Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Cell Rep ; 42(7): 112794, 2023 07 25.
Article in English | MEDLINE | ID: mdl-37459233

ABSTRACT

Relapse of acute myeloid leukemia (AML) remains a significant concern due to persistent leukemia-initiating stem cells (LICs) that are typically not targeted by most existing therapies. Using a murine AML model, human AML cell lines, and patient samples, we show that AML LICs are sensitive to endogenous and exogenous cyclopentenone prostaglandin-J (CyPG), Δ12-PGJ2, and 15d-PGJ2, which are increased upon dietary selenium supplementation via the cyclooxygenase-hematopoietic PGD synthase pathway. CyPGs are endogenous ligands for peroxisome proliferator-activated receptor gamma and GPR44 (CRTH2; PTGDR2). Deletion of GPR44 in a mouse model of AML exacerbated the disease suggesting that GPR44 activation mediates selenium-mediated apoptosis of LICs. Transcriptomic analysis of GPR44-/- LICs indicated that GPR44 activation by CyPGs suppressed KRAS-mediated MAPK and PI3K/AKT/mTOR signaling pathways, to enhance apoptosis. Our studies show the role of GPR44, providing mechanistic underpinnings of the chemopreventive and chemotherapeutic properties of selenium and CyPGs in AML.


Subject(s)
Leukemia, Myeloid, Acute , Selenium , Humans , Mice , Animals , Phosphatidylinositol 3-Kinases , Signal Transduction , Cell Line
2.
Free Radic Biol Med ; 127: 165-171, 2018 11 01.
Article in English | MEDLINE | ID: mdl-29719207

ABSTRACT

Selenium (Se) is incorporated as the 21st amino acid selenocysteine (Sec) into the growing polypeptide chain of proteins involved in redox gatekeeper functions. Erythropoiesis presents a particular problem to redox regulation as the presence of iron, heme, and unpaired globin chains lead to high levels of free radical-mediated oxidative stress, which are detrimental to erythroid development and can lead to anemia. Under homeostatic conditions, bone marrow erythropoiesis produces sufficient erythrocytes to maintain homeostasis. In contrast, anemic stress induces an alternative pathway, stress erythropoiesis, which rapidly produces new erythrocytes at extramedullary sites, such as spleen, to alleviate anemia. Previous studies suggest that dietary Se protects erythrocytes from such oxidative damage and the absence of selenoproteins causes hemolysis of erythrocytes due to oxidative stress. Furthermore, Se deficiency or lack of selenoproteins severely impairs stress erythropoiesis exacerbating the anemia in rodent models and human patients. Interestingly, erythroid progenitors develop in close proximity with macrophages in structures referred to as erythroblastic islands (EBIs), where macrophage expression of selenoproteins appears to be critical for the expression of heme transporters to facilitate export of heme from macrophage stores to the developing erythroid cells. Here we review the role of Se and selenoproteins in the intrinsic development of erythroid cells in addition to their role in the development of the erythropoietic niche that supports the functional role of EBIs in erythroid expansion and maturation in the spleen during recovery from anemia.


Subject(s)
Erythropoiesis/physiology , Selenium/metabolism , Selenoproteins/metabolism , Anemia/metabolism , Animals , Cell Differentiation/physiology , Erythroblasts/metabolism , Humans , Macrophages/metabolism , Oxidation-Reduction , Oxidative Stress/physiology
3.
Blood ; 131(23): 2568-2580, 2018 06 07.
Article in English | MEDLINE | ID: mdl-29615406

ABSTRACT

Micronutrient selenium (Se) plays a key role in redox regulation through its incorporation into selenoproteins as the 21st amino acid selenocysteine (Sec). Because Se deficiency appears to be a cofactor in the anemia associated with chronic inflammatory diseases, we reasoned that selenoproteins may contribute to erythropoietic recovery from anemia, referred to as stress erythropoiesis. Here, we report that loss of selenoproteins through Se deficiency or by mutation of the Sec tRNA (tRNA[Sec]) gene (Trsp) severely impairs stress erythropoiesis at 2 stages. Early stress erythroid progenitors failed to expand and properly differentiate into burst-forming unit-erythroid cells , whereas late-stage erythroid progenitors exhibited a maturation defect that affected the transition of proerythroblasts to basophilic erythroblasts. These defects were, in part, a result of the loss of selenoprotein W (SelenoW), whose expression was reduced at both transcript and protein levels in Se-deficient erythroblasts. Mutation of SelenoW in the bone marrow cells significantly decreased the expansion of stress burst-forming unit-erythroid cell colonies, which recapitulated the phenotypes induced by Se deficiency or mutation of Trsp Similarly, mutation of SelenoW in murine erythroblast (G1E) cell line led to defects in terminal differentiation. In addition to the erythroid defects, the spleens of Se-deficient mice contained fewer red pulp macrophages and exhibited impaired development of erythroblastic island macrophages, which make up the niche supporting erythroblast development. Taken together, these data reveal a critical role of selenoproteins in the expansion and development of stress erythroid progenitors, as well as the erythroid niche during acute anemia recovery.


Subject(s)
Anemia/metabolism , Erythroid Precursor Cells/cytology , Erythropoiesis , Selenium/deficiency , Selenoproteins/metabolism , Anemia/genetics , Animals , Down-Regulation , Erythroblasts/cytology , Erythroblasts/metabolism , Erythroid Precursor Cells/metabolism , Mice, Inbred C57BL , Mutation , Selenium/metabolism , Selenoprotein W/genetics , Selenoprotein W/metabolism , Selenoproteins/genetics , Spleen/cytology , Spleen/metabolism
4.
Adv Cancer Res ; 136: 153-172, 2017.
Article in English | MEDLINE | ID: mdl-29054417

ABSTRACT

Cancer is a complex disease where cancer stem cells (CSCs) maintain unlimited replicative potential, but evade chemotherapy drugs through cellular quiescence. CSCs are able to give rise to bulk tumor cells that have the capability to override antiproliferative signals and evade apoptosis. Numerous pathways are dysregulated in tumor cells, where increased levels of prooxidant reactive oxygen and nitrogen species can lead to localized inflammation to exacerbate all three stages of tumorigenesis: initiation, progression, and metastasis. Modulation of cellular metabolism in tumor cells as well as immune cells in the tumor microenvironment (TME) can impact inflammatory networks. Altering these pathways can potentially serve as a portal for therapy. It is well known that selenium, through selenoproteins, modulates inflammatory pathways in addition to regulating redox homeostasis in cells. Therefore, selenium has the potential to impact the interaction between tumor cells, CSCs, and immune cells. In the sections later, we review the current status of knowledge regarding this interaction, with reference to leukemia stem cells, and the importance of selenium-dependent regulation of inflammation as a potential mechanism to affect the TME and tumor cell survival.


Subject(s)
Inflammation/drug therapy , Neoplasms/drug therapy , Neoplastic Stem Cells/drug effects , Selenium/pharmacology , Signal Transduction/drug effects , Animals , Apoptosis/drug effects , Carcinogenesis/drug effects , Carcinogenesis/metabolism , Carcinogenesis/pathology , Disease Progression , Humans , Inflammation/metabolism , Inflammation/pathology , Neoplasms/metabolism , Neoplasms/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Oxidation-Reduction , Reactive Oxygen Species/metabolism , Selenoproteins/metabolism , Tumor Microenvironment/drug effects
5.
Blood ; 129(13): 1802-1810, 2017 03 30.
Article in English | MEDLINE | ID: mdl-28115365

ABSTRACT

Supplementation with nontoxic doses of micronutrient selenium has been shown to alleviate chronic myelogenous leukemia (CML) via the elimination of leukemia stem cells (LSCs) in mice. This treatment provides a new and novel method for eliminating the LSCs that are otherwise not targeted by existing therapies. The antileukemic effect of selenium was dependent on the production of endogenous cyclopentenone prostaglandins (CyPGs), Δ-12 prostaglandin J2 (Δ12-PGJ2), and 15-deoxy-Δ12,14-prostaglandin J2 (15d-PGJ2). Here, we show that these endogenous CyPGs, produced by mice maintained on selenium-supplemented diets, alleviate the symptoms of CML through their ability to activate the nuclear hormone receptor, peroxisome proliferator activated receptor γ (PPARγ). GW9662, a potent PPARγ antagonist, blocked the antileukemic effect of selenium supplementation by significantly reducing CyPGs. This effect was mediated by an increase in 15-prostaglandin dehydrogenase (15-Pgdh) activity, which oxidizes and inactivates Δ12-PGJ2 and 15d-PGJ2 In contrast, treatment with the PPARγ agonist pioglitazone mimicked selenium supplementation. This treatment led to decreased 15-Pgdh activity and increased CyPG levels, which inhibited CML progression. Selenium-dependent activation of PPARγ mediated by endogenous CyPGs decreased Stat5 expression leading to the downregulation of Cited2, a master regulator of LSC quiescence. These studies suggest a potential role for selenium supplementation as an adjuvant therapy in CML.


Subject(s)
Leukemia/drug therapy , PPAR gamma/metabolism , Prostaglandin D2/analogs & derivatives , Selenium/therapeutic use , Animals , Antineoplastic Agents , Dietary Supplements , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Mice , PPAR gamma/agonists , PPAR gamma/antagonists & inhibitors , Prostaglandin D2/biosynthesis , Prostaglandin D2/physiology , Selenium/pharmacology
6.
Cancer Prev Res (Phila) ; 8(10): 989-99, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26290393

ABSTRACT

Current therapies for treatment of myeloid leukemia do not eliminate leukemia stem cells (LSC), leading to disease relapse. In this study, we supplemented mice with eicosapentaenoic acid (EPA, C20:5), a polyunsaturated omega-3 fatty acid, at pharmacologic levels, to examine whether the endogenous metabolite, cyclopentenone prostaglandin delta-12 PGJ3 (Δ(12)-PGJ3), was effective in targeting LSCs in experimental leukemia. EPA supplementation for 8 weeks resulted in enhanced endogenous production of Δ(12)-PGJ3 that was blocked by indomethacin, a cyclooxygenase (COX) inhibitor. Using a murine model of chronic myelogenous leukemia (CML) induced by bone marrow transplantation of BCR-ABL-expressing hematopoietic stem cells, mice supplemented with EPA showed a decrease in the LSC population, and reduced splenomegaly and leukocytosis, when compared with mice on an oleic acid diet. Supplementation of CML mice carrying the T315I mutation (in BCR-ABL) with EPA resulted in a similar effect. Indomethacin blocked the EPA effect and increased the severity of BCR-ABL-induced CML and decreased apoptosis. Δ(12)-PGJ3 rescued indomethacin-treated BCR-ABL mice and decreased LSCs. Inhibition of hematopoietic-prostaglandin D synthase (H-PGDS) by HQL-79 in EPA-supplemented CML mice also blocked the effect of EPA. In addition, EPA supplementation was effective in a murine model of acute myeloid leukemia. EPA-supplemented mice exhibited a decrease in leukemia burden and a decrease in the LSC colony-forming unit (LSC-CFU). The decrease in LSCs was confirmed through serial transplantation assays in all disease models. The results support a chemopreventive role for EPA in myeloid leukemia, which is dependent on the ability to efficiently convert EPA to endogenous COX-derived prostanoids, including Δ(12)-PGJ3.


Subject(s)
Anticarcinogenic Agents/pharmacology , Dietary Supplements , Eicosapentaenoic Acid/pharmacology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Neoplastic Stem Cells/drug effects , Animals , Chromatography, Liquid , Flow Cytometry , HEK293 Cells , Humans , In Situ Nick-End Labeling , Mice , Mice, Inbred C57BL , Tandem Mass Spectrometry , Transduction, Genetic , Xenograft Model Antitumor Assays
7.
Cancer Res ; 74(14): 3890-901, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24872387

ABSTRACT

Eradicating cancer stem-like cells (CSC) may be essential to fully eradicate cancer. Metabolic changes in CSC could hold a key to their targeting. Here, we report that the dietary micronutrient selenium can trigger apoptosis of CSC derived from chronic or acute myelogenous leukemias when administered at supraphysiologic but nontoxic doses. In leukemia CSC, selenium treatment activated ATM-p53-dependent apoptosis accompanied by increased intracellular levels of reactive oxygen species. Importantly, the same treatment did not trigger apoptosis in hematopoietic stem cells. Serial transplantation studies with BCR-ABL-expressing CSC revealed that the selenium status in mice was a key determinant of CSC survival. Selenium action relied upon the endogenous production of the cyclooxygenase-derived prostaglandins Δ(12)-PGJ2 and 15d-PGJ2. Accordingly, nonsteroidal anti-inflammatory drugs and NADPH oxidase inhibitors abrogated the ability of selenium to trigger apoptosis in leukemia CSC. Our results reveal how selenium-dependent modulation of arachidonic acid metabolism can be directed to trigger apoptosis of primary human and murine CSC in leukemia.


Subject(s)
Eicosanoids/metabolism , Leukemia/metabolism , Selenium/pharmacology , Animals , Apoptosis/drug effects , Arachidonic Acids/metabolism , Cell Transformation, Neoplastic/drug effects , Cytochrome P-450 Enzyme System/metabolism , Humans , Leukemia/pathology , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Selenium/administration & dosage , Signal Transduction/drug effects , Splenomegaly , Tumor Suppressor Protein p53/metabolism
8.
PLoS One ; 8(12): e80622, 2013.
Article in English | MEDLINE | ID: mdl-24312486

ABSTRACT

Previous studies have demonstrated the ability of an eicosapentaenoic acid (EPA)-derived endogenous cyclopentenone prostaglandin (CyPG) metabolite, Δ(12)-PGJ3, to selectively target leukemic stem cells, but not the normal hematopoietic stems cells, in in vitro and in vivo models of chronic myelogenous leukemia (CML). Here we evaluated the stability, bioavailability, and hypersensitivity of Δ(12)-PGJ3. The stability of Δ(12)-PGJ3 was evaluated under simulated conditions using artificial gastric and intestinal juice. The bioavailability of Δ(12)-PGJ3 in systemic circulation was demonstrated upon intraperitoneal injection into mice by LC-MS/MS. Δ(12)-PGJ3 being a downstream metabolite of PGD3 was tested in vitro using primary mouse bone marrow-derived mast cells (BMMCs) and in vivo mouse models for airway hypersensitivity. ZK118182, a synthetic PG analog with potent PGD2 receptor (DP)-agonist activity and a drug candidate in current clinical trials, was used for toxicological comparison. Δ(12)-PGJ3 was relatively more stable in simulated gastric juice than in simulated intestinal juice that followed first-order kinetics of degradation. Intraperitoneal injection into mice revealed that Δ(12)-PGJ3 was bioavailable and well absorbed into systemic circulation with a Cmax of 263 µg/L at 12 h. Treatment of BMMCs with ZK118182 for 12 h resulted in increased production of histamine, while Δ(12)-PGJ3 did not induce degranulation in BMMCs nor increase histamine. In addition, in vivo testing for hypersensitivity in mice showed that ZK118182 induces higher airways hyperresponsiveness when compared Δ(12)-PGJ3 and/or PBS control. Based on the stability studies, our data indicates that intraperitoneal route of administration of Δ(12)-PGJ3 was favorable than oral administration to achieve effective pharmacological levels in the plasma against leukemia. Δ(12)-PGJ3 failed to increase histamine and IL-4 in BMMCs, which is in agreement with reduced airway hyperresponsiveness in mice. In summary, our studies suggest Δ(12)-PGJ3 to be a promising bioactive metabolite for further evaluation as a potential drug candidate for treating CML.


Subject(s)
Antineoplastic Agents , Fatty Acids, Omega-3 , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Prostaglandins , Animals , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Biological Availability , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Cells, Cultured , Drug Hypersensitivity/metabolism , Drug Hypersensitivity/pathology , Drug Screening Assays, Antitumor/methods , Fatty Acids, Omega-3/adverse effects , Fatty Acids, Omega-3/pharmacokinetics , Fatty Acids, Omega-3/pharmacology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Mast Cells/metabolism , Mast Cells/pathology , Mice , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Prostaglandins/adverse effects , Prostaglandins/pharmacokinetics , Prostaglandins/pharmacology
9.
Blood ; 118(26): 6909-19, 2011 Dec 22.
Article in English | MEDLINE | ID: mdl-21967980

ABSTRACT

Targeting cancer stem cells is of paramount importance in successfully preventing cancer relapse. Recently, in silico screening of public gene-expression datasets identified cyclooxygenase-derived cyclopentenone prostaglandins (CyPGs) as likely agents to target malignant stem cells. We show here that Δ(12)-PGJ(3), a novel and naturally produced CyPG from the dietary fish-oil ω-3 polyunsaturated fatty acid eicosapentaenoic acid (EPA; 20:5) alleviates the development of leukemia in 2 well-studied murine models of leukemia. IP administration of Δ(12)-PGJ(3) to mice infected with Friend erythroleukemia virus or those expressing the chronic myelogenous leukemia oncoprotein BCR-ABL in the hematopoietic stem cell pool completely restored normal hematologic parameters, splenic histology, and enhanced survival. More importantly, Δ(12)-PGJ(3) selectively targeted leukemia stem cells (LSCs) for apoptosis in the spleen and BM. This treatment completely eradicated LSCs in vivo, as demonstrated by the inability of donor cells from treated mice to cause leukemia in secondary transplantations. Given the potency of ω-3 polyunsaturated fatty acid-derived CyPGs and the well-known refractoriness of LSCs to currently used clinical agents, Δ(12)-PGJ(3) may represent a new chemotherapeutic for leukemia that targets LSCs.


Subject(s)
Apoptosis/drug effects , Fatty Acids, Omega-3/pharmacology , Leukemia/drug therapy , Neoplastic Stem Cells/drug effects , Prostaglandins/pharmacology , Animals , Ataxia Telangiectasia Mutated Proteins , Blotting, Western , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , Cyclopentanes/chemistry , Cyclopentanes/metabolism , Cyclopentanes/pharmacology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dose-Response Relationship, Drug , Fatty Acids, Omega-3/chemistry , Fatty Acids, Omega-3/metabolism , Leukemia/metabolism , Leukemia/pathology , Leukemia, Erythroblastic, Acute/drug therapy , Leukemia, Erythroblastic, Acute/metabolism , Leukemia, Erythroblastic, Acute/pathology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Molecular Structure , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Prostaglandins/chemistry , Prostaglandins/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Splenomegaly/pathology , Splenomegaly/prevention & control , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
10.
J Biol Chem ; 286(31): 27471-82, 2011 Aug 05.
Article in English | MEDLINE | ID: mdl-21669866

ABSTRACT

The plasticity of macrophages is evident from their dual role in inflammation and resolution of inflammation that are accompanied by changes in the transcriptome and metabolome. Along these lines, we have previously demonstrated that the micronutrient selenium increases macrophage production of arachidonic acid (AA)-derived anti-inflammatory 15-deoxy-Δ(12,14)-prostaglandin J(2) (15d-PGJ(2)) and decreases the proinflammatory PGE(2). Here, we hypothesized that selenium modulated the metabolism of AA by a differential regulation of various prostaglandin (PG) synthases favoring the production of PGD(2) metabolites, Δ(12)-PGJ(2) and 15d-PGJ(2). A dose-dependent increase in the expression of hematopoietic-PGD(2) synthase (H-PGDS) by selenium and a corresponding increase in Δ(12)-PGJ(2) and 15d-PGJ(2) in RAW264.7 macrophages and primary bone marrow-derived macrophages was observed. Studies with organic non-bioavailable forms of selenium and the genetic manipulation of cellular selenium incorporation machinery indicated that selenoproteins were necessary for H-PGDS expression and 15d-PGJ(2) production. Treatment of selenium-deficient macrophages with rosiglitazone, a peroxisome proliferator-activated receptor γ ligand, up-regulated H-PGDS. Furthermore, electrophoretic mobility shift assays indicated the presence of an active peroxisome proliferator-activated receptor-response element in murine Hpgds promoter suggesting a positive feedback mechanism of H-PGDS expression. Alternatively, the expression of nuclear factor-κB-dependent thromboxane synthase and microsomal PGE(2) synthase was down-regulated by selenium. Using a Friend virus infection model of murine leukemia, the onset of leukemia was observed only in selenium-deficient and indomethacin-treated selenium-supplemented mice but not in the selenium-supplemented group or those treated with 15d-PGJ(2). These results suggest the importance of selenium in the shunting of AA metabolism toward the production of PGD(2) metabolites, which may have clinical implications.


Subject(s)
Intramolecular Oxidoreductases/metabolism , Lipocalins/metabolism , Macrophages/enzymology , PPAR gamma/metabolism , Selenoproteins/physiology , Up-Regulation/physiology , Animals , Base Sequence , Cell Line , Chromatography, Liquid , DNA Primers , Intramolecular Oxidoreductases/genetics , Lipocalins/genetics , Mass Spectrometry , Mice , Promoter Regions, Genetic
11.
Am J Physiol Gastrointest Liver Physiol ; 300(3): G470-6, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21193529

ABSTRACT

Dietary iron is particularly critical during periods of rapid growth such as in neonatal development. Human and rodent studies have indicated that iron deficiency or excess during this critical stage of development can have significant long- and short-term consequences. Since the requirement for iron changes during development, the availability of adequate iron is critical for the differentiation and maturation of individual organs participating in iron homeostasis. We have examined in rats the effects of dietary iron supplement following neonatal iron deficiency on tissue iron status in relation to erythropoietic ability during 16 wk of postweaning development. This physiological model indicates that postweaning iron-adequate diet following neonatal iron deficiency adversely affects erythroid differentiation in the bone marrow and promotes splenic erythropoiesis leading to splenomegaly and erythrocytosis. This altered physiology of iron homeostasis during postweaning development is also reflected in the inability to maintain liver and spleen iron concentrations and the altered expression of iron regulatory proteins in the liver. These studies provide critical insights into the consequences of neonatal iron deficiency and the dietary iron-induced cellular signals affecting iron homeostasis during early development.


Subject(s)
Anemia, Iron-Deficiency/blood , Bone Marrow/metabolism , Erythroid Precursor Cells/metabolism , Erythropoiesis , Iron Deficiencies , Iron, Dietary/blood , Liver/metabolism , Spleen/metabolism , Age Factors , Anemia, Iron-Deficiency/diet therapy , Anemia, Iron-Deficiency/pathology , Animals , Animals, Newborn , Bone Marrow/pathology , Erythropoietin/blood , Female , Growth Differentiation Factor 15/blood , Hematocrit , Hemoglobins/metabolism , Homeostasis , Iron, Dietary/administration & dosage , Iron, Dietary/adverse effects , Iron-Regulatory Proteins/genetics , Iron-Regulatory Proteins/metabolism , Male , Maternal Nutritional Physiological Phenomena , Polycythemia/blood , Polycythemia/etiology , Pregnancy , Prenatal Exposure Delayed Effects , Rats , Rats, Sprague-Dawley , Spleen/pathology , Splenomegaly , Transferrin/metabolism , Weaning
12.
Antioxid Redox Signal ; 14(8): 1403-12, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-20969477

ABSTRACT

Redox modulation by antioxidants, such as selenium (Se), has emerged as an important regulator of erythropoiesis. Using Se-deficient (0.04 ppm), Se-adequate (0.1 ppm), and Se-supplemented (0.4 ppm) C57/BL6 mice, we show that Se deficiency caused anemia, when compared to the Se-supplemented and Se-adequate groups. Increased denaturation of hemoglobin, methemoglobin, protein carbonyls, lipid peroxidation, Heinz bodies, and osmotic fragility of erythrocytes were observed in Se-deficient mice. Increased oxidative stress upregulated forkhead transcription factor (FoxO3a) and hypoxia-inducible factor-(HIF)1α in the spleen and kidney of Se-deficient murine as well as in the proerythroblast G1E cells cultured in Se-deficient media. A significant increase in the expression of erythropoietin, a downstream target of HIF1α, and expansion of stress erythroid progenitors (burst forming units-erythroid) were seen in the Se-deficient mice. Despite the increase in erythroid progenitors, lowered reticulocytes suggest a defective erythroid differentiation pathway. While Se deficiency led to increased nuclear levels of active FoxO3a, Se-adequate conditions reversed this effect and increased nuclear export by its binding partner, 14-3-3ßζ, that is under the redox control of selenoproteins. In summary, these results provide insight into the importance of adequate Se nutrition in regulating red cell homeostasis by mitigating oxidative stress-dependent modulation of FoxO3a and HIF1α to effect differentiation of erythroid progenitors.


Subject(s)
Antioxidants/pharmacology , Erythrocytes/drug effects , Erythropoiesis/drug effects , Selenium/pharmacology , Animals , Cells, Cultured , Dietary Supplements , Erythrocytes/metabolism , Forkhead Box Protein O3 , Forkhead Transcription Factors/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Mice, Inbred C57BL , Oxidation-Reduction , Oxidative Stress , Selenium/administration & dosage , Selenium/deficiency
SELECTION OF CITATIONS
SEARCH DETAIL