Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Clin Epigenetics ; 14(1): 59, 2022 05 03.
Article in English | MEDLINE | ID: mdl-35505416

ABSTRACT

BACKGROUND: Unbalanced iron homeostasis in pregnancy is associated with an increased risk of adverse birth and childhood health outcomes. DNA methylation has been suggested as a potential underlying mechanism linking environmental exposures such as micronutrient status during pregnancy with offspring health. We performed a meta-analysis on the association of maternal early-pregnancy serum ferritin concentrations, as a marker of body iron stores, and cord blood DNA methylation. We included 1286 mother-newborn pairs from two population-based prospective cohorts. Serum ferritin concentrations were measured in early pregnancy. DNA methylation was measured with the Infinium HumanMethylation450 BeadChip (Illumina). We examined epigenome-wide associations of maternal early-pregnancy serum ferritin and cord blood DNA methylation using robust linear regression analyses, with adjustment for confounders and performed fixed-effects meta-analyses. We additionally examined whether associations of any CpGs identified in cord blood persisted in the peripheral blood of older children and explored associations with other markers of maternal iron status. We also examined whether similar findings were present in the association of cord blood serum ferritin concentrations with cord blood DNA methylation. RESULTS: Maternal early-pregnancy serum ferritin concentrations were inversely associated with DNA methylation at two CpGs (cg02806645 and cg06322988) in PRR23A and one CpG (cg04468817) in PRSS22. Associations at two of these CpG sites persisted at each of the follow-up time points in childhood. Cord blood serum ferritin concentrations were not associated with cord blood DNA methylation levels at the three identified CpGs. CONCLUSION: Maternal early-pregnancy serum ferritin concentrations were associated with lower cord blood DNA methylation levels at three CpGs and these associations partly persisted in older children. Further studies are needed to uncover the role of these CpGs in the underlying mechanisms of the associations of maternal iron status and offspring health outcomes.


Subject(s)
DNA Methylation , Epigenome , Adolescent , Child , Epigenesis, Genetic , Female , Ferritins/genetics , Genome-Wide Association Study , Humans , Infant, Newborn , Iron , Pregnancy , Prospective Studies
2.
Free Radic Biol Med ; 160: 501-505, 2020 11 20.
Article in English | MEDLINE | ID: mdl-32861780

ABSTRACT

Hepcidin deficiency leads to iron overload by increased dietary iron uptake and iron release from storage cells. The most frequent mutation in Hfe leads to reduced hepcidin expression and thereby causes iron overload. Recent findings suggested that HFE activates hepcidin expression predominantly via the BMP type I receptor ALK3. Here, we investigated whether HFE exclusively utilizes ALK3 or other signaling mechanisms also. We generated mice with double deficiency of Hfe and hepatocyte-specific Alk3 and compared the iron overload phenotypes of these double knockout mice to single hepatocyte-specific Alk3 deficient or Hfe knockout mice. Double Hfe-/-/hepatic Alk3fl/fl;Alb-Cre knockouts develop a similar iron overload phenotype compared to single hepatocyte-specific Alk3 deficient mice hallmarked by serum iron levels, tissue iron content and hepcidin levels of similar grades. HFE protein levels were increased in Alk3fl/fl;Alb-Cre mice compared to Alk3fl/fl mice, which was caused by iron overload - and not by Alk3 deficiency. The data provide evidence by genetic means that 1. HFE exclusively uses the BMP type I receptor ALK3 to induce hepcidin expression and 2. HFE protein expression is induced by iron overload, which further emphasizes the iron sensing function of HFE.


Subject(s)
Hepcidins , Iron Overload , Animals , Bone Morphogenetic Protein Receptors, Type I , Hemochromatosis Protein/genetics , Hepcidins/genetics , Histocompatibility Antigens Class I/genetics , Iron Overload/genetics , Liver/metabolism , Mice , Mice, Knockout , Signal Transduction
3.
Blood Cancer J ; 7(2): e523, 2017 02 03.
Article in English | MEDLINE | ID: mdl-28157215

ABSTRACT

In the search for genes that define critical steps of relapse in pediatric T-cell acute lymphoblastic leukemia (T-ALL) and can serve as prognostic markers, we performed targeted sequencing of 313 leukemia-related genes in 214 patients: 67 samples collected at the time of relapse and 147 at initial diagnosis. As relapse-specific genetic events, we identified activating mutations in NT5C2 (P=0.0001, Fisher's exact test), inactivation of TP53 (P=0.0007, Fisher's exact test) and duplication of chr17:q11.2-24.3 (P=0.0068, Fisher's exact test) in 32/67 of T-ALL relapse samples. Alterations of TP53 were frequently homozygous events, which significantly correlated with higher rates of copy number alterations in other genes compared with wild-type TP53 (P=0.0004, Mann-Whitney's test). We subsequently focused on mutations with prognostic impact and identified genes governing DNA integrity (TP53, n=8; USP7, n=4; MSH6, n=4), having key roles in the RAS signaling pathway (KRAS, NRAS, n=8), as well as IL7R (n=4) and CNOT3 (n=4) to be exclusively mutated in fatal relapses. These markers recognize 24/49 patients with a second event. In 17 of these patients with mostly refractory relapse and dire need for efficient treatment, we identified candidate targets for personalized therapy with p53 reactivating compounds, MEK inhibitors or JAK/STAT-inhibitors that may be incorporated in future treatment strategies.


Subject(s)
Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Child , Child, Preschool , Disease-Free Survival , Humans , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Prognosis , Risk Factors
4.
Bone Marrow Transplant ; 51(1): 89-95, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26367238

ABSTRACT

Elevated serum ferritin contributes to treatment-related morbidity and mortality after allogeneic hematopoietic stem cell transplantation (HSCT). The multicenter DE02 trial assessed the safety, efficacy and impact of deferasirox on iron homeostasis after allogeneic HSCT. Deferasirox was administered at a starting dose of 10 mg/kg per day to 76 recipients of allogeneic HSCT, with subsequent dose adjustments based on efficacy and safety. Deferasirox was initiated at a median of 168 days after HSCT, with 84% of patients still on immunosuppression. Baseline serum ferritin declined from 2045 to 957 ng/mL. Deferasirox induced a negative iron balance in 84% of patients. Hemoglobin increased in the first 3 months, and trough serum cyclosporine levels were stable. Median exposure was 330 days, with a median compliance rate of >80%. The most common investigator-reported drug-related adverse events (AEs) were increased blood creatinine (26.5%), nausea (9.0%) and abdominal discomfort (8.3%). Fifty-four (71.1%) patients experienced drug-related AEs, which occasionally resulted in discontinuation (gastrointestinal (n=6), skin (n=3), elevated transaminases (n=1) and creatinine (n=1)). The incidence of AEs appeared to be dose related, with 7.5 mg/kg per day being the best-tolerated dose. Low-dose deferasirox is an effective chelation therapy after allogeneic HSCT, with a manageable safety profile, even in patients receiving cyclosporine.


Subject(s)
Benzoates/administration & dosage , Benzoates/pharmacokinetics , Ferritins/blood , Hematopoietic Stem Cell Transplantation , Iron Metabolism Disorders , Iron/blood , Triazoles/administration & dosage , Triazoles/pharmacokinetics , Adult , Aged , Allografts , Benzoates/adverse effects , Cyclosporine/administration & dosage , Cyclosporine/adverse effects , Cyclosporine/blood , Deferasirox , Female , Humans , Iron Metabolism Disorders/blood , Iron Metabolism Disorders/drug therapy , Iron Metabolism Disorders/etiology , Male , Middle Aged , Prospective Studies , Triazoles/adverse effects
5.
Exp Clin Endocrinol Diabetes ; 122(4): 201-7, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24623503

ABSTRACT

The clinical symptoms of diabetic neuropathy (DN) manifest in a time dependent manner as a positive symptoms (i. e. pain, hypersensitivity, tingling, cramps, cold feet etc.) during its early stages and by a loss of function (i. e. loss of sensory perception, delayed wound healing etc.) predominating in the later stages. Elevated blood glucose alone cannot explain the development and progression of DN and the lowering of blood glucose is insufficient in preventing and/or reversing neuropathy in patients with type 2 diabetes. Recently it has been shown that the endogenous reactive metabolite methylglyoxal (MG) can contribute to the gain of function via post-translational modification in DN of neuronal ion channels involved in chemosensing and action potential generation in nociceptive nerve endings. Dicarbonyls, such as MG, that are elevated in diabetic patients, modify DNA as well as extra- and intracellular proteins, leading to the formation of advanced glycation endproducts (AGEs). Increased formation of AGEs leads to increased cellular stress, dysfunction and ultimately cell death. The interaction of AGE-modified proteins through cell surface receptors, such as RAGE, can lead to increased cellular activation and sustained inflammatory responses, which are the molecular hallmarks of the later, degenerative, stages of DN. The direct and indirect effects of dicarbonyls on nerves or neuronal microvascular network provides a unifying mechanism for the development and progression of DN. Targeting the accumulation of MG and/or prevention of RAGE interactions may therefore provide new, more effective, therapeutic approaches for the treatment of DN.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Diabetic Neuropathies/metabolism , Diabetic Neuropathies/therapy , Animals , Blood Glucose/metabolism , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/therapy , Diabetic Neuropathies/blood , Glycation End Products, Advanced/blood , Glycation End Products, Advanced/metabolism , Glyoxal/blood , Glyoxal/metabolism , Humans , Receptor for Advanced Glycation End Products , Receptors, Immunologic/blood , Receptors, Immunologic/metabolism
6.
Leukemia ; 24(12): 2005-13, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20944675

ABSTRACT

Precursor T-cell acute lymphoblastic leukemia (T-ALL) remains an important challenge in pediatric oncology. Because of the particularly poor prognosis of relapses, it is vital to identify molecular risk factors allowing early and effective treatment stratification. Activating NOTCH1 mutations signify a favorable prognosis in patients treated on ALL-BFM protocols. We have now tested if NOTCH pathway activation at different steps has similar clinical effects and if multiple mutations in this pathway function synergistically. Analysis of a validation set of 151 T-ALL patients and of the total cohort of 301 patients confirms the low relapse rate generally and the overall favorable effect of activating NOTCH1 mutations. Subgroup analysis shows that the NOTCH1 effect in ALL-BFM is restricted to patients with rapid early treatment response. Inactivation of the ubiquitin ligase FBXW7 is associated with rapid early treatment response and synergizes with NOTCH1 receptor activation. However, the effect of FBXW7 inactivation is separable from NOTCH1 activation by not synergizing with NOTCH1 mutations in predicting favorable long-term outcome, which can probably be explained by the interaction of FBXW7 with other clients. Finally, the comparison with other European protocols suggests that the NOTCH effect is treatment dependent generally and may depend on the intensity of central nervous system-directed therapy specifically.


Subject(s)
Cell Cycle Proteins/genetics , F-Box Proteins/genetics , Mutation , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Prednisone/therapeutic use , Receptor, Notch1/genetics , Ubiquitin-Protein Ligases/genetics , Cell Cycle Proteins/physiology , Child , F-Box Proteins/physiology , F-Box-WD Repeat-Containing Protein 7 , Humans , Treatment Outcome , Ubiquitin-Protein Ligases/physiology
7.
Z Gastroenterol ; 47(12): 1211, 2009 Dec.
Article in German | MEDLINE | ID: mdl-19994473

ABSTRACT

We report on a 23-year-old woman who presented with elevated serum ferritin values at our department. She had undergone cataract surgery at the age of 14 and her family pedigree showed hereditary autosomal-dominant cataract. The combination of isolated hyperferritinemia with autosomal-dominant hereditary cataract led to the diagnosis of the hereditary hyperferritinemia cataract syndrome (HHCS) which we now describe in a German family for the first time. HHCS was confirmed by detection of a causal mutation at position 32 within the iron responsive element (IRE) of L-ferritin leading to a guanine to adenine exchange and the pathognomonic star-shaped cataract. This mutation interrupts the post-transcriptional control of L-ferritin. It prevents binding of the iron regulatory protein 1 (IRP1) to the 5alpha untranslated region of L-ferritin resulting in uncontrolled L-ferritin synthesis and high serum ferritin levels independent of the body iron stores. Premature cataract is eventually caused by deposition of L-ferritin crystals in the lens of the eye. Our family shows the typical autosomal-dominant inheritance of HHCS over four generations affecting a total of 17 family members. The causal mutation, star-shaped cataract and typical laboratory configuration were confirmed in five patients. Thus, in gastroenterological practice, HHCS should be added as a differential diagnosis of hyperferritinemia in Germany. Importantly, patients with HHCS can be spared from invasive diagnostics such as liver biopsy.


Subject(s)
Apoferritins/genetics , Cataract/diagnosis , Cataract/genetics , Iron Metabolism Disorders/diagnosis , Iron Metabolism Disorders/genetics , Adult , Female , Genetic Predisposition to Disease/genetics , Heterozygote , Humans , Polymorphism, Single Nucleotide/genetics , Syndrome
8.
Biotechniques ; 33(3): 620-8, 630, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12238772

ABSTRACT

Gene expression profiling by DNA microarrays has found wide application in many fields of biomedical research. The protocols for this technique are not yet standardized, and for each given step in microarray analysis a number of different protocols are in use. As a consequence, results obtained in different laboratories can be difficult to compare. Of particular importance in this respect are the methods for the preparation of fluorescent cDNA probes that should quantitatively reflect the abundance of different mRNAs in the two samples to be compared. Here we systematically evaluate and compare five different published and/or commercial principles for the synthesis offluorescently labeled probes for microarray analysis (direct labeling, 77 RNA polymerase amplification, aminoallyl labeling, hapten-antibody enzymatic labeling, and 3-D multi-labeled structures). We show that individual labeling methods can significantly influence the expression pattern obtained in a microarray experiment and discuss the respective benefits and limitations of each method.


Subject(s)
DNA Probes/chemical synthesis , Fluorescent Dyes/chemical synthesis , Gene Expression Profiling/methods , Oligonucleotide Array Sequence Analysis/methods , DNA, Complementary/chemistry , HeLa Cells/physiology , Humans , Iron Deficiencies , Reproducibility of Results , Sensitivity and Specificity , Staining and Labeling/methods
9.
Blood ; 94(11): 3915-21, 1999 Dec 01.
Article in English | MEDLINE | ID: mdl-10572108

ABSTRACT

Hereditary hemochromatosis (HH) is a common autosomal-recessive disorder of iron metabolism. More than 80% of HH patients are homozygous for a point mutation in a major histocompatibility complex (MHC) class I type protein (HFE), which results in a lack of HFE expression on the cell surface. A previously identified interaction of HFE and the transferrin receptor suggests a possible regulatory role of HFE in cellular iron absorption. Using an HeLa cell line stably transfected with HFE under the control of a tetracycline-sensitive promoter, we investigated the effect of HFE expression on cellular iron uptake. We demonstrate that the overproduction of HFE results in decreased iron uptake from diferric transferrin. Moreover, HFE expression activates the key regulators of intracellular iron homeostasis, the iron-regulatory proteins (IRPs), implying that HFE can affect the intracellular "labile iron pool." The increase in IRP activity is accompanied by the downregulation of the iron-storage protein, ferritin, and an upregulation of transferrin receptor levels. These findings are discussed in the context of the pathophysiology of HH and a possible role of iron-responsive element (IRE)-containing mRNAs.


Subject(s)
HLA Antigens/genetics , HLA Antigens/metabolism , Hemochromatosis/genetics , Hemochromatosis/metabolism , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/metabolism , Iron-Sulfur Proteins/metabolism , Iron/metabolism , Membrane Proteins , RNA-Binding Proteins/metabolism , Transferrin/metabolism , Down-Regulation , Gene Expression Regulation , Genes, MHC Class I , HeLa Cells , Hemochromatosis Protein , Humans , Iron-Regulatory Proteins , Iron-Sulfur Proteins/genetics , Point Mutation , RNA-Binding Proteins/genetics , Transfection , Transferrin/genetics
10.
RNA ; 4(11): 1321-31, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9814754

ABSTRACT

The cap structure and the poly(A) tail synergistically activate mRNA translation in vivo. Recent work using Saccharomyces cerevisiae spheroplasts and a yeast cell-free translation system revealed that the poly(A) tail can function as an independent promotor for ribosome recruitment, to internal initiation sites within an mRNA. This raises the question of how regulatory upstream open reading frames and translational repressor proteins binding to the 5'UTR can function, as well as how regulated polyadenylation can support faithful activation of protein synthesis. We investigated the function of the regulatory upstream open reading frame 4 from the yeast GCN 4 gene and the effect of IRP-1 binding to an iron-responsive element introduced into the 5' UTR of reporter mRNAs. Both manipulations effectively block cap-dependent translation, whereas ribosome recruitment promoted by the poly(A) tail under non-competitive conditions can efficiently bypass both blocks. We show that the synergistic use of both, the cap structure and the poly-A tail enforced by mRNA competition reinstates the full extent of translational control by both types of 5' UTR regulatory elements. With a view towards regulated polyadenylation, we studied the function of poly(A) tails of defined length on the translation of capped mRNAs. We find that poly(A) tail elongation increases translational efficiency, particularly under competitive conditions. Our results integrate recent findings on the function of the poly(A) tail into an understanding of translational control.


Subject(s)
DNA-Binding Proteins , Gene Expression Regulation, Fungal , Genes, Fungal/genetics , Protein Biosynthesis/genetics , RNA, Messenger/genetics , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae/genetics , Terminator Regions, Genetic/genetics , 5' Untranslated Regions/genetics , Base Sequence , Binding, Competitive , Chloramphenicol O-Acetyltransferase/immunology , Fungal Proteins/genetics , Iron Regulatory Protein 1 , Iron-Regulatory Proteins , Iron-Sulfur Proteins/metabolism , Molecular Sequence Data , Molecular Weight , Mutagenesis, Site-Directed , Open Reading Frames/genetics , Precipitin Tests , Protein Kinases/genetics , RNA Caps/genetics , RNA, Fungal/genetics , RNA, Fungal/metabolism , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Regulatory Sequences, Nucleic Acid/genetics , Transcription, Genetic , Uridine
11.
Mol Cell ; 2(3): 383-8, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9774976

ABSTRACT

Binding of iron regulatory proteins (IRPs) to IREs located in proximity to the cap structure of ferritin H- and L-chain mRNAs blocks ferritin synthesis by preventing the recruitment of the small ribosomal subunit to the mRNA. We have devised a novel procedure to examine the assembly of translation initiation factors (eIFs) on regulated mRNAs. Unexpectedly, we find that the cap binding complex eIF4F (comprising eIF4E, eIF4G, and eIF4A) assembles even when IRP-1 is bound to the cap-proximal IRE. This assembly is futile, because bridging interactions between eIF4F and the small ribosomal subunit cannot be established in the presence of IRP-1. Our findings provide insight into translational control by an mRNA binding protein at the level of translation initiation factors and uncover a key regulatory step in iron homeostasis.


Subject(s)
Ferritins/genetics , Iron-Sulfur Proteins/metabolism , Peptide Chain Initiation, Translational , Peptide Initiation Factors/metabolism , RNA Caps/metabolism , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Ribosomes/metabolism , Animals , Binding Sites , Eukaryotic Initiation Factor-4F , Humans , Iron Regulatory Protein 1 , Iron-Regulatory Proteins , Iron-Sulfur Proteins/chemistry , Models, Genetic , Models, Molecular , Peptide Initiation Factors/chemistry , Protein Biosynthesis , RNA Caps/chemistry , RNA, Messenger/chemistry , RNA, Messenger/genetics , RNA-Binding Proteins/chemistry , Rabbits , Reticulocytes/metabolism , Ribosomes/ultrastructure
12.
EMBO J ; 17(18): 5427-37, 1998 Sep 15.
Article in English | MEDLINE | ID: mdl-9736620

ABSTRACT

Exonucleolytic degradation of the poly(A) tail is often the first step in the decay of eukaryotic mRNAs and is also used to silence certain maternal mRNAs translationally during oocyte maturation and early embryonic development. We previously described the purification of a poly(A)-specific 3'-exoribonuclease (deadenylating nuclease, DAN) from mammalian tissue. Here, the isolation and functional characterization of cDNA clones encoding human DAN is reported. Recombinant DAN overexpressed in Escherichia coli has properties similar to those of the authentic protein. The amino acid sequence of DAN shows homology to the RNase D family of 3'-exonucleases. DAN appears to be localized in both the nucleus and the cytoplasm. It is not stably associated with polysomes or ribosomal subunits. Xenopus oocytes contain nuclear and cytoplasmic DAN isoforms, both of which are closely related to the human DAN. Anti-DAN antibody microinjected into oocytes inhibits default deadenylation during progesterone-induced maturation. Ectopic expression of human DAN in enucleated oocytes rescues maturation-specific deadenylation, indicating that amphibian and mammalian DANs are functionally equivalent.


Subject(s)
Exoribonucleases/metabolism , Meiosis/genetics , RNA, Messenger/metabolism , Amino Acid Sequence , Animals , Cell Nucleus/enzymology , Cloning, Molecular , Cytoplasm/enzymology , DNA, Complementary , Escherichia coli , Exoribonucleases/genetics , Humans , Molecular Sequence Data , Oocytes , Poly(A)-Binding Proteins , RNA-Binding Proteins/metabolism , Recombinant Fusion Proteins , Sequence Homology, Amino Acid , Xenopus
13.
Eur J Biochem ; 254(2): 223-9, 1998 Jun 01.
Article in English | MEDLINE | ID: mdl-9660174

ABSTRACT

Two ferritin cDNAs were cloned from the liver and spinal cord of the sanguivore lamprey Lampetra fluviatilis, an extant representative of the ancient agnathan (jawless) stage in vertebrate evolution. The deduced proteins of 20.2 kDa (H-subunit) and 20.1 kDa (M-subunit) display 73% sequence identity, and both contain the ferroxidase center characteristic of animal H-ferritin. A highly conserved iron-responsive element (IRE) was identified in the 5' untranslated region of lamprey H-ferritin. Lamprey ferritin IRE forms a specific complex with crude lamprey and rat liver extracts, and with recombinant human iron-regulatory protein (IRP-1) in an electrophoretic mobility shift assay. Furthermore, lamprey ferritin IRE competes with labeled human ferritin IRE for binding to IRP in lamprey and mammalian extracts. Two liver cDNA sequences encoding 323 residues and 101 residues of two genetically distinct lamprey IRP were amplified by PCR. Lamprey IRP-1 and IRP-2, which are 72% identical, display about 74% sequence identity to their presumed homologues in mammals. Northern blot analysis shows that two IRP transcripts of 3.6 kb and 5.8 kb are expressed in lamprey liver. Given the ancient lineage of lampreys, the results indicate that the IRE/IRP regulatory system has remained highly conserved during the evolution of vertebrates.


Subject(s)
Evolution, Molecular , Ferritins/genetics , Ferritins/metabolism , Iron-Sulfur Proteins/genetics , Iron-Sulfur Proteins/metabolism , Iron/metabolism , Lampreys/genetics , Lampreys/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , Conserved Sequence , DNA Primers/genetics , DNA, Complementary/genetics , Ferritins/chemistry , Humans , In Vitro Techniques , Iron Regulatory Protein 1 , Iron Regulatory Protein 2 , Iron-Regulatory Proteins , Iron-Sulfur Proteins/chemistry , Molecular Sequence Data , Polymerase Chain Reaction , Protein Conformation , RNA-Binding Proteins/chemistry , Rats , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid
14.
Eur J Biochem ; 254(2): 230-7, 1998 Jun 01.
Article in English | MEDLINE | ID: mdl-9660175

ABSTRACT

Iron-regulatory protein-1 (IRP-1) plays a dual role as a regulatory RNA-binding protein and as a cytoplasmic aconitase. When bound to iron-responsive elements (IRE), IRP-1 post-transcriptionally regulates the expression of mRNAs involved in iron metabolism. IRP have been cloned from several vertebrate species. Using a degenerate-primer PCR strategy and the screening of data bases, we now identify the homologues of IRP-1 in two invertebrate species, Drosophila melanogaster and Caenorhabditis elegans. Comparative sequence analysis shows that these invertebrate IRP are closely related to vertebrate IRP, and that the amino acid residues that have been implicated in aconitase function are particularly highly conserved, suggesting that invertebrate IRP may function as cytoplasmic aconitases. Antibodies raised against recombinant human IRP-1 immunoprecipitate the Drosophila homologue expressed from the cloned cDNA. In contrast to vertebrates, two IRP-1 homologues (Drosophila IRP-1A and Drosophila IRP-1B), displaying 86% identity to each other, are expressed in D. melanogaster. Both of these homologues are distinct from vertebrate IRP-2. In contrast to the mammalian system where the two IRP (IRP-1 and IRP-2) are differentially expressed, Drosophila IRP-1A and Drosophila IRP-1B are not preferentially expressed in specific organs. The localization of Drosophila IRP-1A to position 94C1-8 and of Drosophila IRP-1B to position 86B3-6 on the right arm of chromosome 3 and the availability of an IRP-1 cDNA from C. elegans will facilitate a genetic analysis of the IRE/IRP system, thus opening a new avenue to explore this regulatory network.


Subject(s)
Caenorhabditis elegans/genetics , Drosophila melanogaster/genetics , Iron-Sulfur Proteins/genetics , RNA-Binding Proteins/genetics , Amino Acid Sequence , Animals , Base Sequence , Chromosome Mapping , Cloning, Molecular , Conserved Sequence , DNA Primers/genetics , DNA, Complementary/genetics , Drosophila melanogaster/embryology , Drosophila melanogaster/metabolism , Evolution, Molecular , Gene Expression Regulation, Developmental , Humans , Iron Regulatory Protein 1 , Iron Regulatory Protein 2 , Iron-Regulatory Proteins , Molecular Sequence Data , RNA, Messenger/genetics , RNA, Messenger/metabolism
15.
RNA ; 3(9): 983-95, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9292498

ABSTRACT

The poly(A) tail plays an important role in translation initiation. We report the identification of a mechanism that operates in mammalian somatic cells, and couples mRNA poly(A) tail length with its translation state. The regulation of human ferritin L-chain mRNA by iron-responsive elements (IREs) and iron regulatory proteins (IRPs) is subject to this mechanism: translational repression imposed by IRP binding to the IRE of ferritin L-chain mRNA induces poly(A) tail shortening. For the accumulation of mRNAs with short poly(A) tails, IRP binding to an IRE per se is not sufficient, but must cause translational repression. Interestingly, puromycin and verrucarin (general translation inhibitors that dissociate mRNAs from ribosomes) mimick the negative effect of the specific translational repressor proteins on poly(A) tail length, whereas cycloheximide and anisomycin (general translation inhibitors that maintain the association between mRNAs and ribosomes) preserve long poly(A) tails. Thus, the ribosome association of the mRNA appears to represent the critical determinant. These findings identify a novel mechanism of regulated polyadenylation as a consequence of translational control. They reveal differences in poly(A) tail metabolism between polysomal and mRNP-associated mRNAs. A possible role of this mechanism in the maintenance of translational repression is discussed.


Subject(s)
Poly A/metabolism , Protein Biosynthesis , Repressor Proteins/metabolism , Ribosomes/genetics , Animals , Ferritins/genetics , Ferritins/metabolism , Human Growth Hormone/genetics , Human Growth Hormone/metabolism , Humans , Iron/metabolism , Kinetics , Mammals , Poly A/genetics , RNA Caps , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Repressor Proteins/genetics , Ribosomes/metabolism
17.
Nucleic Acids Res ; 23(3): 405-12, 1995 Feb 11.
Article in English | MEDLINE | ID: mdl-7885836

ABSTRACT

The HIV-1 promoter directs the high level production of transcripts in Xenopus oocytes. However, despite being exported to the cytoplasm, the transcripts are not translated [M. Braddock, A. M. Thorburn, A. Chambers, G. D. Elliott, G. J. Anderson, A. J. Kingsman and S. M. Kingsman (1990) Cell, 62, 1123-1133]. We have shown previously that this is a function of promoter sequences and is independent of the TAR RNA element that is normally located at the 5' end of all HIV mRNAs. We now show that a three nucleotide substitution at position -340, upstream of the RNA start site, reverses the translation inhibition. This site coincides with a sequence that can bind the haematopoietic transcription factor GATA. The inhibition of translation can also be reversed by treatment with inhibitors of casein kinase II or by injection into the nucleus of antibodies specific for the FRGY2 family of RNP proteins. We suggest that the -340 site influences the quality of the transcription complex such that transcripts are diverted to a nucleus-dependent translation inhibition pathway.


Subject(s)
HIV-1/genetics , Promoter Regions, Genetic/genetics , Protein Biosynthesis/genetics , RNA, Messenger/metabolism , Xenopus Proteins , Animals , Base Sequence , Casein Kinase II , Cell Nucleus/metabolism , Cytoplasm/metabolism , DNA/genetics , DNA/metabolism , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/pharmacology , Erythroid-Specific DNA-Binding Factors , HIV Long Terminal Repeat/genetics , Molecular Sequence Data , Mutation/physiology , Oocytes , Protein Biosynthesis/drug effects , Protein Serine-Threonine Kinases/antagonists & inhibitors , Quercetin/pharmacology , RNA, Messenger/biosynthesis , RNA-Binding Proteins/physiology , Rutin/pharmacology , Transcription Factors/metabolism , Transcription Factors/pharmacology , Transcription Factors/physiology , Xenopus laevis
18.
Nucleic Acids Res ; 22(24): 5255-64, 1994 Dec 11.
Article in English | MEDLINE | ID: mdl-7816614

ABSTRACT

The translation of a capped, polyadenylated RNA after injection into the nucleus of Xenopus oocytes occurs only if the RNA contains an intron. A single point mutation in the splice donor site prevents translation. Intron-less RNA is exported efficiently to the cytoplasm and is held, undegraded, in a translationally inert state for several days. Translation can be activated by treating the oocytes with progesterone or by injecting antibodies that bind the FRGY2 class of messenger RNA binding proteins, p56 and p60, but these antibodies are only effective if delivered to the nucleus. Inhibitors of casein kinase II also activate translation whereas phosphatase inhibitors block progesterone-mediated activation of translation. These data suggest the presence of an RNA handling pathway in the nucleus of Xenopus oocytes which is regulated by casein kinase type II phosphorylation and which directs transcripts to be sequestered by p56/p60 or by closely related proteins. This pathway can be bypassed if the RNA contains an intron and it can be reversed by progesterone treatment. These data may have implications for understanding translational control during early development.


Subject(s)
Introns/physiology , Protein Biosynthesis/physiology , RNA, Messenger/metabolism , Xenopus Proteins , Animals , Casein Kinase II , Cell Nucleus/metabolism , Cytoplasm/metabolism , DNA/metabolism , Ethers, Cyclic/pharmacology , Female , Gene Expression Regulation, Developmental/physiology , Microinjections , Okadaic Acid , Oocytes/metabolism , Phosphoprotein Phosphatases/antagonists & inhibitors , Phosphorylation/drug effects , Point Mutation/physiology , Progesterone/pharmacology , Protein Biosynthesis/drug effects , Protein Serine-Threonine Kinases/antagonists & inhibitors , RNA, Messenger/chemical synthesis , RNA, Messenger/genetics , RNA-Binding Proteins/physiology , Recombinant Fusion Proteins/biosynthesis , Transcription Factors/physiology , Xenopus laevis
19.
Nucleic Acids Res ; 22(16): 3365-72, 1994 Aug 25.
Article in English | MEDLINE | ID: mdl-8078772

ABSTRACT

The Tat protein binds to TAR RNA to stimulate the expression of the human immunodeficiency virus type 1 (HIV-1) genome. Tat is an 86 amino acid protein that contains a short region of basic residues (aa49-aa57) that are required for RNA binding and TAR is a 59 nucleotide stem-loop with a tripyrimidine bulge in the upper stem. TAR is located at the 5' end of all viral RNAs. In vitro, Tat specifically interacts with TAR by recognising the sequence of the bulge and upper stem, with no requirement for the loop. However, in vivo the loop sequence is critical for activation, implying a requirement for accessory cellular TAR RNA binding factors. A number of TAR binding cellular factors have been identified in cell extracts and various models for the function of these factors have been suggested, including roles as coactivators and inhibitors. We have now identified a novel 38 kD cellular factor that has little general, single-stranded or double-stranded RNA binding activity, but that specifically recognises the bulge and upper stem region of TAR. The protein, referred to as BBP (bulge binding protein), is conserved in mammalian and amphibian cells and in Schizosaccharomyces pombe but is not found in Saccharomyces cerevisiae. BBP is an effective competitive inhibitor of Tat binding to TAR in vitro. Our data suggest that the bulge-stem recognition motif in TAR is used to mediate cellular factor/RNA interactions and indicates that Tat action might be inhibited by such competing reactions in vivo.


Subject(s)
Gene Products, tat/metabolism , HIV-1/genetics , RNA, Viral/metabolism , RNA-Binding Proteins/analysis , Animals , Base Sequence , Binding Sites , Binding, Competitive , CHO Cells , Cell Nucleus/chemistry , Cricetinae , Factor Xa/metabolism , HeLa Cells , Humans , Molecular Sequence Data , Nucleic Acid Conformation , Peptide Fragments/metabolism , RNA, Viral/chemistry , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism , Species Specificity , Structure-Activity Relationship , tat Gene Products, Human Immunodeficiency Virus
20.
J Virol ; 68(1): 25-33, 1994 Jan.
Article in English | MEDLINE | ID: mdl-8254735

ABSTRACT

Two benzodiazepine compounds, [7-chloro-5-(2-pyrryl)-3H-1,4 benzodiazapin-2-(H)-one] (Ro5-3335) and [7-chloro-5-(1H-pyrrol-2-yl)-3H-benzo[e] [1,4] diazepin-2-yl]- methylamine (Ro24-7429), inhibit human immunodeficiency virus type 1 (HIV-1) replication via a specific effect on the function of the transactivator protein, Tat. To gain further insight into the mechanism of action of these compounds, we have tested their effects in an alternative assay for Tat activation in Xenopus oocytes. In this system, translation of trans-activation response element (TAR)-containing RNA is activated by Tat. Both compounds specifically blocked activation of translation in a dose-dependent fashion, with Ro24-7429 showing the greater potency. In the Xenopus oocyte system, as in mammalian cells, mutation of the TAR loop sequences abolishes Tat action. However, it is possible to obtain TAR-specific, Tat-dependent activation of a target RNA with a mutation in the loop provided that this target is in large excess. This result has been interpreted as indicating that a negative factor has been titrated (M. Braddock, R. Powell, A.D. Blanchard, A.J. Kingsman, and S.M. Kingsman, FASEB J. 7:214-222, 1993). Interestingly Ro24-7429 was unable to inhibit the TAR-specific but loop sequence-independent mode of translational activation. This finding suggests that a specific loop-binding cellular factor may mediate the effects of this inhibitor of Tat action. Consistent with this notion, we could not detect any effect of Ro24-7429 on the efficiency of specific Tat binding to TAR in vitro.


Subject(s)
Antiviral Agents/pharmacology , Benzodiazepines/pharmacology , Gene Expression Regulation, Viral/drug effects , Gene Products, tat/antagonists & inhibitors , HIV-1/drug effects , Protein Biosynthesis/drug effects , RNA, Viral/genetics , Animals , Base Sequence , Benzodiazepinones/pharmacology , Gene Products, tat/metabolism , Humans , Models, Genetic , Molecular Sequence Data , Nucleic Acid Conformation , Oocytes , Pyrroles/pharmacology , RNA, Viral/metabolism , Regulatory Sequences, Nucleic Acid , Transcriptional Activation , Tumor Cells, Cultured , Virus Replication/drug effects , Xenopus , tat Gene Products, Human Immunodeficiency Virus
SELECTION OF CITATIONS
SEARCH DETAIL
...