1
|
Oliveri LM, Buzaleh AM, Gerez EN. Regulation of the expression of ferrochelatase in a murine model of diabetes mellitus type I. Biochem Biophys Rep 2025; 42:101989. [PMID: 40230493 PMCID: PMC11994340 DOI: 10.1016/j.bbrep.2025.101989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 03/17/2025] [Accepted: 03/25/2025] [Indexed: 04/16/2025] Open
Abstract
Background Diabetes produces changes on cellular hemeprotein metabolism. The last enzyme of heme biosynthetic pathway is ferrochelatase (FECH), an enzyme that catalyzes the insertion of ferrous ion into protoporphyrin IX to produce heme. The aim of this work was to investigate whether FECH expression can be other key point in the regulation of heme biosynthesis in diabetic animals. Methods Mice were rendered diabetic with streptozotocin (STZ, 170 mg/kg body weight i.p. for 15 days). Liver FECH protein and mRNA levels were evaluated by Western blot and Northern blot respectively. Vanadate was used as a hypoglycemic agent. The levels of the transcription factor Sp1 bound to the FECH promoter were assessed by chromatin immunoprecipitation (ChIP). Results Hyperglycemia caused an increase in FECH mRNA levels but no changes in FECH protein expression. ChIP analysis revealed that the increase in FECH mRNA levels was due to enhanced Sp1 binding to the FECH promoter in diabetic animals, which was reduced by vanadate administration. Conclusions In diabetic animals, enhanced binding of Sp1 to the FECH promoter may be responsible for the increase in FECH mRNA levels. However, this increase was not reflected in the amount of FECH protein, which would confirm that FECH could be another control point in heme synthesis.
Collapse
Affiliation(s)
- Leda María Oliveri
- Centro de Investigaciones sobre Porfirinas y Porfirias (CIPYP), UBA-CONICET, Hospital de Clínicas José de San Martín, Argentina
| | - Ana Maria Buzaleh
- Centro de Investigaciones sobre Porfirinas y Porfirias (CIPYP), UBA-CONICET, Hospital de Clínicas José de San Martín, Argentina
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Argentina
| | - Esther Noemí Gerez
- Centro de Investigaciones sobre Porfirinas y Porfirias (CIPYP), UBA-CONICET, Hospital de Clínicas José de San Martín, Argentina
- Cátedra Bioquímica General Celular y Molecular, Facultad de Ciencias Médicas. Universidad Católica Argentina (UCA), Buenos Aires, Argentina
| |
Collapse
|
2
|
Qiu L, Frazer DM, Hu M, Song R, Liu X, Qin X, Ma J, Zhou J, Tan Z, Ren F, Collins JF, Wang X. Mechanism and regulation of iron absorption throughout the life cycle. J Adv Res 2025:S2090-1232(25)00002-5. [PMID: 39814221 DOI: 10.1016/j.jare.2025.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 12/24/2024] [Accepted: 01/02/2025] [Indexed: 01/18/2025] Open
Abstract
BACKGROUND Iron plays a crucial role through various life stages of human. Iron homeostasis is primarily regulated by iron absorption which is mediated via divalent metal-ion transporter 1 (DMT1), and iron export protein ferroportin (FPN), as there is no active pathway for iron excretion from the body. Recent studies have shown that the magnitude of iron absorption changes through various life stages to meet changing iron requirements. AIM OF REVIEW This review aims to provide an overview of recent researches on the regulation of iron absorption throughout mammalian life cycle, with the potential to reveal novel molecules and pathways at special stage of life. Such insights may pave the way for new treatments for disorders associated with aberrant iron homeostasis in the future. KEY SCIENTIFIC CONCEPTS OF REVIEW This review first summarize the mechanism and regulation of iron absorption throughout various life stages, highlighting that regulatory mechanisms have developed to precisely align iron absorption to iron requirements. In adults, iron absorption is enhanced when body is deficient of iron, conversely, iron absorption is reduced when iron demand decreases via systemic regulator Hepcidin and cellular regulation. In the elderly, age-related inflammation, hormonal changes, and chronic diseases may affect the production of Hepcidin, affecting iron absorption. In infants, intestinal iron absorption and its regulatory mechanism are different from that in adults and there might be an alternative pathway independent of DMT1 and FPN due to high iron absorption. Unique to the fetus, iron is absorbed from maternal stores for its own use through the placenta and is regulated by maternal iron status. This review also proposes directions for further studies, offering promising avenues for developing new treatments for disorders associated with aberrant iron homeostasis.
Collapse
Affiliation(s)
- Lili Qiu
- College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083 China
| | - David M Frazer
- Molecular Nutrition Laboratory, QIMR Berghofer Medical Research Institute, Herston 4029 Australia
| | - Mengxiao Hu
- College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083 China
| | - Rui Song
- College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083 China
| | - Xiaoxue Liu
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083 China
| | - Xiyu Qin
- College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083 China
| | - Jie Ma
- College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083 China
| | - Jun Zhou
- College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083 China
| | - Zidi Tan
- College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083 China
| | - Fazheng Ren
- College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083 China; Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083 China
| | - James F Collins
- Food Science & Human Nutrition Department, University of Florida, Gainesville, FL 32611, USA
| | - Xiaoyu Wang
- College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083 China; Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083 China.
| |
Collapse
|
3
|
Talkhoncheh MS, Baudet A, Ek F, Subramaniam A, Kao YR, Miharada N, Karlsson C, Oburoglu L, Rydström A, Zemaitis K, Alattar AG, Rak J, Pietras K, Olsson R, Will B, Larsson J. Ciclopirox ethanolamine preserves the immature state of human HSCs by mediating intracellular iron content. Blood Adv 2023; 7:7407-7417. [PMID: 37487020 PMCID: PMC10758717 DOI: 10.1182/bloodadvances.2023009844] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 06/14/2023] [Accepted: 07/05/2023] [Indexed: 07/26/2023] Open
Abstract
Culture conditions in which hematopoietic stem cells (HSCs) can be expanded for clinical benefit are highly sought after. To elucidate regulatory mechanisms governing the maintenance and propagation of human HSCs ex vivo, we screened libraries of annotated small molecules in human cord blood cells using an optimized assay for detection of functional HSCs during culture. We found that the antifungal agent ciclopirox ethanolamine (CPX) selectively supported immature CD34+CD90+ cells during culture and enhanced their long-term in vivo repopulation capacity. Purified HSCs treated with CPX showed a reduced cell division rate and an enrichment of HSC-specific gene expression patterns. Mechanistically, we found that the HSC stimulating effect of CPX was directly mediated by chelation of the intracellular iron pool, which in turn affected iron-dependent proteins and enzymes mediating cellular metabolism and respiration. Our findings unveil a significant impact of iron homeostasis in regulation of human HSCs, with important implications for both basic HSC biology and clinical hematology.
Collapse
Affiliation(s)
| | - Aurélie Baudet
- Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Fredrik Ek
- Chemical Biology and Therapeutics, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | | | - Yun-Ruei Kao
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY
| | - Natsumi Miharada
- Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Christine Karlsson
- Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Leal Oburoglu
- Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Anna Rydström
- Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Kristijonas Zemaitis
- Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Abdul Ghani Alattar
- Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Justyna Rak
- Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Kristian Pietras
- Division of Translational Cancer Research, Medicon Village, Lund University, Lund, Sweden
| | - Roger Olsson
- Chemical Biology and Therapeutics, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Britta Will
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY
- Department of Medicine (Oncology), Albert Einstein College of Medicine, Bronx, NY
| | - Jonas Larsson
- Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden
| |
Collapse
|
4
|
Goralska M, Fleisher LN, McGahan MC. Vitreous Humor Changes Expression of Iron-Handling Proteins in Lens Epithelial Cells. Invest Ophthalmol Vis Sci 2017; 58:1187-1195. [PMID: 28245299 PMCID: PMC5338632 DOI: 10.1167/iovs.16-20610] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose In humans, vitrectomy is associated with development of nuclear cataracts. Iron catalyzes free radical formation causing oxidative damage, which is implicated in cataract formation. This study was designed to determine if vitreous humor, which can initiate differentiation of lens epithelial cells, would have an effect on iron-handling proteins. Methods Cultured canine lens epithelial cells were treated with collected canine vitreous humor. Lysates of treated and control cells were separated by SDS-PAGE. Ferritin H- and L-chains, transferrin receptor 1, and aquaporin 0 were immunodetected and quantitated with specific antibodies. Morphologic changes in treated cells were assessed. Results Treatment of lens epithelial cells with a 33% (vol/vol) solution of vitreous humor changed the morphology of lens cells and induced expression of aquaporin 0, a marker of fiber cell differentiation that was undetectable in control cells. Treatment did not modify the size of iron-handling proteins but significantly increased content of ferritin from 2.9- to 8.8-fold over control and decreased levels of transferrin receptor by 37% to 59%. Conclusions Vitreous humor may significantly limit iron uptake by transferrin/transferrin receptor pathway, and by increasing ferritin levels could profoundly increase the iron-storage capacity of ferritin in lens cells. Vitreous humor may play a significant protective role against iron-catalyzed oxidative damage of lens epithelial cells and therefore in the formation of cataracts.
Collapse
Affiliation(s)
- Malgorzata Goralska
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, North Carolina, United States
| | - Lloyd N Fleisher
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, North Carolina, United States
| | - M Christine McGahan
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, North Carolina, United States
| |
Collapse
|
5
|
Russo J, Lee JE, López CM, Anderson J, Nguyen TMP, Heck AM, Wilusz J, Wilusz CJ. The CELF1 RNA-Binding Protein Regulates Decay of Signal Recognition Particle mRNAs and Limits Secretion in Mouse Myoblasts. PLoS One 2017; 12:e0170680. [PMID: 28129347 PMCID: PMC5271678 DOI: 10.1371/journal.pone.0170680] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 01/09/2017] [Indexed: 02/06/2023] Open
Abstract
We previously identified several mRNAs encoding components of the secretory pathway, including signal recognition particle (SRP) subunit mRNAs, among transcripts associated with the RNA-binding protein CELF1. Through immunoprecipitation of RNAs crosslinked to CELF1 in myoblasts and in vitro binding assays using recombinant CELF1, we now provide evidence that CELF1 directly binds the mRNAs encoding each of the subunits of the SRP. Furthermore, we determined the half-lives of the Srp transcripts in control and CELF1 knockdown myoblasts. Our results indicate CELF1 is a destabilizer of at least five of the six Srp transcripts and that the relative abundance of the SRP proteins is out of balance when CELF1 is depleted. CELF1 knockdown myoblasts exhibit altered secretion of a luciferase reporter protein and are impaired in their ability to migrate and close a wound, consistent with a defect in the secreted extracellular matrix. Importantly, similar defects in wound healing are observed when SRP subunit imbalance is induced by over-expression of SRP68. Our studies support the existence of an RNA regulon containing Srp mRNAs that is controlled by CELF1. One implication is that altered function of CELF1 in myotonic dystrophy may contribute to changes in the extracellular matrix of affected muscle through defects in secretion.
Collapse
Affiliation(s)
- Joseph Russo
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Jerome E. Lee
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
- Program in Cell and Molecular Biology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Carolina M. López
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
| | - John Anderson
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Thuy-mi P. Nguyen
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Adam M. Heck
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
- Program in Cell and Molecular Biology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Jeffrey Wilusz
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
- Program in Cell and Molecular Biology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Carol J. Wilusz
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
- Program in Cell and Molecular Biology, Colorado State University, Fort Collins, Colorado, United States of America
- * E-mail:
| |
Collapse
|
6
|
Kim M, Kim EH, Choi BR, Woo HJ. Differentially expressed genes in iron-induced prion protein conversion. Biochem Biophys Res Commun 2016; 480:734-740. [PMID: 27983987 DOI: 10.1016/j.bbrc.2016.10.117] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 10/27/2016] [Indexed: 01/26/2023]
Abstract
The conversion of the cellular prion protein (PrPC) to the protease-resistant isoform is the key event in chronic neurodegenerative diseases, including transmissible spongiform encephalopathies (TSEs). Increased iron in prion-related disease has been observed due to the prion protein-ferritin complex. Additionally, the accumulation and conversion of recombinant PrP (rPrP) is specifically derived from Fe(III) but not Fe(II). Fe(III)-mediated PK-resistant PrP (PrPres) conversion occurs within a complex cellular environment rather than via direct contact between rPrP and Fe(III). In this study, differentially expressed genes correlated with prion degeneration by Fe(III) were identified using Affymetrix microarrays. Following Fe(III) treatment, 97 genes were differentially expressed, including 85 upregulated genes and 12 downregulated genes (≥1.5-fold change in expression). However, Fe(II) treatment produced moderate alterations in gene expression without inducing dramatic alterations in gene expression profiles. Moreover, functional grouping of identified genes indicated that the differentially regulated genes were highly associated with cell growth, cell maintenance, and intra- and extracellular transport. These findings showed that Fe(III) may influence the expression of genes involved in PrP folding by redox mechanisms. The identification of genes with altered expression patterns in neural cells may provide insights into PrP conversion mechanisms during the development and progression of prion-related diseases.
Collapse
Affiliation(s)
- Minsun Kim
- Laboratory of Immunology and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| | - Eun-Hee Kim
- Laboratory of Immunology and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| | - Bo-Ran Choi
- Laboratory of Immunology and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea; Solomon Snyder Department of Neuroscience, School of Medicine, Johns Hopkins University, PCTB1004, 725 N Wolfe Street, Baltimore, MD, USA
| | - Hee-Jong Woo
- Laboratory of Immunology and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea.
| |
Collapse
|
7
|
Campanella A, Privitera D, Guaraldo M, Rovelli E, Barzaghi C, Garavaglia B, Santambrogio P, Cozzi A, Levi S. Skin fibroblasts from pantothenate kinase-associated neurodegeneration patients show altered cellular oxidative status and have defective iron-handling properties. Hum Mol Genet 2012; 21:4049-59. [PMID: 22692681 DOI: 10.1093/hmg/dds229] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Pantothenate kinase-associated neurodegeneration (PKAN) is a neurodegenerative disease belonging to the group of neurodegeneration with brain iron accumulation disorders. It is characterized by progressive impairments in movement, speech and cognition. The disease is inherited in a recessive manner due to mutations in the Pantothenate Kinase-2 (PANK2) gene that encodes a mitochondrial protein involved in Coenzyme A synthesis. To investigate the link between a PANK2 gene defect and iron accumulation, we analyzed primary skin fibroblasts from three PKAN patients and three unaffected subjects. The oxidative status of the cells and their ability to respond to iron were analyzed in both basal and iron supplementation conditions. In basal conditions, PKAN fibroblasts show an increase in carbonylated proteins and altered expression of antioxidant enzymes with respect to the controls. After iron supplementation, the PKAN fibroblasts had a defective response to the additional iron. Under these conditions, ferritins were up-regulated and Transferrin Receptor 1 (TfR1) was down-regulated to a minor extent in patients compared with the controls. Analysis of iron regulatory proteins (IRPs) reveals that, with respect to the controls, PKAN fibroblasts have a reduced amount of membrane-associated mRNA-bound IRP1, which responds imperfectly to iron. This accounts for the defective expression of ferritin and TfR1 in patients' cells. The inaccurate quantity of these proteins produced a higher bioactive labile iron pool and consequently increased iron-dependent reactive oxygen species formation. Our results suggest that Pank2 deficiency promotes an increased oxidative status that is further enhanced by the addition of iron, potentially causing damage in cells.
Collapse
|
8
|
Prasanthi JRP, Larson T, Schommer J, Ghribi O. Silencing GADD153/CHOP gene expression protects against Alzheimer's disease-like pathology induced by 27-hydroxycholesterol in rabbit hippocampus. PLoS One 2011; 6:e26420. [PMID: 22046282 PMCID: PMC3194795 DOI: 10.1371/journal.pone.0026420] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2011] [Accepted: 09/26/2011] [Indexed: 02/07/2023] Open
Abstract
Endoplasmic reticulum (ER) stress is suggested to play a key role in the pathogenesis of neurodegenerative diseases including Alzheimer's disease (AD). Sustained ER stress leads to activation of the growth arrest and leucine zipper transcription factor, DNA damage inducible gene 153 (gadd153; also called CHOP). Activated gadd153 can generate oxidative damage and reactive oxygen species (ROS), increase β-amyloid (Aβ) levels, disturb iron homeostasis and induce inflammation as well as cell death, which are all pathological hallmarks of AD. Epidemiological and laboratory studies suggest that cholesterol dyshomeostasis contributes to the pathogenesis of AD. We have previously shown that the cholesterol oxidized metabolite 27-hydroxycholesterol (27-OHC) triggers AD-like pathology in organotypic slices. However, the extent to which gadd153 mediates 27-OHC effects has not been determined. We silenced gadd153 gene with siRNA and determined the effects of 27-OHC on AD hallmarks in organotypic slices from adult rabbit hippocampus. siRNA to gadd153 reduced 27-OHC-induced Aβ production by mechanisms involving reduction in levels of β-amyloid precursor protein (APP) and β-secretase (BACE1), the enzyme that initiates cleavage of APP to yield Aβ peptides. Additionally, 27-OHC-induced tau phosphorylation, ROS generation, TNF-α activation, and iron and apoptosis-regulatory protein levels alteration were also markedly reduced by siRNA to gadd153. These data suggest that ER stress-mediated gadd153 activation plays a central role in the triggering of AD pathological hallmarks that result from incubation of hippocampal slices with 27-OHC. Our results add important insights into cellular mechanisms that underlie the potential contribution of cholesterol metabolism in AD pathology, and suggest that preventing gadd153 activation protects against AD related to cholesterol oxidized products.
Collapse
Affiliation(s)
- Jaya R. P. Prasanthi
- Department of Pharmacology, Physiology and Therapeutics, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota, United States of America
| | - Tyler Larson
- Department of Pharmacology, Physiology and Therapeutics, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota, United States of America
| | - Jared Schommer
- Department of Pharmacology, Physiology and Therapeutics, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota, United States of America
| | - Othman Ghribi
- Department of Pharmacology, Physiology and Therapeutics, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota, United States of America
| |
Collapse
|
9
|
Assays for monitoring viral manipulation of host ARE-mRNA turnover. Methods 2011; 55:172-81. [PMID: 21854851 DOI: 10.1016/j.ymeth.2011.08.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2011] [Revised: 08/04/2011] [Accepted: 08/05/2011] [Indexed: 12/22/2022] Open
Abstract
Early host responses to viral infection rapidly induce an antiviral gene expression program that limits viral replication and recruits sentinel cells of the innate immune system. These responses are mediated by cytokines. The mRNAs that encode cytokines typically harbor destabilizing adenine- and uridine-rich elements (AREs) that direct their constitutive degradation in the cytoplasm. In response to a variety of signals, including viral infection, small pools of cytoplasmic ARE-mRNAs are rapidly stabilized and translated. Thus, mRNA stability plays a key role in antiviral gene expression. Intriguingly, recent studies have identified viral proteins that specifically target ARE-mRNAs for stabilization, suggesting that certain proteins encoded by ARE-mRNAs may be advantageous for infection. Here, we discuss the development of a suite of sensitive and complementary assays to monitor ARE-mRNA turnover. These include luciferase- and destabilized-GFP-based assays that can be adapted for high-throughput screening applications.
Collapse
|
10
|
Corallini S, Taranta A, Bellomo F, Palma A, Pastore A, Emma F. Transcriptional and posttranscriptional regulation of the CTNS gene. Pediatr Res 2011; 70:130-5. [PMID: 21508882 DOI: 10.1203/pdr.0b013e3182200187] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Cell cysteine (Cys) levels and/or the [Cys/CySS] redox potential have been shown to regulate mRNA levels of the CTNS gene, which encodes for a lysosomal cystine (CySS) carrier that is defective in cystinosis. To investigate the mechanisms involved CTNS mRNA regulation, different portions of the CTNS promotor were cloned into a luciferase vector and transfected in HK2 cells. A 1.5-2.4-fold increase in luciferase activity was observed when cells were incubated in culture medium containing low CySS concentrations. Conversely, CTNS mRNA levels decreased by 47-56% in the presence of N-acetyl-L-cysteine (NAC). Chase experiments with actinomycin D (ActD) demonstrated a 3-fold stabilization of the CTNS mRNA when cells were cultured in low CySS medium for 48 h. Treatment of control cells with cyclohexamide (CHX) increased CTNS mRNA levels, suggesting that CHX blocked the synthesis of proteins involved in mRNA degradation or in repression of the CTNS gene. Finally, in vitro binding assays showed increased binding (30-110%) of the Sp-1 transcription factor to two regions of the CTNS promotor when cells were incubated in low CySS medium. These results indicate that the CTNS gene is actively regulated at the transcriptional and posttranscriptional levels and suggest that CTNS plays a pivotal role in regulating cell thiol concentrations.
Collapse
Affiliation(s)
- Serena Corallini
- Division of Nephrology and Dialysis, Bambino Gesù Children's Hospital and Research Institute, 00165 Rome, Italy
| | | | | | | | | | | |
Collapse
|
11
|
Ferrari-Toninelli G, Bonini SA, Uberti D, Napolitano F, Stante M, Santoro F, Minopoli G, Zambrano N, Russo T, Memo M. Notch activation induces neurite remodeling and functional modifications in SH-SY5Y neuronal cells. Dev Neurobiol 2009; 69:378-91. [PMID: 19263417 DOI: 10.1002/dneu.20710] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Notch proteins are definitely recognized as key regulators of the neuronal fate during embryo development, but their function in the adult brain is still largely unknown. We have previously demonstrated that Notch pathway stimulation increases microtubules stability followed by the remodeling of neuronal morphology with neurite varicosities loss, thicker neuritis, and enlarged growth cones. Here we show that the neurite remodeling is a dynamic event, dependent on transcription and translation, and with functional implications. Exposure of differentiated human SH-SY5Y neuroblastoma cells to the Notch ligand Jagged1 induces varicosities loss all along the neurites, accompanied by the redistribution of presynaptic vesicles and the decrease in neurotransmitters release. As evaluated by time lapse digital imaging, dynamic changes in neurite morphology were rapidly reversible and dependent on the activation of the Notch signaling pathway. In fact, it was prevented by the inhibition of the proteolytic gamma-secretase enzyme or the transcription machinery, and was mimicked by the transfection of the intracellular domain of Notch. One hour after treatment with Jagged1, several genes were downregulated. Many of these genes encode proteins that are known to be involved in protein synthesis. These data suggest that in adult neurons, Notch pathway activates a transcriptional program that regulates the equilibrium between varicosities formation and varicosities loss in the neuronal presynaptic compartment involving the expression and redistribution of both structural and functional proteins.
Collapse
Affiliation(s)
- Giulia Ferrari-Toninelli
- Department of Biomedical Sciences and Biotechnologies, and National Institute of Neuroscience - Italy, University of Brescia, Brescia, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Hämäläinen M, Korhonen R, Moilanen E. Calcineurin inhibitors down-regulate iNOS expression by destabilising mRNA. Int Immunopharmacol 2009; 9:159-67. [DOI: 10.1016/j.intimp.2008.07.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2007] [Revised: 07/03/2008] [Accepted: 07/16/2008] [Indexed: 01/16/2023]
|
13
|
dos Santos CO, Dore LC, Valentine E, Shelat SG, Hardison RC, Ghosh M, Wang W, Eisenstein RS, Costa FF, Weiss MJ. An iron responsive element-like stem-loop regulates alpha-hemoglobin-stabilizing protein mRNA. J Biol Chem 2008; 283:26956-64. [PMID: 18676996 DOI: 10.1074/jbc.m802421200] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Hemoglobin production during erythropoiesis is mechanistically coupled to the acquisition and metabolism of iron. We discovered that iron regulates the expression of alpha-hemoglobin-stabilizing protein (AHSP), a molecular chaperone that binds and stabilizes free alpha-globin during hemoglobin synthesis. In primates, the 3'-untranslated region (UTR) of AHSP mRNA contains a nucleotide sequence resembling iron responsive elements (IREs), stem-loop structures that regulate gene expression post-transcriptionally by binding iron regulatory proteins (IRPs). The AHSP IRE-like stem-loop deviates from classical consensus sequences and binds IRPs poorly in electrophoretic mobility shift assays. However, in cytoplasmic extracts, AHSP mRNA co-immunoprecipitates with IRPs in a fashion that is dependent on the stem-loop structure and inhibited by iron. Moreover, this interaction enhances AHSP mRNA stability in erythroid and heterologous cells. Our findings demonstrate that IRPs can regulate mRNA expression through non-canonical IREs and extend the repertoire of known iron-regulated genes. In addition, we illustrate a new mechanism through which hemoglobin may be modulated according to iron status.
Collapse
Affiliation(s)
- Camila O dos Santos
- Division of Hematology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Abstract
The recognition of the importance of mRNA turnover in regulating eukaryotic gene expression has mandated the development of reliable, rigorous, and "user-friendly" methods to accurately measure changes in mRNA stability in mammalian cells. Frequently, mRNA stability is studied indirectly by analyzing the steady-state level of mRNA in the cytoplasm; in this case, changes in mRNA abundance are assumed to reflect only mRNA degradation, an assumption that is not always correct. Although direct measurements of mRNA decay rate can be performed with kinetic labeling techniques and transcriptional inhibitors, these techniques often introduce significant changes in cell physiology. Furthermore, many critical mechanistic issues as to deadenylation kinetics, decay intermediates, and precursor-product relationships cannot be readily addressed by these methods. In light of these concerns, we have previously reported transcriptional pulsing methods based on the c-fos serum-inducible promoter and the tetracycline-regulated (Tet-off) promoter systems to better explain mechanisms of mRNA turnover in mammalian cells. In this chapter, we describe and discuss in detail different protocols that use these two transcriptional pulsing methods. The information described here also provides guidelines to help develop optimal protocols for studying mammalian mRNA turnover in different cell types under a wide range of physiologic conditions.
Collapse
Affiliation(s)
- Chyi-Ying A Chen
- Department of Biochemistry and Molecular Biology, The University of Texas Medical School at Houston, Houston, Texas, USA
| | | | | |
Collapse
|
15
|
Kato J, Kobune M, Ohkubo S, Fujikawa K, Tanaka M, Takimoto R, Takada K, Takahari D, Kawano Y, Kohgo Y, Niitsu Y. Iron/IRP-1-dependent regulation of mRNA expression for transferrin receptor, DMT1 and ferritin during human erythroid differentiation. Exp Hematol 2007; 35:879-87. [PMID: 17533042 DOI: 10.1016/j.exphem.2007.03.005] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2006] [Revised: 03/08/2007] [Accepted: 03/08/2007] [Indexed: 11/21/2022]
Abstract
OBJECTIVE We investigated iron regulatory protein (IRP)-dependent expression of transferrin receptor (TfR), divalent metal transporter-1 (DMT1) and ferritin during erythroid differentiation system using an in vitro three-phase liquid culture. METHOD Peripheral blood hematopoietic progenitor cells were cultured with interleukin-3 and stem cell factor (SCF) for 7 days (first phase), subsequently with SCF, erythropoietin (EPO) and insulin-like growth factor-I (IGF-I) for 5 days (second phase), and finally with EPO and IGF-I for 3 days (third phase). Cells were subjected to colony assay, flow-cytometric analysis, mRNA assessment, electrophoretic mobility shift assay (EMSA), immunoblotting, and immunoprecipitation. RESULTS In the second/third phases, erythroid cells serially differentiated. Expression of TfR and DMT1 mRNA, which have iron-responsive elements (IREs) at 3'-UTR, reached a maximum on second phase, and thereafter decreased, while expression of ferritin mRNA, which has an IRE at the 5'-UTR, decreased reciprocally on second phase. IRP in the cytosol after precipitation of polysome decreased on second phase, suggesting that IRP bound to IREs of these mRNAs in the polysome. When cells were incubated with (59)FeCl(3), (59)Fe-bound IRP-1 immunoprecipitated with anti-IRP-1 antibodies was detected on first phase and third phase, but was not detected on second phase. 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.
Collapse
Affiliation(s)
- Junji Kato
- Fourth Department of Internal Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Söderberg M, Raffalli-Mathieu F, Lang MA. Regulation of the murine inducible nitric oxide synthase gene by dexamethasone involves a heterogeneous nuclear ribonucleoprotein I (hnRNPI) dependent pathway. Mol Immunol 2007; 44:3204-10. [DOI: 10.1016/j.molimm.2007.01.029] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2006] [Accepted: 01/20/2007] [Indexed: 10/23/2022]
|
17
|
Transcriptional regulatory network refinement and quantification through kinetic modeling, gene expression microarray data and information theory. BMC Bioinformatics 2007; 8:20. [PMID: 17244365 PMCID: PMC1790715 DOI: 10.1186/1471-2105-8-20] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2006] [Accepted: 01/23/2007] [Indexed: 11/10/2022] Open
Abstract
Background Gene expression microarray and other multiplex data hold promise for addressing the challenges of cellular complexity, refined diagnoses and the discovery of well-targeted treatments. A new approach to the construction and quantification of transcriptional regulatory networks (TRNs) is presented that integrates gene expression microarray data and cell modeling through information theory. Given a partial TRN and time series data, a probability density is constructed that is a functional of the time course of transcription factor (TF) thermodynamic activities at the site of gene control, and is a function of mRNA degradation and transcription rate coefficients, and equilibrium constants for TF/gene binding. Results Our approach yields more physicochemical information that compliments the results of network structure delineation methods, and thereby can serve as an element of a comprehensive TRN discovery/quantification system. The most probable TF time courses and values of the aforementioned parameters are obtained by maximizing the probability obtained through entropy maximization. Observed time delays between mRNA expression and activity are accounted for implicitly since the time course of the activity of a TF is coupled by probability functional maximization, and is not assumed to be proportional to expression level of the mRNA type that translates into the TF. This allows one to investigate post-translational and TF activation mechanisms of gene regulation. Accuracy and robustness of the method are evaluated. A kinetic formulation is used to facilitate the analysis of phenomena with a strongly dynamical character while a physically-motivated regularization of the TF time course is found to overcome difficulties due to omnipresent noise and data sparsity that plague other methods of gene expression data analysis. An application to Escherichia coli is presented. Conclusion Multiplex time series data can be used for the construction of the network of cellular processes and the calibration of the associated physicochemical parameters. We have demonstrated these concepts in the context of gene regulation understood through the analysis of gene expression microarray time series data. Casting the approach in a probabilistic framework has allowed us to address the uncertainties in gene expression microarray data. Our approach was found to be robust to error in the gene expression microarray data and mistakes in a proposed TRN.
Collapse
|
18
|
Christova T, Templeton DM. Effect of hypoxia on the binding and subcellular distribution of iron regulatory proteins. Mol Cell Biochem 2007; 301:21-32. [PMID: 17200797 DOI: 10.1007/s11010-006-9393-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2006] [Accepted: 12/06/2006] [Indexed: 10/23/2022]
Abstract
Iron regulatory proteins 1 and 2 (IRP1, IRP2) are key determinants of uptake and storage of iron by the liver, and are responsive to oxidative stress and hypoxia potentially at the level of both protein concentration and mRNA-binding activity. We examined the effect of hypoxia (1% O(2)) on IRP1 and IRP2 levels (Western blots) and mRNA-binding activity (gel shift assays) in human hepatoma HepG2 cells, and compared them with HEK 293 cells, a renal cell line known to respond to hypoxia. Total IRP binding to an iron responsive element (IRE) mRNA probe was increased several fold by hypoxia in HEK 293 cells, maximally at 4-8 h. An earlier and more modest increase (1.5- to 2-fold, peaking at 2 h and then declining) was seen in HepG2 cells. In both cell lines, IRP1 made a greater contribution to IRE-binding activity than IRP2. IRP1 protein levels were increased slightly by hypoxia in HEK 293 but not in HepG2 cells. IRP1 was distributed between cytosolic and membrane-bound fractions, and in both cells hypoxia increased both the amount and IRE-binding activity of the membrane-associated IRP1 fraction. Further density gradient fractionation of HepG2 membranes revealed that hypoxia caused an increase in total membrane IRP1, with a shift in the membrane-bound fraction from Golgi to an endoplasmic reticulum (ER)-enriched fraction. Translocation of IRP to the ER has previously been shown to stabilize transferrin receptor mRNA, thus increasing iron availability to the cell. Iron depletion with deferoxamine also caused an increase in ER-associated IRP1. Phorbol ester caused serine phosphorylation of IRP1 and increased its association with the ER. The calcium ionophore ionomycin likewise increased ER-associated IRP1, without affecting total IRE-binding activity. We conclude that IRP1 is translocated to the ER by multiple signals in HepG2 cells, including hypoxia, thereby facilitating its role in regulation of hepatic gene expression.
Collapse
Affiliation(s)
- Tania Christova
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | | |
Collapse
|
19
|
Abstract
The ability to regulate cellular gene expression is a key aspect of the lifecycles of a diverse array of viruses. In fact, viral infection often results in a global shutoff of host cellular gene expression; such inhibition serves not only to ensure maximal viral gene expression without competition from the host for essential machinery and substrates but also aids in evasion of immune responses detrimental to successful viral replication and dissemination. Within the herpesvirus family, host shutoff is a prominent feature of both the alpha- and gamma-herpesviruses. Intriguingly, while both classes of herpesviruses block cellular gene expression by inducing decay of messenger RNAs, the viral factors responsible for this phenotype as well as the mechanisms by which it is achieved are quite distinct. However, data suggest that the host shutoff functions of alpha- and gamma-herpesviruses are likely achieved both through the activity of virally encoded nucleases as well as via modulation of cellular RNA degradation pathways. This review highlights the processes governing normal cellular messenger RNA decay and then details the mechanisms by which herpesviruses promote accelerated RNA turnover. Parallels between the viral and cellular degradation systems as well as the known interactions between viral host shutoff factors and the cellular RNA turnover machinery are highlighted.
Collapse
Affiliation(s)
- Britt A Glaunsinger
- Howard Hughes Medical Institute, Department of Microbiology, University of California, San Francisco, 94143, USA
| | | |
Collapse
|
20
|
Nieminen R, Lahti A, Jalonen U, Kankaanranta H, Moilanen E. JNK inhibitor SP600125 reduces COX-2 expression by attenuating mRNA in activated murine J774 macrophages. Int Immunopharmacol 2006; 6:987-96. [PMID: 16644485 DOI: 10.1016/j.intimp.2006.01.009] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2005] [Revised: 10/17/2005] [Accepted: 01/13/2006] [Indexed: 01/15/2023]
Abstract
Inducible prostaglandin synthase (cyclooxygenase-2, COX-2) is highly expressed in inflammation. The signaling mechanisms involved in the up-regulation of COX-2 are not known in detail. In the present study we investigated the role of c-Jun NH2-terminal kinase (JNK), a member of the mitogen-activated protein kinase (MAPK) family in COX-2 expression and prostaglandin (PG) E2 production in murine J774 macrophages activated by bacterial lipopolysaccharide (LPS). LPS caused a transient activation of JNK which was followed by increased COX-2 expression. Anthra(1,9-cd)pyrazol-6(2H)-one (SP600125), an inhibitor of JNK, inhibited phosphorylation of c-Jun with an IC50 of 5-10 microM. At the same concentrations SP600125 suppressed also LPS-induced COX-2 protein levels and PGE2 production. SP600125 did not alter LPS-induced COX-2 mRNA levels when measured 3 h after addition of LPS, whereas mRNA levels were significantly reduced in SP600125-treated cells when measured 24 h after addition of LPS. LPS-induced COX-2 mRNA levels reduced faster in cells treated with SP600125 than in control cells. Cycloheximide (that is known to activate JNK) enhanced COX-2 expression and its effect was inhibited by SP600125. The present results suggest that JNK pathway is involved in the up-regulation of COX-2 expression possibly by a mechanism related to the stability of COX-2 mRNA.
Collapse
Affiliation(s)
- Riina Nieminen
- The Immunopharmacology Research Group, University of Tampere Medical School, and Tampere University Hospital, Research Unit, FIN-33014, Tampere, Finland
| | | | | | | | | |
Collapse
|
21
|
Lahti A, Sareila O, Kankaanranta H, Moilanen E. Inhibition of p38 mitogen-activated protein kinase enhances c-Jun N-terminal kinase activity: implication in inducible nitric oxide synthase expression. BMC Pharmacol 2006; 6:5. [PMID: 16504051 PMCID: PMC1402273 DOI: 10.1186/1471-2210-6-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2006] [Accepted: 02/21/2006] [Indexed: 11/10/2022] Open
Abstract
Background Nitric oxide (NO) is an inflammatory mediator, which acts as a cytotoxic agent and modulates immune responses and inflammation. p38 mitogen-activated protein kinase (MAPK) signal transduction pathway is activated by chemical and physical stress and regulates immune responses. Previous studies have shown that p38 MAPK pathway regulates NO production induced by inflammatory stimuli. The aim of the present study was to investigate the mechanisms involved in the regulation of inducible NO synthesis by p38 MAPK pathway. Results p38 MAPK inhibitors SB203580 and SB220025 stimulated lipopolysaccharide (LPS)-induced inducible nitric oxide synthase (iNOS) expression and NO production in J774.2 murine macrophages. Increased iNOS mRNA expression was associated with reduced degradation of iNOS mRNA. Treatment with SB220025 increased also LPS-induced c-Jun N-terminal kinase (JNK) activity. Interestingly, JNK inhibitor SP600125 reversed the effect of SB220025 on LPS-induced iNOS mRNA expression and NO production. Conclusion The results suggest that inhibition of p38 MAPK by SB220025 results in increased JNK activity, which leads to stabilisation of iNOS mRNA, to enhanced iNOS expression and to increased NO production.
Collapse
Affiliation(s)
- Aleksi Lahti
- The Immunopharmacology Research Group, Medical School, University of Tampere and Tampere University Hospital, Tampere, Finland
| | - Outi Sareila
- The Immunopharmacology Research Group, Medical School, University of Tampere and Tampere University Hospital, Tampere, Finland
| | - Hannu Kankaanranta
- The Immunopharmacology Research Group, Medical School, University of Tampere and Tampere University Hospital, Tampere, Finland
| | - Eeva Moilanen
- The Immunopharmacology Research Group, Medical School, University of Tampere and Tampere University Hospital, Tampere, Finland
| |
Collapse
|
22
|
Schranzhofer M, Schifrer M, Cabrera JA, Kopp S, Chiba P, Beug H, Müllner EW. Remodeling the regulation of iron metabolism during erythroid differentiation to ensure efficient heme biosynthesis. Blood 2006; 107:4159-67. [PMID: 16424395 DOI: 10.1182/blood-2005-05-1809] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Terminal erythropoiesis is accompanied by extreme demand for iron to ensure proper hemoglobinization. Thus, erythroblasts must modify the "standard" post-transcriptional feedback regulation, balancing expression of ferritin (Fer; iron storage) versus transferrin receptor (TfR1; iron uptake) via specific mRNA binding of iron regulatory proteins (IRPs). Although erythroid differentiation involves high levels of incoming iron, TfR1 mRNA stability must be sustained and Fer mRNA translation must not be activated because iron storage would counteract hemoglobinization. Furthermore, translation of the erythroid-specific form of aminolevulinic acid synthase (ALAS-E) mRNA, catalyzing the first step of heme biosynthesis and regulated similarly as Fer mRNA by IRPs, must be ensured. We addressed these questions using mass cultures of primary murine erythroid progenitors from fetal liver, either undergoing sustained proliferation or highly synchronous differentiation. We indeed observed strong inhibition of Fer mRNA translation and efficient ALAS-E mRNA translation in differentiating erythroblasts. Moreover, in contrast to self-renewing cells, TfR1 stability and IRP mRNA binding were no longer modulated by iron supply. These and additional data stemming from inhibition of heme synthesis with succinylacetone or from iron overload suggest that highly efficient utilization of iron in mitochondrial heme synthesis during normal erythropoiesis alters the regulation of iron metabolism via the IRE/IRP system.
Collapse
Affiliation(s)
- Matthias Schranzhofer
- Department of Medical Biochemistry, Division of Molecular Biology, Max F. Perutz Laboratories, University Departments at the Vienna Biocenter, Medical University of Vienna, Dr Bohr-Gasse 9, A-1030 Vienna, Austria
| | | | | | | | | | | | | |
Collapse
|
23
|
Cmejla R, Petrak J, Cmejlova J. A novel iron responsive element in the 3'UTR of human MRCKalpha. Biochem Biophys Res Commun 2006; 341:158-66. [PMID: 16412980 DOI: 10.1016/j.bbrc.2005.12.155] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2005] [Accepted: 12/22/2005] [Indexed: 12/22/2022]
Abstract
Human untranslated region (UTR) databases were searched to identify novel proteins potentially regulated by an iron responsive element (IRE), and found two candidates-cell cycle phosphatase Cdc14A variant 1 and myotonic dystrophy kinase-related Cdc42-binding kinase alpha (MRCKalpha), both possessing a putative IRE in their 3'UTR. In further experiments, we focused on MRCKalpha. Biochemical analyses of the MRCKalpha IRE revealed that it was functional and mediated the response to iron level in the same way as transferrin receptor 1 IREs (TfR) did. Similarly to TfR mRNA, MRCKalpha mRNA is stabilized, when iron supply is low, while it is destabilized under iron-rich conditions. The expression of MRCKalpha mRNA was found to be ubiquitous; the highest levels were noted in testes, the lowest in skeletal muscle. The level of MRCKalpha mRNA in various tissues strongly positively correlates with the level of TfR mRNA, indicating its possible role in the transferrin iron uptake pathway.
Collapse
Affiliation(s)
- Radek Cmejla
- Institute of Hematology and Blood Transfusion, Department of Cell Physiology, U Nemocnice 1, Prague, Czech Republic.
| | | | | |
Collapse
|
24
|
Le NTV, Richardson DR. Iron chelators with high antiproliferative activity up-regulate the expression of a growth inhibitory and metastasis suppressor gene: a link between iron metabolism and proliferation. Blood 2004; 104:2967-75. [PMID: 15251988 DOI: 10.1182/blood-2004-05-1866] [Citation(s) in RCA: 236] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
AbstractIron (Fe) is critical for proliferation, but its precise role in cell cycle progression remains unclear. In this study, we examined the mechanisms involved by assessing the effects of Fe chelators on the expression of molecules that play key roles in this process. In initial studies, gene arrays were used to assess gene expression after incubating cells with 2 Fe chelators, namely, desferrioxamine (DFO) and 2-hydroxy-1-naphthylaldehyde isonicotinoyl hydrazone (311), or the DNA-damaging agent, actinomycin D. From the genes assessed, only the N-myc downstream-regulated gene 1 (Ndrg1) was specifically up-regulated by Fe chelation. Although the function of Ndrg1 is unclear, previous studies showed it markedly slows tumor growth and acts as a potent metastasis suppressor. Incubation of cells with chelators markedly increased Ndrg1 mRNA and protein expression, but this was not found with their Fe complexes or when the Fe-binding site had been inactivated. Increased Ndrg1 expression following Fe chelation was related to the permeability and antiproliferative activity of chelators and could be reversed by Fe repletion. Moreover, Ndrg1 up-regulation after chelation occurred at the transcriptional level and was mediated by hypoxia inducible factor-1α (HIF-1α)-dependent and -independent mechanisms. Our investigation suggests Ndrg1 is a novel link between Fe metabolism and the control of proliferation.
Collapse
Affiliation(s)
- Nghia T V Le
- Children's Cancer Institute Australia for Medical Research, The Iron Metabolism and Chelation Program, PO Box 81, High St, Randwick, Sydney, New South Wales, 2031 Australia
| | | |
Collapse
|
25
|
Christian K, Lang M, Maurel P, Raffalli-Mathieu F. Interaction of heterogeneous nuclear ribonucleoprotein A1 with cytochrome P450 2A6 mRNA: implications for post-transcriptional regulation of the CYP2A6 gene. Mol Pharmacol 2004; 65:1405-14. [PMID: 15155834 DOI: 10.1124/mol.65.6.1405] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The human xenobiotic-metabolizing enzyme cytochrome P450, CYP2A6, catalyzes the bioactivation of a number of carcinogens and drugs and is overexpressed in cases of liver diseases, such as cirrhosis, viral hepatitis, and parasitic infestation, and in certain tumor cells. This suggests that CYP2A6 may be a major liver catalyst in pathological conditions. In the present study, we have addressed molecular mechanisms underlying the regulation of the CYP2A6 gene. We present evidence of several proteins present in human hepatocytes that interact specifically with the 3'-untranslated region (UTR) of CYP2A6 mRNA. Biochemical and immunological evidence show that the RNA-protein complex of highest intensity contains the heterogeneous nuclear ribonucleoprotein (hnRNP) A1 or a closely related protein. Mapping of the hnRNP A1 binding site within CYP2A6 3'-UTR reveals that the smallest portion of RNA supporting significant binding consists of 111 central nucleotides of the 3'-UTR. Our studies also indicate that hnRNPA1 from HepG2 cancer cells exhibits modified binding characteristics to the CYP2A6 3'-UTR compared with primary hepatocytes. We found that the level of CYP2A6 mRNA remains high in conditions of impaired transcription in primary human hepatocytes, showing that CYP2A6 expression can be affected post-transcriptionally in conditions of cellular stress. Our results indicate that the post-transcriptional regulation involves interaction of the hnRNP A1 protein with CYP2A6 mRNA. The present data suggest that hnRNPA1 is a critical regulator of expression of the human CYP2A6 gene and support the notion that this P450 isoform may be of particular significance in stressed human liver cells.
Collapse
Affiliation(s)
- Kyle Christian
- Department of Pharmaceutical Biosciences, Division of Biochemistry, Uppsala University, Uppsala, Sweden
| | | | | | | |
Collapse
|
26
|
Campanella A, Levi S, Cairo G, Biasiotto G, Arosio P. Blotting analysis of native IRP1: a novel approach to distinguish the different forms of IRP1 in cells and tissues. Biochemistry 2004; 43:195-204. [PMID: 14705945 DOI: 10.1021/bi035386f] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Iron regulatory protein 1 (IRP1) is a bifunctional protein, which either has aconitase activity or binds to specific mRNA structures to regulate the expression of iron proteins. Using recombinant human IRP1, we found that the two functional forms are resolved by nondenaturing polyacrylamide gel electrophoresis and that they are distinguished from IRP1/RNA complexes. This allowed us to use specific antibodies to develop a blotting system that recognized the iron-free and iron-containing IRP1 forms in the soluble fraction and the RNA-bound IRP1 in the high-speed precipitate fraction of cell extracts. The system was used to study IRP1 in HeLa, K562 cells, and monocytes/macrophages before and after treatment with iron salts, iron chelators, or hydrogen peroxide, as well as in stomach and duodenum biopsies. The results showed that iron-bound aconitase IRP1 is by far the prevalent form in most cells and that the major effect of cellular iron modifications is a shift between free and RNA-bound IRP1. The fraction of RNA-bound IRP1 was highly variable among different cells and was often a minor one. Furthermore, blotting showed that electrophoretic mobility shift assay, as commonly used, tends to under-evaluate the amount of total IRP1 and to over-evaluate the actual RNA-binding activity of IRP1. In conclusion, blotting analysis of IRP1 is a new, useful, and convenient method to analyze the amount and conformations of the protein that reveals previously undetected differences in IRP1 compartmentalization among various cell types.
Collapse
Affiliation(s)
- Alessandro Campanella
- Department of Biological and Technological Research, IRCCS H. San Raffaele, Milan, Italy
| | | | | | | | | |
Collapse
|
27
|
Glisovic T, Söderberg M, Christian K, Lang M, Raffalli-Mathieu F. Interplay between transcriptional and post-transcriptional regulation of Cyp2a5 expression. Biochem Pharmacol 2003; 65:1653-61. [PMID: 12754101 DOI: 10.1016/s0006-2952(03)00118-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The cytochrome P450 (Cyp) 2a5 gene can be upregulated transcriptionally or by mRNA stabilization. The heterogeneous nuclear ribonucleoprotein (hnRNP) A1 interacting with the CYP2A5 mRNA has been shown to be a key post-transcriptional regulator of the Cyp2a5 gene. The aim of this study was to investigate if the transcriptional and post-transcriptional steps of Cyp2a5 expression are linked. This was done by modifying the transcription rate with transcriptional inducers (phenobarbital and cyclic AMP) and inhibitors (actinomycin D and 5,6-dichloro-1-beta-d-ribofuranosylbenzimidazole) and analyzing the effects upon post-transcriptional events. We found that inhibition of transcription led to relocalization of hnRNP A1 from the nucleus to the cytoplasm, to its strongly increased binding to the cytoplasmic CYP2A5 mRNA and to CYP2A5 mRNA stabilization. In contrast, stimulated transcription resulted in increased binding of nuclear hnRNP A1 to the Cyp2a5 promoter, and overexpression of hnRNP A1 led to stimulated transcription of a Cyp2a5 promoter-driven luciferase recombinant. This strongly suggests that the transcriptional and post-transcriptional stages of Cyp2a5 expression are interrelated and that the nucleocytoplasmic shuttling hnRNP A1 may coordinate these different steps.
Collapse
Affiliation(s)
- Tina Glisovic
- Division of Pharmaceutical Biochemistry, Uppsala Biomedical Centre, Uppsala University, Box 578, SE-751 23, Uppsala, Sweden.
| | | | | | | | | |
Collapse
|
28
|
Bouton C, Chauveau MJ, Lazereg S, Drapier JC. Recycling of RNA binding iron regulatory protein 1 into an aconitase after nitric oxide removal depends on mitochondrial ATP. J Biol Chem 2002; 277:31220-7. [PMID: 12039960 DOI: 10.1074/jbc.m203276200] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Iron regulatory proteins (IRPs) control iron metabolism by specifically interacting with iron-responsive elements (IREs) on mRNAs. Nitric oxide (NO) converts IRP-1 from a [4Fe-4S] aconitase to a trans-regulatory protein through Fe-S cluster disassembly. Here, we have focused on the fate of IRE binding IRP1 from murine macrophages when NO flux stops. We show that virtually all IRP-1 molecules from NO-producing cells dissociated from IRE and recovered aconitase activity after re-assembling a [4Fe-4S] cluster in vitro. The reverse change in IRP-1 activities also occurred in intact cells no longer exposed to NO and did not require de novo protein synthesis. Likewise, inhibition of mitochondrial aconitase via NO-induced Fe-S cluster disassembly was also reversed independently of protein translation after NO removal. Our results provide the first evidence of Fe-S cluster repair of NO-modified aconitases in mammalian cells. Moreover, we show that reverse change in IRP-1 activities and repair of mitochondrial aconitase activity depended on energized mitochondria. Finally, we demonstrate that IRP-1 activation by NO was accompanied by both a drastic decrease in ferritin levels and an increase in transferrin receptor mRNA levels. However, although ferritin expression was recovered upon IRP-1-IRE dissociation, expression of transferrin receptor mRNA continued to rise for several hours after stopping NO flux.
Collapse
Affiliation(s)
- Cécile Bouton
- Institut de Chimie des Substances Naturelles, CNRS, Avenue de la Terrasse, 91190 Gif-sur-Yvette, France
| | | | | | | |
Collapse
|
29
|
Abstract
The steady-state levels of mRNAs depend upon their combined rates of synthesis and processing, transport from the nucleus to cytoplasm, and decay in the cytoplasm. In eukaryotic cells, the degradation of mRNA is an essential determinant in the regulation of gene expression, and it can be modulated in response to developmental, environmental, and metabolic signals. This level of regulation is particularly important for proteins that are active for a brief period, such as growth factors, transcription factors, and proteins that control cell cycle progression. The mechanisms by which mRNAs are degraded and the sequence elements within the mRNAs that affect their stability are the subject of this review. We will summarize the current state of knowledge regarding cis-acting elements in mRNA and trans-acting factors that contribute to mRNA regulation decay. We will then consider the mechanisms by which specific signaling proteins seem to contribute to a dynamic organization of the mRNA degradation machinery in response to physiological stimuli.
Collapse
Affiliation(s)
- Hélène Tourrière
- Institut de génétique moléculaire, UMR5535 du CNRS, IFR 24, 1919, route de Mende, 34293 Montpellier cedex 5, France
| | | | | |
Collapse
|
30
|
Jijon HB, Panenka WJ, Madsen KL, Parsons HG. MAP kinases contribute to IL-8 secretion by intestinal epithelial cells via a posttranscriptional mechanism. Am J Physiol Cell Physiol 2002; 283:C31-41. [PMID: 12055070 DOI: 10.1152/ajpcell.00113.2001] [Citation(s) in RCA: 106] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The intracellular pathways that regulate intestinal epithelial gene expression are poorly understood. In this study we examined the roles of extracellular signal-regulated kinase (ERK) and p38 in the expression of interleukin-8 (IL-8) and intercellular adhesion molecule-1 (ICAM-1) using the human intestinal cell line HT-29. HT-29 cells were treated with tumor necrosis factor-alpha (TNF-alpha) in the presence or absence of ERK and p38 pathway inhibitors. TNF-alpha treatment resulted in increased IL-8 and ICAM-1 protein and mRNA synthesis, increased ERK and p38 activity, and activation of the transcription factors activator protein-1 (AP-1) and nuclear factor-kappaB (NF-kappaB). Inhibition of the ERK and p38 pathways attenuated IL-8 secretion but did not alter ICAM-1 expression. Furthermore, AP-1 and NF-kappaB DNA binding was not affected by ERK and p38 inhibition. In contrast, ERK and p38 inhibition resulted in the accelerated degradation of the IL-8 mRNA, suggesting that in HT-29 cells, p38 and ERK contribute to TNF-alpha-stimulated IL-8 secretion by intestinal epithelial cells via a posttranscriptional mechanism that involves stabilization of the IL-8 transcript.
Collapse
Affiliation(s)
- Humberto B Jijon
- Gastrointestinal Research Group, University of Calgary, Calgary T2N 4N1, Canada T6G 2C2.
| | | | | | | |
Collapse
|
31
|
Piñero DJ, Li N, Hu J, Beard JL, Connor JR. The intracellular location of iron regulatory proteins is altered as a function of iron status in cell cultures and rat brain. J Nutr 2001; 131:2831-6. [PMID: 11694604 DOI: 10.1093/jn/131.11.2831] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Iron regulatory proteins (IRPs) are proteins involved in the regulation of intracellular iron homeostasis that bind to specific mRNA structures termed iron responsive elements (IREs). Because the target mRNAs for the IRPs are both cytosolic and membrane associated, we hypothesize that movement of IRPs between the cytosolic and the membrane associated subcellular fractions occurs in response to intracellular iron changes. We tested this hypothesis in a cell culture model, using mouse fibroblast cells (NIH 3T3) and macrophage cells (J774), and in a rat model of early iron deficiency and excess. This presented the first opportunity to examine IRP binding activity in rat brain during states of dietary iron deficiency and excess. Binding activity for IRPs was demonstrated in both membrane and cytosolic fractions in the cell lines and the rat brain homogenates. Although IRP binding activity is predominantly located in the cytosol (90%), there was increased IRP/IRE binding activity in both cytosolic and membrane fractions when the cells were treated with deferoxamine, and decreased binding activity after treatment with iron. In the rat study, brain cortex, hippocampus and striatum homogenates had more IRP binding activity in iron-deficient rats and less in iron-supplemented rats in a region- and time-specific manner. The intracellular distribution of IRPs also changed between the cytosolic and membrane fractions of the brain homogenates in conjunction with changes in iron. These in vivo studies are consistent with the cell culture analyses showing intracellular redistribution of IRPs as a function of iron status. The results of these experiments extend our understanding of cytoplasmic mRNA binding protein activity and raise questions regarding the mechanism by which mRNA binding proteins can locate their target mRNAs within cells. The elucidation of this mechanism will have a significant impact on our understanding of eukaryotic gene regulation.
Collapse
Affiliation(s)
- D J Piñero
- Department of Neuroscience and Anatomy, G.M. Leader Family Laboratory for Alzheimer's Disease Research, Penn State University College of Medicine, Hershey, PA 17033, USA
| | | | | | | | | |
Collapse
|
32
|
Clement JQ, Maiti S, Wilkinson MF. Localization and stability of introns spliced from the Pem homeobox gene. J Biol Chem 2001; 276:16919-30. [PMID: 11278282 DOI: 10.1074/jbc.m005104200] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
RNA splicing generates two products in equal molar amounts, mature mRNAs and spliced introns. Although the mechanism of RNA splicing and the fate of the spliced mRNA products have been well studied, very little is known about the fate and stability of most spliced introns. Research in this area has been hindered by the widely held view that most vertebrate introns are too unstable to be detectable. Here, we report that we are able to detect all three spliced introns from the coding region of the Pem homeobox gene. By using a tetracycline (tet)-regulated promoter, we found that the half-lives of these Pem introns ranged from 9 to 29 min, comparable with those of short lived mRNAs such as those encoding c-fos and c-myc. The half-lives of the Pem introns correlated with both their length and 5' to 3' orientation in the Pem gene. Subcellular fractionation analysis revealed that spliced Pem introns and pre-mRNA accumulated in the nuclear matrix, high salt-soluble, and DNase-sensitive fractions within the nucleus. Surprisingly, we found that all three of the spliced Pem introns were also in the cytoplasmic fraction, whereas Pem pre-mRNAs, U6 small nuclear RNA, and a spliced intron from another gene were virtually excluded from this fraction. This indicates either that spliced Pem introns are uniquely exported to the cytoplasm for degradation or they reside in a unique soluble nuclear fraction. Our study has implications for understanding the regulation of RNA metabolism, as the stability of introns and the location of their degradation may dictate the following: (i) the stability of nearby mRNAs that compete with spliced introns for rate-limiting nucleases, (ii) the rate at which free nucleotides are available for further rounds of transcription, and (iii) the rate at which splicing factors are recycled.
Collapse
Affiliation(s)
- J Q Clement
- Department of Immunology, the University of Texas, M. D. Anderson Cancer Center, Houston, Texas 77030, USA
| | | | | |
Collapse
|
33
|
Abstract
Mammalian iron homeostasis is maintained through the concerted action of sensory and regulatory networks that modulate the expression of proteins of iron metabolism at the transcriptional and/or post-transcriptional levels. Regulation of gene transcription provides critical developmental, cell cycle, and cell-type-specific controls on iron metabolism. Post-transcriptional control through the action of iron regulatory protein 1 (IRP1) and IRP2 coordinate the use of messenger RNA-encoding proteins that are involved in the uptake, storage, and use of iron in all cells of the body. IRPs may also provide a link between iron availability and cellular citrate use. Multiple factors, including iron, nitric oxide, oxidative stress, phosphorylation, and hypoxia/reoxygenation, influence IRP function. Recent evidence indicates that there is diversity in the function of the IRP system with respect to the response of specific IRPs to the same effector, as well as the selectivity with which IRPs modulate the use of specific messenger RNA.
Collapse
Affiliation(s)
- R S Eisenstein
- Department of Nutritional Sciences, University of Wisconsin, Madison, Wisconsin 53706, USA.
| |
Collapse
|
34
|
Meininghaus M, Chapman RD, Horndasch M, Eick D. Conditional expression of RNA polymerase II in mammalian cells. Deletion of the carboxyl-terminal domain of the large subunit affects early steps in transcription. J Biol Chem 2000; 275:24375-82. [PMID: 10825165 DOI: 10.1074/jbc.m001883200] [Citation(s) in RCA: 66] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The carboxyl-terminal domain (CTD) of the large subunit of mammalian RNA polymerase II contains 52 repeats of a heptapeptide that is the target of a variety of kinases. The hyperphosphorylated CTD recruits important factors for mRNA capping, splicing, and 3'-processing. The role of the CTD for the transcription process in vivo, however, is not yet clear. We have conditionally expressed an alpha-amanitin-resistant large subunit with an almost entirely deleted CTD (LS*Delta5) in B-cells. These cells have a defect in global transcription of cellular genes in the presence of alpha-amanitin. Moreover, pol II harboring LS*Delta5 failed to transcribe up to the promoter-proximal pause sites in the hsp70A and c-fos gene promoters. The results indicate that the CTD is already required for steps that occur before promoter-proximal pausing and maturation of mRNA.
Collapse
Affiliation(s)
- M Meininghaus
- Institute for Clinical Molecular Biology and Tumor Genetics, GSF-Research Center for Environment and Health, Marchioninistrasse 25, D-81377 Munich, Germany
| | | | | | | |
Collapse
|
35
|
Clement JQ, Wilkinson MF. Rapid induction of nuclear transcripts and inhibition of intron decay in response to the polymerase II inhibitor DRB. J Mol Biol 2000; 299:1179-91. [PMID: 10873444 DOI: 10.1006/jmbi.2000.3745] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The transcriptional inhibitor 5, 6-dichloro-1-beta-d-ribofuranosylbenzimidazole (DRB) is an adenosine analog that has been shown to cause premature transcriptional termination and thus has been a useful tool to identify factors important for transcriptional elongation. Here, we establish an efficient system for studying DRB-sensitive steps of transcriptional elongation. In addition, we establish two novel effects of DRB not previously reported: intron stabilization and the induction of long transcripts by a mechanism other than premature termination. We found that DRB had a biphasic effect on T-cell receptor-beta (TCRbeta) transcripts driven by a tetracycline (tet)-responsive promoter in transfected HeLa cells. In the first phase, DRB caused a rapid decrease (within five minutes) of pre-mRNA and its spliced intron (IVS1(Cbeta1)), consistent with the known ability of DRB to inhibit transcription. In the second phase (which began ten minutes to two hours after treatment, depending on the dose), DRB dramatically increased the levels of IVS1(Cbeta1)-containing transcripts by a mechanism requiring de novo RNA synthesis. DRB induced the appearance of short 0.4 to 0.8 kb TCRbeta transcripts in vivo, indicating DRB enhances premature transcriptional termination. A approximately 475 nt prematurely terminated transcript (PT) was characterized that terminated at an internal poly(A) tract in the intron IVS1(Cbeta1). We identified three other effects of DRB. First, we observed that DRB induced the appearance of heterodisperse TCRbeta transcripts that were too long ( approximately 1 kb to >8 kb) to result from the type of premature termination events previously described. Their production was not promoter-specific, as we found that long transcripts were induced by DRB from both the tet-responsive and beta-actin promoters. Second, DRB upregulated full-length normal-sized c-myc mRNA, which provided further evidence that DRB has effects besides regulation of premature termination. Third, DRB stabilized lariat forms of the intron IVS1(Cbeta1), indicating that DRB exerts post-transcriptional actions. We propose that our model system will be useful for elucidating the factors that regulate RNA decay and transcriptional elongation in vivo.
Collapse
Affiliation(s)
- J Q Clement
- Department of Immunology, The University of Texas M. D. Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | | |
Collapse
|
36
|
Xu K, Murphy TJ. Reconstitution of angiotensin receptor mRNA down-regulation in vascular smooth muscle. Post-transcriptional control by protein kinase a but not mitogenic signaling directed by the 5'-untranslated region. J Biol Chem 2000; 275:7604-11. [PMID: 10713068 DOI: 10.1074/jbc.275.11.7604] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Cell surface receptor activation generally leads to changes in mRNA abundance, which may involve regulatory targets in processes working at the post-transcriptional level. Many types of agonists down-regulate vascular smooth muscle angiotensin receptor (AT(1)-R) gene expression, but it is unclear which of these activate post-transcriptional mechanisms. To reconstitute faithfully the normal AT(1)-R mRNA regulatory environment, tetracycline-suppressible promoters drive highly accurate recombinant AT(1)-R mRNA mimics in vascular smooth muscle cells that co-express an endogenous AT(1)-R mRNA. Down-regulation of the latter occurs shortly after stimulating mitogenic receptors or by using forskolin, but only cAMP signaling reduces expression of the recombinant AT(1)-R mRNA. Transcription of the recombinant mRNA is unaffected by cAMP signaling. Deletions of the AT(1)-R mRNA 3'-untranslated region do not impair cAMP-mediated down-regulation. Both loss of function and gain of function mutants show the response is mediated by the 5'-untranslated region. These observations provide the first direct functional evidence for modulation of vascular AT(1)-R gene expression by a mechanism involving a protein kinase A-regulated post-transcriptional process.
Collapse
MESH Headings
- Animals
- Aorta/cytology
- Cells, Cultured
- Colforsin/pharmacology
- Cyclic AMP-Dependent Protein Kinases/metabolism
- Down-Regulation
- Half-Life
- Mitogens/pharmacology
- Mitosis
- Muscle, Smooth, Vascular/metabolism
- Mutation
- RNA Processing, Post-Transcriptional
- RNA Stability
- RNA, Messenger/metabolism
- Rats
- Receptor, Angiotensin, Type 1
- Receptor, Angiotensin, Type 2
- Receptors, Angiotensin/genetics
- Signal Transduction
- Tetracyclines/pharmacology
- Untranslated Regions/metabolism
Collapse
Affiliation(s)
- K Xu
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | | |
Collapse
|
37
|
Yeh KY, Yeh M, Glass J. Glucocorticoids and dietary iron regulate postnatal intestinal heavy and light ferritin expression in rats. Am J Physiol Gastrointest Liver Physiol 2000; 278:G217-26. [PMID: 10666045 DOI: 10.1152/ajpgi.2000.278.2.g217] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
To cope with increasing dietary iron exposure, the intestinal epithelium of weaning rats must control intracellular labile iron pools. Intestinal expression of heavy (H) and light (L) ferritin subunits during early weaning and after cortisone administration and/or iron feeding was investigated. Changes in H and L ferritin gene expression were determined by nuclear runoff transcriptional assay, Northern blot analysis, and metabolic labeling of protein synthesis. H ferritin mRNA levels did not change between days 12 and 15, doubled on day 18, and tripled on day 24. L ferritin mRNA was reduced by 50% on days 18 and 24. The protein level of the H and L subunits paralleled the change in mRNAs. Cortisone treatment on day 12 induced a precocious increase of H and decrease of L mRNA expression on day 15. Nuclear runoff assays showed that cortisone did not change H and reduced L ferritin gene transcription. The increased level of H mRNA by cortisone was not translated, unless the rats were fed an iron-fortified diet, which reduced iron regulatory protein activity and stimulated a three- to sixfold increase of ferritin synthesis. Thus changes in intestinal H and L ferritin expression in weaning rats are modulated by glucocorticoids and iron; the former stabilizes H mRNA and suppresses L ferritin gene transcription, and the latter derepresses translation of ferritin mRNA.
Collapse
Affiliation(s)
- K Y Yeh
- Section of Hematology/Oncology, Department of Medicine, Louisiana State University Health Sciences Center, Shreveport, Louisiana 71130, USA
| | | | | |
Collapse
|
38
|
Khier H, Bartl S, Schuettengruber B, Seiser C. Molecular cloning and characterization of the mouse histone deacetylase 1 gene: integration of a retrovirus in 129SV mice. BIOCHIMICA ET BIOPHYSICA ACTA 1999; 1489:365-73. [PMID: 10673037 DOI: 10.1016/s0167-4781(99)00203-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Reversible histone acetylation plays an important role for chromatin structure and gene expression. The acetylation state of core histones is controlled by histone acetyltransferases and histone deacetylases. Here we report the cloning and characterization of the mouse histone deacetylase 1 (HDAC1) gene. The mouse genome contains several HDAC1-related structures representing the HDAC1 gene and at least three pseudogenes. The HDAC1 gene comprises 14 exons ranging from 49 to 539 bp. Interestingly the murine HDAC1 gene strongly resembles the previously published mouse HDAC2 gene (Zeng et al., J. Biol. Chem. 273 (1998) 28921-28930). The sizes of ten of the 14 exons are identical for both genes and the splicing sites for 11 introns align in identical positions suggesting a gene duplication event. The HDAC1 gene is located only 128 bp downstream from the MARCKS-related protein (MRP) gene in a tail-to-tail orientation. The murine MRP gene was previously mapped to a conserved gene cluster on chromosome 4 sharing linkage homology to human chromosome 1p32-36. The genes for HDAC1 and MRP are co-expressed in a variety of cell types. In the genome of 129SV mice the largest intervening sequence of the HDAC1 gene, intron 3, harbors a complete copy of the endogenous retrovirus MuERV-L. In contrast the HDAC1 gene in other mouse strains such as C57B16, C3H/An and C-RY lacks the retrovirus. Our study provides useful tools for future targeted gene disruption studies.
Collapse
Affiliation(s)
- H Khier
- Institute of Molecular Biology, Vienna Biocenter, University of Vienna, Austria
| | | | | | | |
Collapse
|
39
|
Stoll SW, Elder JT. Differential regulation of EGF-like growth factor genes in human keratinocytes. Biochem Biophys Res Commun 1999; 265:214-21. [PMID: 10548517 DOI: 10.1006/bbrc.1999.1654] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
ErbB signaling increases protein levels of multiple EGF-like growth factors in epithelial cells. To better understand this process, we examined the effects of EGF receptor stimulation on the transcription and mRNA stability of TGF-alpha, amphiregulin (AR), and heparin-binding EGF-like growth factor (HB-EGF) in human keratinocytes. EGF stimulation increased transcription of TGF-alpha, AR, and HB-EGF by 3- to 4-fold within 1 to 2 h. However, AR and HB-EGF mRNA levels peaked at 2 h and then rapidly declined, whereas TGF-alpha transcripts remained elevated for at least 6 h. Actinomycin D decay experiments yielded the rank order of transcript stability TGF-alpha > AR > HB-EGF. Interestingly, ligand treatment appeared to stabilize TGF-alpha and AR mRNAs, whereas HB-EGF transcripts were destabilized. These data demonstrate that gene-specific alterations in gene transcription and mRNA stability play important roles in the temporal regulation of EGF-like growth factor gene expression.
Collapse
Affiliation(s)
- S W Stoll
- Department of Dermatology, University of Michigan, Ann Arbor, Michigan, 48109, USA
| | | |
Collapse
|
40
|
|
41
|
Abstract
Iron is an essential nutrient, yet excess iron can be toxic to cells. The uptake of iron by mammalian cells is post-transcriptionally regulated by the interaction of iron-response proteins (IRP1 and IRP2) with iron-response elements (IREs) found in the mRNAs of genes of iron metabolism, such as ferritin, the transferrin receptor, erythroid aminolevulinic acid synthase, and mitochondrial aconitase. The IRPs are RNA binding proteins that bind to the IRE (found in the mRNAs of the regulated genes) in an iron- dependent manner. Binding of IRPs to the IREs leads to changes in the expression of the regulated genes and subsequent changes in the uptake, utilization, or storage of intracellular iron. Recent work has demonstrated that the binding of the IRPs to the IREs can also be modulated by changes in the redox state or oxidative stress level of the cell. These findings provide an important link between iron metabolism and states of oxidative stress.
Collapse
Affiliation(s)
- D J Haile
- Department of Medicine, University of Texas Health Science Center at San Antonio, 78284-7880, USA
| |
Collapse
|
42
|
Posch M, Sutterluety H, Skern T, Seiser C. Characterization of the translation-dependent step during iron-regulated decay of transferrin receptor mRNA. J Biol Chem 1999; 274:16611-8. [PMID: 10347228 DOI: 10.1074/jbc.274.23.16611] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Iron regulates the stability of the mRNA encoding the transferrin receptor (TfR). When iron is scarce, iron regulatory proteins (IRPs) stabilize TfR mRNA by binding to the 3'-untranslated region. High levels of iron induce degradation of TfR mRNA; the translation inhibitor cycloheximide prevents this. To distinguish between cotranslational mRNA decay and a trans effect of translation inhibitors, we designed a reporter system exploiting the properties of the selectable marker gene thymidine kinase (TK). The 3'-untranslated region of human transferrin receptor, which contains all elements necessary for iron-dependent regulation of mRNA stability, was fused to the TK cDNA. In stably transfected mouse fibroblasts, the expression of the reporter gene was perfectly regulated by iron. Introduction of stop codons in the TK coding sequence or insertion of stable stem-loop structures in the leader sequence did not affect on the iron-dependent regulation of the reporter mRNA. This implies that global translation inhibitors stabilize TfR mRNA in trans. Cycloheximide prevented the destabilization of TfR mRNA only in the presence of active IRPs. Inhibition of IRP inactivation by cycloheximide or by the specific proteasome inhibitor MG132 correlated with the stabilization of TfR mRNA. These observations suggest that inhibition of translation by cycloheximide interferes with the rate-limiting step of iron-induced TfR mRNA decay in a trans-acting mechanism by blocking IRP inactivation.
Collapse
Affiliation(s)
- M Posch
- Institute of Molecular Biology, Vienna Biocenter, University of Vienna, A-1030 Vienna, Austria
| | | | | | | |
Collapse
|
43
|
Brennan LE, Nakagawa J, Egger D, Bienz K, Moroni C. Characterisation and mitochondrial localisation of AUH, an AU-specific RNA-binding enoyl-CoA hydratase. Gene X 1999; 228:85-91. [PMID: 10072761 DOI: 10.1016/s0378-1119(99)00003-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
AU-rich elements function as instability elements which direct rapid mRNA degradation. AUH protein exhibits an AU-specific RNA-binding property and an intrinsic enoyl-CoA hydratase activity and may therefore function to link mRNA decay to metabolic processes (. Proc. Natl. Acad. Sci. USA 92, 2051-2055). The sequence encoding the murine protein, muAUH, was established by cloning, and the corresponding polypeptide predicted to have a molecular mass of 37kDa. As shown for the human protein, muAUH is expressed in a 32kDa form and there is 94% homology between the two species. Recombinant muAUH was shown to be an RNA-binding enoyl-CoA hydratase. All murine cells studied contained a single AUH transcript of approx. 1.7kb and an investigation of tissue-specific expression revealed highest levels in kidney, skeletal muscle, heart, liver and spleen. It was further determined, using immunoelectron microscopy, that AUH is located in the mitochondria of mouse cells.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Blotting, Northern
- Blotting, Western
- DNA, Complementary/chemistry
- DNA, Complementary/genetics
- DNA, Complementary/isolation & purification
- Enoyl-CoA Hydratase/analysis
- Enoyl-CoA Hydratase/genetics
- Fluorescent Antibody Technique
- Gene Expression Regulation, Enzymologic
- Humans
- Kidney/enzymology
- Male
- Mast Cells/cytology
- Mast Cells/enzymology
- Mast Cells/ultrastructure
- Mice
- Mice, Inbred BALB C
- Mitochondria/enzymology
- Mitochondria/ultrastructure
- Molecular Sequence Data
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- RNA-Binding Proteins/analysis
- RNA-Binding Proteins/genetics
- Sequence Analysis, DNA
- Sequence Homology, Amino Acid
- Tissue Distribution
Collapse
Affiliation(s)
- L E Brennan
- Institute for Medical Microbiology, University of Basel, Petersplatz 10, CH-4003, Basel, Switzerland
| | | | | | | | | |
Collapse
|
44
|
Rajagopalan LE, Westmark CJ, Jarzembowski JA, Malter JS. hnRNP C increases amyloid precursor protein (APP) production by stabilizing APP mRNA. Nucleic Acids Res 1998; 26:3418-23. [PMID: 9649628 PMCID: PMC147701 DOI: 10.1093/nar/26.14.3418] [Citation(s) in RCA: 100] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
We have previously shown that heterogeneous nuclear ribonucleoprotein C (hnRNP C) and nucleolin bound specifically to a 29 nt sequence in the 3'-untranslated region of amyloid precursor protein (APP) mRNA. Upon activation of peripheral blood mononuclear cells, hnRNP C and nucleolin acquired APP mRNA binding activity, concurrent with APP mRNA stabilization. These data suggested that the regulated interaction of hnRNP C and nucleolin with APP mRNA controlled its stability. Here we have directly examined the role of the cis element and trans factors in the turnover and translation of APP mRNA in vitro . In a rabbit reticulocyte lysate (RRL) translation system, a mutant APP mRNA lacking the 29 nt element was 3-4-fold more stable and synthesized 2-4-fold more APP as wild-type APP mRNA. Therefore, the 29 nt element functioned as an APP mRNA destabilizer. RNA gel mobility shift assays with the RRL suggested the presence of endogenous nucleolin, but failed to show hnRNP C binding activity. However, wild-type APP mRNA was stabilized and coded for 6-fold more APP when translated in an RRL system supplemented with exogenous active hnRNP C. Control mRNAs lacking the 29 nt element were unaffected by hnRNP C supplementation. Therefore, occupancy of the 29 nt element by hnRNP C stabilized APP mRNA and enhanced its translation.
Collapse
Affiliation(s)
- L E Rajagopalan
- Neuroscience Program, Institute on Aging and Department of Pathology and Laboratory Medicine,University of Wisconsin-Medical School, Madison, WI 53792, USA
| | | | | | | |
Collapse
|
45
|
Affiliation(s)
- J S Malter
- Department of Pathology and Laboratory Medicine, University of Wisconsin Hospital and Clinic, Madison 53792, USA
| |
Collapse
|
46
|
Kühn LC. Iron and gene expression: molecular mechanisms regulating cellular iron homeostasis. Nutr Rev 1998; 56:s11-9; discussion s54-75. [PMID: 9564172 DOI: 10.1111/j.1753-4887.1998.tb01681.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
In recent years, specific post-transcriptional mechanisms in the cytoplasm of vertebrate cells have been elucidated that directly affect the stability and translation of mRNAs coding for central proteins in iron metabolism. This review shall focus primarily on these mechanisms. Other levels of control, either affecting gene transcription and/ or related to the function of iron-capturing substances and transmembrane transport, are also likely to exist and to influence the iron balance and utilization. They are, however, much less clear.
Collapse
Affiliation(s)
- L C Kühn
- Swiss Institute for Experimental Cancer Research, Lausanne, Switzerland
| |
Collapse
|
47
|
Goldenberg HA. Regulation of mammalian iron metabolism: current state and need for further knowledge. Crit Rev Clin Lab Sci 1998; 34:529-72. [PMID: 9439884 DOI: 10.3109/10408369709006425] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Due to its character as an essential element for all forms of life, the biochemistry and physiology of iron has attracted very intensive interest for many decades. In more recent years, the ways that iron metabolism is regulated in mammalian and human organisms have been clarified, and many aspects of iron metabolism have been reviewed. In this article, some newer aspects concerning absorption and intracellular regulation of iron concentration are considered. These include a sorting of possible models for intestinal iron absorption, a description of ways for membrane passage of iron after release from transferrin during receptor-mediated endocytosis, a consideration of possible mechanisms for non-transferrin bound iron uptake and its regulation, and a review of recent knowledge on the properties of iron regulatory proteins and on regulation of iron metabolism by these proteins, changes of their own properties by non-iron-mediated influences, and regulatory events not mediated by these proteins. This somewhat heterogeneous collection of themes is a consequence of the intention to avoid repetition of the many aforementioned reviews already existing and to concentrate on newer findings generated within the last couple of years.
Collapse
Affiliation(s)
- H A Goldenberg
- Department of Medical Chemistry, University of Vienna, Austria
| |
Collapse
|
48
|
Morris BJ. Stabilization of dendritic mRNAs by nitric oxide allows localized, activity-dependent enhancement of hippocampal protein synthesis. Eur J Neurosci 1997; 9:2334-9. [PMID: 9464927 DOI: 10.1111/j.1460-9568.1997.tb01650.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
A small number of mRNA species are not restricted to the neuronal cell body, but are also present in neuronal dendrites. The levels of two of these dendritic mRNAs, encoding the microtubule-associated protein MAP2 and the alpha subunit of calcium/calmodulin-dependent protein kinase II (CamKIIalpha), are increased rapidly by high-frequency synaptic activity or by release of nitric oxide. To test the hypothesis that post-transcriptional mechanisms might contribute to this modulation, primary cultures of rat hippocampal neurons were exposed to s-nitroso-N-acetyl penicillamine (SNAP, 200 microM) or vehicle, and mRNA stability was determined. The stability of both CamKIIalpha mRNA and MAP2 mRNA was increased by SNAP treatment, whereas the stabilities of tubulin T26 mRNA and proenkephalin mRNA were unaffected. When the intensity of staining for MAP2 immunoreactivity and CamKIIalpha immunoreactivity was monitored in cultured hippocampal neurons, nitric oxide-releasing agents induced increases in staining intensity that were dependent on protein synthesis but not on mRNA synthesis. These results show that nitric oxide can selectively stabilize CamKIIalpha mRNA and MAP2 mRNA, leading to increased synthesis of the corresponding proteins. This demonstrates a mechanism whereby the presence of a particular mRNA in the vicinity of a synapse permits the levels of the protein product to be regulated by synaptic activity in a manner that is both prolonged and also highly localized to the region of stimulation. Thus, the dependence of sustained synaptic plasticity on de novo protein synthesis need not entail a loss of anatomical specificity.
Collapse
Affiliation(s)
- B J Morris
- Division of Neuroscience and Biomedical Systems, Institute of Biomedical and Life Sciences, Glasgow University, UK
| |
Collapse
|
49
|
Ralston E, McLaren RS, Horowitz JA. Nuclear domains in skeletal myotubes: the localization of transferrin receptor mRNA is independent of its half-life and restricted by binding to ribosomes. Exp Cell Res 1997; 236:453-62. [PMID: 9367630 DOI: 10.1006/excr.1997.3753] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The retention of mRNAs near the nuclei that synthesize them may be an important feature of the organization of multinucleated skeletal myotubes. Here, we assess the possible role of two factors in this localization. First, we examine the role of mRNA half-life, by studying the distribution of the mRNA for the transferrin receptor (TfR), whose half-life can be manipulated in culture by changing the availability of iron. In situ hybridization of myotubes of the mouse muscle cell line C2 shows that TfR mRNA is concentrated in the core of the myotubes. Its distribution around the nuclei is often asymmetric and its concentration changes abruptly. Stable transcripts display the same asymmetric localization as unstable ones, suggesting that half-life does not determine subcellular localization of TfR mRNA. Differential effects of the protein synthesis inhibitors puromycin and cycloheximide suggest that the mRNA is retained in position by its association with ribosomes. We then examine the distribution of the rough endoplasmic reticulum (RER) and find it to be broader than the distribution of TfR mRNA. In contrast to TfR mRNA, the mRNA for a secreted immunoglobulin kappa light chain has a more uniform distribution. Taken together, the results suggest that TfR mRNA may associate with RER subdomains by specific targeting.
Collapse
Affiliation(s)
- E Ralston
- Laboratory of Neurobiology, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland 20892-4062, USA.
| | | | | |
Collapse
|
50
|
Alonso J, Sánchez de Miguel L, Montón M, Casado S, López-Farré A. Endothelial cytosolic proteins bind to the 3' untranslated region of endothelial nitric oxide synthase mRNA: regulation by tumor necrosis factor alpha. Mol Cell Biol 1997; 17:5719-26. [PMID: 9315630 PMCID: PMC232420 DOI: 10.1128/mcb.17.10.5719] [Citation(s) in RCA: 115] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Changes in endothelial nitric oxide synthase (eNOS) expression may be involved in the endothelium-dependent vasorelaxation dysfunction associated with several vascular diseases. In the present work, we demonstrate that eNOS mRNA contains a previously undescribed cis element in the 3' untranslated region (3' UTR). A U+C-rich segment in the 3' UTR is critical in complex formation with bovine aortic endothelial cell cytosolic proteins. Tumor necrosis factor alpha (TNF-alpha), which destabilizes eNOS mRNA, increased the binding activity of the cytosolic proteins in a time-dependent manner. These data suggest that endothelial cytosolic proteins bind to the 3' UTR of eNOS mRNA. These proteins may play a role in TNF-alpha-induced eNOS mRNA destabilization.
Collapse
Affiliation(s)
- J Alonso
- Nephrology, Hypertension and Cardiovascular Research Laboratory, Instituto de Investigaciones Médicas, Fundación Jiménez Díaz, Madrid, Spain
| | | | | | | | | |
Collapse
|