1
|
Fortuna V, Lima J, Oliveira GF, Oliveira YS, Getachew B, Nekhai S, Aschner M, Tizabi Y. Ferroptosis as an emerging target in sickle cell disease. Curr Res Toxicol 2024; 7:100181. [PMID: 39021403 PMCID: PMC11252799 DOI: 10.1016/j.crtox.2024.100181] [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: 12/30/2023] [Revised: 06/13/2024] [Accepted: 06/17/2024] [Indexed: 07/20/2024] Open
Abstract
Sickle cell disease (SCD) is an inherited hemoglobin disorder marked by red blood cell sickling, resulting in severe anemia, painful episodes, extensive organ damage, and shortened life expectancy. In SCD, increased iron levels can trigger ferroptosis, a specific type of cell death characterized by reactive oxygen species (ROS) and lipid peroxide accumulation, leading to damage and organ impairments. The intricate interplay between iron, ferroptosis, inflammation, and oxidative stress in SCD underscores the necessity of thoroughly understanding these processes for the development of innovative therapeutic strategies. This review highlights the importance of balancing the complex interactions among various factors and exploitation of the knowledge in developing novel therapeutics for this devastating disease.
Collapse
Affiliation(s)
- Vitor Fortuna
- Department of Biochemistry and Biophysics, Health Sciences Institute, Federal University of Bahia, BA 40231-300, Brazil
- Postgraduate Program in Immunology, Health Sciences Institute, Federal University of Bahia, BA 40231-300, Brazil
| | - Jaqueline Lima
- Postgraduate Program in Immunology, Health Sciences Institute, Federal University of Bahia, BA 40231-300, Brazil
| | - Gabriel F. Oliveira
- Postgraduate Program in Immunology, Health Sciences Institute, Federal University of Bahia, BA 40231-300, Brazil
| | - Yasmin S. Oliveira
- Postgraduate Program in Immunology, Health Sciences Institute, Federal University of Bahia, BA 40231-300, Brazil
| | - Bruk Getachew
- Department of Pharmacology, Howard University College of Medicine, Washington, DC, USA
| | - Sergei Nekhai
- Center for Sickle Cell Disease, Departments of Microbiology and Medicine, Howard University College of Medicine, Washington, DC, USA
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Yousef Tizabi
- Department of Pharmacology, Howard University College of Medicine, Washington, DC, USA
| |
Collapse
|
2
|
William N, Acker JP. A perspective on exogenous redox regulation mediated by transfused RBCs subject to the storage lesion. Transfus Apher Sci 2024; 63:103929. [PMID: 38658294 DOI: 10.1016/j.transci.2024.103929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Granted with a potent ability to interact with and tolerate oxidative stressors, RBCs scavenge most reactive oxygen and nitrogen species (RONS) generated in circulation. This essential non-canonical function, however, renders RBCs susceptible to damage when vascular RONS are generated in excess, making vascular redox imbalance a common etiology of anemia, and thus a common indication for transfusion. This accentuates the relevance of impairments in redox metabolism during hypothermic storage, as the exposure to chronic oxidative stressors upon transfusion could be exceedingly deleterious to stored RBCs. Herein, we review the prominent mechanisms of the hypothermic storage lesion that alter the ability of RBCs to scavenge exogenous RONS as well as the associated clinical relevance.
Collapse
Affiliation(s)
- Nishaka William
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, Canada
| | - Jason P Acker
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, Canada; Innovation and Portfolio Management, Canadian Blood Services, Edmonton, Alberta, Canada.
| |
Collapse
|
3
|
Smiianov VV, Fartushok TV, Fedevych YM, Fartushok NV. New view on the compatibility of hemoglobin function in the erythrocytes. WIADOMOSCI LEKARSKIE (WARSAW, POLAND : 1960) 2024; 77:9-16. [PMID: 38431801 DOI: 10.36740/wlek202401101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2024]
Abstract
OBJECTIVE Aim: To study the process of hemoglobin oxidation and the enzymatic reactions associated with it. PATIENTS AND METHODS Materials and Methods: Heparinized human blood (15 IU/ml) was obtained from the clinical department. The concentration of oxy- and methemoglobin, auto-oxidation of hemoglobin was determined spectrophotometrically spectrophotometrically. Autooxidation of hemoglobin was recorded spectrophotometrically, and protein concentration was determined by the Lowry method. Monooxygenase activity of hemoglobin was also measured by the method described by Lowry spectrophotometrically. The concentration of O2 and H2O2 in the reaction media was determined on a biomicroanalyzer OR 210/3 (Redelkis). RESULTS Results: The obtained experimental data allow us to propose a mechanism of "spontaneous autooxidation" of oxyhemoglobin, which can be described by the following equations: Hb2+O2 → Hb3+ + O2 - (1) Hb2+O2 + 2e - + 2H+ →Hb3+ + H2O2 (2) Hb2+O2 + 2e - + 2H+ →Hb2+ + H2O2 (3) Hb2+ + O2 →Hb2+O2 (4) Spectral characteristics of the process of "spontaneous auto-oxidation" indicate the formation of a metform of hemoglobin, the depletion of oxygen by the system was established, at pH 5.6, an increase in the monooxygenase activity of hemoglobin is observed 3-4 times compared to the physiological level. CONCLUSION Сonclusions: In addition to the main, previously known functions of hemoglobin (gas transport, peroxidase, monooxygenase), it catalyzes a two-electron oxidase reaction in which O2 is reduced to H2O2. This is confirmed by experimental data on the formation of one of the products of "spontaneous autoxidation" of oxyhemoglobin _ deoxyform at pH 5.6 _ 8.9.
Collapse
Affiliation(s)
| | | | - Yuri M Fedevych
- DANYLO HALYTSKY LVIV NATIONAL MEDICAL UNIVERSITY, LVIV UKRAINE
| | | |
Collapse
|
4
|
Sharma M, Khan FH, Mahmood R. Nickel chloride generates cytotoxic ROS that cause oxidative damage in human erythrocytes. J Trace Elem Med Biol 2023; 80:127272. [PMID: 37516010 DOI: 10.1016/j.jtemb.2023.127272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 06/07/2023] [Accepted: 07/23/2023] [Indexed: 07/31/2023]
Abstract
BACKGROUND Nickel is a heavy metal that is regarded as a possible hazard to living organisms due to its toxicity and carcinogenicity. Nickel chloride (NiCl2), an inorganic divalent Ni compound, has been shown to cause oxidative stress in cells by altering the redox equilibrium. We have investigated the effect of NiCl2 on isolated human erythrocytes under in vitro condition. METHODS Isolated erythrocytes were treated with different concentrations of NiCl2 (25-500 µM) for 24 h at 37 ºC. Hemolysates were prepared and several biochemical parameters were analyzed in them. RESULTS Treatment of erythrocytes with NiCl2 enhanced the intracellular generation of reactive oxygen species (ROS). A significant increase in hydrogen peroxide levels and oxidation of proteins and lipids was also seen. This was accompanied by a reduction in levels of nitric oxide, glutathione, free amino groups and total sulfhydryl groups. NiCl2 treatment impaired both enzymatic and non-enzymatic defense systems, resulting in lowered antioxidant capacity and diminished ability of cells to quench free radicals and reduce metal ions. NiCl2 exposure also had an inhibitory effect on the activity of enzymes involved in pathways of glucose metabolism (glycolytic and pentose phosphate shunt pathways). Increased level of methemoglobin, which is inactive in oxygen transport, was also seen. The rate of heme breakdown increased resulting in the release of free iron. Exposure to NiCl2 led to considerable cell lysis, indicating damage to the erythrocyte membrane. This was supported by the inhibition of membrane bound enzymes and increase in the osmotic fragility of NiCl2 treated cells. NiCl2 treatment caused severe morphological alterations with the conversion of normal discocytes to echinocytes. All changes were seen in a NiCl2 concentration-dependent manner. CONCLUSION NiCl2 generates cytotoxic ROS in human erythrocytes which cause oxidative damage that can decrease the oxygen carrying capacity of blood and also lead to anemia.
Collapse
Affiliation(s)
- Monika Sharma
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh 202002, U.P., India
| | - Fahim Halim Khan
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh 202002, U.P., India
| | - Riaz Mahmood
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh 202002, U.P., India.
| |
Collapse
|
5
|
Hamali HA. Hypercoagulability in Sickle Cell Disease: A Thrombo-Inflammatory Mechanism. Hemoglobin 2023; 47:205-214. [PMID: 38189099 DOI: 10.1080/03630269.2023.2301026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 12/28/2023] [Indexed: 01/09/2024]
Abstract
Sickle cell disease (SCD) is a group of inherited disorders characterized by the presence of abnormal hemoglobin S. Patients with SCD suffer from frequent episodes of anemia, chronic hemolysis, pain crisis, and vaso-occlusion. Additionally, SCD is associated with diverse and serious clinical complications, including thrombosis, which can lead to organ failure, increased morbidity, and eventually, mortality. SCD is known to be a hypercoagulable condition, and the cause of hypercoagulability is multifactorial, with the molecular basis of hemoglobin S being the main driver. The presence of hemoglobin S induces sickling of the RBCs and their subsequent hemolysis, as well as oxidative stress. Both of these processes can alter the hemostatic system, through the activation of platelets, coagulation system, and fibrinolysis, as well as depletion of coagulation inhibitors. These changes can also induce the formation of microvesicles and expression of tissue factor, leading to activation of WBCs, endothelial cell damage, and inflammatory response. Understanding the various factors that drive hypercoagulability as a thrombo-inflammatory mechanism in SCD can help provide explanations for the pathogenesis and other complications of the disease.
Collapse
Affiliation(s)
- Hassan A Hamali
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, Jazan University, Gizan, Saudi Arabia
| |
Collapse
|
6
|
Georgiou-Siafis SK, Tsiftsoglou AS. The Key Role of GSH in Keeping the Redox Balance in Mammalian Cells: Mechanisms and Significance of GSH in Detoxification via Formation of Conjugates. Antioxidants (Basel) 2023; 12:1953. [PMID: 38001806 PMCID: PMC10669396 DOI: 10.3390/antiox12111953] [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: 09/30/2023] [Revised: 10/18/2023] [Accepted: 10/23/2023] [Indexed: 11/26/2023] Open
Abstract
Glutathione (GSH) is a ubiquitous tripeptide that is biosynthesized in situ at high concentrations (1-5 mM) and involved in the regulation of cellular homeostasis via multiple mechanisms. The main known action of GSH is its antioxidant capacity, which aids in maintaining the redox cycle of cells. To this end, GSH peroxidases contribute to the scavenging of various forms of ROS and RNS. A generally underestimated mechanism of action of GSH is its direct nucleophilic interaction with electrophilic compounds yielding thioether GSH S-conjugates. Many compounds, including xenobiotics (such as NAPQI, simvastatin, cisplatin, and barbital) and intrinsic compounds (such as menadione, leukotrienes, prostaglandins, and dopamine), form covalent adducts with GSH leading mainly to their detoxification. In the present article, we wish to present the key role and significance of GSH in cellular redox biology. This includes an update on the formation of GSH-S conjugates or GSH adducts with emphasis given to the mechanism of reaction, the dependence on GST (GSH S-transferase), where this conjugation occurs in tissues, and its significance. The uncovering of the GSH adducts' formation enhances our knowledge of the human metabolome. GSH-hematin adducts were recently shown to have been formed spontaneously in multiples isomers at hemolysates, leading to structural destabilization of the endogenous toxin, hematin (free heme), which is derived from the released hemoglobin. Moreover, hemin (the form of oxidized heme) has been found to act through the Kelch-like ECH associated protein 1 (Keap1)-nuclear factor erythroid 2-related factor-2 (Nrf2) signaling pathway as an epigenetic modulator of GSH metabolism. Last but not least, the implications of the genetic defects in GSH metabolism, recorded in hemolytic syndromes, cancer and other pathologies, are presented and discussed under the framework of conceptualizing that GSH S-conjugates could be regarded as signatures of the cellular metabolism in the diseased state.
Collapse
Affiliation(s)
| | - Asterios S. Tsiftsoglou
- Laboratory of Pharmacology, Department of Pharmaceutical Sciences, School of Health Sciences, Aristotle University of Thessaloniki (AUTh), 54124 Thessaloniki, Greece;
| |
Collapse
|
7
|
Choi DH, Violet PC, Majumdar S, Levine M. Red Blood Cell Vitamin C Concentration and Its Effect on Deformability in Pediatric Sickle Cell Disease. J Pediatr Hematol Oncol 2023; 45:e936-e939. [PMID: 36897309 PMCID: PMC10423298 DOI: 10.1097/mph.0000000000002631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 12/20/2022] [Indexed: 03/11/2023]
Abstract
Red cell rigidity is common in sickle cell disease (SCD). The contribution of oxidative stress on deformability remains unknown. This study investigated red blood cell (RBC) vitamin C concentrations in pediatric SCD (n=43) compared with healthy controls ( n =23) and developed a protocol to raise RBC vitamin C concentrations to measure the effect on deformability. Sickle cell RBC vitamin C concentrations seem low (20.5 μM, SD: 16.2 vs. 51.7 μM, SD: 15.8; P <0.0001). Vitamin C can be successfully loaded into sickle cell RBCs but seems to have minimal effect on deformability. Future studies are needed to understand the clinical implications of vitamin C deficiency in pediatric SCD.
Collapse
Affiliation(s)
- Daniel H. Choi
- Division of Critical Care Medicine, Department of Pediatrics, Children’s National Hospital, George Washington School of Medicine and Health Sciences, Washington, DC, USA
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, Hasbro Children’s Hospital, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Pierre-Christian Violet
- Molecular and Clinical Nutrition Section, Digestive Disease Branch, Intramural Research Program, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Suvankar Majumdar
- Division of Hematology, Department of Pediatrics, Children’s National Hospital, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Mark Levine
- Molecular and Clinical Nutrition Section, Digestive Disease Branch, Intramural Research Program, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
8
|
Rupa SA, Patwary MAM, Matin MM, Ghann WE, Uddin J, Kazi M. Interaction of mercury species with proteins: towards possible mechanism of mercurial toxicology. Toxicol Res (Camb) 2023; 12:355-368. [PMID: 37397928 PMCID: PMC10311172 DOI: 10.1093/toxres/tfad039] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 05/02/2023] [Accepted: 05/15/2023] [Indexed: 07/04/2023] Open
Abstract
The nature of the binding of mercurials (organic and inorganic) and their subsequent transformations in biological systems is a matter of great debate as several different hypotheses have been proposed and none of them has been conclusively proven to explain the characteristics of Hg binding with the proteins. Thus, the chemical nature of Hg-protein binding through the possible transportation mechanism in living tissues is critically reviewed herein. Emphasis is given to the process of transportation, and binding of Hg species with selenol-containing biomolecules that are appealing for toxicological studies as well as the advancement of environmental and biological research.
Collapse
Affiliation(s)
| | | | | | - William Emmanuel Ghann
- Center for Nanotechnology, Department of Natural Sciences, Coppin State University, Baltimore, MD 21216, USA
| | - Jamal Uddin
- Center for Nanotechnology, Department of Natural Sciences, Coppin State University, Baltimore, MD 21216, USA
| | - Mohsin Kazi
- Department of Pharmaceutics, College of Pharmacy, King Saud University, P.O. BOX-2457, Riyadh 11451, Saudi Arabia
| |
Collapse
|
9
|
Quds R, Iqbal Z, Arif A, Mahmood R. Mancozeb-induced cytotoxicity in human erythrocytes: enhanced generation of reactive species, hemoglobin oxidation, diminished antioxidant power, membrane damage and morphological changes. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2023; 193:105453. [PMID: 37248021 DOI: 10.1016/j.pestbp.2023.105453] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/21/2023] [Accepted: 05/02/2023] [Indexed: 05/31/2023]
Abstract
Mancozeb is an ethylene bis-dithiocarbamate fungicide extensively used in agriculture to safeguard crops from various fungal diseases. The general population is exposed to mancozeb through consumption of contaminated food or water. Here, we have investigated the effect of mancozeb on isolated human erythrocytes under in vitro conditions. Erythrocytes were treated with different concentrations of mancozeb (0, 5, 10, 25, 50, 100 μM) and incubated for 24 h at 37 °C. Analysis of biochemical parameters and cell morphology showed dose-dependent toxicity of mancozeb in human erythrocytes. Mancozeb treatment caused hemoglobin oxidation and heme degradation. Protein and lipid oxidation were enhanced, while a significant decrease was seen in reduced glutathione and total sulfhydryl content. A significant increase in the generation of reactive oxygen and nitrogen species was detected in mancozeb-treated erythrocytes. The antioxidant capacity and the activity of key antioxidant enzymes were greatly diminished, while crucial metabolic pathways were inhibited in erythrocytes. Damage to the erythrocyte membrane on mancozeb treatment was apparent from increased cell lysis and osmotic fragility, along with the impairment of the plasma membrane redox system. Mancozeb also caused morphological alterations and transformed the normal discoid-shaped erythrocytes into echinocytes and stomatocytes. Thus, mancozeb induces oxidative stress in human erythrocytes, impairs the antioxidant defense system, oxidizes cellular components, that will adversely affect erythrocyte structure and function.
Collapse
Affiliation(s)
- Ruhul Quds
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh 202002, UP, India
| | - Zarmin Iqbal
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh 202002, UP, India
| | - Amin Arif
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh 202002, UP, India
| | - Riaz Mahmood
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh 202002, UP, India.
| |
Collapse
|
10
|
Arif A, Quds R, Salam S, Mahmood R. Esculin protects human blood cells from bioallethrin-induced toxicity: An ex vivo study. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2023; 191:105375. [PMID: 36963944 DOI: 10.1016/j.pestbp.2023.105375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 02/21/2023] [Accepted: 02/24/2023] [Indexed: 06/18/2023]
Abstract
Bioallethrin, a household insecticide, is a member of the pyrethroid family and is known for its adverse effects on human health. Human exposure to pyrethroids is unavoidable due to their widespread use in controlling several fatal vector-borne diseases, mostly in developing nations. Bioallethrin is known to induce oxidative stress in target cells, including erythrocytes. Here we have studied the protective effect of dietary antioxidant esculin on bioallethrin-induced damage in isolated human erythrocytes. The cells were incubated with 200 μM bioallethrin, without or with different concentrations of esculin (200, 400 and 600 μM), and the results compared to the untreated control samples. Bioallethrin-treated erythrocytes showed a significant increase in oxidative stress markers, like protein and lipid oxidation, accompanied by decrease in free amino groups and ratio of reduced to oxidized glutathione. There was enhanced generation of reactive oxygen and nitrogen species with changes in plasma membrane integrity. Bioallethrin oxidized hemoglobin to methemoglobin, which cannot transport oxygen. It altered the activities of antioxidant enzymes and lowered the electron donating and free radical quenching ability of erythrocytes. The cell morphology and redox system of erythrocyte membrane were also altered by bioallethrin. Treatment with esculin, prior to incubation with bioallethrin, led to significant restoration in all these parameters in an esculin concentration-dependent manner. Thus esculin attenuated the biolletherin-induced oxidative damage to erythrocytes. Esculin can, therefore, be an effective chemoprotectant against xenobiotic-induced toxicity in human erythrocytes.
Collapse
Affiliation(s)
- Amin Arif
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh 202002, U.P., India
| | - Ruhul Quds
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh 202002, U.P., India
| | - Samreen Salam
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh 202002, U.P., India
| | - Riaz Mahmood
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh 202002, U.P., India.
| |
Collapse
|
11
|
Tantry IQ, Ali A, Mahmood R. Curcumin from Curcuma longa Linn. (Family: Zingiberaceae) attenuates hypochlorous acid-induced cytotoxicity and oxidative damage to human red blood cells. Toxicol In Vitro 2023; 89:105583. [PMID: 36924976 DOI: 10.1016/j.tiv.2023.105583] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 03/06/2023] [Accepted: 03/13/2023] [Indexed: 03/15/2023]
Abstract
Hypochlorous acid (HOCl) is a major oxidant produced by activated neutrophils via the myeloperoxidase catalyzed reaction. The production of HOCl eliminates a wide range of pathogens. However, HOCl can also cause significant oxidative damage in cells and tissues where it is generated. The protective effect of curcumin was studied on HOCl-induced oxidative damage to human red blood cells (RBC). Isolated RBC were incubated with HOCl at 37 °C in absence or presence of different concentrations of curcumin. Hemolysates were prepared and assayed for various biochemical parameters. Treatment of RBC with HOCl alone increased hemolysis, protein carbonyls, heme degradation and chloramines as compared to untreated control cells. This was accompanied by reduction in glutathione level, total sulfhydryls and free amino groups. HOCl also lowered the activities of major antioxidant enzymes and diminished the antioxidant power of RBC. Pre-treatment of RBC with different concentrations of curcumin resulted in concentration-dependent attenuation in all these parameters while curcumin alone had no significant effect. Scanning electron microscopy showed that curcumin prevented HOCl-induced morphological changes in RBC and restored their normal biconcave shape. Thus curcumin can be used as a chemoprotective agent to mitigate HOCl-induced oxidative damage to cells. These results also explain the beneficial effects of curcumin against Helicobacter pylori induced stomach ulcers, caused by excessive production of HOCl at the site of bacterial infection.
Collapse
Affiliation(s)
- Irfan Qadir Tantry
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, 202002, UP, India; Department of Biochemistry, J.N. Medical College, Aligarh Muslim University, Aligarh, 202002, UP, India
| | - Asif Ali
- Department of Biochemistry, J.N. Medical College, Aligarh Muslim University, Aligarh, 202002, UP, India
| | - Riaz Mahmood
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, 202002, UP, India.
| |
Collapse
|
12
|
Wiatr M, Hadzhieva M, Lecerf M, Noé R, Justesen S, Lacroix-Desmazes S, Dragon-Durey MA, Dimitrov JD. Hyperoxidized Species of Heme Have a Potent Capacity to Induce Autoreactivity of Human IgG Antibodies. Int J Mol Sci 2023; 24:ijms24043416. [PMID: 36834827 PMCID: PMC9960230 DOI: 10.3390/ijms24043416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/02/2023] [Accepted: 02/06/2023] [Indexed: 02/11/2023] Open
Abstract
The interaction of some human antibodies with heme results in posttranslational acquisition of binding to various self- and pathogen-derived antigens. The previous studies on this phenomenon were performed with oxidized heme (Fe3+). In the present study, we elucidated the effect of other pathologically relevant species of heme, i.e., species that were formed after contact of heme with oxidizing agents such as hydrogen peroxide, situations in which heme's iron could acquire higher oxidation states. Our data reveal that hyperoxidized species of heme have a superior capacity to heme (Fe3+) in triggering the autoreactivity of human IgG. Mechanistic studies demonstrated that oxidation status of iron was of critical importance for the heme's effect on antibodies. We also demonstrated that hyperoxidized heme species interacted at higher affinities with IgG and that this binding occurred through a different mechanism as compared to heme (Fe3+). Regardless of their profound functional impact on the antigen-binding properties of antibodies, hyperoxidized species of heme did not affect Fc-mediated functions of IgG, such as binding to the neonatal Fc receptor. The obtained data contribute to a better understanding of the pathophysiological mechanism of hemolytic diseases and of the origin of elevated antibody autoreactivity in patients with some hemolytic disorders.
Collapse
Affiliation(s)
- Marie Wiatr
- Centre de Recherche des Cordeliers, INSERM, CNRS, Sorbonne Université, Université Paris Cité, 75006 Paris, France
| | - Maya Hadzhieva
- Centre de Recherche des Cordeliers, INSERM, CNRS, Sorbonne Université, Université Paris Cité, 75006 Paris, France
| | - Maxime Lecerf
- Centre de Recherche des Cordeliers, INSERM, CNRS, Sorbonne Université, Université Paris Cité, 75006 Paris, France
| | - Rémi Noé
- Centre de Recherche des Cordeliers, INSERM, CNRS, Sorbonne Université, Université Paris Cité, 75006 Paris, France
| | - Sune Justesen
- Immunitrack Aps, Lersoe Park Alle 42, 2100 Copenhagen, Denmark
| | - Sébastien Lacroix-Desmazes
- Centre de Recherche des Cordeliers, INSERM, CNRS, Sorbonne Université, Université Paris Cité, 75006 Paris, France
| | - Marie-Agnès Dragon-Durey
- Centre de Recherche des Cordeliers, INSERM, CNRS, Sorbonne Université, Université Paris Cité, 75006 Paris, France
- Service d’Immunologie Biologique, Hôpital Européen Georges Pompidou, Assistance Publique-Hôpitaux de Paris, 75610 Paris, France
| | - Jordan D. Dimitrov
- Centre de Recherche des Cordeliers, INSERM, CNRS, Sorbonne Université, Université Paris Cité, 75006 Paris, France
- Correspondence: ; Tel.: +33-144-278206
| |
Collapse
|
13
|
Xing X, Zhang C, Ji P, Yang J, Li Q, Pan H, An Q. Effects of Different Iron Supplements on Reproductive Performance and Antioxidant Capacity of Pregnant Sows as Well as Iron Content and Antioxidant Gene Expression in Newborn Piglets. Animals (Basel) 2023; 13:ani13030517. [PMID: 36766406 PMCID: PMC9913290 DOI: 10.3390/ani13030517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 01/26/2023] [Accepted: 01/28/2023] [Indexed: 02/05/2023] Open
Abstract
To improve the reproductive performance of sows and the iron nutrition of newborn piglets, we studied the effects of dietary iron on reproductive performance in pregnant sows as well as antioxidant capacity and the visceral iron content of sows and newborn piglets. Forty pregnant sows were divided into four groups, the iron deficiency group (Id group) was fed a basic diet while sows in the treatment groups were fed diets supplemented with 200 mg/kg lactoferrin (LF group), 0.8% heme-iron (Heme-Fe group), or 500 mg/kg iron-glycine complex (Fe-Gly group). The results indicated that (1) different sources of iron had no significant effect on litter size, live litter size, and litter weight of sows; (2) the three additives improved iron nutrition in newborn piglets, with LF and Heme-Fe having better improvement effects; and (3) the addition of different iron sources improved the level of serum antioxidant biochemical indexes of sows and newborn piglets, and it can have an effect on gene level, among which lactoferrin has the best effect. Thus, adding LF, Heme-iron, or Fe-Gly to the diet of sows during the second and third trimester of gestation can improve the antioxidant capacity of the sows. The supplementation of LF in pregnant sow diets can also improve the antioxidant capacity and the iron nutrition of newborn piglets, with better additive effects than in Heme-Fe and Fe-Gly.
Collapse
|
14
|
Popova SA, Shevchenko OG, Chukicheva IY. Synthesis of new coumarin[1,3]oxazine derivatives of 7-hydroxy-6-isobornyl-4-methylcoumarin and their antioxidant activity. Chem Biol Drug Des 2022; 100:994-1004. [PMID: 34553497 DOI: 10.1111/cbdd.13955] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 08/06/2021] [Accepted: 09/06/2021] [Indexed: 01/25/2023]
Abstract
In this work, we synthesized a series of new 9,10-dihydro-2H,8H-chromeno[8,7e][1,3]oxazine-2-on derivatives which incorporate isobornylcoumarin and 1,3-oxazine moieties. A structure-antioxidant activity relationship was analyzed. A comparative evaluation of their radical scavenging activity, antioxidant and membrane-protective properties was carried out in test with DPPH, as well as on the models of Fe2+ /ascorbate-initiated lipid peroxidation and oxidative hemolysis of mammalian red blood cells. The results suggest that all the obtained coumarin[1,3]oxazine derivatives of 7-hydroxy-6-isobornyl-4-methylcoumarin are capable of exhibiting antioxidant activity in various model systems. Compound 7 with a phenyl fragment, combining high radical scavenging activity and the ability to inhibit Fe2+ /ascorbate-initiated peroxidation of animal lipids in a heterogeneous environment, also proved to be the most effective membrane protector and antioxidant in the model of H2 O2 -induced erythrocyte hemolysis.
Collapse
Affiliation(s)
- Svetlana A Popova
- Institute of Chemistry, Komi Scientific Center, Ural Branch of the Russian Academy of Sciences, Syktyvkar, Russian Federation
| | - Oksana G Shevchenko
- Institute of Biology, Komi Scientific Center, Ural Branch of the Russian Academy of Sciences, Syktyvkar, Russian Federation
| | - Irina Yu Chukicheva
- Institute of Chemistry, Komi Scientific Center, Ural Branch of the Russian Academy of Sciences, Syktyvkar, Russian Federation
| |
Collapse
|
15
|
Iqbal Z, Quds R, Mahmood R. Cadmium chloride generates cytotoxic reactive species that cause oxidative damage and morphological changes in human erythrocytes. Biochem Cell Biol 2022; 100:485-498. [PMID: 36288609 DOI: 10.1139/bcb-2022-0188] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Cadmium chloride (CdCl2) is a widely used industrial compound that exhibits multiple organ toxicity. Cadmium is transported through blood where erythrocytes are exposed to its action. Here the effect of CdCl2 on human erythrocytes was examined under in vitro conditions. Human erythrocytes were treated with 0.01-0.5 mM CdCl2 for 24 h at 37 °C. Lysates were made from CdCl2 treated and untreated (control) cells and used for further analysis. CdCl2 treatment resulted in marked hemolysis of erythrocytes and oxidation of hemoglobin to methemoglobin. This will result in anemia and also reduce the oxygen carrying ability of erythrocytes. Hemoglobin oxidation was accompanied by degradation of heme and release of free ferrous iron moiety. Further analysis showed elevated lipid hydroperoxides and formation of advanced oxidation protein products along with reduction in total sulfhydryl content, indicating the generation of oxidative stress condition in the cell. Incubation of erythrocytes with CdCl2 enhanced generation of reactive oxygen and nitrogen species, decreased the antioxidant power and inhibited pathways of glucose metabolism. Plasma membrane was damaged as indicated by enhanced osmotic fragility and inhibition of membrane bound enzymes. This was confirmed by electron microscopy which showed formation of echinocytes. These results show that CdCl2 generates reactive species which impair the antioxidant system resulting in oxidative damage to erythrocytes.
Collapse
Affiliation(s)
- Zarmin Iqbal
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh 202002, U.P, India
| | - Ruhul Quds
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh 202002, U.P, India
| | - Riaz Mahmood
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh 202002, U.P, India
| |
Collapse
|
16
|
Buravlev EV, Shevchenko OG. Novel Mannich Bases of α‐MangostinBearing Methoxyphenyl Moietieswith Antioxidant and Membrane‐protective activity. ChemistrySelect 2022. [DOI: 10.1002/slct.202202474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Evgeny V. Buravlev
- Laboratory of Organic Synthesis and Chemistry of Natural Compounds Institute of Chemistry Komi Scientific Center Ural Branch of the Russian Academy of Sciences Pervomayskaya St. 48 Syktyvkar 167000 Komi Republic Russian Federation
| | - Oksana G. Shevchenko
- Center of Collective Usage ‘Molecular Biology' Institute of Biology Komi Scientific Center Ural Branch of the Russian Academy of Sciences 28, Kommunisticheskaya St. 167982 Syktyvkar Komi Republic Russian Federation
| |
Collapse
|
17
|
Lamarre Y, Nader E, Connes P, Romana M, Garnier Y. Extracellular Vesicles in Sickle Cell Disease: A Promising Tool. Bioengineering (Basel) 2022; 9:bioengineering9090439. [PMID: 36134985 PMCID: PMC9495982 DOI: 10.3390/bioengineering9090439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 08/25/2022] [Accepted: 08/31/2022] [Indexed: 12/12/2022] Open
Abstract
Sickle cell disease (SCD) is the most common hemoglobinopathy worldwide. It is characterized by an impairment of shear stress-mediated vasodilation, a pro-coagulant, and a pro-adhesive state orchestrated among others by the depletion of the vasodilator nitric oxide, by the increased phosphatidylserine exposure and tissue factor expression, and by the increased interactions of erythrocytes with endothelial cells that mediate the overexpression of adhesion molecules such as VCAM-1, respectively. Extracellular vesicles (EVs) have been shown to be novel actors involved in SCD pathophysiological processes. Medium-sized EVs, also called microparticles, which exhibit increased plasma levels in this pathology, were shown to induce the activation of endothelial cells, thereby increasing neutrophil adhesion, a key process potentially leading to the main complication associated with SCD, vaso-occlusive crises (VOCs). Small-sized EVs, also named exosomes, which have also been reported to be overrepresented in SCD, were shown to potentiate interactions between erythrocytes and platelets, and to trigger endothelial monolayer disruption, two processes also known to favor the occurrence of VOCs. In this review we provide an overview of the current knowledge about EVs concentration and role in SCD.
Collapse
Affiliation(s)
- Yann Lamarre
- Université Paris Cité and Université des Antilles, Inserm, BIGR, F-75015 Paris, France
| | - Elie Nader
- Laboratoire Inter-Universitaire de Biologie de la Motricité EA7424, Team “Vascular Biology and Red Blood Cell”, Université Claude Bernard Lyon 1, Université de Lyon, 69622 Lyon, France
| | - Philippe Connes
- Laboratoire Inter-Universitaire de Biologie de la Motricité EA7424, Team “Vascular Biology and Red Blood Cell”, Université Claude Bernard Lyon 1, Université de Lyon, 69622 Lyon, France
| | - Marc Romana
- Université Paris Cité and Université des Antilles, Inserm, BIGR, F-75015 Paris, France
| | - Yohann Garnier
- Université Paris Cité and Université des Antilles, Inserm, BIGR, F-75015 Paris, France
- Correspondence: ; Tel.: +590-590-891530
| |
Collapse
|
18
|
Palomarez A, Jha M, Medina Romero X, Horton RE. Cardiovascular consequences of sickle cell disease. BIOPHYSICS REVIEWS 2022; 3:031302. [PMID: 38505276 PMCID: PMC10903381 DOI: 10.1063/5.0094650] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 07/11/2022] [Indexed: 03/21/2024]
Abstract
Sickle cell disease (SCD) is an inherited blood disorder caused by a single point mutation within the beta globin gene. As a result of this mutation, hemoglobin polymerizes under low oxygen conditions causing red blood cells to deform, become more adhesive, and increase in rigidity, which affects blood flow dynamics. This process leads to enhanced red blood cell interactions with the endothelium and contributes to vaso-occlusion formation. Although traditionally defined as a red blood cell disorder, individuals with SCD are affected by numerous clinical consequences including stroke, painful crisis episodes, bone infarctions, and several organ-specific complications. Elevated cardiac output, endothelium activation along with the sickling process, and the vaso-occlusion events pose strains on the cardiovascular system. We will present a review of the cardiovascular consequences of sickle cell disease and show connections with the vasculopathy related to SCD. We will also highlight biophysical properties and engineering tools that have been used to characterize the disease. Finally, we will discuss therapies for SCD and potential implications on SCD cardiomyopathy.
Collapse
Affiliation(s)
- Alexis Palomarez
- Department of Biomedical Engineering, Cullen College of Engineering, University of Houston, Houston, Texas 77204, USA
| | - Manisha Jha
- Department of Biomedical Engineering, Cullen College of Engineering, University of Houston, Houston, Texas 77204, USA
| | - Ximena Medina Romero
- Department of Biomedical Engineering, Cullen College of Engineering, University of Houston, Houston, Texas 77204, USA
| | - Renita E. Horton
- Department of Biomedical Engineering, Cullen College of Engineering, University of Houston, Houston, Texas 77204, USA
| |
Collapse
|
19
|
Yang H, Yang Z, Yu Z, Xiong C, Zhang Y, Zhang J, Huang Y, Zhou X, Li J, Xu N. SEMA6D, Negatively Regulated by miR-7, Contributes to C28/I2 chondrocyte's Catabolic and Anabolic Activities via p38 Signaling Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:9674221. [PMID: 35757507 PMCID: PMC9225841 DOI: 10.1155/2022/9674221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 05/16/2022] [Accepted: 05/27/2022] [Indexed: 11/17/2022]
Abstract
MiR-7 has been recognized as an osteoarthritis (OA-)-promoting factor, but the specific downstream pathway of miR-7 still remains unknown. Further investigation of the molecular regulatory mechanism of miR-7 might help develop a novel therapeutic method for OA. In this study, we revealed that Semaphorin 6D (SEMA6D) was a direct target gene of miR-7 and presented a negative regulatory relation with SEMA6D in vitro and in vivo. SEMA6D could improve OA in rat OA models, as indicated by H&E and Safranin O-Fast green staining, and also μCT analysis. Further evaluation of SEMA6D suggested that SEMA6D promotes the anabolism and reduces the catabolism of C28/I2 chondrocytes via inhibiting the activation of the p38 pathway. The present research illustrated that SEMA6D is a negatively regulatory factor of miR-7 and a pivotal mediator of catabolism and anabolism in C28/I2 chondrocytes. SEMA6D exerts its function via inhibiting the activation of the p38 pathway.
Collapse
Affiliation(s)
- Haoyu Yang
- Department of Orthopedics, Wuxi 9th People's Hospital Affiliated to Soochow University, Wuxi 214000, China
| | - Zhicheng Yang
- Department of Orthopedics, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou 213000, China
| | - Zhentang Yu
- Department of Orthopedics, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou 213000, China
| | - Chenwei Xiong
- Department of Orthopedics, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou 213000, China
| | - Yi Zhang
- Department of Orthopedics, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou 213000, China
| | - Junjie Zhang
- Department of Orthopedics, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou 213000, China
| | - Yong Huang
- Department of Orthopedics, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou 213000, China
| | - Xindie Zhou
- Department of Orthopedics, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou 213000, China
| | - Jin Li
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Jiaxing University, Jiaxing 314000, China
| | - Nanwei Xu
- Department of Orthopedics, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou 213000, China
| |
Collapse
|
20
|
Golubev D, Zemskaya N, Shevchenko O, Shaposhnikov M, Kukuman D, Patov S, Punegov V, Moskalev A. Honeysuckle extract (Lonicera pallasii L.) exerts antioxidant properties and extends the lifespan and healthspan of Drosophila melanogaster. Biogerontology 2022; 23:215-235. [PMID: 35122571 DOI: 10.1007/s10522-022-09954-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 01/21/2022] [Indexed: 12/14/2022]
Abstract
Honeysuckle Lonicera pallasii (Lonicera caerulea L.) is an excellent source of anthocyanins which have a number of health-promoting properties mainly associated with antioxidant and anti-inflammatory activities. Cyanidin-3-O-glucoside (C3G) is one of the most common anthocyanins naturally found in honeysuckle. The goal of the present study was to investigate antioxidant and anti-aging properties of Lonicera pallasii (Lonicera caerulea L.) extract (LE) and C3G using red blood cells (RBC) and Drosophila melanogaster models. LE and C3G treatment at a concentration of 100 μM induced enhancement of median and maximum lifespan up to 8%. LE and C3G supplementation at a concentration of 100 μM increased stress resistance up to 10%. The locomotor activity decreased during LE and C3G treatment in 4 and 6 weeks up to 52% in females. The integrity of the intestinal barrier was increased by 4% after LE treatment. These effects were accompanied by increased expression of Hif1 (pro-longevity gene) in response to C3G treatment and decreased expression of Keap1 (anti-longevity gene) after C3G and LE supplementation. RNA interference-mediated knockdown of Sirt6 completely abolished the positive effect obtained of LE and C3G supplementation in males which indicates that lifespan-extending effect is associated with Sirt6 activation. The experiments on the various in-vitro models (including radical scavenging activity and oxidative hemolysis of RBC demonstrated antioxidant and membrane-protective activities of LE and C3G. The present study indicates that Lonicera extract can prolong the lifespan and improve the healthspan of Drosophila model through biological and antioxidant activities.
Collapse
Affiliation(s)
- Denis Golubev
- Institute of Biology of Komi Scientific Centre of the Ural Branch of the RAS, Syktyvkar, Russian Federation, 167982
| | - Nadezhda Zemskaya
- Institute of Biology of Komi Scientific Centre of the Ural Branch of the RAS, Syktyvkar, Russian Federation, 167982
| | - Oksana Shevchenko
- Institute of Biology of Komi Scientific Centre of the Ural Branch of the RAS, Syktyvkar, Russian Federation, 167982
| | - Mikhail Shaposhnikov
- Institute of Biology of Komi Scientific Centre of the Ural Branch of the RAS, Syktyvkar, Russian Federation, 167982
| | - Daria Kukuman
- Institute of Biology of Komi Scientific Centre of the Ural Branch of the RAS, Syktyvkar, Russian Federation, 167982
| | - Sergey Patov
- Institute of Chemistry of Komi Scientific Centre of the Ural Branch of the RAS, Syktyvkar, Russian Federation, 167982
| | - Vasily Punegov
- Institute of Biology of Komi Scientific Centre of the Ural Branch of the RAS, Syktyvkar, Russian Federation, 167982
| | - Alexey Moskalev
- Institute of Biology of Komi Scientific Centre of the Ural Branch of the RAS, Syktyvkar, Russian Federation, 167982.
| |
Collapse
|
21
|
Anjum R, Maheshwari N, Mahmood R. 3,4-Dihydroxybenzaldehyde mitigates fluoride-induced cytotoxicity and oxidative damage in human RBC. J Trace Elem Med Biol 2022; 69:126888. [PMID: 34773916 DOI: 10.1016/j.jtemb.2021.126888] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 10/07/2021] [Accepted: 10/28/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND Fluoride is an essential micronutrient that is needed for mineralization of bones and formation of dental enamel. It is a widely dispersed environmental pollutant and chronic exposure to it is toxic, resulting in malignancies and hematological damage in humans. Blood is a major and early target of environmental pollutants and toxicants like fluoride. Fluoride generates reactive oxygen species and free radicals which induce oxidative stress in target cells and mediate its toxic effects. The aim of this study was to determine the mitigating effect of plant antioxidant 3,4-dihydroxybenzaldehyde (DHB) on sodium fluoride (NaF) induced oxidative damage and cytotoxicity in isolated human red blood cells (RBC) METHOD: Isolated human RBC were treated with 0.5 mM NaF, in absence or presence of different concentrations of DHB (0.1-2.5 mM). Several biochemical parameters were analyzed in cell lysates and whole cells. RESULTS Treatment of RBC with NaF increased the formation of reactive oxygen and nitrogen species. It oxidized thiols, proteins and lipids and generated their peroxidative products. Methemoglobin level, heme degradation and lipid peroxidation were increased but cellular antioxidant status declined significantly in NaF alone treated RBC, compared to the control. NaF inhibited antioxidant, membrane bound and glycolytic enzymes in RBC. However, prior incubation of RBC with DHB significantly attenuated the NaF-induced alterations in all these parameters in a DHB concentration-dependent manner. CONCLUSION These results show that DHB mitigates NaF-induced oxidative damage in human RBC, probably because of its antioxidant character.
Collapse
Affiliation(s)
- Ruhi Anjum
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh 202002, U.P., India.
| | - Nikhil Maheshwari
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh 202002, U.P., India.
| | - Riaz Mahmood
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh 202002, U.P., India.
| |
Collapse
|
22
|
Ahmad S, Tufail N, Parveen N, Mahmood R. Attenuation of Hg(II)-induced cellular and DNA damage in human blood cells by uric acid. Biochem Cell Biol 2021; 100:45-58. [PMID: 34653346 DOI: 10.1139/bcb-2021-0229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Mercury (Hg) is a widespread environmental pollutant and toxicant which induces multiple organ damage in humans and animals. Hg toxicity is mediated by the induction of oxidative stress in target cells. We have used uric acid (UA), a potent antioxidant found in biological fluids, to protect human red blood cells (RBC) and lymphocytes against Hg-mediated cell, organelle and genotoxicity. RBC were incubated with HgCl2, an Hg(II) compound, either alone or in presence of UA. Incubation of RBC with only HgCl2 increased production of nitrogen and oxygen radical species, enhanced methemoglobin levels, heme degradation, free ferrous iron, oxidation of proteins and membrane lipids and reduced antioxidant capacity of cells. UA enhanced the antioxidant capacity of RBC and restored metabolic, plasma membrane-bound and antioxidant enzyme activities. Scanning electron microscopy showed that UA prevented HgCl2-mediated morphological changes in RBC. HgCl2 dissipated the mitochondrial membrane potential and increased lysosomal membrane damage in lymphocytes, but UA pre-treatment attenuated these effects. Genotoxicity analysis by comet assay showed that UA protected lymphocyte DNA from HgCl2-induced damage. Importantly, UA itself did not exhibit any deleterious effects in either RBC or lymphocytes. Thus, UA protects human blood cells from Hg(II)-mediated oxidative damage reducing the harmful effects of this extremely toxic metal. We suggest that UA performs a similar protective role in the plasma against heavy metal toxicity.
Collapse
Affiliation(s)
- Shahbaz Ahmad
- Aligarh Muslim University Faculty of Life Sciences, 154014, Aligarh, Uttar Pradesh, India;
| | - Neda Tufail
- Aligarh Muslim University Faculty of Life Sciences, 154014, Aligarh, Uttar Pradesh, India;
| | - Nazia Parveen
- Aligarh Muslim University Faculty of Life Sciences, 154014, Aligarh, Uttar Pradesh, India;
| | - Riaz Mahmood
- Aligarh Muslim University, Department of Biochemistry, Department of Biochemistry, Faculty of Life Sciences, AMU, Aligarh, Aligarh, Uttar Pradesh, India, 202002;
| |
Collapse
|
23
|
Nader E, Garnier Y, Connes P, Romana M. Extracellular Vesicles in Sickle Cell Disease: Plasma Concentration, Blood Cell Types Origin Distribution and Biological Properties. Front Med (Lausanne) 2021; 8:728693. [PMID: 34490315 PMCID: PMC8417591 DOI: 10.3389/fmed.2021.728693] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 07/30/2021] [Indexed: 01/08/2023] Open
Abstract
Prototype of monogenic disorder, sickle cell disease (SCD) is caused by a unique single mutation in the β-globin gene, leading to the production of the abnormal hemoglobin S (HbS). HbS polymerization in deoxygenated condition induces the sickling of red blood cells (RBCs), which become less deformable and more fragile, and thus prone to lysis. In addition to anemia, SCD patients may exhibit a plethora of clinical manifestations ranging from acute complications such as the frequent and debilitating painful vaso-occlusive crisis to chronic end organ damages. Several interrelated pathophysiological processes have been described, including impaired blood rheology, increased blood cell adhesion, coagulation, inflammation and enhanced oxidative stress among others. During the last two decades, it has been shown that extracellular vesicles (EVs), defined as cell-derived anucleated particles delimited by a lipid bilayer, and comprising small EVs (sEVs) and medium/large EVs (m/lEVs); are not only biomarkers but also subcellular actors in SCD pathophysiology. Plasma concentration of m/lEVs, originated mainly from RBCs and platelets (PLTs) but also from the other blood cell types, is higher in SCD patients than in healthy controls. The concentration and the density of externalized phosphatidylserine of those released from RBCs may vary according to clinical status (crisis vs. steady state) and treatment (hydroxyurea). Besides their procoagulant properties initially described, RBC-m/lEVs may promote inflammation through their effects on monocytes/macrophages and endothelial cells. Although less intensely studied, sEVs plasma concentration is increased in SCD and these EVs may cause endothelial damages. In addition, sEVs released from activated PLTs trigger PLT-neutrophil aggregation involved in lung vaso-occlusion in sickle mice. Altogether, these data clearly indicate that EVs are both biomarkers and bio-effectors in SCD, which deserve further studies.
Collapse
Affiliation(s)
- Elie Nader
- Laboratoire Inter-Universitaire de Biologie de la Motricité EA7424, Team "Vascular Biology and Red Blood Cell", Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France.,Laboratoire d'Excellence du Globule Rouge, PRES Sorbonne, Paris, France
| | - Yohann Garnier
- Laboratoire d'Excellence du Globule Rouge, PRES Sorbonne, Paris, France.,Université des Antilles, UMR_S1134, BIGR, Pointe-à-Pitre, France.,Université de Paris, UMR_S1134, BIGR, INSERM, Paris, France
| | - Philippe Connes
- Laboratoire Inter-Universitaire de Biologie de la Motricité EA7424, Team "Vascular Biology and Red Blood Cell", Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France.,Laboratoire d'Excellence du Globule Rouge, PRES Sorbonne, Paris, France
| | - Marc Romana
- Laboratoire d'Excellence du Globule Rouge, PRES Sorbonne, Paris, France.,Université des Antilles, UMR_S1134, BIGR, Pointe-à-Pitre, France.,Université de Paris, UMR_S1134, BIGR, INSERM, Paris, France
| |
Collapse
|
24
|
Buravlev EV, Shevchenko OG, Kutchin AV. Synthesis and comparative evaluation of the antioxidant activity for some new derivatives of 2,6-diisobornylphenol bearing an aminomethyl group at the position 4. Russ Chem Bull 2021. [DOI: 10.1007/s11172-021-3075-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
25
|
Aydemir D, Ulusu NN. People with blood disorders can be more vulnerable during COVID-19 pandemic: A hypothesis paper. Transfus Apher Sci 2021; 60:103080. [PMID: 33608217 PMCID: PMC7874911 DOI: 10.1016/j.transci.2021.103080] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 11/12/2020] [Accepted: 02/06/2021] [Indexed: 11/29/2022]
Abstract
The world has been encountered with COVID-19 pandemic since at the beginning of 2020 and the number of infected people by COVID-19 is increasing every day. Despite various studies conducted by researchers and doctors, no treatment has been developed until now, therefore self-protection and isolation are strongly recommended to stop the spread of the virus. The elderly population and people with chronic diseases such as hypertension, cardiovascular diseases, diabetes, and cancer are categorized as risk groups, however, we suggest that people with hemoglobinopathies or porphyria can be described as risk groups as well. Current in silico studies have revealed that the COVID-19 virus can attack heme and hemoglobin metabolisms which are responsible for the oxygen transport to the tissues, iron metabolism, elevated levels of oxidative stress, and tissue damage. Data of the in silico study have been supported with the biochemistry and hemogram results of the COVID-19 patients, for instance hemoglobin levels decreased and serum ferritin and C-reactive protein levels increased. Indicated biochemistry biomarkers are tightly associated with inflammation, iron overload, and oxidative stress. In conclusion, since people with hemoglobinopathies or porphyria have already impaired heme and hemoglobin metabolism, COVID-19 infection can enhance the adverse effects of impaired hemoglobin metabolism and accelerate the progression of severe symptoms in patients with hemoglobinopathies or porphyria compared to the normal individuals. Thus those people can be considered as a risk group and extra precautions should be applied for them to protect them.
Collapse
Affiliation(s)
- Duygu Aydemir
- Koc University, School of Medicine, Rumelifeneri Yolu, Sariyer, 34450, Istanbul, Turkey; Koc University Research Center for Translational Medicine (KUTTAM), Sariyer, 34450, Istanbul, Turkey
| | - Nuriye Nuray Ulusu
- Koc University, School of Medicine, Rumelifeneri Yolu, Sariyer, 34450, Istanbul, Turkey; Koc University Research Center for Translational Medicine (KUTTAM), Sariyer, 34450, Istanbul, Turkey.
| |
Collapse
|
26
|
Vona R, Sposi NM, Mattia L, Gambardella L, Straface E, Pietraforte D. Sickle Cell Disease: Role of Oxidative Stress and Antioxidant Therapy. Antioxidants (Basel) 2021; 10:antiox10020296. [PMID: 33669171 PMCID: PMC7919654 DOI: 10.3390/antiox10020296] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 02/08/2021] [Accepted: 02/12/2021] [Indexed: 12/11/2022] Open
Abstract
Sickle cell disease (SCD) is the most common hereditary disorder of hemoglobin (Hb), which affects approximately a million people worldwide. It is characterized by a single nucleotide substitution in the β-globin gene, leading to the production of abnormal sickle hemoglobin (HbS) with multi-system consequences. HbS polymerization is the primary event in SCD. Repeated polymerization and depolymerization of Hb causes oxidative stress that plays a key role in the pathophysiology of hemolysis, vessel occlusion and the following organ damage in sickle cell patients. For this reason, reactive oxidizing species and the (end)-products of their oxidative reactions have been proposed as markers of both tissue pro-oxidant status and disease severity. Although more studies are needed to clarify their role, antioxidant agents have been shown to be effective in reducing pathological consequences of the disease by preventing oxidative damage in SCD, i.e., by decreasing the oxidant formation or repairing the induced damage. An improved understanding of oxidative stress will lead to targeted antioxidant therapies that should prevent or delay the development of organ complications in this patient population.
Collapse
Affiliation(s)
- Rosa Vona
- Biomarkers Unit, Center for Gender-Specific Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (R.V.); (N.M.S.); (L.G.)
| | - Nadia Maria Sposi
- Biomarkers Unit, Center for Gender-Specific Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (R.V.); (N.M.S.); (L.G.)
| | - Lorenza Mattia
- Department of Clinical and Molecular Medicine, “La Sapienza” University, 00161 Rome, Italy;
- Endocrine-Metabolic Unit, Sant’Andrea University Hospital, 00189 Rome, Italy
| | - Lucrezia Gambardella
- Biomarkers Unit, Center for Gender-Specific Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (R.V.); (N.M.S.); (L.G.)
| | - Elisabetta Straface
- Biomarkers Unit, Center for Gender-Specific Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (R.V.); (N.M.S.); (L.G.)
- Correspondence: ; Tel.: +39-064-990-2443; Fax: +39-064-990-3690
| | - Donatella Pietraforte
- Core Facilities, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy;
| |
Collapse
|
27
|
Gbotosho OT, Kapetanaki MG, Kato GJ. The Worst Things in Life are Free: The Role of Free Heme in Sickle Cell Disease. Front Immunol 2021; 11:561917. [PMID: 33584641 PMCID: PMC7873693 DOI: 10.3389/fimmu.2020.561917] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 12/04/2020] [Indexed: 12/15/2022] Open
Abstract
Hemolysis is a pathological feature of several diseases of diverse etiology such as hereditary anemias, malaria, and sepsis. A major complication of hemolysis involves the release of large quantities of hemoglobin into the blood circulation and the subsequent generation of harmful metabolites like labile heme. Protective mechanisms like haptoglobin-hemoglobin and hemopexin-heme binding, and heme oxygenase-1 enzymatic degradation of heme limit the toxicity of the hemolysis-related molecules. The capacity of these protective systems is exceeded in hemolytic diseases, resulting in high residual levels of hemolysis products in the circulation, which pose a great oxidative and proinflammatory risk. Sickle cell disease (SCD) features a prominent hemolytic anemia which impacts the phenotypic variability and disease severity. Not only is circulating heme a potent oxidative molecule, but it can act as an erythrocytic danger-associated molecular pattern (eDAMP) molecule which contributes to a proinflammatory state, promoting sickle complications such as vaso-occlusion and acute lung injury. Exposure to extracellular heme in SCD can also augment the expression of placental growth factor (PlGF) and interleukin-6 (IL-6), with important consequences to enthothelin-1 (ET-1) secretion and pulmonary hypertension, and potentially the development of renal and cardiac dysfunction. This review focuses on heme-induced mechanisms that are implicated in disease pathways, mainly in SCD. A special emphasis is given to heme-induced PlGF and IL-6 related mechanisms and their role in SCD disease progression.
Collapse
Affiliation(s)
- Oluwabukola T. Gbotosho
- Division of Hematology-Oncology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Maria G. Kapetanaki
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Gregory J. Kato
- Division of Hematology-Oncology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| |
Collapse
|
28
|
Maheshwari N, Qasim N, Anjum R, Mahmood R. Fluoride enhances generation of reactive oxygen and nitrogen species, oxidizes hemoglobin, lowers antioxidant power and inhibits transmembrane electron transport in isolated human red blood cells. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 208:111611. [PMID: 33396131 DOI: 10.1016/j.ecoenv.2020.111611] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 10/29/2020] [Accepted: 11/02/2020] [Indexed: 06/12/2023]
Abstract
Fluoride is a widespread environmental pollutant that at high levels exerts numerous deleterious effects on human health. The toxic effects of fluoride are a matter of serious concern since many countries have regions of endemic fluorosis. The main source of fluoride exposure for humans is intake of contaminated groundwater. Fluoride is absorbed from the gastrointestinal tract and enters the circulating blood, where the abundant red blood cells (RBC) are an early and major target of fluoride toxicity. Chronic fluoride exposure generates free radicals, reactive species which leads to redox imbalance, cytotoxicity and hematological damage. This study aimed to determine the effect of sodium fluoride (NaF) on human RBC under in vitro conditions. Isolated RBC were incubated with different concentrations of NaF (10-500 µM) for 8 h at 37 °C. Several biochemical parameters were determined in hemolysates or whole cells. Treatment of RBC with NaF enhanced the generation of reactive oxygen and nitrogen species. This increased the oxidation of hemoglobin to yield methemoglobin and oxoferrylhemoglobin, which are inactive in oxygen transport. NaF treatment increased the degradation of heme causing release of free iron from its porphyrin ring. Cellular antioxidant power was significantly decreased in NaF-treated RBC, lowering the metal reducing and free radical quenching ability of cells. The two pathways of glucose metabolism in RBC i.e. glycolysis and hexose monophosphate shunt, were inhibited. NaF also inhibited the plasma membrane redox system, and its associated ascorbate free radical reductase, to disrupt transmembrane electron transport. These results suggest that fluoride generates reactive species that cause extensive oxidative modifications in human RBC.
Collapse
Affiliation(s)
- Nikhil Maheshwari
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh 202002, UP, India
| | - Neha Qasim
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh 202002, UP, India
| | - Ruhi Anjum
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh 202002, UP, India
| | - Riaz Mahmood
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh 202002, UP, India.
| |
Collapse
|
29
|
Haines DD, Tosaki A. Heme Degradation in Pathophysiology of and Countermeasures to Inflammation-Associated Disease. Int J Mol Sci 2020; 21:ijms21249698. [PMID: 33353225 PMCID: PMC7766613 DOI: 10.3390/ijms21249698] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 12/15/2020] [Accepted: 12/16/2020] [Indexed: 12/12/2022] Open
Abstract
The class of tetrapyrrol "coordination complexes" called hemes are prosthetic group components of metalloproteins including hemoglobin, which provide functionality to these physiologically essential macromolecules by reversibly binding diatomic gasses, notably O2, which complexes to ferrous (reduced/Fe(II)) iron within the heme porphyrin ring of hemoglobin in a pH- and PCO2-dependent manner-thus allowing their transport and delivery to anatomic sites of their function. Here, pathologies associated with aberrant heme degradation are explored in the context of their underlying mechanisms and emerging medical countermeasures developed using heme oxygenase (HO), its major degradative enzyme and bioactive metabolites produced by HO activity. Tissue deposits of heme accumulate as a result of the removal of senescent or damaged erythrocytes from circulation by splenic macrophages, which destroy the cells and internal proteins, including hemoglobin, leaving free heme to accumulate, posing a significant toxicogenic challenge. In humans, HO uses NADPH as a reducing agent, along with molecular oxygen, to degrade heme into carbon monoxide (CO), free ferrous iron (FeII), which is sequestered by ferritin protein, and biliverdin, subsequently metabolized to bilirubin, a potent inhibitor of oxidative stress-mediated tissue damage. CO acts as a cellular messenger and augments vasodilation. Nevertheless, disease- or trauma-associated oxidative stressors sufficiently intense to overwhelm HO may trigger or exacerbate a wide range of diseases, including cardiovascular and neurologic syndromes. Here, strategies are described for counteracting the effects of aberrant heme degradation, with a particular focus on "bioflavonoids" as HO inducers, shown to cause amelioration of severe inflammatory diseases.
Collapse
Affiliation(s)
- Donald David Haines
- Advanced Biotherapeutics, London W2 1EB, UK;
- Department of Pharmacology, Faculty of Pharmacy, University of Debrecen, 4032 Debrecen, Hungary
| | - Arpad Tosaki
- Department of Pharmacology, Faculty of Pharmacy, University of Debrecen, 4032 Debrecen, Hungary
- Correspondence: ; Tel./Fax: +36-52-255586
| |
Collapse
|
30
|
Ito H, Kurokawa H, Matsui H. Mitochondrial reactive oxygen species and heme, non-heme iron metabolism. Arch Biochem Biophys 2020; 700:108695. [PMID: 33232715 DOI: 10.1016/j.abb.2020.108695] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 11/17/2020] [Accepted: 11/18/2020] [Indexed: 12/13/2022]
Abstract
Mitochondria are one of the most important organelles for eukaryotes, including humans, to produce energy. In the energy-producing process, mitochondria constantly generate reactive oxygen species as a by-product of electrons leaking out from the electron transport chain react with oxygen. The active oxygen, in turn, plays pivotal roles in mediating several signalings, including those that are implicated in the development of some diseases such as neurodegenerative disease, cardiovascular disease, and carcinogenesis. This signaling, derived from mitochondrial reactive oxygen species, also affects intracellular iron homeostasis by regulating the expression of transporters. Heme iron is incorporated into cells through HCP1, and non-heme iron is transported by DMT1 in absorptive cells. Intracellular iron is exported by ferroportin and bound with transferrin. In most types of cell including erythrocyte, transferrin-bound iron is incorporated through transferrin-transferrin receptor system. We previously reported that the expression of HCP1 and DMT1 was upregulated in cancer cells and that overexpression of manganese superoxide dismutase, which is a mitochondrial-specific superoxide dismutase, downregulated the expression. These findings indicate that mitochondrial reactive oxygen species is associated with iron-related oxidative reactions. Recently, a mitochondria-specific iron transporter, mitoferrin, was identified, and the relationships among mitochondria, iron transportation, and diseases have been increasingly clarified.
Collapse
Affiliation(s)
- Hiromu Ito
- Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan; Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, 890-8544, Japan
| | - Hiromi Kurokawa
- Algae Biomass research and development, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Hirofumi Matsui
- Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan; Algae Biomass research and development, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan.
| |
Collapse
|
31
|
Synthesis and antioxidant properties of N-substituted aminomethyl derivatives of 2-isobornylphenol. Russ Chem Bull 2020. [DOI: 10.1007/s11172-020-2987-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
32
|
Buravlev EV, Fedorova IV, Shevchenko OG, Kutchin AV. Comparative evaluation of the antioxidant activity of some ortho-substituted mono- and dialkylphenols with the para-positioned hydroxymethyl group. Russ Chem Bull 2020. [DOI: 10.1007/s11172-020-2937-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
33
|
Maheshwari N, Mahmood R. 3,4-Dihydroxybenzaldehyde attenuates pentachlorophenol-induced cytotoxicity, DNA damage and collapse of mitochondrial membrane potential in isolated human blood cells. Drug Chem Toxicol 2020; 45:1225-1242. [DOI: 10.1080/01480545.2020.1811722] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Nikhil Maheshwari
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, India
| | - Riaz Mahmood
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, India
| |
Collapse
|
34
|
Rey-Barroso L, Roldán M, Burgos-Fernández FJ, Gassiot S, Ruiz Llobet A, Isola I, Vilaseca M. Spectroscopic Evaluation of Red Blood Cells of Thalassemia Patients with Confocal Microscopy: A Pilot Study. SENSORS 2020; 20:s20144039. [PMID: 32708084 PMCID: PMC7412432 DOI: 10.3390/s20144039] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 07/07/2020] [Accepted: 07/15/2020] [Indexed: 11/16/2022]
Abstract
Hemoglobinopathies represent the most common single-gene defects in the world and pose a major public health problem, particularly in tropical countries, where they occur with high frequency. Diagnosing hemoglobinopathies can sometimes be difficult due to the coexistence of different causes of anemia, such as thalassemia and iron deficiency, and blood transfusions, among other factors, and requires expensive and complex molecular tests. This work explores the possibility of using spectral confocal microscopy as a diagnostic tool for thalassemia in pediatric patients, a disease caused by mutations in the globin genes that result in changes of the globin chains that form hemoglobin-in pediatric patients. Red blood cells (RBCs) from patients with different syndromes of alpha-thalassemia and iron deficiency (including anemia) as well as healthy (control) subjects were analyzed under a Leica TCS SP8 confocal microscope following different image acquisition protocols. We found that diseased RBCs exhibited autofluorescence when excited at 405 nm and their emission was collected in the spectral range from 425 nm to 790 nm. Three experimental descriptors calculated from the mean emission intensities at 502 nm, 579 nm, 628 nm, and 649 nm allowed us to discriminate between diseased and healthy cells. According to the results obtained, spectral confocal microscopy could serve as a tool in the diagnosis of thalassemia.
Collapse
Affiliation(s)
- Laura Rey-Barroso
- Centre for Sensors, Instruments and Systems Development, Technical University of Catalonia, 08222 Terrassa, Spain; (F.J.B.-F.); (M.V.)
- Correspondence: ; Tel.: +34-97-739-8905
| | - Mónica Roldán
- Unit of Confocal Microscopy, Service of Pathological Anatomy, Pediatric Institute of Rare Diseases, Hospital Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain;
- Institute of Pediatric Research, Hospital Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain; (S.G.); (I.I.)
| | - Francisco J. Burgos-Fernández
- Centre for Sensors, Instruments and Systems Development, Technical University of Catalonia, 08222 Terrassa, Spain; (F.J.B.-F.); (M.V.)
| | - Susanna Gassiot
- Institute of Pediatric Research, Hospital Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain; (S.G.); (I.I.)
- Laboratory of Hematology, Service of Laboratory Diagnosis, Hospital Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain
| | - Anna Ruiz Llobet
- Service of Pediatric Hematology, Hospital Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain;
| | - Ignacio Isola
- Institute of Pediatric Research, Hospital Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain; (S.G.); (I.I.)
- Laboratory of Hematology, Service of Laboratory Diagnosis, Hospital Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain
| | - Meritxell Vilaseca
- Centre for Sensors, Instruments and Systems Development, Technical University of Catalonia, 08222 Terrassa, Spain; (F.J.B.-F.); (M.V.)
| |
Collapse
|
35
|
Maheshwari N, Mahmood R. Protective effect of catechin on pentachlorophenol-induced cytotoxicity and genotoxicity in isolated human blood cells. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2020; 27:13826-13843. [PMID: 32036526 DOI: 10.1007/s11356-020-07969-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 02/02/2020] [Indexed: 06/10/2023]
Abstract
Pentachlorophenol (PCP) is an organochlorine compound that is used as pesticide, biocide, and wood preservative. PCP is highly toxic and carcinogenic. It has been detected in food and several consumable products. The toxicity of PCP is thought to be due to generation of oxidative stress in cells. We examined whether the dietary antioxidant catechin can attenuate or protect human erythrocytes and lymphocytes against PCP-induced cytotoxicity and genotoxicity, respectively. Human erythrocytes were treated with increasing concentrations of catechin (0.05-2.5 mM) for 30 min followed by addition of 0.75 mM PCP and further incubation for 4 h at 37 °C. Hemolysates were prepared and assayed for various biochemical parameters. Treatment with PCP alone increased the generation of reactive oxygen and nitrogen species, lipid and protein oxidation, and damaged the plasma membrane, when compared to PCP untreated (control) cells. It significantly decreased glutathione level, total sulfhydryl content, and cellular antioxidant power. PCP treatment lowered the activity of antioxidant enzymes and inhibited enzymes of glucose metabolism. However, prior incubation with catechin attenuated the PCP-induced changes in all these parameters in a catechin concentration-dependent manner. Scanning electron microscopy of erythrocytes confirmed these biochemical results. PCP treatment converted the normal discoidal erythrocytes to irregularly contracted cells, acanthocytes, and echinocytes but the presence of catechin inhibited these morphological changes and erythrocytes retained their biconcave shape to a large extent. Genotoxicity was studied in human lymphocytes by single-cell gel electrophoresis (comet assay). It showed strand breaks and longer comet tail length in PCP alone treated cells. The comet tail length was reduced in the catechin +PCP-treated lymphocytes showing that catechin protected cells from PCP-induced DNA damage. These results show that catechin protects human blood cells against PCP-induced oxidative damage.
Collapse
Affiliation(s)
- Nikhil Maheshwari
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, U. P., 202002, India
| | - Riaz Mahmood
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, U. P., 202002, India.
| |
Collapse
|
36
|
Salam S, Arif A, Mahmood R. Thiram-induced cytotoxicity and oxidative stress in human erythrocytes: an in vitro study. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2020; 164:14-25. [PMID: 32284119 DOI: 10.1016/j.pestbp.2019.12.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 12/05/2019] [Accepted: 12/15/2019] [Indexed: 06/11/2023]
Abstract
Tetramethylthiuram disulfide, commonly known as thiram, is an organosulfur compound which is used as a bactericide, fungicide and ectoparasiticide to prevent disease in seeds and crops. Being a fungicide there is a high probability of human occupational exposure to thiram and also via consumption of contaminated food. In this work, the cytotoxicity of thiram was studied under in vitro conditions using human erythrocytes as the cellular model. Erythrocytes were incubated with different concentrations of thiram (25-500 μM) for 4 h at 37 °C. Control cells (thiram untreated) were similarly incubated at 37 °C. Whole cells and hemolysates were analyzed for various biochemical parameters. Treatment of erythrocytes with thiram increased protein and lipid oxidation and hydrogen peroxide level in hemolysates but decreased glutathione and total sulfhydryl group content. This was accompanied by hemoglobin oxidation, heme degradation and release of free iron. Activities of all major antioxidant enzymes were inhibited. The antioxidant power of thiram treated erythrocytes was lowered resulting in decreased metal reducing and free radical quenching ability. These results suggest that thiram enhances the generation of reactive species that cause oxidative modification of cell components. This was confirmed by experiments that showed enhanced generation of reactive oxygen and nitrogen species in thiram treated erythrocytes. Activities of marker enzymes of glucose metabolism and erythrocyte membrane were also inhibited. All effects were seen in a thiram concentration-dependent manner. Electron microscopy further supported the damaging effect of thiram on erythrocytes. Thus thiram induces oxidative stress condition in human erythrocytes and causes oxidative modification of cell components.
Collapse
Affiliation(s)
- Samreen Salam
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh 202002, U.P., India
| | - Amin Arif
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh 202002, U.P., India
| | - Riaz Mahmood
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh 202002, U.P., India.
| |
Collapse
|
37
|
Arif A, Salam S, Mahmood R. Bioallethrin-induced generation of reactive species and oxidative damage in isolated human erythrocytes. Toxicol In Vitro 2020; 65:104810. [PMID: 32097678 DOI: 10.1016/j.tiv.2020.104810] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 02/18/2020] [Accepted: 02/21/2020] [Indexed: 12/12/2022]
Abstract
Bioallethrin is an insecticide that is widely used to control mosquitoes, fleas and cockroaches. The widespread use of bioallethrin has resulted in both occupational and non-occupational human exposure. Bioallethrin enters blood, regardless of the route of exposure, where it can interact with erythrocytes. We have studied the effect of bioallethrin on isolated human erythrocytes under in vitro conditions. Erythrocytes were incubated with increasing concentrations of bioallethrin (10-200 μM) for 4 h at 37 °C. Several biochemical parameters were analyzed in bioallethrin treated and untreated (control) cells. Incubation of erythrocytes with bioallethrin increased protein oxidation, lipid peroxidation and depleted sulfhydryl group content. Membrane damage was evident from cell lysis, osmotic fragility, inhibition of bound enzymes and transmembrane electron transport system. Bioallethrin also increased hemoglobin oxidation, heme degradation and the release of free iron moiety. This will decrease the oxygen transporting ability of blood. Bioallethrin treatment altered the specific activities of antioxidant enzymes and diminished the antioxidant power of cells. Scanning electron microscopy showed that bioallethrin treatment also altered erythrocyte mophology. Almost all changes were in a bioallethrin concentration dependent manner. The cytotoxicity of bioallethrin is probably mediated by reactive oxygen and nitrogen species whose formation was significantly enhanced in treated erythrocytes. Thus bioallethrin enhances the generation of reactive species which cause oxidative damage of cell components in human erythrocytes.
Collapse
Affiliation(s)
- Amin Arif
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh 202002, U.P, India
| | - Samreen Salam
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh 202002, U.P, India
| | - Riaz Mahmood
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh 202002, U.P, India.
| |
Collapse
|
38
|
Buravlev EV, Dvornikova IA, Schevchenko OG, Kutchin AV. Synthesis and Antioxidant Ability of Novel Derivatives Based on
para
‐Coumaric Acid Containing Isobornyl Groups. Chem Biodivers 2019; 16:e1900362. [DOI: 10.1002/cbdv.201900362] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Accepted: 08/10/2019] [Indexed: 01/13/2023]
Affiliation(s)
- Evgeny V. Buravlev
- Institute of Chemistry, Komi Scientific CenterUral Branch of the Russian Academy of Sciences, 48 Pervomayskaya St. 167000 Syktyvkar, Komi Republic Russian Federation
| | - Irina A. Dvornikova
- Institute of Chemistry, Komi Scientific CenterUral Branch of the Russian Academy of Sciences, 48 Pervomayskaya St. 167000 Syktyvkar, Komi Republic Russian Federation
| | - Oksana G. Schevchenko
- Institute of Biology, Komi Scientific CenterUral Branch of the Russian Academy of Sciences, 28 Kommunisticheskaya St. 167982 Syktyvkar, Komi Republic Russian Federation
| | - Aleksandr V. Kutchin
- Institute of Chemistry, Komi Scientific CenterUral Branch of the Russian Academy of Sciences, 48 Pervomayskaya St. 167000 Syktyvkar, Komi Republic Russian Federation
| |
Collapse
|
39
|
Detterich JA, Liu H, Suriany S, Kato RM, Chalacheva P, Tedla B, Shah PM, Khoo MC, Wood JC, Coates TD, Milne GL, Oh JY, Patel RP, Forman HJ. Erythrocyte and plasma oxidative stress appears to be compensated in patients with sickle cell disease during a period of relative health, despite the presence of known oxidative agents. Free Radic Biol Med 2019; 141:408-415. [PMID: 31279092 PMCID: PMC6750280 DOI: 10.1016/j.freeradbiomed.2019.07.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 06/03/2019] [Accepted: 07/03/2019] [Indexed: 01/17/2023]
Abstract
Sickle cell disease (SCD) is a monogenetic disease that results in the formation of hemoglobin S. Due to more rapid oxidation of hemoglobin S due to intracellular heme and adventitious iron in SCD, it has been thought that an inherent property of SCD red cells would be an imbalance in antioxidant defenses and oxidant production. Less deformable and fragile RBC in SCD results in intravascular hemolysis and release of free hemoglobin (PFHb) in the plasma, which might be expected to produce oxidative stress in the plasma. Thus, we aimed to characterize intracellular and vascular oxidative stress in whole blood and plasma samples from adult SCD patients and controls recruited into a large study of SCD at Children's Hospital of Los Angeles. We evaluated the cellular content of metHb and several components of the antioxidant system in RBC as well as oxidation of GSH and Prx-2 oxidation in RBC after challenge with hydroperoxides. Plasma markers included PFHb, low molecular weight protein bound heme (freed heme), hemopexin, isoprostanes, and protein carbonyls. While GSH was slightly lower in SCD RBC, protein carbonyls, NADH, NAD+ and total NADP+ + NADPH were not different. Furthermore, GSH or Prx-2 oxidation was not different after oxidative challenge in SCD vs. Control. Elevated freed heme and PFHb had a significant negative, non-linear association with hemopexin. There appeared to be a threshold effect for hemopexin (200 μg/ml), under which the freed heme rose acutely. Plasma F2-isoprostanes were not significantly elevated in SCD. Despite significant release of Hb and elevation of freed heme in SCD when hemopexin was apparently saturated, there was no clear indication of uncompensated vascular oxidative stress. This somewhat surprising result, suggests that oxidative stress is well compensated in RBCs and plasma during a period of relative health.
Collapse
Affiliation(s)
- Jon A Detterich
- Division of Cardiology, Children's Hospital of Los Angeles, USA.
| | - Honglei Liu
- Division of Cardiology, Children's Hospital of Los Angeles, USA
| | - Silvie Suriany
- Division of Cardiology, Children's Hospital of Los Angeles, USA; Division of Hematology, Children's Hospital of Los Angeles, USA
| | | | | | - Bruke Tedla
- Division of Cardiology, Children's Hospital of Los Angeles, USA
| | - Payal M Shah
- Division of Hematology, Children's Hospital of Los Angeles, USA
| | - Michael C Khoo
- Viterbi School of Engineering, University of Southern California, USA
| | - John C Wood
- Division of Cardiology, Children's Hospital of Los Angeles, USA; Viterbi School of Engineering, University of Southern California, USA
| | - Thomas D Coates
- Division of Hematology, Children's Hospital of Los Angeles, USA
| | - Ginger L Milne
- Division of Clinical Pharmacology, Vanderbilt University, USA
| | - Joo-Yeun Oh
- Department of Pathology and Center for Free Radical Biology, University of Alabama School of Medicine, USA
| | - Rakesh P Patel
- Department of Pathology and Center for Free Radical Biology, University of Alabama School of Medicine, USA
| | - Henry Jay Forman
- Leonard Davis School of Gerontology, University of Southern California, USA
| |
Collapse
|
40
|
Synthesis and antioxidant properties of benzimidazole derivatives with isobornylphenol fragments. Russ Chem Bull 2019. [DOI: 10.1007/s11172-019-2510-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
41
|
Buravlev EV, Fedorova IV, Shevchenko OG. Comparative evaluation of antioxidant activity of 2-alkyl-4-methylphenols and their 6-n-octylaminomethyl derivatives. Russ Chem Bull 2019. [DOI: 10.1007/s11172-019-2508-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
42
|
Maheshwari N, Khan FH, Mahmood R. Pentachlorophenol-induced cytotoxicity in human erythrocytes: enhanced generation of ROS and RNS, lowered antioxidant power, inhibition of glucose metabolism, and morphological changes. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2019; 26:12985-13001. [PMID: 30895543 DOI: 10.1007/s11356-019-04736-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 02/27/2019] [Indexed: 06/09/2023]
Abstract
Pentachlorophenol (PCP) is a class 2B human carcinogen that is used as an insecticide, herbicide, and wood preservative. PCP is rapidly absorbed and enters the blood where it can interact with erythrocytes. We have examined the effect of PCP on human erythrocytes. Treatment of erythrocytes with PCP increased the intracellular generation of reactive oxygen and nitrogen species. It also increased lipid and protein oxidation accompanied by decrease in glutathione levels and total sulfhydryl content. The activities of all major antioxidant enzymes were altered. The antioxidant power was significantly impaired resulting in lower free radical quenching and metal reducing ability of the PCP-treated cells. PCP exposure also inhibited the activities of enzymes of glycolysis and pentose phosphate shunt, the two pathways of glucose metabolism in erythrocytes. Heme degradation was enhanced leading to the release of free iron. Incubation of erythrocytes with PCP caused significant cell lysis suggesting plasma membrane damage which was also evident from inhibition of bound enzymes. Scanning electron microscopy of erythrocytes confirmed these biochemical results and showed that PCP treatment converted the normal biconcave discoids to echinocytes and other irregularly shaped cells. Thus, PCP induces oxidative and nitrosative stress in erythrocytes, alters the enzymatic and nonenzymatic antioxidant defense systems, inhibits glucose metabolism, and causes significant modifications in cellular morphology.
Collapse
Affiliation(s)
- Nikhil Maheshwari
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, Uttar Pradesh, 202002, India
| | - Fahim Halim Khan
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, Uttar Pradesh, 202002, India
| | - Riaz Mahmood
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, Uttar Pradesh, 202002, India.
| |
Collapse
|
43
|
2-Hydroxy-3-isobornyl-5-methylbenzaldehyde derivatives: synthesis and antioxidant activity in vitro. Russ Chem Bull 2019. [DOI: 10.1007/s11172-019-2419-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
44
|
Ajibola KA, Adedokun KA, Oduola T, Oparinde DP, Ayelagbe OG, Ojokuku HO. Assessment of iron status and interplay between lipid peroxidation and antioxidant capacity in common hemoglobin variants in Osun State, southwestern Nigeria. Kaohsiung J Med Sci 2019; 35:358-364. [DOI: 10.1002/kjm2.12062] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 03/07/2019] [Indexed: 01/09/2023] Open
Affiliation(s)
- Kabiru A. Ajibola
- Department of Chemical PathologyLadoke Akintola University of Technology Osogbo Osun State Nigeria
| | - Kamoru A. Adedokun
- Department of Chemical PathologyCollege of Medicine, University of Ibadan Ibadan Oyo State Nigeria
- Department of Oral PathologyKing Saud University Medical City, DUH Riyadh Saudi Arabia
| | - Taofeeq Oduola
- Department of Chemical PathologyFaculty of Medical Laboratory Sciences, Usmanu Danfodiyo University Sokoto Nigeria
| | - Dolapo P. Oparinde
- Department of Chemical PathologyLadoke Akintola University of Technology Osogbo Osun State Nigeria
| | - Olubunmi G. Ayelagbe
- Department of Chemical PathologyLadoke Akintola University of Technology Osogbo Osun State Nigeria
| | - Hammed O. Ojokuku
- Department of Medical LaboratoryReddington Multispecialist Hospital Victoria Island, Lagos Nigeria
| |
Collapse
|
45
|
Ahmad S, Mahmood R. Mercury chloride toxicity in human erythrocytes: enhanced generation of ROS and RNS, hemoglobin oxidation, impaired antioxidant power, and inhibition of plasma membrane redox system. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2019; 26:5645-5657. [PMID: 30612358 DOI: 10.1007/s11356-018-04062-5] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 12/20/2018] [Indexed: 06/09/2023]
Abstract
Mercury is among the most toxic heavy metals and a widespread environmental pollutant. Mercury chloride (HgCl2) is an inorganic compound of mercury which is easily absorbed in the gastrointestinal tract and then enters the blood where it can interact with erythrocytes. In this study, the effect of HgCl2 on human erythrocytes was studied under in vitro conditions. Erythrocytes were treated with different concentrations of HgCl2 (1-100 μM) for 1 h at 37 °C. Cell lysates were prepared and assayed for several biochemical parameters. HgCl2 treatment resulted in oxidation of ferrous iron of hemoglobin to ferric form giving methemoglobin which is inactive as an oxygen transporter. However, the activity of methemoglobin reductase was increased. Hemoglobin oxidation was accompanied by heme degradation and the release of free iron. Protein oxidation was greatly increased with a simultaneous decrease in free amino and sulfhydryl groups and glutathione content. The antioxidant power of HgCl2-treated erythrocytes was impaired resulting in lowered metal reducing and free radical quenching ability of these cells. This suggests that HgCl2 induces oxidative stress in human erythrocytes. This was confirmed when superoxide anion, hydrogen peroxide, peroxynitrite, and nitric oxide generation were found to be dose-dependently increased in HgCl2-treated erythrocytes. Glycolysis and pentose phosphate pathway, the two major pathways of glucose metabolism in erythrocytes, were also inhibited. HgCl2 treatment also inhibited the plasma membrane redox system while the activities of AMP deaminase and glyoxalase-I were increased. These results show that HgCl2 induces oxidative and nitrosative stress, oxidizes hemoglobin, impairs the antioxidant defense mechanism, and alters metabolic pathways in human erythrocytes.
Collapse
Affiliation(s)
- Shahbaz Ahmad
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, U.P., 202002, India
| | - Riaz Mahmood
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, U.P., 202002, India.
| |
Collapse
|
46
|
Antwi-Boasiako C, Dankwah GB, Aryee R, Hayfron-Benjamin C, Donkor ES, Campbell AD. Oxidative Profile of Patients with Sickle Cell Disease. Med Sci (Basel) 2019; 7:medsci7020017. [PMID: 30691006 PMCID: PMC6410293 DOI: 10.3390/medsci7020017] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 01/09/2019] [Accepted: 01/18/2019] [Indexed: 01/29/2023] Open
Abstract
Oxidative stress plays a very significant role in the pathophysiology of sickle cell disease (SCD) and associated complications. Oxidative stress, which is often experienced by SCD patients as a result of continuous production of reactive oxygen species (ROS), may lead to endothelial dysfunction and acute inflammation. Antioxidant enzymes, such as superoxide dismutase (SOD) and catalase (CAT), often play a protective role. The current study aimed at determining the oxidative profile of persons with SCD at a tertiary hospital in Ghana. This was a case-control study involving 90 patients with SCD (34 HbSS patients at steady state, 30 HbSC at steady state, 15 HbSS with vaso-occlusive crisis, 11 HbSC with vaso-occlusive crisis), and 50 HbAA control group. Whole blood samples were collected from the study participants and analyzed for full blood counts. The blood samples were assayed for SOD and CAT as a measure of antioxidant defense, while lipid peroxidation was quantified as malondialdehyde (MDA). The results showed that the levels of SOD and CAT were significantly lower in SCD patients as compared to the control group. Patients with HbSS vaso-occlusive crisis had the lowest levels of SOD and CAT. The difference in SOD levels between HbSS at steady state and HbSC with vaso-occlusive crisis was, however, not significant (p = 0.228). The MDA level was significantly higher in SCD patients compared to the control group. This study concludes that the levels of various antioxidant enzymes (erythrocyte SOD and erythrocyte CAT) and oxidative marker (MDA) and are altered in SCD patients.
Collapse
Affiliation(s)
- Charles Antwi-Boasiako
- Department of Physiology, School of Biomedical and Allied Health Sciences, University of Ghana, Accra +233, Ghana.
| | - Gifty B Dankwah
- Department of Physiology, School of Biomedical and Allied Health Sciences, University of Ghana, Accra +233, Ghana.
| | - Robert Aryee
- Department of Physiology, School of Biomedical and Allied Health Sciences, University of Ghana, Accra +233, Ghana.
| | - Charles Hayfron-Benjamin
- Department of Physiology, School of Biomedical and Allied Health Sciences, University of Ghana, Accra +233, Ghana.
- Department of Anaesthesia, School of Medicine and Dentistry, University of Ghana, Accra +233, Ghana.
| | - Eric S Donkor
- Department of Medical Microbiology, School of Biomedical and Allied Health Sciences, University of Ghana, Accra +233, Ghana.
| | - Andrew D Campbell
- Center for Cancer and Blood Disorders Children's National Medical Center George Washington University School of Medicine and Health Sciences, Washington, DC 20052, USA.
| |
Collapse
|
47
|
Membrane protein carbonylation of Plasmodium falciparum infected erythrocytes under conditions of sickle cell trait and G6PD deficiency. Mol Biochem Parasitol 2018; 227:5-14. [PMID: 30472238 DOI: 10.1016/j.molbiopara.2018.11.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 11/09/2018] [Accepted: 11/20/2018] [Indexed: 12/13/2022]
Abstract
Deficiency of glucose-6-phosphate dehydrogenase (G6PD) and sickle cell trait (SCT) are described as the polymorphic disorders prevalent in erythrocytes. Both are considered the result of the selective pressure exerted by Plasmodium parasites over human genome, due to a certain degree of resistance to the clinical symptoms of severe malaria. There exist in both a prooxidant environment that favors the oxidative damage on membrane proteins, which probably is part of molecular protector mechanisms. Nevertheless, mechanisms are not completely understood at molecular level for each polymorphism yet, and even less if are commons for several of them. Here, synchronous cultures at high parasitemia levels of P. falciparum 3D7 were used to quantify oxidative damage in membrane proteins of erythrocytes with G6PD deficient and SCT. Carbonyl index by dot blot assay was used to calculate the variation of oxidative damage during the asexual phases. Besides, protein carbonylation profiles were obtained by Western blot and complemented with mass spectrometry using MALDI-TOF-TOF analysis. Erythrocytes with G6PD deficient and SCT showed higher carbonyl index values than control and similar profiles of carbonylated proteins; moreover, cytoskeletal and stress response proteins were identified as the main targets of oxidative damage. Therefore, both polymorphisms promote carbonylation on the same membrane proteins. Finally, these results allowed to reinforce the hypothesis of oxidative damage in erythrocyte membrane proteins as molecular mechanism of human adaptation to malaria infection.
Collapse
|
48
|
Rivers A, Jagadeeswaran R, Lavelle D. Potential role of LSD1 inhibitors in the treatment of sickle cell disease: a review of preclinical animal model data. Am J Physiol Regul Integr Comp Physiol 2018; 315:R840-R847. [PMID: 30067082 DOI: 10.1152/ajpregu.00440.2017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Sickle cell disease (SCD) is caused by a mutation of the β-globin gene (Ingram VM. Nature 180: 326-328, 1957), which triggers the polymerization of deoxygenated sickle hemoglobin (HbS). Approximately 100,000 SCD patients in the United States and millions worldwide (Piel FB, et al. PLoS Med 10: e1001484, 2013) suffer from chronic hemolytic anemia, painful crises, multisystem organ damage, and reduced life expectancy (Rees DC, et al. Lancet 376: 2018-2031, 2010; Serjeant GR. Cold Spring Harb Perspect Med 3: a011783, 2013). Hematopoietic stem cell transplantation can be curative, but the majority of patients do not have a suitable donor (Talano JA, Cairo MS. Eur J Haematol 94: 391-399, 2015). Advanced gene-editing technologies also offer the possibility of a cure (Goodman MA, Malik P. Ther Adv Hematol 7: 302-315, 2016; Lettre G, Bauer DE. Lancet 387: 2554-2564, 2016), but the likelihood that these strategies can be mobilized to treat the large numbers of patients residing in developing countries is remote. A pharmacological treatment to increase fetal hemoglobin (HbF) as a therapy for SCD has been a long-sought goal, because increased levels of HbF (α2γ2) inhibit the polymerization of HbS (Poillin WN, et al. Proc Natl Acad Sci USA 90: 5039-5043, 1993; Sunshine HR, et al. J Mol Biol 133: 435-467, 1979) and are associated with reduced symptoms and increased lifespan of SCD patients (Platt OS, et al. N Engl J Med 330: 1639-1644, 1994; Platt OS, et al. N Engl J Med 325: 11-16, 1991). Only two drugs, hydroxyurea and l-glutamine, are approved by the US Food and Drug Administration for treatment of SCD. Hydroxyurea is ineffective at HbF induction in ~50% of patients (Charache S, et al. N Engl J Med 332: 1317-1322, 1995). While polymerization of HbS has been traditionally considered the driving force in the hemolysis of SCD, the excessive reactive oxygen species generated from red blood cells, with further amplification by intravascular hemolysis, also are a major contributor to SCD pathology. This review highlights a new class of drugs, lysine-specific demethylase (LSD1) inhibitors, that induce HbF and reduce reactive oxygen species.
Collapse
Affiliation(s)
- Angela Rivers
- Department of Pediatrics, University of Illinois at Chicago , Chicago, Illinois.,Jesse Brown Veterans Affairs Medical Center , Chicago, Illinois
| | - Ramasamy Jagadeeswaran
- Department of Pediatrics, University of Illinois at Chicago , Chicago, Illinois.,Jesse Brown Veterans Affairs Medical Center , Chicago, Illinois
| | - Donald Lavelle
- Department of Medicine, University of Illinois at Chicago , Chicago, Illinois.,Jesse Brown Veterans Affairs Medical Center , Chicago, Illinois
| |
Collapse
|
49
|
Shchukina OV, Chukicheva IY, Shevchenko OG, Kolegova TA, Suponitsky KY, Kutchin AV. Synthesis and Properties of Functional Derivatives of 2,6-Diisobornylphenol and 2-Isobornyl-6-(1-phenylethyl)phenol. RUSS J GEN CHEM+ 2018. [DOI: 10.1134/s1070363218040096] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
50
|
Louie JE, Anderson CJ, Fayaz M. Fomani K, Henry A, Killeen T, Mohandas N, Yazdanbakhsh K, Belcher JD, Vercellotti GM, Shi PA. Case series supporting heme detoxification via therapeutic plasma exchange in acute multiorgan failure syndrome resistant to red blood cell exchange in sickle cell disease. Transfusion 2017; 58:470-479. [DOI: 10.1111/trf.14407] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 10/08/2017] [Accepted: 10/08/2017] [Indexed: 01/25/2023]
Affiliation(s)
- James E. Louie
- Long Island Jewish Medical Center, Northwell Health; New Hyde Park New York
| | - Caitlin J. Anderson
- Lindley F. Kimball Research Institute, New York Blood Center; New York New York
| | | | - Alonye Henry
- Lindley F. Kimball Research Institute, New York Blood Center; New York New York
| | - Trevor Killeen
- Department of Hematology, Oncology, and Transplantation; University of Minnesota Medical School; Minneapolis Minnesota
| | - Narla Mohandas
- Lindley F. Kimball Research Institute, New York Blood Center; New York New York
| | - Karina Yazdanbakhsh
- Lindley F. Kimball Research Institute, New York Blood Center; New York New York
| | - John D. Belcher
- Department of Hematology, Oncology, and Transplantation; University of Minnesota Medical School; Minneapolis Minnesota
| | - Gregory M. Vercellotti
- Department of Hematology, Oncology, and Transplantation; University of Minnesota Medical School; Minneapolis Minnesota
| | - Patricia A. Shi
- Lindley F. Kimball Research Institute, New York Blood Center; New York New York
| |
Collapse
|