1
|
Khalid M, Suhail M, Faisal A, Poombal F. β-Thalassemia Major Complicated by Acute Myeloid Leukemia. Cureus 2024; 16:e69557. [PMID: 39421114 PMCID: PMC11486485 DOI: 10.7759/cureus.69557] [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] [Accepted: 09/16/2024] [Indexed: 10/19/2024] Open
Abstract
This report describes the rare co-occurrence of acute myeloid leukemia (AML) French-American-British type M2 in a 4.5-year-old boy with previously diagnosed thalassemia major, an inherited hemoglobinopathy, typically presenting with severe, transfusion-dependent anemia. Chronic transfusions, though lifesaving, can lead to iron overload, which may generate free radicals and potentially contribute to malignancy. Our case highlights the importance of close monitoring for secondary malignancies in thalassemia patients. Our patient born to consanguineous parents, presented with persistent fever, abdominal pain, and splenomegaly. Hematological investigations revealed severe cytopenias (low blood cell counts) and many immature blood cells (blasts). Bone marrow examination confirmed AML M2, characterized by an overabundance of myeloid blasts. Despite the initiation of myeloid leukemia-directed aggressive chemotherapy, the patient, unfortunately, succumbed to the disease within a month of diagnosis.
Collapse
Affiliation(s)
- Mehreen Khalid
- Department of Hematopathology, Armed Forces Institute of Pathology, Rawalpindi, PAK
| | - Maymoona Suhail
- Department of Hematology, Armed Forces Institute of Pathology, Rawalpindi, PAK
| | - Alizah Faisal
- Department of Hematology, Rawalpindi Medical University, Rawalpindi, PAK
| | - Fnu Poombal
- Department of Pathology, Nishtar Medical University, Multan, PAK
| |
Collapse
|
2
|
Settakorn K, Hantrakool S, Petiwathayakorn T, Hutachok N, Tantiworawit A, Charoenkwan P, Chalortham N, Chompupoung A, Paradee N, Koonyosying P, Srichairatanakool S. A randomized placebo-controlled clinical trial of oral green tea epigallocatechin 3-gallate on erythropoiesis and oxidative stress in transfusion-dependent β-thalassemia patients. Front Mol Biosci 2024; 10:1248742. [PMID: 38328786 PMCID: PMC10848917 DOI: 10.3389/fmolb.2023.1248742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 12/28/2023] [Indexed: 02/09/2024] Open
Abstract
β-Thalassemia patients suffer from ineffective erythropoiesis and increased red blood cell (RBC) hemolysis. Blood transfusion, erythropoietic enhancement, and antioxidant supplementation can ameliorate chronic anemia. Green tea extract (GTE) is comprised of catechin derivatives, of which epigallocatechin-3-gallate (EGCG) is the most abundant, presenting free-radical scavenging, iron-chelating, and erythropoiesis-protective effects. The present study aimed to evaluate the effects of GTE tablets on the primary outcome of erythropoiesis and oxidative stress parameters in transfusion-dependent β-thalassemia (TDT) patients. Twenty-seven TDT patients were randomly divided into placebo and GTE tablet (50 and 100 mg EGCG equivalent) groups and assigned to consume the product once daily for 60 days. Blood was collected for analysis of hematological, biochemical, and oxidative stress parameters. Accordingly, consumption of GTE tablets improved blood hemoglobin levels when compared with the placebo; however, there were more responders to the GTE tablets. Interestingly, amounts of nonheme iron in RBC membranes tended to decrease in both GTE tablet groups when compared with the placebo. Importantly, consumption of GTE tablets lowered plasma levels of erythroferrone (p < 0.05) and reduced bilirubin non-significantly and dose-independently. Thus, GTE tablets could improve RBC hemolysis and modulate erythropoiesis regulators in transfusion-dependent thalassemia patients.
Collapse
Affiliation(s)
- Kornvipa Settakorn
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Sasinee Hantrakool
- Division of Hematology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Thalassemia and Hematology Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | | | - Nuntouchaporn Hutachok
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Adisak Tantiworawit
- Division of Hematology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Thalassemia and Hematology Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Pimlak Charoenkwan
- Division of Hematology and Oncology, Department of Pediatrics, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Nopphadol Chalortham
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai, Thailand
| | | | - Narisara Paradee
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Pimpisid Koonyosying
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | | |
Collapse
|
3
|
Settakorn K, Kongkarnka S, Chompupoung A, Svasti S, Fucharoen S, Porter JB, Srichairatanakool S, Koonyosying P. Effects of green tea extract treatment on erythropoiesis and iron parameters in iron-overloaded β-thalassemic mice. Front Physiol 2022; 13:1053060. [PMID: 36620219 PMCID: PMC9816339 DOI: 10.3389/fphys.2022.1053060] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Accepted: 11/29/2022] [Indexed: 12/24/2022] Open
Abstract
β-Thalassemia is characterized by ineffective erythropoiesis leading to chronic anemia. Thus, increased iron absorption from the duodenum and via blood transfusions is required to maintain normal blood hemoglobin (Hb) levels and iron chelators in the removal of excessive iron. Certain agents are also needed for the improvement of stress erythropoiesis and iron dysregulation. Green tea extract (GTE), which is rich in epigallocatechin-3-gallate (EGCG), is known to possess radical scavenging and iron-chelating activities. We aimed to assess the effects of green tea extract on erythroid regulators, iron mobilization and anti-lipid peroxidation in the liver, spleen, and kidneys of iron-loaded β-globin gene knockout thalassemic (BKO) mice. Our results indicate that treatments of green tea extract and/or deferiprone (DFP) diminished levels of plasma erythropoietin (EPO) and erythroferrone (ERFE), and consistently suppressed kidney Epo and spleen Erfe mRNA expressions (p < .05) in iron- loaded BKO mice when compared with untreated mice. Coincidently, the treatments decreased plasma ferritin (Ft) levels, iron content levels in the liver (p < .05), spleen (p < .05), and kidney tissues of iron-loaded BKO mice. Furthermore, lipid-peroxidation products in the tissues and plasma were also decreased when compared with untreated mice. This is the first evidence of the orchestral role of green tea extract abundant with epigallocatechin-3-gallate in improving ineffective erythropoiesis, iron dysregulation and oxidative stress in iron-overloaded β-thalassemic mice.
Collapse
Affiliation(s)
- Kornvipa Settakorn
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Sarawut Kongkarnka
- Department of Pathology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | | | - Saovaros Svasti
- Thalassemia Research Center, Institute of Molecular Biosciences, Mahidol University Salaya Campus, Nakorn Pathom, Thailand
| | - Suthat Fucharoen
- Thalassemia Research Center, Institute of Molecular Biosciences, Mahidol University Salaya Campus, Nakorn Pathom, Thailand
| | - John B. Porter
- Red Cell Disorder Unit, Department of Haematology, University College London, London, United Kingdom
| | - Somdet Srichairatanakool
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand,*Correspondence: Somdet Srichairatanakool, ; Pimpisid Koonyosying,
| | - Pimpisid Koonyosying
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand,*Correspondence: Somdet Srichairatanakool, ; Pimpisid Koonyosying,
| |
Collapse
|
4
|
The Effect of Low-Energy Laser-Driven Ultrashort Pulsed Electron Beam Irradiation on Erythropoiesis and Oxidative Stress in Rats. Int J Mol Sci 2022; 23:ijms23126692. [PMID: 35743135 PMCID: PMC9223873 DOI: 10.3390/ijms23126692] [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: 05/18/2022] [Revised: 06/06/2022] [Accepted: 06/10/2022] [Indexed: 12/04/2022] Open
Abstract
Anemia is a commonly observed consequence of whole-body exposure to a dose of X-ray or gamma irradiation of the order of the mean lethal dose in mammals, and it is an important factor for the determination of the survival of animals. The aim of this study was to unravel the effect of laser-driven ultrashort pulsed electron beam (UPEB) irradiation on the process of erythropoiesis and the redox state in the organism. Wistar rats were exposed to laser-driven UPEB irradiation, after which the level of oxidative stress and the activities of different antioxidant enzymes, as well as blood smears, bone marrow imprints and sections, erythroblastic islets, hemoglobin and hematocrit, hepatic iron, DNA, and erythropoietin levels, were assessed on the 1st, 3rd, 7th, 14th, and 28th days after irradiation. Despite the fact that laser-driven UPEB irradiation requires quite low doses and repetition rates to achieve the LD50 in rats, our findings suggest that whole-body exposure with this new type of irradiation causes relatively mild anemia in rats, with subsequent fast recovery up to the 28th day. Moreover, this novel type of irradiation causes highly intense processes of oxidative stress, which, despite being relatively extinguished, did not reach the physiologically stable level even at the 28th day after irradiation due to the violations in the antioxidant system of the organism.
Collapse
|
5
|
Correcting β-thalassemia by combined therapies that restrict iron and modulate erythropoietin activity. Blood 2021; 136:1968-1979. [PMID: 32556142 DOI: 10.1182/blood.2019004719] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Accepted: 05/21/2020] [Indexed: 11/20/2022] Open
Abstract
β-Thalassemia intermedia is a disorder characterized by ineffective erythropoiesis (IE), anemia, splenomegaly, and systemic iron overload. Novel approaches are being explored based on the modulation of pathways that reduce iron absorption (ie, using hepcidin activators like Tmprss6-antisense oligonucleotides [ASOs]) or increase erythropoiesis (by erythropoietin [EPO] administration or modulating the ability of transferrin receptor 2 [Tfr2] to control red blood cell [RBC] synthesis). Targeting Tmprss6 messenger RNA by Tmprss6-ASO was proven to be effective in improving IE and splenomegaly by inducing iron restriction. However, we postulated that combinatorial strategies might be superior to single therapies. Here, we combined Tmprss6-ASO with EPO administration or removal of a single Tfr2 allele in the bone marrow of animals affected by β-thalassemia intermedia (Hbbth3/+). EPO administration alone or removal of a single Tfr2 allele increased hemoglobin levels and RBCs. However, EPO or Tfr2 single-allele deletion alone, respectively, exacerbated or did not improve splenomegaly in β-thalassemic mice. To overcome this issue, we postulated that some level of iron restriction (by targeting Tmprss6) would improve splenomegaly while preserving the beneficial effects on RBC production mediated by EPO or Tfr2 deletion. While administration of Tmprss6-ASO alone improved the anemia, the combination of Tmprss6-ASO + EPO or Tmprss6-ASO + Tfr2 single-allele deletion produced significantly higher hemoglobin levels and reduced splenomegaly. In conclusion, our results clearly indicate that these combinatorial approaches are superior to single treatments in ameliorating IE and anemia in β-thalassemia and could provide guidance to translate some of these approaches into viable therapies.
Collapse
|
6
|
Fibach E. The Redox Balance and Membrane Shedding in RBC Production, Maturation, and Senescence. Front Physiol 2021; 12:604738. [PMID: 33664673 PMCID: PMC7920951 DOI: 10.3389/fphys.2021.604738] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 01/18/2021] [Indexed: 12/25/2022] Open
Abstract
Membrane shedding in the form of extracellular vesicles plays a key role in normal physiology and pathology. Partial disturbance of the membrane-cytoskeleton linkage and increased in the intracellular Ca content are considered to be mechanisms underlying the process, but it is questionable whether they constitute the primary initiating steps. Homeostasis of the redox system, which depends on the equilibrium between oxidants and antioxidants, is crucial for many cellular processes. Excess oxidative power results in oxidative stress, which affects many cellular components, including the membrane. Accumulating evidence suggests that oxidative stress indirectly affects membrane shedding most probably by affecting the membrane-cytoskeleton and the Ca content. In red blood cells (RBCs), changes in both the redox system and membrane shedding occur throughout their life-from birth-their production in the bone marrow, to death-aging in the peripheral blood and removal by macrophages in sites of the reticuloendothelial system. Both oxidative stress and membrane shedding are disturbed in diseases affecting the RBC, such as the hereditary and acquired hemolytic anemias (i.e., thalassemia, sickle cell anemia, and autoimmune hemolytic anemia). Herein, I review some data-based and hypothetical possibilities that await experimental confirmation regarding some aspects of the interaction between the redox system and membrane shedding and its role in the normal physiology and pathology of RBCs.
Collapse
Affiliation(s)
- Eitan Fibach
- Department of Hematology, Hadassah University Hospital, Jerusalem, Israel
| |
Collapse
|
7
|
Oxidative DNA Damage, Inflammatory Signature, and Altered Erythrocytes Properties in Diamond-Blackfan Anemia. Int J Mol Sci 2020; 21:ijms21249652. [PMID: 33348919 PMCID: PMC7768356 DOI: 10.3390/ijms21249652] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 12/15/2020] [Indexed: 12/12/2022] Open
Abstract
Molecular pathophysiology of Diamond-Blackfan anemia (DBA) involves disrupted erythroid-lineage proliferation, differentiation and apoptosis; with the activation of p53 considered as a key component. Recently, oxidative stress was proposed to play an important role in DBA pathophysiology as well. CRISPR/Cas9-created Rpl5- and Rps19-deficient murine erythroleukemia (MEL) cells and DBA patients' samples were used to evaluate proinflammatory cytokines, oxidative stress, DNA damage and DNA damage response. We demonstrated that the antioxidant defense capacity of Rp-mutant cells is insufficient to meet the greater reactive oxygen species (ROS) production which leads to oxidative DNA damage, cellular senescence and activation of DNA damage response signaling in the developing erythroblasts and altered characteristics of mature erythrocytes. We also showed that the disturbed balance between ROS formation and antioxidant defense is accompanied by the upregulation of proinflammatory cytokines. Finally, the alterations detected in the membrane of DBA erythrocytes may cause their enhanced recognition and destruction by reticuloendothelial macrophages, especially during infections. We propose that the extent of oxidative stress and the ability to activate antioxidant defense systems may contribute to high heterogeneity of clinical symptoms and response to therapy observed in DBA patients.
Collapse
|
8
|
Georgatzakou HT, Tzounakas VL, Velentzas AD, Papassideri IS, Kokkalis AC, Stamoulis KE, Kriebardis AG, Antonelou MH. Recipient's effects on stored red blood cell performance: the case of uremic plasma. Transfusion 2019; 59:1900-1906. [PMID: 30888086 DOI: 10.1111/trf.15257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 02/21/2019] [Accepted: 02/22/2019] [Indexed: 11/30/2022]
Abstract
BACKGROUND Despite universal administration of erythropoiesis-stimulating agents, patients with end-stage renal disease (ESRD) are at high risk for presenting persistent anemia. Due to ambiguities in optimal hemoglobin targets and evidence of recombinant human erythropoietin (EPO)-related toxicity, an increase in blood transfusions has been observed in chronic renal disease over the past years. The probable effects of uremic plasma on the performance of stored red blood cells (RBCs) after transfusion have not been investigated. STUDY DESIGN AND METHODS Leukoreduced RBCs after short or long storage in CPD-SAGM (n = 5) were assessed for hemolysis, surface removal signaling, reactive oxygen species (ROS) accumulation, and shape distortions before and after reconstitution with healthy (n = 10) or uremic plasma from ESRD patients (n = 20) for 24 hours at physiologic temperature, by using a previously reported in vitro model of transfusion. RESULTS Temperature and cell environment shifts from blood bag to plasma independently and in synergy affected the RBC physiology. Outcome measures at transfusion-simulating conditions might not be analogous to timing of storage lesion. The uremic plasma ameliorated the susceptibility of stored RBCs to hemolysis, phosphatidylserine externalization, and ROS generation after stimulation by oxidants, but negatively affected shape homeostasis versus healthy plasma. Creatinine, uric acid, and EPO levels had correlations with the performance of stored RBCs in ESRD plasma. CONCLUSION Renal insufficiency and EPO supplementation likely affect the recovery of donor RBCs and the reactivity of RBCs after transfusion by exerting both toxic and cytoprotective influences on them. ESRD patients constitute a specific recipient group that deserves further examination.
Collapse
Affiliation(s)
- Hara T Georgatzakou
- Department of Biology, School of Science, National & Kapodistrian University of Athens (NKUA), Athens, Greece
| | - Vassilis L Tzounakas
- Department of Biology, School of Science, National & Kapodistrian University of Athens (NKUA), Athens, Greece
| | - Athanassios D Velentzas
- Department of Biology, School of Science, National & Kapodistrian University of Athens (NKUA), Athens, Greece
| | - Issidora S Papassideri
- Department of Biology, School of Science, National & Kapodistrian University of Athens (NKUA), Athens, Greece
| | | | | | - Anastasios G Kriebardis
- Department of Biomedical Science, School of Health & Caring Science, University of West Attica (UniWA), Egaleo, Greece
| | - Marianna H Antonelou
- Department of Biology, School of Science, National & Kapodistrian University of Athens (NKUA), Athens, Greece
| |
Collapse
|
9
|
Ferreira FA, Benites BD, Costa FF, Gilli S, Olalla-Saad ST. Recombinant erythropoietin as alternative to red cell transfusion in sickle cell disease. Vox Sang 2019; 114:178-181. [PMID: 30695814 DOI: 10.1111/vox.12750] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 12/25/2018] [Accepted: 12/27/2018] [Indexed: 11/28/2022]
Abstract
Disturbances in the physiological regulation of erythropoietin (EPO) in patients with sickle cell disease (SCD) may contribute to worsening anaemia and increased transfusion requirements, but the use of recombinant EPO in this group of patients is controversial. The objective of this study was to evaluate the use of this drug in adult patients with SCD and its effects on haemoglobin levels and transfusion requirements. We conducted a retrospective analysis at the University of Campinas, with nineteen adults with a diagnosis of SCD (HbSS and HbS/β+ thalassaemia), who had received at least 1 year of EPO therapy between 2007 and 2014. Haemoglobin concentrations and trends of variation in transfused RBC volumes were compared before and after EPO administration. We observed that seven patients had a good response to treatment (Hb increment higher than 1·5 g/dl) and nine had a partial response (0·5-1·5 g/dl increment) and there was a significant decrease in the need for transfusion amongst those who usually required regular transfusions. There were no increases in the rates of vaso-occlusive crisis or venous thromboembolism in comparison to the year before the onset of the therapy. Erythropoietin therapy led to a marked increase in haemoglobin concentration with a concomitant decrease in the demand for transfusion. Considering all complications related to allogeneic transfusion, we believe that EPO therapy represents an important therapeutic tool in sickle cell anaemia.
Collapse
Affiliation(s)
- Francisco A Ferreira
- Hematology and Transfusion Medicine Center, University of Campinas, Campinas, Brazil
| | - Bruno D Benites
- Hematology and Transfusion Medicine Center, University of Campinas, Campinas, Brazil
| | - Fernando F Costa
- Hematology and Transfusion Medicine Center, University of Campinas, Campinas, Brazil
| | - Simone Gilli
- Hematology and Transfusion Medicine Center, University of Campinas, Campinas, Brazil
| | - Sara T Olalla-Saad
- Hematology and Transfusion Medicine Center, University of Campinas, Campinas, Brazil
| |
Collapse
|
10
|
Management of Children With β-Thalassemia Intermedia: Overview, Recent Advances, and Treatment Challenges. J Pediatr Hematol Oncol 2018; 40:253-268. [PMID: 29629992 DOI: 10.1097/mph.0000000000001148] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Our knowledge of the various clinical morbidities that thalassemia intermedia (TI) patients endure has substantially increased over the past decade. It is mandatory to grasp a solid understanding of disease-specific complications in order to tailor management. The optimal course of management for TI patients has been hard to identify, and several controversies remain with regard to the best treatment plan. Although advances in TI are moving at a fast pace, many complications remain with no treatment guidelines. Studies that expand our understanding of the mechanisms and risk factors, as well as clinical trials evaluating the roles of available treatments, will help establish management guidelines that improve patient care. Novel therapeutic modalities are now emerging. This article focuses on the management of children with β-TI. We present various clinical morbidities and their association with the underlying disease pathophysiology and risk factors. All therapeutic options, recent advances, and treatment challenges were reviewed.
Collapse
|
11
|
Georgatzakou HT, Tzounakas VL, Kriebardis AG, Velentzas AD, Papageorgiou EG, Voulgaridou AI, Kokkalis AC, Antonelou MH, Papassideri IS. Pathophysiological aspects of red blood cells in end-stage renal disease patients resistant to recombinant human erythropoietin therapy. Eur J Haematol 2017; 98:590-600. [DOI: 10.1111/ejh.12875] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/02/2017] [Indexed: 12/13/2022]
Affiliation(s)
- Hara T. Georgatzakou
- Department of Biology; Section of Cell Biology & Biophysics; School of Science; National and Kapodistrian University of Athens (NKUA); Athens Greece
| | - Vassilis L. Tzounakas
- Department of Biology; Section of Cell Biology & Biophysics; School of Science; National and Kapodistrian University of Athens (NKUA); Athens Greece
| | - Anastasios G. Kriebardis
- Department of Medical Laboratories; Faculty of Health and Caring Professions; Technological and Educational Institute (TEI) of Athens; Athens Greece
| | - Athanassios D. Velentzas
- Department of Biology; Section of Cell Biology & Biophysics; School of Science; National and Kapodistrian University of Athens (NKUA); Athens Greece
| | - Effie G. Papageorgiou
- Department of Medical Laboratories; Faculty of Health and Caring Professions; Technological and Educational Institute (TEI) of Athens; Athens Greece
| | | | | | - Marianna H. Antonelou
- Department of Biology; Section of Cell Biology & Biophysics; School of Science; National and Kapodistrian University of Athens (NKUA); Athens Greece
| | - Issidora S. Papassideri
- Department of Biology; Section of Cell Biology & Biophysics; School of Science; National and Kapodistrian University of Athens (NKUA); Athens Greece
| |
Collapse
|
12
|
Sundem L, Chris Tseng KC, Li H, Ketz J, Noble M, Elfar J. Erythropoietin Enhanced Recovery After Traumatic Nerve Injury: Myelination and Localized Effects. J Hand Surg Am 2016; 41:999-1010. [PMID: 27593486 PMCID: PMC5053901 DOI: 10.1016/j.jhsa.2016.08.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 06/08/2016] [Accepted: 08/06/2016] [Indexed: 02/02/2023]
Abstract
PURPOSE We previously found that administration of erythropoietin (EPO) shortens the course of recovery after experimental crush injury to the mouse sciatic nerve. The course of recovery was more rapid than would be expected if EPO's effects were caused by axonal regeneration, which raised the question of whether recovery was instead the result of promoting remyelination and/or preserving myelin on injured neurons. This study tested the hypothesis that EPO has a direct and local effect on myelination in vivo and in vitro. METHODS Animals were treated with EPO after standard calibrated sciatic nerve crush injury; immunohistochemical analysis was performed to assay for myelinated axons. Combined in vitro neuron-Schwann cell co-cultures were performed to assess EPO-mediated effects directly on myelination and putative protective effects against oxidative stress. In vivo local administration of EPO in a fibrin glue carrier was used to demonstrate early local effects of EPO treatment well in advance of possible neuroregenerative effects. RESULTS Systemic Administration of EPO maintained more in vivo myelinated axons at the site of nerve crush injury. In vitro, EPO treatment promoted myelin formation and protected myelin from the effects of nitric oxide exposure in co-cultures of Schwann cells and dorsal root ganglion neurons. In a novel, surgically applicable local treatment using Food and Drug Administration-approved fibrin glue as a vehicle, EPO was as effective as systemic EPO administration at time points earlier than those explainable using standard models of neuroregeneration. CONCLUSIONS In nerve crush injury, EPO may be exerting a primary influence on myelin status to promote functional recovery. CLINICAL RELEVANCE Mixed injury to myelin and axons may allow the opportunity for the repurposing of EPO for use as a myeloprotective agent in which injuries spare a requisite number of axons to allow early functional recovery.
Collapse
Affiliation(s)
- Leigh Sundem
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY
| | | | - Haiyan Li
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY
| | - John Ketz
- Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY
| | - Mark Noble
- Department of Biomedical Genetics, Institute for Stem Cell and Regenerative Medicine, University of Rochester Medical Center, Rochester, NY
| | - John Elfar
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY; Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY.
| |
Collapse
|
13
|
Georgatzakou HT, Antonelou MH, Papassideri IS, Kriebardis AG. Red blood cell abnormalities and the pathogenesis of anemia in end-stage renal disease. Proteomics Clin Appl 2016; 10:778-90. [PMID: 26948278 DOI: 10.1002/prca.201500127] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 01/14/2016] [Accepted: 02/29/2016] [Indexed: 12/20/2022]
Abstract
Anemia is the most common hematologic complication in end-stage renal disease (ESRD). It is ascribed to decreased erythropoietin production, shortened red blood cell (RBC) lifespan, and inflammation. Uremic toxins severely affect RBC lifespan; however, the implicated molecular pathways are poorly understood. Moreover, current management of anemia in ESRD is controversial due to the "anemia paradox" phenomenon, which underlines the need for a more individualized approach to therapy. RBCs imprint the adverse effects of uremic, inflammatory, and oxidative stresses in a context of structural and functional deterioration that is associated with RBC removal signaling and morbidity risk. RBCs circulate in hostile plasma by raising elegant homeostatic defenses. Variability in primary defect, co-morbidity, and therapeutic approaches add complexity to the pathophysiological background of the anemic ESRD patient. Several blood components have been suggested as biomarkers of anemia-related morbidity and mortality risk in ESRD. However, a holistic view of blood cell and plasma modifications through integrated omics approaches and high-throughput studies might assist the development of new diagnostic tests and therapies that will target the underlying pathophysiologic processes of ESRD anemia.
Collapse
Affiliation(s)
- Hara T Georgatzakou
- Department of Cell Biology and Biophysics, Faculty of Biology, National and Kapodistrian University of Athens, Greece
| | - Marianna H Antonelou
- Department of Cell Biology and Biophysics, Faculty of Biology, National and Kapodistrian University of Athens, Greece
| | - Issidora S Papassideri
- Department of Cell Biology and Biophysics, Faculty of Biology, National and Kapodistrian University of Athens, Greece
| | - Anastasios G Kriebardis
- Department of Medical Laboratories, Faculty of Health and Caring Professions, Technological and Educational Institute of Athens, Greece
| |
Collapse
|
14
|
Fibach E, Dana M. Oxidative stress in paroxysmal nocturnal hemoglobinuria and other conditions of complement-mediated hemolysis. Free Radic Biol Med 2015; 88:63-9. [PMID: 25937178 DOI: 10.1016/j.freeradbiomed.2015.04.027] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Revised: 02/23/2015] [Accepted: 04/21/2015] [Indexed: 12/11/2022]
Abstract
The complement (C') system and redox status play important roles in the physiological functioning of the body, such as the defense system, but they are also involved in various pathological conditions, including hemolytic anemia. Herein, we review the interaction between the C' and the redox systems in C'-mediated hemolytic anemias, paroxysmal nocturnal hemoglobinuria (PNH) and autoimmune hemolytic anemia, including acute hemolytic transfusion reaction. Blood cells in these diseases have been shown to have increased oxidative status, which was further elevated by interaction with activated C'. The results suggest that oxidative stress, in conjunction with activated C', may cause the underlying symptoms of these diseases, such as intra- and extravascular hemolysis and thrombotic complications. Antioxidants ameliorate oxidative stress by preventing generation of free radicals, by scavenging and preventing their accumulation, and by correcting their cellular damage. Antioxidants have been shown to reduce the oxidative stress and inhibit hemolysis as well as platelet activation mediated by activated C'. This raises the possibility that treatment with antioxidants might be considered as a potential therapeutic modality for C'-mediated hemolytic anemias. Currently, eculizumab, a humanized monoclonal antibody that specifically targets the C' protein C5, is the main treatment modality for PNH. However, because antioxidants are well tolerated and relatively inexpensive, they might be considered as potential adjuvants or an alternative therapeutic modality for PNH and other C'-mediated hemolytic anemias.
Collapse
Affiliation(s)
- Eitan Fibach
- Department of Hematology, Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel.
| | - Mutaz Dana
- Department of Hematology, Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel
| |
Collapse
|
15
|
Wang L, Di L, Noguchi CT. Erythropoietin, a novel versatile player regulating energy metabolism beyond the erythroid system. Int J Biol Sci 2014; 10:921-39. [PMID: 25170305 PMCID: PMC4147225 DOI: 10.7150/ijbs.9518] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2014] [Accepted: 06/04/2014] [Indexed: 12/12/2022] Open
Abstract
Erythropoietin (EPO), the required cytokine for promoting the proliferation and differentiation of erythroid cells to stimulate erythropoiesis, has been reported to act as a pleiotropic cytokine beyond hematopoietic system. The various activities of EPO are determined by the widespread distribution of its cell surface EPO receptor (EpoR) in multiple tissues including endothelial, neural, myoblasts, adipocytes and other cell types. EPO activity has been linked to angiogenesis, neuroprotection, cardioprotection, stress protection, anti-inflammation and especially the energy metabolism regulation that is recently revealed. The investigations of EPO activity in animals and the expression analysis of EpoR provide more insights on the potential of EPO in regulating energy metabolism and homeostasis. The findings of crosstalk between EPO and some important energy sensors and the regulation of EPO in the cellular respiration and mitochondrial function further provide molecular mechanisms for EPO activity in metabolic activity regulation. In this review, we will summarize the roles of EPO in energy metabolism regulation and the activity of EPO in tissues that are tightly associated with energy metabolism. We will also discuss the effects of EPO in regulating oxidative metabolism and mitochondrial function, the interactions between EPO and important energy regulation factors, and the protective role of EPO from stresses that are related to metabolism, providing a brief overview of previously less appreciated EPO biological function in energy metabolism and homeostasis.
Collapse
Affiliation(s)
- Li Wang
- 1. Faculty of Health Sciences, University of Macau, SAR of People's Republic of China
| | - Lijun Di
- 1. Faculty of Health Sciences, University of Macau, SAR of People's Republic of China
| | - Constance Tom Noguchi
- 2. Molecular Medicine Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, U.S.A
| |
Collapse
|
16
|
Wang L, Di L, Noguchi CT. AMPK is involved in mediation of erythropoietin influence on metabolic activity and reactive oxygen species production in white adipocytes. Int J Biochem Cell Biol 2014; 54:1-9. [PMID: 24953559 DOI: 10.1016/j.biocel.2014.06.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Revised: 05/29/2014] [Accepted: 06/15/2014] [Indexed: 12/15/2022]
Abstract
Erythropoietin, discovered for its indispensable role during erythropoiesis, has been used in therapy for selected red blood cell disorders in erythropoietin-deficient patients. The biological activities of erythropoietin have been found in animal models to extend to non-erythroid tissues due to the expression of erythropoietin receptor. We previously demonstrated that erythropoietin promotes metabolic activity and white adipocytes browning to increase mitochondrial function and energy expenditure via peroxisome proliferator-activated receptor alpha and Sirtuin1. Here we report that AMP-activated protein kinase was activated by erythropoietin possibly via Ca(2+)/calmodulin-dependent protein kinase kinase in adipocytes as well as in white adipose tissue from diet induced obese mice. Erythropoietin increased cellular nicotinamide adenine dinucleotide via increased AMP-activated protein kinase activity, possibly leading to Sirtuin1 activation. AMP-activated protein kinase knock down reduced erythropoietin mediated increase in cellular oxidative function including the increased oxygen consumption rate, fatty acid utilization and induction of key metabolic genes. Under hypoxia, adipocytes were found to generate more reactive oxygen species, and erythropoietin reduced the reactive oxygen species and increased antioxidant gene expression, suggesting that erythropoietin may provide protection from oxidative stress in adipocytes. Erythropoietin also reversed increased nicotinamide adenine dinucleotide by hypoxia via increased AMP-activated protein kinase. Additionally, AMP-activated protein kinase is found to be involved in erythropoietin stimulated increase in oxygen consumption rate, fatty acid oxidation and mitochondrial gene expression. AMP-activated protein kinase knock down impaired erythropoietin stimulated increases in antioxidant gene expression. Collectively, our findings identify the AMP-activated protein kinase involvement in erythropoietin signaling in regulating adipocyte cellular redox status and metabolic activity.
Collapse
Affiliation(s)
- Li Wang
- Faculty of Health Sciences, University of Macau, Macau SAR, China.
| | - Lijun Di
- Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Constance Tom Noguchi
- Molecular Medicine Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Building 10, Room 9N319, 10 CENTER DR MSC-1822, Bethesda, MD 20892-1822, USA.
| |
Collapse
|
17
|
Fibach E, Rachmilewitz EA. Does erythropoietin have a role in the treatment of β-hemoglobinopathies? Hematol Oncol Clin North Am 2014; 28:249-63. [PMID: 24589265 DOI: 10.1016/j.hoc.2013.11.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
This review presents the indications and contraindications (pros and cons) for the potential use of erythropoietin (Epo) as a treatment in β-thalassemia and sickle cell anemia (SCA). Its high cost and route of administration (by injection) are obvious obstacles, especially in underdeveloped countries, where thalassemia is prevalent. We believe that from the data summarized in this review, the time has come to define, by studying in vitro and in vivo models, as well as by controlled clinical trials, the rationale for treating patients with various forms of thalassemia and SCA with Epo alone or in combination with other medications.
Collapse
Affiliation(s)
- Eitan Fibach
- Department of Hematology, Hadassah-Hebrew University Medical Center, Ein-Kerem, Jerusalem 91120, Israel.
| | | |
Collapse
|
18
|
Elalfy MS, Adly AAM, Ismail EA, Elhenawy YI, Elghamry IR. Therapeutic superiority and safety of combined hydroxyurea with recombinant human erythropoietin over hydroxyurea in young β-thalassemia intermedia patients. Eur J Haematol 2013; 91:522-33. [PMID: 23927461 DOI: 10.1111/ejh.12182] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/29/2013] [Indexed: 11/27/2022]
Abstract
OBJECTIVE To assess the efficacy and safety of combined hydroxyurea (HU) and recombinant human erythropoietin (rHuEPO) in β-thalassemia intermedia (TI) patients compared with single HU therapy. METHODS An interventional prospective randomized study registered in the ClinicalTrials.gov (NCT01624038) was performed on 80 TI patients (≤ 18 yr) divided into group A (40 patients received combined HU and rHuEPO) and group B (40 patients received single HU therapy). Baseline serum EPO levels were measured, and both groups were followed up for a mean period of 1 yr with regular assessment of transfusion requirements, blood pressure, ferritin, liver and renal functions, hemoglobin, and HbF. Quality of life (QoL) was assessed at the start and end of the study. RESULTS Transfusion frequency and index were significantly decreased, while QoL was increased in group A compared with group B where 85% of patients showed improvement on combined therapy compared with 50% of patients on HU. Hemoglobin and HbF were significantly increased in both TI groups; however, this was more evident in group A than in group B. Also, 37.5% of patients in group A became transfusion-independent compared with 15% in group B. EPO levels were negatively related to increments of hemoglobin and HbF. Splenectomized patients and those with initial HbF% >40% had the best response to combined therapy. No serious adverse events necessitating discontinuation of therapy in both groups. CONCLUSIONS HU was effective in management of TI; however, combination with rHuEPO gave a superior therapeutic effect resulting in the best clinical and hematological responses without adverse events.
Collapse
Affiliation(s)
- Mohsen S Elalfy
- Pediatrics Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | | | | | | | | |
Collapse
|
19
|
Abstract
It is common knowledge that thalassemic patients are under significant oxidative stress. Chronic hemolysis, frequent blood transfusion, and increased intestinal absorption of iron are the main factors that result in iron overload with its subsequent pathophysiologic complications. Iron overload frequently associates with the generation of redox-reactive labile iron, which in turn promotes the production of other reactive oxygen species (ROS). If not neutralized, uncontrolled production of ROS often leads to damage of various intra- and extracellular components such as DNA, proteins, lipids, and small antioxidant molecules among others. A number of endogenous and exogenous defense mechanisms can neutralize and counteract the damaging effects of labile iron and the reactive substances associated with it. Endogenous antioxidant enzymes, such as superoxide dismutase, catalase, glutathione peroxidase, and ferroxidase, may directly or sequentially terminate the activities of ROS. Nonenzymatic endogenous defense mechanisms include metal binding proteins (ceruloplasmin, haptoglobin, albumin, and others) and endogenously produced free radical scavengers (glutathione (GSH), ubiquinols, and uric acid). Exogenous agents that are known to function as antioxidants (vitamins C and E, selenium, and zinc) are mostly diet-derived. In this review, we explore recent findings related to various antioxidative mechanisms operative in thalassemic patients with special emphasis on protein antioxidants.
Collapse
Affiliation(s)
- Samir Awadallah
- Department of Medical Laboratory Sciences, College of Health Sciences, University of Sharjah, Sharjah, United Arab Emirates.
| |
Collapse
|