Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Front Cell Dev Biol ; 11: 1151108, 2023.
Article in English | MEDLINE | ID: mdl-37325564

ABSTRACT

Cadmium (Cd) is a toxic heavy metal found throughout the environment and one of the top ten toxicants of major public health concern identified by the World Health Organization. In utero Cd exposure causes fetal growth restriction, malformation, and spontaneous abortion; however, the mechanisms by which Cd impacts these outcomes are poorly understood. Cd accumulates in the placenta, suggesting that these negative outcomes may be a consequence of disrupted placental function and placental insufficiency. To understand the impact of Cd on gene expression within the placenta, we developed a mouse model of Cd-induced fetal growth restriction through maternal consumption of CdCl2 and performed RNA-seq on control and CdCl2 exposed placentae. The top differentially expressed transcript was the Tcl1 Upstream Neuron-Associated (Tuna) long non-coding RNA, which was upregulated over 25-fold in CdCl2 exposed placentae. Tuna has been shown to be critical for neural stem cell differentiation. However, within the placenta, there is no evidence that Tuna is normally expressed or functional at any developmental stage. To determine the spatial expression of Cd-activated Tuna within the placenta, we used in situ hybridization as well as placental layer-specific RNA isolation and analysis. Both methods confirmed the absence of Tuna expression in control samples and determined that Cd-induced Tuna expression is specific to the junctional zone. Since many lncRNAs regulate gene expression, we hypothesized that Tuna forms part of the mechanism of Cd-induced transcriptomic changes. To test this, we over-expressed Tuna in cultured choriocarcinoma cells and compared gene expression profiles to those of control and CdCl2 exposed cells. We demonstrate significant overlap between genes activated by Tuna overexpression and genes activated by CdCl2 exposure, with enrichment in the NRF2-mediated oxidative stress response. Herein we analyze the NRF2 pathway and show that Tuna increases NRF2/NRF2 both at the transcript and protein levels. Tuna drives increased NRF2 target gene expression, a result that is abrogated with the use of an NRF2 inhibitor, confirming that Tuna activates oxidative stress response genes through this pathway. This work identifies the lncRNA Tuna as a potential novel player in Cd-induced placental insufficiency.

2.
Int J Mol Sci ; 24(4)2023 Feb 10.
Article in English | MEDLINE | ID: mdl-36835018

ABSTRACT

Investigation of RNA- and DNA-binding proteins to a defined regulatory sequence, such as an AU-rich RNA and a DNA enhancer element, is important for understanding gene regulation through their interactions. For in vitro binding studies, an electrophoretic mobility shift assay (EMSA) was widely used in the past. In line with the trend toward using non-radioactive materials in various bioassays, end-labeled biotinylated RNA and DNA oligonucleotides can be more practical probes to study protein-RNA and protein-DNA interactions; thereby, the binding complexes can be pulled down with streptavidin-conjugated resins and identified by Western blotting. However, setting up RNA and DNA pull-down assays with biotinylated probes in optimum protein binding conditions remains challenging. Here, we demonstrate the step-by step optimization of pull-down for IRP (iron-responsive-element-binding protein) with a 5'-biotinylated stem-loop IRE (iron-responsive element) RNA, HuR, and AUF1 with an AU-rich RNA element and Nrf2 binding to an antioxidant-responsive element (ARE) enhancer in the human ferritin H gene. This study was designed to address key technical questions in RNA and DNA pull-down assays: (1) how much RNA and DNA probes we should use; (2) what binding buffer and cell lysis buffer we can use; (3) how to verify the specific interaction; (4) what streptavidin resin (agarose or magnetic beads) works; and (5) what Western blotting results we can expect from varying to optimum conditions. We anticipate that our optimized pull-down conditions can be applicable to other RNA- and DNA-binding proteins along with emerging non-coding small RNA-binding proteins for their in vitro characterization.


Subject(s)
Biotinylation , Carrier Proteins , Chromatography, Affinity , DNA , RNA , Humans , Carrier Proteins/chemistry , DNA/chemistry , Iron/metabolism , Iron Regulatory Protein 1/chemistry , Iron Regulatory Protein 2/chemistry , NF-E2-Related Factor 2/chemistry , RNA/chemistry , Streptavidin/metabolism , Chromatography, Affinity/methods
3.
PLoS One ; 15(7): e0235563, 2020.
Article in English | MEDLINE | ID: mdl-32645092

ABSTRACT

Western blotting has been widely used for investigation of protein expression, posttranslational modifications, and interactions. Because western blotting usually involves heat-denaturation of samples prior to gel loading, clarification of detailed procedures for sample preparation have been omitted or neglected in many publications. We show here the case that even excellent primary antibodies failed to detect a specific protein of interest due to a routine heating practice of protein samples. We performed western blotting for transmembrane iron transporter proteins; SLC11A2 (divalent metal transporter 1, DMT1), SLC40A1 (ferroportin 1, Fpn1), and transferrin receptor-1 (TfR1), along with cytoplasmic iron storage protein ferritin H. Our results in 12 human culture cell lysates indicated that only unheated samples prior to gel loading gave rise to clear resolution of DMT1 protein, while heated samples (95°C, 5min) caused the loss of resolution due to DMT1 protein aggregates. Unheated samples also resulted in better resolution for Fpn1 and TfR1 western blots. Conversely, only heated samples allowed to detect ferritin H, otherwise ferritin polymers failed to get into the gel. Neither different lysis/sample loading buffers nor sonication improved the resolution of DMT1 and Fpn1 western blots. Thus, heating samples most critically affected the outcome of western blotting, suggesting the similar cases for thousands of other transmembrane and heat-sensitive proteins.


Subject(s)
Blotting, Western/methods , Cation Transport Proteins/analysis , Cell Fractionation/methods , A549 Cells , Antibodies/immunology , Caco-2 Cells , Cation Transport Proteins/immunology , HEK293 Cells , HeLa Cells , Hep G2 Cells , Humans , Jurkat Cells , Limit of Detection , MCF-7 Cells
4.
RNA ; 25(11): 1416-1420, 2019 11.
Article in English | MEDLINE | ID: mdl-31511321

ABSTRACT

We read with great interest the Divergent Views article by Connell and colleagues disputing our recent publication describing a role for two microRNAs in the iron-mediated regulation of transferrin receptor 1 (TfR1) mRNA stability. Our publication sought to shed light on a long-standing question in the field of cellular iron metabolism, and we welcome commentary and critique. However, there are several critical issues contained in the article by Connell and colleagues that require further consideration. We appreciate the opportunity to reply here.


Subject(s)
Iron , MicroRNAs , RNA Stability , RNA, Messenger , Receptors, Transferrin
5.
Cell Chem Biol ; 26(1): 85-97.e4, 2019 01 17.
Article in English | MEDLINE | ID: mdl-30449675

ABSTRACT

Cisplatin is classically known to exhibit anticancer activity through DNA damage in the nucleus. Here we found a mechanism by which cisplatin affects iron metabolism, leading to toxicity and cell death. Cisplatin causes intracellular iron deficiency through direct inhibition of the master regulator of iron metabolism, iron regulatory protein 2 (IRP2) with marginal effects on IRP1. Cisplatin, but not carboplatin or transplatin, binds human IRP2 at Cys512 and Cys516 and impairs IRP2 binding to iron-responsive elements of ferritin and transferrin receptor-1 (TfR1) mRNAs. IRP2 inhibition by cisplatin caused ferritin upregulation and TfR1 downregulation leading to sustained intracellular iron deficiency. Cys512/516Ala mutant IRP2 made cells more resistant to cisplatin. Furthermore, combination of cisplatin and the iron chelator desferrioxamine enhanced cytotoxicity through augmented iron depletion in culture and xenograft mouse model. Collectively, cisplatin is an inhibitor of IRP2 that induces intracellular iron deficiency.


Subject(s)
Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Iron Regulatory Protein 2/antagonists & inhibitors , Iron/metabolism , Adolescent , Adult , Aged , Animals , Antineoplastic Agents/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Cisplatin/metabolism , Drug Screening Assays, Antitumor , Female , Fetus , Humans , Iron Deficiencies , Iron Regulatory Protein 2/genetics , Iron Regulatory Protein 2/metabolism , Male , Mice , Middle Aged , Molecular Structure , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology
6.
RNA ; 24(4): 468-479, 2018 04.
Article in English | MEDLINE | ID: mdl-29295890

ABSTRACT

Intracellular iron is tightly regulated by coordinated expression of iron transport and storage genes, such as transferrin receptor-1 (TfR1) and ferritin. They are primarily regulated by iron through iron-induced dissociation of iron-regulatory proteins (IRPs) from iron-responsive elements (IREs) in the 3'-UTR (untranslated region) of TfR1 or 5'-UTR of ferritin mRNA, resulting in destabilization of TfR1 mRNA and release of ferritin translation block. Thus high iron decreases iron transport via TfR1 mRNA degradation and increases iron storage via ferritin translational up-regulation. However, the molecular mechanism of TfR1 mRNA destabilization in response to iron remains elusive. Here, we demonstrate that miR-7-5p and miR-141-3p target 3'-TfR1 IREs and down-regulate TfR1 mRNA and protein expression. Conversely, miR-7-5p and miR-141-3p antagomiRs partially but significantly blocked iron- or IRP knockdown-induced down-regulation of TfR1 mRNA, suggesting the interplay between these microRNAs and IRPs along with involvement of another uncharacterized mechanism in TfR1 mRNA degradation. Luciferase reporter assays using 3'-UTR TfR1 IRE mutants suggested that the IREs C and E are targets of miR-7-5p and miR-141-3p, respectively. Furthermore, miR-7 expression was inversely correlated with TfR1 mRNA in human pancreatic adenocarcinoma patient samples. These results suggest a role of microRNAs in the TfR1 regulation in the IRP-IRE system.


Subject(s)
Antigens, CD/genetics , Iron-Regulatory Proteins/genetics , MicroRNAs/genetics , RNA, Messenger/biosynthesis , Receptors, Transferrin/genetics , 3T3 Cells , Animals , Antigens, CD/biosynthesis , Cell Proliferation/genetics , Ferritins/genetics , Humans , Iron/metabolism , Mice , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Receptors, Transferrin/biosynthesis
7.
Cell Signal ; 33: 69-78, 2017 05.
Article in English | MEDLINE | ID: mdl-28229933

ABSTRACT

Lamins are important constituents of the nuclear inner membrane and provide a platform for transcription factors and chromatin. Progerin, a C-terminal truncated lamin A mutant, causes premature aging termed Hutchinson-Gilford Progeria Syndrome (HGPS). Oxidative stress appears to be involved in the pathogenesis of HGPS, although the mechanistic role of progerin remains elusive. Here we examined whether nuclear lamins are important for a cellular antioxidant mechanism, and whether progerin compromises it. We investigated the activation of nuclear factor-E2-related factor 2 (Nrf2) which regulates various antioxidant genes including heme oxygenase-1 (HMOX1), following exposure to sodium arsenite or cadmium chloride in lamin knockdown human cell lines and primary HGPS human fibroblasts. Knocking down lamin A/C, or B, or all nuclear lamins simultaneously in three human cell lines (HaCaT, SW480, and K562) did not impair arsenite- or cadmium-induced activation of Nrf2. Progerin-expressing human primary HGPS fibroblasts showed lower basal levels of HMOX1 and NQO1 expression; however, in response to arsenic stress both normal and HGPS primary fibroblasts showed Nrf2 nuclear accumulation along with upregulation and phosphorylation of p62/SQSTM1 at Ser351, downregulation of Keap1, and comparable expression of an array of downstream Nrf2-regulated antioxidant genes. We also observed new forms of cleaved lamin A, B1 and B2 induced by cadmium stress although their roles in the Nrf2 antioxidant system need further investigation. These results suggest that the nuclear lamins and progerin have marginal roles in the activation of the antioxidant Nrf2 response to arsenic and cadmium.


Subject(s)
Antioxidants/metabolism , Arsenic/toxicity , Cadmium/toxicity , Cell Nucleus/metabolism , Lamin Type A/metabolism , Lamins/metabolism , NF-E2-Related Factor 2/metabolism , Cell Line, Tumor , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression Profiling , Gene Knockdown Techniques , Heme Oxygenase-1/metabolism , Humans , Kelch-Like ECH-Associated Protein 1/metabolism , Oxidative Stress/drug effects , Oxidative Stress/genetics , Phosphorylation/drug effects , Progeria/metabolism , Progeria/pathology , Sequestosome-1 Protein/metabolism
8.
J Mol Biol ; 429(1): 64-78, 2017 01 06.
Article in English | MEDLINE | ID: mdl-27884605

ABSTRACT

Cyclic AMP-response element-binding protein (CREB) plays key transcriptional roles in cell metabolism, proliferation, and survival. Ser133 phosphorylation by protein kinase A (PKA) is a well-characterized CREB activation mechanism. Homeodomain-interacting protein kinase (HIPK) 2, a nuclear serine/threonine kinase, activates CREB through Ser271 phosphorylation; however, the regulatory mechanism remains uncharacterized. Transfection of CREB in HEK293 cells together with the kinase demonstrated that HIPK2 phosphorylated CREB at Ser271 but not Ser133; likewise, PKA phosphorylated CREB at Ser133 but not Ser271, suggesting two distinct CREB regulatory mechanisms by HIPK2 and PKA. In vitro kinase assay revealed that HIPK2, and HIPK1 and HIPK3, directly phosphorylated CREB. Cells exposed to 10µM sodium arsenite increased the stability of HIPK1 and HIPK2 proteins, leading to CREB activation via Ser271 phosphorylation. Phospho-Ser271 CREB showed facilitated interaction with the TFIID subunit coactivator TAF4 assessed by immunoprecipitation. Furthermore, a focused gene array between cells transfected with CREB alone and CREB plus HIPK2 over empty vector-transfected control displayed 14- and 32-fold upregulation of cyclin A1, respectively, while no upregulation was displayed by HIPK2 alone. These results suggest that the HIPK2-phospho-Ser271 CREB axis is a new arsenic-responsive CREB activation mechanism in parallel with the PKA-phospho-Ser133 CREB axis.


Subject(s)
Arsenic/metabolism , Carrier Proteins/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/metabolism , Transcriptional Activation , Cell Line , Gene Expression Profiling , Humans , Phosphorylation , Serine/metabolism
9.
Trends Mol Med ; 22(12): 1077-1090, 2016 12.
Article in English | MEDLINE | ID: mdl-27825668

ABSTRACT

Iron is an essential nutrient for life. During infection, a fierce battle of iron acquisition occurs between the host and bacterial pathogens. Bacteria acquire iron by secreting siderophores, small ferric iron-binding molecules. In response, host immune cells secrete lipocalin 2 (also known as siderocalin), a siderophore-binding protein, to prevent bacterial reuptake of iron-loaded siderophores. To counter this threat, some bacteria can produce lipocalin 2-resistant siderophores. This review discusses the recently described molecular mechanisms of siderophore iron trafficking between host and bacteria, highlighting the therapeutic potential of exploiting pathogen siderophore machinery for the treatment of antibiotic-resistant bacterial infections. Because the latter reflect a persistent problem in hospital settings, siderophore-targeting or siderophore-based compounds represent a promising avenue to combat such infections.


Subject(s)
Bacteria/metabolism , Bacterial Infections/metabolism , Host-Pathogen Interactions , Iron/metabolism , Lipocalin-2/metabolism , Siderophores/metabolism , Animals , Anti-Bacterial Agents/pharmacology , Bacteria/drug effects , Bacteria/immunology , Bacterial Infections/drug therapy , Bacterial Infections/immunology , Drug Discovery , Host-Pathogen Interactions/drug effects , Humans , Iron/immunology , Lipocalin-2/immunology , Siderophores/immunology
10.
Antioxid Redox Signal ; 25(17): 953-964, 2016 12 10.
Article in English | MEDLINE | ID: mdl-27245349

ABSTRACT

AIMS: Nuclear factor erythroid 2 (NF-E2)-related factor 2 (Nrf2) is the master transcriptional regulator of antioxidant gene expression. On increased oxidative stress, an adaptor for Nrf2 degradation, Kelch-like ECH-associated protein 1 (Keap1), is directly modulated by oxidants in the cytoplasm, which results in stabilization and activation of Nrf2. Nrf2 is also constitutively active, to some extent, in the absence of exogenous oxidative stress. We have previously demonstrated that intestinal epithelium-specific TGF-ß-activated kinase 1 (TAK1) deletion downregulates the level of Nrf2 protein, resulting in an increase of reactive oxygen species (ROS) in a mouse model. We aim at determining the mechanism by which TAK1 modulates the level of Nrf2. RESULTS: We found that TAK1 upregulated serine 351 phosphorylation of an autophagic adaptor protein, p62/Sequestosome-1 (SQSTM1), which facilitates interaction between p62/SQSTM1 and Keap1 and subsequent Keap1 degradation. This, ultimately, causes increased Nrf2. Tak1 deficiency reduced the phosphorylation of p62/SQSTM1, resulting in decreased steady-state levels of Nrf2 along with increased Keap1. We also found that this regulation is independent of the canonical redox-mediated Nrf2 activation mechanism. In Tak1-deficient intestinal epithelium, a synthetic phenolic electrophile, butylated hydroxyanisole still effectively upregulated Nrf2 and reduced ROS. INNOVATION: Our results identify for the first time that TAK1 is a modulator of p62/SQSTM1-dependent Keap1 degradation and maintains the steady state-level of Nrf2. CONCLUSION: TAK1 regulates Nrf2 through modulation of Keap-p62/SQSTM1 interaction. This regulation is important for homeostatic antioxidant protection in the intestinal epithelium. Antioxid. Redox Signal. 25, 953-964.


Subject(s)
Antioxidants/metabolism , MAP Kinase Kinase Kinases/metabolism , NF-E2-Related Factor 2/metabolism , Sequestosome-1 Protein/metabolism , Animals , Cell Line , Gene Expression Regulation , Humans , Intestinal Mucosa/metabolism , Kelch-Like ECH-Associated Protein 1/metabolism , MAP Kinase Kinase Kinases/genetics , Mice , Mice, Knockout , Models, Biological , NF-E2-Related Factor 2/genetics , Oxidative Stress , Protein Binding , Proteolysis , Reactive Oxygen Species/metabolism
11.
Trends Biochem Sci ; 41(3): 274-286, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26725301

ABSTRACT

Iron is necessary for life, but can also cause cell death. Accordingly, cells evolved a robust, tightly regulated suite of genes for maintaining iron homeostasis. Previous mechanistic studies on iron homeostasis have granted insight into the role of iron in human health and disease. We highlight new regulators of iron metabolism, including iron-trafficking proteins [solute carrier family 39, SLC39, also known as ZRT/IRT-like protein, ZIP; and poly-(rC)-binding protein, PCBP] and a cargo receptor (NCOA4) that is crucial for release of ferritin-bound iron. We also discuss emerging roles of iron in apoptosis and a novel iron-dependent cell death pathway termed 'ferroptosis', the dysregulation of iron metabolism in human pathologies, and the use of iron chelators in cancer therapy.


Subject(s)
Cell Death , Homeostasis , Iron/metabolism , Humans
12.
Biochim Biophys Acta ; 1849(10): 1277-88, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26291278

ABSTRACT

Expression of the antioxidant gene heme oxygenase-1 (HO-1) is primarily induced through NF-E2-related factor 2 (Nrf2)-mediated activation of the antioxidant response element (ARE). Gene transcription is coordinately regulated by transcription factor activity at enhancer elements and epigenetic alterations such as the posttranslational modification of histone proteins. However, the role of histone modifications in the Nrf2-ARE axis remains largely uncharacterized. The environmental contaminant arsenite is a potent inducer of both HO-1 expression and phosphorylation of histone H3 serine 10 (H3S10); therefore, we investigated the relationships between Nrf2 and H3S10 phosphorylation in arsenite-induced, ARE-dependent, transcriptional activation of the human HO-1 gene. Arsenite increased phosphorylation of H3S10 both globally and at the HO-1 promoter concomitantly with HO-1 transcription in human HaCaT keratinocytes. Conversely, arsenite-induced H3S10 phosphorylation and HO-1 expression were blocked by N-acetylcysteine (NAC), the c-Jun N-terminal kinase (JNK) inhibitor SP600125, and JNK knockdown (siJNK). Interestingly, ablation of arsenite-induced H3S10 phosphorylation by SP600125 or siJNK did not inhibit Nrf2 nuclear accumulation nor ARE binding, despite inhibiting HO-1 expression. In response to arsenite, binding of Nrf2 to the HO-1 ARE preceded phosphorylation of H3S10 at the HO-1 ARE. Furthermore, arsenite-mediated occupancy of phosphorylated H3S10 at the HO-1 ARE was decreased in Nrf2-deficient mouse embryonic fibroblasts. These results suggest the involvement of H3S10 phosphorylation in the Nrf2-ARE axis by proposing that Nrf2 may influence H3S10 phosphorylation at the HO-1 ARE and additional promoter regions. Our data highlights the complex interplay between Nrf2 and H3S10 phosphorylation in arsenite-activated HO-1 transcription.


Subject(s)
Antioxidant Response Elements/genetics , Heme Oxygenase-1/biosynthesis , Histones/genetics , NF-E2-Related Factor 2/genetics , Transcriptional Activation/genetics , Animals , Arsenites/pharmacology , Gene Expression Regulation, Enzymologic , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Histones/metabolism , Humans , Keratinocytes/metabolism , Mice , NF-E2-Related Factor 2/metabolism , Oxidative Stress/genetics , Phosphorylation , Promoter Regions, Genetic , Protein Binding , Signal Transduction , Transcriptional Activation/drug effects
13.
Cell Signal ; 26(12): 2702-9, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25172425

ABSTRACT

Cobalt chloride has been used as a hypoxia mimetic because it stabilizes hypoxia inducible factor-1α (HIF1-α) and activates gene transcription through a hypoxia responsive element (HRE). However, differences between hypoxia and hypoxia mimetic cobalt chloride in gene regulation remain elusive. Expression of ferritin, the major iron storage protein, is regulated at the transcriptional and posttranscriptional levels through DNA and RNA regulatory elements. Here we demonstrate that hypoxia and cobalt chloride regulate ferritin heavy chain (ferritin H) expression by two distinct mechanisms. Both hypoxia and cobalt chloride increased HIF1-α but a putative HRE in the human ferritin H gene was not activated. Instead, cobalt chloride but not hypoxia activated ferritin H transcription through an antioxidant responsive element (ARE), to which Nrf2 was recruited. Intriguingly, cobalt chloride downregulated ferritin H protein expression while it upregulated other ARE-regulated antioxidant genes in K562 cells. Further characterization demonstrated that cobalt chloride increased interaction between iron regulatory proteins (IRP1 and IRP2) and iron responsive element (IRE) in the 5'UTR of ferritin H mRNA, resulting in translational block of the accumulated ferritin H mRNA. In contrast, hypoxia had marginal effect on ferritin H transcription but increased its translation through decreased IRP1-IRE interaction. These results suggest that hypoxia and hypoxia mimetic cobalt chloride employ distinct regulatory mechanisms through the interplay between DNA and mRNA elements at the transcriptional and post-transcriptional levels.


Subject(s)
Cobalt/pharmacology , Ferritins/genetics , Hypoxia/genetics , Protein Processing, Post-Translational/genetics , Transcription, Genetic/genetics , Apoferritins/genetics , Cell Line, Tumor , Gene Expression Regulation/genetics , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Iron-Regulatory Proteins/genetics , Protein Processing, Post-Translational/drug effects , RNA, Messenger/genetics , Transcription, Genetic/drug effects , Transcriptional Activation/drug effects , Transcriptional Activation/genetics
14.
Mol Biol Cell ; 25(13): 2116-27, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24807908

ABSTRACT

The mammalian Shc family, composed of p46, p52, and p66 isoforms, serves as an adaptor protein in cell growth and stress response. p66Shc was shown to be a negative lifespan regulator by acting as a prooxidant protein in mitochondria; however, the regulatory mechanisms of p66Shc expression and function are incompletely understood. This study provides evidence for new features of p66Shc serving as an antioxidant and critical protein in cell differentiation. Unique among the Shc family, transcription of p66Shc is activated through the antioxidant response element (ARE)-nuclear factor erythroid 2-related factor 2 (Nrf2) pathway in K562 human erythroleukemia and other cell types after treatment with hemin, an iron-containing porphyrin. Phosphorylated p66Shc at Ser-36, previously reported to be prone to mitochondrial localization, is increased by hemin treatment, but p66Shc remains exclusively in the cytoplasm. p66Shc knockdown inhibits hemin-induced erythroid differentiation, in which reactive oxygen species production and apoptosis are significantly enhanced in conjunction with suppression of other ARE-dependent antioxidant genes. Conversely, p66Shc overexpression is sufficient for inducing erythroid differentiation. Collectively these results demonstrate the isoform-specific regulation of the Shc gene by the Nrf2-ARE pathway and a new antioxidant role of p66Shc in the cytoplasm. Thus p66Shc is a bifunctional protein involved in cellular oxidative stress response and differentiation.


Subject(s)
Antioxidants/physiology , Shc Signaling Adaptor Proteins/physiology , Transcriptional Activation , Apoptosis , Cell Differentiation , Erythroid Cells/metabolism , Hemin/pharmacology , Humans , K562 Cells , Mitochondria/metabolism , NF-E2-Related Factor 2/metabolism , Oxidative Stress , Phosphorylation , Protein Isoforms/physiology , Protein Processing, Post-Translational , Protein Transport , Reactive Oxygen Species/metabolism , Response Elements , Src Homology 2 Domain-Containing, Transforming Protein 1 , Transcription, Genetic
15.
Biochemistry ; 52(30): 5075-83, 2013 Jul 30.
Article in English | MEDLINE | ID: mdl-23829535

ABSTRACT

Resveratrol, a natural polyphenol, increases cellular antioxidant capacity by inducing the expression of a battery of cytoprotective genes through an antioxidant responsive element (ARE). However, upstream signaling events initiated by resveratrol leading to the activation of an ARE enhancer, particularly in immune cells, have not been fully elucidated. In this study, ARE-dependent transcriptional activation of the ferritin heavy chain (ferritin H) gene by resveratrol was further investigated in Jurkat T cells and human peripheral blood mononuclear cells. We found that AMP-activated protein kinase (AMPK) plays a key role in the activation of nuclear factor E2-related factor (Nrf2) and subsequent ARE-dependent ferritin H gene transcription by resveratrol. A chromatin immunoprecipitation assay for Nrf2 after AMPKα knockdown with siRNA revealed that Nrf2 nuclear accumulation and subsequent binding to the ferritin H ARE induced by resveratrol were dependent on activation of AMPKα, but not PI3K/AKT. Furthermore, AMPKα knockdown blocked resveratrol-induced phosphorylation of glycogen synthase kinase 3ß (GSK3ß) at Ser9 as well as ARE-dependent transcriptional activation of the ferritin H and HO-1 genes, suggesting that AMPKα is an upstream kinase for GSK3ß phosphorylation and activation of the Nrf2-ARE pathway. Consistently, GSK3ß knockdown by siRNA enhanced resveratrol-mediated ferritin H mRNA induction, and the inhibition of AMPKα by compound C or siRNA weakened the protective effect of resveratrol against oxidative stress-induced cytotoxicity in CD3+ T cells. Collectively, these results suggest that AMPKα plays a significant role in ARE-dependent transcription of ferritin H genes by resveratrol and may influence the redox status in immune cells.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Antioxidants/pharmacology , Apoferritins/metabolism , Gene Expression Regulation/drug effects , Stilbenes/pharmacology , T-Lymphocytes/drug effects , AMP-Activated Protein Kinases/antagonists & inhibitors , AMP-Activated Protein Kinases/chemistry , AMP-Activated Protein Kinases/genetics , Antioxidants/chemistry , Apoferritins/genetics , Enzyme Activation/drug effects , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Jurkat Cells , K562 Cells , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , RNA Interference , Response Elements/drug effects , Resveratrol , Serine/metabolism , Stilbenes/antagonists & inhibitors , T-Lymphocytes/metabolism
16.
FASEB J ; 27(9): 3763-74, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23699174

ABSTRACT

Antioxidant genes such as ferritin are transcriptionally activated in oxidative stress via the antioxidant responsive element (ARE), to which nuclear factor-E2-related factor 2 (Nrf2) binds and activates transcription. Histone modification plays a cooperative and essential role in transcriptional regulation; however, its role in antioxidant gene transcription remains elusive. Arsenic exposure activated ferritin transcription via the ARE concomitant with increased methylation of histones H4Arg3 (H4R3) and H3Arg17 (H3R17). To test our hypothesis that histone H4R3 and H3R17 methylation regulates ferritin transcription, H4R3 and H3R17 protein arginine (R) methyltransferases 1 and 4 (PRMT1 and PRMT4) were investigated. Arsenic exposure of human HaCaT keratinocytes induced nuclear accumulation of PRMT1 and PRMT4, histone H4R3 and H3R17 methylation proximal to the ARE, but not to the non-ARE regions of ferritin genes. PRMT1 or PRMT4 knockdown did not block Nrf2 nuclear accumulation but inhibited Nrf2 binding to the AREs by ∼40% (P<0.05), thus diminishing ferritin transcription in HaCaT and human primary keratinocytes and fibroblasts, causing enhanced cellular susceptibility to arsenic toxicity as evidenced by 2-fold caspase 3 activation. Focused microarray further characterized several oxidative stress response genes are subject to PRMT1 or PRMT4 regulation. Collectively, PRMT1 and PRMT4 regulate the ARE and cellular antioxidant response to arsenic.


Subject(s)
Ferritins/genetics , NF-E2-Related Factor 2/metabolism , Protein-Arginine N-Methyltransferases/metabolism , Repressor Proteins/metabolism , Antioxidants/metabolism , Arsenic/toxicity , Cell Line , Cells, Cultured , Fibroblasts/cytology , Fibroblasts/metabolism , Histones/metabolism , Humans , Keratinocytes/cytology , Keratinocytes/metabolism , Methylation , NF-E2-Related Factor 2/genetics , Oxidative Stress/drug effects , Oxidative Stress/genetics , Protein-Arginine N-Methyltransferases/genetics , Repressor Proteins/genetics , Transcription, Genetic/drug effects , Transcription, Genetic/genetics
17.
Clin Med Insights Oncol ; 7: 21-30, 2013.
Article in English | MEDLINE | ID: mdl-23439317

ABSTRACT

INTRODUCTION: This multicenter phase II study determined the efficacy and safety of new daily oral S-1 and weekly irinotecan (CPT-11) combination schedule in patients with previously untreated advanced or recurrent colorectal cancer. PATIENTS AND METHODS: Patients received first-line chemotherapy comprising S-1 80 mg/m(2)/day given on days 3 to 7, 10 to 14, and 17 to 21 and 60 mg/m(2) CPT-11 administered intravenously on days 1, 8, and 15 of a 28-day cycle. RESULTS: A total of 45 eligible patients were enrolled in this study. The overall response rate was 48.9%. Median progression-free survival and median overall survival was 8.1 months and 20.9 months, respectively. The rates of grade 3 or 4 toxicity were as follows: neutropenia, 8.9%; anemia, 4.4%; anorexia, 6.7%; and diarrhea, 6.7%. CONCLUSIONS: This new S-1 and irinotecan combination schedule appeared to be an effective, well-tolerated, and convenient regimen in patients with advanced colorectal cancer as compared with conventional regimens such as FOLFIRI and IRIS.

18.
Cell Signal ; 24(5): 981-90, 2012 May.
Article in English | MEDLINE | ID: mdl-22286106

ABSTRACT

Reactive oxygen species (ROS) are generated during mitochondrial oxidative metabolism as well as in cellular response to xenobiotics, cytokines, and bacterial invasion. Oxidative stress refers to the imbalance due to excess ROS or oxidants over the capability of the cell to mount an effective antioxidant response. Oxidative stress results in macromolecular damage and is implicated in various disease states such as atherosclerosis, diabetes, cancer, neurodegeneration, and aging. Paradoxically, accumulating evidence indicates that ROS also serve as critical signaling molecules in cell proliferation and survival. While there is a large body of research demonstrating the general effect of oxidative stress on signaling pathways, less is known about the initial and direct regulation of signaling molecules by ROS, or what we term the "oxidative interface." Cellular ROS sensing and metabolism are tightly regulated by a variety of proteins involved in the redox (reduction/oxidation) mechanism. This review focuses on the molecular mechanisms through which ROS directly interact with critical signaling molecules to initiate signaling in a broad variety of cellular processes, such as proliferation and survival (MAP kinases, PI3 kinase, PTEN, and protein tyrosine phosphatases), ROS homeostasis and antioxidant gene regulation (thioredoxin, peroxiredoxin, Ref-1, and Nrf-2), mitochondrial oxidative stress, apoptosis, and aging (p66Shc), iron homeostasis through iron-sulfur cluster proteins (IRE-IRP), and ATM-regulated DNA damage response.


Subject(s)
Homeostasis , Reactive Oxygen Species/metabolism , Signal Transduction , Amino Acid Sequence , Animals , Gene Expression Regulation , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/physiology , Molecular Sequence Data , Oxidation-Reduction , Oxidative Stress
19.
J Cell Sci ; 123(Pt 22): 3863-71, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20980392

ABSTRACT

ATF1 (activating transcription factor 1), a stimulus-induced CREB family transcription factor, plays important roles in cell survival and proliferation. Phosphorylation of ATF1 at Ser63 by PKA (cAMP-dependent protein kinase) and related kinases was the only known post-translational regulatory mechanism of ATF1. Here, we found that HIPK2 (homeodomain-interacting protein kinase 2), a DNA-damage-responsive nuclear kinase, is a new ATF1 kinase that phosphorylates Ser198 but not Ser63. ATF1 phosphorylation by HIPK2 activated ATF1 transcription function in the GAL4-reporter system. ATF1 is a transcriptional repressor of ferritin H, the major intracellular iron storage gene, through an ARE (antioxidant-responsive element). HIPK2 overrode the ATF1-mediated ARE repression in a kinase-activity-dependent manner in HepG2 cells. Furthermore, DNA-damage-inducing agents doxorubicin, etoposide and sodium arsenite induced ferritin H mRNA expression in HIPK2(+/+) MEF cells, whereas it was significantly impaired in HIPK2(-/-) MEF cells. Induction of other ARE-regulated detoxification genes such as NQO1 (NADPH quinone oxidoreductase 1), GST (glutathione S-transferase) and HO1 (heme oxygenase 1) by genotoxic stress was also decreased in HIPK2-deficient cells. Taken together, these results suggest that HIPK2 is a new ATF1 kinase involved in the regulation of ferritin H and other antioxidant detoxification genes in genotoxic stress conditions.


Subject(s)
Activating Transcription Factor 1/genetics , Carrier Proteins/genetics , DNA Damage , Ferritins/genetics , Protein Serine-Threonine Kinases/genetics , Transcription Factors/genetics , Activating Transcription Factor 1/metabolism , Carrier Proteins/metabolism , Cell Growth Processes/genetics , Cell Survival/genetics , Ferritins/metabolism , Gene Expression Regulation , Humans , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Transcription Factors/metabolism
20.
Mol Biol Cell ; 21(16): 2966-74, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20573984

ABSTRACT

CREB (cyclic AMP response element-binding protein) is a stimulus-induced transcription factor that plays pivotal roles in cell survival and proliferation. The transactivation function of CREB is primarily regulated through Ser-133 phosphorylation by cAMP-dependent protein kinase A (PKA) and related kinases. Here we found that homeodomain-interacting protein kinase 2 (HIPK2), a DNA-damage responsive nuclear kinase, is a new CREB kinase for phosphorylation at Ser-271 but not Ser-133, and activates CREB transactivation function including brain-derived neurotrophic factor (BDNF) mRNA expression. Ser-271 to Glu-271 substitution potentiated the CREB transactivation function. ChIP assays in SH-SY5Y neuroblastoma cells demonstrated that CREB Ser-271 phosphorylation by HIPK2 increased recruitment of a transcriptional coactivator CBP (CREB binding protein) without modulation of CREB binding to the BDNF CRE sequence. HIPK2-/- MEF cells were more susceptible to apoptosis induced by etoposide, a DNA-damaging agent, than HIPK2+/+ cells. Etoposide activated CRE-dependent transcription in HIPK2+/+ MEF cells but not in HIPK2-/- cells. HIPK2 knockdown in SH-SY5Y cells decreased etoposide-induced BDNF mRNA expression. These results demonstrate that HIPK2 is a new CREB kinase that regulates CREB-dependent transcription in genotoxic stress.


Subject(s)
Carrier Proteins/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , DNA Damage , Protein Serine-Threonine Kinases/metabolism , Serine/metabolism , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Blotting, Western , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , CREB-Binding Protein/genetics , CREB-Binding Protein/metabolism , Carrier Proteins/genetics , Cell Line, Tumor , Cells, Cultured , Cyclic AMP Response Element-Binding Protein/genetics , Embryo, Mammalian/cytology , Etoposide/pharmacology , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , HEK293 Cells , Humans , K562 Cells , Mice , Mice, Knockout , Phosphorylation , Protein Binding , Protein Serine-Threonine Kinases/genetics , RNA Interference , Serine/genetics , Transcription, Genetic/drug effects , Two-Hybrid System Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...