Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
J Biol Chem ; 287(5): 3156-64, 2012 Jan 27.
Article in English | MEDLINE | ID: mdl-22167198

ABSTRACT

The highly conserved DYNLL1 (LC8) protein was originally discovered as a light chain of the dynein motor complex, but is increasingly emerging as a sequence-specific regulator of protein dimerization with hundreds of targets and wide-ranging cellular functions. Despite its important roles, DYNLL1's own regulation remains poorly understood. Here we identify ASCIZ (ATMIN/ZNF822), an essential Zn(2+) finger protein with dual roles in the DNA base damage response and as a developmental transcription factor, as a conserved regulator of Dynll1 gene expression. DYNLL1 levels are reduced by ∼10-fold in the absence of ASCIZ in human, mouse and chicken cells. ASCIZ binds directly to the Dynll1 promoter and regulates its activity in a Zn(2+) finger-dependent manner. DYNLL1 protein in turn interacts with ten binding sites in the ASCIZ transcription activation domain, and high DYNLL1 levels inhibit the transcriptional activity of ASCIZ. In addition, DYNLL1 was also required for DNA damage-induced ASCIZ focus formation. The dual ability of ASCIZ to activate Dynll1 gene expression and to sense free DYNLL1 protein levels enables a simple dynamic feedback loop to adjust DYNLL1 levels to cellular needs. The ASCIZ-DYNLL1 feedback loop represents a novel mechanism for auto-regulation of gene expression, where the gene product directly inhibits the transcriptional activator while bound at its own promoter.


Subject(s)
Carrier Proteins/metabolism , Cytoplasmic Dyneins/biosynthesis , Gene Expression Regulation, Enzymologic/physiology , Nuclear Proteins/metabolism , Promoter Regions, Genetic/physiology , Zinc/metabolism , Animals , Binding Sites , Carrier Proteins/genetics , Cell Line , Chickens , Cytoplasmic Dyneins/genetics , Humans , Mice , Nuclear Proteins/genetics , Transcription Factors , Transcription, Genetic/physiology , Zinc Fingers
2.
PLoS Genet ; 6(10): e1001170, 2010 Oct 21.
Article in English | MEDLINE | ID: mdl-20975950

ABSTRACT

Zn²(+)-finger proteins comprise one of the largest protein superfamilies with diverse biological functions. The ATM substrate Chk2-interacting Zn²(+)-finger protein (ASCIZ; also known as ATMIN and ZNF822) was originally linked to functions in the DNA base damage response and has also been proposed to be an essential cofactor of the ATM kinase. Here we show that absence of ASCIZ leads to p53-independent late-embryonic lethality in mice. Asciz-deficient primary fibroblasts exhibit increased sensitivity to DNA base damaging agents MMS and H2O2, but Asciz deletion knock-down does not affect ATM levels and activation in mouse, chicken, or human cells. Unexpectedly, Asciz-deficient embryos also exhibit severe respiratory tract defects with complete pulmonary agenesis and severe tracheal atresia. Nkx2.1-expressing respiratory precursors are still specified in the absence of ASCIZ, but fail to segregate properly within the ventral foregut, and as a consequence lung buds never form and separation of the trachea from the oesophagus stalls early. Comparison of phenotypes suggests that ASCIZ functions between Wnt2-2b/ß-catenin and FGF10/FGF-receptor 2b signaling pathways in the mesodermal/endodermal crosstalk regulating early respiratory development. We also find that ASCIZ can activate expression of reporter genes via its SQ/TQ-cluster domain in vitro, suggesting that it may exert its developmental functions as a transcription factor. Altogether, the data indicate that, in addition to its role in the DNA base damage response, ASCIZ has separate developmental functions as an essential regulator of respiratory organogenesis.


Subject(s)
Carrier Proteins/physiology , DNA Repair/physiology , Lung/embryology , Nuclear Proteins/physiology , Organogenesis/physiology , Animals , Blotting, Western , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line , Cell Survival/drug effects , Cell Survival/radiation effects , Cells, Cultured , Cellular Senescence , DNA Damage , Embryo, Mammalian/abnormalities , Embryo, Mammalian/metabolism , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Genotype , Humans , Hydrogen Peroxide/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oxidants/pharmacology , Time Factors , Trachea/embryology , Transcription Factors , Ultraviolet Rays
3.
Mol Cell Biol ; 22(8): 2821-9, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11909974

ABSTRACT

Ca(2+) signaling plays a central role in cardiac contractility and adaptation to increased hemodynamic demand. We have generated mice with a targeted deletion of the S100A1 gene coding for the major cardiac isoform of the large multigenic S100 family of EF hand Ca(2+)-binding proteins. S100A1(-/-) mice have normal cardiac function under baseline conditions but have significantly reduced contraction rate and relaxation rate responses to beta-adrenergic stimulation that are associated with a reduced Ca(2+) sensitivity. In S100A1(-/-) mice, basal left-ventricular contractility deteriorated following 3-week pressure overload by thoracic aorta constriction despite a normal adaptive hypertrophy. Surprisingly, heterozygotes also had an impaired response to acute beta-adrenergic stimulation but maintained normal contractility in response to chronic pressure overload that coincided with S100A1 upregulation to wild-type levels. In contrast to other genetic models with impaired cardiac contractility, loss of S100A1 did not lead to cardiac hypertrophy or dilation in aged mice. The data demonstrate that high S100A1 protein levels are essential for the cardiac reserve and adaptation to acute and chronic hemodynamic stress in vivo.


Subject(s)
Calcium-Binding Proteins/deficiency , Calcium-Binding Proteins/genetics , Myocardial Contraction/physiology , Adrenergic beta-Agonists/pharmacology , Aging/physiology , Animals , Calcium Signaling/genetics , Calcium Signaling/physiology , Calcium-Binding Proteins/physiology , Gene Targeting , Heart Ventricles/drug effects , Hemodynamics , Heterozygote , Isoproterenol/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardial Contraction/drug effects , Myocardial Contraction/genetics , Receptors, Adrenergic, beta/physiology , S100 Proteins , Stress, Physiological/physiopathology , Ventricular Function
4.
Genetics ; 169(1): 65-75, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15466434

ABSTRACT

DNA damage checkpoints regulate gene expression at the transcriptional and post-transcriptional level. Some components of the yeast Ccr4-Not complex, which regulates transcription as well as transcript turnover, have previously been linked to DNA damage responses, but it is unclear if this involves transcriptional or post-transcriptional functions. Here we show that CCR4 and CAF1, which together encode the major cytoplasmic mRNA deadenylase complex, have complex genetic interactions with the checkpoint genes DUN1, MRC1, RAD9, and RAD17 in response to DNA-damaging agents hydroxyurea (HU) and methylmethane sulfonate (MMS). The exonuclease-inactivating ccr4-1 point mutation mimics ccr4Delta phenotypes, including synthetic HU hypersensitivity with dun1Delta, demonstrating that Ccr4-Not mRNA deadenylase activity is required for DNA damage responses. However, ccr4Delta and caf1Delta DNA damage phenotypes and genetic interactions with checkpoint genes are not identical, and deletions of some Not components that are believed to predominantly function at the transcriptional level rather than mRNA turnover, e.g., not5Delta, also lead to increased DNA damage sensitivity and synthetic HU hypersensitivity with dun1Delta. Taken together, our data thus suggest that both transcriptional and post-transcriptional functions of the Ccr4-Not complex contribute to the DNA damage response affecting gene expression in a complex manner.


Subject(s)
DNA Damage , Ribonucleases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Hydroxyurea/pharmacology , Methyl Methanesulfonate/pharmacology , Mutagens/pharmacology , Nucleic Acid Synthesis Inhibitors/pharmacology , Phenotype , Point Mutation , Ribonucleases/genetics , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Transcription, Genetic
5.
Genetics ; 196(2): 443-53, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24336747

ABSTRACT

The essential zinc finger protein ASCIZ (also known as ATMIN, ZNF822) plays critical roles during lung organogenesis and B cell development in mice, where it regulates the expression of dynein light chain (DYNLL1/LC8), but its functions in other species including invertebrates are largely unknown. Here we report the identification of the Drosophila ortholog of ASCIZ (dASCIZ) and show that loss of dASCIZ function leads to pronounced mitotic delays with centrosome and spindle positioning defects during development, reminiscent of impaired dynein motor functions. Interestingly, similar mitotic and developmental defects were observed upon knockdown of the DYNLL/LC8-type dynein light chain Cutup (Ctp), and dASCIZ loss-of-function phenotypes could be suppressed by ectopic Ctp expression. Consistent with a genetic function of dASCIZ upstream of Ctp, we show that loss of dASCIZ led to reduced endogenous Ctp mRNA and protein levels and dramatically reduced Ctp-LacZ reporter gene activity in vivo, indicating that dASCIZ regulates development and mitosis as a Ctp transcription factor. We speculate that the more severe mitotic defects in the absence of ASCIZ in flies compared to mice may be due to redundancy with a second, ASCIZ-independent, Dynll2 gene in mammals in contrast to a single Ctp gene in Drosophila. Altogether, our data demonstrate that ASCIZ is an evolutionary highly conserved transcriptional regulator of dynein light-chain levels and a novel regulator of mitosis in flies.


Subject(s)
Drosophila/genetics , Drosophila/metabolism , Dyneins/genetics , Gene Expression Regulation , Mitosis , Zinc Fingers/physiology , Animals , Apoptosis/genetics , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Dyneins/metabolism , Female , Gene Knockdown Techniques , Larva/genetics , Larva/metabolism , Male , Organogenesis/genetics , Phenotype , RNA Interference , Spindle Apparatus/genetics , Spindle Apparatus/metabolism , Wings, Animal/growth & development
6.
EMBO J ; 24(13): 2447-57, 2005 Jul 06.
Article in English | MEDLINE | ID: mdl-15933716

ABSTRACT

Nuclear Rad51 focus formation is required for homology-directed repair of DNA double-strand breaks (DSBs), but its regulation in response to non-DSB lesions is poorly understood. Here we report a novel human SQ/TQ cluster domain-containing protein termed ASCIZ that forms Rad51-containing foci in response to base-modifying DNA methylating agents but not in response to DSB-inducing agents. ASCIZ foci seem to form prior to Rad51 recruitment, and an ASCIZ core domain can concentrate Rad51 in focus-like structures independently of DNA damage. ASCIZ depletion dramatically increases apoptosis after methylating DNA damage and impairs Rad51 focus formation in response to methylating agents but not after ionizing radiation. ASCIZ focus formation and increased apoptosis in ASCIZ-depleted cells depend on the mismatch repair protein MLH1. Interestingly, ASCIZ foci form efficiently during G1 phase, when sister chromatids are unavailable as recombination templates. We propose that ASCIZ acts as a lesion-specific focus scaffold in a Rad51-dependent pathway that resolves cytotoxic repair intermediates, most likely single-stranded DNA gaps, resulting from MLH1-dependent processing of base lesions.


Subject(s)
Apoptosis , DNA Damage , DNA Methylation , DNA Repair , DNA-Binding Proteins/metabolism , Nuclear Proteins/physiology , Adaptor Proteins, Signal Transducing , Base Pair Mismatch , Carrier Proteins , Cell Line, Tumor , Cell Survival , Humans , MutL Protein Homolog 1 , Neoplasm Proteins/physiology , Protein Structure, Tertiary , Rad51 Recombinase , Signal Transduction
7.
J Biol Chem ; 279(38): 39636-44, 2004 Sep 17.
Article in English | MEDLINE | ID: mdl-15271990

ABSTRACT

Saccharomyces cerevisiae Rad53 has crucial functions in many aspects of the cellular response to DNA damage and replication blocks. To coordinate these diverse roles, Rad53 has two forkhead-associated (FHA) phosphothreonine-binding domains in addition to a kinase domain. Here, we show that the conserved N-terminal FHA1 domain is essential for the function of Rad53 to prevent the firing of late replication origins in response to replication blocks. However, the FHA1 domain is not required for Rad53 activation during S phase, and as a consequence of defective downstream signaling, Rad53 containing an inactive FHA1 domain is hyperphosphorylated in response to replication blocks. The FHA1 mutation dramatically hypersensitizes strains with defects in the cell cycle-wide checkpoint pathways (rad9Delta and rad17Delta) to DNA damage, but it is largely epistatic with defects in the replication checkpoint (mrc1Delta). Altogether, our data indicate that the FHA1 domain links activated Rad53 to downstream effectors in the replication checkpoint. The results reveal an important mechanistic difference to the homologous Schizosaccharomyces pombe FHA domain that is required for Mrc1-dependent activation of the corresponding Cds1 kinase. Surprisingly, despite the severely impaired replication checkpoint and also G(2)/M checkpoint functions, the FHA1 mutation by itself leads to only moderate viability defects in response to DNA damage, highlighting the importance of functionally redundant pathways.


Subject(s)
Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , DNA Replication/physiology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/enzymology , Binding Sites , Cell Cycle Proteins/chemistry , Checkpoint Kinase 2 , DNA-Binding Proteins , Forkhead Transcription Factors , Genes, cdc/physiology , Mutation , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Phosphoprotein Phosphatases/metabolism , Phosphorylation , Phosphothreonine/metabolism , Protein Phosphatase 2C , Protein Serine-Threonine Kinases/chemistry , Protein Structure, Tertiary , Replication Origin , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/chemistry , Transcription Factors/chemistry , Transcription Factors/genetics
8.
IUBMB Life ; 55(1): 23-7, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12716058

ABSTRACT

Forkhead-associated (FHA) domains are present in >200 diverse proteins in all phyla from bacteria to mammals and seem to be particularly prevalent in proteins with cell cycle control functions. Recent work from several laboratories has considerably improved our understanding of the structure and function of these domains that were virtually unknown a few years ago, and the first disease associations of FHA domains have now emerged. FHA domains form 11-stranded beta-sandwiches that contain some 100-180 amino acid residues with a high degree of sequence diversity. FHA domains act as phosphorylation-dependent protein-protein interaction modules that preferentially bind to phospho-threonine residues in their targets. Interestingly, point mutations in the human CHK2 gene that lead to single-residue amino acid substitutions in the FHA domain of this cell cycle checkpoint kinase have been found to cause a subset of cases of the Li-Fraumeni multi-cancer syndrome.


Subject(s)
Phosphothreonine/metabolism , Signal Transduction/physiology , Amino Acid Sequence , Animals , Cell Cycle/physiology , Humans , Models, Molecular , Molecular Sequence Data , Protein Binding , Protein Structure, Tertiary , Sequence Alignment
SELECTION OF CITATIONS
SEARCH DETAIL