Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
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.
Cancer Rep (Hoboken) ; 6(11): e1863, 2023 11.
Article in English | MEDLINE | ID: mdl-37489647

ABSTRACT

BACKGROUND AND AIM: There is an increased risk of colon cancer associated with inflammatory bowel disease (IBD). Dietary fibers (DFs) naturally present in vegetables and whole grains offer numerous beneficial effects on intestinal health. However, the effects of refined DFs on intestinal health remain unclear. Therefore, we elucidated the impact of the refined DF inulin on colonic inflammation and tumorigenesis. METHODS: Four-week-old wild-type (WT) mice were fed diets containing insoluble DF cellulose (control) or refined DF inulin for 4 weeks. A subgroup of mice was then switched to drinking water containing dextran sulfate sodium (DSS, 1.4% wt/vol) for colitis induction. In another subgroup of mice, colitis-associated colorectal cancer (CRC) was initiated with three 7-day alternate cycles of DSS following an initial dose of mutagenic substance azoxymethane (AOM; 7.5 mg/kg body weight; i.p.). Post 7 weeks of AOM treatment, mice were euthanized and examined for CRC development. RESULTS: Mice consuming inulin-containing diet exhibited severe colitis upon DSS administration, as evidenced by more body weight loss, rectal bleeding, and increased colonic inflammation than the DSS-treated control group. Correspondingly, histological analysis revealed extensive disruption of colon architecture and massive infiltration of immune cells in the inulin-fed group. We next examined the effect of inulin on CRC development. Surprisingly, significant mortality (~50%) was observed in the inulin-fed but not in the control group during the DSS cycle. Consequently, the remaining inulin-fed mice, which completed the study exhibited extensive colon tumorigenesis. Immunohistochemical characterization showed comparatively high expression of the cell proliferation marker Ki67 and activation of the Wnt signaling in tumor sections obtained from the inulin-fed group. Gut microbiota and metabolite analysis revealed expansion of succinate producers and elevated cecal succinate in inulin-fed mice. Human colorectal carcinoma cells (HCT116) proliferated more rapidly when supplemented with succinate in an inflamed environment, suggesting that elevated luminal succinate may contribute to tumorigenesis. CONCLUSIONS: Our study uncovers that supplementation of diet with refined inulin induces abnormal succinate accumulation in the intestinal lumen, which in part contributes to promoting colon inflammation and tumorigenesis.


Subject(s)
Colitis , Colonic Neoplasms , Colorectal Neoplasms , Humans , Animals , Mice , Inulin , Succinic Acid , Dextran Sulfate/toxicity , Inflammation/complications , Inflammation/pathology , Colitis/complications , Colitis/metabolism , Colitis/pathology , Colonic Neoplasms/chemically induced , Colorectal Neoplasms/chemically induced , Carcinogenesis , Cell Transformation, Neoplastic
3.
J Clin Invest ; 133(13)2023 07 03.
Article in English | MEDLINE | ID: mdl-37395277

ABSTRACT

Some studies suggest that the trace element selenium protects against colorectal cancer (CRC). However, the contribution of selenoprotein P (SELENOP), a unique selenocysteine-containing protein, to sporadic colorectal carcinogenesis challenges this paradigm. SELENOP is predominately secreted by the liver but is also expressed in various cells of the small intestine and colon in mice and humans. In this issue of the JCI, Pilat et al. demonstrate that increased SELENOP expression promoted the progression of conventional adenomas to carcinoma. SELENOP functioned as a modulator of canonical WNT signaling activity through interactions with WNT3A and its coreceptor LDL receptor-related protein 5/6 (LRP5/6). Secreted SELENOP formed a concentration gradient along the gut crypt axis, which might amplify WNT signaling activity by binding to LRPL5/6. The mechanism for WNT control via SELENOP may affect colorectal tumorigenesis and provide therapeutic targets for CRC.


Subject(s)
Colorectal Neoplasms , Selenium , Humans , Mice , Animals , Selenoprotein P/genetics , Selenoprotein P/metabolism , Carcinogenesis/genetics , Carcinogenesis/metabolism , Liver/metabolism , Cell Transformation, Neoplastic/metabolism , Selenium/metabolism , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Low Density Lipoprotein Receptor-Related Protein-5/genetics , Low Density Lipoprotein Receptor-Related Protein-5/metabolism , Wnt3A Protein/genetics , Wnt3A Protein/metabolism
4.
Redox Biol ; 59: 102571, 2023 02.
Article in English | MEDLINE | ID: mdl-36516721

ABSTRACT

Macrophages play a pivotal role in mediating inflammation and subsequent resolution of inflammation. The availability of selenium as a micronutrient and the subsequent biosynthesis of selenoproteins, containing the 21st amino acid selenocysteine (Sec), are important for the physiological functions of macrophages. Selenoproteins regulate the redox tone in macrophages during inflammation, the early onset of which involves oxidative burst of reactive oxygen and nitrogen species. SELENOW is a highly expressed selenoprotein in bone marrow-derived macrophages (BMDMs). Beyond its described general role as a thiol and peroxide reductase and as an interacting partner for 14-3-3 proteins, its cellular functions, particularly in macrophages, remain largely unknown. In this study, we utilized Selenow knock-out (KO) murine bone marrow-derived macrophages (BMDMs) to address the role of SELENOW in inflammation following stimulation with bacterial endotoxin lipopolysaccharide (LPS). RNAseq-based temporal analyses of expression of selenoproteins and the Sec incorporation machinery genes suggested no major differences in the selenium utilization pathway in the Selenow KO BMDMs compared to their wild-type counterparts. However, selective enrichment of oxidative stress-related selenoproteins and increased ROS in Selenow-/- BMDMs indicated anomalies in redox homeostasis associated with hierarchical expression of selenoproteins. Selenow-/- BMDMs also exhibited reduced expression of arginase-1, a key enzyme associated with anti-inflammatory (M2) phenotype necessary to resolve inflammation, along with a significant decrease in efferocytosis of neutrophils that triggers pathways of resolution. Parallel targeted metabolomics analysis also confirmed an impairment in arginine metabolism in Selenow-/- BMDMs. Furthermore, Selenow-/- BMDMs lacked the ability to enhance characteristic glycolytic metabolism during inflammation. Instead, these macrophages atypically relied on oxidative phosphorylation for energy production when glucose was used as an energy source. These findings suggest that SELENOW expression in macrophages may have important implications on cellular redox processes and bioenergetics during inflammation and its resolution.


Subject(s)
Selenium , Selenoprotein W , Mice , Animals , Selenoprotein W/genetics , Selenoprotein W/metabolism , Selenium/metabolism , Selenoproteins/genetics , Selenoproteins/metabolism , Macrophages/metabolism , Oxidation-Reduction , Inflammation/genetics
5.
Biol Trace Elem Res ; 200(12): 4949-4954, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35028869

ABSTRACT

Asthma is characterized by reversible airway obstruction, increased bronchial hyper-responsiveness and chronic inflammation, as well as higher levels of oxidative stress mainly due to decreased antioxidant defenses. Our primary aim was to investigate the correlation of serum selenium (Se) levels with the severity of asthma across gender, age, family history, and prevalence from childhood. Selenium levels in blood samples in 103 asthmatic patients and 103 healthy individuals were evaluated. The obtained data indicated that the mean serum Se levels in asthma patients were found to be twofold lower as compared to the controls (p < 0.001). However, there were no significant differences in the asthmatic patients when gender and age were considered. Patients characterized by family history of asthma and inhaler usage had 8% and 7% lower serum Se concentrations, although the difference was only border significant (p = 0.05). Multiple regression analysis demonstrated a significant inverse association of inhaler usage (ß = - 0.226; p < 0.001) with serum Se levels even after adjustment for asthma severity (ß = - 0.644; p < 0.001). While this report clearly necessitates a more detailed study, it is plausible that Se deficiency leads to impaired immune response, and therefore, Se supplementation might modulate oxidative stress in the lung and could potentially alleviate asthma pathophysiology.


Subject(s)
Asthma , Selenium , Antioxidants/metabolism , Asthma/drug therapy , Asthma/epidemiology , Child , Humans , Lung/metabolism , Oxidative Stress
6.
Front Immunol ; 12: 701341, 2021.
Article in English | MEDLINE | ID: mdl-34777335

ABSTRACT

The essential micronutrient Selenium (Se) is co-translationally incorporated as selenocysteine into proteins. Selenoproteins contain one or more selenocysteines and are vital for optimum immunity. Interestingly, many pathogenic bacteria utilize Se for various biological processes suggesting that Se may play a role in bacterial pathogenesis. A previous study had speculated that Francisella tularensis, a facultative intracellular bacterium and the causative agent of tularemia, sequesters Se by upregulating Se-metabolism genes in type II alveolar epithelial cells. Therefore, we investigated the contribution of host vs. pathogen-associated selenoproteins in bacterial disease using F. tularensis as a model organism. We found that F. tularensis was devoid of any Se utilization traits, neither incorporated elemental Se, nor exhibited Se-dependent growth. However, 100% of Se-deficient mice (0.01 ppm Se), which express low levels of selenoproteins, succumbed to F. tularensis-live vaccine strain pulmonary challenge, whereas 50% of mice on Se-supplemented (0.4 ppm Se) and 25% of mice on Se-adequate (0.1 ppm Se) diet succumbed to infection. Median survival time for Se-deficient mice was 8 days post-infection while Se-supplemented and -adequate mice was 11.5 and >14 days post-infection, respectively. Se-deficient macrophages permitted significantly higher intracellular bacterial replication than Se-supplemented macrophages ex vivo, corroborating in vivo observations. Since Francisella replicates in alveolar macrophages during the acute phase of pneumonic infection, we hypothesized that macrophage-specific host selenoproteins may restrict replication and systemic spread of bacteria. F. tularensis infection led to an increased expression of several macrophage selenoproteins, suggesting their key role in limiting bacterial replication. Upon challenge with F. tularensis, mice lacking selenoproteins in macrophages (TrspM) displayed lower survival and increased bacterial burden in the lung and systemic tissues in comparison to WT littermate controls. Furthermore, macrophages from TrspM mice were unable to restrict bacterial replication ex vivo in comparison to macrophages from littermate controls. We herein describe a novel function of host macrophage-specific selenoproteins in restriction of intracellular bacterial replication. These data suggest that host selenoproteins may be considered as novel targets for modulating immune response to control a bacterial infection.


Subject(s)
Francisella tularensis/immunology , Host-Pathogen Interactions/immunology , Macrophages/immunology , Macrophages/metabolism , Selenoproteins/metabolism , Tularemia/etiology , Tularemia/metabolism , Animals , Disease Models, Animal , Disease Susceptibility , Francisella tularensis/genetics , Francisella tularensis/pathogenicity , Mice , Pneumonia/immunology , Pneumonia/metabolism , Pneumonia/microbiology , Pneumonia/pathology , Tularemia/mortality , Virulence/genetics , Virulence Factors/genetics
7.
J Biol Chem ; 296: 100410, 2021.
Article in English | MEDLINE | ID: mdl-33581115

ABSTRACT

Trace element selenium (Se) is incorporated as the 21st amino acid, selenocysteine, into selenoproteins through tRNA[Ser]Sec. Selenoproteins act as gatekeepers of redox homeostasis and modulate immune function to effect anti-inflammation and resolution. However, mechanistic underpinnings involving metabolic reprogramming during inflammation and resolution remain poorly understood. Bacterial endotoxin lipopolysaccharide (LPS) activation of murine bone marrow-derived macrophages cultured in the presence or absence of Se (as selenite) was used to examine temporal changes in the proteome and metabolome by multiplexed tandem mass tag-quantitative proteomics, metabolomics, and machine-learning approaches. Kinetic deltagram and clustering analysis indicated that addition of Se led to extensive reprogramming of cellular metabolism upon stimulation with LPS enhancing the pentose phosphate pathway, tricarboxylic acid cycle, and oxidative phosphorylation, to aid in the phenotypic transition toward alternatively activated macrophages, synonymous with resolution of inflammation. Remodeling of metabolic pathways and consequent metabolic adaptation toward proresolving phenotypes began with Se treatment at 0 h and became most prominent around 8 h after LPS stimulation that included succinate dehydrogenase complex, pyruvate kinase, and sedoheptulokinase. Se-dependent modulation of these pathways predisposed bone marrow-derived macrophages to preferentially increase oxidative phosphorylation to efficiently regulate inflammation and its timely resolution. The use of macrophages lacking selenoproteins indicated that all three metabolic nodes were sensitive to selenoproteome expression. Furthermore, inhibition of succinate dehydrogenase complex with dimethylmalonate affected the proresolving effects of Se by increasing the resolution interval in a murine peritonitis model. In summary, our studies provide novel insights into the role of cellular Se via metabolic reprograming to facilitate anti-inflammation and proresolution.


Subject(s)
Selenium/metabolism , Selenoproteins/metabolism , Animals , Disease Susceptibility/metabolism , Inflammation/metabolism , Inflammation/physiopathology , Lipopolysaccharides/metabolism , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Peritonitis/drug therapy , Peritonitis/immunology , Proteome/metabolism , Proteomics , Selenium/pharmacology , Selenoproteins/genetics , Selenoproteins/physiology , Succinate Dehydrogenase/metabolism
8.
Front Nutr ; 7: 96, 2020.
Article in English | MEDLINE | ID: mdl-32775340

ABSTRACT

Enteropathogenic Escherichia coli (EPEC) leads to adverse colonic inflammation associated with poor resolution of inflammation and loss of epithelial integrity. Micronutrient trace element selenium (Se) is incorporated into selenoproteins as the 21st amino acid, selenocysteine (Sec). Previous studies have shown that such an incorporation of Sec into the selenoproteome is key for the anti-inflammatory functions of Se in macrophages and other immune cells. An intriguing mechanism underlying the anti-inflammatory and pro-resolving effects of Se stems from the ability of selenoproteins to skew arachidonic acid metabolism from pro-inflammatory mediators, prostaglandin E2 (PGE2) toward anti-inflammatory mediators derived from PGD2, such as 15-deoxy-Δ12, 14- prostaglandin J2 (15d-PGJ2), via eicosanoid class switching of bioactive lipids. The impact of Se and such an eicosanoid-class switching mechanism was tested in an enteric infection model of gut inflammation by C. rodentium, a murine equivalent of EPEC. C57BL/6 mice deficient in Se (Se-D) experienced higher mortality when compared to those on Se adequate (0.08 ppm Se) and Se supplemented (0.4 ppm Se) diets following infection. Decreased survival was associated with decreased group 3 innate lymphoid cells (ILC3s) and T helper 17 (Th17) cells in colonic lamina propria of Se-D mice along with deceased expression of epithelial barrier protein Zo-1. Inhibition of metabolic inactivation of PGE2 by 15-prostaglandin dehydrogenase blocked the Se-dependent increase in ILC3 and Th17 cells in addition to reducing epithelial barrier integrity, as seen by increased systemic levels of FITC-dextran following oral administration; while 15d-PGJ2 administration in Se-D mice alleviated the effects by increasing ILC3 and Th17 cells. Mice lacking selenoproteins in monocyte/macrophages via the conditional deletion of the tRNA[Sec] showed increased mortality post infection. Our studies indicate a crucial role for dietary Se in the protection against inflammation following enteric infection via immune mechanisms involving epithelial barrier integrity.

9.
Food Funct ; 10(5): 2839-2846, 2019 May 22.
Article in English | MEDLINE | ID: mdl-31062009

ABSTRACT

Cell signaling is necessary for the organs to co-ordinate with the whole body and it includes response to external stimuli, inflammation, hormonal secretions and other various metabolic functions. In the present study, we have focused on the inflammatory signals modulated by the reactive oxygen and nitrogen species (RONS). Under homeostatic conditions, these species turn on the COX-1-dependent arachidonic acid (AA) pathway towards the release of anti-inflammatory enzymes. However, the excess release of these ions induces negative effects in the form of inflammation by turning on the COX-2-dependent AA pathway to release pro-inflammatory enzymes. In the present study, we observed the shunting of the COX-2-dependent AA pathway towards the release of anti-inflammatory enzymes with the supplementation of organic dietary selenium in the form of seleniferous maize extracts. We observed that 500 nM selenium concentration in Se-maize extracts downregulated the COX-2 and mPGES-1 expressions by 3.8- and 3.2-fold and upregulated the GPx-1 and H-PGDS expressions by 5.0- and 5.4-fold, respectively. To facilitate more availability of Se from the dietary matrices, Se-maize extracts were incubated with rMETase. It was observed that the enzyme-treated cells increased the downregulation of COX-2 and mPGES-1 expressions by 24.8- and 21.0-fold and the upregulation of GPx-1 and H-PGDS expressions by 13.2- and 16.5-fold, respectively.


Subject(s)
Macrophages/drug effects , Prostaglandins/metabolism , Selenium/pharmacology , Zea mays/chemistry , Animals , Arachidonic Acid/metabolism , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Gene Expression/drug effects , Lipopolysaccharides/adverse effects , Macrophages/metabolism , Mice , Prostaglandin-E Synthases/genetics , Prostaglandin-E Synthases/metabolism , RAW 264.7 Cells , Selenium/analysis
10.
J Am Soc Mass Spectrom ; 30(7): 1276-1283, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30972724

ABSTRACT

Selenium (Se) functions as a cellular redox gatekeeper through its incorporation into proteins as the 21st amino acid, selenocysteine (Sec). Supplementation of macrophages with exogenous Se (as sodium selenite) downregulates inflammation and intracellular oxidative stress by effectively restoring redox homeostasis upon challenge with bacterial endotoxin lipopolysaccharide (LPS). Here, we examined the use of a standard Tandem Mass Tag (TMT)-labeling mass spectrometry-based proteomic workflow to quantitate and examine temporal regulation of selenoproteins in such inflamed cells. Se-deficient murine primary bone marrow-derived macrophages (BMDMs) exposed to LPS in the presence or absence of selenite treatment for various time periods (0-20 h) were used to analyze the selenoproteome expression using isobaric labeling and shotgun proteomic workflow. To overcome the challenge of identification of Sec peptides, we used the identification of non-Sec containing peptides downstream of Sec as a reliable evidence of ribosome readthrough indicating efficient decoding of Sec codon. Results indicated a temporal regulation of the selenoproteome with a general increase in their expression in inflamed cells in a Se-dependent manner. Selenow, Gpx1, Msrb1, and Selenom were highly upregulated upon stimulation with LPS when compared to other selenoproteins. Interestingly, Selenow appeared to be one amongst the highly regulated selenoproteins in macrophages that was previously thought to be mainly restricted to myocytes. Collectively, TMT-labeling method of non-Sec peptides offers a reliable method to quantitate and study temporal regulation of selenoproteins; however, further optimization to include Sec-peptides could make this strategy more robust and sensitive compared to other semi-quantitative or qualitative methods. Graphical Abstract.


Subject(s)
Macrophages/chemistry , Selenoproteins/analysis , Amino Acid Sequence , Animals , Inflammation/immunology , Lipopolysaccharides/immunology , Macrophages/immunology , Male , Mice, Inbred C57BL , Proteomics/methods , Selenoproteins/immunology , Tandem Mass Spectrometry/methods
11.
Crit Rev Biochem Mol Biol ; 54(6): 484-516, 2019 12.
Article in English | MEDLINE | ID: mdl-31996052

ABSTRACT

Selenium (Se) is an essential trace element that functions in the form of the 21st amino acid, selenocysteine (Sec) in a defined set of proteins. Se deficiency is associated with pathological conditions in humans and animals, where incorporation of Sec into selenoproteins is reduced along with their expression and catalytic activity. Supplementation of Se-deficient population with Se has shown health benefits suggesting the importance of Se in physiology. An interesting paradigm to explain, in part, the health benefits of Se stems from the observations that selenoprotein-dependent modulation of inflammation and efficient resolution of inflammation relies on mechanisms involving a group of bioactive lipid mediators, prostanoids, which orchestrate a concerted action toward maintenance and restoration of homeostatic immune responses. Such an effect involves the interaction of various immune cells with these lipid mediators where cellular redox gatekeeper functions of selenoproteins further aid in not only dampening inflammation, but also initiating an effective and active resolution process. Here we have summarized the current literature on the multifaceted roles of Se/selenoproteins in the regulation of these bioactive lipid mediators and their immunomodulatory effects.


Subject(s)
Prostaglandins/immunology , Prostaglandins/metabolism , Selenium/administration & dosage , Selenoproteins/immunology , Selenoproteins/metabolism , Animals , Humans , Inflammation/drug therapy , Inflammation/immunology , Inflammation/metabolism , Lipid Metabolism , Randomized Controlled Trials as Topic , Selenium/immunology , Selenium/metabolism , Signal Transduction
12.
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
13.
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
14.
Food Funct ; 9(4): 1998-2004, 2018 Apr 25.
Article in English | MEDLINE | ID: mdl-29644347

ABSTRACT

Selenium (Se) is an essential dietary supplement that resolves inflammatory responses and offers antioxidant cytoprotection. In this study, we present the data on the cytoprotective effect of Se-rich mustard protein isolated from mustard cultivated in seleniferous soils in Punjab, India. The concentrations of total Se in mustard seed, oil-free mustard cake, and mustard protein were 110.0 ± 3.04, 143.0 ± 5.18, and 582.3 ± 6.23 µg g-1, respectively. The cytoprotective effect of Se-rich mustard protein was studied on tert-butyl hydroperoxide (TBHP)-induced cytotoxicity in a mouse melanoma cell line (B16-F10). When compared with TBHP treated cells (where no viable cells were found), Se-rich protein made bioaccessible through simulated gastrointestinal digestion protected melanoma cells from cytotoxicity with decreased levels of oxidative stress resulting in 73% cell viability. Such an effect was associated with a significant increase in glutathione peroxidase activity as a function of bioaccessible Se and its response towards cytoprotection.


Subject(s)
Food , Melanoma/prevention & control , Models, Biological , Mustard Plant/chemistry , Oxidative Stress , Seeds/chemistry , Selenium/administration & dosage , Animals , Cell Line, Tumor , Cell Survival/drug effects , Crops, Agricultural/chemistry , Crops, Agricultural/growth & development , Cytoprotection , Digestion , Glutathione Peroxidase/chemistry , Glutathione Peroxidase/metabolism , Humans , India , Melanoma/immunology , Melanoma/metabolism , Melanoma/pathology , Mice , Mustard Plant/growth & development , Nutritive Value , Oxidative Stress/drug effects , Plant Proteins, Dietary/metabolism , Seeds/growth & development , Selenium/analysis , Selenium/metabolism , tert-Butylhydroperoxide/toxicity
15.
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
16.
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
17.
Food Agric Immunol ; 28(6): 1374-1392, 2017.
Article in English | MEDLINE | ID: mdl-29563666

ABSTRACT

The accessibility of selenium from naturally enriched sources such as cereals crops can potentially be used as selenium supplements to support nutritional requirements. Dietary selenium supplementation, as Se-rich wheat extracts, on RAW264.7 macrophage cells enhanced the antioxidant capacity via augmentation of cellular selenoprotein glutathione peroxidase 1 (GPx-1) expression in the absence or presence of lipopolysaccharide (LPS) treatment. Cells were supplemented with Se in the form of sodium selenite (SS), seleniferous wheat extract (SeW) and seleniferous wheat extract with rMETase treatment (SeW+rMET) at three different concentrations. Cells supplemented with SS and SeW+rMET showed increase in GPx-1 expression as compared to SeW treated cells. SeW+rMET, further, down-regulated the LPS-induced expression of cyclooxygenase-2, microsomal PGE synthase-1 and inducible nitric oxide synthase w.r.t. Se-deficient cells, while the expression of hematopoietic PGD synthase was upregulated. This demonstrates SeSup effectively modulates the expression inflammatory responses, indicating the potential benefits of dietary selenium supplementation.

19.
J Biol Chem ; 291(6): 2787-98, 2016 Feb 05.
Article in English | MEDLINE | ID: mdl-26644468

ABSTRACT

The plasticity of macrophages is evident in helminthic parasite infections, providing protection from inflammation. Previously we demonstrated that the micronutrient selenium induces a phenotypic switch in macrophage activation from a classically activated (pro-inflammatory; M1/CAM) toward an alternatively activated (anti-inflammatory; M2/AAM) phenotype, where cyclooxygenase (COX)-dependent cyclopentenone prostaglandin J2 (15d-PGJ2) plays a key role. Here, we hypothesize that dietary selenium modulates macrophage polarization toward an AAM phenotype to assist in the increasing clearance of adult Nippostrongylus brasiliensis, a gastrointestinal nematode parasite. Mice on a selenium-adequate (0.08 ppm) diet significantly augmented intestinal AAM presence while decreasing adult worms and fecal egg production when compared with infection of mice on selenium-deficient (<0.01 ppm) diet. Further increase in dietary selenium to supraphysiological levels (0.4 ppm) had very little or no impact on worm expulsion. Normal adult worm clearance and enhanced AAM marker expression were observed in the selenium-supplemented Trsp(fl/fl)Cre(WT) mice that express selenoproteins driven by tRNA(Sec) (Trsp), whereas N. brasiliensis-infected Trsp(fl/fl)Cre(LysM) selenium-supplemented mice showed a decreased clearance, with lowered intestinal expression of several AAM markers. Inhibition of the COX pathway with indomethacin resulted in delayed worm expulsion in selenium-adequate mice. This was rescued with 15d-PGJ2, which partially recapitulated the effect of selenium supplementation on fecal egg output in addition to increasing markers of AAMs in the small intestine. Antagonism of PPARγ blocked the effect of selenium. These results suggest that optimal expression of selenoproteins and selenium-dependent production of COX-derived endogenous prostanoids, such as Δ(12)-PGJ2 and 15d-PGJ2, may regulate AAM activation to enhance anti-helminthic parasite responses.


Subject(s)
Gene Expression Regulation/immunology , Macrophage Activation , Macrophages/immunology , Nippostrongylus/immunology , Selenoproteins/immunology , Strongylida Infections/immunology , Animals , Dietary Supplements , Macrophages/parasitology , Macrophages/pathology , Male , Mice , Prostaglandin D2/analogs & derivatives , Prostaglandin D2/immunology , Selenium/pharmacology , Strongylida Infections/drug therapy
20.
Biochemistry ; 54(33): 5121-4, 2015 Aug 25.
Article in English | MEDLINE | ID: mdl-26252619

ABSTRACT

Tetrathionate, a polythionate oxidation product of microbial hydrogen sulfide and reactive oxygen species from immune cells in the gut, serves as a terminal electron acceptor to confer a growth advantage for Salmonella and other enterobacteria. Here we show that the rat liver selenoenzyme thioredoxin reductase (Txnrd1, TR1) efficiently reduces tetrathionate in vitro. Furthermore, lysates of selenium-supplemented murine macrophages also displayed activity toward tetrathionate, while cells lacking TR1 were unable to reduce tetrathionate. These studies suggest that upregulation of TR1 expression, via selenium supplementation, may modulate the gut microbiome, particularly during inflammation, by regulating the levels of tetrathionate.


Subject(s)
Tetrathionic Acid/metabolism , Thioredoxin-Disulfide Reductase/metabolism , Animals , Liver/enzymology , Oxidation-Reduction , Rats , Selenium/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL