Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
1.
Mol Cell ; 65(2): 247-259, 2017 Jan 19.
Article in English | MEDLINE | ID: mdl-27986371

ABSTRACT

Monoubiquitination and deubiquitination of FANCD2:FANCI heterodimer is central to DNA repair in a pathway that is defective in the cancer predisposition syndrome Fanconi anemia (FA). The "FA core complex" contains the RING-E3 ligase FANCL and seven other essential proteins that are mutated in various FA subtypes. Here, we purified recombinant FA core complex to reveal the function of these other proteins. The complex contains two spatially separate FANCL molecules that are dimerized by FANCB and FAAP100. FANCC and FANCE act as substrate receptors and restrict monoubiquitination to the FANCD2:FANCI heterodimer in only a DNA-bound form. FANCA and FANCG are dispensable for maximal in vitro ubiquitination. Finally, we show that the reversal of this reaction by the USP1:UAF1 deubiquitinase only occurs when DNA is disengaged. Our work reveals the mechanistic basis for temporal and spatial control of FANCD2:FANCI monoubiquitination that is critical for chemotherapy responses and prevention of Fanconi anemia.


Subject(s)
Fanconi Anemia Complementation Group D2 Protein/metabolism , Fanconi Anemia Complementation Group Proteins/metabolism , Fanconi Anemia/metabolism , Ubiquitination , Cell Line , DNA/genetics , DNA/metabolism , DNA-Binding Proteins/metabolism , Fanconi Anemia/genetics , Fanconi Anemia Complementation Group A Protein/metabolism , Fanconi Anemia Complementation Group C Protein/metabolism , Fanconi Anemia Complementation Group D2 Protein/genetics , Fanconi Anemia Complementation Group E Protein/metabolism , Fanconi Anemia Complementation Group G Protein/metabolism , Fanconi Anemia Complementation Group L Protein/metabolism , Fanconi Anemia Complementation Group Proteins/genetics , Humans , Inhibitor of Differentiation Protein 2/metabolism , Multiprotein Complexes , Nuclear Proteins/metabolism , Protein Binding , Protein Multimerization , Recombinant Proteins/metabolism , Substrate Specificity , Time Factors , Transfection , Ubiquitin-Specific Proteases/metabolism
2.
PLoS Genet ; 15(7): e1008266, 2019 07.
Article in English | MEDLINE | ID: mdl-31276497

ABSTRACT

Rothmund-Thomson syndrome (RTS) is a rare autosomal recessive disorder characterized by skin rash (poikiloderma), skeletal dysplasia, small stature, juvenile cataracts, sparse or absent hair, and predisposition to specific malignancies such as osteosarcoma and hematological neoplasms. RTS is caused by germ-line mutations in RECQL4, a RecQ helicase family member. In vitro studies have identified functions for the ATP-dependent helicase of RECQL4. However, its specific role in vivo remains unclear. To determine the physiological requirement and the biological functions of Recql4 helicase activity, we generated mice with an ATP-binding-deficient knock-in mutation (Recql4K525A). Recql4K525A/K525A mice were strikingly normal in terms of embryonic development, body weight, hematopoiesis, B and T cell development, and physiological DNA damage repair. However, mice bearing two distinct truncating mutations Recql4G522Efs and Recql4R347*, that abolished not only the helicase but also the C-terminal domain, developed a profound bone marrow failure and decrease in survival similar to a Recql4 null allele. These results demonstrate that the ATP-dependent helicase activity of Recql4 is not essential for its physiological functions and that other domains might contribute to this phenotype. Future studies need to be performed to elucidate the complex interactions of RECQL4 domains and its contribution to the development of RTS.


Subject(s)
Adenosine Triphosphate/metabolism , RecQ Helicases/genetics , RecQ Helicases/metabolism , Rothmund-Thomson Syndrome/genetics , Animals , B-Lymphocytes/metabolism , Binding Sites , Body Weight , DNA Damage , Disease Models, Animal , Embryonic Development , Gene Knock-In Techniques , Hematopoiesis , Humans , Mice , Phenotype , Protein Domains , RecQ Helicases/chemistry , T-Lymphocytes/metabolism
3.
Hum Mol Genet ; 28(15): 2573-2588, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31009951

ABSTRACT

Mutations in subunits of the cilia-specific cytoplasmic dynein-2 (CD2) complex cause short-rib thoracic dystrophy syndromes (SRTDs), characterized by impaired bone growth and life-threatening perinatal respiratory complications. Different SRTD mutations result in varying disease severities. It remains unresolved whether this reflects the extent of retained hypomorphic protein functions or relative importance of the affected subunits for the activity of the CD2 holoenzyme. To define the contribution of the LC8-type dynein light chain subunit to the CD2 complex, we have generated Dynll1-deficient mouse strains, including the first-ever conditional knockout (KO) mutant for any CD2 subunit. Germline Dynll1 KO mice exhibit a severe ciliopathy-like phenotype similar to mice lacking another CD2 subunit, Dync2li1. Limb mesoderm-specific loss of Dynll1 results in severe bone shortening similar to human SRTD patients. Mechanistically, loss of Dynll1 leads to a partial depletion of other SRTD-related CD2 subunits, severely impaired retrograde intra-flagellar transport, significant thickening of primary cilia and cilia signaling defects. Interestingly, phenotypes of Dynll1-deficient mice are very similar to entirely cilia-deficient Kif3a/Ift88-null mice, except that they never present with polydactyly and retain relatively higher signaling outputs in parts of the hedgehog pathway. Compared to complete loss of Dynll1, maintaining very low DYNLL1 levels in mice lacking the Dynll1-transcription factor ASCIZ (ATMIN) results in significantly attenuated phenotypes and improved CD2 protein levels. The results suggest that primary cilia can maintain some functionality in the absence of intact CD2 complexes and provide a viable animal model for the analysis of the underlying bone development defects of SRTDs.


Subject(s)
Bone Diseases, Developmental/metabolism , Cilia/metabolism , Ciliopathies/metabolism , Cytoplasmic Dyneins/genetics , Osteogenesis , Animals , Bone Diseases, Developmental/genetics , Bone Diseases, Developmental/physiopathology , Cells, Cultured , Cilia/physiology , Ciliopathies/genetics , Ciliopathies/physiopathology , Cytoplasmic Dyneins/metabolism , Cytoplasmic Dyneins/physiology , Extremities/pathology , Extremities/physiopathology , Hedgehog Proteins/metabolism , Male , Mice , Mice, Knockout , Phenotype , Signal Transduction , Transcription Factors/metabolism
4.
PLoS Genet ; 13(9): e1007010, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28922373

ABSTRACT

Mechanistic differences in the development and function of adaptive, high-affinity antibody-producing B-2 cells and innate-like, "natural" antibody-producing B-1a cells remain poorly understood. Here we show that the multi-functional dynein light chain (DYNLL1/LC8) plays important roles in the establishment of B-1a cells in the peritoneal cavity and in the ongoing development of B-2 lymphoid cells in the bone marrow of mice. Epistasis analyses indicate that Dynll1 regulates B-1a and early B-2 cell development in a single, linear pathway with its direct transcriptional activator ASCIZ (ATMIN/ZNF822), and that the two genes also have complementary functions during late B-2 cell development. The B-2 cell defects caused by loss of DYNLL1 were associated with lower levels of the anti-apoptotic protein BCL-2, and could be supressed by deletion of pro-apoptotic BIM which is negatively regulated by both DYNLL1 and BCL-2. Defects in B cell development caused by loss of DYNLL1 could also be partially suppressed by a pre-arranged SWHEL Igm-B cell receptor transgene. In contrast to the rescue of B-2 cell numbers, the B-1a cell deficiency in Dynll1-deleted mice could not be suppressed by the loss of Bim, and was further compounded by the SWHEL transgene. Conversely, oncogenic MYC expression, which is synthetic lethal with Dynll1 deletion in B-2 cells, did not further reduce B-1a cell numbers in Dynll1-defcient mice. Finally, we found that the ASCIZ-DYNLL1 axis was also required for the early-juvenile development of aggressive MYC-driven and p53-deficient B cell lymphomas. These results identify ASCIZ and DYNLL1 as the core of a transcriptional circuit that differentially regulates the development of the B-1a and B-2 B lymphoid cell lineages and plays a critical role in lymphomagenesis.


Subject(s)
B-Lymphocytes/metabolism , Dyneins/genetics , Lymphoma, B-Cell/genetics , Transcription Factors/genetics , Animals , B-Lymphocytes/immunology , Cell Lineage/genetics , Cytoplasmic Dyneins , Dyneins/metabolism , Gene Expression Regulation , Humans , Lymphocytes/metabolism , Lymphocytes/pathology , Lymphoma, B-Cell/pathology , Mice , Peritoneal Cavity , Proto-Oncogene Proteins c-myc/genetics , Transcription Factors/metabolism , Tumor Suppressor Protein p53/genetics
5.
Mol Cell ; 30(6): 767-78, 2008 Jun 20.
Article in English | MEDLINE | ID: mdl-18570878

ABSTRACT

Forkhead-associated (FHA) domains recognize phosphothreonines, and SQ/TQ cluster domains (SCDs) contain concentrated phosphorylation sites for ATM/ATR-like DNA-damage-response kinases. The Rad53-SCD1 has dual functions in regulating the activation of the Rad53-Dun1 checkpoint kinase cascade but with unknown molecular mechanisms. Here we present structural, biochemical, and genetic evidence that Dun1-FHA possesses an unprecedented diphosphothreonine-binding specificity. The Dun1-FHA has >100-fold increased affinity for diphosphorylated relative to monophosphorylated Rad53-SCD1 due to the presence of two separate phosphothreonine-binding pockets. In vivo, any single threonine of Rad53-SCD1 is sufficient for Rad53 activation and RAD53-dependent survival of DNA damage, but two adjacent phosphothreonines in the Rad53-SCD1 and two phosphothreonine-binding sites in the Dun1-FHA are necessary for Dun1 activation and DUN1-dependent transcriptional responses to DNA damage. The results uncover a phospho-counting mechanism that regulates the specificity of SCD, and provide mechanistic insight into a role of multisite phosphorylation in DNA-damage signaling.


Subject(s)
Cell Cycle Proteins/metabolism , Phosphothreonine/metabolism , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/enzymology , Binding Sites , Cell Cycle Proteins/chemistry , Checkpoint Kinase 2 , DNA Damage , DNA, Fungal/genetics , Enzyme Activation , Kinetics , Ligands , Phosphothreonine/chemistry , Protein Binding , Protein Kinases/chemistry , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/chemistry , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/chemistry , Sensitivity and Specificity
6.
Mol Cell Proteomics ; 13(2): 551-65, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24302356

ABSTRACT

The cell cycle checkpoint kinases play central roles in the genome maintenance of eukaryotes. Activation of the yeast checkpoint kinase Rad53 involves Rad9 or Mrc1 adaptor-mediated phospho-priming by Mec1 kinase, followed by auto-activating phosphorylation within its activation loop. However, the mechanisms by which these adaptors regulate priming phosphorylation of specific sites and how this then leads to Rad53 activation remain poorly understood. Here we used quantitative mass spectrometry to delineate the stepwise phosphorylation events in the activation of endogenous Rad53 in response to S phase alkylation DNA damage, and we show that the two Rad9 and Mrc1 adaptors, the four N-terminal Mec1-target TQ sites of Rad53 (Rad53-SCD1), and Rad53-FHA2 coordinate intimately for optimal priming phosphorylation to support substantial Rad53 auto-activation. Rad9 or Mrc1 alone can mediate surprisingly similar Mec1 target site phosphorylation patterns of Rad53, including previously undetected tri- and tetraphosphorylation of Rad53-SCD1. Reducing the number of TQ motifs turns the SCD1 into a proportionally poorer Mec1 target, which then requires the presence of both Mrc1 and Rad9 for sufficient priming and auto-activation. The phosphothreonine-interacting Rad53-FHA domains, particularly FHA2, regulate phospho-priming by interacting with the checkpoint mediators but do not seem to play a major role in the phospho-SCD1-dependent auto-activation step. Finally, mutation of all four SCD1 TQ motifs greatly reduces Rad53 activation but does not eliminate it, and residual Rad53 activity in this mutant is dependent on Rad9 but not Mrc1. Altogether, our results provide a paradigm for how phosphorylation site clusters and checkpoint mediators can be involved in the regulation of signaling relay in protein kinase cascades in vivo and elucidate an SCD1-independent Rad53 auto-activation mechanism through the Rad9 pathway. The work also demonstrates the power of mass spectrometry for in-depth analyses of molecular mechanisms in cellular signaling in vivo.


Subject(s)
Cell Cycle Proteins/metabolism , Checkpoint Kinase 2/metabolism , Mass Spectrometry/methods , Protein Kinases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Alkylating Agents/pharmacology , Catalytic Domain , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/genetics , Checkpoint Kinase 2/chemistry , Checkpoint Kinase 2/genetics , DNA Damage/physiology , Enzyme Activation , Homeostasis , Organisms, Genetically Modified , Phosphorylation , Protein Interaction Mapping , Protein Processing, Post-Translational , S Phase/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics
7.
PLoS Genet ; 9(2): e1003298, 2013.
Article in English | MEDLINE | ID: mdl-23408915

ABSTRACT

Single-stranded DNA binding proteins (SSBs) regulate multiple DNA transactions, including replication, transcription, and repair. We recently identified SSB1 as a novel protein critical for the initiation of ATM signaling and DNA double-strand break repair by homologous recombination. Here we report that germline Ssb1(-/-) embryos die at birth from respiratory failure due to severe rib cage malformation and impaired alveolar development, coupled with additional skeletal defects. Unexpectedly, Ssb1(-/-) fibroblasts did not exhibit defects in Atm signaling or γ-H2ax focus kinetics in response to ionizing radiation (IR), and B-cell specific deletion of Ssb1 did not affect class-switch recombination in vitro. However, conditional deletion of Ssb1 in adult mice led to increased cancer susceptibility with broad tumour spectrum, impaired male fertility with testicular degeneration, and increased radiosensitivity and IR-induced chromosome breaks in vivo. Collectively, these results demonstrate essential roles of Ssb1 in embryogenesis, spermatogenesis, and genome stability in vivo.


Subject(s)
Carrier Proteins , DNA Breaks, Double-Stranded/radiation effects , DNA Repair , Nuclear Proteins , Suppressor of Cytokine Signaling Proteins , Animals , B-Lymphocytes/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Chromosome Breakage/radiation effects , DNA Repair/genetics , DNA Repair/radiation effects , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression Regulation, Developmental , Genomic Instability/genetics , Histones/genetics , Histones/metabolism , Homologous Recombination/genetics , Humans , Infertility, Male/genetics , Male , Mice , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Radiation Tolerance/genetics , Radiation, Ionizing , Signal Transduction/genetics , Spermatogenesis , Suppressor of Cytokine Signaling Proteins/deficiency , Suppressor of Cytokine Signaling Proteins/genetics , Suppressor of Cytokine Signaling Proteins/metabolism , Transcription Factors
8.
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
9.
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
10.
Cell Genom ; 3(8): 100349, 2023 Aug 09.
Article in English | MEDLINE | ID: mdl-37601968

ABSTRACT

Meiotic crossovers are required for accurate chromosome segregation and producing new allelic combinations. Meiotic crossover numbers are tightly regulated within a narrow range, despite an excess of initiating DNA double-strand breaks. Here, we reveal the tumor suppressor FANCM as a meiotic anti-crossover factor in mammals. We use unique large-scale crossover analyses with both single-gamete sequencing and pedigree-based bulk-sequencing datasets to identify a genome-wide increase in crossover frequencies in Fancm-deficient mice. Gametogenesis is heavily perturbed in Fancm loss-of-function mice, which is consistent with the reproductive defects reported in humans with biallelic FANCM mutations. A portion of the gametogenesis defects can be attributed to the cGAS-STING pathway after birth. Despite the gametogenesis phenotypes in Fancm mutants, both sexes are capable of producing offspring. We propose that the anti-crossover function and role in gametogenesis of Fancm are separable and will inform diagnostic pathways for human genomic instability disorders.

11.
Cell Rep ; 41(10): 111749, 2022 12 06.
Article in English | MEDLINE | ID: mdl-36476850

ABSTRACT

Co-transcriptional R loops arise from stalling of RNA polymerase, leading to the formation of stable DNA:RNA hybrids. Unresolved R loops promote genome instability but are counteracted by helicases and nucleases. Here, we show that branchpoint translocases are a third class of R-loop-displacing enzyme in vitro. In cells, deficiency in the Fanconi-anemia-associated branchpoint translocase FANCM causes R-loop accumulation, particularly after treatment with DNA:RNA-hybrid-stabilizing agents. This correlates with FANCM localization at R-loop-prone regions of the genome. Moreover, other branchpoint translocases associated with human disease, such as SMARCAL1 and ZRANB3, and those from lower organisms can also remove R loops in vitro. Branchpoint translocases are more potent than helicases in resolving R loops, indicating their evolutionary important role in R-loop suppression. In human cells, FANCM, SMARCAL1, and ZRANB3 depletion causes additive effects on R-loop accumulation and DNA damage. Our work reveals a mechanistic basis for R-loop displacement that is linked to genome stability.


Subject(s)
R-Loop Structures , RNA , Humans , DNA Helicases/genetics
12.
Mol Cell Biol ; 41(12): e0025121, 2021 11 22.
Article in English | MEDLINE | ID: mdl-34543116

ABSTRACT

Toll-like receptors (TLRs) and interleukin-1 (IL-1) receptors regulate immune and inflammatory responses by activating the NF-κB pathway. Here, we report that B-cell-specific loss of dynein light chain 1 (DYNLL1, LC8) or its designated transcription factor ASCIZ (ATMIN) leads to severely reduced in vivo antibody responses to TLR4-dependent but not T-cell-dependent antigens in mice. This defect was independent of DYNLL1's established roles in modulating BIM-dependent apoptosis and 53BP1-dependent antibody class-switch recombination. In B cells and fibroblasts, the ASCIZ-DYNLL1 axis was required for TLR4-, IL-1-, and CD40-mediated NF-κB pathway activation but dispensable for antigen receptor and tumor necrosis factor α (TNF-α) signaling. In contrast to previous reports that overexpressed DYNLL1 directly inhibits the phosphorylation and degradation of the NF-κB inhibitor IκBα, we found here that under physiological conditions, DYNLL1 is required for signal-specific activation of the NF-κB pathway upstream of IκBα. Our data identify DYNLL1 as a signal-specific regulator of the NF-κB pathway and indicate that it may act as a universal modulator of TLR4 (and IL-1) signaling with wide-ranging roles in inflammation and immunity.


Subject(s)
Antibody Formation/immunology , Cytoplasmic Dyneins/metabolism , NF-kappa B/metabolism , Toll-Like Receptor 4/immunology , Transcription Factors/metabolism , Animals , B-Lymphocytes/immunology , CD40 Antigens/metabolism , Cells, Cultured , Cytoplasmic Dyneins/genetics , Immunoglobulin Class Switching/immunology , Mice , Mice, Inbred C57BL , NF-KappaB Inhibitor alpha/metabolism , T-Lymphocytes/immunology , Transcription Factors/genetics , Tumor Necrosis Factor-alpha/metabolism , Tumor Suppressor p53-Binding Protein 1/immunology
13.
Yeast ; 27(1): 41-52, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19894211

ABSTRACT

Recent evidence indicates considerable cross-talk between genome maintenance and cell integrity control pathways. The RNA recognition motif (RRM)- and SQ/TQ cluster domain (SCD)-containing protein Mdt1 is required for repair of 3'-blocked DNA double-strand breaks (DSBs) and efficient recombinational maintenance of telomeres in budding yeast. Here we show that deletion of MDT1 (PIN4/YBL051C) leads to severe synthetic sickness in the absence of the genes for the central cell integrity MAP kinases Bck1 and Slt2/Mpk1. Consistent with a cell integrity function, mdt1Delta cells are hypersensitive to the cell wall toxin calcofluor white and the Bck1-Slt2 pathway activator caffeine. An RRM-deficient mdt1-RRM0 allele shares the severe bleomycin hypersensitivity, inefficient recombinational telomere maintenance and slt2 synthetic sickness phenotypes, but not the cell wall toxin hypersensitivity with mdt1Delta. However, the mdt1-RRM(3A) allele, where only the RNA-binding site is mutated, behaves similarly to the wild-type, suggesting that the Mdt1 RRM functions as a protein-protein interaction rather than a nucleic acid-binding module. Surprisingly, in a strain background where double mutants are sick but still viable, bck1Deltamdt1Delta and slt2Deltamdt1Delta mutants differ in some of their phenotypes, consistent with the emerging concept of flexible signal entry and exit points in the Bck1-Mkk1/2-Slt2 pathway. Overall, the results indicate that Mdt1 has partially separable functions in both cell wall and genome integrity pathways.


Subject(s)
Gene Expression Regulation, Fungal/physiology , Rad52 DNA Repair and Recombination Protein/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/metabolism , Benzenesulfonates , Binding Sites , Bleomycin , Caffeine , Gene Deletion , Genome, Fungal , MAP Kinase Signaling System/genetics , MAP Kinase Signaling System/physiology , Protein Binding , RNA, Fungal/metabolism , Rad52 DNA Repair and Recombination Protein/genetics , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Sorbitol
14.
Circ Res ; 102(7): 786-94, 2008 Apr 11.
Article in English | MEDLINE | ID: mdl-18292599

ABSTRACT

S100A1, a Ca(2+)-binding protein of the EF-hand type, is known to modulate sarcoplasmic reticulum Ca(2+) handling in skeletal muscle and cardiomyocytes. Recently, S100A1 has been shown to be expressed in endothelial cells (ECs). Because intracellular Ca(2+) ([Ca(2+)](i)) transients can be involved in important EC functions and endothelial NO synthase activity, we sought to investigate the impact of endothelial S100A1 on the regulation of endothelial and vascular function. Thoracic aortas from S100A1 knockout mice (SKO) showed significantly reduced relaxation in response to acetylcholine compared with wild-type vessels, whereas direct vessel relaxation using sodium nitroprusside was unaltered. Endothelial dysfunction attributable to the lack of S100A1 expression could also be demonstrated in vivo and translated into hypertension of SKO. Mechanistically, both basal and acetylcholine-induced endothelial NO release of SKO aortas was significantly reduced compared with wild type. Impaired endothelial NO production in SKO could be attributed, at least in part, to diminished agonist-induced [Ca(2+)](i) transients in ECs. Consistently, silencing endothelial S100A1 expression in wild type also reduced [Ca(2+)](i) and NO generation. Moreover, S100A1 overexpression in ECs further increased NO generation that was blocked by the inositol-1,4,5-triphosphate receptor blocker 2-aminoethoxydiphenylborate. Finally, cardiac endothelial S100A1 expression was shown to be downregulated in heart failure in vivo. Collectively, endothelial S100A1 critically modulates vascular function because lack of S100A1 expression leads to decreased [Ca(2+)](i) and endothelial NO release, which contributes, at least partially, to impaired endothelium-dependent vascular relaxation and hypertension in SKO mice. Targeting endothelial S100A1 expression may, therefore, be a novel therapeutic means to improve endothelial function in vascular disease or heart failure.


Subject(s)
Endothelium, Vascular/metabolism , Nitric Oxide/metabolism , S100 Proteins/metabolism , Vasoconstriction/physiology , Vasodilation/physiology , Acetylcholine/pharmacology , Animals , Aorta, Thoracic/metabolism , Aorta, Thoracic/pathology , Aorta, Thoracic/physiopathology , Calcium/metabolism , Disease Models, Animal , Endothelium, Vascular/pathology , Endothelium, Vascular/physiopathology , Female , Heart Failure/metabolism , Heart Failure/pathology , Heart Failure/physiopathology , Humans , Hypertension/metabolism , Hypertension/pathology , Hypertension/physiopathology , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitroprusside/pharmacology , Rats , Rats, Wistar , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Vasoconstriction/drug effects , Vasodilation/drug effects , Vasodilator Agents/pharmacology
15.
Mol Cell Biol ; 27(18): 6532-45, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17636027

ABSTRACT

DNA recombination plays critical roles in DNA repair and alternative telomere maintenance. Here we show that absence of the SQ/TQ cluster domain-containing protein Mdt1 (Ybl051c) renders Saccharomyces cerevisiae particularly hypersensitive to bleomycin, a drug that causes 3'-phospho-glycolate-blocked DNA double-strand breaks (DSBs). mdt1Delta also hypersensitizes partially recombination-defective cells to camptothecin-induced 3'-phospho-tyrosyl protein-blocked DSBs. Remarkably, whereas mdt1Delta cells are unable to restore broken chromosomes after bleomycin treatment, they efficiently repair "clean" endonuclease-generated DSBs. Epistasis analyses indicate that MDT1 acts in the repair of bleomycin-induced DSBs by regulating the efficiency of the homologous recombination pathway as well as telomere-related functions of the KU complex. Moreover, mdt1Delta leads to severe synthetic growth defects with a deletion of the recombination facilitator and telomere-positioning factor gene CTF18 already in the absence of exogenous DNA damage. Importantly, mdt1Delta causes a dramatic shift from the usually prevalent type II to the less-efficient type I pathway of recombinational telomere maintenance in the absence of telomerase in liquid senescence assays. As telomeres resemble protein-blocked DSBs, the results indicate that Mdt1 acts in a novel blocked-end-specific recombination pathway that is required for the efficiency of both drug-induced DSB repair and telomerase-independent telomere maintenance.


Subject(s)
DNA Damage , DNA Repair/physiology , Rad52 DNA Repair and Recombination Protein/metabolism , Recombination, Genetic , Saccharomyces cerevisiae Proteins/physiology , Telomere/metabolism , Antibiotics, Antineoplastic/toxicity , Antigens, Nuclear/genetics , Antigens, Nuclear/physiology , Antineoplastic Agents, Phytogenic/toxicity , Bleomycin/toxicity , Camptothecin/toxicity , DNA Repair/genetics , DNA, Fungal/drug effects , DNA, Fungal/genetics , DNA, Fungal/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Epistasis, Genetic , Ku Autoantigen , Rad52 DNA Repair and Recombination Protein/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Telomere/genetics
16.
Biochem Biophys Res Commun ; 371(2): 225-9, 2008 Jun 27.
Article in English | MEDLINE | ID: mdl-18433721

ABSTRACT

ASCIZ (ATMIN) was recently identified as a novel DNA damage response protein. Here we report that ASCIZ-deficient chicken DT40 B lymphocyte lines displayed markedly increased Ig gene conversion rates, whereas overexpression of human ASCIZ reduced Ig gene conversion below wild-type levels. However, neither the efficiency of double-strand break repair nor hypermutation was affected by ASCIZ levels, indicating that ASCIZ does not directly control homologous recombination or formation of abasic sites. Loss of ASCIZ led to mild sensitivity to the base damaging agent methylmethane sulfonate (MMS), yet remarkably, suppressed the dramatic MMS hypersensitivity of polbeta-deficient cells. These data suggest that ASCIZ may affect the choice between competing base repair pathways in a manner that reduces the amount of substrates available for Ig gene conversion.


Subject(s)
Alkylating Agents/pharmacology , Carrier Proteins/metabolism , DNA Damage/genetics , DNA Repair/genetics , Gene Conversion , Genes, Immunoglobulin/genetics , Animals , Carrier Proteins/genetics , Cell Line , DNA Polymerase beta/genetics , Drug Resistance/genetics , Humans , Methyl Methanesulfonate/pharmacology , Mice , Mice, Transgenic , Mutagens/pharmacology , Nuclear Proteins , Suppression, Genetic , Transcription Factors
17.
Front Immunol ; 9: 592, 2018.
Article in English | MEDLINE | ID: mdl-29623080

ABSTRACT

The proapoptotic BH3-only protein BIM (Bcl2l11) plays key roles in the maintenance of multiple hematopoietic cell types. In mice, germline knockout or conditional pan-hematopoietic deletion of Bim results in marked splenomegaly and significantly increased numbers of B cells. However, it has remained unclear whether these abnormalities reflect the loss of cell-intrinsic functions of BIM within the B lymphoid lineage and, if so, which stages in the lifecycle of B cells are most impacted by the loss of BIM. Here, we show that B lymphoid-specific conditional deletion of Bim during early development (i.e., in pro-B cells using Mb1-Cre) or during the final differentiation steps (i.e., in transitional B cells using Cd23-Cre) led to a similar >2-fold expansion of the mature follicular B cell pool. Notably, while the expansion of mature B cells was quantitatively similar in conditional and germline Bim-deficient mice, the splenomegaly was significantly attenuated after B lymphoid-specific compared to global Bim deletion. In vitro, conditional loss of Bim substantially increased the survival of mature B cells that were refractory to activation by lipopolysaccharide. Finally, we also found that conditional deletion of just one Bim allele by Mb1-Cre dramatically accelerated the development of Myc-driven B cell lymphoma, in a manner that was comparable to the effect of germline Bim heterozygosity. These data indicate that, under physiological conditions, BIM regulates B cell homeostasis predominantly by limiting the life span of non-activated mature B cells, and that it can have additional effects on developing B cells under pathological conditions.


Subject(s)
Apoptosis/genetics , Apoptosis/immunology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Bcl-2-Like Protein 11/genetics , Homeostasis , Animals , Cell Survival/genetics , Cell Survival/immunology , Gene Deletion , Genes, myc , Genotype , Immunophenotyping , Lymphopoiesis/genetics , Lymphopoiesis/immunology , Mice , Phenotype
18.
Elife ; 72018 05 01.
Article in English | MEDLINE | ID: mdl-29714690

ABSTRACT

The transcription factor ASCIZ (ATMIN, ZNF822) has an unusually high number of recognition motifs for the product of its main target gene, the hub protein LC8 (DYNLL1). Using a combination of biophysical methods, structural analysis by NMR and electron microscopy, and cellular transcription assays, we developed a model that proposes a concerted role of intrinsic disorder and multiple LC8 binding events in regulating LC8 transcription. We demonstrate that the long intrinsically disordered C-terminal domain of ASCIZ binds LC8 to form a dynamic ensemble of complexes with a gradient of transcriptional activity that is inversely proportional to LC8 occupancy. The preference for low occupancy complexes at saturating LC8 concentrations with both human and Drosophila ASCIZ indicates that negative cooperativity is an important feature of ASCIZ-LC8 interactions. The prevalence of intrinsic disorder and multivalency among transcription factors suggests that formation of heterogeneous, dynamic complexes is a widespread mechanism for tuning transcriptional regulation.


Subject(s)
Cytoplasmic Dyneins/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Dyneins/metabolism , Gene Expression Regulation , Intrinsically Disordered Proteins/metabolism , Transcription Factors/metabolism , Animals , Cytoplasmic Dyneins/chemistry , Cytoplasmic Dyneins/genetics , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Drosophila melanogaster/growth & development , Drosophila melanogaster/physiology , Dyneins/chemistry , Dyneins/genetics , Humans , Intrinsically Disordered Proteins/genetics , Transcription Factors/chemistry , Transcription Factors/genetics
19.
Nat Commun ; 9(1): 5406, 2018 12 17.
Article in English | MEDLINE | ID: mdl-30559443

ABSTRACT

53BP1 controls a specialized non-homologous end joining (NHEJ) pathway that is essential for adaptive immunity, yet oncogenic in BRCA1 mutant cancers. Intra-chromosomal DNA double-strand break (DSB) joining events during immunoglobulin class switch recombination (CSR) require 53BP1. However, in BRCA1 mutant cells, 53BP1 blocks homologous recombination (HR) and promotes toxic NHEJ, resulting in genomic instability. Here, we identify the protein dimerization hub-DYNLL1-as an organizer of multimeric 53BP1 complexes. DYNLL1 binding stimulates 53BP1 oligomerization, and promotes 53BP1's recruitment to, and interaction with, DSB-associated chromatin. Consequently, DYNLL1 regulates 53BP1-dependent NHEJ: CSR is compromised upon deletion of Dynll1 or its transcriptional regulator Asciz, or by mutation of DYNLL1 binding motifs in 53BP1; furthermore, Brca1 mutant cells and tumours are rendered resistant to poly-ADP ribose polymerase (PARP) inhibitor treatments upon deletion of Dynll1 or Asciz. Thus, our results reveal a mechanism that regulates 53BP1-dependent NHEJ and the therapeutic response of BRCA1-deficient cancers.


Subject(s)
BRCA1 Protein/genetics , Cytoplasmic Dyneins/metabolism , DNA End-Joining Repair/genetics , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Transcription Factors/metabolism , Tumor Suppressor p53-Binding Protein 1/genetics , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , CRISPR-Cas Systems , Cell Line, Tumor , DNA Breaks, Double-Stranded , Female , Genomic Instability/genetics , HEK293 Cells , Humans , MCF-7 Cells , Mice , Mice, Inbred C57BL , Mice, Knockout
20.
Circulation ; 114(12): 1258-68, 2006 Sep 19.
Article in English | MEDLINE | ID: mdl-16952982

ABSTRACT

BACKGROUND: Diminished cardiac S100A1 protein levels are characteristic of ischemic and dilated human cardiomyopathy. Because S100A1 has recently been identified as a Ca2+-dependent inotropic factor in the heart, this study sought to explore the pathophysiological relevance of S100A1 levels in development and progression of postischemic heart failure (HF). METHODS AND RESULTS: S100A1-transgenic (STG) and S100A1-knockout (SKO) mice were subjected to myocardial infarction (MI) by surgical left anterior descending coronary artery ligation, and survival, cardiac function, and remodeling were compared with nontransgenic littermate control (NLC) and wild-type (WT) animals up to 4 weeks. Although MI size was similar in all groups, infarcted S100A1-deficient hearts (SKO-MI) responded with acute contractile decompensation and accelerated transition to HF, rapid onset of cardiac remodeling with augmented apoptosis, and excessive mortality. NLC/WT-MI mice, displaying a progressive decrease in cardiac S100A1 expression, showed a later onset of cardiac remodeling and progression to HF. Infarcted S100A1-overexpressing hearts (STG-MI), however, showed preserved global contractile performance, abrogated apoptosis, and prevention from cardiac hypertrophy and HF with superior survival compared with NLC/WT-MI and SKO-MI. Both Gq-protein-dependent signaling and protein kinase C activation resulted in decreased cardiac S100A1 mRNA and protein levels, whereas Gs-protein-related signaling exerted opposite effects on cardiac S100A1 abundance. Mechanistically, sarcoplasmic reticulum Ca2+ cycling and beta-adrenergic signaling were severely impaired in SKO-MI myocardium but preserved in STG-MI. CONCLUSIONS: Our novel proof-of-concept study provides evidence that downregulation of S100A1 protein critically contributes to contractile dysfunction of the diseased heart, which is potentially responsible for driving the progressive downhill clinical course of patients with HF.


Subject(s)
Cardiac Output, Low/physiopathology , Myocardial Contraction/physiology , Myocardial Infarction/physiopathology , Myocardium/metabolism , S100 Proteins/metabolism , Animals , Apoptosis/physiology , Calcium-Transporting ATPases/genetics , Calcium-Transporting ATPases/metabolism , Cardiac Output, Low/etiology , Cardiac Output, Low/prevention & control , Cyclic AMP/physiology , Disease Progression , Down-Regulation , GTP-Binding Protein alpha Subunits, Gs/physiology , Mice , Mice, Knockout , Mice, Transgenic , Myocardial Contraction/genetics , Myocardial Infarction/complications , Myocardial Infarction/pathology , Myocardium/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Adrenergic, beta/physiology , S100 Proteins/genetics , Sarcoplasmic Reticulum Calcium-Transporting ATPases , Ventricular Remodeling/physiology
SELECTION OF CITATIONS
SEARCH DETAIL