. Recent Advances: The ability of Asc to act as a reductant also plays an important part in its overall role in iron metabolism, where Asc induces both nontransferrin-bound iron and transferrin-bound iron uptake at physiological concentrations (∼50 μM). Moreover, Asc has emerged to play an important role in multiple diseases and its effects at pharmacological doses could be important for their treatment. Critical Issues: Asc's role as a regulator of cellular iron metabolism, along with its cytotoxic effects and different roles at pharmacological concentrations, makes it a candidate as an anticancer agent. Ever since the controversy regarding the studies from the Mayo Clinic was finally explained, there has been a renewed interest in using Asc as a therapeutic approach toward cancer due to its minimal side effects. Numerous studies have been able to demonstrate the anticancer activity of Asc through selective oxidative stress toward cancer cells via H2O2 generation at pharmacological concentrations. Studies have demonstrated that Asc's cytotoxic mechanism at concentrations (>1 mM) has been associated with decreased cellular iron uptake. Future Directions: Recent studies have also suggested other mechanisms, such as Asc's effects on autophagy, polyamine metabolism, and the cell cycle. Clearly, more has yet to be discovered about Asc's mechanism of action to facilitate safe and effective treatment options for cancer and other diseases.
Collapse
Affiliation(s)
- Dong-Hun Bae
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, The University of Sydney, Sydney, Australia
| | - Mahan Gholam Azad
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, The University of Sydney, Sydney, Australia
| | - Danuta S Kalinowski
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, The University of Sydney, Sydney, Australia
| | - Darius J R Lane
- The Florey Institute of Neuroscience and Mental Health, Melbourne Dementia Research Centre, The University of Melbourne, Parkville, Australia
| | - Patric J Jansson
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, The University of Sydney, Sydney, Australia
| | - Des R Richardson
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, The University of Sydney, Sydney, Australia.,Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Showa-ku, Japan
| |
Collapse
|
13
|
Sherman HG, Jovanovic C, Stolnik S, Baronian K, Downard AJ, Rawson FJ. New Perspectives on Iron Uptake in Eukaryotes. Front Mol Biosci 2018; 5:97. [PMID: 30510932 PMCID: PMC6254016 DOI: 10.3389/fmolb.2018.00097] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 10/23/2018] [Indexed: 12/20/2022] Open
Abstract
All eukaryotic organisms require iron to function. Malfunctions within iron homeostasis have a range of physiological consequences, and can lead to the development of pathological conditions that can result in an excess of non-transferrin bound iron (NTBI). Despite extensive understanding of iron homeostasis, the links between the “macroscopic” transport of iron across biological barriers (cellular membranes) and the chemistry of redox changes that drive these processes still needs elucidating. This review draws conclusions from the current literature, and describes some of the underlying biophysical and biochemical processes that occur in iron homeostasis. By first taking a broad view of iron uptake within the gut and subsequent delivery to tissues, in addition to describing the transferrin and non-transferrin mediated components of these processes, we provide a base of knowledge from which we further explore NTBI uptake. We provide concise up-to-date information of the transplasma electron transport systems (tPMETSs) involved within NTBI uptake, and highlight how these systems are not only involved within NTBI uptake for detoxification but also may play a role within the reduction of metabolic stress through regeneration of intracellular NAD(P)H/NAD(P)+ levels. Furthermore, we illuminate the thermodynamics that governs iron transport, namely the redox potential cascade and electrochemical behavior of key components of the electron transport systems that facilitate the movement of electrons across the plasma membrane to the extracellular compartment. We also take account of kinetic changes that occur to transport iron into the cell, namely membrane dipole change and their consequent effects within membrane structure that act to facilitate transport of ions.
Collapse
Affiliation(s)
- Harry G Sherman
- Division of Regenerative Medicine and Cellular Therapies, School of Pharmacy, University of Nottingham, Nottingham, United Kingdom
| | | | - Snow Stolnik
- Division of Molecular Therapeutics and Formulation, School of Pharmacy, University of Nottingham, Nottingham, United Kingdom
| | - Kim Baronian
- School of Biological Sciences, University of Canterbury, Christchurch, New Zealand
| | - Alison J Downard
- MacDiarmid Institute for Advanced Materials and Nanotechnology, School of Physical and Chemical Sciences, University of Canterbury, Christchurch, New Zealand
| | - Frankie J Rawson
- Division of Regenerative Medicine and Cellular Therapies, School of Pharmacy, University of Nottingham, Nottingham, United Kingdom
| |
Collapse
|
14
|
Ferroportin deficiency in erythroid cells causes serum iron deficiency and promotes hemolysis due to oxidative stress. Blood 2018; 132:2078-2087. [PMID: 30213870 DOI: 10.1182/blood-2018-04-842997] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 09/06/2018] [Indexed: 02/07/2023] Open
Abstract
Ferroportin (FPN), the only known vertebrate iron exporter, transports iron from intestinal, splenic, and hepatic cells into the blood to provide iron to other tissues and cells in vivo. Most of the circulating iron is consumed by erythroid cells to synthesize hemoglobin. Here we found that erythroid cells not only consumed large amounts of iron, but also returned significant amounts of iron to the blood. Erythroblast-specific Fpn knockout (Fpn KO) mice developed lower serum iron levels in conjunction with tissue iron overload and increased FPN expression in spleen and liver without changing hepcidin levels. Our results also showed that Fpn KO mice, which suffer from mild hemolytic anemia, were sensitive to phenylhydrazine-induced oxidative stress but were able to tolerate iron deficiency upon exposure to a low-iron diet and phlebotomy, supporting that the anemia of Fpn KO mice resulted from erythrocytic iron overload and resulting oxidative injury rather than a red blood cell (RBC) production defect. Moreover, we found that the mean corpuscular volume (MCV) values of gain-of-function FPN mutation patients were positively associated with serum transferrin saturations, whereas MCVs of loss-of-function FPN mutation patients were not, supporting that erythroblasts donate iron to blood through FPN in response to serum iron levels. Our results indicate that FPN of erythroid cells plays an unexpectedly essential role in maintaining systemic iron homeostasis and protecting RBCs from oxidative stress, providing insight into the pathophysiology of FPN diseases.
Collapse
|
15
|
Schlottmann F, Vera-Aviles M, Latunde-Dada GO. Duodenal cytochrome b (Cybrd1) ferric reductase functional studies in cells. Metallomics 2018; 9:1389-1393. [PMID: 28937159 DOI: 10.1039/c7mt00254h] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Dietary non-heme ferric iron is reduced by the ferric reductase enzyme, duodenal cytochrome b (Dcytb), before absorption by the divalent metal transporter 1 (DMT1). A single nucleotide polymorphism (SNP rs10455 mutant) that is located in the last exon of the Dcytb gene was reported in C282Y haemochromatosis HFE subjects. The present work therefore investigated the phenotype of this mutant Dcytb in Chinese hamster ovary (CHO) cells. These cultured cells were transfected with either wild type (WT) or the SNP vector plasmids of Dcytb. Ferric reductase assays were performed in Dcytb transgenic CHO cells using the ferrozine spectrophometric assay protocol. The Dcytb SNP rs10455 showed a gain-of-function capability since ferric reductase activity increased significantly (p < 0.01) in the transgenic cells. Varying ferric reductase activity was found when CHO cells were pretreated with modulators of Dcytb protein expression. Although ferric reductase in endogenous CHO cells increased with deferoxamine or CoCl2, iron loading with ferric ammonium citrate (FAC) had the opposite effect. Taken together, the study reveals a gain-of-function phenotype for Dcytb rs10455 mutation that could be a putative modifier of colorectal cancer risk, with attendant variability in penetrance among human HFE C282Y homozygotes.
Collapse
Affiliation(s)
- F Schlottmann
- King's College London, Department of Nutritional Sciences, Faculty of Life Sciences and Medicine, Franklin-Wilkins Building, 150 Stamford Street, London, SE1 9NH, UK.
| | | | | |
Collapse
|
16
|
Sherman HG, Jovanovic C, Stolnik S, Rawson FJ. Electrochemical System for the Study of Trans-Plasma Membrane Electron Transport in Whole Eukaryotic Cells. Anal Chem 2018; 90:2780-2786. [DOI: 10.1021/acs.analchem.7b04853] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Harry G. Sherman
- Division
of Regenerative Medicine and Cellular Therapies, School of Pharmacy, University of Nottingham, Nottingham, NG7 2RD, United Kingdom
| | | | - Snow Stolnik
- Division
of Molecular Therapeutics and Formulation, School of Pharmacy, University of Nottingham, Nottingham, NG7 2RD, United Kingdom
| | - Frankie J. Rawson
- Division
of Regenerative Medicine and Cellular Therapies, School of Pharmacy, University of Nottingham, Nottingham, NG7 2RD, United Kingdom
| |
Collapse
|
17
|
Chemical Transport Knockout for Oxidized Vitamin C, Dehydroascorbic Acid, Reveals Its Functions in vivo. EBioMedicine 2017; 23:125-135. [PMID: 28851583 PMCID: PMC5605377 DOI: 10.1016/j.ebiom.2017.08.017] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 08/15/2017] [Accepted: 08/17/2017] [Indexed: 12/24/2022] Open
Abstract
Despite its transport by glucose transporters (GLUTs) in vitro, it is unknown whether dehydroascorbic acid (oxidized vitamin C, DHA) has any in vivo function. To investigate, we created a chemical transport knockout model using the vitamin C analog 6-bromo-ascorbate. This analog is transported on sodium-dependent vitamin C transporters but its oxidized form, 6-bromo-dehydroascorbic acid, is not transported by GLUTs. Mice (gulo−/−) unable to synthesize ascorbate (vitamin C) were raised on 6-bromo-ascorbate. Despite normal survival, centrifugation of blood produced hemolysis secondary to near absence of red blood cell (RBC) ascorbate/6-bromo-ascorbate. Key findings with clinical implications were that RBCs in vitro transported dehydroascorbic acid but not bromo-dehydroascorbic acid; RBC ascorbate in vivo was obtained only via DHA transport; ascorbate via DHA transport in vivo was necessary for RBC structural integrity; and internal RBC ascorbate was essential to maintain ascorbate plasma concentrations in vitro/in vivo. Red cells in vivo obtain vitamin C (ascorbate) by dehydroascorbic acid transport. Red blood cell ascorbate is necessary to maintain red blood cell structural integrity. Red blood cell ascorbate maintains external plasma ascorbate concentrations in vivo by transmembrane electron transfer.
In animals and humans, it is unknown whether the oxidized form of vitamin C, termed dehydroascorbic acid, has a physiologic purpose. Using a mouse model and a custom-synthesized vitamin C analog, we show that red blood cells obtain their vitamin C by transport of dehydroascorbic acid, instead of vitamin C itself. The transported material is reduced inside and has at least two physiologic functions. One is to maintain structural integrity of red blood cells, and the other is to maintain vitamin C in the liquid part of blood, plasma.
Collapse
|
18
|
Kuhn V, Diederich L, Keller TCS, Kramer CM, Lückstädt W, Panknin C, Suvorava T, Isakson BE, Kelm M, Cortese-Krott MM. Red Blood Cell Function and Dysfunction: Redox Regulation, Nitric Oxide Metabolism, Anemia. Antioxid Redox Signal 2017; 26:718-742. [PMID: 27889956 PMCID: PMC5421513 DOI: 10.1089/ars.2016.6954] [Citation(s) in RCA: 266] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
SIGNIFICANCE Recent clinical evidence identified anemia to be correlated with severe complications of cardiovascular disease (CVD) such as bleeding, thromboembolic events, stroke, hypertension, arrhythmias, and inflammation, particularly in elderly patients. The underlying mechanisms of these complications are largely unidentified. Recent Advances: Previously, red blood cells (RBCs) were considered exclusively as transporters of oxygen and nutrients to the tissues. More recent experimental evidence indicates that RBCs are important interorgan communication systems with additional functions, including participation in control of systemic nitric oxide metabolism, redox regulation, blood rheology, and viscosity. In this article, we aim to revise and discuss the potential impact of these noncanonical functions of RBCs and their dysfunction in the cardiovascular system and in anemia. CRITICAL ISSUES The mechanistic links between changes of RBC functional properties and cardiovascular complications related to anemia have not been untangled so far. FUTURE DIRECTIONS To allow a better understanding of the complications associated with anemia in CVD, basic and translational science studies should be focused on identifying the role of noncanonical functions of RBCs in the cardiovascular system and on defining intrinsic and/or systemic dysfunction of RBCs in anemia and its relationship to CVD both in animal models and clinical settings. Antioxid. Redox Signal. 26, 718-742.
Collapse
Affiliation(s)
- Viktoria Kuhn
- 1 Cardiovascular Research Laboratory, Division of Cardiology, Pneumology, and Vascular Medicine, Medical Faculty, Heinrich Heine University of Düsseldorf , Düsseldorf, Germany
| | - Lukas Diederich
- 1 Cardiovascular Research Laboratory, Division of Cardiology, Pneumology, and Vascular Medicine, Medical Faculty, Heinrich Heine University of Düsseldorf , Düsseldorf, Germany
| | - T C Stevenson Keller
- 2 Department of Molecular Physiology and Biological Physics, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine , Charlottesville, Virginia
| | - Christian M Kramer
- 1 Cardiovascular Research Laboratory, Division of Cardiology, Pneumology, and Vascular Medicine, Medical Faculty, Heinrich Heine University of Düsseldorf , Düsseldorf, Germany
| | - Wiebke Lückstädt
- 1 Cardiovascular Research Laboratory, Division of Cardiology, Pneumology, and Vascular Medicine, Medical Faculty, Heinrich Heine University of Düsseldorf , Düsseldorf, Germany
| | - Christina Panknin
- 1 Cardiovascular Research Laboratory, Division of Cardiology, Pneumology, and Vascular Medicine, Medical Faculty, Heinrich Heine University of Düsseldorf , Düsseldorf, Germany
| | - Tatsiana Suvorava
- 1 Cardiovascular Research Laboratory, Division of Cardiology, Pneumology, and Vascular Medicine, Medical Faculty, Heinrich Heine University of Düsseldorf , Düsseldorf, Germany
| | - Brant E Isakson
- 2 Department of Molecular Physiology and Biological Physics, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine , Charlottesville, Virginia
| | - Malte Kelm
- 1 Cardiovascular Research Laboratory, Division of Cardiology, Pneumology, and Vascular Medicine, Medical Faculty, Heinrich Heine University of Düsseldorf , Düsseldorf, Germany
| | - Miriam M Cortese-Krott
- 1 Cardiovascular Research Laboratory, Division of Cardiology, Pneumology, and Vascular Medicine, Medical Faculty, Heinrich Heine University of Düsseldorf , Düsseldorf, Germany
| |
Collapse
|
19
|
Lemler DJ, Lynch ML, Tesfay L, Deng Z, Paul BT, Wang X, Hegde P, Manz DH, Torti SV, Torti FM. DCYTB is a predictor of outcome in breast cancer that functions via iron-independent mechanisms. Breast Cancer Res 2017; 19:25. [PMID: 28270217 PMCID: PMC5341190 DOI: 10.1186/s13058-017-0814-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Accepted: 02/09/2017] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Duodenal cytochrome b (DCYTB) is a ferrireductase that functions together with divalent metal transporter 1 (DMT1) to mediate dietary iron reduction and uptake in the duodenum. DCYTB is also a member of a 16-gene iron regulatory gene signature (IRGS) that predicts metastasis-free survival in breast cancer patients. To better understand the relationship between DCYTB and breast cancer, we explored in detail the prognostic significance and molecular function of DCYTB in breast cancer. METHODS The prognostic significance of DCYTB expression was evaluated using publicly available microarray data. Signaling Pathway Impact Analysis (SPIA) of microarray data was used to identify potential novel functions of DCYTB. The role of DCYTB was assessed using immunohistochemistry and measurements of iron uptake, iron metabolism, and FAK signaling. RESULTS High DCYTB expression was associated with prolonged survival in two large independent cohorts, together totaling 1610 patients (cohort #1, p = 1.6e-11, n = 741; cohort #2, p = 1.2e-05, n = 869; log-rank test) as well as in the Gene expression-based Outcome for Breast cancer Online (GOBO) cohort (p < 1.0e-05, n = 1379). High DCYTB expression was also associated with increased survival in homogeneously treated groups of patients who received either tamoxifen or chemotherapy. Immunohistochemistry revealed that DCYTB is localized on the plasma membrane of breast epithelial cells, and that expression is dramatically reduced in high-grade tumors. Surprisingly, neither overexpression nor knockdown of DCYTB affected levels of ferritin H, transferrin receptor, labile iron or total cellular iron in breast cancer cells. Because SPIA pathway analysis of patient microarray data revealed an association between DCYTB and the focal adhesion pathway, we examined the influence of DCYTB on FAK activation in breast cancer cells. These experiments reveal that DCYTB reduces adhesion and activation of focal adhesion kinase (FAK) and its adapter protein paxillin. CONCLUSIONS DCYTB is an important predictor of outcome and is associated with response to therapy in breast cancer patients. DCYTB does not affect intracellular iron in breast cancer cells. Instead, DCYTB may retard cancer progression by reducing activation of FAK, a kinase that plays a central role in tumor cell adhesion and metastasis.
Collapse
Affiliation(s)
- David J. Lemler
- Department of Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, CT 06030 USA
- Present address: Department of Molecular Biomedical Sciences, North Carolina State University, CVM Research Building 474, Raleigh, NC 27695 USA
| | - Miranda L. Lynch
- Center for Quantitative Medicine, University of Connecticut Health Center, Farmington, CT 06030 USA
- Present address: Statistical Sciences Group CCS-6, Los Alamos National Laboratory, Los Alamos, NM 87545 USA
| | - Lia Tesfay
- Department of Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, CT 06030 USA
| | - Zhiyong Deng
- Department of Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, CT 06030 USA
| | - Bibbin T. Paul
- Department of Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, CT 06030 USA
| | - Xiaohong Wang
- Department of Pathology, University of Connecticut Health Center, Farmington, CT 06030 USA
| | - Poornima Hegde
- Department of Pathology, University of Connecticut Health Center, Farmington, CT 06030 USA
| | - David H. Manz
- Department of Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, CT 06030 USA
- School of Dental Medicine, University of Connecticut Health Center, Farmington, CT 06030 USA
| | - Suzy V. Torti
- Department of Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, CT 06030 USA
| | - Frank M. Torti
- Department of Medicine, University of Connecticut Health Center, Farmington, CT 06030 USA
| |
Collapse
|
20
|
Padayatty SJ, Levine M. Vitamin C: the known and the unknown and Goldilocks. Oral Dis 2016; 22:463-93. [PMID: 26808119 PMCID: PMC4959991 DOI: 10.1111/odi.12446] [Citation(s) in RCA: 441] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 01/08/2016] [Indexed: 12/11/2022]
Abstract
Vitamin C (Ascorbic Acid), the antiscorbutic vitamin, cannot be synthesized by humans and other primates, and has to be obtained from diet. Ascorbic acid is an electron donor and acts as a cofactor for fifteen mammalian enzymes. Two sodium-dependent transporters are specific for ascorbic acid, and its oxidation product dehydroascorbic acid is transported by glucose transporters. Ascorbic acid is differentially accumulated by most tissues and body fluids. Plasma and tissue vitamin C concentrations are dependent on amount consumed, bioavailability, renal excretion, and utilization. To be biologically meaningful or to be clinically relevant, in vitro and in vivo studies of vitamin C actions have to take into account physiologic concentrations of the vitamin. In this paper, we review vitamin C physiology; the many phenomena involving vitamin C where new knowledge has accrued or where understanding remains limited; raise questions about the vitamin that remain to be answered; and explore lines of investigations that are likely to be fruitful.
Collapse
Affiliation(s)
- S J Padayatty
- Molecular and Clinical Nutrition Section, Digestive Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - M Levine
- Molecular and Clinical Nutrition Section, Digestive Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
21
|
Tedesco I, Moccia S, Volpe S, Alfieri G, Strollo D, Bilotto S, Spagnuolo C, Di Renzo M, Aquino RP, Russo GL. Red wine activates plasma membrane redox system in human erythrocytes. Free Radic Res 2016; 50:557-69. [PMID: 26866566 DOI: 10.3109/10715762.2016.1152629] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
In the present study, we report that polyphenols present in red wine obtained by a controlled microvinification process are able to protect human erythrocytes from oxidative stress and to activate Plasma Membrane Redox System (PMRS). Human plasma obtained from healthy subjects was incubated in the presence of whole red wine at a concentration corresponding to 9.13-73 μg/ml gallic acid equivalents to verify the capacity to protect against hypochlorous acid (HOCl)-induced plasma oxidation and to minimize chloramine formation. Red wine reduced hemolysis and chloramine formation induced by HOCl of 40 and 35%, respectively. PMRS present on human erythrocytes transfers electrons from intracellular molecules to extracellular electron acceptors. We demonstrated that whole red wine activated PMRS activity in human erythrocytes isolated from donors in a dose-dependent manner with a maximum at about 70-100 μg/ml gallic acid equivalents. We also showed that red wine increased glutathione (GSH) levels and erythrocytic antioxidant capacity, measured by 2,2-diphenyl-1-picrylhydrazyl (DPPH) quenching assay. Furthermore, we reported that GSH played a crucial role in regulating PMRS activity in erythrocytes. In fact, the effect of iodoacetamide, an alkylating agent that induces depletion of intracellular GSH, was completely counteracted by red wine. Bioactive compounds present in red wine, such as gallic acid, resveratrol, catechin, and quercetin were unable to activate PMRS when tested at the concentrations normally present in aged red wines. On the contrary, the increase of PMRS activity was associated with the anthocyanin fraction, suggesting the capacity of this class of compounds to positively modulate PMRS enzymatic activity.
Collapse
Affiliation(s)
- Idolo Tedesco
- a Institute of Food Sciences, National Research Council , Avellino , Italy
| | - Stefania Moccia
- a Institute of Food Sciences, National Research Council , Avellino , Italy
| | - Silvestro Volpe
- b Division of Onco-Hematology , S.G. Moscati Hospital , Avellino , Italy
| | - Giovanna Alfieri
- b Division of Onco-Hematology , S.G. Moscati Hospital , Avellino , Italy
| | | | - Stefania Bilotto
- a Institute of Food Sciences, National Research Council , Avellino , Italy
| | - Carmela Spagnuolo
- a Institute of Food Sciences, National Research Council , Avellino , Italy
| | | | - Rita P Aquino
- d Department of Pharmacy , University of Salerno , Fisciano (SA) , Italy
| | - Gian Luigi Russo
- a Institute of Food Sciences, National Research Council , Avellino , Italy
| |
Collapse
|
22
|
Veskoukis AS, Goutianos G, Paschalis V, Margaritelis NV, Tzioura A, Dipla K, Zafeiridis A, Vrabas IS, Kyparos A, Nikolaidis MG. The rat closely mimics oxidative stress and inflammation in humans after exercise but not after exercise combined with vitamin C administration. Eur J Appl Physiol 2016; 116:791-804. [PMID: 26856335 DOI: 10.1007/s00421-016-3336-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2015] [Accepted: 01/25/2016] [Indexed: 02/08/2023]
Abstract
PURPOSE The purpose of the present study was to directly compare oxidative stress and inflammation responses between rats and humans. METHODS We contrasted rat and human oxidative stress and inflammatory responses to exercise (pro-oxidant stimulus) and/or vitamin C (anti-oxidant stimulus) administration. Vitamin C was administered orally in both species (16 mg kg(-1) of body weight). Twelve redox biomarkers and seven inflammatory biomarkers were determined in plasma and erythrocytes pre- and post-exercise or pre- and post-exercise combined with vitamin C administration. RESULTS Exercise increased oxidative stress and induced an inflammatory state in rats and humans. There were only 1/19 significant species × exercise interactions (catalase), indicating similar responses to exercise between rats and humans in redox and inflammatory biomarkers. Vitamin C decreased oxidative stress and increased antioxidant capacity only in humans and did not affect the redox state of rats. In contrast, vitamin C induced an anti-inflammatory state only in rats and did not affect the inflammatory state of humans. There were 10/19 significant species × vitamin C interactions, indicating that rats poorly mimic human oxidative stress and inflammatory responses to vitamin C administration. Exercise after acute vitamin C administration altered redox state only in humans and did not affect the redox state of rats. On the contrary, inflammation biomarkers changed similarly after exercise combined with vitamin C in both rats and humans. CONCLUSIONS The rat adequately mimics human responses to exercise in basic blood redox/inflammatory profile, yet this is not the case after exercise combined with vitamin C administration.
Collapse
Affiliation(s)
- Aristidis S Veskoukis
- Department of Physical Education and Sports Science at Serres, Aristotle University of Thessaloniki, Agios Ioannis, 62110, Serres, Greece
| | - Georgios Goutianos
- Department of Physical Education and Sports Science at Serres, Aristotle University of Thessaloniki, Agios Ioannis, 62110, Serres, Greece
| | - Vassilis Paschalis
- Department of Physical Education and Sport Science, University of Thessaly, Karies, Trikala, Greece.,Department of Health Sciences, School of Sciences, European University Cyprus, Nicosia, Cyprus
| | - Nikos V Margaritelis
- Department of Physical Education and Sports Science at Serres, Aristotle University of Thessaloniki, Agios Ioannis, 62110, Serres, Greece.,Intensive Care Unit, 424 General Military Hospital of Thessaloniki, Thessaloniki, Greece
| | - Aikaterini Tzioura
- Department of Physical Education and Sports Science at Serres, Aristotle University of Thessaloniki, Agios Ioannis, 62110, Serres, Greece.,Department of Hematology, Blood Bank, General Hospital of Serres, Serres, Greece
| | - Konstantina Dipla
- Department of Physical Education and Sports Science at Serres, Aristotle University of Thessaloniki, Agios Ioannis, 62110, Serres, Greece
| | - Andreas Zafeiridis
- Department of Physical Education and Sports Science at Serres, Aristotle University of Thessaloniki, Agios Ioannis, 62110, Serres, Greece
| | - Ioannis S Vrabas
- Department of Physical Education and Sports Science at Serres, Aristotle University of Thessaloniki, Agios Ioannis, 62110, Serres, Greece
| | - Antonios Kyparos
- Department of Physical Education and Sports Science at Serres, Aristotle University of Thessaloniki, Agios Ioannis, 62110, Serres, Greece
| | - Michalis G Nikolaidis
- Department of Physical Education and Sports Science at Serres, Aristotle University of Thessaloniki, Agios Ioannis, 62110, Serres, Greece.
| |
Collapse
|
23
|
Gibson SW, Todd CD. Arabidopsis AIR12 influences root development. PHYSIOLOGY AND MOLECULAR BIOLOGY OF PLANTS : AN INTERNATIONAL JOURNAL OF FUNCTIONAL PLANT BIOLOGY 2015; 21:479-89. [PMID: 26600675 PMCID: PMC4646869 DOI: 10.1007/s12298-015-0323-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Revised: 09/13/2015] [Accepted: 09/21/2015] [Indexed: 05/05/2023]
Abstract
Arabidopsis AUXIN INDUCED IN ROOTS (AIR 12) is a predicted to encode a glycosylphosphatidylinositol tail anchored protein. It has been associated with extracellular redox processes, but little is known about its physiological role. An air12 mutant line demonstrated increased germination rates in the presence of a range of abiotic stress factors and hormones, but not in the presence of ABA. Disruption of AIR12 also affected primary and lateral root development and was linked to changes in root catalase activity and superoxide production. We suggest AIR12 is an extracellular constituent linking both hormone and reactive oxygen signaling in plants.
Collapse
Affiliation(s)
| | - Christopher D. Todd
- Department of Biology, University of Saskatchewan, 112 Science Place, Saskatoon, SK S7N 5E2 Canada
| |
Collapse
|
24
|
Sadowska-Bartosz I, Bartosz G. Ascorbic acid and protein glycation in vitro. Chem Biol Interact 2015; 240:154-62. [PMID: 26163454 DOI: 10.1016/j.cbi.2015.07.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 05/14/2015] [Accepted: 07/06/2015] [Indexed: 02/07/2023]
Abstract
The aim of the study was to compare the effects of ascorbic acid (AA) in vitro in the absence and in the presence of cell-dependent recycling. In a cell-free system, AA enhanced glycoxidation of bovine serum albumin (BSA) by glucose and induced BSA glycation in the absence of sugars. On the other hand, AA did not affect erythrocyte hemolysis, glycation of hemoglobin and erythrocyte membranes, and inactivation of catalase, protected against inactivation of acetylcholinesterase of erythrocytes incubated with high glucose concentrations and enhanced the loss of glutathione. These results can be explained by assumption that AA acts as a proglycating agent in the absence of recycling while is an antiglycating agent when metabolic recycling occurs.
Collapse
Affiliation(s)
- Izabela Sadowska-Bartosz
- Department of Biochemistry and Cell Biology, University of Rzeszow, Zelwerowicza St. 4, PL 35-601 Rzeszow, Poland.
| | - Grzegorz Bartosz
- Department of Biochemistry and Cell Biology, University of Rzeszow, Zelwerowicza St. 4, PL 35-601 Rzeszow, Poland; Department of Molecular Biophysics, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland
| |
Collapse
|
25
|
Duodenal cytochrome b (DCYTB) in iron metabolism: an update on function and regulation. Nutrients 2015; 7:2274-96. [PMID: 25835049 PMCID: PMC4425144 DOI: 10.3390/nu7042274] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2014] [Revised: 03/03/2015] [Accepted: 03/05/2015] [Indexed: 01/01/2023] Open
Abstract
Iron and ascorbate are vital cellular constituents in mammalian systems. The bulk-requirement for iron is during erythropoiesis leading to the generation of hemoglobin-containing erythrocytes. Additionally, both iron and ascorbate are required as co-factors in numerous metabolic reactions. Iron homeostasis is controlled at the level of uptake, rather than excretion. Accumulating evidence strongly suggests that in addition to the known ability of dietary ascorbate to enhance non-heme iron absorption in the gut, ascorbate regulates iron homeostasis. The involvement of ascorbate in dietary iron absorption extends beyond the direct chemical reduction of non-heme iron by dietary ascorbate. Among other activities, intra-enterocyte ascorbate appears to be involved in the provision of electrons to a family of trans-membrane redox enzymes, namely those of the cytochrome b561 class. These hemoproteins oxidize a pool of ascorbate on one side of the membrane in order to reduce an electron acceptor (e.g., non-heme iron) on the opposite side of the membrane. One member of this family, duodenal cytochrome b (DCYTB), may play an important role in ascorbate-dependent reduction of non-heme iron in the gut prior to uptake by ferrous-iron transporters. This review discusses the emerging relationship between cellular iron homeostasis, the emergent “IRP1-HIF2α axis”, DCYTB and ascorbate in relation to iron metabolism.
Collapse
|
26
|
Lane DJR, Richardson DR. The active role of vitamin C in mammalian iron metabolism: much more than just enhanced iron absorption! Free Radic Biol Med 2014; 75:69-83. [PMID: 25048971 DOI: 10.1016/j.freeradbiomed.2014.07.007] [Citation(s) in RCA: 163] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Revised: 07/04/2014] [Accepted: 07/08/2014] [Indexed: 01/18/2023]
Abstract
Ascorbate is a cofactor in numerous metabolic reactions. Humans cannot synthesize ascorbate owing to inactivation of the gene encoding the enzyme l-gulono-γ-lactone oxidase, which is essential for ascorbate synthesis. Accumulating evidence strongly suggests that in addition to the known ability of dietary ascorbate to enhance nonheme iron absorption in the gut, ascorbate within mammalian systems can regulate cellular iron uptake and metabolism. Ascorbate modulates iron metabolism by stimulating ferritin synthesis, inhibiting lysosomal ferritin degradation, and decreasing cellular iron efflux. Furthermore, ascorbate cycling across the plasma membrane is responsible for ascorbate-stimulated iron uptake from low-molecular-weight iron-citrate complexes, which are prominent in the plasma of individuals with iron-overload disorders. Importantly, this iron-uptake pathway is of particular relevance to astrocyte brain iron metabolism and tissue iron loading in disorders such as hereditary hemochromatosis and β-thalassemia. Recent evidence also indicates that ascorbate is a novel modulator of the classical transferrin-iron uptake pathway, which provides almost all iron for cellular demands and erythropoiesis under physiological conditions. Ascorbate acts to stimulate transferrin-dependent iron uptake by an intracellular reductive mechanism, strongly suggesting that it may act to stimulate iron mobilization from the endosome. The ability of ascorbate to regulate transferrin iron uptake could help explain the metabolic defect that contributes to ascorbate-deficiency-induced anemia.
Collapse
Affiliation(s)
- Darius J R Lane
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, NSW 2006, Australia.
| | - Des R Richardson
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, NSW 2006, Australia.
| |
Collapse
|
27
|
Hwang MW, Kim BJ. Apoptotic Effects and Involvement of TRPM7 Channels of the Traditional Herbal Medicine, Dangkwisoo-San in Gastric Cancer Cells. INT J PHARMACOL 2014; 10:398-405. [DOI: 10.3923/ijp.2014.398.405] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
28
|
Sakai H, Li B, Lim WL, Iga Y. Red blood cells donate electrons to methylene blue mediated chemical reduction of methemoglobin compartmentalized in liposomes in blood. Bioconjug Chem 2014; 25:1301-10. [PMID: 24877769 DOI: 10.1021/bc500153x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Electron-energy-rich coenzymes in cells, NADH and NADPH, are re-energized repeatedly through the Embden-Meyerhof and pentose-phosphate glycolytic pathways, respectively. This study demonstrates extraction of their electron energies in red blood cells (RBCs) for in vivo extracellular chemical reactions using an electron mediator shuttling across the biomembrane. Hemoglobin-vesicles (HbVs) are an artificial oxygen carrier encapsulating purified and concentrated Hb solution in liposomes. Because of the absence of a metHb-reducing enzymatic system in HbV, HbO2 gradually autoxidizes to form metHb. Wistar rats received HbV suspension (10 mL/kg body weight) intravenously. At the metHb level of around 50%, methylene blue [MB(+); 3,7-bis(dimethylamino)phenothiazinium chloride] was injected. The level of metHb quickly decreased to around 16% in 40 min, remaining for more than 5 h. In vitro mixing of HbV/MB(+) with RBCs recreated the in vivo metHb reduction, but not with plasma. NAD(P)H levels in RBCs decreased after metHb reduction. The addition of glucose facilitated metHb reduction. Liposome-encapsulated NAD(P)H, a model of RBC, reduced metHb in HbV in the presence of MB(+). These results indicate that (i) NAD(P)H in RBCs reacts with MB(+) to convert it to leukomethylene blue (MBH); (ii) MB(+) and MBH shuttle freely between RBC and HbV across the hydrophobic lipid membranes; and (iii) MBH is transferred into HbV and reduces metHb in HbV. Four other electron mediators with appropriate redox potentials appeared to be as effective as MB(+) was, indicating the possibility for further optimization of electron mediators. We established an indirect enzymatic metHb reducing system for HbV using unlimited endogenous electrons created in RBCs in combination with an effective electron mediator that prolongs the functional lifespan of HbV in blood circulation.
Collapse
Affiliation(s)
- Hiromi Sakai
- Department of Chemistry, Nara Medical University , 840 Shijo-cho, Kashihara, Nara 634-8521, Japan
| | | | | | | |
Collapse
|
29
|
Lee HJ, Kim MC, Lim B, Kim BJ. Buxus Microphylla var. Koreana Nakai Extract for the Treatment of Gastric Cancer. J Pharmacopuncture 2013; 16:39-45. [PMID: 25780674 PMCID: PMC4331968 DOI: 10.3831/kpi.2013.16.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Accepted: 06/12/2013] [Indexed: 11/24/2022] Open
Abstract
OBJECTIVES Buxus Microphylla var. Koreana Nakai Extract (BMKNE) is used as a folk remedy for malaria and veneral disease. In the present study, we investigated the effects of BMKNE in the growth and the survival of AGS cells, the most common human gastric adenocarcinoma cell lines. METHODS The AGS cells were treated with varying concentrations of BMKNE. Analyses of the sub G1 peak, the caspase-3 and -9 activities, and the mitochondrial depolarization were conducted to determine whether AGS cell death occured by apoptosis. Also, to identify the role of transient receptor potential melastatin (TRPM) 7 channels in AGS cell growth and survival, we used human embryonic kidney (HEK) 293 cells overexpressed with TRPM7 channels. RESULTS Experimental results showed that the sub G1 peak, the caspase-3 and -9 activities, and the mitochondrial depolarization were increased. Therefore, BMKNE was found to induce the apoptosis of these cells, and this apoptosis was inhibited by SB203580 (a p38 mitogen-activated protein kinase (MAPK) inhibitor), and by a c-jun NH2-terminal kinase (JNK) II inhibitor. Furthermore, BMKNE inhibited TRPM7 currents and TRPM7 channel over-expressions in HEK 293 cells, exacerbating BMKNE-induced cell death. CONCLUSIONS These findings indicate that BMKNE inhibits the growth and the survival of gastric cancer cells due to a blockade of the TRPM7 channel's activity and MAPK signaling. Therefore, BMKNE is a potential drug for treatment of gastric cancer, and both the TRPM7 channel and MAPK signaling may play an important role in survival in gastric cancer cells.
Collapse
Affiliation(s)
- Hee Jung Lee
- Division of Longevity and Biofunctional Medicine, Pusan National University School of Korean Medicine, Yangsan, Korea
| | - Min Chul Kim
- Division of Longevity and Biofunctional Medicine, Pusan National University School of Korean Medicine, Yangsan, Korea
| | - Bora Lim
- Division of Longevity and Biofunctional Medicine, Pusan National University School of Korean Medicine, Yangsan, Korea
| | - Byung Joo Kim
- Division of Longevity and Biofunctional Medicine, Pusan National University School of Korean Medicine, Yangsan, Korea
| |
Collapse
|
30
|
Luo X, Hill M, Johnson A, Latunde-Dada GO. Modulation of Dcytb (Cybrd 1) expression and function by iron, dehydroascorbate and Hif-2α in cultured cells. Biochim Biophys Acta Gen Subj 2013; 1840:106-12. [PMID: 23981688 DOI: 10.1016/j.bbagen.2013.08.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Revised: 07/24/2013] [Accepted: 08/17/2013] [Indexed: 01/25/2023]
Abstract
BACKGROUND Duodenal cytochrome b (Dcytb) is a mammalian plasma ferric reductase enzyme that catalyses the reduction of ferric to ferrous ion in the process of iron absorption. The current study investigates the relationship between Dcytb, iron, dehydroascorbate (DHA) and Hif-2α in cultured cell lines. METHODS Dcytb and Hif-2α protein expression was analysed by Western blot technique while gene regulation was determined by quantitative PCR. Functional analyses were carried out by ferric reductase and (59)Fe uptake assays. RESULTS Iron and dehydroascorbic acid treatment of cells inhibited Dcytb mRNA and protein expression. Desferrioxamine also enhanced Dcytb mRNA level after cells were treated overnight. Dcytb knockdown in HuTu cells resulted in reduced mRNA expression and lowered reductase activity. Preloading cells with DHA (to enhance intracellular ascorbate levels) did not stimulate reductase activity fully in Dcytb-silenced cells, implying a Dcytb-dependence of ascorbate-mediated ferrireduction. Moreover, Hif-2α knockdown in HuTu cells led to a reduction in reductase activity and iron uptake. CONCLUSIONS Taken together, this study shows the functional regulation of Dcytb reductase activity by DHA and Hif-2α. GENERAL SIGNIFICANCE Dcytb is a plasma membrane protein that accepts electrons intracellularly from DHA/ascorbic acid for ferrireduction at the apical surface of cultured cells and enterocytes.
Collapse
Affiliation(s)
- Xiaomin Luo
- Diabetes and Nutritional Sciences Division, King's College London, London, UK
| | | | | | | |
Collapse
|
31
|
Lim B, Lee HJ, Kim MC, Kim BJ. Effects of Ulmi Pumilae Cortex on AGS Gastric Cancer Cells. J Pharmacopuncture 2013; 16:55-61. [PMID: 25780669 PMCID: PMC4331962 DOI: 10.3831/kpi.2013.16.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Accepted: 04/19/2013] [Indexed: 11/09/2022] Open
Abstract
OBJECTIVE Ulmi Pumilae Cortex(UPC) is a deciduous tree with uneven pinnate leaves and is classified as a subfamily of Ulmuceae and contains many pharmacologically active constituents. The aim of this study was to investigate the effects of UPC on the growth and survival of AGS cells, the most common human gastric adenocarcinoma cell lines. METHODS The AGS cells were treated with varying concentrations of UPC. Analyses of the sub G1, caspase-3 activity, and mitochondrial depolarization were conducted to determine whether AGS cell death occured by apoptosis. Furthermore, to identify the role of the transient receptor potential melastatin (TRPM) 7 channels in AGS cell growth and survival, we used human embryonic kidney (HEK) 293 cells overexpressed with TRPM7 channels. RESULTS The addition of UPC to a culture medium inhibited AGS cell growth and survival. Experimental results showed that the sub G1, caspase-3 activity, and mitochondrial depolarization were increased. Furthermore, TRPM7 channel overexpression in HEK 293 cells exacerbated UPC-induced cell death. CONCLUSION These findings indicate that UPC inhibits the growth and survival of gastric cancer cells due to a blockade of the TRPM7 channel activity. Therefore, UPC is a potential drug for treatment of gastric cancer, and TRPM7 channels may play an important role in survival in cases of gastric cancer.
Collapse
Affiliation(s)
- Bora Lim
- Division of Longevity and Biofunctional Medicine, Pusan National University School of Korean Medicine, Yangsan, Korea
| | - Hee Jung Lee
- Division of Longevity and Biofunctional Medicine, Pusan National University School of Korean Medicine, Yangsan, Korea
| | - Min Chul Kim
- Division of Longevity and Biofunctional Medicine, Pusan National University School of Korean Medicine, Yangsan, Korea
| | - Byung Joo Kim
- Division of Longevity and Biofunctional Medicine, Pusan National University School of Korean Medicine, Yangsan, Korea
| |
Collapse
|
32
|
Loke SY, Siddiqi NJ, Alhomida AS, Kim HC, Ong WY. Expression and localization of duodenal cytochrome b in the rat hippocampus after kainate-induced excitotoxicity. Neuroscience 2013; 245:179-90. [PMID: 23597830 DOI: 10.1016/j.neuroscience.2013.04.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2012] [Revised: 03/30/2013] [Accepted: 04/06/2013] [Indexed: 11/24/2022]
Abstract
Brain iron accumulation and oxidative stress are common features of many neurodegenerative diseases, and could be due in part to increased iron influx across the blood-brain interface. The iron transport protein, divalent metal transporter 1 (DMT1) is found in reactive astrocytes of the lesioned hippocampal CA fields after excitotoxicity induced by the glutamate analog kainate (KA), but in order for iron to be transported by DMT1, it must be converted from the ferric to the ferrous form. The present study was carried out to investigate the expression of a ferric reductase, duodenal cytochrome b (DCYTB), in the rat hippocampus after KA injury. Quantitative reverse transcriptase-polymerase chain reaction showed significant increases in DCYTB mRNA expression of 2.5, 2.7, and 5.2-fold in the hippocampus at 1week, 2weeks and 1month post-KA lesions respectively compared to untreated controls, and 3.0-fold compared to 1month post-saline injection. DCYTB-positive cells were double labeled with glial fibrillary acidic protein, and electron microscopy showed that the DCYTB-positive cells had dense bundles of glial filaments, characteristic of astrocytes, and were present as end-feet around unlabeled brain capillary endothelial cells. DMT1 labeling in astrocytes and increased iron staining were also observed in the lesioned hippocampus. Together, the present findings of DCYTB and DMT1 localization in astrocytes suggest that DCYTB is a ferric reductase for reduction of ferric iron, for transport by DMT1 into the brain. We postulate that the coordinated action of these two proteins could be important in iron influx across the blood-brain interface, in areas undergoing neurodegeneration.
Collapse
Affiliation(s)
- S-Y Loke
- Department of Anatomy, National University of Singapore, Singapore 119260, Singapore
| | | | | | | | | |
Collapse
|
33
|
Kim BJ. Involvement of melastatin type transient receptor potential 7 channels in ginsenoside Rd-induced apoptosis in gastric and breast cancer cells. J Ginseng Res 2013; 37:201-209. [PMID: 23717173 PMCID: PMC3659640 DOI: 10.5142/jgr.2013.37.201] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2012] [Revised: 12/05/2012] [Accepted: 12/05/2012] [Indexed: 01/16/2023] Open
Abstract
Ginsenoside, one of the active ingredients of Panax ginseng, has a variety of physiologic and pharmacologic effects. The purpose of this study was to explore the effects of ginsenoside Rd (G-Rd) on melastatin type transient receptor potential 7 (TRPM7) channels with respect to the proliferation and survival of AGS and MCF-7 cells (a gastric and a breast cancer cell line, respectively). AGS and MCF-7 cells were treated with different concentrations of G-Rd, and caspase-3 activities, mitochondrial depolarizations, and sub-G1 fractions were analyzed to determine if cell death occurred by apoptosis. In addition, human embryonic kidney (HEK) 293 cells overexpressing TRPM7 channels were used to confirm the role of TRPM7 channels. G-Rd inhibited the proliferation and survival of AGS and MCF-7 cells and enhanced caspase-3 activity, mitochondrial depolarization, and sub-G1 populations. In addition, G-Rd inhibited TRPM7-like currents in AGS and MCF-7 cells and in TRPM7 channel overexpressing HEK 293 cells, as determined by whole cell voltage-clamp recordings. Furthermore, TRPM7 overexpression in HEK 293 cells promoted G-Rd induced cell death. These findings suggest that G-Rd inhibits the proliferation and survival of gastric and breast cancer cells by inhibiting TRPM7 channel activity.
Collapse
Affiliation(s)
- Byung Joo Kim
- Division of Longevity and Biofunctional Medicine, School of Korean Medicine, Pusan National University, Yangsan 626-870, Korea
| |
Collapse
|
34
|
Raval JS, Fontes J, Banerjee U, Yazer MH, Mank E, Palmer AF. Ascorbic acid improves membrane fragility and decreases haemolysis during red blood cell storage. Transfus Med 2013; 23:87-93. [PMID: 23406333 DOI: 10.1111/tme.12013] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2012] [Revised: 12/15/2012] [Accepted: 01/12/2013] [Indexed: 01/29/2023]
Abstract
BACKGROUND Changes that occur to red blood cells (RBCs) during routine blood bank storage include decreased deformability, increased haemolysis and oxidative damage. Oxidative injury to the RBC membrane and haemoglobin can affect changes in shape and deformability. Ascorbic acid (AA) is an antioxidant that maintains haemoglobin in a reduced state and minimises RBC oxidative injury. We hypothesised that AA would improve membrane fragility and decrease haemolysis during storage. METHODS Whole blood derived, AS-5 preserved, pre-storage leucoreduced RBC units were exposed to either AA or saline control solutions. Several rheological and biochemical parameters were measured serially during storage, including RBC membrane mechanical fragility, percent haemolysis and methaemoglobin levels. RESULTS AA exposure significantly reduced mechanical fragility and haemolysis over the entire storage period. The highest two concentrations of AA affected the greatest reductions in mechanical fragility and percent haemolysis. Addition of AA to the RBCs did not significantly alter their biochemical parameters compared to control RBCs incubated with saline. CONCLUSION AA reduced RBC membrane fragility and decreased haemolysis during storage without adversely affecting other RBC biochemical parameters. The clinical significance of these findings needs to be determined.
Collapse
Affiliation(s)
- J S Raval
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | | | | | | | | | | |
Collapse
|
35
|
Corpe CP, Eck P, Wang J, Al-Hasani H, Levine M. Intestinal dehydroascorbic acid (DHA) transport mediated by the facilitative sugar transporters, GLUT2 and GLUT8. J Biol Chem 2013; 288:9092-101. [PMID: 23396969 DOI: 10.1074/jbc.m112.436790] [Citation(s) in RCA: 100] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Intestinal vitamin C (Asc) absorption was believed to be mediated by the Na(+)-dependent ascorbic acid transporter SVCT1. However, Asc transport across the intestines of SVCT1 knock-out mice is normal indicating that alternative ascorbic acid transport mechanisms exist. To investigate these mechanisms, rodents were gavaged with Asc or its oxidized form dehydroascorbic acid (DHA), and plasma Asc concentrations were measured. Asc concentrations doubled following DHA but not Asc gavage. We hypothesized that the transporters responsible were facilitated glucose transporters (GLUTs). Using Xenopus oocyte expression, we investigated whether facilitative glucose transporters GLUT2 and GLUT5-12 transported DHA. Only GLUT2 and GLUT8, known to be expressed in intestines, transported DHA with apparent transport affinities (Km) of 2.33 and 3.23 mm and maximal transport rates (Vmax) of 25.9 and 10.1 pmol/min/oocyte, respectively. Maximal rates for DHA transport mediated by GLUT2 and GLUT8 in oocytes were lower than maximal rates for 2-deoxy-d-glucose (Vmax of 224 and 32 pmol/min/oocyte for GLUT2 and GLUT8, respectively) and fructose (Vmax of 406 and 116 pmol/min/oocyte for GLUT2 and GLUT8, respectively). These findings may be explained by differences in the exofacial binding of substrates, as shown by inhibition studies with ethylidine glucose. DHA transport activity in GLUT2- and GLUT8-expressing oocytes was inhibited by glucose, fructose, and by the flavonoids phloretin and quercetin. These studies indicate intestinal DHA transport may be mediated by the facilitative sugar transporters GLUT2 and GLUT8. Furthermore, dietary sugars and flavonoids in fruits and vegetables may modulate Asc bioavailability via inhibition of small intestinal GLUT2 and GLUT8.
Collapse
Affiliation(s)
- Christopher P Corpe
- Molecular and Clinical Nutrition Section, Digestive Diseases Branch, Intramural Research Program, NIDDK, National Institutes of Health, Bethesda, MD 20892-1372, USA
| | | | | | | | | |
Collapse
|
36
|
Choi J, Masaratana P, Latunde-Dada GO, Arno M, Simpson RJ, McKie AT. Duodenal reductase activity and spleen iron stores are reduced and erythropoiesis is abnormal in Dcytb knockout mice exposed to hypoxic conditions. J Nutr 2012; 142:1929-34. [PMID: 22990466 DOI: 10.3945/jn.112.160358] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Duodenal cytochrome b (Dcytb, Cybrd1) is a ferric reductase localized in the duodenum that is highly upregulated in circumstances of increased iron absorption. To address the contribution of Dcytb to total duodenal ferric reductase activity as well as its wider role in iron metabolism, we first measured duodenal ferric reductase activity in wild-type (WT) and Dcytb knockout (Dcytb(-/-)) mice under 3 conditions known to induce gut ferric reductase: dietary iron deficiency, hypoxia, and pregnancy. Dcytb(-/-) and WT mice were randomly assigned to control (iron deficiency experiment, 48 mg/kg dietary iron; hypoxia experiment, normal atmospheric pressure; pregnancy experiment, nonpregnant animals) or treatment (iron deficiency experiment, 2-3 mg/kg dietary iron; hypoxia experiment, 53.3 kPa pressure; pregnancy experiment, d 20 of pregnancy) groups and duodenal reductase activity measured. We found no induction of ferric reductase activity in Dcytb(-/-) mice under any of these conditions, indicating there are no other inducible ferric reductases present in the duodenum. To test whether Dcytb was required for iron absorption in conditions with increased erythropoietic demand, we also measured tissue nonheme iron levels and hematological indices in WT and Dcytb(-/-) mice exposed to hypoxia. There was no evidence of gross alterations in iron absorption, hemoglobin, or total liver nonheme iron in Dcytb(-/-) mice exposed to hypoxia compared with WT mice. However, spleen nonheme iron was significantly less (6.7 ± 1.0 vs. 12.7 ± 0.9 nmol · mg tissue(-1); P < 0.01, n = 7-8) in hypoxic Dcytb(-/-) compared with hypoxic WT mice and there was evidence of impaired reticulocyte hemoglobinization with a lower reticulocyte mean corpuscular hemoglobin (276 ± 1 vs. 283 ± 2 g · L(-1); P < 0.05, n = 7-8) in normoxic Dcytb(-/-) compared with normoxic WT mice. We therefore conclude that DCYTB is the primary iron-regulated duodenal ferric reductase in the gut and that Dcytb is necessary for optimal iron metabolism.
Collapse
Affiliation(s)
- Jeehyea Choi
- Diabetes and Nutritional Sciences Division, School of Biomedical and Health Sciences, King’s College, London, UK
| | | | | | | | | | | |
Collapse
|
37
|
da Silva GFZ, Shinkarev VP, Kamensky YA, Palmer G. Spectroscopic Evidence of the Role of an Axial Ligand Histidinate in the Mechanism of Adrenal Cytochrome b561. Biochemistry 2012; 51:8730-42. [DOI: 10.1021/bi301127k] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Giordano F. Z. da Silva
- Department
of Biochemistry and Cell Biology, Rice University, Houston, Texas 77251, United States
| | - Vladimir P. Shinkarev
- Department
of Biochemistry and Cell Biology, Rice University, Houston, Texas 77251, United States
| | - Yury A. Kamensky
- Department
of Biochemistry and Cell Biology, Rice University, Houston, Texas 77251, United States
| | - Graham Palmer
- Department
of Biochemistry and Cell Biology, Rice University, Houston, Texas 77251, United States
| |
Collapse
|
38
|
Sadowska-Woda I, Bartosz G. How do erythrocytes contribute to the ABTS* scavenging capacity of blood? Free Radic Res 2012; 47:35-43. [PMID: 23025487 DOI: 10.3109/10715762.2012.734611] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
It has been suggested lately that erythrocytes contribute significantly to the oxidant scavenging capacity (OSC) of blood and that surface adsorption of polyphenols enhances the antioxidant capacity of erythrocytes. The aim of this study was to examine the contribution of erythrocytes to the OSC of whole blood measured with a substrate not penetrating into the cells. Comparison of reduction of 2,2'-azino-bis(3-ethylbenzothiazoline-6-sulphonic acid) radical (ABTS*) by whole blood and blood plasma indicates that erythrocytes do contribute to ABTS* reduction but their contribution is lower with respect to plasma. ABTS* reduction by erythrocytes and its enhancement by polyphenols were inhibited by thiol reagents (N-ethylmaleimide and iodacetate). These reagents inhibited also the reduction of extracellular ferricyanide by erythrocytes and its enhancement by polyphenols. On this basis we postulate that the contribution of erythrocytes to the blood OSC estimated by ABTS* decolorization is at least partly due to the transmembrane-reducing system, which activity is routinely assayed by ferricyanide reduction.
Collapse
|
39
|
Kim BJ. Involvement of Transient Receptor Potential Melastatin 7 Channels in Sophorae Radix-induced Apoptosis in Cancer Cells: Sophorae Radix and TRPM7. J Pharmacopuncture 2012; 15:31-38. [PMID: 25780645 PMCID: PMC4331944 DOI: 10.3831/kpi.2012.15.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2012] [Accepted: 09/10/2012] [Indexed: 12/15/2022] Open
Abstract
UNLABELLED Sophorae Radix (SR) plays a role in a number of physiologic and pharmacologic functions in many organs. OBJECTIVE The aim of this study was to clarify the potential role for transient receptor potential melastatin 7 (TRPM7) channels in SR-inhibited growth and survival of AGS and MCF-7 cells, the most common human gastric and breast adenocarcinoma cell lines. METHODS The AGS and the MCF-7 cells were treated with varying concentrations of SR. Analyses of the caspase-3 and - 9 activity, the mitochondrial depolarization and the poly (ADPribose) polymerase (PARP) cleavage were conducted to determine if AGS and MCF-7 cell death occured by apoptosis. TRPM7 channel blockers (Gd(3+) or 2-APB) and small interfering RNA (siRNA) were used in this study to confirm the role of TRPM7 channels. Furthermore, TRPM7 channels were overexpressed in human embryonic kidney (HEK) 293 cells to identify the role of TRPM7 channels in AGS and MCF-7 cell growth and survival. RESULTS The addition of SR to a culture medium inhibited AGS and MCF-7 cell growth and survival. Experimental results showed that the caspase-3 and -9 activity, the mitochondrial depolarization, and the degree of PARP cleavage was increased. TRPM7 channel blockade, either by Gd(3+) or 2-APB or by suppressing TRPM7 expression with small interfering RNA, blocked the SR-induced inhibition of cell growth and survival. Furthermore, TRPM7 channel overexpression in HEK 293 cells exacerbated SR-induced cell death. CONCLUSIONS These findings indicate that SR inhibits the growth and survival of gastric and breast cancer cells due to a blockade of the TRPM7 channel activity. Therefore, TRPM7 channels may play an important role in the survival of patients with gastric and breast cancer.
Collapse
Affiliation(s)
- Byung Joo Kim
- Division of Longevity and Biofunctional Medicine, Pusan National University School of Korean Medicine, Yangsan, Korea
| |
Collapse
|
40
|
Li H, Tu H, Wang Y, Levine M. Vitamin C in mouse and human red blood cells: an HPLC assay. Anal Biochem 2012; 426:109-17. [PMID: 22522059 DOI: 10.1016/j.ab.2012.04.014] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2011] [Revised: 03/22/2012] [Accepted: 04/06/2012] [Indexed: 11/27/2022]
Abstract
Although vitamin C (ascorbate) is present in whole blood, measurements in red blood cells (RBCs) are problematic because of interference, instability, limited sensitivity, and sample volume requirements. We describe a new technique using HPLC with coulometric electrochemical detection for ascorbate measurement in RBCs of humans, wild-type mice, and mice unable to synthesize ascorbate. Exogenously added ascorbate was fully recovered even when endogenous RBC ascorbate was below the detection threshold (25 nM). Twenty microliters of whole blood or 10 μl of packed RBCs was sufficient for assay. RBC ascorbate was stable for 24h from whole-blood samples at 4°C. Processed, stored samples were stable for >1 month at -80°C. Unlike other tissues, ascorbate concentrations in human and mouse RBCs were linear in relation to plasma concentrations (R=0.8 and 0.9, respectively). In healthy humans, RBC ascorbate concentrations were 9-57 μM, corresponding to ascorbate plasma concentrations of 15-90 μM. Mouse data were similar. In human blood stored as if for transfusion, initial RBC ascorbate concentrations varied approximately sevenfold and decreased 50% after 6 weeks of storage under clinical conditions. With this assay, it becomes possible for the first time to characterize ascorbate function in relation to endogenous concentrations in RBCs.
Collapse
Affiliation(s)
- Hongyan Li
- Molecular and Clinical Nutrition Section, Digestive Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892-1372, USA
| | | | | | | |
Collapse
|
41
|
Cenacchi L, Busch M, Schleidt PG, Müller FG, Stumpp TVM, Mäntele W, Trost P, Lancaster CRD. Heterologous production and characterisation of two distinct dihaem-containing membrane integral cytochrome b(561) enzymes from Arabidopsis thaliana in Pichia pastoris and Escherichia coli cells. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2011; 1818:679-88. [PMID: 22085541 DOI: 10.1016/j.bbamem.2011.10.030] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2011] [Revised: 10/29/2011] [Accepted: 10/31/2011] [Indexed: 01/07/2023]
Abstract
Cytochrome (cyt) b(561) proteins are dihaem-containing membrane proteins, belonging to the CYBASC (cytochrome-b(561)-ascorbate-reducible) family, and are proposed to be involved in ascorbate recycling and/or the facilitation of iron absorption. Here, we present the heterologous production of two cyt b(561) paralogs from Arabidopsis thaliana (Acytb(561)-A, Acytb(561)-B) in Escherichia coli and Pichia pastoris, their purification, and initial characterisation. Spectra indicated that Acytb(561)-A resembles the best characterised member of the CYBASC family, the cytochrome b(561) from adrenomedullary chromaffin vesicles, and that Acytb(561)-B is atypical compared to other CYBASC proteins. Haem oxidation-reduction midpoint potential (E(M)) values were found to be fully consistent with ascorbate oxidation activities and Fe(3+)-chelates reductase activities. The ascorbate dependent reduction and protein stability of both paralogs were found to be sensitive to alkaline pH values as reported for the cytochrome b(561) from chromaffin vesicles. For both paralogs, ascorbate-dependent reduction was inhibited and the low-potential haem E(M) values were affected significantly by incubation with diethyl pyrocarbonate (DEPC) in the absence of ascorbate. Modification with DEPC in the presence of ascorbate left the haem E(M) values unaltered compared to the unmodified proteins. However, ascorbate reduction was inhibited. We concluded that the ascorbate-binding site is located near the low-potential haem with the Fe(3+)-chelates reduction-site close to the high-potential haem. Furthermore, inhibition of ascorbate oxidation by DEPC treatment occurs not only by lowering the haem E(M) values but also by an additional modification affecting ascorbate binding and/or electron transfer. Analytical gel filtration experiments suggest that both cyt b(561) paralogs exist as homodimers.
Collapse
Affiliation(s)
- Lucia Cenacchi
- Max Planck Institute of Biophysics, Department of Molecular Membrane Biology, Frankfurt, Germany
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Kim BJ, Nah SY, Jeon JH, So I, Kim SJ. Transient receptor potential melastatin 7 channels are involved in ginsenoside Rg3-induced apoptosis in gastric cancer cells. Basic Clin Pharmacol Toxicol 2011; 109:233-239. [PMID: 21443732 DOI: 10.1111/j.1742-7843.2011.00706.x] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Ginsenosides play a role in a number of physiological and pharmacological functions in the gastrointestinal tract. The aim of this study was to clarify the potential role for transient receptor potential melastatin 7 (TRPM7) channels in ginsenoside Rg3-inhibited growth and survival of AGS cells, the most common human gastric adenocarcinoma cell line. The AGS cells were treated with varying concentrations of Rg3. Sub-G1 analysis, caspase-3 activity and poly(ADP-ribose) polymerase (PARP) cleavage analysis were conducted to determine whether AGS cell death occurs by apoptosis. TRPM7 channel blockers (La(3+) or 2-APB) and small interfering RNA (siRNA) were used in this study to confirm the role of TRPM7 channels. Furthermore, TRPM7 channels were over-expressed in human embryonic kidney (HEK) 293 cells to identify the role of TRPM7 channels in AGS cell growth and survival. The addition of Rg3 to the culture medium inhibited AGS growth and survival. Experimental results showed sub-G1 was markedly increased, caspase-3 activity was elevated, and degree of PARP cleavage was increased. TRPM7 channel blockade, either by La(3+) or 2-APB or by suppressing TRPM7 expression with siRNA, blocked the Rg3-induced inhibition of cell growth and survival. Furthermore, TRPM7 channel over-expression in HEK 293 cells exacerbated Rg3-induced cell death. These findings indicate that ginsenoside Rg3 inhibits the growth and survival of gastric cancer cell which is because of the blockade of TRPM7 channel activity. Therefore, TRPM7 channels may play an important role in the survival of gastric cancer.
Collapse
Affiliation(s)
- Byung Joo Kim
- Division of Longevity and Biofunctional Medicine, Pusan National University School of Korean Medicine, Yangsan, Korea
| | | | | | | | | |
Collapse
|
43
|
Liu W, Wu G, Tsai AL, Kulmacz RJ. High-yield production, purification and characterization of functional human duodenal cytochrome b in an Escherichia coli system. Protein Expr Purif 2011; 79:115-21. [PMID: 21501687 PMCID: PMC3133858 DOI: 10.1016/j.pep.2011.04.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2011] [Revised: 03/29/2011] [Accepted: 04/01/2011] [Indexed: 11/15/2022]
Abstract
Human duodenal cytochrome b (Dcytb) is a transmembrane hemoprotein found in the duodenal brush border membrane and in erythrocytes. Dcytb has been linked to uptake of dietary iron and to ascorbate recycling in erythrocytes. Detailed biophysical and biochemical characterization of Dcytb has been limited by difficulties in expressing sufficient amounts of functional recombinant protein in yeast and insect cell systems. We have developed an Escherichia coli Rosetta-gami B(DE3) cell system for production of recombinant His-tagged human Dcytb with a yield of ∼26 mg of purified, ascorbate-reducible cytochrome per liter of culture. The recombinant protein is readily solubilized with n-dodecyl-β-D-maltoside and purified to electrophoretic homogeneity by one-step chromatography on cobalt affinity resin. The purified recombinant Dcytb has a heme to protein ratio very close to the theoretical value of 2 and retains functional reactivity with ascorbate, as assessed by spectroscopic and kinetic measurements. Ascorbate showed a marked kinetic selectivity for the high-potential heme center over the low-potential heme center in purified Dcytb. This new E. coli expression system for Dcytb offers ∼7-fold improvement in yield and other substantial advantages over existing expression systems for reliable production of functional Dcytb at levels suitable for biochemical, biophysical and structural characterization.
Collapse
Affiliation(s)
- Wen Liu
- Department of Internal Medicine, University of Texas Health Science Center, Houston, TX 77030, United States.
| | | | | | | |
Collapse
|
44
|
Del Principe D, Avigliano L, Savini I, Catani MV. Trans-plasma membrane electron transport in mammals: functional significance in health and disease. Antioxid Redox Signal 2011; 14:2289-318. [PMID: 20812784 DOI: 10.1089/ars.2010.3247] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Trans-plasma membrane electron transport (t-PMET) has been established since the 1960s, but it has only been subject to more intensive research in the last decade. The discovery and characterization at the molecular level of its novel components has increased our understanding of how t-PMET regulates distinct cellular functions. This review will give an update on t-PMET, with particular emphasis on how its malfunction relates to some diseases, such as cancer, abnormal cell death, cardiovascular diseases, aging, obesity, neurodegenerative diseases, pulmonary fibrosis, asthma, and genetically linked pathologies. Understanding these relationships may provide novel therapeutic approaches for pathologies associated with unbalanced redox state.
Collapse
Affiliation(s)
- Domenico Del Principe
- Department of Experimental Medicine and Biochemical Sciences, University of Rome Tor Vergata, Rome, Italy.
| | | | | | | |
Collapse
|
45
|
Liu W, da Silva GF, Wu G, Palmer G, Tsai AL, Kulmacz RJ. Functional and structural roles of residues in the third extramembrane segment of adrenal cytochrome b561. Biochemistry 2011; 50:3149-60. [PMID: 21401125 PMCID: PMC3099539 DOI: 10.1021/bi101796m] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Several residues in the third extramembrane segment (EM3) of adrenal cytochrome b(561) have been proposed to be involved in this cytochrome's interaction with ascorbate, but there has been no systematic evaluation of residues in the segment. We used alanine scanning mutagenesis to assess the functional and structural roles of the EM3 residues and several adjacent residues (residues 70-85) in the bovine cytochrome. Each alanine mutant was expressed in a bacterial system, solubilized with detergent, and affinity-purified. The recombinant proteins contained approximately two hemes per monomer and, except for R74A, retained basic functionality (≥ 94% reduced by 20 mM ascorbate). Equilibrium spectrophotometric titrations with ascorbate were used to analyze the α-band line shape and amplitude during reduction of the high- and low-potential heme centers (b(H) and b(L), respectively) and the midpoint ascorbate concentrations for the b(H) and b(L) transitions (C(H) and C(L), respectively). Y73A and K85A markedly narrowed the b(H) α-band peak; other mutants had weaker effects or no effect on b(H) or b(L) spectra. Relative changes in C(H) for the mutants were larger than changes in C(L), with 1.5-2.9-fold increases in C(H) for L70A, L71A, Y73A, R74A, N78A, and K85A. The amounts of functional b(H) and b(L) centers in additional Arg74 mutants, assessed by ascorbate titration and EPR spectroscopy, declined in concert in the following order: wild type > R74K > R74Q > R74T and R74Y > R74E. The results of this first comprehensive experimental test of the proposed roles of EM3 residues have identified residues with a direct or indirect impact on ascorbate interactions, on the environment of the b(H) heme center, and on formation of the native b(H)-b(L) unit. Surprisingly, no individual EM3 residue was by itself indispensable for the interaction with ascorbate, and the role of the segment appears to be more subtle than previously thought. These results also support our topological model of the adrenal cytochrome, which positions b(H) near the cytoplasmic side of the membrane.
Collapse
Affiliation(s)
- Wen Liu
- Department of Internal Medicine, University of Texas Health Science Center, Houston, TX 77030
| | - Giordano F.Z. da Silva
- Department of Internal Medicine, University of Texas Health Science Center, Houston, TX 77030
- Department of Biochemistry & Cell Biology, Rice University, Houston, TX 77251
| | - Gang Wu
- Department of Internal Medicine, University of Texas Health Science Center, Houston, TX 77030
| | - Graham Palmer
- Department of Biochemistry & Cell Biology, Rice University, Houston, TX 77251
| | - Ah-Lim Tsai
- Department of Internal Medicine, University of Texas Health Science Center, Houston, TX 77030
| | - Richard J. Kulmacz
- Department of Internal Medicine, University of Texas Health Science Center, Houston, TX 77030
| |
Collapse
|
46
|
Chepelev NL, Willmore WG. Regulation of iron pathways in response to hypoxia. Free Radic Biol Med 2011; 50:645-66. [PMID: 21185934 DOI: 10.1016/j.freeradbiomed.2010.12.023] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2010] [Revised: 12/13/2010] [Accepted: 12/15/2010] [Indexed: 10/24/2022]
Abstract
Constituting an integral part of a heme's porphyrin ring, iron is essential for supplying cells and tissues with oxygen. Given tight links between oxygen delivery and iron availability, it is not surprising that iron deprivation and oxygen deprivation (hypoxia) have very similar consequences at the molecular level. Under hypoxia, the expression of major iron homeostasis genes including transferrin, transferrin receptor, ceruloplasmin, and heme oxygenase-1 is activated by hypoxia-inducible factors to provide increased iron availability for erythropoiesis in an attempt to enhance oxygen uptake and delivery to hypoxic cells. Iron-response proteins (IRP1 and IRP2) and "cap-n-collar" bZIP transcriptional factors (NE-F2 p45; Nrf1, 2, and 3; Bach1 and 2) also control gene and protein expression of the key iron homeostasis proteins. In this article, we give an overview of the mechanisms by which iron pathways are regulated by hypoxia at multiple levels. In addition, potential clinical benefits of manipulating iron pathways in the hypoxia-related conditions anemia and ischemia are discussed.
Collapse
|
47
|
Rizvi SI, Pandey KB. Activation of the erythrocyte plasma membrane redox system by resveratrol: a possible mechanism for antioxidant properties. Pharmacol Rep 2011; 62:726-32. [PMID: 20885013 DOI: 10.1016/s1734-1140(10)70330-3] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2009] [Revised: 01/11/2010] [Indexed: 12/13/2022]
Abstract
Resveratrol is one of the most widely studied of all the plant-produced polyphenols and has diverse, beneficial health effects including anti-cancer and cardio-protective effects. Many of the biological actions of this polyphenol have been attributed to its antioxidant properties. Erythrocytes contain a plasma membrane redox system (PMRS), which transfers electrons from intracellular donors (NADH and/or ascorbate (ASC)) to extracellular acceptors. There is evidence that the intracellular ASC donates electrons to extracellular ascorbate free radicals (AFRs) via the PMRS, which encompasses an AFR reductase; such a redox system enables the cells to effectively counteract oxidative processes.We present evidence to show that human erythrocytes take up resveratrol, and once inside the cell, resveratrol can donate electrons to extracellular electron acceptors through the erythrocyte PMRS and AFR reductase. Incubating human erythrocytes with resveratrol (10 μM) caused a significant activation of the PMRS (41%) and AFR reductase (30%) over (basal level) the control; the effect of resveratrol was concentration-dependent. The electron donating ability of resveratrol is slightly less than that observed with quercetin. The role of resveratrol in activating the erythrocyte PMRS and AFR reductase may assume significance in all disease conditions in which there is a decrease in plasma antioxidant potential.
Collapse
Affiliation(s)
- Syed Ibrahim Rizvi
- Department of Biochemistry, University of Allahabad, Allahabad 211002, India.
| | | |
Collapse
|
48
|
Buehler PW, D’Agnillo F, Schaer DJ. Hemoglobin-based oxygen carriers: from mechanisms of toxicity and clearance to rational drug design. Trends Mol Med 2010; 16:447-57. [DOI: 10.1016/j.molmed.2010.07.006] [Citation(s) in RCA: 105] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2010] [Revised: 07/14/2010] [Accepted: 07/16/2010] [Indexed: 12/19/2022]
|
49
|
A role for Na+/H+ exchangers and intracellular pH in regulating vitamin C-driven electron transport across the plasma membrane. Biochem J 2010; 428:191-200. [DOI: 10.1042/bj20100064] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Ascorbate (vitamin C) is the major electron donor to a tPMET (transplasma membrane electron transport) system that was originally identified in human erythrocytes. This plasma membrane redox system appears to transfer electrons from intracellular ascorbate to extracellular oxidants (e.g. non-transferrin-bound iron). Although this phenomenon has been observed in nucleated cells, its mechanism and regulation are not well understood. In the present study we have examined both facets of this phenomenon in K562 cells and primary astrocyte cultures. Using ferricyanide as the analytical oxidant we demonstrate that tPMET is enhanced by dehydroascorbate uptake via facilitative glucose transporters, and subsequent accumulation of intracellular ascorbate. Additionally, we demonstrate that this stimulation is not due to ascorbate that is released from the cells, but is dependent only on a restricted intracellular pool of the vitamin. Substrate-saturation kinetics suggest an enzyme-catalysed reaction across the plasma membrane by an as-yet-unidentified reductase that relies on extensive recycling of intracellular ascorbate. Inhibition of ascorbate-stimulated tPMET by the NHE (Na+/H+-exchanger) inhibitors amiloride and 5-(N-ethyl-N-isopropyl)amiloride, which is diminished by bicarbonate, suggests that tPMET activity may be regulated by intracellular pH. In support of this hypothesis, tPMET in astrocytes was significantly inhibited by ammonium chloride-pulse-induced intracellular acidification, whereas it was significantly stimulated by bicarbonate-induced intracellular alkalinization. These results suggest that ascorbate-dependent tPMET is enzyme-catalysed and is modulated by NHE activity and intracellular pH.
Collapse
|
50
|
Activation of erythrocyte plasma membrane redox system provides a useful method to evaluate antioxidant potential of plant polyphenols. Methods Mol Biol 2010. [PMID: 20072929 DOI: 10.1007/978-1-60761-411-1_24] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2024]
Abstract
Plant polyphenols are known to possess antioxidant activities. In recent years, there have been numerous reports confirming the efficacy of these compounds to improve plasma antioxidant capacity in humans. Current methods to evaluate the antioxidant potential of polyphenols are based on in vitro assay procedures (TEAC, ORAC, FRAP, DPPH). However, the antioxidant potential assessed by these methods does not correlate with the biological activity observed in vivo. Eukaryotic cells display a plasma membrane redox system (PMRS) that transfers electrons from intracellular substrates to extracellular electron acceptors. Here, we describe a method to evaluate the antioxidant potential of plant polyphenols based on their ability to enter the erythrocytes and donate electrons to PMRS. We also present results to show the potentiating effect of quercetin, EGCG, EC and catechin on erythrocyte PMRS activity.
Collapse
|