PMID-sentid Pub_year Sent_text comp_official_name comp_offsetprotein_name organism prot_offset 25727755-1 2015 Transferrin receptor (TfR1) and divalent metal transporter 1 (DMT1) are important proteins for cellular iron uptake, and both are regulated transcriptionally through the binding of hypoxia-inducible factor 1 (HIF-1) to hypoxia-responsive elements (HREs) under hypoxic conditions. Iron 104-108 solute carrier family 11 member 2 Homo sapiens 32-60 27669335-7 2016 Levels of mRNA for Tfr1 and Slc11a2, iron-responsive genes involved in iron uptake, were significantly elevated in DFO-treated cultures at 11DIV and 18DIV, indicating a degree of neuronal ID similar to that seen in rodent ID models. Iron 37-41 solute carrier family 11 member 2 Homo sapiens 28-35 27669335-7 2016 Levels of mRNA for Tfr1 and Slc11a2, iron-responsive genes involved in iron uptake, were significantly elevated in DFO-treated cultures at 11DIV and 18DIV, indicating a degree of neuronal ID similar to that seen in rodent ID models. Iron 71-75 solute carrier family 11 member 2 Homo sapiens 28-35 26289753-0 2015 Iron-induced reactive oxygen species mediate transporter DMT1 endocytosis and iron uptake in intestinal epithelial cells. Iron 0-4 solute carrier family 11 member 2 Homo sapiens 57-61 26289753-1 2015 Recent evidence shows that iron induces the endocytosis of the iron transporter dimetal transporter 1 (DMT1) during intestinal absorption. Iron 27-31 solute carrier family 11 member 2 Homo sapiens 103-107 26289753-2 2015 We, and others, have proposed that iron-induced DMT1 internalization underlies the mucosal block phenomena, a regulatory response that downregulates intestinal iron uptake after a large oral dose of iron. Iron 35-39 solute carrier family 11 member 2 Homo sapiens 48-52 26289753-2 2015 We, and others, have proposed that iron-induced DMT1 internalization underlies the mucosal block phenomena, a regulatory response that downregulates intestinal iron uptake after a large oral dose of iron. Iron 160-164 solute carrier family 11 member 2 Homo sapiens 48-52 26289753-2 2015 We, and others, have proposed that iron-induced DMT1 internalization underlies the mucosal block phenomena, a regulatory response that downregulates intestinal iron uptake after a large oral dose of iron. Iron 160-164 solute carrier family 11 member 2 Homo sapiens 48-52 26289753-4 2015 By means of selective surface protein biotinylation of polarized Caco-2 cells, we determined the kinetics of DMT1 internalization from the apical membrane after an iron challenge. Iron 164-168 solute carrier family 11 member 2 Homo sapiens 109-113 26289753-5 2015 The initial decrease in DMT1 levels in the apical membrane induced by iron was followed at later times by increased levels of DMT1. Iron 70-74 solute carrier family 11 member 2 Homo sapiens 24-28 26289753-9 2015 The decrease of DMT1 levels at the apical membrane induced by iron was associated with decreased iron uptake rates. Iron 62-66 solute carrier family 11 member 2 Homo sapiens 16-20 26289753-9 2015 The decrease of DMT1 levels at the apical membrane induced by iron was associated with decreased iron uptake rates. Iron 97-101 solute carrier family 11 member 2 Homo sapiens 16-20 26289753-11 2015 The model qualitatively captures the experimental observations and accurately describes the effect of iron, NAC, and DMSO on the apical distribution of DMT1. Iron 102-106 solute carrier family 11 member 2 Homo sapiens 152-156 26289753-12 2015 Taken together, our data suggest that iron uptake induces the production of ROS, which modify DMT1 endocytic cycling, thus changing the iron transport activity at the apical membrane. Iron 38-42 solute carrier family 11 member 2 Homo sapiens 94-98 26289753-12 2015 Taken together, our data suggest that iron uptake induces the production of ROS, which modify DMT1 endocytic cycling, thus changing the iron transport activity at the apical membrane. Iron 136-140 solute carrier family 11 member 2 Homo sapiens 94-98 26208637-0 2015 Dictyostelium Nramp1, which is structurally and functionally similar to mammalian DMT1 transporter, mediates phagosomal iron efflux. Iron 120-124 solute carrier family 11 member 2 Homo sapiens 82-86 26208637-4 2015 Nramp2 is located exclusively in the contractile vacuole membrane and controls, synergistically with Nramp1, iron homeostasis. Iron 109-113 solute carrier family 11 member 2 Homo sapiens 0-6 26878799-0 2016 Iron mitigates DMT1-mediated manganese cytotoxicity via the ASK1-JNK signaling axis: Implications of iron supplementation for manganese toxicity. Iron 0-4 solute carrier family 11 member 2 Homo sapiens 15-19 26878799-0 2016 Iron mitigates DMT1-mediated manganese cytotoxicity via the ASK1-JNK signaling axis: Implications of iron supplementation for manganese toxicity. Iron 101-105 solute carrier family 11 member 2 Homo sapiens 15-19 26551072-2 2016 The divalent metal transporter 1 (DMT1) is a major metal transporter involved in the absorption and metabolism of several essential metals like iron and manganese. Iron 144-148 solute carrier family 11 member 2 Homo sapiens 4-32 26551072-2 2016 The divalent metal transporter 1 (DMT1) is a major metal transporter involved in the absorption and metabolism of several essential metals like iron and manganese. Iron 144-148 solute carrier family 11 member 2 Homo sapiens 34-38 26572590-1 2015 Divalent metal-ion transporter 1 (DMT1) has been found to play an important role in the iron metabolism and hemogenesis. Iron 88-92 solute carrier family 11 member 2 Homo sapiens 0-32 26572590-1 2015 Divalent metal-ion transporter 1 (DMT1) has been found to play an important role in the iron metabolism and hemogenesis. Iron 88-92 solute carrier family 11 member 2 Homo sapiens 34-38 26572590-7 2015 Taken together, our studies demonstrate that decreased expression of DMT1 in intestinal mucosa leads to compromised absorption and transportation of iron and that blockade of TNF could rescue anemia and promote DMT1 expression in gut mucosa. Iron 149-153 solute carrier family 11 member 2 Homo sapiens 69-73 25753988-0 2015 Increased duodenal iron absorption through up-regulation of divalent metal transporter 1 from enhancement of iron regulatory protein 1 activity in patients with nonalcoholic steatohepatitis. Iron 19-23 solute carrier family 11 member 2 Homo sapiens 60-88 25753988-12 2015 CONCLUSION: In spite of elevation of serum hepcidin, iron absorption from the GI tract increased through up-regulation of DMT1 by IRP1 activation by humoral factor(s) in sera of patients with NASH. Iron 53-57 solute carrier family 11 member 2 Homo sapiens 122-126 25727755-1 2015 Transferrin receptor (TfR1) and divalent metal transporter 1 (DMT1) are important proteins for cellular iron uptake, and both are regulated transcriptionally through the binding of hypoxia-inducible factor 1 (HIF-1) to hypoxia-responsive elements (HREs) under hypoxic conditions. Iron 104-108 solute carrier family 11 member 2 Homo sapiens 62-66 25727755-3 2015 In iron-deficient cells, IRP1-IRE interactions stabilize TfR1 and DMT1 mRNAs, enhancing iron uptake. Iron 3-7 solute carrier family 11 member 2 Homo sapiens 66-70 25727755-3 2015 In iron-deficient cells, IRP1-IRE interactions stabilize TfR1 and DMT1 mRNAs, enhancing iron uptake. Iron 88-92 solute carrier family 11 member 2 Homo sapiens 66-70 25817364-5 2015 Divalent metal transporter 1 (DMT1) is one protein responsible for iron transport. Iron 67-71 solute carrier family 11 member 2 Homo sapiens 0-28 26106291-0 2015 Divalent metal transporter 1 (DMT1) in the brain: implications for a role in iron transport at the blood-brain barrier, and neuronal and glial pathology. Iron 77-81 solute carrier family 11 member 2 Homo sapiens 0-28 26106291-0 2015 Divalent metal transporter 1 (DMT1) in the brain: implications for a role in iron transport at the blood-brain barrier, and neuronal and glial pathology. Iron 77-81 solute carrier family 11 member 2 Homo sapiens 30-34 26106291-2 2015 In this review, we focus on iron transport in the brain and the role of the divalent metal transporter 1 (DMT1) vital for iron uptake in most cells. Iron 122-126 solute carrier family 11 member 2 Homo sapiens 76-104 26106291-2 2015 In this review, we focus on iron transport in the brain and the role of the divalent metal transporter 1 (DMT1) vital for iron uptake in most cells. Iron 122-126 solute carrier family 11 member 2 Homo sapiens 106-110 26106291-3 2015 DMT1 locates to cellular membranes and endosomal membranes, where it is a key player in non-transferrin bound iron uptake and transferrin-bound iron uptake, respectively. Iron 110-114 solute carrier family 11 member 2 Homo sapiens 0-4 26106291-3 2015 DMT1 locates to cellular membranes and endosomal membranes, where it is a key player in non-transferrin bound iron uptake and transferrin-bound iron uptake, respectively. Iron 144-148 solute carrier family 11 member 2 Homo sapiens 0-4 26106291-4 2015 Four isoforms of DMT1 exist, and their respective characteristics involve a complex cell-specific regulatory machinery all controlling iron transport across these membranes. Iron 135-139 solute carrier family 11 member 2 Homo sapiens 17-21 26106291-9 2015 This supports existing evidence that iron uptake at the BBB occurs by means of transferrin-receptor mediated endocytosis followed by detachment of iron from transferrin inside the acidic compartment of the endosome and DMT1-mediated pumping iron into the cytosol. Iron 37-41 solute carrier family 11 member 2 Homo sapiens 219-223 25843360-5 2015 Following exposure to VOSO4, there was an increase (336+-73%) in RNA for divalent metal transporter 1 (DMT1), a major iron importer. Iron 118-122 solute carrier family 11 member 2 Homo sapiens 73-101 25843360-5 2015 Following exposure to VOSO4, there was an increase (336+-73%) in RNA for divalent metal transporter 1 (DMT1), a major iron importer. Iron 118-122 solute carrier family 11 member 2 Homo sapiens 103-107 26047847-1 2015 Divalent metal transporter-1 (SLC11A2/DMT1) uses the H(+) electrochemical gradient as the driving force to transport divalent metal ions such as Fe(2+), Mn(2+) and others metals into mammalian cells. Iron 145-147 solute carrier family 11 member 2 Homo sapiens 30-37 26047847-1 2015 Divalent metal transporter-1 (SLC11A2/DMT1) uses the H(+) electrochemical gradient as the driving force to transport divalent metal ions such as Fe(2+), Mn(2+) and others metals into mammalian cells. Iron 145-147 solute carrier family 11 member 2 Homo sapiens 38-42 25817364-5 2015 Divalent metal transporter 1 (DMT1) is one protein responsible for iron transport. Iron 67-71 solute carrier family 11 member 2 Homo sapiens 30-34 26756421-6 2015 Expression of the iron-related proteins ferritin, DMT1, and FPN1 was elevated in lung tissue from the six asbestosis patients relative to controls. Iron 18-22 solute carrier family 11 member 2 Homo sapiens 50-54 25318588-12 2015 The increased duodenal DcytB, DMT1, and FPN1 expression can enhance intestinal iron absorption to meet the high iron requirements in infants. Iron 79-83 solute carrier family 11 member 2 Homo sapiens 30-34 25483413-0 2015 Evaluation of the effect of divalent metal transporter 1 gene polymorphism on blood iron, lead and cadmium levels. Iron 84-88 solute carrier family 11 member 2 Homo sapiens 28-56 25483413-1 2015 Divalent metal transporter 1 (DMT1), a member of the proton-coupled metal ion transporter family, mediates transport of ferrous iron from the lumen of the intestine into the enterocyte and export of iron from endocytic vesicles. Iron 128-132 solute carrier family 11 member 2 Homo sapiens 0-28 25483413-1 2015 Divalent metal transporter 1 (DMT1), a member of the proton-coupled metal ion transporter family, mediates transport of ferrous iron from the lumen of the intestine into the enterocyte and export of iron from endocytic vesicles. Iron 128-132 solute carrier family 11 member 2 Homo sapiens 30-34 25483413-1 2015 Divalent metal transporter 1 (DMT1), a member of the proton-coupled metal ion transporter family, mediates transport of ferrous iron from the lumen of the intestine into the enterocyte and export of iron from endocytic vesicles. Iron 199-203 solute carrier family 11 member 2 Homo sapiens 0-28 25483413-1 2015 Divalent metal transporter 1 (DMT1), a member of the proton-coupled metal ion transporter family, mediates transport of ferrous iron from the lumen of the intestine into the enterocyte and export of iron from endocytic vesicles. Iron 199-203 solute carrier family 11 member 2 Homo sapiens 30-34 25483413-5 2015 To study the possible association of DMT1 gene with the blood levels of some divalent cations such as iron, lead and cadmium, a single nucleotide polymorphism (SNP) (IVS4+44C/A) in DMT1 gene was investigated in 486 unrelated and healthy individuals in a Turkish population by method of polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP). Iron 102-106 solute carrier family 11 member 2 Homo sapiens 37-41 25483413-9 2015 Highly statistically significant associations were detected between IVS4+44 C/A polymorphism in the DMT1 gene and iron and lead levels (p=0.001 and p=0.036, respectively), but no association was found with cadmium level (p=0.344). Iron 114-118 solute carrier family 11 member 2 Homo sapiens 100-104 25483413-10 2015 This study suggested that DMT1 IVS4+44 C/A polymorphism is associated with inter-individual variations in blood iron, lead and cadmium levels. Iron 112-116 solute carrier family 11 member 2 Homo sapiens 26-30 25467637-4 2015 Further study showed that the decrease of Ndfip1 occurred earlier than the increase of DMT1 with iron-responsive element (DMT1 + IRE) in 6-OHDA-treated MES23.5 cells, indicating that the decrease of Ndfip1 might be involved in the increase of DMT1 + IRE. Iron 97-101 solute carrier family 11 member 2 Homo sapiens 87-91 25467637-4 2015 Further study showed that the decrease of Ndfip1 occurred earlier than the increase of DMT1 with iron-responsive element (DMT1 + IRE) in 6-OHDA-treated MES23.5 cells, indicating that the decrease of Ndfip1 might be involved in the increase of DMT1 + IRE. Iron 97-101 solute carrier family 11 member 2 Homo sapiens 122-126 25467637-4 2015 Further study showed that the decrease of Ndfip1 occurred earlier than the increase of DMT1 with iron-responsive element (DMT1 + IRE) in 6-OHDA-treated MES23.5 cells, indicating that the decrease of Ndfip1 might be involved in the increase of DMT1 + IRE. Iron 97-101 solute carrier family 11 member 2 Homo sapiens 122-126 25467637-5 2015 In addition, we demonstrated that overexpression of Ndfip1 caused DMT1 + IRE downregulation, resulting in the decreased iron influx and iron-induced neurotoxicity. Iron 120-124 solute carrier family 11 member 2 Homo sapiens 66-70 25467637-5 2015 In addition, we demonstrated that overexpression of Ndfip1 caused DMT1 + IRE downregulation, resulting in the decreased iron influx and iron-induced neurotoxicity. Iron 136-140 solute carrier family 11 member 2 Homo sapiens 66-70 25263405-10 2015 It is here (and in the duodenum) that divalent metal transporter 1 (DMT1), which is primarily responsible for the passage of iron (or uranium?) Iron 125-129 solute carrier family 11 member 2 Homo sapiens 38-66 25263405-10 2015 It is here (and in the duodenum) that divalent metal transporter 1 (DMT1), which is primarily responsible for the passage of iron (or uranium?) Iron 125-129 solute carrier family 11 member 2 Homo sapiens 68-72 25467637-8 2015 These data suggest that decreased Ndfip1 expression might contribute to the pathogenesis of 6-OHDA-induced iron accumulation and Ndfip1 could attenuate 6-OHDA-induced iron accumulation via regulating the degradation of DMT1. Iron 107-111 solute carrier family 11 member 2 Homo sapiens 219-223 25467637-8 2015 These data suggest that decreased Ndfip1 expression might contribute to the pathogenesis of 6-OHDA-induced iron accumulation and Ndfip1 could attenuate 6-OHDA-induced iron accumulation via regulating the degradation of DMT1. Iron 167-171 solute carrier family 11 member 2 Homo sapiens 219-223 25239763-4 2014 6-hydroxydopamine-induced enhanced ferrous iron influx via improper up-regulation of divalent metal transporter 1 with iron responsive element (DMT1+IRE) was consistently relieved by BDNF and GDNF. Iron 43-47 solute carrier family 11 member 2 Homo sapiens 144-152 25239763-4 2014 6-hydroxydopamine-induced enhanced ferrous iron influx via improper up-regulation of divalent metal transporter 1 with iron responsive element (DMT1+IRE) was consistently relieved by BDNF and GDNF. Iron 119-123 solute carrier family 11 member 2 Homo sapiens 144-152 24854545-8 2014 Iron-associated DMT1 could interact with PCBP2 in vitro, whereas iron-chelated DMT1 could not. Iron 65-69 solute carrier family 11 member 2 Homo sapiens 79-83 25127743-2 2014 Reduction of dietary iron from the ferric to the ferrous form is required for uptake by solute carrier family 11 (proton-coupled divalent metal ion transporters), member 2 (Slc11a2) into the enterocytes. Iron 21-25 solute carrier family 11 member 2 Homo sapiens 173-180 24854545-9 2014 These results indicate that ferrous iron imported by DMT1 is transferred directly to PCBP2. Iron 36-40 solute carrier family 11 member 2 Homo sapiens 53-57 24854545-11 2014 These findings suggest that PCBP2 can transfer ferrous iron from DMT1 to the appropriate intracellular sites or ferroportin and could function as an iron chaperone. Iron 55-59 solute carrier family 11 member 2 Homo sapiens 65-69 24854545-1 2014 DMT1 (divalent metal transporter 1) is the main iron importer found in animals, and ferrous iron is taken up by cells via DMT1. Iron 48-52 solute carrier family 11 member 2 Homo sapiens 0-4 24854545-1 2014 DMT1 (divalent metal transporter 1) is the main iron importer found in animals, and ferrous iron is taken up by cells via DMT1. Iron 48-52 solute carrier family 11 member 2 Homo sapiens 6-34 24854545-1 2014 DMT1 (divalent metal transporter 1) is the main iron importer found in animals, and ferrous iron is taken up by cells via DMT1. Iron 92-96 solute carrier family 11 member 2 Homo sapiens 0-4 24521359-7 2014 The pro-inflammatory cytokine IL-1beta facilitates divalent metal transporter 1 (DMT1)-induced beta-cell iron uptake and consequently ROS formation and apoptosis, and we propose that this mechanism provides the relay between inflammation and oxidative beta-cell damage. Iron 105-109 solute carrier family 11 member 2 Homo sapiens 51-79 24854545-1 2014 DMT1 (divalent metal transporter 1) is the main iron importer found in animals, and ferrous iron is taken up by cells via DMT1. Iron 92-96 solute carrier family 11 member 2 Homo sapiens 6-34 24854545-1 2014 DMT1 (divalent metal transporter 1) is the main iron importer found in animals, and ferrous iron is taken up by cells via DMT1. Iron 92-96 solute carrier family 11 member 2 Homo sapiens 122-126 24854545-7 2014 Iron uptake and subsequent ferritin expression were suppressed by either DMT1 or PCBP2 knockdown. Iron 0-4 solute carrier family 11 member 2 Homo sapiens 73-77 24854545-8 2014 Iron-associated DMT1 could interact with PCBP2 in vitro, whereas iron-chelated DMT1 could not. Iron 0-4 solute carrier family 11 member 2 Homo sapiens 16-20 24505080-1 2014 Divalent metal ion transporter 1 (DMT1) is a proton-coupled Fe(2+)transporter that is essential for iron uptake in enterocytes and for transferrin-associated endosomal iron transport in many other cell types. Iron 100-104 solute carrier family 11 member 2 Homo sapiens 34-38 24505080-1 2014 Divalent metal ion transporter 1 (DMT1) is a proton-coupled Fe(2+)transporter that is essential for iron uptake in enterocytes and for transferrin-associated endosomal iron transport in many other cell types. Iron 168-172 solute carrier family 11 member 2 Homo sapiens 34-38 24448823-10 2014 We suggest that DMT1 not only exports iron from endosomes, but also serves to import the metal into the mitochondria. Iron 38-42 solute carrier family 11 member 2 Homo sapiens 16-20 24521359-7 2014 The pro-inflammatory cytokine IL-1beta facilitates divalent metal transporter 1 (DMT1)-induced beta-cell iron uptake and consequently ROS formation and apoptosis, and we propose that this mechanism provides the relay between inflammation and oxidative beta-cell damage. Iron 105-109 solute carrier family 11 member 2 Homo sapiens 81-85 22580996-12 2012 Ineffective erythropoiesis associated with defective erythroid iron utilization due to DMT1 mutations has specific biological and clinical features. Iron 63-67 solute carrier family 11 member 2 Homo sapiens 87-91 24318355-2 2014 Four isoforms of DMT1 have been identified in mammalian cells encoded by a single gene that differ both in their N- and C-terminal sequences with two mRNA isoforms possessing an iron response element (IRE) motif downstream from the stop codon on the message. Iron 178-182 solute carrier family 11 member 2 Homo sapiens 17-21 25276251-0 2014 The role of hepcidin, ferroportin, HCP1, and DMT1 protein in iron absorption in the human digestive tract. Iron 61-65 solute carrier family 11 member 2 Homo sapiens 45-49 25276251-4 2014 Two transporters: Hem Carrier Protein 1 (HCP1) and Divalent Metal Transporter 1 (DMT1) appear to mediate the entry of most if not all dietary iron into these mucosal cells. Iron 142-146 solute carrier family 11 member 2 Homo sapiens 51-79 25276251-4 2014 Two transporters: Hem Carrier Protein 1 (HCP1) and Divalent Metal Transporter 1 (DMT1) appear to mediate the entry of most if not all dietary iron into these mucosal cells. Iron 142-146 solute carrier family 11 member 2 Homo sapiens 81-85 24066281-11 2013 Nramp1 and Nramp2 localization in distinct compartments suggests that both proteins synergistically regulate iron homeostasis. Iron 109-113 solute carrier family 11 member 2 Homo sapiens 11-17 23506870-4 2013 SLC11A2 is a key player in iron metabolism and is ubiquitously expressed, most notably in the proximal duodenum, immature erythroid cells, brain, placenta and kidney. Iron 27-31 solute carrier family 11 member 2 Homo sapiens 0-7 23506870-5 2013 Intestinal iron absorption is mediated by SLC11A2 at the apical membrane of enterocytes, followed by basolateral exit via SLC40A1. Iron 11-15 solute carrier family 11 member 2 Homo sapiens 42-49 23506870-7 2013 Both SLC11A1 and SLC11A2 play an important role in macrophage iron recycling. Iron 62-66 solute carrier family 11 member 2 Homo sapiens 17-24 23506870-8 2013 SLC11A2 also transports iron into the cytosol across the membrane of endocytotic vesicles of the transferrin receptor-cycle. Iron 24-28 solute carrier family 11 member 2 Homo sapiens 0-7 23506870-12 2013 The roles of SLC11A2 and/or SLC40A1 in iron-associated disorders such as hemochromatosis, neurodegenerative diseases, and breast cancer are also summarized. Iron 39-43 solute carrier family 11 member 2 Homo sapiens 13-20 24327293-1 2014 Divalent Metal Transporter 1 (DMT1) is an apical Fe transporter in the duodenum and is involved in endosomal Fe export. Iron 49-51 solute carrier family 11 member 2 Homo sapiens 0-28 24327293-1 2014 Divalent Metal Transporter 1 (DMT1) is an apical Fe transporter in the duodenum and is involved in endosomal Fe export. Iron 49-51 solute carrier family 11 member 2 Homo sapiens 30-34 25483589-1 2014 The divalent metal transporter (DMT1) is well known for its roles in duodenal iron absorption across the apical enterocyte membrane, in iron efflux from the endosome during transferrin-dependent cellular iron acquisition, as well as in uptake of non-transferrin bound iron in many cells. Iron 78-82 solute carrier family 11 member 2 Homo sapiens 32-36 25483589-1 2014 The divalent metal transporter (DMT1) is well known for its roles in duodenal iron absorption across the apical enterocyte membrane, in iron efflux from the endosome during transferrin-dependent cellular iron acquisition, as well as in uptake of non-transferrin bound iron in many cells. Iron 136-140 solute carrier family 11 member 2 Homo sapiens 32-36 25483589-1 2014 The divalent metal transporter (DMT1) is well known for its roles in duodenal iron absorption across the apical enterocyte membrane, in iron efflux from the endosome during transferrin-dependent cellular iron acquisition, as well as in uptake of non-transferrin bound iron in many cells. Iron 136-140 solute carrier family 11 member 2 Homo sapiens 32-36 25483589-1 2014 The divalent metal transporter (DMT1) is well known for its roles in duodenal iron absorption across the apical enterocyte membrane, in iron efflux from the endosome during transferrin-dependent cellular iron acquisition, as well as in uptake of non-transferrin bound iron in many cells. Iron 136-140 solute carrier family 11 member 2 Homo sapiens 32-36 25483589-4 2014 Here we provide additional support for a role of DMT1 in mitochondrial iron acquisition by immunofluorescence colocalization with mitochondrial markers in cells and isolated mitochondria, as well as flow cytometric quantification of DMT1-positive mitochondria from an inducible expression system. Iron 71-75 solute carrier family 11 member 2 Homo sapiens 49-53 24332855-5 2013 NRAMP2 is the major iron transporter in cells, and loss of NRAMP2 attenuates intracellular iron transport. Iron 20-24 solute carrier family 11 member 2 Homo sapiens 0-6 24332855-5 2013 NRAMP2 is the major iron transporter in cells, and loss of NRAMP2 attenuates intracellular iron transport. Iron 20-24 solute carrier family 11 member 2 Homo sapiens 59-65 24089420-3 2013 Divalent metal transporter 1 (Dmt1), the predominant iron importer in the mammalian duodenum, also transports other metal ions, possibly including copper. Iron 53-57 solute carrier family 11 member 2 Homo sapiens 0-28 24089420-3 2013 Divalent metal transporter 1 (Dmt1), the predominant iron importer in the mammalian duodenum, also transports other metal ions, possibly including copper. Iron 53-57 solute carrier family 11 member 2 Homo sapiens 30-34 24089420-10 2013 Additional experiments in Dmt1 overexpressing HEK-293 cells showed that copper ((64)Cu) uptake was stimulated (~3-fold) in the presence of an iron chelator. Iron 142-146 solute carrier family 11 member 2 Homo sapiens 26-30 23771608-6 2013 In accordance with these iron traffic modulations, both mRNA and protein levels of iron importer divalent metal transporter 1 with iron responsive element (DMT1+IRE) and exporter ferroportin 1 (FPN1) were up-regulated in these cells. Iron 25-29 solute carrier family 11 member 2 Homo sapiens 156-164 23771608-6 2013 In accordance with these iron traffic modulations, both mRNA and protein levels of iron importer divalent metal transporter 1 with iron responsive element (DMT1+IRE) and exporter ferroportin 1 (FPN1) were up-regulated in these cells. Iron 83-87 solute carrier family 11 member 2 Homo sapiens 156-164 23303856-1 2013 OBJECTIVE: The present study aimed to evaluate alterations in the levels of iron, divalent metal transporter 1 (DMT1) with the iron-responsive element (IRE), transferrin receptor 1 (TfR1), ferroportin 1 (FPN1), and iron regulatory protein 1 (IRP1) in the temporal cortex of human brains with Parkinson disease (PD). Iron 127-131 solute carrier family 11 member 2 Homo sapiens 82-110 23303856-1 2013 OBJECTIVE: The present study aimed to evaluate alterations in the levels of iron, divalent metal transporter 1 (DMT1) with the iron-responsive element (IRE), transferrin receptor 1 (TfR1), ferroportin 1 (FPN1), and iron regulatory protein 1 (IRP1) in the temporal cortex of human brains with Parkinson disease (PD). Iron 127-131 solute carrier family 11 member 2 Homo sapiens 112-116 22736759-1 2012 Divalent metal-ion transporter-1 (DMT1) is a H(+)-coupled metal-ion transporter that plays essential roles in iron homeostasis. Iron 110-114 solute carrier family 11 member 2 Homo sapiens 0-32 23055972-8 2012 The divalent metal transporter-1 (DMT1) is involved in the uptake of both iron and copper. Iron 74-78 solute carrier family 11 member 2 Homo sapiens 4-32 23055972-8 2012 The divalent metal transporter-1 (DMT1) is involved in the uptake of both iron and copper. Iron 74-78 solute carrier family 11 member 2 Homo sapiens 34-38 23055972-9 2012 Furthermore, copper is an essential co-factor in numerous proteins that are vital for iron homeostasis and affects the binding of iron-response proteins to iron-response elements in the mRNA of the transferrin receptor, DMT1, and ferroportin, all highly involved in iron transport. Iron 130-134 solute carrier family 11 member 2 Homo sapiens 220-224 23055972-9 2012 Furthermore, copper is an essential co-factor in numerous proteins that are vital for iron homeostasis and affects the binding of iron-response proteins to iron-response elements in the mRNA of the transferrin receptor, DMT1, and ferroportin, all highly involved in iron transport. Iron 130-134 solute carrier family 11 member 2 Homo sapiens 220-224 22310887-1 2012 Divalent metal ion transporter (DMT1) is the major transporter for iron entrance into mammalian cells and iron exit from endosomes during the transferrin cycle. Iron 67-71 solute carrier family 11 member 2 Homo sapiens 32-36 22310887-1 2012 Divalent metal ion transporter (DMT1) is the major transporter for iron entrance into mammalian cells and iron exit from endosomes during the transferrin cycle. Iron 106-110 solute carrier family 11 member 2 Homo sapiens 32-36 22137264-4 2012 We show that divalent metal transporter-1 (DMT-1) delocalizes from the plasma membrane upon iron or zinc depletion, but its apical abundance increases with zinc supplementation. Iron 92-96 solute carrier family 11 member 2 Homo sapiens 13-41 22137264-4 2012 We show that divalent metal transporter-1 (DMT-1) delocalizes from the plasma membrane upon iron or zinc depletion, but its apical abundance increases with zinc supplementation. Iron 92-96 solute carrier family 11 member 2 Homo sapiens 43-48 22137264-5 2012 This translocation of DMT-1 coincides with an increase in iron uptake upon zinc supplementation, as previously reported by us. Iron 58-62 solute carrier family 11 member 2 Homo sapiens 22-27 22736759-1 2012 Divalent metal-ion transporter-1 (DMT1) is a H(+)-coupled metal-ion transporter that plays essential roles in iron homeostasis. Iron 110-114 solute carrier family 11 member 2 Homo sapiens 34-38 22736759-6 2012 Radiotracer and continuous measurement of transport by fluorescence assays revealed that DMT1 mediates the transport of several metal ions that were ranked in selectivity by using the ratio I(max)/K(0.5) (determined from evoked currents at -70 mV): Cd(2+) > Fe(2+) > Co(2+), Mn(2+) >> Zn(2+), Ni(2+), VO(2+). Iron 261-263 solute carrier family 11 member 2 Homo sapiens 89-93 22198321-3 2012 There is also evidence showing that a series of genes with important functions in iron metabolism, including transferrin receptor (TfR1) and divalent metal transporter 1 (DMT1), are regulated by HIF-1alpha in response to hypoxia in extra-neural organs or cells. Iron 82-86 solute carrier family 11 member 2 Homo sapiens 141-169 22634399-0 2012 Ebselen inhibits iron-induced tau phosphorylation by attenuating DMT1 up-regulation and cellular iron uptake. Iron 17-21 solute carrier family 11 member 2 Homo sapiens 65-69 22313374-0 2012 The homozygous mutation G75R in the human SLC11A2 gene leads to microcytic anaemia and iron overload. Iron 87-91 solute carrier family 11 member 2 Homo sapiens 42-49 22198321-3 2012 There is also evidence showing that a series of genes with important functions in iron metabolism, including transferrin receptor (TfR1) and divalent metal transporter 1 (DMT1), are regulated by HIF-1alpha in response to hypoxia in extra-neural organs or cells. Iron 82-86 solute carrier family 11 member 2 Homo sapiens 171-175 21947861-1 2012 Divalent metal transporter 1 (DMT1) is likely responsible for the release of iron from endosomes to the cytoplasm in placental syncytiotrophoblasts (STB). Iron 77-81 solute carrier family 11 member 2 Homo sapiens 0-28 21947861-1 2012 Divalent metal transporter 1 (DMT1) is likely responsible for the release of iron from endosomes to the cytoplasm in placental syncytiotrophoblasts (STB). Iron 77-81 solute carrier family 11 member 2 Homo sapiens 30-34 21947861-2 2012 To determine the localization and the regulation of DMT1 expression by iron directly in placenta, the expression of DMT1 in human term placental tissues and BeWo cells (human placental choriocarcinoma cell line) was detected and the change in expression in response to different iron treatments on BeWo cells was observed. Iron 71-75 solute carrier family 11 member 2 Homo sapiens 52-56 21947861-5 2012 Further, DMT1 mRNA responded more significantly to treatments if it possessed an iron-responsive element than mRNA without this element. Iron 81-85 solute carrier family 11 member 2 Homo sapiens 9-13 21947861-6 2012 This study indicated that DMT1 is likely involved in endosomal iron transport in placental STB and placental DMT1 + IRE expression was primarily regulated by the IRE/IRP mechanism. Iron 63-67 solute carrier family 11 member 2 Homo sapiens 26-30 21871825-0 2011 A novel N491S mutation in the human SLC11A2 gene impairs protein trafficking and in association with the G212V mutation leads to microcytic anemia and liver iron overload. Iron 157-161 solute carrier family 11 member 2 Homo sapiens 36-43 23177986-1 2012 Divalent metal-ion transporter-1 (DMT1) is a widely expressed, iron-preferring membrane transport protein. Iron 63-67 solute carrier family 11 member 2 Homo sapiens 0-32 23177986-1 2012 Divalent metal-ion transporter-1 (DMT1) is a widely expressed, iron-preferring membrane transport protein. Iron 63-67 solute carrier family 11 member 2 Homo sapiens 34-38 23177986-2 2012 Animal models establish that DMT1 plays indispensable roles in intestinal nonheme-iron absorption and iron acquisition by erythroid precursor cells. Iron 82-86 solute carrier family 11 member 2 Homo sapiens 29-33 23177986-2 2012 Animal models establish that DMT1 plays indispensable roles in intestinal nonheme-iron absorption and iron acquisition by erythroid precursor cells. Iron 102-106 solute carrier family 11 member 2 Homo sapiens 29-33 23177986-9 2012 An iron-responsive element (IRE) in the mRNA 3"-untranslated region permits the regulation of some isoforms by iron status, and additional mechanisms by which DMT1 is regulated are emerging. Iron 3-7 solute carrier family 11 member 2 Homo sapiens 159-163 23177986-11 2012 The principal or only intestinal nonheme-iron transporter, DMT1 is a validated therapeutic target in hereditary hemochromatosis (HHC) and other iron-overload disorders. Iron 41-45 solute carrier family 11 member 2 Homo sapiens 59-63 22121954-0 2012 Expression of divalent metal transporter 1 in primary hippocampal neurons: reconsidering its role in non-transferrin-bound iron influx. Iron 123-127 solute carrier family 11 member 2 Homo sapiens 14-42 22121954-1 2012 The divalent metal transporter 1 (DMT1) is the best characterized Fe2+ transporter involved in cellular iron uptake in mammals. Iron 104-108 solute carrier family 11 member 2 Homo sapiens 4-32 22121954-1 2012 The divalent metal transporter 1 (DMT1) is the best characterized Fe2+ transporter involved in cellular iron uptake in mammals. Iron 104-108 solute carrier family 11 member 2 Homo sapiens 34-38 22121954-3 2012 Despite the general importance of DMT1 in controlling iron homeostasis, the distribution and the role of the transporter in the CNS is still controversial. Iron 54-58 solute carrier family 11 member 2 Homo sapiens 34-38 22121954-5 2012 We found that the main isoform endogenously expressed is DMT1-1B/IRE+, which shows cytoplasmic distribution, colocalization with late endosome/lysosome markers and iron regulation, as expected from the presence of an iron responsive element. Iron 164-168 solute carrier family 11 member 2 Homo sapiens 57-61 22121954-5 2012 We found that the main isoform endogenously expressed is DMT1-1B/IRE+, which shows cytoplasmic distribution, colocalization with late endosome/lysosome markers and iron regulation, as expected from the presence of an iron responsive element. Iron 217-221 solute carrier family 11 member 2 Homo sapiens 57-61 22121954-7 2012 Overall, our results argue against a physiological role of the endogenous DMT1 in neuronal iron uptake but do not exclude that, under pathological conditions, the expression of other DMT1 isoforms might contribute to iron overload. Iron 217-221 solute carrier family 11 member 2 Homo sapiens 183-187 22509377-1 2012 BACKGROUND: Iron-refractory iron deficiency anaemia (IRIDA) is a rare disorder which was linked to mutations in two genes (SLC11A2 and TMPRSS6). Iron 12-16 solute carrier family 11 member 2 Homo sapiens 123-130 22509377-10 2012 One individual exhibiting the joint SLC11A2/TMPRSS6 profile of the anaemic son had iron and ferritin levels lying below the 5(th) percentile of the population"s iron and ferritin level distribution. Iron 83-87 solute carrier family 11 member 2 Homo sapiens 36-43 22509377-10 2012 One individual exhibiting the joint SLC11A2/TMPRSS6 profile of the anaemic son had iron and ferritin levels lying below the 5(th) percentile of the population"s iron and ferritin level distribution. Iron 161-165 solute carrier family 11 member 2 Homo sapiens 36-43 22068728-0 2012 Iron, copper, and zinc transport: inhibition of divalent metal transporter 1 (DMT1) and human copper transporter 1 (hCTR1) by shRNA. Iron 0-4 solute carrier family 11 member 2 Homo sapiens 48-76 22068728-0 2012 Iron, copper, and zinc transport: inhibition of divalent metal transporter 1 (DMT1) and human copper transporter 1 (hCTR1) by shRNA. Iron 0-4 solute carrier family 11 member 2 Homo sapiens 78-82 22068728-6 2012 Both shRNA-DMT1 and shRNA-hCTR1 cells had lower apical Fe uptake (a decrease of 51% and 41%, respectively), Cu uptake (a decrease of 25.8% and 38.5%, respectively), and Zn content (a decrease of 23.1% and 22.7%, respectively) compared to control cells. Iron 55-57 solute carrier family 11 member 2 Homo sapiens 11-15 22068728-7 2012 These results confirm that DMT1 is involved in active transport of Fe, Cu, and Zn although Zn showed a different relative capacity. Iron 67-69 solute carrier family 11 member 2 Homo sapiens 27-31 22154351-5 2012 These three series represent promising starting points for lead optimization efforts aimed at the discovery of DMT1 blockers as iron overload therapeutics. Iron 128-132 solute carrier family 11 member 2 Homo sapiens 111-115 22666436-3 2012 We hypothesized that the increase of iron through a NF-kappaB-regulated 1B isoform of the divalent metal transporter-1 (1B/DMT1) might contribute to post-ischemic neuronal damage. Iron 37-41 solute carrier family 11 member 2 Homo sapiens 123-127 22666436-5 2012 Either OGD or over-expression of 1B/(-)IRE DMT1 isoform significantly increased iron uptake, as detected by total reflection X-ray fluorescence, and iron-dependent cell death. Iron 80-84 solute carrier family 11 member 2 Homo sapiens 43-47 22666436-5 2012 Either OGD or over-expression of 1B/(-)IRE DMT1 isoform significantly increased iron uptake, as detected by total reflection X-ray fluorescence, and iron-dependent cell death. Iron 149-153 solute carrier family 11 member 2 Homo sapiens 43-47 22666436-6 2012 Iron chelation by deferoxamine treatment or (-)IRE DMT1 RNA silencing displayed significant neuroprotection against OGD which concomitantly decreased intracellular iron levels. Iron 164-168 solute carrier family 11 member 2 Homo sapiens 51-55 21871825-1 2011 BACKGROUND: DMT1 is a transmembrane iron transporter involved in iron duodenal absorption and cellular iron uptake. Iron 36-40 solute carrier family 11 member 2 Homo sapiens 12-16 21871825-1 2011 BACKGROUND: DMT1 is a transmembrane iron transporter involved in iron duodenal absorption and cellular iron uptake. Iron 65-69 solute carrier family 11 member 2 Homo sapiens 12-16 21871825-2 2011 Mutations in the human SLC11A2 gene coding DMT1 lead to microcytic anemia and hepatic iron overload, with unexpectedly low levels of plasma ferritin in the presence of iron stores. Iron 86-90 solute carrier family 11 member 2 Homo sapiens 23-30 21871825-2 2011 Mutations in the human SLC11A2 gene coding DMT1 lead to microcytic anemia and hepatic iron overload, with unexpectedly low levels of plasma ferritin in the presence of iron stores. Iron 86-90 solute carrier family 11 member 2 Homo sapiens 43-47 21871825-2 2011 Mutations in the human SLC11A2 gene coding DMT1 lead to microcytic anemia and hepatic iron overload, with unexpectedly low levels of plasma ferritin in the presence of iron stores. Iron 168-172 solute carrier family 11 member 2 Homo sapiens 23-30 21871825-2 2011 Mutations in the human SLC11A2 gene coding DMT1 lead to microcytic anemia and hepatic iron overload, with unexpectedly low levels of plasma ferritin in the presence of iron stores. Iron 168-172 solute carrier family 11 member 2 Homo sapiens 43-47 21795716-3 2011 Notably, nigral dopaminergic neurons are enriched in iron, the uptake of which is facilitated by the divalent metal ion transporter DMT1. Iron 53-57 solute carrier family 11 member 2 Homo sapiens 132-136 22060282-1 2011 OBJECTIVES: The absorption of commonly used ferrous iron salts from intestinal segments at neutral to slightly alkaline pH is low, mainly because soluble ferrous iron is easily oxidized to poorly soluble ferric iron and ferrous iron but not ferric iron is carried by the divalent metal transporter DMT-1. Iron 44-56 solute carrier family 11 member 2 Homo sapiens 298-303 22060282-1 2011 OBJECTIVES: The absorption of commonly used ferrous iron salts from intestinal segments at neutral to slightly alkaline pH is low, mainly because soluble ferrous iron is easily oxidized to poorly soluble ferric iron and ferrous iron but not ferric iron is carried by the divalent metal transporter DMT-1. Iron 154-166 solute carrier family 11 member 2 Homo sapiens 298-303 21795716-5 2011 We found that DMT1 overexpression dramatically enhances Fe(2+) uptake, which concomitantly promotes cell death. Iron 56-58 solute carrier family 11 member 2 Homo sapiens 14-18 21276595-2 2011 SLC11A2 gene encodes the divalent metal transport 1 (DMT1) mediating iron transport in cerebral endosomal compartments. Iron 69-73 solute carrier family 11 member 2 Homo sapiens 0-7 21438013-5 2011 Our results demonstrated that astrocytes, when treated with hepcidin peptide or infected with hepcidin expression adenovirus (ad-hepcidin), showed a significant ability in reducing iron uptake (both Tf-Fe and NTBI), and iron release, which were accompanied by decreased expressions of TfR1, DMT1, and Fpn1. Iron 181-185 solute carrier family 11 member 2 Homo sapiens 291-295 19913089-6 2011 Pb transport displayed properties that were associated with iron response element (IRE) positive isoform of DMT1. Iron 60-64 solute carrier family 11 member 2 Homo sapiens 108-112 21276595-9 2011 CONCLUSIONS: Our findings support an implication for iron metabolism in ALS and suggest that the genotype of the SLC11A2 gene could modulate the duration of the disease in French SALS patients. Iron 53-57 solute carrier family 11 member 2 Homo sapiens 113-120 21276595-2 2011 SLC11A2 gene encodes the divalent metal transport 1 (DMT1) mediating iron transport in cerebral endosomal compartments. Iron 69-73 solute carrier family 11 member 2 Homo sapiens 25-51 21276595-2 2011 SLC11A2 gene encodes the divalent metal transport 1 (DMT1) mediating iron transport in cerebral endosomal compartments. Iron 69-73 solute carrier family 11 member 2 Homo sapiens 53-57 20819077-0 2010 Two routes of iron accumulation in astrocytes: ascorbate-dependent ferrous iron uptake via the divalent metal transporter (DMT1) plus an independent route for ferric iron. Iron 14-18 solute carrier family 11 member 2 Homo sapiens 123-127 22123640-8 2011 Iron in the developing fetus is accumulated against a concentration gradient and, in the case of maternal iron deficiency, the placenta can protect the fetus significantly through the increased expression of placental transferrin receptor together with a rise in divalent metal transporter 1 (DMT1). Iron 0-4 solute carrier family 11 member 2 Homo sapiens 263-291 22123640-8 2011 Iron in the developing fetus is accumulated against a concentration gradient and, in the case of maternal iron deficiency, the placenta can protect the fetus significantly through the increased expression of placental transferrin receptor together with a rise in divalent metal transporter 1 (DMT1). Iron 0-4 solute carrier family 11 member 2 Homo sapiens 293-297 20819077-0 2010 Two routes of iron accumulation in astrocytes: ascorbate-dependent ferrous iron uptake via the divalent metal transporter (DMT1) plus an independent route for ferric iron. Iron 67-79 solute carrier family 11 member 2 Homo sapiens 123-127 20819077-6 2010 The acquisition of this additional iron depends on effluxed ascorbate and can be blocked by the DMT1 inhibitor ferristatin/NSC306711. Iron 35-39 solute carrier family 11 member 2 Homo sapiens 96-100 20007457-0 2010 Iron supply determines apical/basolateral membrane distribution of intestinal iron transporters DMT1 and ferroportin 1. Iron 0-4 solute carrier family 11 member 2 Homo sapiens 96-100 20394798-0 2010 Both Nramp1 and DMT1 are necessary for efficient macrophage iron recycling. Iron 60-64 solute carrier family 11 member 2 Homo sapiens 16-20 20394798-6 2010 Compared to macrophages singly deficient in either DMT1 or Nramp1 transport ability, macrophages where DMT1 and Nramp1 were both compromised exhibited an abrogated increase in labile iron pool content, released less iron, and experienced diminished upregulation of ferroportin and heme-oxygenase 1 levels following erythrophagocytosis. Iron 183-187 solute carrier family 11 member 2 Homo sapiens 103-107 20394798-7 2010 CONCLUSIONS: These results suggest that although the loss of either Nramp1 or DMT1 transport ability results in minor impairment after erythrophagocytosis, the simultaneous loss of both Nramp1 and DMT1 iron transport activity is detrimental to the iron recycling capacity of the macrophage. Iron 202-206 solute carrier family 11 member 2 Homo sapiens 197-201 20394798-7 2010 CONCLUSIONS: These results suggest that although the loss of either Nramp1 or DMT1 transport ability results in minor impairment after erythrophagocytosis, the simultaneous loss of both Nramp1 and DMT1 iron transport activity is detrimental to the iron recycling capacity of the macrophage. Iron 248-252 solute carrier family 11 member 2 Homo sapiens 197-201 20410187-1 2010 BACKGROUND: Divalent metal transporter 1 (DMT1) is a widely expressed metal-iron transporter gene encoding four variant mRNA transcripts, differing for alternative promoter at 5" (DMT1 1A and 1B) and alternative splicing at 3" UTR, differing by a specific sequence either containing or lacking an iron regulatory element (+IRE and -IRE, respectively). Iron 76-80 solute carrier family 11 member 2 Homo sapiens 12-40 20410187-1 2010 BACKGROUND: Divalent metal transporter 1 (DMT1) is a widely expressed metal-iron transporter gene encoding four variant mRNA transcripts, differing for alternative promoter at 5" (DMT1 1A and 1B) and alternative splicing at 3" UTR, differing by a specific sequence either containing or lacking an iron regulatory element (+IRE and -IRE, respectively). Iron 76-80 solute carrier family 11 member 2 Homo sapiens 42-46 20410187-1 2010 BACKGROUND: Divalent metal transporter 1 (DMT1) is a widely expressed metal-iron transporter gene encoding four variant mRNA transcripts, differing for alternative promoter at 5" (DMT1 1A and 1B) and alternative splicing at 3" UTR, differing by a specific sequence either containing or lacking an iron regulatory element (+IRE and -IRE, respectively). Iron 76-80 solute carrier family 11 member 2 Homo sapiens 180-184 20410187-10 2010 CONCLUSIONS: Overall, these data suggest that miR-Let-7d participates in the finely tuned regulation of iron metabolism by targeting DMT1-IRE isoform in erythroid cells. Iron 104-108 solute carrier family 11 member 2 Homo sapiens 133-137 20089134-3 2010 The major transport protein responsible for uptake of iron is divalent metal transporter 1 (DMT1) and recent studies demonstrate that the 1B species is regulated post-translationally by degradation via the proteasomal pathway. Iron 54-58 solute carrier family 11 member 2 Homo sapiens 62-90 20089134-3 2010 The major transport protein responsible for uptake of iron is divalent metal transporter 1 (DMT1) and recent studies demonstrate that the 1B species is regulated post-translationally by degradation via the proteasomal pathway. Iron 54-58 solute carrier family 11 member 2 Homo sapiens 92-96 20152801-3 2010 We tested the hypothesis that divalent metal-ion transporter-1 (DMT1)-the principal or only mechanism by which nonheme iron is taken up at the intestinal brush border-is shared also by calcium. Iron 119-123 solute carrier family 11 member 2 Homo sapiens 30-62 20152801-3 2010 We tested the hypothesis that divalent metal-ion transporter-1 (DMT1)-the principal or only mechanism by which nonheme iron is taken up at the intestinal brush border-is shared also by calcium. Iron 119-123 solute carrier family 11 member 2 Homo sapiens 64-68 20152801-8 2010 The alkaline-earth metal ions Ba2+, Sr2+, and Mg2+ also inhibited DMT1-mediated iron-transport activity. Iron 80-84 solute carrier family 11 member 2 Homo sapiens 66-70 21462112-5 2010 The interaction between Ca and Fe may be a lumenal event, affecting Fe uptake through DMT1 (divalent metal transporter 1) at the apical membrane. Iron 31-33 solute carrier family 11 member 2 Homo sapiens 86-90 21462112-5 2010 The interaction between Ca and Fe may be a lumenal event, affecting Fe uptake through DMT1 (divalent metal transporter 1) at the apical membrane. Iron 31-33 solute carrier family 11 member 2 Homo sapiens 92-120 21462112-5 2010 The interaction between Ca and Fe may be a lumenal event, affecting Fe uptake through DMT1 (divalent metal transporter 1) at the apical membrane. Iron 68-70 solute carrier family 11 member 2 Homo sapiens 86-90 21462112-5 2010 The interaction between Ca and Fe may be a lumenal event, affecting Fe uptake through DMT1 (divalent metal transporter 1) at the apical membrane. Iron 68-70 solute carrier family 11 member 2 Homo sapiens 92-120 20631677-6 2010 In Caco-2 cells, the addition of erythropoietin significantly increased the expression of apical divalent metal transporter 1 (DMT1) and basolateral ferroportin and, consequently, iron transport across the monolayer. Iron 180-184 solute carrier family 11 member 2 Homo sapiens 127-131 20681027-0 2010 Hypoxia regulates the ferrous iron uptake and reactive oxygen species level via divalent metal transporter 1 (DMT1) Exon1B by hypoxia-inducible factor-1. Iron 22-34 solute carrier family 11 member 2 Homo sapiens 80-108 20681027-0 2010 Hypoxia regulates the ferrous iron uptake and reactive oxygen species level via divalent metal transporter 1 (DMT1) Exon1B by hypoxia-inducible factor-1. Iron 22-34 solute carrier family 11 member 2 Homo sapiens 110-114 20681027-2 2010 Divalent metal transporter 1 (DMT1) is a transmembrane protein that is important in divalent metal ion transport, in particular iron. Iron 128-132 solute carrier family 11 member 2 Homo sapiens 0-28 20681027-2 2010 Divalent metal transporter 1 (DMT1) is a transmembrane protein that is important in divalent metal ion transport, in particular iron. Iron 128-132 solute carrier family 11 member 2 Homo sapiens 30-34 20681027-6 2010 Both the total cellular iron and ferrous uptake increased after hypoxia, decreased after DMT1 RNA interference. Iron 24-28 solute carrier family 11 member 2 Homo sapiens 89-93 20681027-7 2010 Reactive oxygen species (ROS) were also changed by +iron responsive element (IRE) DMT1 exon1B overexpression. Iron 52-56 solute carrier family 11 member 2 Homo sapiens 82-86 20681027-9 2010 Hypoxia might affect cellular iron uptake through regulating the expression of DMT1. Iron 30-34 solute carrier family 11 member 2 Homo sapiens 79-83 20509117-6 2010 Transport of non-haem iron from the proximal intestinal lumen into the enterocytes is mediated by the divalent metal transporter 1 (DMT1). Iron 22-26 solute carrier family 11 member 2 Homo sapiens 102-130 20509117-6 2010 Transport of non-haem iron from the proximal intestinal lumen into the enterocytes is mediated by the divalent metal transporter 1 (DMT1). Iron 22-26 solute carrier family 11 member 2 Homo sapiens 132-136 20007457-1 2010 Intestinal iron absorption comprises the coordinated activity of the influx transporter divalent metal transporter 1 (DMT1) and the efflux transporter ferroportin (FPN). Iron 11-15 solute carrier family 11 member 2 Homo sapiens 118-122 20007457-2 2010 In this work, we studied the movement of DMT1 and FPN between cellular compartments as a function of iron supply. Iron 101-105 solute carrier family 11 member 2 Homo sapiens 41-45 20007457-5 2010 In Caco-2 cells, the apical-to-basal movement of cyan fluorescent protein-tagged DMT1 was complete 90 min after the addition of iron. Iron 128-132 solute carrier family 11 member 2 Homo sapiens 81-85 20007457-6 2010 Steady-state membrane localization studies in Caco-2 cells revealed that iron status determined the apical/basolateral membrane distribution of DMT1 and FPN. Iron 73-77 solute carrier family 11 member 2 Homo sapiens 144-148 20007457-8 2010 Antisense oligonucleotides targeted to DMT1 or FPN inhibited basolateral iron uptake and apical iron efflux, respectively, indicating the participation of DMT1 and FPN in these fluxes. Iron 73-77 solute carrier family 11 member 2 Homo sapiens 39-43 20007457-8 2010 Antisense oligonucleotides targeted to DMT1 or FPN inhibited basolateral iron uptake and apical iron efflux, respectively, indicating the participation of DMT1 and FPN in these fluxes. Iron 73-77 solute carrier family 11 member 2 Homo sapiens 155-159 20007457-8 2010 Antisense oligonucleotides targeted to DMT1 or FPN inhibited basolateral iron uptake and apical iron efflux, respectively, indicating the participation of DMT1 and FPN in these fluxes. Iron 96-100 solute carrier family 11 member 2 Homo sapiens 39-43 20007457-10 2010 These findings suggest a novel mechanism of regulation of intestinal iron absorption based on inward and outward fluxes at both membrane domains, and repositioning of DMT1 and FPN between membrane and intracellular compartments as a function of iron supply. Iron 245-249 solute carrier family 11 member 2 Homo sapiens 167-171 20392994-1 2010 Since malignant cells often have a high demand for iron, we hypothesize that breast cancer cells may alter the expression of iron transporter genes including iron importers [transferrin receptor (TFRC) and solute carrier family 11 (proton-coupled divalent metal ion transporters), member 2 (SLC11A2)] and the iron exporter SLC40A1 (ferroportin), and additionally that the growth of breast cancer can be inhibited by manipulating iron transporter gene expression. Iron 125-129 solute carrier family 11 member 2 Homo sapiens 291-298 20392994-1 2010 Since malignant cells often have a high demand for iron, we hypothesize that breast cancer cells may alter the expression of iron transporter genes including iron importers [transferrin receptor (TFRC) and solute carrier family 11 (proton-coupled divalent metal ion transporters), member 2 (SLC11A2)] and the iron exporter SLC40A1 (ferroportin), and additionally that the growth of breast cancer can be inhibited by manipulating iron transporter gene expression. Iron 125-129 solute carrier family 11 member 2 Homo sapiens 291-298 20232409-0 2010 Regulatory mechanisms of intestinal iron absorption-uncovering of a fast-response mechanism based on DMT1 and ferroportin endocytosis. Iron 36-40 solute carrier family 11 member 2 Homo sapiens 101-105 19786204-1 2009 Divalent metal transporter 1 (DMT1) is the protein that allows elemental iron entry into the duodenal cell. Iron 73-77 solute carrier family 11 member 2 Homo sapiens 0-28 19306086-12 2009 Elevations in cell iron can possibly affect an increased expression of DMT1 and ferritin which function to diminish oxidative stress. Iron 19-23 solute carrier family 11 member 2 Homo sapiens 71-75 20619018-2 2010 These defects are located in genes that encode for the cellular iron importing protein Divalent Metal Transporter 1 (DMT1), the iron exporting protein ferroportin, the mitochondrial enzyme glutaredoxin-5 and the hepatocyte membrane protein matriptase-2. Iron 64-68 solute carrier family 11 member 2 Homo sapiens 87-115 20619018-2 2010 These defects are located in genes that encode for the cellular iron importing protein Divalent Metal Transporter 1 (DMT1), the iron exporting protein ferroportin, the mitochondrial enzyme glutaredoxin-5 and the hepatocyte membrane protein matriptase-2. Iron 64-68 solute carrier family 11 member 2 Homo sapiens 117-121 19786204-1 2009 Divalent metal transporter 1 (DMT1) is the protein that allows elemental iron entry into the duodenal cell. Iron 73-77 solute carrier family 11 member 2 Homo sapiens 30-34 19786204-12 2009 In this review we analyze the role of DMT1 in iron metabolism and the major causes of reduction and their consequences in animal models as well in humans, and we attempt to define the correct treatment for human mutants. Iron 46-50 solute carrier family 11 member 2 Homo sapiens 38-42 19179627-1 2009 Divalent metal transporter-1 (DMT1) is a divalent cation transporter that plays a key role in iron metabolism by mediating ferrous iron uptake across the small intestine. Iron 94-98 solute carrier family 11 member 2 Homo sapiens 0-28 19553145-0 2009 Not all DMT1 mutations lead to iron overload. Iron 31-35 solute carrier family 11 member 2 Homo sapiens 8-12 19553145-2 2009 Results from animal models and from patients have shown that DMT1 is required for intestinal iron absorption and iron acquisition by erythrocytes. Iron 93-97 solute carrier family 11 member 2 Homo sapiens 61-65 19553145-2 2009 Results from animal models and from patients have shown that DMT1 is required for intestinal iron absorption and iron acquisition by erythrocytes. Iron 113-117 solute carrier family 11 member 2 Homo sapiens 61-65 19706893-3 2009 The divalent metal transporter 1 (DMT1) plays a central role in the regulation of Fe as well as other metals; hence, failure of DMT1 regulation is linked to human brain pathology. Iron 82-84 solute carrier family 11 member 2 Homo sapiens 4-32 19706893-3 2009 The divalent metal transporter 1 (DMT1) plays a central role in the regulation of Fe as well as other metals; hence, failure of DMT1 regulation is linked to human brain pathology. Iron 82-84 solute carrier family 11 member 2 Homo sapiens 34-38 19706893-3 2009 The divalent metal transporter 1 (DMT1) plays a central role in the regulation of Fe as well as other metals; hence, failure of DMT1 regulation is linked to human brain pathology. Iron 82-84 solute carrier family 11 member 2 Homo sapiens 128-132 19706893-6 2009 Using human neurons we show the Ndfip1 is upregulated and binds to DMT1 in response to Fe and cobalt (Co) exposure. Iron 87-89 solute carrier family 11 member 2 Homo sapiens 67-71 19788062-9 2009 Based on these results, a two-transporter model of iron uptake, comprising the apical iron uptake transporter divalent metal ion transporter-1 (DMT-1) and an unknown putative transporter was derived. Iron 51-55 solute carrier family 11 member 2 Homo sapiens 110-142 19788062-9 2009 Based on these results, a two-transporter model of iron uptake, comprising the apical iron uptake transporter divalent metal ion transporter-1 (DMT-1) and an unknown putative transporter was derived. Iron 51-55 solute carrier family 11 member 2 Homo sapiens 144-149 19788062-9 2009 Based on these results, a two-transporter model of iron uptake, comprising the apical iron uptake transporter divalent metal ion transporter-1 (DMT-1) and an unknown putative transporter was derived. Iron 86-90 solute carrier family 11 member 2 Homo sapiens 110-142 19788062-9 2009 Based on these results, a two-transporter model of iron uptake, comprising the apical iron uptake transporter divalent metal ion transporter-1 (DMT-1) and an unknown putative transporter was derived. Iron 86-90 solute carrier family 11 member 2 Homo sapiens 144-149 19788062-12 2009 DMT-1 might not simultaneously transport iron and zinc, providing a mechanistic basis for observed interactions. Iron 41-45 solute carrier family 11 member 2 Homo sapiens 0-5 19179627-1 2009 Divalent metal transporter-1 (DMT1) is a divalent cation transporter that plays a key role in iron metabolism by mediating ferrous iron uptake across the small intestine. Iron 94-98 solute carrier family 11 member 2 Homo sapiens 30-34 19179627-1 2009 Divalent metal transporter-1 (DMT1) is a divalent cation transporter that plays a key role in iron metabolism by mediating ferrous iron uptake across the small intestine. Iron 123-135 solute carrier family 11 member 2 Homo sapiens 0-28 19179627-1 2009 Divalent metal transporter-1 (DMT1) is a divalent cation transporter that plays a key role in iron metabolism by mediating ferrous iron uptake across the small intestine. Iron 123-135 solute carrier family 11 member 2 Homo sapiens 30-34 19179627-4 2009 One compound, NSC306711, inhibited DMT1-mediated iron uptake in a reversible and competitive manner. Iron 49-53 solute carrier family 11 member 2 Homo sapiens 35-39 19179627-8 2009 This study characterizes important pharmacological tools that can be used to probe DMT1"s mechanism of iron transport and its role in iron metabolism. Iron 103-107 solute carrier family 11 member 2 Homo sapiens 83-87 19179627-8 2009 This study characterizes important pharmacological tools that can be used to probe DMT1"s mechanism of iron transport and its role in iron metabolism. Iron 134-138 solute carrier family 11 member 2 Homo sapiens 83-87 19147412-2 2009 Duodenal cytochrome b (DcytB) and divalent metal transporter 1 (DMT1) are regulators of iron absorption. Iron 88-92 solute carrier family 11 member 2 Homo sapiens 34-62 19147412-2 2009 Duodenal cytochrome b (DcytB) and divalent metal transporter 1 (DMT1) are regulators of iron absorption. Iron 88-92 solute carrier family 11 member 2 Homo sapiens 64-68 19147412-4 2009 Hypoxia-inducible factor (HIF) signaling was induced in the intestine following acute iron deficiency in the duodenum, resulting in activation of DcytB and DMT1 expression and an increase in iron uptake. Iron 86-90 solute carrier family 11 member 2 Homo sapiens 156-160 19240805-8 2009 CONCLUSION: The up-regulation of DMT1-IRE and the increase in DMT1-IRE-mediated iron influx play a key role in L-DOPA neurotoxicity in cortical neurons. Iron 80-84 solute carrier family 11 member 2 Homo sapiens 62-66 18838961-10 2008 However, there are complex alterations in DMT1 and ferroportin expression in cirrhotic liver, including decreases in ferroportin and DMT1 at the protein level that may play a role in aberrant regulation of iron metabolism in cirrhosis. Iron 206-210 solute carrier family 11 member 2 Homo sapiens 133-137 18815723-13 2008 Western Blot analyses revealed a long term down-regulating effect of ascorbic acid on iron independent and iron dependent Nramp2 and Dcytb expression. Iron 107-111 solute carrier family 11 member 2 Homo sapiens 122-128 19002083-14 2008 A number of proteins is involved in iron metabolism including: ferritin, transferrin,transferrin receptor, divalent metal transporter (DMT1), cytochrome b, ferroportin, hephaestin, hepcidin and lactoferrin (LF). Iron 36-40 solute carrier family 11 member 2 Homo sapiens 135-139 18815723-16 2008 However, the impact of iron alone on Nramp2 up-regulation seems to be greater in the absence of ascorbic acid. Iron 23-27 solute carrier family 11 member 2 Homo sapiens 37-43 18815723-20 2008 Similarly, the short term up-regulation of Nramp2 and Dcytb seems to agree with the improvement in iron uptake shown in humans when single doses of ascorbic acid were administrated. Iron 99-103 solute carrier family 11 member 2 Homo sapiens 43-49 18281333-1 2008 Divalent metal ion transporter 1 (DMT1; also known as SLC11A2) can transport several metals including Fe and Cu in mammalian systems. Iron 102-104 solute carrier family 11 member 2 Homo sapiens 0-32 18492824-2 2008 It is thought to act as a ferric reductase that furnishes Fe(II), the specific and selective iron species transported by divalent metal transporter 1 (DMT1) in the duodenal enterocytes. Iron 93-97 solute carrier family 11 member 2 Homo sapiens 121-149 18492824-2 2008 It is thought to act as a ferric reductase that furnishes Fe(II), the specific and selective iron species transported by divalent metal transporter 1 (DMT1) in the duodenal enterocytes. Iron 93-97 solute carrier family 11 member 2 Homo sapiens 151-155 18492824-9 2008 Cotransfection of Dcytb and DMT1 resulted in an additive increase in iron uptake by the cells. Iron 69-73 solute carrier family 11 member 2 Homo sapiens 28-32 18082289-4 2008 The aim of the present study is to construct recombinant adenovirus encoding human DMT1 with iron responsive element (DMT1+IRE) and infect MES23.5 dopaminergic cells in order to investigate the relationship between increased DMT1+IRE expression and iron accumulation. Iron 93-97 solute carrier family 11 member 2 Homo sapiens 83-87 18082289-4 2008 The aim of the present study is to construct recombinant adenovirus encoding human DMT1 with iron responsive element (DMT1+IRE) and infect MES23.5 dopaminergic cells in order to investigate the relationship between increased DMT1+IRE expression and iron accumulation. Iron 93-97 solute carrier family 11 member 2 Homo sapiens 118-122 18082289-4 2008 The aim of the present study is to construct recombinant adenovirus encoding human DMT1 with iron responsive element (DMT1+IRE) and infect MES23.5 dopaminergic cells in order to investigate the relationship between increased DMT1+IRE expression and iron accumulation. Iron 93-97 solute carrier family 11 member 2 Homo sapiens 118-122 18082289-4 2008 The aim of the present study is to construct recombinant adenovirus encoding human DMT1 with iron responsive element (DMT1+IRE) and infect MES23.5 dopaminergic cells in order to investigate the relationship between increased DMT1+IRE expression and iron accumulation. Iron 249-253 solute carrier family 11 member 2 Homo sapiens 83-87 18082289-13 2008 These results suggested that increased DMT1+IRE expression in MES23.5 cells caused the increased intracellular iron accumulation. Iron 111-115 solute carrier family 11 member 2 Homo sapiens 39-43 18419598-2 2008 Divalent metal transporter 1 (DMT1) is also a key transporter of iron under physiological conditions. Iron 65-69 solute carrier family 11 member 2 Homo sapiens 0-28 18419598-2 2008 Divalent metal transporter 1 (DMT1) is also a key transporter of iron under physiological conditions. Iron 65-69 solute carrier family 11 member 2 Homo sapiens 30-34 18281333-1 2008 Divalent metal ion transporter 1 (DMT1; also known as SLC11A2) can transport several metals including Fe and Cu in mammalian systems. Iron 102-104 solute carrier family 11 member 2 Homo sapiens 34-38 18281333-1 2008 Divalent metal ion transporter 1 (DMT1; also known as SLC11A2) can transport several metals including Fe and Cu in mammalian systems. Iron 102-104 solute carrier family 11 member 2 Homo sapiens 54-61 17533042-0 2007 Iron/IRP-1-dependent regulation of mRNA expression for transferrin receptor, DMT1 and ferritin during human erythroid differentiation. Iron 0-4 solute carrier family 11 member 2 Homo sapiens 77-81 18223212-10 2008 Iron loading OE33 and SEG-1 cells caused increased cellular proliferation, which was associated with increased H-ferritin and decreased transferrin receptor 1 and DMT1 expression. Iron 0-4 solute carrier family 11 member 2 Homo sapiens 163-167 17510944-2 2007 Animal models with mutation in (DMT1) divalent metal transporter 1 gene, an important brain iron transporter, demonstrate a similar iron deficiency profile as found in RLS brain. Iron 92-96 solute carrier family 11 member 2 Homo sapiens 32-36 17719162-1 2007 Low iron status is known to increase the uptake of dietary cadmium in both adolescents and adults and there are indications that cadmium is absorbed from the intestine by the two major iron transporters divalent metal transporter 1 (DMT1) and ferroportin 1 (FPN1). Iron 4-8 solute carrier family 11 member 2 Homo sapiens 203-231 17719162-1 2007 Low iron status is known to increase the uptake of dietary cadmium in both adolescents and adults and there are indications that cadmium is absorbed from the intestine by the two major iron transporters divalent metal transporter 1 (DMT1) and ferroportin 1 (FPN1). Iron 4-8 solute carrier family 11 member 2 Homo sapiens 233-237 17533042-8 2007 CONCLUSION: These results suggest that IRP-1/IRE interactions, which are supposedly induced after sensing a decrease of the intracellular non-Heme iron levels, play a crucial role on the posttranscriptional regulation of TfR, DMT1, and ferritin mRNAs during differentiation of normal human erythropoietic cells. Iron 147-151 solute carrier family 11 member 2 Homo sapiens 226-230 17109629-1 2007 DMT1 (divalent metal-ion transporter 1) is a widely expressed metal-ion transporter that is vital for intestinal iron absorption and iron utilization by most cell types throughout the body, including erythroid precursors. Iron 113-117 solute carrier family 11 member 2 Homo sapiens 0-4 16927405-5 2007 We found that iron uptake in this cell type is mediated by divalent-metal transporter 1 (DMT1) and transferrin receptor-1 (TfR) whereas Ferroportin-1 is very weakly expressed. Iron 14-18 solute carrier family 11 member 2 Homo sapiens 59-87 16927405-5 2007 We found that iron uptake in this cell type is mediated by divalent-metal transporter 1 (DMT1) and transferrin receptor-1 (TfR) whereas Ferroportin-1 is very weakly expressed. Iron 14-18 solute carrier family 11 member 2 Homo sapiens 89-93 16927405-7 2007 The expression of DMT1 and HFE barely varies upon endotoxin-induced maturation but TfR is up-regulated and the iron export molecule Ferroportin-1 is down-regulated. Iron 111-115 solute carrier family 11 member 2 Homo sapiens 18-22 16927405-9 2007 Our results indicate that the uptake of iron during DCs development and maturation is mediated by a strong expression of iron-uptake molecules such as DMT1 and TfR as well as a down-regulation of iron export molecules such as Ferroportin-1. Iron 40-44 solute carrier family 11 member 2 Homo sapiens 151-155 17109629-1 2007 DMT1 (divalent metal-ion transporter 1) is a widely expressed metal-ion transporter that is vital for intestinal iron absorption and iron utilization by most cell types throughout the body, including erythroid precursors. Iron 113-117 solute carrier family 11 member 2 Homo sapiens 6-38 17109629-1 2007 DMT1 (divalent metal-ion transporter 1) is a widely expressed metal-ion transporter that is vital for intestinal iron absorption and iron utilization by most cell types throughout the body, including erythroid precursors. Iron 133-137 solute carrier family 11 member 2 Homo sapiens 0-4 17109629-1 2007 DMT1 (divalent metal-ion transporter 1) is a widely expressed metal-ion transporter that is vital for intestinal iron absorption and iron utilization by most cell types throughout the body, including erythroid precursors. Iron 133-137 solute carrier family 11 member 2 Homo sapiens 6-38 16737442-1 2006 DMT1 (divalent metal transporter; also known as SLC11A2, DCT1 or Nramp2) is responsible for ferrous iron uptake in the duodenum, iron exit from endosomes during the transferrin cycle and some transferrin-independent iron uptake in many cells. Iron 100-104 solute carrier family 11 member 2 Homo sapiens 0-4 20020877-6 2007 For example, dietary iron uptake across the intestinal duodenal mucosa is mediated by an intramembrane divalent metal transporter 1 (DMT1), and cellular iron efflux involves ferroportin, the only known iron exporter. Iron 21-25 solute carrier family 11 member 2 Homo sapiens 103-131 20020877-6 2007 For example, dietary iron uptake across the intestinal duodenal mucosa is mediated by an intramembrane divalent metal transporter 1 (DMT1), and cellular iron efflux involves ferroportin, the only known iron exporter. Iron 21-25 solute carrier family 11 member 2 Homo sapiens 133-137 17236123-1 2007 It is hypothesized that a homozygous C282Y mutation of the HFE gene prohibits the assembly of the transferrin-receptor 1 (TFR1) with the divalent metal transporter (DMT1) as the main iron update complex in hepatocytes membrane. Iron 183-187 solute carrier family 11 member 2 Homo sapiens 165-169 17215883-5 2006 Several isoforms of NRAMP2 (SLC11A2, DMT1, DCT1) are expressed ubiquitously in recycling endosomes or specifically at the apical membrane of epithelial cells in intestine and kidneys, and can contribute to iron overload, whereas mutations impairing NRAMP2 function cause a form of congenital microcytic hypochromic anemia. Iron 206-210 solute carrier family 11 member 2 Homo sapiens 20-26 17215883-5 2006 Several isoforms of NRAMP2 (SLC11A2, DMT1, DCT1) are expressed ubiquitously in recycling endosomes or specifically at the apical membrane of epithelial cells in intestine and kidneys, and can contribute to iron overload, whereas mutations impairing NRAMP2 function cause a form of congenital microcytic hypochromic anemia. Iron 206-210 solute carrier family 11 member 2 Homo sapiens 28-35 17215883-5 2006 Several isoforms of NRAMP2 (SLC11A2, DMT1, DCT1) are expressed ubiquitously in recycling endosomes or specifically at the apical membrane of epithelial cells in intestine and kidneys, and can contribute to iron overload, whereas mutations impairing NRAMP2 function cause a form of congenital microcytic hypochromic anemia. Iron 206-210 solute carrier family 11 member 2 Homo sapiens 37-41 17215883-5 2006 Several isoforms of NRAMP2 (SLC11A2, DMT1, DCT1) are expressed ubiquitously in recycling endosomes or specifically at the apical membrane of epithelial cells in intestine and kidneys, and can contribute to iron overload, whereas mutations impairing NRAMP2 function cause a form of congenital microcytic hypochromic anemia. Iron 206-210 solute carrier family 11 member 2 Homo sapiens 43-47 16737442-1 2006 DMT1 (divalent metal transporter; also known as SLC11A2, DCT1 or Nramp2) is responsible for ferrous iron uptake in the duodenum, iron exit from endosomes during the transferrin cycle and some transferrin-independent iron uptake in many cells. Iron 100-104 solute carrier family 11 member 2 Homo sapiens 48-55 16737442-1 2006 DMT1 (divalent metal transporter; also known as SLC11A2, DCT1 or Nramp2) is responsible for ferrous iron uptake in the duodenum, iron exit from endosomes during the transferrin cycle and some transferrin-independent iron uptake in many cells. Iron 100-104 solute carrier family 11 member 2 Homo sapiens 57-61 16737442-1 2006 DMT1 (divalent metal transporter; also known as SLC11A2, DCT1 or Nramp2) is responsible for ferrous iron uptake in the duodenum, iron exit from endosomes during the transferrin cycle and some transferrin-independent iron uptake in many cells. Iron 100-104 solute carrier family 11 member 2 Homo sapiens 65-71 16737442-1 2006 DMT1 (divalent metal transporter; also known as SLC11A2, DCT1 or Nramp2) is responsible for ferrous iron uptake in the duodenum, iron exit from endosomes during the transferrin cycle and some transferrin-independent iron uptake in many cells. Iron 129-133 solute carrier family 11 member 2 Homo sapiens 0-4 16737442-1 2006 DMT1 (divalent metal transporter; also known as SLC11A2, DCT1 or Nramp2) is responsible for ferrous iron uptake in the duodenum, iron exit from endosomes during the transferrin cycle and some transferrin-independent iron uptake in many cells. Iron 129-133 solute carrier family 11 member 2 Homo sapiens 48-55 16737442-1 2006 DMT1 (divalent metal transporter; also known as SLC11A2, DCT1 or Nramp2) is responsible for ferrous iron uptake in the duodenum, iron exit from endosomes during the transferrin cycle and some transferrin-independent iron uptake in many cells. Iron 129-133 solute carrier family 11 member 2 Homo sapiens 57-61 16737442-1 2006 DMT1 (divalent metal transporter; also known as SLC11A2, DCT1 or Nramp2) is responsible for ferrous iron uptake in the duodenum, iron exit from endosomes during the transferrin cycle and some transferrin-independent iron uptake in many cells. Iron 129-133 solute carrier family 11 member 2 Homo sapiens 65-71 16737442-1 2006 DMT1 (divalent metal transporter; also known as SLC11A2, DCT1 or Nramp2) is responsible for ferrous iron uptake in the duodenum, iron exit from endosomes during the transferrin cycle and some transferrin-independent iron uptake in many cells. Iron 129-133 solute carrier family 11 member 2 Homo sapiens 0-4 16737442-1 2006 DMT1 (divalent metal transporter; also known as SLC11A2, DCT1 or Nramp2) is responsible for ferrous iron uptake in the duodenum, iron exit from endosomes during the transferrin cycle and some transferrin-independent iron uptake in many cells. Iron 129-133 solute carrier family 11 member 2 Homo sapiens 48-55 16737442-1 2006 DMT1 (divalent metal transporter; also known as SLC11A2, DCT1 or Nramp2) is responsible for ferrous iron uptake in the duodenum, iron exit from endosomes during the transferrin cycle and some transferrin-independent iron uptake in many cells. Iron 129-133 solute carrier family 11 member 2 Homo sapiens 57-61 16737442-1 2006 DMT1 (divalent metal transporter; also known as SLC11A2, DCT1 or Nramp2) is responsible for ferrous iron uptake in the duodenum, iron exit from endosomes during the transferrin cycle and some transferrin-independent iron uptake in many cells. Iron 129-133 solute carrier family 11 member 2 Homo sapiens 65-71 16984886-0 2006 Small-molecule screening identifies the selanazal drug ebselen as a potent inhibitor of DMT1-mediated iron uptake. Iron 102-106 solute carrier family 11 member 2 Homo sapiens 88-92 16984886-1 2006 HEK293T cells overexpressing divalent metal transporter-1 (DMT1) were established to screen for small-molecule inhibitors of iron uptake. Iron 125-129 solute carrier family 11 member 2 Homo sapiens 29-57 16984886-1 2006 HEK293T cells overexpressing divalent metal transporter-1 (DMT1) were established to screen for small-molecule inhibitors of iron uptake. Iron 125-129 solute carrier family 11 member 2 Homo sapiens 59-63 16707273-0 2006 Functional role of DMT1 in transferrin-independent iron uptake by human hepatocyte and hepatocellular carcinoma cell, HLF. Iron 51-55 solute carrier family 11 member 2 Homo sapiens 19-23 16908409-4 2006 PAP7 in turn binds to the divalent metal transporter (DMT1), an iron import channel. Iron 64-68 solute carrier family 11 member 2 Homo sapiens 54-58 16908409-5 2006 We have identified a signaling cascade in neurons whereby stimulation of NMDA receptors activates nNOS, leading to S-nitrosylation and activation of Dexras1, which, via PAP7 and DMT1, physiologically induces iron uptake. Iron 208-212 solute carrier family 11 member 2 Homo sapiens 178-182 16908409-6 2006 As selective iron chelation prevents NMDA neurotoxicity in cortical cultures, the NMDA-NO-Dexras1-PAP7-DMT1-iron uptake signaling cascade also appears to mediate NMDA neurotoxicity. Iron 108-112 solute carrier family 11 member 2 Homo sapiens 103-107 16707273-2 2006 Dietary iron is transported into intestinal enterocytes by divalent metal transporter 1 (DMT1), which also transports iron from transferrin receptor 1 (TfR1)-mediated recycling endosome to intracytoplasm. Iron 8-12 solute carrier family 11 member 2 Homo sapiens 59-87 16707273-2 2006 Dietary iron is transported into intestinal enterocytes by divalent metal transporter 1 (DMT1), which also transports iron from transferrin receptor 1 (TfR1)-mediated recycling endosome to intracytoplasm. Iron 8-12 solute carrier family 11 member 2 Homo sapiens 89-93 16707273-2 2006 Dietary iron is transported into intestinal enterocytes by divalent metal transporter 1 (DMT1), which also transports iron from transferrin receptor 1 (TfR1)-mediated recycling endosome to intracytoplasm. Iron 118-122 solute carrier family 11 member 2 Homo sapiens 59-87 16707273-2 2006 Dietary iron is transported into intestinal enterocytes by divalent metal transporter 1 (DMT1), which also transports iron from transferrin receptor 1 (TfR1)-mediated recycling endosome to intracytoplasm. Iron 118-122 solute carrier family 11 member 2 Homo sapiens 89-93 16707273-3 2006 We made an antiserum against human DMT1 protein derived from mRNA with the iron responsive element (IRE). Iron 75-79 solute carrier family 11 member 2 Homo sapiens 35-39 16707273-5 2006 In addition, iron-depleted HLF increased membrane expression of DMT1, suggesting that the intracellular iron concentration regulated the DMT1 expression in hepatocytes via the iron regulatory protein (IRP)/IRE system. Iron 13-17 solute carrier family 11 member 2 Homo sapiens 64-68 16707273-5 2006 In addition, iron-depleted HLF increased membrane expression of DMT1, suggesting that the intracellular iron concentration regulated the DMT1 expression in hepatocytes via the iron regulatory protein (IRP)/IRE system. Iron 13-17 solute carrier family 11 member 2 Homo sapiens 137-141 16707273-5 2006 In addition, iron-depleted HLF increased membrane expression of DMT1, suggesting that the intracellular iron concentration regulated the DMT1 expression in hepatocytes via the iron regulatory protein (IRP)/IRE system. Iron 104-108 solute carrier family 11 member 2 Homo sapiens 64-68 16707273-5 2006 In addition, iron-depleted HLF increased membrane expression of DMT1, suggesting that the intracellular iron concentration regulated the DMT1 expression in hepatocytes via the iron regulatory protein (IRP)/IRE system. Iron 104-108 solute carrier family 11 member 2 Homo sapiens 137-141 16584902-0 2006 A novel R416C mutation in human DMT1 (SLC11A2) displays pleiotropic effects on function and causes microcytic anemia and hepatic iron overload. Iron 129-133 solute carrier family 11 member 2 Homo sapiens 32-36 16449358-1 2006 The H+-coupled polyligand transport protein divalent metal transporter 1 (DMT1) plays a key role in mammalian iron homeostasis. Iron 110-114 solute carrier family 11 member 2 Homo sapiens 44-72 16449358-1 2006 The H+-coupled polyligand transport protein divalent metal transporter 1 (DMT1) plays a key role in mammalian iron homeostasis. Iron 110-114 solute carrier family 11 member 2 Homo sapiens 74-78 16584902-0 2006 A novel R416C mutation in human DMT1 (SLC11A2) displays pleiotropic effects on function and causes microcytic anemia and hepatic iron overload. Iron 129-133 solute carrier family 11 member 2 Homo sapiens 38-45 16584902-1 2006 A patient suffering from microcytic anemia and hepatic iron overload was found to be compound heterozygote for polymorphisms in the iron transporter DMT1 (Nramp2, SLC11A2), including a 3-bp deletion (DMT1(delCTT)) in intron 4 that partially impairs splicing and an amino acid substitution (DMT1(C1246T), R416C) at a conserved residue in transmembrane domain 9 of the protein. Iron 55-59 solute carrier family 11 member 2 Homo sapiens 149-153 16584902-1 2006 A patient suffering from microcytic anemia and hepatic iron overload was found to be compound heterozygote for polymorphisms in the iron transporter DMT1 (Nramp2, SLC11A2), including a 3-bp deletion (DMT1(delCTT)) in intron 4 that partially impairs splicing and an amino acid substitution (DMT1(C1246T), R416C) at a conserved residue in transmembrane domain 9 of the protein. Iron 55-59 solute carrier family 11 member 2 Homo sapiens 155-161 16584902-1 2006 A patient suffering from microcytic anemia and hepatic iron overload was found to be compound heterozygote for polymorphisms in the iron transporter DMT1 (Nramp2, SLC11A2), including a 3-bp deletion (DMT1(delCTT)) in intron 4 that partially impairs splicing and an amino acid substitution (DMT1(C1246T), R416C) at a conserved residue in transmembrane domain 9 of the protein. Iron 55-59 solute carrier family 11 member 2 Homo sapiens 163-170 16584902-1 2006 A patient suffering from microcytic anemia and hepatic iron overload was found to be compound heterozygote for polymorphisms in the iron transporter DMT1 (Nramp2, SLC11A2), including a 3-bp deletion (DMT1(delCTT)) in intron 4 that partially impairs splicing and an amino acid substitution (DMT1(C1246T), R416C) at a conserved residue in transmembrane domain 9 of the protein. Iron 55-59 solute carrier family 11 member 2 Homo sapiens 200-204 16584902-1 2006 A patient suffering from microcytic anemia and hepatic iron overload was found to be compound heterozygote for polymorphisms in the iron transporter DMT1 (Nramp2, SLC11A2), including a 3-bp deletion (DMT1(delCTT)) in intron 4 that partially impairs splicing and an amino acid substitution (DMT1(C1246T), R416C) at a conserved residue in transmembrane domain 9 of the protein. Iron 55-59 solute carrier family 11 member 2 Homo sapiens 200-204 16584902-5 2006 We propose that DMT1(C1246T) (R416C) represents a complete loss-of-function, and that a quantitative reduction in DMT1 expression is the cause of the microcytic anemia and iron overload in the patient. Iron 172-176 solute carrier family 11 member 2 Homo sapiens 114-118 16651563-1 2006 Nramp (Slc11a1) genes in mammals are associated with the transport of iron and other divalent cations; Nramp1 in macrophages involved in the innate immune response against intracellular pathogens, and Nramp2 with duodenal iron uptake and the transferrin-transferrin-receptor pathway of iron assimilation. Iron 70-74 solute carrier family 11 member 2 Homo sapiens 201-207 16651563-1 2006 Nramp (Slc11a1) genes in mammals are associated with the transport of iron and other divalent cations; Nramp1 in macrophages involved in the innate immune response against intracellular pathogens, and Nramp2 with duodenal iron uptake and the transferrin-transferrin-receptor pathway of iron assimilation. Iron 222-226 solute carrier family 11 member 2 Homo sapiens 201-207 16651563-1 2006 Nramp (Slc11a1) genes in mammals are associated with the transport of iron and other divalent cations; Nramp1 in macrophages involved in the innate immune response against intracellular pathogens, and Nramp2 with duodenal iron uptake and the transferrin-transferrin-receptor pathway of iron assimilation. Iron 222-226 solute carrier family 11 member 2 Homo sapiens 201-207 16651563-7 2006 Expression within the gut was of significant interest, as mammalian Nramp2 in the gut plays a primary role in the acquisition of dietary iron. Iron 137-141 solute carrier family 11 member 2 Homo sapiens 68-74 16474007-2 2006 Iron absorption across the brush-border membrane requires divalent metal transporter 1 (DMT1), whereas ferroportin (FPN) and hephaestin are required for exit across the basolateral membrane. Iron 0-4 solute carrier family 11 member 2 Homo sapiens 58-86 16272461-2 2006 We tested the hypothesis that the cellular response to asbestos includes the transport and sequestration of this iron through (1) generation of superoxide for ferrireduction, (2) up-regulation of divalent metal transporter-1 (DMT1) for intracellular transport of Fe2+, and (3) increased production of cellular ferritin where the metal is stored in a catalytically less reactive state. Iron 113-117 solute carrier family 11 member 2 Homo sapiens 196-224 16272461-2 2006 We tested the hypothesis that the cellular response to asbestos includes the transport and sequestration of this iron through (1) generation of superoxide for ferrireduction, (2) up-regulation of divalent metal transporter-1 (DMT1) for intracellular transport of Fe2+, and (3) increased production of cellular ferritin where the metal is stored in a catalytically less reactive state. Iron 113-117 solute carrier family 11 member 2 Homo sapiens 226-230 16759552-2 2006 The objective of the present study was to ascertain whether the competitive blockade of DMT1 by the administration of high doses of oral Mg2+ reduces iron absorption in patients homozygous for the C282Y mutation. Iron 150-154 solute carrier family 11 member 2 Homo sapiens 88-92 16474007-2 2006 Iron absorption across the brush-border membrane requires divalent metal transporter 1 (DMT1), whereas ferroportin (FPN) and hephaestin are required for exit across the basolateral membrane. Iron 0-4 solute carrier family 11 member 2 Homo sapiens 88-92 16474007-5 2006 With iron feeding, DMT1 undergoes endocytosis and FPN translocates from the apical cytosol to the basolateral membrane. Iron 5-9 solute carrier family 11 member 2 Homo sapiens 19-23 16267047-0 2006 Two isoforms of a divalent metal transporter (DMT1) in Schistosoma mansoni suggest a surface-associated pathway for iron absorption in schistosomes. Iron 116-120 solute carrier family 11 member 2 Homo sapiens 46-50 16431145-9 2006 Altering luminal saline pH from 7.0 to 5.5 did not affect ferric or ferrous iron uptake, suggesting that if iron is entering via DMT1 in marine fish intestine this transporter works efficiently under circumneutral conditions. Iron 108-112 solute carrier family 11 member 2 Homo sapiens 129-133 16475818-1 2006 The metal transporter DMT1 (Slc11a2) plays a vital role in iron metabolism. Iron 59-63 solute carrier family 11 member 2 Homo sapiens 22-26 16475818-1 2006 The metal transporter DMT1 (Slc11a2) plays a vital role in iron metabolism. Iron 59-63 solute carrier family 11 member 2 Homo sapiens 28-35 16475818-2 2006 Alternative splicing of the 3" exon generates two DMT1 isoforms with different C-terminal protein sequences and a 3" untranslated region harboring (isoform I, +IRE) or not (isoform II, -IRE), an iron-responsive element. Iron 195-199 solute carrier family 11 member 2 Homo sapiens 50-54 16232120-2 2006 Protein and mRNA expression for the +/-IRE (iron response element) forms of DMT1, but not the 1A isoform, were down-regulated within the first few hours upon removal of RA, at which time the cells began to differentiate. Iron 44-48 solute carrier family 11 member 2 Homo sapiens 76-80 16267047-6 2006 An iron-responsive element, present at the 3"-untranslated region of many DMT1 molecules, is not present in schistosome mRNAs studied here. Iron 3-7 solute carrier family 11 member 2 Homo sapiens 74-78 17134988-10 2006 The inflammatory cells in the endomysium were reactive for CD68, cytosolic ferritin, and the DMT1 isoform(s) translated from messenger ribonucleic acids containing iron-responsive elements (DMT1+). Iron 164-168 solute carrier family 11 member 2 Homo sapiens 93-97 16629172-3 2006 The apical divalent metal transporter-1 (DMT1) transports ferrous iron from the lumen into the cells, while the basolateral transporter ferroportin extrudes iron from the enterocytes into the circulation. Iron 66-70 solute carrier family 11 member 2 Homo sapiens 11-39 16629172-3 2006 The apical divalent metal transporter-1 (DMT1) transports ferrous iron from the lumen into the cells, while the basolateral transporter ferroportin extrudes iron from the enterocytes into the circulation. Iron 66-70 solute carrier family 11 member 2 Homo sapiens 41-45 16629172-3 2006 The apical divalent metal transporter-1 (DMT1) transports ferrous iron from the lumen into the cells, while the basolateral transporter ferroportin extrudes iron from the enterocytes into the circulation. Iron 157-161 solute carrier family 11 member 2 Homo sapiens 11-39 16629172-3 2006 The apical divalent metal transporter-1 (DMT1) transports ferrous iron from the lumen into the cells, while the basolateral transporter ferroportin extrudes iron from the enterocytes into the circulation. Iron 157-161 solute carrier family 11 member 2 Homo sapiens 41-45 16629172-8 2006 These control sites may be influenced by gene therapeutic approaches; one general therapy for hemochromatosis of different etiologies is the inhibition of DMT1 synthesis by antisense-generating genes, which has been shown to markedly inhibit apical iron uptake by intestinal epithelial cells. Iron 249-253 solute carrier family 11 member 2 Homo sapiens 155-159 17134988-10 2006 The inflammatory cells in the endomysium were reactive for CD68, cytosolic ferritin, and the DMT1 isoform(s) translated from messenger ribonucleic acids containing iron-responsive elements (DMT1+). Iron 164-168 solute carrier family 11 member 2 Homo sapiens 190-194 16160008-0 2006 Microcytic anemia and hepatic iron overload in a child with compound heterozygous mutations in DMT1 (SCL11A2). Iron 30-34 solute carrier family 11 member 2 Homo sapiens 95-99 16091957-1 2006 The H(+) -coupled divalent metal-ion transporter DMT1 serves as both the primary entry point for iron into the body (intestinal brush-border uptake) and the route by which transferrin-associated iron is mobilized from endosomes to cytosol in erythroid precursors and other cells. Iron 97-101 solute carrier family 11 member 2 Homo sapiens 49-53 16160008-1 2006 Divalent metal transporter 1 (DMT1) mediates apical iron uptake in duodenal enterocytes and iron transfer from the transferrin receptor endosomal cycle into the cytosol in erythroid cells. Iron 52-56 solute carrier family 11 member 2 Homo sapiens 0-28 16160008-1 2006 Divalent metal transporter 1 (DMT1) mediates apical iron uptake in duodenal enterocytes and iron transfer from the transferrin receptor endosomal cycle into the cytosol in erythroid cells. Iron 52-56 solute carrier family 11 member 2 Homo sapiens 30-34 16160008-1 2006 Divalent metal transporter 1 (DMT1) mediates apical iron uptake in duodenal enterocytes and iron transfer from the transferrin receptor endosomal cycle into the cytosol in erythroid cells. Iron 92-96 solute carrier family 11 member 2 Homo sapiens 0-28 16160008-1 2006 Divalent metal transporter 1 (DMT1) mediates apical iron uptake in duodenal enterocytes and iron transfer from the transferrin receptor endosomal cycle into the cytosol in erythroid cells. Iron 92-96 solute carrier family 11 member 2 Homo sapiens 30-34 16160008-3 2006 We report the hematologic phenotype of a child, compound heterozygote for 2 DMT1 mutations, who was affected by severe anemia since birth and showed hepatic iron overload. Iron 157-161 solute carrier family 11 member 2 Homo sapiens 76-80 16160008-8 2006 The early onset of iron overload indicates that, as in animal models, DMT1 is dispensable for liver iron uptake, whereas its deficiency in the gut is likely bypassed by the up-regulation of other pathways of iron use. Iron 19-23 solute carrier family 11 member 2 Homo sapiens 70-74 16091957-1 2006 The H(+) -coupled divalent metal-ion transporter DMT1 serves as both the primary entry point for iron into the body (intestinal brush-border uptake) and the route by which transferrin-associated iron is mobilized from endosomes to cytosol in erythroid precursors and other cells. Iron 195-199 solute carrier family 11 member 2 Homo sapiens 49-53 16091957-2 2006 Elucidating the molecular mechanisms of DMT1 will therefore increase our understanding of iron metabolism and the etiology of iron overload disorders. Iron 90-94 solute carrier family 11 member 2 Homo sapiens 40-44 16091957-2 2006 Elucidating the molecular mechanisms of DMT1 will therefore increase our understanding of iron metabolism and the etiology of iron overload disorders. Iron 126-130 solute carrier family 11 member 2 Homo sapiens 40-44 16091957-12 2006 Since plasma membrane expression of DMT1 is upregulated in liver of hemochromatosis patients, this H(+) -uncoupled facilitative Fe(2+) transport via DMT1 can account for the uptake of nontransferrin-bound plasma iron characteristic of iron overload disorders. Iron 212-216 solute carrier family 11 member 2 Homo sapiens 36-40 16091957-12 2006 Since plasma membrane expression of DMT1 is upregulated in liver of hemochromatosis patients, this H(+) -uncoupled facilitative Fe(2+) transport via DMT1 can account for the uptake of nontransferrin-bound plasma iron characteristic of iron overload disorders. Iron 212-216 solute carrier family 11 member 2 Homo sapiens 149-153 16091957-12 2006 Since plasma membrane expression of DMT1 is upregulated in liver of hemochromatosis patients, this H(+) -uncoupled facilitative Fe(2+) transport via DMT1 can account for the uptake of nontransferrin-bound plasma iron characteristic of iron overload disorders. Iron 235-239 solute carrier family 11 member 2 Homo sapiens 36-40 16091957-12 2006 Since plasma membrane expression of DMT1 is upregulated in liver of hemochromatosis patients, this H(+) -uncoupled facilitative Fe(2+) transport via DMT1 can account for the uptake of nontransferrin-bound plasma iron characteristic of iron overload disorders. Iron 235-239 solute carrier family 11 member 2 Homo sapiens 149-153 16298746-7 2005 Increased expression of DMT-1 and IREG-1 was associated with iron accumulation and oxidative stress. Iron 61-65 solute carrier family 11 member 2 Homo sapiens 24-29 16091455-0 2005 Functional consequences of the human DMT1 (SLC11A2) mutation on protein expression and iron uptake. Iron 87-91 solute carrier family 11 member 2 Homo sapiens 37-41 16091455-0 2005 Functional consequences of the human DMT1 (SLC11A2) mutation on protein expression and iron uptake. Iron 87-91 solute carrier family 11 member 2 Homo sapiens 43-50 16091455-6 2005 We hypothesize that the residual protein in hematopoietic cells represents the functional E399D DMT1 variant, but because of its quantitative reduction, the iron uptake activity of DMT1 in the patient"s erythroid cells is severely suppressed. Iron 157-161 solute carrier family 11 member 2 Homo sapiens 181-185 16123094-2 2005 Two isoforms, which differ by the presence (DMT1-IRE) or absence (DMT1-nonIRE) of an iron-responsive element (IRE) in their 3" untranslated region, are implicated in apical iron transport and endosomal iron transport respectively. Iron 85-89 solute carrier family 11 member 2 Homo sapiens 66-70 16123094-2 2005 Two isoforms, which differ by the presence (DMT1-IRE) or absence (DMT1-nonIRE) of an iron-responsive element (IRE) in their 3" untranslated region, are implicated in apical iron transport and endosomal iron transport respectively. Iron 173-177 solute carrier family 11 member 2 Homo sapiens 44-48 16123094-2 2005 Two isoforms, which differ by the presence (DMT1-IRE) or absence (DMT1-nonIRE) of an iron-responsive element (IRE) in their 3" untranslated region, are implicated in apical iron transport and endosomal iron transport respectively. Iron 173-177 solute carrier family 11 member 2 Homo sapiens 66-70 16123094-2 2005 Two isoforms, which differ by the presence (DMT1-IRE) or absence (DMT1-nonIRE) of an iron-responsive element (IRE) in their 3" untranslated region, are implicated in apical iron transport and endosomal iron transport respectively. Iron 173-177 solute carrier family 11 member 2 Homo sapiens 44-48 16123094-2 2005 Two isoforms, which differ by the presence (DMT1-IRE) or absence (DMT1-nonIRE) of an iron-responsive element (IRE) in their 3" untranslated region, are implicated in apical iron transport and endosomal iron transport respectively. Iron 173-177 solute carrier family 11 member 2 Homo sapiens 66-70 16123094-10 2005 Predominant DMT1-IRE isoform expression in placenta suggests an iron-regulatory mechanism reminiscent of that in the adult duodenum. Iron 64-68 solute carrier family 11 member 2 Homo sapiens 12-16 16023393-2 2005 A single patient with severe microcytic anemia and iron overload was recently reported to carry a mutation in exon 12 of DMT1 (1285G>C). Iron 51-55 solute carrier family 11 member 2 Homo sapiens 121-125 15901240-6 2005 TNFalpha mediated an early induction in both iron import and iron export, which were associated with increased DMT-1 and IREG-1 mRNA and protein expression (P<0.05). Iron 45-49 solute carrier family 11 member 2 Homo sapiens 111-116 16023393-0 2005 Functional characterization of the E399D DMT1/NRAMP2/SLC11A2 protein produced by an exon 12 mutation in a patient with microcytic anemia and iron overload. Iron 141-145 solute carrier family 11 member 2 Homo sapiens 41-45 16023393-0 2005 Functional characterization of the E399D DMT1/NRAMP2/SLC11A2 protein produced by an exon 12 mutation in a patient with microcytic anemia and iron overload. Iron 141-145 solute carrier family 11 member 2 Homo sapiens 46-52 16283525-3 2005 Here we verify the ability of nickel to enter the cell via the divalent metal ion transporter 1 (DMT1) and disturb cellular iron homeostasis. Iron 124-128 solute carrier family 11 member 2 Homo sapiens 97-101 15880641-4 2005 Several iron transporters and regulators were recently characterized, including DMT1 and ferroportin/Ireg1 that transport iron through membranes, and HFE that regulates TfR-mediated iron uptake. Iron 8-12 solute carrier family 11 member 2 Homo sapiens 80-84 16023393-0 2005 Functional characterization of the E399D DMT1/NRAMP2/SLC11A2 protein produced by an exon 12 mutation in a patient with microcytic anemia and iron overload. Iron 141-145 solute carrier family 11 member 2 Homo sapiens 53-60 16023393-1 2005 DMT1 (Nramp2, Slc11a2) mediates iron uptake at the intestinal brush border and across the membrane of acidified endosomes. Iron 32-36 solute carrier family 11 member 2 Homo sapiens 0-4 16023393-1 2005 DMT1 (Nramp2, Slc11a2) mediates iron uptake at the intestinal brush border and across the membrane of acidified endosomes. Iron 32-36 solute carrier family 11 member 2 Homo sapiens 6-12 16023393-1 2005 DMT1 (Nramp2, Slc11a2) mediates iron uptake at the intestinal brush border and across the membrane of acidified endosomes. Iron 32-36 solute carrier family 11 member 2 Homo sapiens 14-21 15880641-4 2005 Several iron transporters and regulators were recently characterized, including DMT1 and ferroportin/Ireg1 that transport iron through membranes, and HFE that regulates TfR-mediated iron uptake. Iron 122-126 solute carrier family 11 member 2 Homo sapiens 80-84 15929194-3 2005 RESULTS: Whereas in hereditary hemochromatosis patients without CD, DMT1 expression was up-regulated leading to excessive uptake of iron, we identified a significant reduction in protein and mRNA expression of DMT1 as a compensatory mechanism in this patient with HH and CD. Iron 132-136 solute carrier family 11 member 2 Homo sapiens 68-72 16085548-1 2005 In the last few years, the field of iron metabolism has exploded with the discovery of many new proteins including ferroportin, hephaestin, hepcidin, duodenal cytochrome b and the topic of this review, divalent metal ion transporter 1 (DMT1). Iron 36-40 solute carrier family 11 member 2 Homo sapiens 202-234 16085548-1 2005 In the last few years, the field of iron metabolism has exploded with the discovery of many new proteins including ferroportin, hephaestin, hepcidin, duodenal cytochrome b and the topic of this review, divalent metal ion transporter 1 (DMT1). Iron 36-40 solute carrier family 11 member 2 Homo sapiens 236-240 16085548-2 2005 DMT1 functions in transport of ferrous iron, and some, but not all divalent metal ions across the plasma membrane and/or out of the endosomal compartment. Iron 39-43 solute carrier family 11 member 2 Homo sapiens 0-4 16085548-4 2005 Rodents with defects in iron absorption and utilization were identified long before it was determined that the defect was due to a single nucleotide mutation in DMT1. Iron 24-28 solute carrier family 11 member 2 Homo sapiens 161-165 16085548-5 2005 Study of these animals reveals that transport of iron and other divalent metal ions by DMT1 is pH dependent, but the exact manner in which pH exerts its effect is unknown. Iron 49-53 solute carrier family 11 member 2 Homo sapiens 87-91 16085548-9 2005 At least one signal for DMT1 expression appears to be intracellular iron status, however, other, as yet undefined signals may also contribute to DMT1 expression. Iron 68-72 solute carrier family 11 member 2 Homo sapiens 24-28 16085548-10 2005 Interestingly, DMT1 function may differ subtly between humans and other animals; the spontaneous DMT1 mutation found in mice and rats appears to limit iron uptake in the intestine and iron utilization in red cell precursors, whereas the only known human mutation has its primary effect on iron utilization by erythroid cells. Iron 151-155 solute carrier family 11 member 2 Homo sapiens 15-19 16085548-10 2005 Interestingly, DMT1 function may differ subtly between humans and other animals; the spontaneous DMT1 mutation found in mice and rats appears to limit iron uptake in the intestine and iron utilization in red cell precursors, whereas the only known human mutation has its primary effect on iron utilization by erythroid cells. Iron 151-155 solute carrier family 11 member 2 Homo sapiens 97-101 16085548-10 2005 Interestingly, DMT1 function may differ subtly between humans and other animals; the spontaneous DMT1 mutation found in mice and rats appears to limit iron uptake in the intestine and iron utilization in red cell precursors, whereas the only known human mutation has its primary effect on iron utilization by erythroid cells. Iron 184-188 solute carrier family 11 member 2 Homo sapiens 15-19 16085548-10 2005 Interestingly, DMT1 function may differ subtly between humans and other animals; the spontaneous DMT1 mutation found in mice and rats appears to limit iron uptake in the intestine and iron utilization in red cell precursors, whereas the only known human mutation has its primary effect on iron utilization by erythroid cells. Iron 184-188 solute carrier family 11 member 2 Homo sapiens 97-101 16085548-10 2005 Interestingly, DMT1 function may differ subtly between humans and other animals; the spontaneous DMT1 mutation found in mice and rats appears to limit iron uptake in the intestine and iron utilization in red cell precursors, whereas the only known human mutation has its primary effect on iron utilization by erythroid cells. Iron 184-188 solute carrier family 11 member 2 Homo sapiens 15-19 16085548-10 2005 Interestingly, DMT1 function may differ subtly between humans and other animals; the spontaneous DMT1 mutation found in mice and rats appears to limit iron uptake in the intestine and iron utilization in red cell precursors, whereas the only known human mutation has its primary effect on iron utilization by erythroid cells. Iron 184-188 solute carrier family 11 member 2 Homo sapiens 97-101 16085548-11 2005 The importance of DMT1 function at the level of the whole organism and the individual cell and mechanisms of its regulation on a molecular scale are only beginning to be understood; an appreciation of these process will lead to an understanding of the role of iron in various cellular processes and improved treatments for both anemia and iron-overload. Iron 260-264 solute carrier family 11 member 2 Homo sapiens 18-22 15749739-1 2005 Regulation of the metal transport protein divalent metal transporter-1 (DMT1) may contribute to the uptake and detoxification of iron by cells resident in the respiratory tract. Iron 129-133 solute carrier family 11 member 2 Homo sapiens 42-70 15749739-1 2005 Regulation of the metal transport protein divalent metal transporter-1 (DMT1) may contribute to the uptake and detoxification of iron by cells resident in the respiratory tract. Iron 129-133 solute carrier family 11 member 2 Homo sapiens 72-76 15878745-11 2005 Whilst the consequence of altered DMT1 expression on renal iron handling and oxidant damage remains to be determined, the attenuation of the putative lysosomal iron exit pathway in proximal tubules could potentially explain lysosomal iron accumulation reported in human diabetes and STZ-diabetic animals. Iron 59-63 solute carrier family 11 member 2 Homo sapiens 34-38 15896335-3 2005 The iron transporter divalent metal transporter-1 (DMT-1) of enterocytes is responsible for iron uptake from the intestinal lumen; iron is further extruded into the blood by the basolateral transporter ferroportin-1. Iron 4-8 solute carrier family 11 member 2 Homo sapiens 21-49 15896335-3 2005 The iron transporter divalent metal transporter-1 (DMT-1) of enterocytes is responsible for iron uptake from the intestinal lumen; iron is further extruded into the blood by the basolateral transporter ferroportin-1. Iron 4-8 solute carrier family 11 member 2 Homo sapiens 51-56 15896335-3 2005 The iron transporter divalent metal transporter-1 (DMT-1) of enterocytes is responsible for iron uptake from the intestinal lumen; iron is further extruded into the blood by the basolateral transporter ferroportin-1. Iron 92-96 solute carrier family 11 member 2 Homo sapiens 21-49 15896335-3 2005 The iron transporter divalent metal transporter-1 (DMT-1) of enterocytes is responsible for iron uptake from the intestinal lumen; iron is further extruded into the blood by the basolateral transporter ferroportin-1. Iron 92-96 solute carrier family 11 member 2 Homo sapiens 51-56 15896335-4 2005 A therapeutic approach for HH could start with a long-term reduction of iron transport by reduction of DMT-1 levels. Iron 72-76 solute carrier family 11 member 2 Homo sapiens 103-108 15896335-5 2005 We designed an AAV vector coding for a short antisense RNA (AAV-DMT-1-AS) against DMT-1, which reduced iron uptake by 50-60% in human intestinal cells (Caco-2). Iron 103-107 solute carrier family 11 member 2 Homo sapiens 64-69 15896335-5 2005 We designed an AAV vector coding for a short antisense RNA (AAV-DMT-1-AS) against DMT-1, which reduced iron uptake by 50-60% in human intestinal cells (Caco-2). Iron 103-107 solute carrier family 11 member 2 Homo sapiens 82-87 15896335-7 2005 DMT-1 mRNA levels returned to normal at higher infection levels, indicating that an additional mechanism of mRNA occupation, able to block DMT-1 translation and to avoid feedback regulation by iron responsive elements (IRE), also exists. Iron 193-197 solute carrier family 11 member 2 Homo sapiens 0-5 15459009-0 2005 Identification of a human mutation of DMT1 in a patient with microcytic anemia and iron overload. Iron 83-87 solute carrier family 11 member 2 Homo sapiens 38-42 15792797-1 2005 A number of regulatory factors including dietary iron levels can dramatically alter the expression of the intestinal iron transporter DMT1. Iron 49-53 solute carrier family 11 member 2 Homo sapiens 134-138 15792797-2 2005 Here we show that Caco-2 cells exposed to iron for 4h exhibited a significant decrease in plasma membrane DMT1 protein, though total cellular DMT1 levels were unaltered. Iron 42-46 solute carrier family 11 member 2 Homo sapiens 106-110 15792797-2 2005 Here we show that Caco-2 cells exposed to iron for 4h exhibited a significant decrease in plasma membrane DMT1 protein, though total cellular DMT1 levels were unaltered. Iron 42-46 solute carrier family 11 member 2 Homo sapiens 142-146 15792797-3 2005 Following biotinylation of cell surface proteins, there was a significant increase in intracellular biotin-labelled DMT1 in iron-exposed cells. Iron 124-128 solute carrier family 11 member 2 Homo sapiens 116-120 15792797-4 2005 Furthermore, iron-treatment increased levels of DMT1 co-localised with LAMP1, suggesting that the initial response of intestinal epithelial cells to iron involves internalisation and targeting of DMT1 transporter protein towards a late endosomal/lysosomal compartment. Iron 13-17 solute carrier family 11 member 2 Homo sapiens 48-52 15792797-4 2005 Furthermore, iron-treatment increased levels of DMT1 co-localised with LAMP1, suggesting that the initial response of intestinal epithelial cells to iron involves internalisation and targeting of DMT1 transporter protein towards a late endosomal/lysosomal compartment. Iron 13-17 solute carrier family 11 member 2 Homo sapiens 196-200 15792797-4 2005 Furthermore, iron-treatment increased levels of DMT1 co-localised with LAMP1, suggesting that the initial response of intestinal epithelial cells to iron involves internalisation and targeting of DMT1 transporter protein towards a late endosomal/lysosomal compartment. Iron 149-153 solute carrier family 11 member 2 Homo sapiens 48-52 15686373-1 2005 The mammalian iron transporter, divalent metal transporter (DMT1), is a 12-transmembrane domain integral protein, responsible for dietary iron uptake in the duodenum and iron acquisition from transferrin in peripheral tissues. Iron 14-18 solute carrier family 11 member 2 Homo sapiens 60-64 15686373-1 2005 The mammalian iron transporter, divalent metal transporter (DMT1), is a 12-transmembrane domain integral protein, responsible for dietary iron uptake in the duodenum and iron acquisition from transferrin in peripheral tissues. Iron 138-142 solute carrier family 11 member 2 Homo sapiens 60-64 15459009-1 2005 Divalent metal transporter 1 (DMT1) is a transmembrane protein crucial for duodenal iron absorption and erythroid iron transport. Iron 84-88 solute carrier family 11 member 2 Homo sapiens 0-28 15459009-1 2005 Divalent metal transporter 1 (DMT1) is a transmembrane protein crucial for duodenal iron absorption and erythroid iron transport. Iron 84-88 solute carrier family 11 member 2 Homo sapiens 30-34 15459009-1 2005 Divalent metal transporter 1 (DMT1) is a transmembrane protein crucial for duodenal iron absorption and erythroid iron transport. Iron 114-118 solute carrier family 11 member 2 Homo sapiens 0-28 15459009-1 2005 Divalent metal transporter 1 (DMT1) is a transmembrane protein crucial for duodenal iron absorption and erythroid iron transport. Iron 114-118 solute carrier family 11 member 2 Homo sapiens 30-34 15459009-4 2005 We report here the first human mutation of DMT1 identified in a female with severe hypochromic microcytic anemia and iron overload. Iron 117-121 solute carrier family 11 member 2 Homo sapiens 43-47 15128303-1 2004 The divalent metal transporter (DMT1) is a 12-transmembrane domain protein responsible for dietary iron uptake in the duodenum and iron acquisition from transferrin in peripheral tissues. Iron 99-103 solute carrier family 11 member 2 Homo sapiens 32-36 15247188-0 2004 Increased DMT1 but not IREG1 or HFE mRNA following iron depletion therapy in hereditary haemochromatosis. Iron 51-55 solute carrier family 11 member 2 Homo sapiens 10-14 16240664-3 2005 Iron absorption is regulated by divalent metal ion transporter 1 (DMT1) and ferroportin 1 (FPN1). Iron 0-4 solute carrier family 11 member 2 Homo sapiens 32-64 16240664-3 2005 Iron absorption is regulated by divalent metal ion transporter 1 (DMT1) and ferroportin 1 (FPN1). Iron 0-4 solute carrier family 11 member 2 Homo sapiens 66-70 15178582-4 2004 This was accompanied by a decrease in both the protein and the mRNA expression of the iron-response element containing variant of the divalent metal transporter (DMT1[+IRE]). Iron 86-90 solute carrier family 11 member 2 Homo sapiens 162-166 14988066-10 2004 These findings strongly suggest a relationship between Cu and Fe homeostasis in Hep-G2 cells in which Cu accumulation downregulates DMT1 activity. Iron 62-64 solute carrier family 11 member 2 Homo sapiens 132-136 15128303-1 2004 The divalent metal transporter (DMT1) is a 12-transmembrane domain protein responsible for dietary iron uptake in the duodenum and iron acquisition from transferrin in peripheral tissues. Iron 131-135 solute carrier family 11 member 2 Homo sapiens 32-36 14770366-4 2004 Intestinal iron absorption by mature duodenal enterocytes requires Fe (III) iron reduction by Dcytb and Fe (II) iron transport through apical membranes by the iron transporter Nramp2/DMT1. Iron 11-15 solute carrier family 11 member 2 Homo sapiens 176-182 15105261-7 2004 Indeed, recent mechanistic studies found that the intestinal transporter for nonheme iron, divalent metal transporter 1 (DMT1), mediates the transport of Pb and Cd. Iron 85-89 solute carrier family 11 member 2 Homo sapiens 91-119 15105261-7 2004 Indeed, recent mechanistic studies found that the intestinal transporter for nonheme iron, divalent metal transporter 1 (DMT1), mediates the transport of Pb and Cd. Iron 85-89 solute carrier family 11 member 2 Homo sapiens 121-125 15105261-8 2004 DMT1 is regulated, in part, by dietary iron, and chemical species of Cd and Pb that are transported by DMT1 would be made available through digestion and are also found in plasma. Iron 39-43 solute carrier family 11 member 2 Homo sapiens 0-4 15105261-9 2004 Accordingly, the involvement of DMT1 in metal uptake offers a mechanistic explanation for why an iron-deficient diet is a risk factor for Pb and Cd poisoning. Iron 97-101 solute carrier family 11 member 2 Homo sapiens 32-36 14530973-7 2004 DMT1 is up-regulated dramatically in the intestine by dietary iron restriction and, despite high serum iron levels, is not appropriately down-regulated in hereditary hemochromatosis. Iron 62-66 solute carrier family 11 member 2 Homo sapiens 0-4 14530973-7 2004 DMT1 is up-regulated dramatically in the intestine by dietary iron restriction and, despite high serum iron levels, is not appropriately down-regulated in hereditary hemochromatosis. Iron 103-107 solute carrier family 11 member 2 Homo sapiens 0-4 15014911-3 2004 The functional properties and localization in plasma membrane of cells and endosomes suggest an important role for the divalent metal transporter DMT1 (also known as DCT1 and Nramp2) in iron transport and cellular iron homeostasis. Iron 186-190 solute carrier family 11 member 2 Homo sapiens 146-150 15014911-3 2004 The functional properties and localization in plasma membrane of cells and endosomes suggest an important role for the divalent metal transporter DMT1 (also known as DCT1 and Nramp2) in iron transport and cellular iron homeostasis. Iron 186-190 solute carrier family 11 member 2 Homo sapiens 166-170 15014911-3 2004 The functional properties and localization in plasma membrane of cells and endosomes suggest an important role for the divalent metal transporter DMT1 (also known as DCT1 and Nramp2) in iron transport and cellular iron homeostasis. Iron 186-190 solute carrier family 11 member 2 Homo sapiens 175-181 15014911-3 2004 The functional properties and localization in plasma membrane of cells and endosomes suggest an important role for the divalent metal transporter DMT1 (also known as DCT1 and Nramp2) in iron transport and cellular iron homeostasis. Iron 214-218 solute carrier family 11 member 2 Homo sapiens 146-150 15081108-4 2004 With regard to iron metabolism, we showed that HEPH, SLC11A2, SLC11A3, and TF are significantly up-regulated, while ATP7B and SLC39A1 (and SFT) are down-regulated and ACO1, dCYTb, FECH, and FTH1 show constant expression. Iron 15-19 solute carrier family 11 member 2 Homo sapiens 53-60 15105252-2 2004 The concurrent expression of the ferrous iron transporter, divalent metal transporter I (DMT1), in neurons suggests that the internalization of transferrin is followed by detachment of iron within recycling endosomes and transport into the cytosol via DMT1. Iron 41-45 solute carrier family 11 member 2 Homo sapiens 89-93 15105252-2 2004 The concurrent expression of the ferrous iron transporter, divalent metal transporter I (DMT1), in neurons suggests that the internalization of transferrin is followed by detachment of iron within recycling endosomes and transport into the cytosol via DMT1. Iron 41-45 solute carrier family 11 member 2 Homo sapiens 252-256 15105252-3 2004 To enable DMT1-mediated export of iron from the endosome to the cytosol, ferric iron must be reduced to its ferrous form, which could be mediated by a ferric reductase. Iron 34-38 solute carrier family 11 member 2 Homo sapiens 10-14 15105252-10 2004 Pharmacological blockage of the transferrin receptor/DMT1-mediated uptake could be a target to prevent further iron uptake. Iron 111-115 solute carrier family 11 member 2 Homo sapiens 53-57 14768003-3 2004 The current study investigated the duodenal expression of the iron transport molecules divalent metal transporter 1 (DMT1 [IRE]), iron-regulated gene 1 (Ireg1 [ferroportin]), hephaestin, and duodenal cytochrome b (Dyctb) in 46 patients with cirrhosis and 20 control subjects. Iron 62-66 solute carrier family 11 member 2 Homo sapiens 117-121 14768003-5 2004 Expression of DMT1 and Ireg1 was increased 1.5 to 3-fold in subjects with cirrhosis compared with iron-replete control subjects. Iron 98-102 solute carrier family 11 member 2 Homo sapiens 14-18 14768003-12 2004 Increased expression of DMT1 may play an important role in the pathogenesis of cirrhosis-associated hepatic iron overload. Iron 108-112 solute carrier family 11 member 2 Homo sapiens 24-28 15014911-3 2004 The functional properties and localization in plasma membrane of cells and endosomes suggest an important role for the divalent metal transporter DMT1 (also known as DCT1 and Nramp2) in iron transport and cellular iron homeostasis. Iron 214-218 solute carrier family 11 member 2 Homo sapiens 166-170 15014911-3 2004 The functional properties and localization in plasma membrane of cells and endosomes suggest an important role for the divalent metal transporter DMT1 (also known as DCT1 and Nramp2) in iron transport and cellular iron homeostasis. Iron 214-218 solute carrier family 11 member 2 Homo sapiens 175-181 15014911-4 2004 Although iron metabolism is strictly controlled and the activity of DMT1 is central in controlling iron homeostasis, no regulatory mechanisms for DMT1 have been so far identified. Iron 99-103 solute carrier family 11 member 2 Homo sapiens 68-72 14770366-4 2004 Intestinal iron absorption by mature duodenal enterocytes requires Fe (III) iron reduction by Dcytb and Fe (II) iron transport through apical membranes by the iron transporter Nramp2/DMT1. Iron 11-15 solute carrier family 11 member 2 Homo sapiens 183-187 12907459-8 2003 Comparative Northern analyses of iron-related genes after erythrophagocytosis revealed a 16-fold increase in FPN1 levels after 3 hours, a 10-fold increase in heme oxygenase-1 (HO-1) after 3 hours, a 2-fold increase in natural resistance macrophage-associated protein 1 (Nramp1) levels after 6 hours, but no change in divalent metal ion transporter 1 (DMT1) levels. Iron 33-37 solute carrier family 11 member 2 Homo sapiens 317-349 15381278-3 2004 We observed a general correlation between levels of DMT1, and iron staining. Iron 62-66 solute carrier family 11 member 2 Homo sapiens 52-56 15381278-7 2004 The high levels of DMT1 expression in some of the nuclei of the basal ganglia, particularly the caudate nucleus, putamen, and substantia nigra pars reticulata, may account for the high levels of iron in these regions. Iron 195-199 solute carrier family 11 member 2 Homo sapiens 19-23 12907459-8 2003 Comparative Northern analyses of iron-related genes after erythrophagocytosis revealed a 16-fold increase in FPN1 levels after 3 hours, a 10-fold increase in heme oxygenase-1 (HO-1) after 3 hours, a 2-fold increase in natural resistance macrophage-associated protein 1 (Nramp1) levels after 6 hours, but no change in divalent metal ion transporter 1 (DMT1) levels. Iron 33-37 solute carrier family 11 member 2 Homo sapiens 351-355 12724326-1 2003 Nramp2 (natural resistance-associated macrophage protein 2, also called DMT1 and Slc11a2) is a proton-dependent cation transporter, which plays a central role in iron homeostasis. Iron 162-166 solute carrier family 11 member 2 Homo sapiens 0-6 12876064-0 2003 Altered dietary iron intake is a strong modulator of renal DMT1 expression. Iron 16-20 solute carrier family 11 member 2 Homo sapiens 59-63 12876064-1 2003 Divalent metal transporter1 (DMT1; also known as DCT1 or NRAMP2) is an important component of the cellular machinery responsible for dietary iron absorption in the duodenum. Iron 141-145 solute carrier family 11 member 2 Homo sapiens 0-27 12876064-1 2003 Divalent metal transporter1 (DMT1; also known as DCT1 or NRAMP2) is an important component of the cellular machinery responsible for dietary iron absorption in the duodenum. Iron 141-145 solute carrier family 11 member 2 Homo sapiens 29-33 12876064-1 2003 Divalent metal transporter1 (DMT1; also known as DCT1 or NRAMP2) is an important component of the cellular machinery responsible for dietary iron absorption in the duodenum. Iron 141-145 solute carrier family 11 member 2 Homo sapiens 49-53 12876064-1 2003 Divalent metal transporter1 (DMT1; also known as DCT1 or NRAMP2) is an important component of the cellular machinery responsible for dietary iron absorption in the duodenum. Iron 141-145 solute carrier family 11 member 2 Homo sapiens 57-63 12876064-2 2003 DMT1 is also highly expressed in the kidney where it has been suggested to play a role in urinary iron handling. Iron 98-102 solute carrier family 11 member 2 Homo sapiens 0-4 12876064-3 2003 In this study, we determined the effect on renal DMT1 expression of feeding an iron-restricted diet (50 mg/kg) or an iron-enriched diet (5 g/kg) for 4 wk and measured urinary and fecal iron excretion rates. Iron 79-83 solute carrier family 11 member 2 Homo sapiens 49-53 12876064-4 2003 Feeding the low-iron diet caused a reduction in serum iron concentration and fecal iron output rate with an increase in renal DMT1 expression. Iron 16-20 solute carrier family 11 member 2 Homo sapiens 126-130 12876064-6 2003 Therefore, DMT1 expression in the kidney is sensitive to dietary iron intake, and the level of expression is inversely related to the dietary iron content. Iron 65-69 solute carrier family 11 member 2 Homo sapiens 11-15 12876064-6 2003 Therefore, DMT1 expression in the kidney is sensitive to dietary iron intake, and the level of expression is inversely related to the dietary iron content. Iron 142-146 solute carrier family 11 member 2 Homo sapiens 11-15 12876064-8 2003 Increased DMT1 expression was accompanied by a decrease in urinary iron excretion rate and vice versa when DMT1 expression was reduced. Iron 67-71 solute carrier family 11 member 2 Homo sapiens 10-14 12876064-9 2003 Together, these findings suggest that modulation of renal DMT1 expression may influence renal iron excretion rate. Iron 94-98 solute carrier family 11 member 2 Homo sapiens 58-62 12949720-6 2003 Spearman rank correlations showed that Dcytb, hephaestin, FPN1, and DMT1 mRNA expression are positively related to each other independently of the underlying disease, which ensures an efficient transepithelial transport of absorbed iron. Iron 232-236 solute carrier family 11 member 2 Homo sapiens 68-72 12743117-7 2003 We also provide evidence that the divalent metal influx transporter DMT1 is expressed by astrocytes and is likely to mediate iron influx into these glial cells. Iron 125-129 solute carrier family 11 member 2 Homo sapiens 68-72 12724326-1 2003 Nramp2 (natural resistance-associated macrophage protein 2, also called DMT1 and Slc11a2) is a proton-dependent cation transporter, which plays a central role in iron homeostasis. Iron 162-166 solute carrier family 11 member 2 Homo sapiens 8-58 12724326-1 2003 Nramp2 (natural resistance-associated macrophage protein 2, also called DMT1 and Slc11a2) is a proton-dependent cation transporter, which plays a central role in iron homeostasis. Iron 162-166 solute carrier family 11 member 2 Homo sapiens 72-76 12724326-1 2003 Nramp2 (natural resistance-associated macrophage protein 2, also called DMT1 and Slc11a2) is a proton-dependent cation transporter, which plays a central role in iron homeostasis. Iron 162-166 solute carrier family 11 member 2 Homo sapiens 81-88 12724326-10 2003 Their subcellular colocalization and parallel trafficking suggest that Nramp2 and transferrin receptors are functionally coupled to effect pH-dependent iron uptake across the endosomal membrane. Iron 152-156 solute carrier family 11 member 2 Homo sapiens 71-77 12734107-2 2003 DMT1, also known as Nramp2 and DCT1, is the transporter responsible for intestinal iron uptake. Iron 83-87 solute carrier family 11 member 2 Homo sapiens 0-4 12801950-1 2003 BACKGROUND AND AIMS: In HFE associated hereditary haemochromatosis, the duodenal enterocyte behaves as if iron deficient and previous reports have shown increased duodenal expression of divalent metal transporter 1 (DMT1) and iron regulated gene 1 (Ireg1) in affected subjects. Iron 106-110 solute carrier family 11 member 2 Homo sapiens 186-214 12801950-5 2003 RESULTS: Expression of DMT1 (IRE) and Ireg1 was increased 3-5-fold in iron deficient subjects compared with iron replete subjects. Iron 70-74 solute carrier family 11 member 2 Homo sapiens 23-27 12801950-5 2003 RESULTS: Expression of DMT1 (IRE) and Ireg1 was increased 3-5-fold in iron deficient subjects compared with iron replete subjects. Iron 108-112 solute carrier family 11 member 2 Homo sapiens 23-27 12801950-6 2003 Duodenal expression of DMT1 (IRE) and Ireg1 was similar in haemochromatosis patients and iron replete subjects but in haemochromatosis patients with elevated serum ferritin concentrations, both DMT1 (IRE) and Ireg1 expression were inappropriately increased relative to serum ferritin concentration. Iron 89-93 solute carrier family 11 member 2 Homo sapiens 23-27 12801950-9 2003 CONCLUSIONS: These findings are consistent with DMT1 (IRE) and Ireg1 playing primary roles in the adaptive response to iron deficiency. Iron 119-123 solute carrier family 11 member 2 Homo sapiens 48-52 12787877-10 2003 Further the increase in the expression of the transporter DMT-1 in HepG2 cells after iron treatment is in contrast to the regulation in the duodenum and may be involved in the upregulated uptake of potentially toxic non-transferrin bound iron from the circulation to store it in the non-toxic form of ferritin. Iron 85-89 solute carrier family 11 member 2 Homo sapiens 58-63 12787877-10 2003 Further the increase in the expression of the transporter DMT-1 in HepG2 cells after iron treatment is in contrast to the regulation in the duodenum and may be involved in the upregulated uptake of potentially toxic non-transferrin bound iron from the circulation to store it in the non-toxic form of ferritin. Iron 238-242 solute carrier family 11 member 2 Homo sapiens 58-63 12646040-1 2003 Divalent-metal transporter 1 (DMT1) is involved in the intestinal iron absorption and in iron transport in the transferrin cycle. Iron 66-70 solute carrier family 11 member 2 Homo sapiens 0-28 12646040-1 2003 Divalent-metal transporter 1 (DMT1) is involved in the intestinal iron absorption and in iron transport in the transferrin cycle. Iron 66-70 solute carrier family 11 member 2 Homo sapiens 30-34 12646040-1 2003 Divalent-metal transporter 1 (DMT1) is involved in the intestinal iron absorption and in iron transport in the transferrin cycle. Iron 89-93 solute carrier family 11 member 2 Homo sapiens 0-28 12646040-1 2003 Divalent-metal transporter 1 (DMT1) is involved in the intestinal iron absorption and in iron transport in the transferrin cycle. Iron 89-93 solute carrier family 11 member 2 Homo sapiens 30-34 12734107-5 2003 Treatment of cells with a DMT1 antisense oligonucleotide resulted in 80 and 48% inhibition of iron and copper uptake, respectively. Iron 94-98 solute carrier family 11 member 2 Homo sapiens 26-30 12734107-11 2003 These results demonstrate that DMT1 is a physiologically relevant Cu(1+) transporter in intestinal cells, indicating that intestinal absorption of copper and iron are intertwined. Iron 158-162 solute carrier family 11 member 2 Homo sapiens 31-35 12522007-0 2003 Iron transport by Nramp2/DMT1: pH regulation of transport by 2 histidines in transmembrane domain 6. Iron 0-4 solute carrier family 11 member 2 Homo sapiens 18-24 12522007-0 2003 Iron transport by Nramp2/DMT1: pH regulation of transport by 2 histidines in transmembrane domain 6. Iron 0-4 solute carrier family 11 member 2 Homo sapiens 25-29 12522007-1 2003 Mutations at natural resistance-associated macrophage protein 1 (Nramp1) impair phagocyte function and cause susceptibility to infections while mutations at Nramp2 (divalent metal transporter 1 [DMT1]) affect iron homeostasis and cause severe microcytic anemia. Iron 209-213 solute carrier family 11 member 2 Homo sapiens 157-163 12522007-1 2003 Mutations at natural resistance-associated macrophage protein 1 (Nramp1) impair phagocyte function and cause susceptibility to infections while mutations at Nramp2 (divalent metal transporter 1 [DMT1]) affect iron homeostasis and cause severe microcytic anemia. Iron 209-213 solute carrier family 11 member 2 Homo sapiens 165-200 12730455-2 2003 Several new potential mRNA targets for IRP were recently identified: divalent metal transporter-1 (DMT-1) and ferroportin, which are critical regulators of iron absorption in the gut and of iron cycling between various tissues of the body. Iron 156-160 solute carrier family 11 member 2 Homo sapiens 69-97 12730455-2 2003 Several new potential mRNA targets for IRP were recently identified: divalent metal transporter-1 (DMT-1) and ferroportin, which are critical regulators of iron absorption in the gut and of iron cycling between various tissues of the body. Iron 156-160 solute carrier family 11 member 2 Homo sapiens 99-104 12730464-7 2003 Divalent metal transporter 1 (DMT1) mRNA in the iron-responsive element (IRE) regulated, but not the non-IRE regulated form is increased, as is the placenta Cu oxidase. Iron 48-52 solute carrier family 11 member 2 Homo sapiens 0-28 12730464-7 2003 Divalent metal transporter 1 (DMT1) mRNA in the iron-responsive element (IRE) regulated, but not the non-IRE regulated form is increased, as is the placenta Cu oxidase. Iron 48-52 solute carrier family 11 member 2 Homo sapiens 30-34 12730455-2 2003 Several new potential mRNA targets for IRP were recently identified: divalent metal transporter-1 (DMT-1) and ferroportin, which are critical regulators of iron absorption in the gut and of iron cycling between various tissues of the body. Iron 190-194 solute carrier family 11 member 2 Homo sapiens 69-97 12730455-2 2003 Several new potential mRNA targets for IRP were recently identified: divalent metal transporter-1 (DMT-1) and ferroportin, which are critical regulators of iron absorption in the gut and of iron cycling between various tissues of the body. Iron 190-194 solute carrier family 11 member 2 Homo sapiens 99-104 12388109-0 2003 Effect of DMT1 knockdown on iron, cadmium, and lead uptake in Caco-2 cells. Iron 28-32 solute carrier family 11 member 2 Homo sapiens 10-14 12572661-4 2003 The transporter DMT1 (divalent metal transporter 1) clearly plays a major part in iron uptake and trafficking. Iron 82-86 solute carrier family 11 member 2 Homo sapiens 16-20 12572661-4 2003 The transporter DMT1 (divalent metal transporter 1) clearly plays a major part in iron uptake and trafficking. Iron 82-86 solute carrier family 11 member 2 Homo sapiens 22-50 12572662-18 2003 The recent identification of other proteins of iron and copper metabolism, for example, copper ATPases and the membrane iron transporters DCT1/DMT1/Nramp2 and IREG1/MTP1/ferroportinl, are likely to fill crucial pathway gaps. Iron 47-51 solute carrier family 11 member 2 Homo sapiens 138-142 12572662-18 2003 The recent identification of other proteins of iron and copper metabolism, for example, copper ATPases and the membrane iron transporters DCT1/DMT1/Nramp2 and IREG1/MTP1/ferroportinl, are likely to fill crucial pathway gaps. Iron 47-51 solute carrier family 11 member 2 Homo sapiens 143-147 12572662-18 2003 The recent identification of other proteins of iron and copper metabolism, for example, copper ATPases and the membrane iron transporters DCT1/DMT1/Nramp2 and IREG1/MTP1/ferroportinl, are likely to fill crucial pathway gaps. Iron 47-51 solute carrier family 11 member 2 Homo sapiens 148-154 12753426-0 2003 Effect of iron treatment on nickel absorption and gene expression of the divalent metal transporter (DMT1) by human intestinal Caco-2 cells. Iron 10-14 solute carrier family 11 member 2 Homo sapiens 101-105 12753426-1 2003 Divalent Metal Transporter 1 (DMT1) is a transmembrane transporter located at the apical membrane of enterocytes and implicated in the duodenal uptake of iron. Iron 154-158 solute carrier family 11 member 2 Homo sapiens 0-28 12753426-1 2003 Divalent Metal Transporter 1 (DMT1) is a transmembrane transporter located at the apical membrane of enterocytes and implicated in the duodenal uptake of iron. Iron 154-158 solute carrier family 11 member 2 Homo sapiens 30-34 12753426-6 2003 Iron treatment resulted in decreased DMT1 gene expression which correlated well with the uptake of 59Fe and 63Ni into fully differentiated Caco-2 cells. Iron 0-4 solute carrier family 11 member 2 Homo sapiens 37-41 12388109-1 2003 DMT1 (divalent metal transporter 1) is a hydrogen-coupled divalent metal transporter with a substrate preference for iron, although the protein when expressed in frog oocytes transports a broad range of metals, including the toxic metals cadmium and lead. Iron 117-121 solute carrier family 11 member 2 Homo sapiens 0-4 12388109-1 2003 DMT1 (divalent metal transporter 1) is a hydrogen-coupled divalent metal transporter with a substrate preference for iron, although the protein when expressed in frog oocytes transports a broad range of metals, including the toxic metals cadmium and lead. Iron 117-121 solute carrier family 11 member 2 Homo sapiens 6-34 12388109-8 2003 These results show that DMT1 is important in iron and cadmium transport in Caco-2 cells but that lead enters these cells through an independent hydrogen-driven mechanism. Iron 45-49 solute carrier family 11 member 2 Homo sapiens 24-28 12547239-4 2002 The effects of iron perturbations on DMT1 and FPN1 expression were investigated in CaCo2 cells and in primary tissue cultures of human duodenal biopsies by means of Northern Blot, Western Blot, RNA-bandshift and Nuclear Run off analysis. Iron 15-19 solute carrier family 11 member 2 Homo sapiens 37-41 12376346-7 2002 This is similar to our previous findings with divalent metal transporter-1 (DMT1), an iron transporter that is required for iron uptake and intracellular iron trafficking. Iron 86-90 solute carrier family 11 member 2 Homo sapiens 46-74 12376346-7 2002 This is similar to our previous findings with divalent metal transporter-1 (DMT1), an iron transporter that is required for iron uptake and intracellular iron trafficking. Iron 86-90 solute carrier family 11 member 2 Homo sapiens 76-80 12376346-7 2002 This is similar to our previous findings with divalent metal transporter-1 (DMT1), an iron transporter that is required for iron uptake and intracellular iron trafficking. Iron 124-128 solute carrier family 11 member 2 Homo sapiens 46-74 12376346-7 2002 This is similar to our previous findings with divalent metal transporter-1 (DMT1), an iron transporter that is required for iron uptake and intracellular iron trafficking. Iron 124-128 solute carrier family 11 member 2 Homo sapiens 76-80 12547214-2 2002 The identification of the HFE gene and the apical iron transporter divalent metal transporter-1, DMT-1, provide a direct method to address the mechanisms of iron overload in this disease. Iron 50-54 solute carrier family 11 member 2 Homo sapiens 97-102 12547214-13 2002 The observed twofold upregulation of the DMT-1 is consistent with the slow but steady increase in body iron stores observed in those presenting with clinical features of hereditary hemochromatosis. Iron 103-107 solute carrier family 11 member 2 Homo sapiens 41-46 12475959-1 2002 Divalent metal transporter 1 (DMT1) is responsible for dietary-iron absorption from apical plasma membrane in the duodenum and iron acquisition from the transferrin cycle endosomes in peripheral tissues. Iron 63-67 solute carrier family 11 member 2 Homo sapiens 0-28 12475959-1 2002 Divalent metal transporter 1 (DMT1) is responsible for dietary-iron absorption from apical plasma membrane in the duodenum and iron acquisition from the transferrin cycle endosomes in peripheral tissues. Iron 63-67 solute carrier family 11 member 2 Homo sapiens 30-34 12475959-1 2002 Divalent metal transporter 1 (DMT1) is responsible for dietary-iron absorption from apical plasma membrane in the duodenum and iron acquisition from the transferrin cycle endosomes in peripheral tissues. Iron 127-131 solute carrier family 11 member 2 Homo sapiens 0-28 12475959-1 2002 Divalent metal transporter 1 (DMT1) is responsible for dietary-iron absorption from apical plasma membrane in the duodenum and iron acquisition from the transferrin cycle endosomes in peripheral tissues. Iron 127-131 solute carrier family 11 member 2 Homo sapiens 30-34 12475959-11 2002 Our data indicate that the cell type-specific expression patterns and the distinct subcellular localizations of two DMT1 isoforms may be involved in the different iron acquisition steps from the subcellular membranes in various cell types. Iron 163-167 solute carrier family 11 member 2 Homo sapiens 116-120 12547239-0 2002 Mechanisms of iron mediated regulation of the duodenal iron transporters divalent metal transporter 1 and ferroportin 1. Iron 14-18 solute carrier family 11 member 2 Homo sapiens 73-101 12547239-2 2002 Identification of divalent metal transporter 1 (DMT1) and ferroportin 1 (FPN1) has improved our understanding of iron transport across the intestinal epithelium. Iron 113-117 solute carrier family 11 member 2 Homo sapiens 18-46 12547239-2 2002 Identification of divalent metal transporter 1 (DMT1) and ferroportin 1 (FPN1) has improved our understanding of iron transport across the intestinal epithelium. Iron 113-117 solute carrier family 11 member 2 Homo sapiens 48-52 12547239-3 2002 Although DMT1 and FPN1 mRNA bear an iron responsive element (IRE) within its untranslated regions which should cause susceptibility to iron mediated posttranscriptional regulation the latter has not been shown so far. Iron 36-40 solute carrier family 11 member 2 Homo sapiens 9-13 12547239-3 2002 Although DMT1 and FPN1 mRNA bear an iron responsive element (IRE) within its untranslated regions which should cause susceptibility to iron mediated posttranscriptional regulation the latter has not been shown so far. Iron 135-139 solute carrier family 11 member 2 Homo sapiens 9-13 12547239-8 2002 Nuclear run-off analysis then demonstrated that the effects of iron and desferrioxamine on DMT1 and FPN1 mRNA expression are rather due to modulation of transcription of these genes. Iron 63-67 solute carrier family 11 member 2 Homo sapiens 91-95 12547239-9 2002 Our results demonstrate that iron unidirectionally regulates the expression of the two ferrous ion transporters DMT1 and FPN1 by affecting their transcription. Iron 29-33 solute carrier family 11 member 2 Homo sapiens 112-116 12139757-0 2002 Erythroid 5-aminolevulinate synthase, ferrochelatase and DMT1 expression in erythroid progenitors: differential pathways for erythropoietin and iron-dependent regulation. Iron 144-148 solute carrier family 11 member 2 Homo sapiens 57-61 12223357-0 2002 The transcytosis of divalent metal transporter 1 and apo-transferrin during iron uptake in intestinal epithelium. Iron 76-80 solute carrier family 11 member 2 Homo sapiens 20-48 12223357-3 2002 With the addition of iron to the apical surface, divalent metal transporter 1 (DMT1) on the brush-border membrane (BBM) undergoes endocytosis. Iron 21-25 solute carrier family 11 member 2 Homo sapiens 49-77 12223357-3 2002 With the addition of iron to the apical surface, divalent metal transporter 1 (DMT1) on the brush-border membrane (BBM) undergoes endocytosis. Iron 21-25 solute carrier family 11 member 2 Homo sapiens 79-83 12223357-4 2002 These findings suggest that in Caco-2 cells DMT1 and apo-transferrin may cooperate in iron transport through transcytosis. Iron 86-90 solute carrier family 11 member 2 Homo sapiens 44-48 12223357-5 2002 To prove this hypothesis, we determined by confocal microscopy that, after addition of iron to the apical chamber, DMT1 from the BBM and Texas red apo-transferrin from the basal chamber colocalized in a perinuclear compartment. Iron 87-91 solute carrier family 11 member 2 Homo sapiens 115-119 12220667-3 2002 Our data demonstrate that copper could compete with iron for uptake via DMT1 and that DMT1 protein and mRNA expression were decreased following exposure (24 h) to high copper. Iron 52-56 solute carrier family 11 member 2 Homo sapiens 72-76 12196176-2 2002 Dcytb is highly expressed in duodenal brush-border membrane and is implicated in dietary iron absorption by reducing dietary ferric iron to the ferrous form for transport via Nramp2/DCT1 (divalent-cation transporter 1)/DMT1 (divalent metal-transporter 1). Iron 89-93 solute carrier family 11 member 2 Homo sapiens 182-186 12196176-2 2002 Dcytb is highly expressed in duodenal brush-border membrane and is implicated in dietary iron absorption by reducing dietary ferric iron to the ferrous form for transport via Nramp2/DCT1 (divalent-cation transporter 1)/DMT1 (divalent metal-transporter 1). Iron 89-93 solute carrier family 11 member 2 Homo sapiens 188-217 12209011-1 2002 Divalent metal transporter 1 (DMT1) mediates apical iron uptake into duodenal enterocytes and also transfers iron from the endosome into the cytosol after cellular uptake via the transferrin receptor. Iron 52-56 solute carrier family 11 member 2 Homo sapiens 0-28 12209011-1 2002 Divalent metal transporter 1 (DMT1) mediates apical iron uptake into duodenal enterocytes and also transfers iron from the endosome into the cytosol after cellular uptake via the transferrin receptor. Iron 52-56 solute carrier family 11 member 2 Homo sapiens 30-34 12209011-1 2002 Divalent metal transporter 1 (DMT1) mediates apical iron uptake into duodenal enterocytes and also transfers iron from the endosome into the cytosol after cellular uptake via the transferrin receptor. Iron 109-113 solute carrier family 11 member 2 Homo sapiens 0-28 12209011-1 2002 Divalent metal transporter 1 (DMT1) mediates apical iron uptake into duodenal enterocytes and also transfers iron from the endosome into the cytosol after cellular uptake via the transferrin receptor. Iron 109-113 solute carrier family 11 member 2 Homo sapiens 30-34 12209011-3 2002 DMT1 mRNA levels are increased in the duodenum of iron-deficient animals. Iron 50-54 solute carrier family 11 member 2 Homo sapiens 0-4 12209011-5 2002 Here, we show that iron regulation of DMT1 involves the expression of a previously unrecognized upstream 5" exon (exon 1A) of the human and murine DMT1 gene. Iron 19-23 solute carrier family 11 member 2 Homo sapiens 38-42 12209011-9 2002 We show that two regulatory regions, the 5" promoter/exon 1A region and the IRE-containing terminal exon participate in iron regulation of DMT1 expression, which operate in a tissue-specific way. Iron 120-124 solute carrier family 11 member 2 Homo sapiens 139-143 12209011-10 2002 These results uncover an unexpected complexity of DMT1 expression and regulation, with implications for understanding the physiology, cell biology, and pathophysiology of mammalian iron metabolism. Iron 181-185 solute carrier family 11 member 2 Homo sapiens 50-54 12127992-2 2002 Here, we provide direct evidence demonstrating that DMT1 expressed in yeast mutants defective for high affinity iron transport facilitates the transport of iron with an "apparent K(m)" of approximately 1.2 microM, and transport of lead with an "apparent K(m)" of approximately 1.8 microM. Iron 112-116 solute carrier family 11 member 2 Homo sapiens 52-56 12127992-2 2002 Here, we provide direct evidence demonstrating that DMT1 expressed in yeast mutants defective for high affinity iron transport facilitates the transport of iron with an "apparent K(m)" of approximately 1.2 microM, and transport of lead with an "apparent K(m)" of approximately 1.8 microM. Iron 156-160 solute carrier family 11 member 2 Homo sapiens 52-56 11897618-6 2002 In controls, we found inverse relationships between the DCT1 splice form containing an iron-responsive element (IRE) and blood hemoglobin, serum transferrin saturation, or ferritin. Iron 87-91 solute carrier family 11 member 2 Homo sapiens 56-60 12127992-3 2002 DMT1-dependent lead transport was H(+)-dependent and was inhibited by iron. Iron 70-74 solute carrier family 11 member 2 Homo sapiens 0-4 12127992-5 2002 These results show that DMT1 transports lead and iron with similar affinity in a yeast model suggesting that DMT1 is a transporter for lead. Iron 49-53 solute carrier family 11 member 2 Homo sapiens 24-28 12127992-5 2002 These results show that DMT1 transports lead and iron with similar affinity in a yeast model suggesting that DMT1 is a transporter for lead. Iron 49-53 solute carrier family 11 member 2 Homo sapiens 109-113 11943663-1 2002 The capacity of natural resistance-associated macrophage protein-2 [Nramp2; also called divalent metal transporter-1 (DMT1) and divalent cation transporter-1 (DCT1)] to transport iron and its ubiquitous expression make it a likely candidate for transferrin-independent uptake of iron in peripheral tissues. Iron 179-183 solute carrier family 11 member 2 Homo sapiens 16-66 11943663-1 2002 The capacity of natural resistance-associated macrophage protein-2 [Nramp2; also called divalent metal transporter-1 (DMT1) and divalent cation transporter-1 (DCT1)] to transport iron and its ubiquitous expression make it a likely candidate for transferrin-independent uptake of iron in peripheral tissues. Iron 179-183 solute carrier family 11 member 2 Homo sapiens 68-74 11943663-1 2002 The capacity of natural resistance-associated macrophage protein-2 [Nramp2; also called divalent metal transporter-1 (DMT1) and divalent cation transporter-1 (DCT1)] to transport iron and its ubiquitous expression make it a likely candidate for transferrin-independent uptake of iron in peripheral tissues. Iron 179-183 solute carrier family 11 member 2 Homo sapiens 88-116 11943663-1 2002 The capacity of natural resistance-associated macrophage protein-2 [Nramp2; also called divalent metal transporter-1 (DMT1) and divalent cation transporter-1 (DCT1)] to transport iron and its ubiquitous expression make it a likely candidate for transferrin-independent uptake of iron in peripheral tissues. Iron 179-183 solute carrier family 11 member 2 Homo sapiens 118-122 11943663-1 2002 The capacity of natural resistance-associated macrophage protein-2 [Nramp2; also called divalent metal transporter-1 (DMT1) and divalent cation transporter-1 (DCT1)] to transport iron and its ubiquitous expression make it a likely candidate for transferrin-independent uptake of iron in peripheral tissues. Iron 179-183 solute carrier family 11 member 2 Homo sapiens 128-157 11943663-1 2002 The capacity of natural resistance-associated macrophage protein-2 [Nramp2; also called divalent metal transporter-1 (DMT1) and divalent cation transporter-1 (DCT1)] to transport iron and its ubiquitous expression make it a likely candidate for transferrin-independent uptake of iron in peripheral tissues. Iron 179-183 solute carrier family 11 member 2 Homo sapiens 159-163 11943663-1 2002 The capacity of natural resistance-associated macrophage protein-2 [Nramp2; also called divalent metal transporter-1 (DMT1) and divalent cation transporter-1 (DCT1)] to transport iron and its ubiquitous expression make it a likely candidate for transferrin-independent uptake of iron in peripheral tissues. Iron 279-283 solute carrier family 11 member 2 Homo sapiens 16-66 11943663-1 2002 The capacity of natural resistance-associated macrophage protein-2 [Nramp2; also called divalent metal transporter-1 (DMT1) and divalent cation transporter-1 (DCT1)] to transport iron and its ubiquitous expression make it a likely candidate for transferrin-independent uptake of iron in peripheral tissues. Iron 279-283 solute carrier family 11 member 2 Homo sapiens 68-74 11943663-1 2002 The capacity of natural resistance-associated macrophage protein-2 [Nramp2; also called divalent metal transporter-1 (DMT1) and divalent cation transporter-1 (DCT1)] to transport iron and its ubiquitous expression make it a likely candidate for transferrin-independent uptake of iron in peripheral tissues. Iron 279-283 solute carrier family 11 member 2 Homo sapiens 88-116 11943663-1 2002 The capacity of natural resistance-associated macrophage protein-2 [Nramp2; also called divalent metal transporter-1 (DMT1) and divalent cation transporter-1 (DCT1)] to transport iron and its ubiquitous expression make it a likely candidate for transferrin-independent uptake of iron in peripheral tissues. Iron 279-283 solute carrier family 11 member 2 Homo sapiens 118-122 11943663-1 2002 The capacity of natural resistance-associated macrophage protein-2 [Nramp2; also called divalent metal transporter-1 (DMT1) and divalent cation transporter-1 (DCT1)] to transport iron and its ubiquitous expression make it a likely candidate for transferrin-independent uptake of iron in peripheral tissues. Iron 279-283 solute carrier family 11 member 2 Homo sapiens 128-157 11943663-1 2002 The capacity of natural resistance-associated macrophage protein-2 [Nramp2; also called divalent metal transporter-1 (DMT1) and divalent cation transporter-1 (DCT1)] to transport iron and its ubiquitous expression make it a likely candidate for transferrin-independent uptake of iron in peripheral tissues. Iron 279-283 solute carrier family 11 member 2 Homo sapiens 159-163 11943663-2 2002 We tested the hypothesis that non-transferrin-bound iron uptake by airway epithelial cells is associated with Nramp2/DMT1/DCT1 and that exposure to iron can increase Nramp2/DMT1/DCT1 mRNA and protein expression and transport of this metal. Iron 52-56 solute carrier family 11 member 2 Homo sapiens 110-116 11943663-2 2002 We tested the hypothesis that non-transferrin-bound iron uptake by airway epithelial cells is associated with Nramp2/DMT1/DCT1 and that exposure to iron can increase Nramp2/DMT1/DCT1 mRNA and protein expression and transport of this metal. Iron 52-56 solute carrier family 11 member 2 Homo sapiens 117-121 11943663-2 2002 We tested the hypothesis that non-transferrin-bound iron uptake by airway epithelial cells is associated with Nramp2/DMT1/DCT1 and that exposure to iron can increase Nramp2/DMT1/DCT1 mRNA and protein expression and transport of this metal. Iron 52-56 solute carrier family 11 member 2 Homo sapiens 122-126 11943663-2 2002 We tested the hypothesis that non-transferrin-bound iron uptake by airway epithelial cells is associated with Nramp2/DMT1/DCT1 and that exposure to iron can increase Nramp2/DMT1/DCT1 mRNA and protein expression and transport of this metal. Iron 148-152 solute carrier family 11 member 2 Homo sapiens 166-172 11943663-2 2002 We tested the hypothesis that non-transferrin-bound iron uptake by airway epithelial cells is associated with Nramp2/DMT1/DCT1 and that exposure to iron can increase Nramp2/DMT1/DCT1 mRNA and protein expression and transport of this metal. Iron 148-152 solute carrier family 11 member 2 Homo sapiens 173-177 11943663-2 2002 We tested the hypothesis that non-transferrin-bound iron uptake by airway epithelial cells is associated with Nramp2/DMT1/DCT1 and that exposure to iron can increase Nramp2/DMT1/DCT1 mRNA and protein expression and transport of this metal. Iron 148-152 solute carrier family 11 member 2 Homo sapiens 178-182 11943663-5 2002 The Nramp2/DMT1/DCT1 mRNA isoform without an iron-response element (IRE) increased with exposure of BEAS-2B cells to FAC. Iron 45-49 solute carrier family 11 member 2 Homo sapiens 4-10 11943663-5 2002 The Nramp2/DMT1/DCT1 mRNA isoform without an iron-response element (IRE) increased with exposure of BEAS-2B cells to FAC. Iron 45-49 solute carrier family 11 member 2 Homo sapiens 11-15 11943663-5 2002 The Nramp2/DMT1/DCT1 mRNA isoform without an iron-response element (IRE) increased with exposure of BEAS-2B cells to FAC. Iron 45-49 solute carrier family 11 member 2 Homo sapiens 16-20 11943663-10 2002 We conclude that airway epithelial cells increase mRNA and expression of the Nramp2/DMT1/DCT1 without an IRE after exposure to iron. Iron 127-131 solute carrier family 11 member 2 Homo sapiens 77-83 11943663-10 2002 We conclude that airway epithelial cells increase mRNA and expression of the Nramp2/DMT1/DCT1 without an IRE after exposure to iron. Iron 127-131 solute carrier family 11 member 2 Homo sapiens 84-88 11943663-10 2002 We conclude that airway epithelial cells increase mRNA and expression of the Nramp2/DMT1/DCT1 without an IRE after exposure to iron. Iron 127-131 solute carrier family 11 member 2 Homo sapiens 89-93 11897618-11 2002 Our data show that expression levels of human DCT1 mRNA, and to a lesser extent IREG1 mRNA, are regulated in an iron-dependent manner, whereas mRNA of hephaestin is not affected. Iron 112-116 solute carrier family 11 member 2 Homo sapiens 46-50 11925462-3 2002 Recently, a number of novel genes involved in iron metabolism, such as the iron uptake transporter DMT1/DCT1/Nramp2 and the iron export transporter IREG1/ferroportin1/MTP1, have been identified, providing important insights about molecular aspects of intestinal iron absorption and its regulation. Iron 75-79 solute carrier family 11 member 2 Homo sapiens 99-103 11925462-3 2002 Recently, a number of novel genes involved in iron metabolism, such as the iron uptake transporter DMT1/DCT1/Nramp2 and the iron export transporter IREG1/ferroportin1/MTP1, have been identified, providing important insights about molecular aspects of intestinal iron absorption and its regulation. Iron 75-79 solute carrier family 11 member 2 Homo sapiens 104-108 11925462-0 2002 Iron treatment downregulates DMT1 and IREG1 mRNA expression in Caco-2 cells. Iron 0-4 solute carrier family 11 member 2 Homo sapiens 29-33 11925462-3 2002 Recently, a number of novel genes involved in iron metabolism, such as the iron uptake transporter DMT1/DCT1/Nramp2 and the iron export transporter IREG1/ferroportin1/MTP1, have been identified, providing important insights about molecular aspects of intestinal iron absorption and its regulation. Iron 75-79 solute carrier family 11 member 2 Homo sapiens 109-115 11925462-3 2002 Recently, a number of novel genes involved in iron metabolism, such as the iron uptake transporter DMT1/DCT1/Nramp2 and the iron export transporter IREG1/ferroportin1/MTP1, have been identified, providing important insights about molecular aspects of intestinal iron absorption and its regulation. Iron 46-50 solute carrier family 11 member 2 Homo sapiens 99-103 11925462-4 2002 The aim of this study was to investigate the effects of iron treatment on DMT1 and IREG1 mRNA expression in Caco-2 cells, a human intestinal cell line. Iron 56-60 solute carrier family 11 member 2 Homo sapiens 74-78 11925462-3 2002 Recently, a number of novel genes involved in iron metabolism, such as the iron uptake transporter DMT1/DCT1/Nramp2 and the iron export transporter IREG1/ferroportin1/MTP1, have been identified, providing important insights about molecular aspects of intestinal iron absorption and its regulation. Iron 46-50 solute carrier family 11 member 2 Homo sapiens 104-108 11925462-5 2002 Exposure of the cells to iron (200 micromol/L ferric nitrilotriacetic acid for 72 h) significantly decreased transferrin receptor mRNA (80%), DMT1 mRNA (57%) and IREG1 mRNA (52%). Iron 25-29 solute carrier family 11 member 2 Homo sapiens 142-146 11925462-3 2002 Recently, a number of novel genes involved in iron metabolism, such as the iron uptake transporter DMT1/DCT1/Nramp2 and the iron export transporter IREG1/ferroportin1/MTP1, have been identified, providing important insights about molecular aspects of intestinal iron absorption and its regulation. Iron 46-50 solute carrier family 11 member 2 Homo sapiens 109-115 11925462-3 2002 Recently, a number of novel genes involved in iron metabolism, such as the iron uptake transporter DMT1/DCT1/Nramp2 and the iron export transporter IREG1/ferroportin1/MTP1, have been identified, providing important insights about molecular aspects of intestinal iron absorption and its regulation. Iron 75-79 solute carrier family 11 member 2 Homo sapiens 99-103 11925462-3 2002 Recently, a number of novel genes involved in iron metabolism, such as the iron uptake transporter DMT1/DCT1/Nramp2 and the iron export transporter IREG1/ferroportin1/MTP1, have been identified, providing important insights about molecular aspects of intestinal iron absorption and its regulation. Iron 75-79 solute carrier family 11 member 2 Homo sapiens 104-108 11925462-3 2002 Recently, a number of novel genes involved in iron metabolism, such as the iron uptake transporter DMT1/DCT1/Nramp2 and the iron export transporter IREG1/ferroportin1/MTP1, have been identified, providing important insights about molecular aspects of intestinal iron absorption and its regulation. Iron 75-79 solute carrier family 11 member 2 Homo sapiens 109-115 11925462-3 2002 Recently, a number of novel genes involved in iron metabolism, such as the iron uptake transporter DMT1/DCT1/Nramp2 and the iron export transporter IREG1/ferroportin1/MTP1, have been identified, providing important insights about molecular aspects of intestinal iron absorption and its regulation. Iron 75-79 solute carrier family 11 member 2 Homo sapiens 99-103 11925462-3 2002 Recently, a number of novel genes involved in iron metabolism, such as the iron uptake transporter DMT1/DCT1/Nramp2 and the iron export transporter IREG1/ferroportin1/MTP1, have been identified, providing important insights about molecular aspects of intestinal iron absorption and its regulation. Iron 75-79 solute carrier family 11 member 2 Homo sapiens 104-108 11925462-3 2002 Recently, a number of novel genes involved in iron metabolism, such as the iron uptake transporter DMT1/DCT1/Nramp2 and the iron export transporter IREG1/ferroportin1/MTP1, have been identified, providing important insights about molecular aspects of intestinal iron absorption and its regulation. Iron 75-79 solute carrier family 11 member 2 Homo sapiens 109-115 11842004-9 2002 Since paraferritin functions to reduce newly transported ferric iron to ferrous iron and DMT-1 can transport ferrous iron, these findings suggest a role for DMT-1 in conveyance of iron from paraferritin to ferrochelatase, the enzyme utilizing ferrous iron for the synthesis of heme in the mitochondrion. Iron 80-84 solute carrier family 11 member 2 Homo sapiens 157-162 11755534-1 2002 A divalent metal transporter, DMT1, located on the apical membrane of intestinal enterocytes is the major pathway for the absorption of dietary non-haem iron. Iron 153-157 solute carrier family 11 member 2 Homo sapiens 30-34 11842003-6 2002 These results suggest that DMT1, Fpn1, and Heph are involved in the iron uptake process modulated by copper status. Iron 68-72 solute carrier family 11 member 2 Homo sapiens 27-31 11842004-9 2002 Since paraferritin functions to reduce newly transported ferric iron to ferrous iron and DMT-1 can transport ferrous iron, these findings suggest a role for DMT-1 in conveyance of iron from paraferritin to ferrochelatase, the enzyme utilizing ferrous iron for the synthesis of heme in the mitochondrion. Iron 80-84 solute carrier family 11 member 2 Homo sapiens 157-162 11842004-9 2002 Since paraferritin functions to reduce newly transported ferric iron to ferrous iron and DMT-1 can transport ferrous iron, these findings suggest a role for DMT-1 in conveyance of iron from paraferritin to ferrochelatase, the enzyme utilizing ferrous iron for the synthesis of heme in the mitochondrion. Iron 80-84 solute carrier family 11 member 2 Homo sapiens 157-162 11755534-2 2002 Using human intestinal Caco-2 TC7 cells, we have shown that iron uptake and DMT1 protein in the plasma membrane were significantly decreased by exposure to high iron for 24 h, in a concentration-dependent manner, whereas whole cell DMT1 protein abundance was unaltered. Iron 161-165 solute carrier family 11 member 2 Homo sapiens 76-80 11755534-2 2002 Using human intestinal Caco-2 TC7 cells, we have shown that iron uptake and DMT1 protein in the plasma membrane were significantly decreased by exposure to high iron for 24 h, in a concentration-dependent manner, whereas whole cell DMT1 protein abundance was unaltered. Iron 161-165 solute carrier family 11 member 2 Homo sapiens 232-236 11755534-3 2002 This suggests that part of the response to high iron involved redistribution of DMT1 between the cytosol and cell membrane. Iron 48-52 solute carrier family 11 member 2 Homo sapiens 80-84 11755534-4 2002 These events preceded changes in DMT1 mRNA, which was only decreased following 72 h exposure to high iron. Iron 101-105 solute carrier family 11 member 2 Homo sapiens 33-37 11502556-5 2001 Increased uptake of Fe and Cd was observed in the HEK-293 cell line overexpressing DMT1. Iron 20-22 solute carrier family 11 member 2 Homo sapiens 83-87 11741608-1 2001 The first step in intestinal iron absorption is mediated by the H(+)-coupled Fe(2+) transporter called divalent cation transporter 1/divalent metal ion transporter 1 (DCT1/DMT1) (also known as natural resistance-associated macrophage protein 2). Iron 29-33 solute carrier family 11 member 2 Homo sapiens 103-165 11741608-1 2001 The first step in intestinal iron absorption is mediated by the H(+)-coupled Fe(2+) transporter called divalent cation transporter 1/divalent metal ion transporter 1 (DCT1/DMT1) (also known as natural resistance-associated macrophage protein 2). Iron 29-33 solute carrier family 11 member 2 Homo sapiens 167-171 11741608-1 2001 The first step in intestinal iron absorption is mediated by the H(+)-coupled Fe(2+) transporter called divalent cation transporter 1/divalent metal ion transporter 1 (DCT1/DMT1) (also known as natural resistance-associated macrophage protein 2). Iron 29-33 solute carrier family 11 member 2 Homo sapiens 172-176 11741608-1 2001 The first step in intestinal iron absorption is mediated by the H(+)-coupled Fe(2+) transporter called divalent cation transporter 1/divalent metal ion transporter 1 (DCT1/DMT1) (also known as natural resistance-associated macrophage protein 2). Iron 29-33 solute carrier family 11 member 2 Homo sapiens 193-243 11741608-2 2001 DCT1/DMT1 mRNA levels in the duodenum strongly increase in response to iron depletion. Iron 71-75 solute carrier family 11 member 2 Homo sapiens 0-4 11741608-2 2001 DCT1/DMT1 mRNA levels in the duodenum strongly increase in response to iron depletion. Iron 71-75 solute carrier family 11 member 2 Homo sapiens 5-9 11741608-3 2001 To study the mechanism of iron-dependent DCT1/DMT1 mRNA regulation, we investigated the endogenous expression of DCT1/DMT1 mRNA in various cell types. Iron 26-30 solute carrier family 11 member 2 Homo sapiens 41-45 11741608-3 2001 To study the mechanism of iron-dependent DCT1/DMT1 mRNA regulation, we investigated the endogenous expression of DCT1/DMT1 mRNA in various cell types. Iron 26-30 solute carrier family 11 member 2 Homo sapiens 46-50 11741608-3 2001 To study the mechanism of iron-dependent DCT1/DMT1 mRNA regulation, we investigated the endogenous expression of DCT1/DMT1 mRNA in various cell types. Iron 26-30 solute carrier family 11 member 2 Homo sapiens 113-117 11741608-3 2001 To study the mechanism of iron-dependent DCT1/DMT1 mRNA regulation, we investigated the endogenous expression of DCT1/DMT1 mRNA in various cell types. Iron 26-30 solute carrier family 11 member 2 Homo sapiens 118-122 11741608-4 2001 We found that only the iron responsive element (IRE)-containing form, which corresponds to one of two splice forms of DCT1/DMT1, is responsive to iron treatment and this responsiveness was cell type specific. Iron 23-27 solute carrier family 11 member 2 Homo sapiens 118-122 11741608-4 2001 We found that only the iron responsive element (IRE)-containing form, which corresponds to one of two splice forms of DCT1/DMT1, is responsive to iron treatment and this responsiveness was cell type specific. Iron 23-27 solute carrier family 11 member 2 Homo sapiens 123-127 11741608-4 2001 We found that only the iron responsive element (IRE)-containing form, which corresponds to one of two splice forms of DCT1/DMT1, is responsive to iron treatment and this responsiveness was cell type specific. Iron 146-150 solute carrier family 11 member 2 Homo sapiens 118-122 11741608-4 2001 We found that only the iron responsive element (IRE)-containing form, which corresponds to one of two splice forms of DCT1/DMT1, is responsive to iron treatment and this responsiveness was cell type specific. Iron 146-150 solute carrier family 11 member 2 Homo sapiens 123-127 11741608-7 2001 We propose that regulation of DCT1/DMT1 mRNA by iron involves post-transcriptional regulation through the binding of IRP1 to the transporter"s IRE, as well as other as yet unknown factors. Iron 48-52 solute carrier family 11 member 2 Homo sapiens 30-34 11741608-7 2001 We propose that regulation of DCT1/DMT1 mRNA by iron involves post-transcriptional regulation through the binding of IRP1 to the transporter"s IRE, as well as other as yet unknown factors. Iron 48-52 solute carrier family 11 member 2 Homo sapiens 35-39 11684086-3 2001 Several studies have shown that increasing dietary iron downregulates DMT1. Iron 51-55 solute carrier family 11 member 2 Homo sapiens 70-74 11502556-9 2001 Our results suggest that Fe and Cd are transported in MDCK cells by a transporter with biochemical properties similar to those of DMT1. Iron 25-27 solute carrier family 11 member 2 Homo sapiens 130-134 11389698-5 2001 Similarly, expression of the iron-responsive element (IRE)-regulated form of the divalent metal transporter 1 (DMT1) was also increased. Iron 29-33 solute carrier family 11 member 2 Homo sapiens 81-109 11514223-2 2001 NRAMP2 (DMT1) acts as an iron-uptake protein in both the duodenum and in peripheral tissues. Iron 25-29 solute carrier family 11 member 2 Homo sapiens 0-6 11514223-2 2001 NRAMP2 (DMT1) acts as an iron-uptake protein in both the duodenum and in peripheral tissues. Iron 25-29 solute carrier family 11 member 2 Homo sapiens 8-12 11439223-1 2001 Divalent metal transporter 1 (DMT1) is a transmembrane, proton-coupled metal ion transporter that is upregulated in the duodenum of iron-deficient rodents and in hereditary hemochromatosis patients, suggesting that it may constitute a key factor in the uptake of dietary iron. Iron 132-136 solute carrier family 11 member 2 Homo sapiens 0-28 11439223-1 2001 Divalent metal transporter 1 (DMT1) is a transmembrane, proton-coupled metal ion transporter that is upregulated in the duodenum of iron-deficient rodents and in hereditary hemochromatosis patients, suggesting that it may constitute a key factor in the uptake of dietary iron. Iron 132-136 solute carrier family 11 member 2 Homo sapiens 30-34 11439223-1 2001 Divalent metal transporter 1 (DMT1) is a transmembrane, proton-coupled metal ion transporter that is upregulated in the duodenum of iron-deficient rodents and in hereditary hemochromatosis patients, suggesting that it may constitute a key factor in the uptake of dietary iron. Iron 271-275 solute carrier family 11 member 2 Homo sapiens 0-28 11439223-1 2001 Divalent metal transporter 1 (DMT1) is a transmembrane, proton-coupled metal ion transporter that is upregulated in the duodenum of iron-deficient rodents and in hereditary hemochromatosis patients, suggesting that it may constitute a key factor in the uptake of dietary iron. Iron 271-275 solute carrier family 11 member 2 Homo sapiens 30-34 11439223-7 2001 No effects were observed at 24 h. Gene expression of DMT1 in the iron-treated Caco-2 cells was reduced by about 50% at 3 and 7 days and thus, correlated well with the uptake of cadmium. Iron 65-69 solute carrier family 11 member 2 Homo sapiens 53-57 11389698-5 2001 Similarly, expression of the iron-responsive element (IRE)-regulated form of the divalent metal transporter 1 (DMT1) was also increased. Iron 29-33 solute carrier family 11 member 2 Homo sapiens 111-115 11313311-2 2001 The identification of divalent-metal transporter 1 (DMT1) and ferroportin 1 (FP1) has improved our understanding of transmembrane iron trafficking. Iron 130-134 solute carrier family 11 member 2 Homo sapiens 22-50 11313311-2 2001 The identification of divalent-metal transporter 1 (DMT1) and ferroportin 1 (FP1) has improved our understanding of transmembrane iron trafficking. Iron 130-134 solute carrier family 11 member 2 Homo sapiens 52-56 11313311-0 2001 Expression of the duodenal iron transporters divalent-metal transporter 1 and ferroportin 1 in iron deficiency and iron overload. Iron 27-31 solute carrier family 11 member 2 Homo sapiens 45-73 11313311-6 2001 Moreover, DMT1 and FP1 mRNA levels were significantly increased in patients with iron deficiency, HFE and non-HFE hemochromatosis, whereas they were unchanged in patients with secondary iron overload. Iron 81-85 solute carrier family 11 member 2 Homo sapiens 10-14 11313311-8 2001 In patients with normal iron status or iron deficiency, significant negative correlations between DMT1, FP1 mRNA, and serum iron parameters were found, which were absent in subjects with primary hemochromatosis. Iron 39-43 solute carrier family 11 member 2 Homo sapiens 98-102 11313311-9 2001 CONCLUSIONS: DMT1 and FP1 are centrally involved in iron uptake/transfer in the duodenum and in the adaptive changes of iron homeostasis to iron deficiency and overload. Iron 52-56 solute carrier family 11 member 2 Homo sapiens 13-17 11313311-9 2001 CONCLUSIONS: DMT1 and FP1 are centrally involved in iron uptake/transfer in the duodenum and in the adaptive changes of iron homeostasis to iron deficiency and overload. Iron 120-124 solute carrier family 11 member 2 Homo sapiens 13-17 11095929-3 2000 The aim of this study was to determine whether the recently characterized endosomal membrane iron transporter, divalent metal ion transporter-1 (DMT-1), is expressed in human syncytiotrophoblast, and whether its cellular localization would support roles for cytoplasmic and placental-fetal iron transport. Iron 93-97 solute carrier family 11 member 2 Homo sapiens 111-143 10942769-0 2000 Nramp 2 (DCT1/DMT1) expressed at the plasma membrane transports iron and other divalent cations into a calcein-accessible cytoplasmic pool. Iron 64-68 solute carrier family 11 member 2 Homo sapiens 0-7 10942769-0 2000 Nramp 2 (DCT1/DMT1) expressed at the plasma membrane transports iron and other divalent cations into a calcein-accessible cytoplasmic pool. Iron 64-68 solute carrier family 11 member 2 Homo sapiens 9-13 10942769-0 2000 Nramp 2 (DCT1/DMT1) expressed at the plasma membrane transports iron and other divalent cations into a calcein-accessible cytoplasmic pool. Iron 64-68 solute carrier family 11 member 2 Homo sapiens 14-18 10942769-1 2000 Nramp2, also known as DMT1 and DCT1, is a 12-transmembrane (TM) domain protein responsible for dietary iron uptake in the duodenum and iron acquisition from transferrin in peripheral tissues. Iron 103-107 solute carrier family 11 member 2 Homo sapiens 0-6 10942769-1 2000 Nramp2, also known as DMT1 and DCT1, is a 12-transmembrane (TM) domain protein responsible for dietary iron uptake in the duodenum and iron acquisition from transferrin in peripheral tissues. Iron 103-107 solute carrier family 11 member 2 Homo sapiens 22-26 10942769-1 2000 Nramp2, also known as DMT1 and DCT1, is a 12-transmembrane (TM) domain protein responsible for dietary iron uptake in the duodenum and iron acquisition from transferrin in peripheral tissues. Iron 103-107 solute carrier family 11 member 2 Homo sapiens 31-35 10942769-1 2000 Nramp2, also known as DMT1 and DCT1, is a 12-transmembrane (TM) domain protein responsible for dietary iron uptake in the duodenum and iron acquisition from transferrin in peripheral tissues. Iron 135-139 solute carrier family 11 member 2 Homo sapiens 0-6 10942769-1 2000 Nramp2, also known as DMT1 and DCT1, is a 12-transmembrane (TM) domain protein responsible for dietary iron uptake in the duodenum and iron acquisition from transferrin in peripheral tissues. Iron 135-139 solute carrier family 11 member 2 Homo sapiens 22-26 10942769-1 2000 Nramp2, also known as DMT1 and DCT1, is a 12-transmembrane (TM) domain protein responsible for dietary iron uptake in the duodenum and iron acquisition from transferrin in peripheral tissues. Iron 135-139 solute carrier family 11 member 2 Homo sapiens 31-35 10942769-7 2000 Iron transport at the plasma membrane was time- and pH-dependent, saturable, and proportional to the amount of Nramp2 expression. Iron 0-4 solute carrier family 11 member 2 Homo sapiens 111-117 10942769-8 2000 Iron uptake by Nramp2 at the plasma membrane was into the nonferritin-bound, calcein-accessible so-called "labile iron pool." Iron 0-4 solute carrier family 11 member 2 Homo sapiens 15-21 10942769-8 2000 Iron uptake by Nramp2 at the plasma membrane was into the nonferritin-bound, calcein-accessible so-called "labile iron pool." Iron 114-118 solute carrier family 11 member 2 Homo sapiens 15-21 10942769-11 2000 Monitoring the effect of Nramp2 on the calcein-sensisitve labile iron pool allows a simple, rapid, and nonisotopic approach to the functional study of this protein. Iron 65-69 solute carrier family 11 member 2 Homo sapiens 25-31 11207374-7 2001 Recent identification and characterization of the hemochromatosis protein HFE, the iron importer Nramp2, the iron exporter ferroportin1, and the second transferrin-binding and -transport protein transferrin receptor 2, have demonstrated their important roles in maintaining body"s iron homeostasis. Iron 83-87 solute carrier family 11 member 2 Homo sapiens 97-103 11095929-3 2000 The aim of this study was to determine whether the recently characterized endosomal membrane iron transporter, divalent metal ion transporter-1 (DMT-1), is expressed in human syncytiotrophoblast, and whether its cellular localization would support roles for cytoplasmic and placental-fetal iron transport. Iron 93-97 solute carrier family 11 member 2 Homo sapiens 145-150 11095929-8 2000 Subsequent transport of iron out of the endosome and across the basal membrane to the fetus may occur via DMT-1. Iron 24-28 solute carrier family 11 member 2 Homo sapiens 106-111 10911382-8 2000 Ferrous iron uptake is facilitated by DMT-1 (Nramp-2, DCT-1) in a pathway shared with manganese. Iron 8-12 solute carrier family 11 member 2 Homo sapiens 38-43 10984552-1 2000 The recent discovery of HFE, the MHC-Class-I-like gene mutated in up to 90% of patients with hereditary haemochromatosis, and the gene encoding the Nramp2/divalent metal transporter-1 (DMT-1) implicated in ferrous iron transport holds promise for a greater understanding of human iron metabolism. Iron 214-218 solute carrier family 11 member 2 Homo sapiens 148-183 10984552-1 2000 The recent discovery of HFE, the MHC-Class-I-like gene mutated in up to 90% of patients with hereditary haemochromatosis, and the gene encoding the Nramp2/divalent metal transporter-1 (DMT-1) implicated in ferrous iron transport holds promise for a greater understanding of human iron metabolism. Iron 214-218 solute carrier family 11 member 2 Homo sapiens 185-190 10984552-1 2000 The recent discovery of HFE, the MHC-Class-I-like gene mutated in up to 90% of patients with hereditary haemochromatosis, and the gene encoding the Nramp2/divalent metal transporter-1 (DMT-1) implicated in ferrous iron transport holds promise for a greater understanding of human iron metabolism. Iron 280-284 solute carrier family 11 member 2 Homo sapiens 148-183 10984552-1 2000 The recent discovery of HFE, the MHC-Class-I-like gene mutated in up to 90% of patients with hereditary haemochromatosis, and the gene encoding the Nramp2/divalent metal transporter-1 (DMT-1) implicated in ferrous iron transport holds promise for a greater understanding of human iron metabolism. Iron 280-284 solute carrier family 11 member 2 Homo sapiens 185-190 10984552-5 2000 Immunohistochemical staining showed that DMT-1 protein localized to the brush border of human duodenum where it is predicted to serve as the principal transporter of ferrous iron from the intestinal lumen. Iron 174-178 solute carrier family 11 member 2 Homo sapiens 41-46 10984552-7 2000 This interaction may be critical in small-intestinal crypt cells which express HFE, where it may function to modulate their intrinsic iron status thereby programming iron absorption by DMT-1 in the mature enterocyte. Iron 166-170 solute carrier family 11 member 2 Homo sapiens 185-190 10984552-8 2000 In undifferentiated Caco-2 cells, DMT-1 localized to a discrete late endosome compartment distinct from that occupied by HFE where, in addition to brush-border iron uptake, it may function to regulate the availability of iron delivery to intracellular iron pools. Iron 160-164 solute carrier family 11 member 2 Homo sapiens 34-39 10984552-8 2000 In undifferentiated Caco-2 cells, DMT-1 localized to a discrete late endosome compartment distinct from that occupied by HFE where, in addition to brush-border iron uptake, it may function to regulate the availability of iron delivery to intracellular iron pools. Iron 221-225 solute carrier family 11 member 2 Homo sapiens 34-39 10984552-8 2000 In undifferentiated Caco-2 cells, DMT-1 localized to a discrete late endosome compartment distinct from that occupied by HFE where, in addition to brush-border iron uptake, it may function to regulate the availability of iron delivery to intracellular iron pools. Iron 221-225 solute carrier family 11 member 2 Homo sapiens 34-39 10984552-10 2000 Identification of the molecular interactions of HFE with DMT-1 and other key components of the iron transport pathway has implications for a mechanistic understanding of the pathophysiology of human iron storage diseases as well as the regulation of normal iron balance. Iron 95-99 solute carrier family 11 member 2 Homo sapiens 57-62 10984552-10 2000 Identification of the molecular interactions of HFE with DMT-1 and other key components of the iron transport pathway has implications for a mechanistic understanding of the pathophysiology of human iron storage diseases as well as the regulation of normal iron balance. Iron 199-203 solute carrier family 11 member 2 Homo sapiens 57-62 10984552-10 2000 Identification of the molecular interactions of HFE with DMT-1 and other key components of the iron transport pathway has implications for a mechanistic understanding of the pathophysiology of human iron storage diseases as well as the regulation of normal iron balance. Iron 199-203 solute carrier family 11 member 2 Homo sapiens 57-62 10966897-1 2000 BACKGROUND: Divalent metal transporter 1 (DMT1), HFE, and stimulator of iron transport (SFT) are transmembrane proteins that have been implicated in the regulation of iron homeostasis. Iron 72-76 solute carrier family 11 member 2 Homo sapiens 12-40 10966897-1 2000 BACKGROUND: Divalent metal transporter 1 (DMT1), HFE, and stimulator of iron transport (SFT) are transmembrane proteins that have been implicated in the regulation of iron homeostasis. Iron 72-76 solute carrier family 11 member 2 Homo sapiens 42-46 10966897-1 2000 BACKGROUND: Divalent metal transporter 1 (DMT1), HFE, and stimulator of iron transport (SFT) are transmembrane proteins that have been implicated in the regulation of iron homeostasis. Iron 167-171 solute carrier family 11 member 2 Homo sapiens 12-40 10966897-1 2000 BACKGROUND: Divalent metal transporter 1 (DMT1), HFE, and stimulator of iron transport (SFT) are transmembrane proteins that have been implicated in the regulation of iron homeostasis. Iron 167-171 solute carrier family 11 member 2 Homo sapiens 42-46 10966897-2 2000 OBJECTIVE: The objective of this study was to investigate whether absorption and transepithelial movement of iron correlated with gene expression of DMT1, HFE, and SFT in an experimental model of human absorptive enterocytes. Iron 109-113 solute carrier family 11 member 2 Homo sapiens 149-153 10966897-5 2000 In the absence of serum, iron treatment was associated with a reduction of DMT1 expression by 50% at 72 and 168 h. HFE expression was dependent on serum, but iron treatment did not alter HFE expression. Iron 25-29 solute carrier family 11 member 2 Homo sapiens 75-79 10911382-8 2000 Ferrous iron uptake is facilitated by DMT-1 (Nramp-2, DCT-1) in a pathway shared with manganese. Iron 8-12 solute carrier family 11 member 2 Homo sapiens 45-52 10911382-8 2000 Ferrous iron uptake is facilitated by DMT-1 (Nramp-2, DCT-1) in a pathway shared with manganese. Iron 8-12 solute carrier family 11 member 2 Homo sapiens 54-59 17024033-9 2000 Nramp2, also called DMT1 (divalent metal ion transporter), seems to be a major regulator of transferrin-independent, nonheme iron uptake. Iron 125-129 solute carrier family 11 member 2 Homo sapiens 0-6 10861241-1 2000 Natural resistance-associated macrophage protein 2 (Nramp2) has been suggested to be involved in transferrin-independent iron uptake. Iron 121-125 solute carrier family 11 member 2 Homo sapiens 0-50 10861241-1 2000 Natural resistance-associated macrophage protein 2 (Nramp2) has been suggested to be involved in transferrin-independent iron uptake. Iron 121-125 solute carrier family 11 member 2 Homo sapiens 52-58 10751401-0 2000 Human NRAMP2/DMT1, which mediates iron transport across endosomal membranes, is localized to late endosomes and lysosomes in HEp-2 cells. Iron 34-38 solute carrier family 11 member 2 Homo sapiens 6-12 10751401-0 2000 Human NRAMP2/DMT1, which mediates iron transport across endosomal membranes, is localized to late endosomes and lysosomes in HEp-2 cells. Iron 34-38 solute carrier family 11 member 2 Homo sapiens 13-17 17024033-9 2000 Nramp2, also called DMT1 (divalent metal ion transporter), seems to be a major regulator of transferrin-independent, nonheme iron uptake. Iron 125-129 solute carrier family 11 member 2 Homo sapiens 20-24 10625641-0 2000 Nramp2 expression is associated with pH-dependent iron uptake across the apical membrane of human intestinal Caco-2 cells. Iron 50-54 solute carrier family 11 member 2 Homo sapiens 0-6 10625641-4 2000 Here we demonstrate that Nramp2 is expressed in the apical membrane of the human intestinal epithelial cell line, Caco 2 TC7, and is associated with functional iron transport in these cells with a substrate preference for iron over other divalent cations. Iron 160-164 solute carrier family 11 member 2 Homo sapiens 25-31 10625641-4 2000 Here we demonstrate that Nramp2 is expressed in the apical membrane of the human intestinal epithelial cell line, Caco 2 TC7, and is associated with functional iron transport in these cells with a substrate preference for iron over other divalent cations. Iron 222-226 solute carrier family 11 member 2 Homo sapiens 25-31 10625641-6 2000 Taken together, these data suggest that the expression of the Nramp2 transporter in human enterocytes may play an important role in intestinal iron absorption. Iron 143-147 solute carrier family 11 member 2 Homo sapiens 62-68 10522550-8 1999 However, studies with blocking antibodies, observations in rodents with disorders of iron metabolism, and studies in tissue culture cells suggest that the DCT-1 pathway is dominant in embryonic cells and is involved with cellular uptake of ferrous iron, whereas the mobilferrin-integrin pathway facilitates absorption of dietary inorganic ferric iron. Iron 248-252 solute carrier family 11 member 2 Homo sapiens 155-160 10582331-5 1999 isolated DMT1 through an expression cloning strategy looking for mRNAs that stimulated iron uptake by Xenopus oocytes. Iron 87-91 solute carrier family 11 member 2 Homo sapiens 9-13 10582331-6 1999 Taken together, these data indicate that the twelve transmembrane domain protein DMT1 transfers iron across the apical surface of intestinal cells and out of transferrin cycle endosomes. Iron 96-100 solute carrier family 11 member 2 Homo sapiens 81-85 10582331-7 1999 Human DMT1 may be a good target for pharmacological intervention in patients with iron overload disorders attributable to increased iron absorption. Iron 82-86 solute carrier family 11 member 2 Homo sapiens 6-10 9915882-1 1999 The newly identified hemochromatosis gene, HFE, and a candidate iron transporter gene, Nramp2, have been proposed as key factors responsible for the regulation of intestinal iron absorption. Iron 64-68 solute carrier family 11 member 2 Homo sapiens 87-93 10417735-10 1999 The Nramp2 protein was subsequently shown to be the major transferrin-independent iron uptake system of the intestine. Iron 82-86 solute carrier family 11 member 2 Homo sapiens 4-10 10382697-2 1999 Iron absorption is regulated by the duodenal metal transporter, DMT-1, also called NRAMP-2. Iron 0-4 solute carrier family 11 member 2 Homo sapiens 64-69 10382697-2 1999 Iron absorption is regulated by the duodenal metal transporter, DMT-1, also called NRAMP-2. Iron 0-4 solute carrier family 11 member 2 Homo sapiens 83-90 10382697-11 1999 INTERPRETATION: Increased NRAMP-2 mRNA expression in duodenal mucosa of patients with hereditary haemochromatosis may promote duodenal iron uptake and lead to iron overload. Iron 135-139 solute carrier family 11 member 2 Homo sapiens 26-33 10382697-11 1999 INTERPRETATION: Increased NRAMP-2 mRNA expression in duodenal mucosa of patients with hereditary haemochromatosis may promote duodenal iron uptake and lead to iron overload. Iron 159-163 solute carrier family 11 member 2 Homo sapiens 26-33 9915882-3 1999 Thus, we examined whether the expression of the HFE and Nramp2 genes are regulated by iron status in the human intestinal cell line Caco-2. Iron 86-90 solute carrier family 11 member 2 Homo sapiens 56-62 10429185-0 1999 The effect of intracellular iron concentration and nitrogen monoxide on Nramp2 expression and non-transferrin-bound iron uptake. Iron 28-32 solute carrier family 11 member 2 Homo sapiens 72-78 10429185-1 1999 Recent studies have demonstrated that the protein product (natural resistance associated macrophage protein 2, Nramp2) encoded by the gene Nramp2 acts as an Fe transporter involved in the uptake of Fe from transferrin (Tf) and low Mr Fe complexes. Iron 157-159 solute carrier family 11 member 2 Homo sapiens 59-109 10429185-1 1999 Recent studies have demonstrated that the protein product (natural resistance associated macrophage protein 2, Nramp2) encoded by the gene Nramp2 acts as an Fe transporter involved in the uptake of Fe from transferrin (Tf) and low Mr Fe complexes. Iron 157-159 solute carrier family 11 member 2 Homo sapiens 111-117 10429185-1 1999 Recent studies have demonstrated that the protein product (natural resistance associated macrophage protein 2, Nramp2) encoded by the gene Nramp2 acts as an Fe transporter involved in the uptake of Fe from transferrin (Tf) and low Mr Fe complexes. Iron 157-159 solute carrier family 11 member 2 Homo sapiens 139-145 10429185-1 1999 Recent studies have demonstrated that the protein product (natural resistance associated macrophage protein 2, Nramp2) encoded by the gene Nramp2 acts as an Fe transporter involved in the uptake of Fe from transferrin (Tf) and low Mr Fe complexes. Iron 198-200 solute carrier family 11 member 2 Homo sapiens 59-109 10429185-1 1999 Recent studies have demonstrated that the protein product (natural resistance associated macrophage protein 2, Nramp2) encoded by the gene Nramp2 acts as an Fe transporter involved in the uptake of Fe from transferrin (Tf) and low Mr Fe complexes. Iron 198-200 solute carrier family 11 member 2 Homo sapiens 111-117 10429185-1 1999 Recent studies have demonstrated that the protein product (natural resistance associated macrophage protein 2, Nramp2) encoded by the gene Nramp2 acts as an Fe transporter involved in the uptake of Fe from transferrin (Tf) and low Mr Fe complexes. Iron 198-200 solute carrier family 11 member 2 Homo sapiens 139-145 10429185-2 1999 Interestingly, there are two splice variants of Nramp2, one with a putative iron-responsive element (IRE) in its 3" untranslated region (UTR) and another without. Iron 76-80 solute carrier family 11 member 2 Homo sapiens 48-54 10429185-3 1999 Due to the importance of Nramp2 in Fe transport, and the presence of an IRE in its 3"-UTR, we have examined the effect of Fe-deprivation, Fe-loading, and nitrogen monoxide on the expression of Nramp2 mRNA. Iron 122-124 solute carrier family 11 member 2 Homo sapiens 193-199 10429185-3 1999 Due to the importance of Nramp2 in Fe transport, and the presence of an IRE in its 3"-UTR, we have examined the effect of Fe-deprivation, Fe-loading, and nitrogen monoxide on the expression of Nramp2 mRNA. Iron 122-124 solute carrier family 11 member 2 Homo sapiens 193-199 10429185-10 1999 As Nramp2 can transport Fe from non-Tf-bound Fe, the effect of preincubation with DFO and FAC was also examined on Fe uptake from [59Fe]nitrilotriacetate and [59Fe]citrate. Iron 24-26 solute carrier family 11 member 2 Homo sapiens 3-9 10429185-10 1999 As Nramp2 can transport Fe from non-Tf-bound Fe, the effect of preincubation with DFO and FAC was also examined on Fe uptake from [59Fe]nitrilotriacetate and [59Fe]citrate. Iron 45-47 solute carrier family 11 member 2 Homo sapiens 3-9 10429185-10 1999 As Nramp2 can transport Fe from non-Tf-bound Fe, the effect of preincubation with DFO and FAC was also examined on Fe uptake from [59Fe]nitrilotriacetate and [59Fe]citrate. Iron 45-47 solute carrier family 11 member 2 Homo sapiens 3-9 10361139-0 1999 Cellular and subcellular localization of the Nramp2 iron transporter in the intestinal brush border and regulation by dietary iron. Iron 52-56 solute carrier family 11 member 2 Homo sapiens 45-51 10361139-1 1999 Genetic studies in animal models of microcytic anemia and biochemical studies of transport have implicated the Nramp2 gene in iron transport. Iron 126-130 solute carrier family 11 member 2 Homo sapiens 111-117 10361139-2 1999 Nramp2 generates two alternatively spliced mRNAs that differ at their 3" untranslated region by the presence or absence of an iron-response element (IRE) and that encode two proteins with distinct carboxy termini. Iron 126-130 solute carrier family 11 member 2 Homo sapiens 0-6 10361139-6 1999 Dietary iron starvation results in a dramatic upregulation of the Nramp2 isoform I in the proximal portion of the duodenum only, whereas expression in the rest of the small intestine and in kidney remains largely unchanged in response to the lack of dietary iron. Iron 8-12 solute carrier family 11 member 2 Homo sapiens 66-72 10361139-7 1999 In proximal duodenum, immunostaining studies of tissue sections show that Nramp2 protein expression is abundant under iron deplete condition and limited to the villi and is absent in the crypts. Iron 118-122 solute carrier family 11 member 2 Homo sapiens 74-80 10361139-10 1999 These results strongly suggest that Nramp2 is indeed responsible for transferrin-independent iron uptake in the duodenum. Iron 93-97 solute carrier family 11 member 2 Homo sapiens 36-42 10049947-3 1999 Nramp2 has a much broader range of tissue expression and mutations at Nramp2 result in iron deficiency, indicating a role for Nramp2 in iron metabolism. Iron 87-91 solute carrier family 11 member 2 Homo sapiens 70-76 10049947-3 1999 Nramp2 has a much broader range of tissue expression and mutations at Nramp2 result in iron deficiency, indicating a role for Nramp2 in iron metabolism. Iron 87-91 solute carrier family 11 member 2 Homo sapiens 70-76 10049947-9 1999 These findings suggest that Nramp2 plays a key role in the metabolism of transferrin-bound iron by transporting free Fe2+ across the endosomal membrane and into the cytoplasm. Iron 91-95 solute carrier family 11 member 2 Homo sapiens 28-34 9915882-4 1999 HFE mRNA and HFE protein were increased and Nramp2 mRNA was decreased by increasing cellular iron status in Caco-2 cells. Iron 93-97 solute carrier family 11 member 2 Homo sapiens 44-50 9915882-8 1999 This reciprocal modification of the HFE and Nramp2 gene expression during both iron treatment and cell differentiation in Caco-2 cells is consistent with an opposing role for these proteins in homeostatic regulation of human intestinal iron absorption. Iron 79-83 solute carrier family 11 member 2 Homo sapiens 44-50 9915882-8 1999 This reciprocal modification of the HFE and Nramp2 gene expression during both iron treatment and cell differentiation in Caco-2 cells is consistent with an opposing role for these proteins in homeostatic regulation of human intestinal iron absorption. Iron 236-240 solute carrier family 11 member 2 Homo sapiens 44-50 9521625-9 1998 The divalent cation transporter 1 (DCT1) is regulated by iron, but exhibits transport activity for a number of trace elements including zinc. Iron 57-61 solute carrier family 11 member 2 Homo sapiens 4-33 9808632-5 1998 Nramp2 is likely to be the membrane transporter that functions in controlling iron entry across the apical membrane and in the export of iron out of endosomal vesicles. Iron 78-82 solute carrier family 11 member 2 Homo sapiens 0-6 9808632-5 1998 Nramp2 is likely to be the membrane transporter that functions in controlling iron entry across the apical membrane and in the export of iron out of endosomal vesicles. Iron 137-141 solute carrier family 11 member 2 Homo sapiens 0-6 9808632-6 1998 The observation that the expression of both HFE and Nramp2 mRNAs are reciprocally regulated by cellular iron status in Caco-2 cells, a human intestinal cell line, lends additional credence to the notion that these proteins may work in concert to regulate intestinal iron absorption. Iron 104-108 solute carrier family 11 member 2 Homo sapiens 52-58 9808632-6 1998 The observation that the expression of both HFE and Nramp2 mRNAs are reciprocally regulated by cellular iron status in Caco-2 cells, a human intestinal cell line, lends additional credence to the notion that these proteins may work in concert to regulate intestinal iron absorption. Iron 266-270 solute carrier family 11 member 2 Homo sapiens 52-58 9642100-1 1998 Nramp2 is a gene encoding a transmembrane protein that is important in metal transport, in particular iron. Iron 102-106 solute carrier family 11 member 2 Homo sapiens 0-6 9642100-3 1998 Nramp2 contains a classical iron responsive element in the 3" untranslated region that confers iron dependent mRNA stabilization. Iron 28-32 solute carrier family 11 member 2 Homo sapiens 0-6 9642100-3 1998 Nramp2 contains a classical iron responsive element in the 3" untranslated region that confers iron dependent mRNA stabilization. Iron 95-99 solute carrier family 11 member 2 Homo sapiens 0-6 9642100-4 1998 In this report, we describe a splice variant form of human nramp2 that has the carboxyl terminal 18 amino acids substituted with 25 novel amino acids and has a new 3" untranslated region lacking a classical iron-responsive element. Iron 207-211 solute carrier family 11 member 2 Homo sapiens 59-65 9658732-11 1998 However, other proteins are involved in iron uptake, as the recently cloned Nramp2, the first iron transporter discovered in mammalians. Iron 40-44 solute carrier family 11 member 2 Homo sapiens 76-82 9658732-12 1998 Nramp2 has a recognized role both in the intestinal iron uptake and in the iron transport within the erythroblast. Iron 52-56 solute carrier family 11 member 2 Homo sapiens 0-6 9658732-12 1998 Nramp2 has a recognized role both in the intestinal iron uptake and in the iron transport within the erythroblast. Iron 75-79 solute carrier family 11 member 2 Homo sapiens 0-6 9729418-1 1998 New findings on the role of LfR (lactotransferrin receptor), MTf (melanotransferrin), CP (ceruloplasmin) and DCT1 (Divalent Cation Transporter) in brain iron transport, obtained during the past 3 years, are important advances in the fields of physiology and pathophysiology of brain iron metabolism. Iron 153-157 solute carrier family 11 member 2 Homo sapiens 109-113 9729418-3 1998 Further studies on the involvement of LfR, MTf and DCT1 in iron uptake by and CP in iron egress from different types of brain cells as well as control mechanisms of expression of these proteins in the brain are critical for elucidating the causes of excessive accumulation of iron in the brain and neuronal death in neurodegenerative diseases. Iron 59-63 solute carrier family 11 member 2 Homo sapiens 51-55 9729418-3 1998 Further studies on the involvement of LfR, MTf and DCT1 in iron uptake by and CP in iron egress from different types of brain cells as well as control mechanisms of expression of these proteins in the brain are critical for elucidating the causes of excessive accumulation of iron in the brain and neuronal death in neurodegenerative diseases. Iron 84-88 solute carrier family 11 member 2 Homo sapiens 51-55 9729418-3 1998 Further studies on the involvement of LfR, MTf and DCT1 in iron uptake by and CP in iron egress from different types of brain cells as well as control mechanisms of expression of these proteins in the brain are critical for elucidating the causes of excessive accumulation of iron in the brain and neuronal death in neurodegenerative diseases. Iron 84-88 solute carrier family 11 member 2 Homo sapiens 51-55 9521625-9 1998 The divalent cation transporter 1 (DCT1) is regulated by iron, but exhibits transport activity for a number of trace elements including zinc. Iron 57-61 solute carrier family 11 member 2 Homo sapiens 35-39 35181698-2 2022 Dietary non-heme iron is physiologically absorbed via the divalent metal transporter-1 (DMT1) pathway. Iron 17-21 solute carrier family 11 member 2 Homo sapiens 58-86 35457224-2 2022 DMT1 has a vital role in iron homeostasis by mediating iron uptake in the intestine and kidneys and by recovering iron from recycling endosomes after transferrin endocytosis. Iron 25-29 solute carrier family 11 member 2 Homo sapiens 0-4 35457224-2 2022 DMT1 has a vital role in iron homeostasis by mediating iron uptake in the intestine and kidneys and by recovering iron from recycling endosomes after transferrin endocytosis. Iron 55-59 solute carrier family 11 member 2 Homo sapiens 0-4 35457224-2 2022 DMT1 has a vital role in iron homeostasis by mediating iron uptake in the intestine and kidneys and by recovering iron from recycling endosomes after transferrin endocytosis. Iron 114-118 solute carrier family 11 member 2 Homo sapiens 0-4 35457224-3 2022 Mutations in SLC11A2 cause an ultra-rare hypochromic microcytic anemia with iron overload (AHMIO1), which has been described in eight patients so far. Iron 76-80 solute carrier family 11 member 2 Homo sapiens 13-20 35457224-3 2022 Mutations in SLC11A2 cause an ultra-rare hypochromic microcytic anemia with iron overload (AHMIO1), which has been described in eight patients so far. Iron 76-80 solute carrier family 11 member 2 Homo sapiens 91-97 9564183-0 1998 Identification of Nramp2 as an iron transport protein: another piece of the intestinal iron absorption puzzle. Iron 31-35 solute carrier family 11 member 2 Homo sapiens 18-24 9564183-0 1998 Identification of Nramp2 as an iron transport protein: another piece of the intestinal iron absorption puzzle. Iron 87-91 solute carrier family 11 member 2 Homo sapiens 18-24 9242408-7 1997 DCT1 is upregulated by dietary iron deficiency, and may represent a key mediator of intestinal iron absorption. Iron 31-35 solute carrier family 11 member 2 Homo sapiens 0-4 34286752-1 2021 A cocktail (1 + 2) dual-fluorescent probe system was developed to realize the real-time visualization of dynamic iron state changes between Fe2+ and Fe3+ at the cellular level and in multicellular organisms, providing insights into the effect of DMT1 and ferroportin on iron regulation. Iron 113-117 solute carrier family 11 member 2 Homo sapiens 246-250 34286752-1 2021 A cocktail (1 + 2) dual-fluorescent probe system was developed to realize the real-time visualization of dynamic iron state changes between Fe2+ and Fe3+ at the cellular level and in multicellular organisms, providing insights into the effect of DMT1 and ferroportin on iron regulation. Iron 270-274 solute carrier family 11 member 2 Homo sapiens 246-250 34360779-1 2021 Pro-inflammatory cytokines promote cellular iron-import through enhanced divalent metal transporter-1 (DMT1) expression in pancreatic beta-cells, consequently cell death. Iron 44-48 solute carrier family 11 member 2 Homo sapiens 73-101 34360779-1 2021 Pro-inflammatory cytokines promote cellular iron-import through enhanced divalent metal transporter-1 (DMT1) expression in pancreatic beta-cells, consequently cell death. Iron 44-48 solute carrier family 11 member 2 Homo sapiens 103-107 34360779-2 2021 Inhibition of beta-cell iron-import by DMT1 silencing protects against apoptosis in animal models of diabetes. Iron 24-28 solute carrier family 11 member 2 Homo sapiens 39-43 35061889-11 2022 Additional experimentation suggested that intestinal DMT1 was required for enhancement of iron transport by the 4AAs. Iron 90-94 solute carrier family 11 member 2 Homo sapiens 53-57 35061889-12 2022 Select AA thus enhance iron absorption by inducing DMT1 trafficking onto the apical membrane of duodenal enterocytes. Iron 23-27 solute carrier family 11 member 2 Homo sapiens 51-55 35181698-2 2022 Dietary non-heme iron is physiologically absorbed via the divalent metal transporter-1 (DMT1) pathway. Iron 17-21 solute carrier family 11 member 2 Homo sapiens 88-92 33925597-4 2021 Dietary iron is taken up by the divalent metal transporter 1 (DMT1) in enterocytes and transported to portal blood via ferroportin (FPN), where it is bound to transferrin and taken up by hepatocytes, macrophages and bone marrow cells via transferrin receptor 1 (TfR1). Iron 8-12 solute carrier family 11 member 2 Homo sapiens 32-60 33925597-4 2021 Dietary iron is taken up by the divalent metal transporter 1 (DMT1) in enterocytes and transported to portal blood via ferroportin (FPN), where it is bound to transferrin and taken up by hepatocytes, macrophages and bone marrow cells via transferrin receptor 1 (TfR1). Iron 8-12 solute carrier family 11 member 2 Homo sapiens 62-66 33834418-6 2021 These effects were all attenuated significantly by the endolysosome-specific iron chelator deferoxamine, by inhibitors of endolysosome-resident two-pore channels and divalent metal transporter-1 (DMT-1), and by inhibitors of mitochondria-resident DMT-1 and mitochondrial permeability transition pores. Iron 77-81 solute carrier family 11 member 2 Homo sapiens 196-201 33834418-6 2021 These effects were all attenuated significantly by the endolysosome-specific iron chelator deferoxamine, by inhibitors of endolysosome-resident two-pore channels and divalent metal transporter-1 (DMT-1), and by inhibitors of mitochondria-resident DMT-1 and mitochondrial permeability transition pores. Iron 77-81 solute carrier family 11 member 2 Homo sapiens 247-252 33536631-4 2021 Variants at DUOX2, F5, SLC11A2 and TMPRSS6 associate with iron deficiency anemia, while variants at TF, HFE, TFR2 and TMPRSS6 associate with iron overload. Iron 58-62 solute carrier family 11 member 2 Homo sapiens 23-30 33683742-6 2021 Divalent metal transporter 1(DMT1) decreased significantly, ferroportin 1 and ferritin increased significantly in the liver of UL iron group (p < 0.05). Iron 130-134 solute carrier family 11 member 2 Homo sapiens 29-33 33450374-6 2021 In accordance with iron-replete condition, proteins involved in iron uptake, transport and storage including divalent metal ion transporter 1 (DMT1), transferrin receptor 1 (TFR1), ferritin, poly(rC)-binding proteins 1 and 2 (PCBP1 and 2) and nuclear factor E2-related factor 2 (NRF2) all increase in ovarian cancer spheroids. Iron 64-68 solute carrier family 11 member 2 Homo sapiens 143-147 33443215-3 2021 Divalent metal transporter 1 (DMT1)-mediated iron overload involves NMDA-induced neurotoxicity in males. Iron 45-49 solute carrier family 11 member 2 Homo sapiens 30-34 33739032-8 2021 Ferrous iron is absorbed in the brush border of duodenal enterocytes through a carrier protein, divalent metal transporter 1 (DMT1). Iron 8-12 solute carrier family 11 member 2 Homo sapiens 96-124 33739032-8 2021 Ferrous iron is absorbed in the brush border of duodenal enterocytes through a carrier protein, divalent metal transporter 1 (DMT1). Iron 8-12 solute carrier family 11 member 2 Homo sapiens 126-130 32668256-6 2020 We identify that ebselen, a selective divalent metal transporter 1 (DMT1) inhibitor and antioxidant, could prevent the observed iron overload phenotypes, supporting the role of DMT1 in iron uptake into the human myocardium. Iron 185-189 solute carrier family 11 member 2 Homo sapiens 177-181 32968192-13 2020 These results indicate that the S-nitrosylation of parkin inhibits its E3 ubiquitin ligase activity for the ubiquitination of DMT1, which contributes to iron accumulation and degenerative process in PD. Iron 153-157 solute carrier family 11 member 2 Homo sapiens 126-130 32450084-2 2020 Iron obtained from daily diet is passed through the intestinal enterocyte apical membrane via divalent metal transporter 1 (DMT1), which is either stored as ferritin or moved into the plasma by hepcidin-ferroportin (Fpn) as an exporter. Iron 0-4 solute carrier family 11 member 2 Homo sapiens 94-122 32450084-2 2020 Iron obtained from daily diet is passed through the intestinal enterocyte apical membrane via divalent metal transporter 1 (DMT1), which is either stored as ferritin or moved into the plasma by hepcidin-ferroportin (Fpn) as an exporter. Iron 0-4 solute carrier family 11 member 2 Homo sapiens 124-128 32152203-6 2020 RESULTS: By investigating PCPG cells with low SDHB levels, we show that pseudohypoxia resulted in elevated expression of iron transport proteins, including transferrin (TF), transferrin receptor 2 (TFR2) and the divalent metal transporter 1 (SLC11A2; DMT1), leading to iron accumulation. Iron 121-125 solute carrier family 11 member 2 Homo sapiens 242-249 32152203-6 2020 RESULTS: By investigating PCPG cells with low SDHB levels, we show that pseudohypoxia resulted in elevated expression of iron transport proteins, including transferrin (TF), transferrin receptor 2 (TFR2) and the divalent metal transporter 1 (SLC11A2; DMT1), leading to iron accumulation. Iron 121-125 solute carrier family 11 member 2 Homo sapiens 251-255 32841622-1 2021 As a member of natural resistance-associated macrophage protein (Nramp) family, Nramp2 conservatively exists in the cell membrane across species and is essential for normal iron homeostasis in an H+-dependent manner. Iron 173-177 solute carrier family 11 member 2 Homo sapiens 80-86 32984336-6 2020 Yet we recently showed iron and manganese import at the OMM involves divalent metal transporter 1 (DMT1), an H+-coupled metal ion transporter. Iron 23-27 solute carrier family 11 member 2 Homo sapiens 69-97 32984336-6 2020 Yet we recently showed iron and manganese import at the OMM involves divalent metal transporter 1 (DMT1), an H+-coupled metal ion transporter. Iron 23-27 solute carrier family 11 member 2 Homo sapiens 99-103 32984336-9 2020 Furthermore, the environmental toxicant cadmium selectively damages kidney mitochondria by "ionic mimicry" utilizing iron and calcium transporters, such as OMM DMT1 or IMM calcium uniporter, and by disrupting the electron transport chain. Iron 20-24 solute carrier family 11 member 2 Homo sapiens 160-164 32379419-0 2020 alpha-Synuclein regulates iron homeostasis via preventing parkin-mediated DMT1 ubiquitylation in Parkinson"s disease models. Iron 26-30 solute carrier family 11 member 2 Homo sapiens 74-78 32083771-0 2020 DMT1 Inhibitors Kill Cancer Stem Cells by Blocking Lysosomal Iron Translocation. Iron 61-65 solute carrier family 11 member 2 Homo sapiens 0-4 32083771-3 2020 Here, inhibitors of the divalent metal transporter 1 (DMT1) have been identified that selectively target CSC by blocking lysosomal iron translocation. Iron 131-135 solute carrier family 11 member 2 Homo sapiens 24-52 32083771-3 2020 Here, inhibitors of the divalent metal transporter 1 (DMT1) have been identified that selectively target CSC by blocking lysosomal iron translocation. Iron 131-135 solute carrier family 11 member 2 Homo sapiens 54-58 32379419-3 2020 Based on our previous findings that up-regulation of divalent metal transporter 1 (DMT1) accounted for the nigral iron accumulation in PD, this raised the question whether alpha-Syn disturbed iron homeostasis by modulating DMT1 expression. Iron 114-118 solute carrier family 11 member 2 Homo sapiens 53-81 32379419-3 2020 Based on our previous findings that up-regulation of divalent metal transporter 1 (DMT1) accounted for the nigral iron accumulation in PD, this raised the question whether alpha-Syn disturbed iron homeostasis by modulating DMT1 expression. Iron 114-118 solute carrier family 11 member 2 Homo sapiens 83-87 33479694-2 2020 SLC11A2 (hDMT1) mediates intestinal iron uptake and its inhibition might be used to treat iron overload diseases such as hereditary hemochromatosis. Iron 36-40 solute carrier family 11 member 2 Homo sapiens 0-7 33479694-2 2020 SLC11A2 (hDMT1) mediates intestinal iron uptake and its inhibition might be used to treat iron overload diseases such as hereditary hemochromatosis. Iron 36-40 solute carrier family 11 member 2 Homo sapiens 9-14 33479694-2 2020 SLC11A2 (hDMT1) mediates intestinal iron uptake and its inhibition might be used to treat iron overload diseases such as hereditary hemochromatosis. Iron 90-94 solute carrier family 11 member 2 Homo sapiens 0-7 33479694-2 2020 SLC11A2 (hDMT1) mediates intestinal iron uptake and its inhibition might be used to treat iron overload diseases such as hereditary hemochromatosis. Iron 90-94 solute carrier family 11 member 2 Homo sapiens 9-14 33479694-3 2020 Here we report a micromolar (IC50 = 1.1 muM) pyrazolyl-pyrimidone inhibitor of radiolabeled iron uptake in hDMT1 overexpressing HEK293 cells acting by a non-competitive mechanism, which however does not affect the electrophysiological properties of the transporter. Iron 92-96 solute carrier family 11 member 2 Homo sapiens 107-112 32184317-4 2020 The airway tissue expression of the iron sequestration molecules divalent metal transporter 1 (DMT1) and transferrin receptor 1 (TFR1) are increased in asthma with TFR1 expression correlating with reduced lung function and increased type 2 (T2) inflammatory responses in the airways. Iron 36-40 solute carrier family 11 member 2 Homo sapiens 65-93 32184317-4 2020 The airway tissue expression of the iron sequestration molecules divalent metal transporter 1 (DMT1) and transferrin receptor 1 (TFR1) are increased in asthma with TFR1 expression correlating with reduced lung function and increased type 2 (T2) inflammatory responses in the airways. Iron 36-40 solute carrier family 11 member 2 Homo sapiens 95-99 32000000-9 2020 This association could be explained by an increased expression of DMT1, a divalent metal transporter that captures higher levels of iron in deficiency states of this metal. Iron 132-136 solute carrier family 11 member 2 Homo sapiens 66-70 32223474-2 2022 During pregnancy, placental transport protein Divalent metal transporter1 (DMT1) plays a crucial role in transit of iron across placenta. Iron 116-120 solute carrier family 11 member 2 Homo sapiens 46-73 32223474-2 2022 During pregnancy, placental transport protein Divalent metal transporter1 (DMT1) plays a crucial role in transit of iron across placenta. Iron 116-120 solute carrier family 11 member 2 Homo sapiens 75-79 32096669-1 2021 Background: During pregnancy, iron is transferred from mother to fetus with placental iron transport proteins (Transferrin receptor, Divalent metal transporter/DMT1, ferroportin/FPN1 and Zyklopen). Iron 30-34 solute carrier family 11 member 2 Homo sapiens 160-164 31871928-1 2019 The DMT1 gene encodes divalent metal transporter 1, a membrane iron transport protein. Iron 63-67 solute carrier family 11 member 2 Homo sapiens 4-8 31721611-0 2020 Increased divalent metal ion transporter-1 (DMT1) and ferroportin-1 (FPN1) expression with enhanced iron absorption in ulcerative colitis human colon. Iron 100-104 solute carrier family 11 member 2 Homo sapiens 44-48 31721611-2 2020 Most iron absorption occurs in the duodenum via divalent metal transporter 1 (DMT1)-mediated uptake and ferroportin 1 (FPN1)-mediated export across the apical and basolateral membranes, respectively. Iron 5-9 solute carrier family 11 member 2 Homo sapiens 48-76 31721611-2 2020 Most iron absorption occurs in the duodenum via divalent metal transporter 1 (DMT1)-mediated uptake and ferroportin 1 (FPN1)-mediated export across the apical and basolateral membranes, respectively. Iron 5-9 solute carrier family 11 member 2 Homo sapiens 78-82 31721611-10 2020 Increased DMT1 expression was associated with enhanced 2-(3-carbamimidoylsulfanylmethyl-benzyl)-isothiourea (CISMBI, DMT1 specific inhibitor)-sensitive 59Fe uptake in UC human colon. Iron 152-156 solute carrier family 11 member 2 Homo sapiens 10-14 31721611-10 2020 Increased DMT1 expression was associated with enhanced 2-(3-carbamimidoylsulfanylmethyl-benzyl)-isothiourea (CISMBI, DMT1 specific inhibitor)-sensitive 59Fe uptake in UC human colon. Iron 152-156 solute carrier family 11 member 2 Homo sapiens 117-121 31106422-3 2019 Iron enters the cardiomyocyte through the classic pathway, by binding to the transferrin 1 receptor (TfR1), but also through other routes: T-type calcium channel (TTCC), divalent metal transporter 1 (DMT1), L-type calcium channel (LTCC), Zrt-, Irt-like Proteins (ZIP) 8 and 14. Iron 0-4 solute carrier family 11 member 2 Homo sapiens 170-198 31106422-3 2019 Iron enters the cardiomyocyte through the classic pathway, by binding to the transferrin 1 receptor (TfR1), but also through other routes: T-type calcium channel (TTCC), divalent metal transporter 1 (DMT1), L-type calcium channel (LTCC), Zrt-, Irt-like Proteins (ZIP) 8 and 14. Iron 0-4 solute carrier family 11 member 2 Homo sapiens 200-204 31368012-2 2019 Divalent metal-ion transporter (DMT1) is the major iron importer in enterocytes and erythroblasts. Iron 51-55 solute carrier family 11 member 2 Homo sapiens 32-36 31626992-0 2019 Site-specific intestinal DMT1 silencing to mitigate iron absorption using pH-sensitive multi-compartmental nanoparticulate oral delivery system. Iron 52-56 solute carrier family 11 member 2 Homo sapiens 25-29 31626992-3 2019 While the divalent metal transporter 1 (DMT1) plays a well-established role in the absorption of dietary iron, up-regulation of intestinal DMT1 is associated with iron overload in both humans and rodents. Iron 105-109 solute carrier family 11 member 2 Homo sapiens 10-38 31626992-3 2019 While the divalent metal transporter 1 (DMT1) plays a well-established role in the absorption of dietary iron, up-regulation of intestinal DMT1 is associated with iron overload in both humans and rodents. Iron 105-109 solute carrier family 11 member 2 Homo sapiens 40-44 31626992-3 2019 While the divalent metal transporter 1 (DMT1) plays a well-established role in the absorption of dietary iron, up-regulation of intestinal DMT1 is associated with iron overload in both humans and rodents. Iron 163-167 solute carrier family 11 member 2 Homo sapiens 139-143 31626992-5 2019 Using the gelatin NPs coated with Eudragit L100-55, we demonstrated that DMT1 siRNA-loaded MCPs down-regulated DMT1 mRNA levels in the duodenum, which was consistent with decreased intestinal absorption of orally-administered 59Fe. Iron 227-231 solute carrier family 11 member 2 Homo sapiens 74-78 31316397-1 2019 Divalent metal transporter 1 (DMT1) is a key transporter of iron uptake and delivering in human and animals. Iron 60-64 solute carrier family 11 member 2 Homo sapiens 0-28 31412634-7 2019 Studies in intestinal cell culture models demonstrate that zinc induces iron uptake and transcellular transport via induction of divalent metal iron transporter-1 (DMT1) and ferroportin (FPN1) expression, respectively. Iron 72-76 solute carrier family 11 member 2 Homo sapiens 164-168 31412634-9 2019 Therefore, zinc appears to be modulating the iron metabolism possibly via regulating the DMT1 and FPN1 levels. Iron 45-49 solute carrier family 11 member 2 Homo sapiens 89-93 31412634-10 2019 Herein we critically reviewed the available evidence to hypothesize novel mechanism of Zinc-DMT1/FPN1 axis in regulating intestinal iron absorption and tissue iron accumulation to facilitate future research aimed at understanding the yet elusive mechanisms of iron and zinc interactions. Iron 132-136 solute carrier family 11 member 2 Homo sapiens 92-96 31412634-10 2019 Herein we critically reviewed the available evidence to hypothesize novel mechanism of Zinc-DMT1/FPN1 axis in regulating intestinal iron absorption and tissue iron accumulation to facilitate future research aimed at understanding the yet elusive mechanisms of iron and zinc interactions. Iron 159-163 solute carrier family 11 member 2 Homo sapiens 92-96 31412634-10 2019 Herein we critically reviewed the available evidence to hypothesize novel mechanism of Zinc-DMT1/FPN1 axis in regulating intestinal iron absorption and tissue iron accumulation to facilitate future research aimed at understanding the yet elusive mechanisms of iron and zinc interactions. Iron 159-163 solute carrier family 11 member 2 Homo sapiens 92-96 31316397-1 2019 Divalent metal transporter 1 (DMT1) is a key transporter of iron uptake and delivering in human and animals. Iron 60-64 solute carrier family 11 member 2 Homo sapiens 30-34 31181103-0 2019 Mathematical modeling of the relocation of the divalent metal transporter DMT1 in the intestinal iron absorption process. Iron 97-101 solute carrier family 11 member 2 Homo sapiens 74-78 31181103-5 2019 Two phenomenological models are proposed for the iron absorption process: DMT1"s binary switching mechanism model and DMT1"s swinging-mechanism model, which represent the absorption mechanism for iron uptake in intestinal cells. Iron 49-53 solute carrier family 11 member 2 Homo sapiens 74-78 31181103-5 2019 Two phenomenological models are proposed for the iron absorption process: DMT1"s binary switching mechanism model and DMT1"s swinging-mechanism model, which represent the absorption mechanism for iron uptake in intestinal cells. Iron 49-53 solute carrier family 11 member 2 Homo sapiens 118-122 31181103-5 2019 Two phenomenological models are proposed for the iron absorption process: DMT1"s binary switching mechanism model and DMT1"s swinging-mechanism model, which represent the absorption mechanism for iron uptake in intestinal cells. Iron 196-200 solute carrier family 11 member 2 Homo sapiens 118-122 31092704-5 2019 Zinc-induced DMT1 expression and iron absorption were inhibited by siRNA silencing of DMT1. Iron 33-37 solute carrier family 11 member 2 Homo sapiens 86-90 31181103-8 2019 Both models are capable of capturing the kinetics of iron absorption and represent empirical observations, but the DMT1"s swinging-mechanism model exhibits a better correlation with experimental data and is able to capture the regulatory phenomenon of mucosal block. Iron 53-57 solute carrier family 11 member 2 Homo sapiens 115-119 31181103-9 2019 The DMT1 swinging-mechanism model is the first phenomenological model reported to effectively represent the complexity of the iron absorption process, as it can predict the behavior of iron absorption fluxes after challenging cells with an initial dose of iron, and the reduction in iron uptake observed as a result of mucosal block after a second iron dose. Iron 126-130 solute carrier family 11 member 2 Homo sapiens 4-8 31092704-12 2019 Based on these findings, we conclude that zinc-induced iron absorption involves elevation of DMT1 expression by stabilization of its mRNA, by a PI3K/IRP2-dependent mechanism. Iron 55-59 solute carrier family 11 member 2 Homo sapiens 93-97 31181103-9 2019 The DMT1 swinging-mechanism model is the first phenomenological model reported to effectively represent the complexity of the iron absorption process, as it can predict the behavior of iron absorption fluxes after challenging cells with an initial dose of iron, and the reduction in iron uptake observed as a result of mucosal block after a second iron dose. Iron 185-189 solute carrier family 11 member 2 Homo sapiens 4-8 31181103-9 2019 The DMT1 swinging-mechanism model is the first phenomenological model reported to effectively represent the complexity of the iron absorption process, as it can predict the behavior of iron absorption fluxes after challenging cells with an initial dose of iron, and the reduction in iron uptake observed as a result of mucosal block after a second iron dose. Iron 185-189 solute carrier family 11 member 2 Homo sapiens 4-8 31181103-9 2019 The DMT1 swinging-mechanism model is the first phenomenological model reported to effectively represent the complexity of the iron absorption process, as it can predict the behavior of iron absorption fluxes after challenging cells with an initial dose of iron, and the reduction in iron uptake observed as a result of mucosal block after a second iron dose. Iron 185-189 solute carrier family 11 member 2 Homo sapiens 4-8 31181103-9 2019 The DMT1 swinging-mechanism model is the first phenomenological model reported to effectively represent the complexity of the iron absorption process, as it can predict the behavior of iron absorption fluxes after challenging cells with an initial dose of iron, and the reduction in iron uptake observed as a result of mucosal block after a second iron dose. Iron 185-189 solute carrier family 11 member 2 Homo sapiens 4-8 30870050-3 2019 Divalent metal transporter 1 (DMT1) mediates the uptake of iron into the cell. Iron 59-63 solute carrier family 11 member 2 Homo sapiens 0-28 30132882-4 2019 We demonstrated that the iron exporter ferroportin 1 (FPN1) and iron importer divalent metal transporter 1 (DMT1) were upregulated and iron content was decreased after estrogen treatment for 12 hr in primary cultured astrocytes. Iron 64-68 solute carrier family 11 member 2 Homo sapiens 78-106 30132882-4 2019 We demonstrated that the iron exporter ferroportin 1 (FPN1) and iron importer divalent metal transporter 1 (DMT1) were upregulated and iron content was decreased after estrogen treatment for 12 hr in primary cultured astrocytes. Iron 64-68 solute carrier family 11 member 2 Homo sapiens 108-112 31231185-2 2019 The development of different neurodegenerative diseases is associated with alterations of the intracellular transport of iron and heavy metals, principally mediated by Divalent Metal Transporter 1 (DMT1), responsible for Non-Transferrin Bound Iron transport (NTBI). Iron 121-125 solute carrier family 11 member 2 Homo sapiens 168-196 31231185-2 2019 The development of different neurodegenerative diseases is associated with alterations of the intracellular transport of iron and heavy metals, principally mediated by Divalent Metal Transporter 1 (DMT1), responsible for Non-Transferrin Bound Iron transport (NTBI). Iron 121-125 solute carrier family 11 member 2 Homo sapiens 198-202 31231185-2 2019 The development of different neurodegenerative diseases is associated with alterations of the intracellular transport of iron and heavy metals, principally mediated by Divalent Metal Transporter 1 (DMT1), responsible for Non-Transferrin Bound Iron transport (NTBI). Iron 243-247 solute carrier family 11 member 2 Homo sapiens 168-196 31231185-2 2019 The development of different neurodegenerative diseases is associated with alterations of the intracellular transport of iron and heavy metals, principally mediated by Divalent Metal Transporter 1 (DMT1), responsible for Non-Transferrin Bound Iron transport (NTBI). Iron 243-247 solute carrier family 11 member 2 Homo sapiens 198-202 31231185-4 2019 This review highlights the contribution of DMT1 to the physiological exchange and distribution of body iron and heavy metals during aging and neurodegenerative diseases. Iron 103-107 solute carrier family 11 member 2 Homo sapiens 43-47 31231185-5 2019 DMT1 also mediates the crosstalk between central nervous system and peripheral tissues, by systemic diffusion through the Blood Brain Barrier (BBB), with the involvement of peripheral iron homeostasis in association with inflammation. Iron 184-188 solute carrier family 11 member 2 Homo sapiens 0-4 30870050-3 2019 Divalent metal transporter 1 (DMT1) mediates the uptake of iron into the cell. Iron 59-63 solute carrier family 11 member 2 Homo sapiens 30-34 30415773-1 2018 BACKGROUND INFORMATION: Divalent metal transporter 1 (DMT1) and transferrin receptor (TfR1) are vital proteins for cellular iron uptake. Iron 124-128 solute carrier family 11 member 2 Homo sapiens 24-52 30055235-1 2019 Many past and recent advances in the field of iron metabolism have relied upon the discovery of divalent metal transporter 1, DMT1 in 1997. Iron 46-50 solute carrier family 11 member 2 Homo sapiens 126-130 30055235-2 2019 DMT1 is the major iron transporter and contributes non-heme iron uptake in most types of cell. Iron 18-22 solute carrier family 11 member 2 Homo sapiens 0-4 30055235-3 2019 Each DMT1 isoform exhibits different expression patterns in cell-type specificity and distinct subcellular distribution, which enables cells to uptake both transferrin-bound and non-transferrin-bound irons efficiently. Iron 200-205 solute carrier family 11 member 2 Homo sapiens 5-9 30055235-4 2019 DMT1 expression is regulated by iron through the translational and degradation pathways to ensure iron homeostasis. Iron 32-36 solute carrier family 11 member 2 Homo sapiens 0-4 30055235-4 2019 DMT1 expression is regulated by iron through the translational and degradation pathways to ensure iron homeostasis. Iron 98-102 solute carrier family 11 member 2 Homo sapiens 0-4 30055235-5 2019 It is considered that mammalian iron transporters including DMT1 cannot transport ferric iron but ferrous iron. Iron 98-110 solute carrier family 11 member 2 Homo sapiens 60-64 30055235-8 2019 We summarize DMT1 expression depending on the types of cell or tissue and the function and mechanism of one of the iron chaperones, PCBP2. Iron 115-119 solute carrier family 11 member 2 Homo sapiens 13-17 30527495-3 2019 Accumulating evidence showing a link between carcinogenesis and increased expression of iron import proteins and intracellular iron prompted us to investigate a role of divalent metal-ion transporter-1 (DMT1) that binds and regulates a variety of divalent metals in HCC. Iron 88-92 solute carrier family 11 member 2 Homo sapiens 203-207 30527495-3 2019 Accumulating evidence showing a link between carcinogenesis and increased expression of iron import proteins and intracellular iron prompted us to investigate a role of divalent metal-ion transporter-1 (DMT1) that binds and regulates a variety of divalent metals in HCC. Iron 127-131 solute carrier family 11 member 2 Homo sapiens 203-207 31456203-4 2019 Divalent metal transporter 1 (DMT1) at the apical membrane of intestinal enterocyte brings in non-heme iron from the diet, whereas ferroportin 1 (FPN1) at the basal membrane exports iron into the circulation. Iron 103-107 solute carrier family 11 member 2 Homo sapiens 0-28 31456203-4 2019 Divalent metal transporter 1 (DMT1) at the apical membrane of intestinal enterocyte brings in non-heme iron from the diet, whereas ferroportin 1 (FPN1) at the basal membrane exports iron into the circulation. Iron 103-107 solute carrier family 11 member 2 Homo sapiens 30-34 31456203-6 2019 After binding to transferrin receptor 1 (TfR1), the complex is endocytosed into the cell, where iron enters the cytoplasm via DMT1 on the endosomal membrane. Iron 96-100 solute carrier family 11 member 2 Homo sapiens 126-130 31456203-10 2019 Inside the cells, iron regulatory proteins (IRPs) modulate the expressions of DMT1, TfR1, ferritin, and FPN1 via binding to the iron-responsive element (IRE) in their mRNAs. Iron 18-22 solute carrier family 11 member 2 Homo sapiens 78-82 31456203-10 2019 Inside the cells, iron regulatory proteins (IRPs) modulate the expressions of DMT1, TfR1, ferritin, and FPN1 via binding to the iron-responsive element (IRE) in their mRNAs. Iron 128-132 solute carrier family 11 member 2 Homo sapiens 78-82 30813537-0 2019 Iron Transport from Ferrous Bisglycinate and Ferrous Sulfate in DMT1-Knockout Human Intestinal Caco-2 Cells. Iron 0-4 solute carrier family 11 member 2 Homo sapiens 64-68 30813537-2 2019 The divalent metal transporter 1 (DMT1)-knockout Caco-2 cell line was developed by Crispr-Cas9, and then the cells were treated with ferrous sulfate (FeSO4) or Fe-Gly to observe the labile iron pool and determine their iron transport. Iron 189-193 solute carrier family 11 member 2 Homo sapiens 4-32 30813537-2 2019 The divalent metal transporter 1 (DMT1)-knockout Caco-2 cell line was developed by Crispr-Cas9, and then the cells were treated with ferrous sulfate (FeSO4) or Fe-Gly to observe the labile iron pool and determine their iron transport. Iron 189-193 solute carrier family 11 member 2 Homo sapiens 34-38 30813537-2 2019 The divalent metal transporter 1 (DMT1)-knockout Caco-2 cell line was developed by Crispr-Cas9, and then the cells were treated with ferrous sulfate (FeSO4) or Fe-Gly to observe the labile iron pool and determine their iron transport. Iron 219-223 solute carrier family 11 member 2 Homo sapiens 4-32 30813537-2 2019 The divalent metal transporter 1 (DMT1)-knockout Caco-2 cell line was developed by Crispr-Cas9, and then the cells were treated with ferrous sulfate (FeSO4) or Fe-Gly to observe the labile iron pool and determine their iron transport. Iron 219-223 solute carrier family 11 member 2 Homo sapiens 34-38 30813537-4 2019 DMT1-knockout suppressed the synthesis of ferritin and inhibited the response of iron regulatory protein 1 (IRP-1) and IRP-2 to these two iron sources. Iron 81-85 solute carrier family 11 member 2 Homo sapiens 0-4 30813537-7 2019 These results indicated that iron from Fe-Gly was probably mainly transported into enterocytes via DMT1 like FeSO4; Zip14 may play a certain role in the intestinal iron transport. Iron 29-33 solute carrier family 11 member 2 Homo sapiens 99-103 30415773-1 2018 BACKGROUND INFORMATION: Divalent metal transporter 1 (DMT1) and transferrin receptor (TfR1) are vital proteins for cellular iron uptake. Iron 124-128 solute carrier family 11 member 2 Homo sapiens 54-58 30415773-3 2018 Besides, iron regulatory protein 1 (IRP1) regulates DMT1 and TfR1 by binding to iron-responsive elements (IREs) present in their mRNAs to control cellular iron homeostasis. Iron 9-13 solute carrier family 11 member 2 Homo sapiens 52-56 30415773-3 2018 Besides, iron regulatory protein 1 (IRP1) regulates DMT1 and TfR1 by binding to iron-responsive elements (IREs) present in their mRNAs to control cellular iron homeostasis. Iron 80-84 solute carrier family 11 member 2 Homo sapiens 52-56 30415773-5 2018 Ferrous iron uptake was elevated by DMT1(+IRE) and TfR1 under acute hypoxia. Iron 0-12 solute carrier family 11 member 2 Homo sapiens 36-40 30809369-10 2019 These results indicate that the impairment of the DMT1 traffic machinery affects subcellular iron homeostasis, promoting Fe(ii) leakage at the Golgi and lysosomal accumulation of Fe(ii) through missorting of DMT1. Iron 93-97 solute carrier family 11 member 2 Homo sapiens 50-54 30555088-6 2018 Solute carrier family-11 member-2 (SLC11A2) functions to transport ferrous iron and some divalent metal ions throughout the plasmamembrane and across endosomalmembranes. Iron 75-79 solute carrier family 11 member 2 Homo sapiens 0-33 30555088-6 2018 Solute carrier family-11 member-2 (SLC11A2) functions to transport ferrous iron and some divalent metal ions throughout the plasmamembrane and across endosomalmembranes. Iron 75-79 solute carrier family 11 member 2 Homo sapiens 35-42 30555088-7 2018 Functional polymorphisms in the SLC11A2 gene have been reported to cause excess storage of iron, resulting in iron overload. Iron 91-95 solute carrier family 11 member 2 Homo sapiens 32-39 30555088-7 2018 Functional polymorphisms in the SLC11A2 gene have been reported to cause excess storage of iron, resulting in iron overload. Iron 110-114 solute carrier family 11 member 2 Homo sapiens 32-39 30809369-10 2019 These results indicate that the impairment of the DMT1 traffic machinery affects subcellular iron homeostasis, promoting Fe(ii) leakage at the Golgi and lysosomal accumulation of Fe(ii) through missorting of DMT1. Iron 93-97 solute carrier family 11 member 2 Homo sapiens 208-212 30193229-1 2018 Iron transport through the duodenum is regulated by carrier proteins, one of which is the ubiquitously distributed divalent metal transporter (DMT1) which is responsible for the uptake of iron across the apical surface of the duodenal enterocyte. Iron 0-4 solute carrier family 11 member 2 Homo sapiens 143-147 30193229-1 2018 Iron transport through the duodenum is regulated by carrier proteins, one of which is the ubiquitously distributed divalent metal transporter (DMT1) which is responsible for the uptake of iron across the apical surface of the duodenal enterocyte. Iron 188-192 solute carrier family 11 member 2 Homo sapiens 143-147 30193229-4 2018 The differences in binding modes between ScaDMT1 and hDMT1 were noted for a set of 7 iron containing compounds, including ferrous sulphate. Iron 85-89 solute carrier family 11 member 2 Homo sapiens 53-58 29063108-4 2017 HIFs play a key role in iron metabolism by regulating the expression of iron-related proteins, such as divalent metal transporter 1 (DMT1), ferroportin 1 (FPN1), duodenal cytochrome b (Dcytb), and transferrin receptor (TfR). Iron 24-28 solute carrier family 11 member 2 Homo sapiens 133-137 29110513-9 2018 The results showed that ABS-derived iron influenced transcriptions of iron-regulated marker genes, including divalent metal transporter ( Dmt1), transferrin receptor ( TfR), ankyrin repeat domain 37 ( Ankrd37), and hepcidin ( Hamp) in IDA-induced Caco-2 and HepG2 cells. Iron 36-40 solute carrier family 11 member 2 Homo sapiens 138-142 29110513-9 2018 The results showed that ABS-derived iron influenced transcriptions of iron-regulated marker genes, including divalent metal transporter ( Dmt1), transferrin receptor ( TfR), ankyrin repeat domain 37 ( Ankrd37), and hepcidin ( Hamp) in IDA-induced Caco-2 and HepG2 cells. Iron 70-74 solute carrier family 11 member 2 Homo sapiens 138-142 29082606-8 2018 CONCLUSION: The expression of ferroportin and SLC11A2 is increased in the intestine of patients with T2D in association with iron stores and serum hepcidin levels. Iron 125-129 solute carrier family 11 member 2 Homo sapiens 46-53 29070551-3 2017 Although the absorption of heme iron is poorly understood, nonheme iron is transported across the apical membrane of the intestinal enterocyte by divalent metal-ion transporter 1 (DMT1) and is exported into the circulation via ferroportin 1 (FPN1). Iron 67-71 solute carrier family 11 member 2 Homo sapiens 180-184 29070551-5 2017 Iron-loaded transferrin binds to transferrin receptor 1 on the surface of most body cells, and after endocytosis of the complex, iron enters the cytoplasm via DMT1 in the endosomal membrane. Iron 0-4 solute carrier family 11 member 2 Homo sapiens 159-163 29070551-5 2017 Iron-loaded transferrin binds to transferrin receptor 1 on the surface of most body cells, and after endocytosis of the complex, iron enters the cytoplasm via DMT1 in the endosomal membrane. Iron 129-133 solute carrier family 11 member 2 Homo sapiens 159-163 29070561-10 2017 The major iron transporters in the small intestine divalent metal-ion transporter 1 (DMT1) and ferroportin were not affected by pup iron status at 10 d of age but were strongly affected by iron status at 20 d of age. Iron 10-14 solute carrier family 11 member 2 Homo sapiens 85-89 30104344-0 2018 S-Nitrosylation of Divalent Metal Transporter 1 Enhances Iron Uptake to Mediate Loss of Dopaminergic Neurons and Motoric Deficit. Iron 57-61 solute carrier family 11 member 2 Homo sapiens 19-47 29939292-8 2018 Results: Pea ferritin exposed to gastric pH treatment was degraded, and the released iron was transported into Caco-2 cells by DMT-1. Iron 85-89 solute carrier family 11 member 2 Homo sapiens 127-132 29939292-9 2018 Inhibitors of DMT-1 and nonheme iron absorption reduced iron uptake by 26-40%. Iron 56-60 solute carrier family 11 member 2 Homo sapiens 14-19 29362518-1 2018 Iron transport across the intestinal epithelium is facilitated by the divalent metal transporter 1 (DMT1) on the brush border membrane (BBM). Iron 0-4 solute carrier family 11 member 2 Homo sapiens 70-98 29362518-1 2018 Iron transport across the intestinal epithelium is facilitated by the divalent metal transporter 1 (DMT1) on the brush border membrane (BBM). Iron 0-4 solute carrier family 11 member 2 Homo sapiens 100-104 29054412-5 2017 The role of the endosomal divalent metal transporter 1 (DMT1) in BBB iron acquisition and transport has been questioned. Iron 69-73 solute carrier family 11 member 2 Homo sapiens 26-54 29054412-5 2017 The role of the endosomal divalent metal transporter 1 (DMT1) in BBB iron acquisition and transport has been questioned. Iron 69-73 solute carrier family 11 member 2 Homo sapiens 56-60 29054412-6 2017 Here, we show that inhibition of DMT1 alters the transport of iron and Tf across the endothelial cells. Iron 62-66 solute carrier family 11 member 2 Homo sapiens 33-37 29063108-4 2017 HIFs play a key role in iron metabolism by regulating the expression of iron-related proteins, such as divalent metal transporter 1 (DMT1), ferroportin 1 (FPN1), duodenal cytochrome b (Dcytb), and transferrin receptor (TfR). Iron 72-76 solute carrier family 11 member 2 Homo sapiens 103-131 29063108-4 2017 HIFs play a key role in iron metabolism by regulating the expression of iron-related proteins, such as divalent metal transporter 1 (DMT1), ferroportin 1 (FPN1), duodenal cytochrome b (Dcytb), and transferrin receptor (TfR). Iron 72-76 solute carrier family 11 member 2 Homo sapiens 133-137 28655775-3 2017 Recently, we demonstrated that the iron chaperone poly(rC)-binding protein 2 (PCBP2) can directly receive ferrous iron from DMT1 or transfer iron to the iron exporter, ferroportin 1. Iron 35-39 solute carrier family 11 member 2 Homo sapiens 124-128 29023457-2 2017 Iron enters the enterocytes through an apical divalent metal transporter, DMT1. Iron 0-4 solute carrier family 11 member 2 Homo sapiens 74-78 29023457-3 2017 Different DMT1 transcripts have been identified, depending on the presence of an iron-responsive element that allows DMT1 up-regulation during iron starvation. Iron 81-85 solute carrier family 11 member 2 Homo sapiens 10-14 29023457-3 2017 Different DMT1 transcripts have been identified, depending on the presence of an iron-responsive element that allows DMT1 up-regulation during iron starvation. Iron 81-85 solute carrier family 11 member 2 Homo sapiens 117-121 29023457-3 2017 Different DMT1 transcripts have been identified, depending on the presence of an iron-responsive element that allows DMT1 up-regulation during iron starvation. Iron 143-147 solute carrier family 11 member 2 Homo sapiens 10-14 29023457-3 2017 Different DMT1 transcripts have been identified, depending on the presence of an iron-responsive element that allows DMT1 up-regulation during iron starvation. Iron 143-147 solute carrier family 11 member 2 Homo sapiens 117-121 29023457-4 2017 An intronic DMT1 polymorphism, IVS4+44C>A, has been associated with metal toxicity, and the CC-carriers show high iron levels. Iron 117-121 solute carrier family 11 member 2 Homo sapiens 12-16 29023457-8 2017 Moreover, we found that A-allele is associated to preferential expression of the DMT1 transcripts lacking the iron-responsive element, thus limiting the DMT1 overexpression that normally occurs to respond to iron starvation. Iron 110-114 solute carrier family 11 member 2 Homo sapiens 81-85 29023457-8 2017 Moreover, we found that A-allele is associated to preferential expression of the DMT1 transcripts lacking the iron-responsive element, thus limiting the DMT1 overexpression that normally occurs to respond to iron starvation. Iron 110-114 solute carrier family 11 member 2 Homo sapiens 153-157 29023457-8 2017 Moreover, we found that A-allele is associated to preferential expression of the DMT1 transcripts lacking the iron-responsive element, thus limiting the DMT1 overexpression that normally occurs to respond to iron starvation. Iron 208-212 solute carrier family 11 member 2 Homo sapiens 81-85 29023457-8 2017 Moreover, we found that A-allele is associated to preferential expression of the DMT1 transcripts lacking the iron-responsive element, thus limiting the DMT1 overexpression that normally occurs to respond to iron starvation. Iron 208-212 solute carrier family 11 member 2 Homo sapiens 153-157 29023457-9 2017 DISCUSSION: Possibly, the IVS4+44-AA-related dysregulation of the iron-induced changes in DMT1 expression is not able to impair iron absorption in physiological condition. Iron 66-70 solute carrier family 11 member 2 Homo sapiens 90-94 28655775-2 2017 Moreover, intestinal iron is incorporated as ferrous iron, which is transported via the iron importer, divalent metal transporter 1 (DMT1). Iron 21-25 solute carrier family 11 member 2 Homo sapiens 103-131 28655775-2 2017 Moreover, intestinal iron is incorporated as ferrous iron, which is transported via the iron importer, divalent metal transporter 1 (DMT1). Iron 21-25 solute carrier family 11 member 2 Homo sapiens 133-137 28655775-2 2017 Moreover, intestinal iron is incorporated as ferrous iron, which is transported via the iron importer, divalent metal transporter 1 (DMT1). Iron 53-57 solute carrier family 11 member 2 Homo sapiens 103-131 28655775-2 2017 Moreover, intestinal iron is incorporated as ferrous iron, which is transported via the iron importer, divalent metal transporter 1 (DMT1). Iron 53-57 solute carrier family 11 member 2 Homo sapiens 133-137 28655775-2 2017 Moreover, intestinal iron is incorporated as ferrous iron, which is transported via the iron importer, divalent metal transporter 1 (DMT1). Iron 53-57 solute carrier family 11 member 2 Homo sapiens 103-131 28655775-2 2017 Moreover, intestinal iron is incorporated as ferrous iron, which is transported via the iron importer, divalent metal transporter 1 (DMT1). Iron 53-57 solute carrier family 11 member 2 Homo sapiens 133-137 28655775-3 2017 Recently, we demonstrated that the iron chaperone poly(rC)-binding protein 2 (PCBP2) can directly receive ferrous iron from DMT1 or transfer iron to the iron exporter, ferroportin 1. Iron 114-118 solute carrier family 11 member 2 Homo sapiens 124-128 28655775-3 2017 Recently, we demonstrated that the iron chaperone poly(rC)-binding protein 2 (PCBP2) can directly receive ferrous iron from DMT1 or transfer iron to the iron exporter, ferroportin 1. Iron 114-118 solute carrier family 11 member 2 Homo sapiens 124-128 28754960-1 2017 In humans, the H+-coupled Fe2+ transporter DMT1 (SLC11A2) is essential for proper maintenance of iron homeostasis. Iron 97-101 solute carrier family 11 member 2 Homo sapiens 43-47 28754960-1 2017 In humans, the H+-coupled Fe2+ transporter DMT1 (SLC11A2) is essential for proper maintenance of iron homeostasis. Iron 97-101 solute carrier family 11 member 2 Homo sapiens 49-56 28534724-9 2017 These results indicate that maternal protein restriction during pregnancy and lactation influences growth of female offspring at weaning, reduces duodenal expression of Fe transporters (DMT1 and FPN) and decreases serum Fe level in male weaning piglets. Iron 169-171 solute carrier family 11 member 2 Homo sapiens 186-190 28270217-1 2017 BACKGROUND: Duodenal cytochrome b (DCYTB) is a ferrireductase that functions together with divalent metal transporter 1 (DMT1) to mediate dietary iron reduction and uptake in the duodenum. Iron 146-150 solute carrier family 11 member 2 Homo sapiens 91-119 28424608-3 2017 We observed a significant downregulation of genes related to hemoglobin including, hemoglobin delta (HBD), alpha hemoglobin stabilizing protein (ASHP), genes implicated in iron metabolism including, solute carrier family 11 member 2 (SLC11A2), ferrochelatase (FECH), and erythrocyte-specific genes including erythrocyte membrane protein (EPB42), and 5"-aminolevulinate synthase 2 (ALAS2). Iron 172-176 solute carrier family 11 member 2 Homo sapiens 199-232 28270217-1 2017 BACKGROUND: Duodenal cytochrome b (DCYTB) is a ferrireductase that functions together with divalent metal transporter 1 (DMT1) to mediate dietary iron reduction and uptake in the duodenum. Iron 146-150 solute carrier family 11 member 2 Homo sapiens 121-125 28005311-13 2017 2 and 6 mum ELT increased Tfr1 and Slc11a2 (iron-responsive genes involved in neuronal iron uptake) mRNA levels, indicating neuronal ID. Iron 44-48 solute carrier family 11 member 2 Homo sapiens 35-42 28005311-13 2017 2 and 6 mum ELT increased Tfr1 and Slc11a2 (iron-responsive genes involved in neuronal iron uptake) mRNA levels, indicating neuronal ID. Iron 87-91 solute carrier family 11 member 2 Homo sapiens 35-42 28955733-10 2017 GENERAL SIGNIFICANCE: These results suggest that exogenous PP IX disrupts iron metabolism by decreasing the protein expression levels of PAP7, DMT1 and C/EBPalpha. Iron 74-78 solute carrier family 11 member 2 Homo sapiens 143-147 28046154-1 2016 Two experiments were conducted with 28-d-old commercial male broilers to study the kinetics of iron (Fe) absorption and the effect of Fe treatment on divalent metal transporter 1 (DMT1) and ferroportin 1 (FPN1) mRNA levels in in situ ligated segments from different small intestinal regions of broilers. Iron 134-136 solute carrier family 11 member 2 Homo sapiens 150-178 28367088-13 2017 These results suggest that the increased expression of DMT1 induces iron overload and iron overload induces osteoblast autophagy and apoptosis, thus affecting the pathological processes of osteoporosis. Iron 68-72 solute carrier family 11 member 2 Homo sapiens 55-59 28955733-4 2017 We have demonstrated that PBR-Associated Protein 7 (PAP7) bound to the Iron Responsive Element (IRE) isoform of divalent metal transporter 1 (DMT1). Iron 71-75 solute carrier family 11 member 2 Homo sapiens 112-140 28955733-4 2017 We have demonstrated that PBR-Associated Protein 7 (PAP7) bound to the Iron Responsive Element (IRE) isoform of divalent metal transporter 1 (DMT1). Iron 71-75 solute carrier family 11 member 2 Homo sapiens 142-146 28261264-6 2017 An altered pattern of iron transporters with iron overload is highlighted in BPAN human fibroblasts, supporting for a role of DMT1 in NBIA. Iron 22-26 solute carrier family 11 member 2 Homo sapiens 126-130 27903581-4 2017 In the present study, we assessed the contribution of the metal-ion transporters ZRT/IRT-like protein 14 and 8 (ZIP14 and ZIP8) and divalent metal-ion transporter-1 (DMT1) to iron uptake by human beta-cells. Iron 175-179 solute carrier family 11 member 2 Homo sapiens 166-170 28046154-1 2016 Two experiments were conducted with 28-d-old commercial male broilers to study the kinetics of iron (Fe) absorption and the effect of Fe treatment on divalent metal transporter 1 (DMT1) and ferroportin 1 (FPN1) mRNA levels in in situ ligated segments from different small intestinal regions of broilers. Iron 134-136 solute carrier family 11 member 2 Homo sapiens 180-184 27681840-10 2016 Since anti-inflammatory actions mediated through CB2 would be associated with reduced DMT1 phosphorylation, we postulate that this pathway provides a means to reduce oxidative stress by limiting iron uptake. Iron 195-199 solute carrier family 11 member 2 Homo sapiens 86-90 27681840-1 2016 Divalent metal transporter-1 (DMT1) mediates dietary iron uptake across the intestinal mucosa and facilitates peripheral delivery of iron released by transferrin in the endosome. Iron 53-57 solute carrier family 11 member 2 Homo sapiens 0-28 27681840-1 2016 Divalent metal transporter-1 (DMT1) mediates dietary iron uptake across the intestinal mucosa and facilitates peripheral delivery of iron released by transferrin in the endosome. Iron 53-57 solute carrier family 11 member 2 Homo sapiens 30-34 26911670-6 2016 This result was supported by the observation that two duodenal iron transporters, divalent metal transporter 1 (DMT1) and ferroportin, were downregulated by iron loading, although the levels of expression of ferroportin in iron storage tissues were not changed by iron loading under erythropoietic stimulation by epoetin-beta pegol (C.E.R.A., a long-acting erythropoiesis-stimulating agent). Iron 63-67 solute carrier family 11 member 2 Homo sapiens 82-110 27681840-1 2016 Divalent metal transporter-1 (DMT1) mediates dietary iron uptake across the intestinal mucosa and facilitates peripheral delivery of iron released by transferrin in the endosome. Iron 133-137 solute carrier family 11 member 2 Homo sapiens 0-28 27681840-1 2016 Divalent metal transporter-1 (DMT1) mediates dietary iron uptake across the intestinal mucosa and facilitates peripheral delivery of iron released by transferrin in the endosome. Iron 133-137 solute carrier family 11 member 2 Homo sapiens 30-34 27681840-8 2016 Among kinase inhibitors that affected DMT1-mediated iron uptake, staurosporine also reduced DMT1 phosphorylation confirming a role for serine phosphorylation in iron transport regulation. Iron 52-56 solute carrier family 11 member 2 Homo sapiens 38-42 27646472-0 2016 Activation of ATP-sensitive potassium channels enhances DMT1-mediated iron uptake in SK-N-SH cells in vitro. Iron 70-74 solute carrier family 11 member 2 Homo sapiens 56-60 27646472-1 2016 Iron importer divalent metal transporter 1 (DMT1) plays a crucial role in the nigal iron accumulation in Parkinson"s disease (PD). Iron 84-88 solute carrier family 11 member 2 Homo sapiens 14-42 27646472-1 2016 Iron importer divalent metal transporter 1 (DMT1) plays a crucial role in the nigal iron accumulation in Parkinson"s disease (PD). Iron 84-88 solute carrier family 11 member 2 Homo sapiens 44-48 27646472-5 2016 Therefore, it is of vital importance to study the effects of activation of KATP channels on DMT1-mediated iron uptake function. Iron 106-110 solute carrier family 11 member 2 Homo sapiens 92-96 27646472-6 2016 In the present study, activation of KATP channels by diazoxide resulted in the hyperpolarization of the membrane potential and increased DMT1-mediated iron uptake in SK-N-SH cells. Iron 151-155 solute carrier family 11 member 2 Homo sapiens 137-141 27646472-8 2016 Delayed inactivation of the Fe(2+)-evoked currents by diazoxide was recorded by patch clamp in HEK293 cells, which demonstrated that diazoxide could prolonged DMT1-facilitated iron transport. Iron 176-180 solute carrier family 11 member 2 Homo sapiens 159-163 27302059-5 2016 Subsequently, PCBP2 receives iron from DMT1 and then disengages from the transporter. Iron 29-33 solute carrier family 11 member 2 Homo sapiens 39-43 27117373-14 2016 In turn, endometriosis-related conditions, as iron overload and inflammation (IL-1beta), enhance endometriosis patients endometrial DMT1 expression, creating a vicious circle on DMT-1-modulated pathways. Iron 46-50 solute carrier family 11 member 2 Homo sapiens 132-136 27117373-14 2016 In turn, endometriosis-related conditions, as iron overload and inflammation (IL-1beta), enhance endometriosis patients endometrial DMT1 expression, creating a vicious circle on DMT-1-modulated pathways. Iron 46-50 solute carrier family 11 member 2 Homo sapiens 178-183 27117373-13 2016 CONCLUSION(S): Divalent metal transporter-1 overexpression in endometriosis patients" endometrium can increase iron influx to endometrial cells, inducing oxidative stress-mediated proinflammatory signaling. Iron 111-115 solute carrier family 11 member 2 Homo sapiens 15-43 26911670-6 2016 This result was supported by the observation that two duodenal iron transporters, divalent metal transporter 1 (DMT1) and ferroportin, were downregulated by iron loading, although the levels of expression of ferroportin in iron storage tissues were not changed by iron loading under erythropoietic stimulation by epoetin-beta pegol (C.E.R.A., a long-acting erythropoiesis-stimulating agent). Iron 63-67 solute carrier family 11 member 2 Homo sapiens 112-116 26911670-6 2016 This result was supported by the observation that two duodenal iron transporters, divalent metal transporter 1 (DMT1) and ferroportin, were downregulated by iron loading, although the levels of expression of ferroportin in iron storage tissues were not changed by iron loading under erythropoietic stimulation by epoetin-beta pegol (C.E.R.A., a long-acting erythropoiesis-stimulating agent). Iron 157-161 solute carrier family 11 member 2 Homo sapiens 82-110 26911670-6 2016 This result was supported by the observation that two duodenal iron transporters, divalent metal transporter 1 (DMT1) and ferroportin, were downregulated by iron loading, although the levels of expression of ferroportin in iron storage tissues were not changed by iron loading under erythropoietic stimulation by epoetin-beta pegol (C.E.R.A., a long-acting erythropoiesis-stimulating agent). Iron 157-161 solute carrier family 11 member 2 Homo sapiens 112-116 26911670-6 2016 This result was supported by the observation that two duodenal iron transporters, divalent metal transporter 1 (DMT1) and ferroportin, were downregulated by iron loading, although the levels of expression of ferroportin in iron storage tissues were not changed by iron loading under erythropoietic stimulation by epoetin-beta pegol (C.E.R.A., a long-acting erythropoiesis-stimulating agent). Iron 157-161 solute carrier family 11 member 2 Homo sapiens 82-110 26911670-6 2016 This result was supported by the observation that two duodenal iron transporters, divalent metal transporter 1 (DMT1) and ferroportin, were downregulated by iron loading, although the levels of expression of ferroportin in iron storage tissues were not changed by iron loading under erythropoietic stimulation by epoetin-beta pegol (C.E.R.A., a long-acting erythropoiesis-stimulating agent). Iron 157-161 solute carrier family 11 member 2 Homo sapiens 112-116 26827808-5 2016 Iron depletion increased expression of iron-regulated proteins (TfR, transferrin receptor and DMT1, divalent metal transporter, as predicted, but it also promoted a marked reduction in growth and proliferation of Caco-2 cells. Iron 0-4 solute carrier family 11 member 2 Homo sapiens 94-98 26827808-5 2016 Iron depletion increased expression of iron-regulated proteins (TfR, transferrin receptor and DMT1, divalent metal transporter, as predicted, but it also promoted a marked reduction in growth and proliferation of Caco-2 cells. Iron 39-43 solute carrier family 11 member 2 Homo sapiens 94-98