Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Free Radic Biol Med ; 204: 243-251, 2023 08 01.
Article in English | MEDLINE | ID: mdl-37179033

ABSTRACT

The redox sensitive transcription factor NRF2 is a central regulator of the transcriptional response to reactive oxygen species (ROS). NRF2 is widely recognized for its ROS-responsive upregulation of antioxidant genes that are essential for mitigating the damaging effects of oxidative stress. However, multiple genome-wide approaches have suggested that NRF2's regulatory reach extends well beyond the canonical antioxidant genes, with the potential to regulate many noncanonical target genes. Recent work from our lab and others suggests HIF1A, which encodes the hypoxia-responsive transcription factor HIF1α, is one such noncanonical NRF2 target. These studies found that NRF2 activity is associated with high HIF1A expression in multiple cellular contexts, HIF1A expression is partially dependent on NRF2, and there is a putative NRF2 binding site (antioxidant response element, or ARE) approximately 30 kilobases upstream of HIF1A. These findings all support a model in which HIF1A is a direct target of NRF2, but did not confirm the functional importance of the upstream ARE in HIF1A expression. Here we use CRISPR/Cas9 genome editing to mutate this ARE in its genomic context and test the impact on HIF1A expression. We find that mutation of this ARE in a breast cancer cell line (MDA-MB-231) eliminates NRF2 binding and decreases HIF1A expression at the transcript and protein levels, and disrupts HIF1α target genes as well as phenotypes driven by these HIF1α targets. Taken together, these results indicate that this NRF2 targeted ARE plays an important role in the expression of HIF1A and activity of the HIF1α axis in MDA-MB-231 cells.


Subject(s)
Antioxidant Response Elements , Gene Expression Regulation, Neoplastic , Hypoxia-Inducible Factor 1, alpha Subunit , Humans , Cell Line, Tumor , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Mutation , NF-E2-Related Factor 2/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism
2.
Neuron ; 111(4): 493-507.e6, 2023 02 15.
Article in English | MEDLINE | ID: mdl-36577403

ABSTRACT

Spinocerebellar ataxia type 1 (SCA1) is a dominant trinucleotide repeat neurodegenerative disease characterized by motor dysfunction, cognitive impairment, and premature death. Degeneration of cerebellar Purkinje cells is a frequent and prominent pathological feature of SCA1. We previously showed that transport of ATXN1 to Purkinje cell nuclei is required for pathology, where mutant ATXN1 alters transcription. To examine the role of ATXN1 nuclear localization broadly in SCA1-like disease pathogenesis, CRISPR-Cas9 was used to develop a mouse with an amino acid alteration (K772T) in the nuclear localization sequence of the expanded ATXN1 protein. Characterization of these mice indicates that proper nuclear localization of mutant ATXN1 contributes to many disease-like phenotypes including motor dysfunction, cognitive deficits, and premature lethality. RNA sequencing analysis of genes with expression corrected to WT levels in Atxn1175QK772T/2Q mice indicates that transcriptomic aspects of SCA1 pathogenesis differ between the cerebellum, brainstem, cerebral cortex, hippocampus, and striatum.


Subject(s)
Ataxin-1 , Spinocerebellar Ataxias , Transcriptome , Animals , Mice , Ataxin-1/genetics , Ataxin-1/metabolism , Brain/metabolism , Cerebellum/metabolism , Disease Models, Animal , Mice, Transgenic , Nerve Tissue Proteins/genetics , Phenotype , Protein Transport/genetics , Purkinje Cells/metabolism , Spinocerebellar Ataxias/genetics , Spinocerebellar Ataxias/metabolism
3.
Sci Adv ; 8(16): eabj5227, 2022 Apr 22.
Article in English | MEDLINE | ID: mdl-35452291

ABSTRACT

Here, we report that the LynB splice variant of the Src-family kinase Lyn exerts a dominant immunosuppressive function in vivo, whereas the LynA isoform is uniquely required to restrain autoimmunity in female mice. We used CRISPR-Cas9 gene editing to constrain lyn splicing and expression, generating single-isoform LynA knockout (LynAKO) or LynBKO mice. Autoimmune disease in total LynKO mice is characterized by production of antinuclear antibodies, glomerulonephritis, impaired B cell development, and overabundance of activated B cells and proinflammatory myeloid cells. Expression of LynA or LynB alone uncoupled the developmental phenotype from the autoimmune disease: B cell transitional populations were restored, but myeloid cells and differentiated B cells were dysregulated. These changes were isoform-specific, sexually dimorphic, and distinct from the complete LynKO. Despite the apparent differences in disease etiology and penetrance, loss of either LynA or LynB had the potential to induce severe autoimmune disease with parallels to human systemic lupus erythematosus (SLE).

4.
DNA Repair (Amst) ; 85: 102738, 2020 01.
Article in English | MEDLINE | ID: mdl-31731258

ABSTRACT

The repair of DNA double-stranded breaks (DSBs) is an essential function performed by the Classical Non-Homologous End-Joining (C-NHEJ) pathway in higher eukaryotes. C-NHEJ, in fact, does double duty as it is also required for the repair of the intermediates formed during lymphoid B- and T-cell recombination. Consequently, the failure to properly repair DSBs leads to both genomic instability and immunodeficiency. A critical DSB protein required for C-NHEJ is the DNA Ligase IV (LIGIV) accessory factor, X-Ray Cross Complementing 4 (XRCC4). XRCC4 is believed to stabilize LIGIV, participate in LIGIV activation, and to help tether the broken DSB ends together. XRCC4's role in these processes has been muddied by the identification of two additional XRCC4 paralogs, XRCC4-Like Factor (XLF), and Paralog of XRCC4 and XLF (PAXX). The roles that these paralogs play in C-NHEJ is partially understood, but, in turn, has itself been obscured by species-specific differences observed in the absence of one or the other paralogs. In order to investigate the role(s) that XRCC4 may play, with or without XLF and/or PAXX, in lymphoid variable(diversity)joining [V(D)J] recombination as well as in DNA DSB repair in human somatic cells, we utilized gene targeting to inactivate the XRCC4 gene in both parental and XLF- HCT116 cells and then inactivated PAXX in those same cell lines. The loss of XRCC4 expression by itself led, as anticipated, to increased sensitivity to DNA damaging agents as well as an increased dependence on microhomology-mediated DNA repair whether in the context of DSB repair or during V(D)J recombination. The additional loss of XLF in these cell lines sensitized the cells even more whereas the presence or absence of PAXX was scarcely negligible. These studies demonstrate that, of the three LIG4 accessory factor paralogs, the absence of XRCC4 influences DNA repair and recombination the most in human cells.


Subject(s)
DNA Repair Enzymes/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Etoposide/adverse effects , DNA Breaks, Double-Stranded , DNA End-Joining Repair , DNA Ligase ATP/metabolism , Gene Targeting , HCT116 Cells , Humans , V(D)J Recombination
5.
Nucleic Acids Res ; 47(5): 2402-2424, 2019 03 18.
Article in English | MEDLINE | ID: mdl-30590694

ABSTRACT

Fusion of critically short or damaged telomeres is associated with the genomic rearrangements that support malignant transformation. We have demonstrated the fundamental contribution of DNA ligase 4-dependent classical non-homologous end-joining to long-range inter-chromosomal telomere fusions. In contrast, localized genomic recombinations initiated by sister chromatid fusion are predominantly mediated by alternative non-homologous end-joining activity that may employ either DNA ligase 3 or DNA ligase 1. In this study, we sought to discriminate the relative involvement of these ligases in sister chromatid telomere fusion through a precise genetic dissociation of functional activity. We have resolved an essential and non-redundant role for DNA ligase 1 in the fusion of sister chromatids bearing targeted double strand DNA breaks that is entirely uncoupled from its requisite engagement in DNA replication. Importantly, this fusogenic repair occurs in cells fully proficient for non-homologous end-joining and is not compensated by DNA ligases 3 or 4. The dual functions of DNA ligase 1 in replication and non-homologous end-joining uniquely position and capacitate this ligase for DNA repair at stalled replication forks, facilitating mitotic progression.


Subject(s)
Chromatids/genetics , DNA End-Joining Repair/genetics , DNA Ligase ATP/genetics , Mitosis/genetics , DNA Breaks, Double-Stranded , DNA Repair/genetics , DNA Replication/genetics , G2 Phase Cell Cycle Checkpoints/genetics , HCT116 Cells , Humans , Poly-ADP-Ribose Binding Proteins/genetics , Sister Chromatid Exchange/genetics , Telomere/genetics
6.
Nucleic Acids Res ; 46(17): 8926-8939, 2018 09 28.
Article in English | MEDLINE | ID: mdl-30113698

ABSTRACT

The Artemis nuclease and tyrosyl-DNA phosphodiesterase (TDP1) are each capable of resolving protruding 3'-phosphoglycolate (PG) termini of DNA double-strand breaks (DSBs). Consequently, both a knockout of Artemis and a knockout/knockdown of TDP1 rendered cells sensitive to the radiomimetic agent neocarzinostatin (NCS), which induces 3'-PG-terminated DSBs. Unexpectedly, however, a knockdown or knockout of TDP1 in Artemis-null cells did not confer any greater sensitivity than either deficiency alone, indicating a strict epistasis between TDP1 and Artemis. Moreover, a deficiency in Artemis, but not TDP1, resulted in a fraction of unrepaired DSBs, which were assessed as 53BP1 foci. Conversely, a deficiency in TDP1, but not Artemis, resulted in a dramatic increase in dicentric chromosomes following NCS treatment. An inhibitor of DNA-dependent protein kinase, a key regulator of the classical nonhomologous end joining (C-NHEJ) pathway sensitized cells to NCS, but eliminated the sensitizing effects of both TDP1 and Artemis deficiencies. These results suggest that TDP1 and Artemis perform different functions in the repair of terminally blocked DSBs by the C-NHEJ pathway, and that whereas an Artemis deficiency prevents end joining of some DSBs, a TDP1 deficiency tends to promote DSB mis-joining.


Subject(s)
DNA End-Joining Repair , DNA/genetics , Endonucleases/genetics , Epistasis, Genetic , Nuclear Proteins/genetics , Phosphoric Diester Hydrolases/genetics , Cell Survival/drug effects , Cytotoxins/pharmacology , DNA/chemistry , DNA/metabolism , DNA Breaks, Double-Stranded , DNA-Binding Proteins , Endonucleases/antagonists & inhibitors , Endonucleases/deficiency , HCT116 Cells , HEK293 Cells , Humans , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/deficiency , Nucleic Acid Synthesis Inhibitors/pharmacology , Phosphoric Diester Hydrolases/deficiency , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Tumor Suppressor p53-Binding Protein 1/genetics , Tumor Suppressor p53-Binding Protein 1/metabolism , Zinostatin/pharmacology
7.
Genome Res ; 27(7): 1099-1111, 2017 07.
Article in English | MEDLINE | ID: mdl-28356322

ABSTRACT

The use of programmable meganucleases is transforming genome editing and functional genomics. CRISPR/Cas9 was developed such that targeted genomic lesions could be introduced in vivo with unprecedented ease. In the presence of homology donors, these lesions facilitate high-efficiency precise genome editing (PGE) via homology-directed repair (HDR) pathways. However, the identity and hierarchy of the HDR (sub)pathways leading to the formation of PGE products remain elusive. Here, we established a green to blue fluorescent protein conversion system to systematically characterize oligodeoxynucleotide (ODN)-mediated PGE using Cas9 and its nickase variants in human cells. We demonstrate that, unlike double-stranded DNA (dsDNA) donors with central heterologies, ODNs generated short conversion tracts with Gaussian-like distributions. Interestingly, single-nick-induced PGE using ODN donors produced conversion tracts biased either mostly uni- or bidirectional depending on the relative strandedness of the ODNs and the nick. Moreover, the ODNs were physically incorporated into the genome only in the bidirectional, but not in the unidirectional, conversion pathway. In the presence of double-stranded genomic lesions, the unidirectional conversion pathway was preferentially utilized even though the knock-in mutation could theoretically have been converted by both pathways. Collectively, our results suggest that ODN-mediated PGE utilizes synthesis-dependent strand annealing and single-stranded DNA incorporation pathways. Both of these pathways generate short conversion tracts with Gaussian-like distributions. Although synthesis-dependent strand annealing is preferentially utilized, our work unequivocally establishes the existence of a single-stranded DNA incorporation pathway in human cells. This work extends the paradigms of HDR-mediated gene conversion and establishes guidelines for PGE in human cells.


Subject(s)
CRISPR-Cas Systems , DNA , Gene Editing/methods , Oligodeoxyribonucleotides/chemistry , Cell Line , DNA/genetics , DNA/metabolism , Humans , Oligodeoxyribonucleotides/genetics
8.
Genome Res ; 26(5): 588-600, 2016 05.
Article in English | MEDLINE | ID: mdl-26941250

ABSTRACT

Telomeres shorten with each cell division and can ultimately become substrates for nonhomologous end-joining repair, leading to large-scale genomic rearrangements of the kind frequently observed in human cancers. We have characterized more than 1400 telomere fusion events at the single-molecule level, using a combination of high-throughput sequence analysis together with experimentally induced telomeric double-stranded DNA breaks. We show that a single chromosomal dysfunctional telomere can fuse with diverse nontelomeric genomic loci, even in the presence of an otherwise stable genome, and that fusion predominates in coding regions. Fusion frequency was markedly increased in the absence of TP53 checkpoint control and significantly modulated by the cellular capacity for classical, versus alternative, nonhomologous end joining (NHEJ). We observed a striking reduction in inter-chromosomal fusion events in cells lacking DNA ligase 4, in contrast to a remarkably consistent profile of intra-chromosomal fusion in the context of multiple genetic knockouts, including DNA ligase 3 and 4 double-knockouts. We reveal distinct mutational signatures associated with classical NHEJ-mediated inter-chromosomal, as opposed to alternative NHEJ-mediated intra-chromosomal, telomere fusions and evidence for an unanticipated sufficiency of DNA ligase 1 for these intra-chromosomal events. Our findings have implications for mechanisms driving cancer genome evolution.


Subject(s)
Chromatids , Chromosomes, Human , DNA Breaks, Double-Stranded , DNA End-Joining Repair , DNA Ligase ATP , Neoplasms , Telomere , Cell Line, Tumor , Chromatids/genetics , Chromatids/metabolism , Chromosomes, Human/genetics , Chromosomes, Human/metabolism , Gene Deletion , Humans , Neoplasms/genetics , Neoplasms/metabolism , Telomere/genetics , Telomere/metabolism , Tumor Suppressor Protein p53
9.
Mol Cell ; 55(6): 829-842, 2014 Sep 18.
Article in English | MEDLINE | ID: mdl-25201414

ABSTRACT

Breakpoint junctions of the chromosomal translocations that occur in human cancers display hallmarks of nonhomologous end-joining (NHEJ). In mouse cells, translocations are suppressed by canonical NHEJ (c-NHEJ) components, which include DNA ligase IV (LIG4), and instead arise from alternative NHEJ (alt-NHEJ). Here we used designer nucleases (ZFNs, TALENs, and CRISPR/Cas9) to introduce DSBs on two chromosomes to study translocation joining mechanisms in human cells. Remarkably, translocations were altered in cells deficient for LIG4 or its interacting protein XRCC4. Translocation junctions had significantly longer deletions and more microhomology, indicative of alt-NHEJ. Thus, unlike mouse cells, translocations in human cells are generated by c-NHEJ. Human cancer translocations induced by paired Cas9 nicks also showed a dependence on c-NHEJ, despite having distinct joining characteristics. These results demonstrate an unexpected and striking species-specific difference for common genomic rearrangements associated with tumorigenesis.


Subject(s)
DNA End-Joining Repair , DNA Ligases/genetics , DNA-Binding Proteins/genetics , Deoxyribonucleases/physiology , Translocation, Genetic/genetics , Animals , Chromosomes, Human , DNA Ligase ATP , Humans , Mice , Sequence Deletion , Species Specificity , Tumor Cells, Cultured
10.
PLoS Genet ; 10(4): e1004251, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24699519

ABSTRACT

Gene targeting in human somatic cells is of importance because it can be used to either delineate the loss-of-function phenotype of a gene or correct a mutated gene back to wild-type. Both of these outcomes require a form of DNA double-strand break (DSB) repair known as homologous recombination (HR). The mechanism of HR leading to gene targeting, however, is not well understood in human cells. Here, we demonstrate that a two-end, ends-out HR intermediate is valid for human gene targeting. Furthermore, the resolution step of this intermediate occurs via the classic DSB repair model of HR while synthesis-dependent strand annealing and Holliday Junction dissolution are, at best, minor pathways. Moreover, and in contrast to other systems, the positions of Holliday Junction resolution are evenly distributed along the homology arms of the targeting vector. Most unexpectedly, we demonstrate that when a meganuclease is used to introduce a chromosomal DSB to augment gene targeting, the mechanism of gene targeting is inverted to an ends-in process. Finally, we demonstrate that the anti-recombination activity of mismatch repair is a significant impediment to gene targeting. These observations significantly advance our understanding of HR and gene targeting in human cells.


Subject(s)
DNA Repair/genetics , DNA/genetics , Recombination, Genetic/genetics , Cell Line, Tumor , DNA Breaks, Double-Stranded , DNA, Cruciform/genetics , Gene Targeting/methods , Genetic Vectors/genetics , HCT116 Cells , Humans
11.
Mol Cell Biol ; 28(20): 6182-95, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18710952

ABSTRACT

The DNA-dependent protein kinase (DNA-PK) complex is a serine/threonine protein kinase comprised of a 469-kDa catalytic subunit (DNA-PK(cs)) and the DNA binding regulatory heterodimeric (Ku70/Ku86) complex Ku. DNA-PK functions in the nonhomologous end-joining pathway for the repair of DNA double-stranded breaks (DSBs) introduced by either exogenous DNA damage or endogenous processes, such as lymphoid V(D)J recombination. Not surprisingly, mutations in Ku70, Ku86, or DNA-PK(cs) result in animals that are sensitive to agents that cause DSBs and that are also immune deficient. While these phenotypes have been validated in several model systems, an extension of them to humans has been missing due to the lack of patients with mutations in any one of the three DNA-PK subunits. The worldwide lack of patients suggests that during mammalian evolution this complex has become uniquely essential in primates. This hypothesis was substantiated by the demonstration that functional inactivation of either Ku70 or Ku86 in human somatic cell lines is lethal. Here we report on the functional inactivation of DNA-PK(cs) in human somatic cells. Surprisingly, DNA-PK(cs) does not appear to be essential, although the cell line lacking this gene has profound proliferation and genomic stability deficits not observed for other mammalian systems.


Subject(s)
Catalytic Domain , DNA-Activated Protein Kinase/metabolism , Genomic Instability , Telomere/metabolism , Biomarkers/metabolism , Cell Cycle/drug effects , Cell Proliferation/drug effects , DNA Damage , DNA-Activated Protein Kinase/deficiency , Etoposide/pharmacology , Gene Targeting , Genomic Instability/drug effects , HCT116 Cells , Heterozygote , Homozygote , Humans
12.
DNA Repair (Amst) ; 7(5): 762-74, 2008 May 03.
Article in English | MEDLINE | ID: mdl-18387344

ABSTRACT

NHEJ (non-homologous end joining) is the predominant mechanism for repairing DNA double-stranded breaks in human cells. One essential NHEJ factor is the Ku heterodimer, which is composed of Ku70 and Ku86. Here we have generated heterozygous loss-of-function mutations for each of these genes in two different human somatic cell lines, HCT116 and NALM-6, using gene targeting. Previous work had suggested that phenotypic differences might exist between the genes and/or between the cell lines. By providing a side-by-each comparison of the four cell lines, we demonstrate that there are indeed subtle differences between loss-of-function mutations for Ku70 versus Ku86, which is accentuated by whether the mutations were derived in the HCT116 or NALM-6 genetic background. Overall, however, the phenotypes of the four lines are quite similar and they provide a compelling argument for the hypothesis that Ku loss-of-function mutations in human somatic cells result in demonstrable haploinsufficiencies. Collectively, these studies demonstrate the importance of proper biallelic expression of these genes for NHEJ and telomere maintenance and they provide insights into why these genes are uniquely essential for primates.


Subject(s)
Antigens, Nuclear/genetics , DNA Repair/genetics , DNA-Binding Proteins/genetics , Mutation/genetics , Cell Line, Tumor , Cell Proliferation , DNA Damage/genetics , Genomic Instability/genetics , HCT116 Cells , Humans , Ku Autoantigen , Radiation Tolerance , Radiation, Ionizing , Telomere/genetics
13.
J Biol Chem ; 283(1): 398-404, 2008 Jan 04.
Article in English | MEDLINE | ID: mdl-17965019

ABSTRACT

Protein phosphatase I (PP1) is an essential eukaryotic serine/threonine phosphatase required for many cellular processes, including cell division, signaling, and metabolism. In mammalian cells there are three major isoforms of the PP1 catalytic subunit (PP1alpha, PP1beta, and PP1gamma) that are over 90% identical. Despite this high degree of identity, the PP1 catalytic subunits show distinct localization patterns in interphase cells; PP1alpha is primarily nuclear and largely excluded from nucleoli, whereas PP1gamma and to a lesser extent PP1beta concentrate in the nucleoli. The subcellular localization and the substrate specificity of PP1 catalytic subunits are determined by their interaction with targeting subunits, most of which bind PP1 through a so-called "RVXF" sequence. Although PP1 targeting subunits have been identified that direct PP1 to a number of subcellular locations and/or substrates, no targeting subunit has been identified that localizes PP1 to the nucleolus. Identification of nucleolar PP1 targeting subunit(s) is important because all three PP1 isoforms are included in the nucleolar proteome, enzymatically active PP1 is present in nucleoli, and PP1gamma is highly concentrated in nucleoli of interphase cells. In this study, we identify NOM1 (nucleolar protein with MIF4G domain 1) as a PP1-interacting protein and further identify the NOM1 RVXF motif required for its binding to PP1. We also define the NOM1 nucleolar localization sequence. Finally, we demonstrate that NOM1 can target PP1 to the nucleolus and show that a specific NOM1 RVXF motif and the NOM1 nucleolar localization sequence are required for this targeting activity. We therefore conclude that NOM1 is a PP1 nucleolar targeting subunit, the first identified in eukaryotic cells.


Subject(s)
Cell Nucleolus/metabolism , Nuclear Proteins/metabolism , Protein Phosphatase 1/metabolism , RNA-Binding Proteins/metabolism , Animals , Binding Sites/genetics , Blotting, Western , Cell Line , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Immunoprecipitation , Mice , Microscopy, Confocal , Mutation , NIH 3T3 Cells , Nuclear Proteins/genetics , Protein Binding , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Phosphatase 1/genetics , RNA-Binding Proteins/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Two-Hybrid System Techniques
14.
Gene ; 347(1): 137-45, 2005 Feb 28.
Article in English | MEDLINE | ID: mdl-15715967

ABSTRACT

Proteins that contain the recently described MIF4G and/or MA3 domains function in translation, cell growth, proliferation, transformation, and apoptosis. Examples of MIF4G/MA3 containing proteins and their functions include eIF4G, which serves as a scaffold for assembly of factors required for translation initiation, programmed cell death protein 4 (Pdcd4) that inhibits translation and functions as a tumor suppressor, and NMD2, which is essential for nonsense-mediated mRNA decay. MIF4G and MA3 domains serve as binding sites for one or more isoforms of the eIF4A family of ATP-dependent DEAD-box RNA helicases that are required for translation and for nonsense-mediated decay. In this report, we describe the characterization of a novel MIF4G/MA3 family member called NOM1 (nucleolar protein with MIF4G domain 1) that was identified at the chromosome 7q36 breakpoint involved in 7;12 translocations associated with certain acute leukemias of childhood. NOM1, which includes a previously described EST called c7orf3, encodes a ubiquitously expressed transcript composed of 11 exons and an approximately 3 kb 3' UTR that contains several Alu repeats. The predicted NOM1 protein contains one MIF4G domain and one MA3 domain and, consistent with data obtained with other MIF4G/MA3 proteins, interacts with members of the eIF4A family of helicases. Database searches reveal that NOM1 homologs exist in several organisms and that at least two of these are essential genes. Finally, like its Saccharomyces cerevisiae homolog Sgd1p, NOM1 localizes predominantly to the nucleolus. These data demonstrate that NOM1 is a new member of the MIF4G/MA3 family of proteins and suggest that it may provide an essential function in metazoans.


Subject(s)
Chromosomes, Human, Pair 7/genetics , Eukaryotic Initiation Factor-4A/metabolism , Exons/genetics , Leukemia, Myeloid, Acute/genetics , Nuclear Proteins/genetics , RNA-Binding Proteins/genetics , Alu Elements/genetics , Alu Elements/physiology , Apoptosis/genetics , Apoptosis/physiology , Cell Nucleus/metabolism , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Chromosome Breakage/genetics , Codon, Nonsense/genetics , Codon, Nonsense/metabolism , Eukaryotic Initiation Factor-4A/genetics , Exons/physiology , Gene Expression Regulation, Leukemic/genetics , Gene Expression Regulation, Leukemic/physiology , Humans , Leukemia, Myeloid, Acute/metabolism , Nuclear Proteins/metabolism , Protein Binding/genetics , Protein Binding/physiology , RNA Stability/genetics , RNA Stability/physiology , RNA-Binding Proteins/metabolism , Transcription, Genetic/genetics , Transcription, Genetic/physiology , Translocation, Genetic/genetics , Translocation, Genetic/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...