Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20.422
Filter
Add more filters

Publication year range
1.
Annu Rev Immunol ; 34: 299-316, 2016 05 20.
Article in English | MEDLINE | ID: mdl-27168240

ABSTRACT

The discovery of tissue-resident innate lymphoid cell populations effecting different forms of type 1, 2, and 3 immunity; tissue repair; and immune regulation has transformed our understanding of mucosal immunity and allergy. The emerging complexity of these populations along with compounding issues of redundancy and plasticity raise intriguing questions about their precise lineage relationship. Here we review advances in mapping the emergence of these lineages from early lymphoid precursors. We discuss the identification of a common innate lymphoid cell precursor characterized by transient expression of the transcription factor PLZF, and the lineage relationships of innate lymphoid cells with conventional natural killer cells and lymphoid tissue inducer cells. We also review the rapidly growing understanding of the network of transcription factors that direct the development of these lineages.


Subject(s)
Cell Differentiation , Hypersensitivity/immunology , Immunity, Innate , Lymphocytes/immunology , Lymphoid Progenitor Cells/immunology , Animals , Cell Lineage , Cytokines/metabolism , Gene Expression Regulation/immunology , Gene Regulatory Networks , Humans , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Promyelocytic Leukemia Zinc Finger Protein , Th1 Cells/immunology , Th2 Cells/immunology
2.
Cell ; 186(18): 3810-3825.e18, 2023 08 31.
Article in English | MEDLINE | ID: mdl-37552983

ABSTRACT

A ubiquitous feature of eukaryotic transcriptional regulation is cooperative self-assembly between transcription factors (TFs) and DNA cis-regulatory motifs. It is thought that this strategy enables specific regulatory connections to be formed in gene networks between otherwise weakly interacting, low-specificity molecular components. Here, using synthetic gene circuits constructed in yeast, we find that high regulatory specificity can emerge from cooperative, multivalent interactions among artificial zinc-finger-based TFs. We show that circuits "wired" using the strategy of cooperative TF assembly are effectively insulated from aberrant misregulation of the host cell genome. As we demonstrate in experiments and mathematical models, this mechanism is sufficient to rescue circuit-driven fitness defects, resulting in genetic and functional stability of circuits in long-term continuous culture. Our naturally inspired approach offers a simple, generalizable means for building high-fidelity, evolutionarily robust gene circuits that can be scaled to a wide range of host organisms and applications.


Subject(s)
Gene Regulatory Networks , Transcription Factors , Transcription Factors/genetics , Saccharomyces cerevisiae/genetics , Genome
3.
Annu Rev Immunol ; 33: 291-353, 2015.
Article in English | MEDLINE | ID: mdl-25861976

ABSTRACT

Ion channels and transporters mediate the transport of charged ions across hydrophobic lipid membranes. In immune cells, divalent cations such as calcium, magnesium, and zinc have important roles as second messengers to regulate intracellular signaling pathways. By contrast, monovalent cations such as sodium and potassium mainly regulate the membrane potential, which indirectly controls the influx of calcium and immune cell signaling. Studies investigating human patients with mutations in ion channels and transporters, analysis of gene-targeted mice, or pharmacological experiments with ion channel inhibitors have revealed important roles of ionic signals in lymphocyte development and in innate and adaptive immune responses. We here review the mechanisms underlying the function of ion channels and transporters in lymphocytes and innate immune cells and discuss their roles in lymphocyte development, adaptive and innate immune responses, and autoimmunity, as well as recent efforts to develop pharmacological inhibitors of ion channels for immunomodulatory therapy.


Subject(s)
Adaptive Immunity/physiology , Immunity, Innate/physiology , Ion Channels/metabolism , Animals , Calcium Channels/genetics , Calcium Channels/metabolism , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Humans , Hypersensitivity/genetics , Hypersensitivity/immunology , Hypersensitivity/metabolism , Immunologic Deficiency Syndromes/drug therapy , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/immunology , Immunologic Deficiency Syndromes/metabolism , Immunotherapy/methods , Ion Channels/genetics , Lymphocytes/cytology , Lymphocytes/immunology , Lymphocytes/metabolism , Mast Cells/immunology , Mast Cells/metabolism , Molecular Targeted Therapy , Mutation , Signal Transduction
4.
Cell ; 185(12): 2148-2163.e27, 2022 06 09.
Article in English | MEDLINE | ID: mdl-35584702

ABSTRACT

Zinc (Zn) is an essential micronutrient and cofactor for up to 10% of proteins in living organisms. During Zn limitation, specialized enzymes called metallochaperones are predicted to allocate Zn to specific metalloproteins. This function has been putatively assigned to G3E GTPase COG0523 proteins, yet no Zn metallochaperone has been experimentally identified in any organism. Here, we functionally characterize a family of COG0523 proteins that is conserved across vertebrates. We identify Zn metalloprotease methionine aminopeptidase 1 (METAP1) as a COG0523 client, leading to the redesignation of this group of COG0523 proteins as the Zn-regulated GTPase metalloprotein activator (ZNG1) family. Using biochemical, structural, genetic, and pharmacological approaches across evolutionarily divergent models, including zebrafish and mice, we demonstrate a critical role for ZNG1 proteins in regulating cellular Zn homeostasis. Collectively, these data reveal the existence of a family of Zn metallochaperones and assign ZNG1 an important role for intracellular Zn trafficking.


Subject(s)
Metalloendopeptidases/metabolism , Zinc , Animals , GTP Phosphohydrolases/metabolism , Homeostasis , Metallochaperones/metabolism , Metalloproteins/genetics , Mice , Zebrafish/metabolism , Zinc/metabolism
5.
Cell ; 179(3): 659-670.e13, 2019 10 17.
Article in English | MEDLINE | ID: mdl-31587896

ABSTRACT

P2X receptors are trimeric, non-selective cation channels activated by extracellular ATP. The P2X7 receptor subtype is a pharmacological target because of involvement in apoptotic, inflammatory, and tumor progression pathways. It is the most structurally and functionally distinct P2X subtype, containing a unique cytoplasmic domain critical for the receptor to initiate apoptosis and not undergo desensitization. However, lack of structural information about the cytoplasmic domain has hindered understanding of the molecular mechanisms underlying these processes. We report cryoelectron microscopy structures of full-length rat P2X7 receptor in apo and ATP-bound states. These structures reveal how one cytoplasmic element, the C-cys anchor, prevents desensitization by anchoring the pore-lining helix to the membrane with palmitoyl groups. They show a second cytoplasmic element with a unique fold, the cytoplasmic ballast, which unexpectedly contains a zinc ion complex and a guanosine nucleotide binding site. Our structures provide first insights into the architecture and function of a P2X receptor cytoplasmic domain.


Subject(s)
Lipoylation , Receptors, Purinergic P2X7/chemistry , Adenosine Triphosphate/metabolism , Animals , Binding Sites , Cryoelectron Microscopy , Guanosine/metabolism , HEK293 Cells , Humans , Molecular Docking Simulation , Molecular Dynamics Simulation , Protein Binding , Receptors, Purinergic P2X7/metabolism , Sf9 Cells , Spodoptera , Xenopus , Zinc/metabolism
6.
Cell ; 175(6): 1520-1532.e15, 2018 11 29.
Article in English | MEDLINE | ID: mdl-30500536

ABSTRACT

N-methyl-D-aspartate receptors (NMDARs) play essential roles in memory formation, neuronal plasticity, and brain development, with their dysfunction linked to a range of disorders from ischemia to schizophrenia. Zinc and pH are physiological allosteric modulators of NMDARs, with GluN2A-containing receptors inhibited by nanomolar concentrations of divalent zinc and by excursions to low pH. Despite the widespread importance of zinc and proton modulation of NMDARs, the molecular mechanism by which these ions modulate receptor activity has proven elusive. Here, we use cryoelectron microscopy to elucidate the structure of the GluN1/GluN2A NMDAR in a large ensemble of conformations under a range of physiologically relevant zinc and proton concentrations. We show how zinc binding to the amino terminal domain elicits structural changes that are transduced though the ligand-binding domain and result in constriction of the ion channel gate.


Subject(s)
Multiprotein Complexes/chemistry , Protons , Receptors, N-Methyl-D-Aspartate/chemistry , Zinc/chemistry , Allosteric Regulation , Animals , Cryoelectron Microscopy , Hydrogen-Ion Concentration , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Multiprotein Complexes/ultrastructure , Protein Domains , Rats , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Sf9 Cells , Spodoptera , Zinc/metabolism
7.
Cell ; 173(1): 221-233.e12, 2018 03 22.
Article in English | MEDLINE | ID: mdl-29551271

ABSTRACT

Tandem zinc finger (ZF) proteins are the largest and most rapidly diverging family of DNA-binding transcription regulators in mammals. ZFP568 represses a transcript of placental-specific insulin like growth factor 2 (Igf2-P0) in mice. ZFP568 binds a 24-base pair sequence-specific element upstream of Igf2-P0 via the eleven-ZF array. Both DNA and protein conformations deviate from the conventional one finger-three bases recognition, with individual ZFs contacting 2, 3, or 4 bases and recognizing thymine on the opposite strand. These interactions arise from a shortened minor groove caused by an AT-rich stretch, suggesting adaptability of ZF arrays to sequence variations. Despite conservation in mammals, mutations at Igf2 and ZFP568 reduce their binding affinity in chimpanzee and humans. Our studies provide important insights into the evolutionary and structural dynamics of ZF-DNA interactions that play a key role in mammalian development and evolution.


Subject(s)
DNA/metabolism , Nuclear Proteins/metabolism , Amino Acid Sequence , Animals , Base Sequence , Binding Sites , Carrier Proteins/chemistry , Carrier Proteins/classification , Carrier Proteins/genetics , Carrier Proteins/metabolism , DNA/chemistry , Humans , Insulin-Like Growth Factor II/chemistry , Insulin-Like Growth Factor II/genetics , Insulin-Like Growth Factor II/metabolism , Mice , Molecular Dynamics Simulation , Nuclear Proteins/chemistry , Nuclear Proteins/classification , Nuclear Proteins/genetics , Nucleic Acid Conformation , Pan troglodytes , Phylogeny , Polymorphism, Single Nucleotide , Protein Binding , Protein Structure, Tertiary , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Sequence Alignment
8.
Cell ; 173(2): 430-442.e17, 2018 04 05.
Article in English | MEDLINE | ID: mdl-29606353

ABSTRACT

Fetal hemoglobin (HbF, α2γ2) level is genetically controlled and modifies severity of adult hemoglobin (HbA, α2ß2) disorders, sickle cell disease, and ß-thalassemia. Common genetic variation affects expression of BCL11A, a regulator of HbF silencing. To uncover how BCL11A supports the developmental switch from γ- to ß- globin, we use a functional assay and protein binding microarray to establish a requirement for a zinc-finger cluster in BCL11A in repression and identify a preferred DNA recognition sequence. This motif appears in embryonic and fetal-expressed globin promoters and is duplicated in γ-globin promoters. The more distal of the duplicated motifs is mutated in individuals with hereditary persistence of HbF. Using the CUT&RUN approach to map protein binding sites in erythroid cells, we demonstrate BCL11A occupancy preferentially at the distal motif, which can be disrupted by editing the promoter. Our findings reveal that direct γ-globin gene promoter repression by BCL11A underlies hemoglobin switching.


Subject(s)
Carrier Proteins/metabolism , Fetal Hemoglobin/genetics , Nuclear Proteins/metabolism , Base Sequence , Binding Sites , Carrier Proteins/genetics , Cell Line , Chromatin/metabolism , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Erythroid Cells/cytology , Erythroid Cells/metabolism , Gene Editing , Humans , Nuclear Proteins/genetics , Promoter Regions, Genetic , Protein Isoforms/genetics , Protein Isoforms/metabolism , Repressor Proteins , Zinc Fingers/genetics , beta-Globins/genetics , beta-Thalassemia/genetics , beta-Thalassemia/pathology , gamma-Globins/genetics
9.
Mol Cell ; 2024 Aug 19.
Article in English | MEDLINE | ID: mdl-39173638

ABSTRACT

Partitioning of repressive from actively transcribed chromatin in mammalian cells fosters cell-type-specific gene expression patterns. While this partitioning is reconstructed during differentiation, the chromatin occupancy of the key insulator, CCCTC-binding factor (CTCF), is unchanged at the developmentally important Hox clusters. Thus, dynamic changes in chromatin boundaries must entail other activities. Given its requirement for chromatin loop formation, we examined cohesin-based chromatin occupancy without known insulators, CTCF and Myc-associated zinc-finger protein (MAZ), and identified a family of zinc-finger proteins (ZNFs), some of which exhibit tissue-specific expression. Two such ZNFs foster chromatin boundaries at the Hox clusters that are distinct from each other and from MAZ. PATZ1 was critical to the thoracolumbar boundary in differentiating motor neurons and mouse skeleton, while ZNF263 contributed to cervicothoracic boundaries. We propose that these insulating activities act with cohesin, alone or combinatorially, with or without CTCF, to implement precise positional identity and cell fate during development.

10.
Cell ; 166(6): 1500-1511.e9, 2016 Sep 08.
Article in English | MEDLINE | ID: mdl-27610572

ABSTRACT

Reversing the dysfunctional T cell state that arises in cancer and chronic viral infections is the focus of therapeutic interventions; however, current therapies are effective in only some patients and some tumor types. To gain a deeper molecular understanding of the dysfunctional T cell state, we analyzed population and single-cell RNA profiles of CD8(+) tumor-infiltrating lymphocytes (TILs) and used genetic perturbations to identify a distinct gene module for T cell dysfunction that can be uncoupled from T cell activation. This distinct dysfunction module is downstream of intracellular metallothioneins that regulate zinc metabolism and can be identified at single-cell resolution. We further identify Gata-3, a zinc-finger transcription factor in the dysfunctional module, as a regulator of dysfunction, and we use CRISPR-Cas9 genome editing to show that it drives a dysfunctional phenotype in CD8(+) TILs. Our results open novel avenues for targeting dysfunctional T cell states while leaving activation programs intact.


Subject(s)
CD8-Positive T-Lymphocytes/pathology , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/pathology , Animals , CD8-Positive T-Lymphocytes/immunology , CRISPR-Cas Systems , Carcinogenesis/genetics , Carcinogenesis/immunology , Female , GATA3 Transcription Factor/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/genetics , Humans , Melanoma/immunology , Melanoma/physiopathology , Metallothionein/deficiency , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
11.
Mol Cell ; 83(2): 167-185.e9, 2023 Jan 19.
Article in English | MEDLINE | ID: mdl-36577401

ABSTRACT

The DNA double-strand break repair complex Mre11-Rad50-Nbs1 (MRN) detects and nucleolytically processes DNA ends, activates the ATM kinase, and tethers DNA at break sites. How MRN can act both as nuclease and scaffold protein is not well understood. The cryo-EM structure of MRN from Chaetomium thermophilum reveals a 2:2:1 complex with a single Nbs1 wrapping around the autoinhibited Mre11 nuclease dimer. MRN has two DNA-binding modes, one ATP-dependent mode for loading onto DNA ends and one ATP-independent mode through Mre11's C terminus, suggesting how it may interact with DSBs and intact DNA. MRNs two 60-nm-long coiled-coil domains form a linear rod structure, the apex of which is assembled by the two joined zinc-hook motifs. Apices from two MRN complexes can further dimerize, forming 120-nm spanning MRN-MRN structures. Our results illustrate the architecture of MRN and suggest how it mechanistically integrates catalytic and tethering functions.


Subject(s)
DNA Repair , DNA , Cryoelectron Microscopy , DNA/genetics , Acid Anhydride Hydrolases/genetics , DNA Breaks, Double-Stranded , DNA Repair Enzymes/metabolism , Adenosine Triphosphate/metabolism , MRE11 Homologue Protein/genetics , MRE11 Homologue Protein/metabolism , Cell Cycle Proteins/metabolism
12.
Annu Rev Biochem ; 84: 685-709, 2015.
Article in English | MEDLINE | ID: mdl-26034892

ABSTRACT

Hv1 is a voltage-gated proton-selective channel that plays critical parts in host defense, sperm motility, and cancer progression. Hv1 contains a conserved voltage-sensor domain (VSD) that is shared by a large family of voltage-gated ion channels, but it lacks a pore domain. Voltage sensitivity and proton conductivity are conferred by a unitary VSD that consists of four transmembrane helices. The architecture of Hv1 differs from that of cation channels that form a pore in the center among multiple subunits (as in most cation channels) or homologous repeats (as in voltage-gated sodium and calcium channels). Hv1 forms a dimer in which a cytoplasmic coiled coil underpins the two protomers and forms a single, long helix that is contiguous with S4, the transmembrane voltage-sensing segment. The closed-state structure of Hv1 was recently solved using X-ray crystallography. In this article, we discuss the gating mechanism of Hv1 and focus on cooperativity within dimers and their sensitivity to metal ions.


Subject(s)
Ion Channels/chemistry , Ion Channels/metabolism , Animals , Crystallography, X-Ray , Humans , Models, Molecular
13.
Genes Dev ; 37(15-16): 675-677, 2023 08 01.
Article in English | MEDLINE | ID: mdl-37673460

ABSTRACT

Specialized enzymes add methyl groups to the nitrogens of the amino acid histidine, altering the chemical properties of its imidazole ring and, in turn, the function of the modified (poly)peptide. In this issue of Genes & Development, Shimazu and colleagues (pp. 724-742) make the remarkable discovery that CARNMT1 acts as a dual-specificity histidine methyltransferase, modifying both the small-molecule dipeptide carnosine and a set of proteins, predominantly within RNA-binding C3H zinc finger (C3H ZF) motifs. As a result, CARNMT1 modulates the activity of its protein targets to affect RNA processing and metabolism, ultimately contributing an essential function during mammalian development.


Subject(s)
Amino Acids , Histidine , Animals , Methylation , Methyltransferases , Organogenesis , Mammals
14.
Genes Dev ; 37(15-16): 724-742, 2023 08 01.
Article in English | MEDLINE | ID: mdl-37612136

ABSTRACT

Histidine (His) residues are methylated in various proteins, but their roles and regulation mechanisms remain unknown. Here, we show that carnosine N-methyltransferase 1 (CARNMT1), a known His methyltransferase of dipeptide carnosine (ßAla-His), is a major His N1-position-specific methyltransferase. We found that 52 His sites in 20 proteins underwent CARNMT1-mediated methylation. The consensus methylation site for CARNMT1 was identified as Cx(F/Y)xH, a C3H zinc finger (C3H ZF) motif. CARNMT1-deficient and catalytically inactive mutant mice showed embryonic lethality. Among the CARNMT1 target C3H ZF proteins, RNA degradation mediated by Roquin and tristetraprolin (TTP) was affected by CARNMT1 and its enzymatic activity. Furthermore, the recognition of the 3' splice site of the CARNMT1 target C3H ZF protein U2AF1 was perturbed, and pre-mRNA alternative splicing (AS) was affected by CARNMT1 deficiency. These findings indicate that CARNMT1-mediated protein His methylation, which is essential for embryogenesis, plays roles in diverse aspects of RNA metabolism by targeting C3H ZF-type RNA-binding proteins and modulating their functions, including pre-mRNA AS and mRNA degradation regulation.


Subject(s)
Carnosine , Animals , Mice , Mice, Inbred C3H , Histidine/genetics , RNA Precursors , Methyltransferases/genetics , RNA Splice Sites , Zinc Fingers
15.
Immunity ; 54(7): 1417-1432.e7, 2021 07 13.
Article in English | MEDLINE | ID: mdl-34004142

ABSTRACT

The transcriptional repressor ZEB2 regulates development of many cell fates among somatic, neural, and hematopoietic lineages, but the basis for its requirement in these diverse lineages is unclear. Here, we identified a 400-basepair (bp) region located 165 kilobases (kb) upstream of the Zeb2 transcriptional start site (TSS) that binds the E proteins at several E-box motifs and was active in hematopoietic lineages. Germline deletion of this 400-bp region (Zeb2Δ-165mice) specifically prevented Zeb2 expression in hematopoietic stem cell (HSC)-derived lineages. Zeb2Δ-165 mice lacked development of plasmacytoid dendritic cells (pDCs), monocytes, and B cells. All macrophages in Zeb2Δ-165 mice were exclusively of embryonic origin. Using single-cell chromatin profiling, we identified a second Zeb2 enhancer located at +164-kb that was selectively active in embryonically derived lineages, but not HSC-derived ones. Thus, Zeb2 expression in adult, but not embryonic, hematopoiesis is selectively controlled by the -165-kb Zeb2 enhancer.


Subject(s)
Enhancer Elements, Genetic/genetics , Hematopoiesis/genetics , Transcription, Genetic/genetics , Zinc Finger E-box Binding Homeobox 2/genetics , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , Chromatin/genetics , Dendritic Cells/physiology , Female , Humans , Male , Mice , Mice, Inbred C57BL , Monocytes/physiology
16.
Mol Cell ; 82(3): 598-615.e8, 2022 02 03.
Article in English | MEDLINE | ID: mdl-34998453

ABSTRACT

An increasing number of genetic diseases are linked to deregulation of E3 ubiquitin ligases. Loss-of-function mutations in the RING-between-RING (RBR) family E3 ligase RNF216 (TRIAD3) cause Gordon-Holmes syndrome (GHS) and related neurodegenerative diseases. Functionally, RNF216 assembles K63-linked ubiquitin chains and has been implicated in regulation of innate immunity signaling pathways and synaptic plasticity. Here, we report crystal structures of key RNF216 reaction states including RNF216 in complex with ubiquitin and its reaction product, K63 di-ubiquitin. Our data provide a molecular explanation for chain-type specificity and reveal the molecular basis for disruption of RNF216 function by pathogenic GHS mutations. Furthermore, we demonstrate how RNF216 activity and chain-type specificity are regulated by phosphorylation and that RNF216 is allosterically activated by K63-linked di-ubiquitin. These molecular insights expand our understanding of RNF216 function and its role in disease and further define the mechanistic diversity of the RBR E3 ligase family.


Subject(s)
Cerebellar Ataxia/enzymology , Gonadotropin-Releasing Hormone/deficiency , Hypogonadism/enzymology , Protein Processing, Post-Translational , Ubiquitin-Protein Ligases/metabolism , Allosteric Regulation , Binding Sites , Catalysis , Cerebellar Ataxia/genetics , Crystallography, X-Ray , Genetic Predisposition to Disease , Gonadotropin-Releasing Hormone/genetics , HEK293 Cells , Humans , Hypogonadism/genetics , Loss of Function Mutation , Lysine , Models, Molecular , Phenotype , Phosphorylation , Protein Binding , Protein Conformation , Structure-Activity Relationship , Ubiquitin-Protein Ligases/genetics , Ubiquitination
17.
Mol Cell ; 82(17): 3135-3150.e9, 2022 09 01.
Article in English | MEDLINE | ID: mdl-35914531

ABSTRACT

Alternative polyadenylation (APA) enhances gene regulatory potential by increasing the diversity of mRNA transcripts. 3' UTR shortening through APA correlates with enhanced cellular proliferation and is a widespread phenomenon in tumor cells. Here, we show that the ubiquitously expressed transcription factor Sp1 binds RNA in vivo and is a common repressor of distal poly(A) site usage. RNA sequencing identified 2,344 genes (36% of the total mapped mRNA transcripts) with lengthened 3' UTRs upon Sp1 depletion. Sp1 preferentially binds the 3' UTRs of such lengthened transcripts and inhibits cleavage at distal sites by interacting with the subunits of the core cleavage and polyadenylation (CPA) machinery. The 3' UTR lengths of Sp1 target genes in breast cancer patient RNA-seq data correlate with Sp1 expression levels, implicating Sp1-mediated APA regulation in modulating tumorigenic properties. Taken together, our findings provide insights into the mechanism for dynamic APA regulation by unraveling a previously unknown function of the DNA-binding transcription factor Sp1.


Subject(s)
Poly A , Polyadenylation , 3' Untranslated Regions , Humans , Poly A/metabolism , RNA, Messenger/metabolism , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/metabolism , Zinc/metabolism
18.
Annu Rev Biochem ; 83: 409-39, 2014.
Article in English | MEDLINE | ID: mdl-24606144

ABSTRACT

Current technology enables the production of highly specific genome modifications with excellent efficiency and specificity. Key to this capability are targetable DNA cleavage reagents and cellular DNA repair pathways. The break made by these reagents can produce localized sequence changes through inaccurate nonhomologous end joining (NHEJ), often leading to gene inactivation. Alternatively, user-provided DNA can be used as a template for repair by homologous recombination (HR), leading to the introduction of desired sequence changes. This review describes three classes of targetable cleavage reagents: zinc-finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs), and CRISPR/Cas RNA-guided nucleases (RGNs). As a group, these reagents have been successfully used to modify genomic sequences in a wide variety of cells and organisms, including humans. This review discusses the properties, advantages, and limitations of each system, as well as the specific considerations required for their use in different biological systems.


Subject(s)
Endonucleases/genetics , Genetic Engineering/methods , Genome , Animals , Arabidopsis , DNA/chemistry , DNA Damage , DNA End-Joining Repair , DNA Repair , Drosophila , Drosophila melanogaster , Gene Deletion , Genomics , Humans , Mice , Protein Engineering/methods , Protein Structure, Tertiary , Rats , Recombination, Genetic , Zebrafish , Zinc Fingers
19.
Mol Cell ; 81(22): 4591-4604.e8, 2021 11 18.
Article in English | MEDLINE | ID: mdl-34592134

ABSTRACT

Protein ADP-ribosylation is a reversible post-translational modification that transfers ADP-ribose from NAD+ onto acceptor proteins. Poly(ADP-ribosyl)ation (PARylation), catalyzed by poly(ADP-ribose) polymerases (PARPs) and poly(ADP-ribose) glycohydrolases (PARGs), which remove the modification, regulates diverse cellular processes. However, the chemistry and physiological functions of mono(ADP-ribosyl)ation (MARylation) remain elusive. Here, we report that Arabidopsis zinc finger proteins SZF1 and SZF2, key regulators of immune gene expression, are MARylated by the noncanonical ADP-ribosyltransferase SRO2. Immune elicitation promotes MARylation of SZF1/SZF2 via dissociation from PARG1, which has an unconventional activity in hydrolyzing both poly(ADP-ribose) and mono(ADP-ribose) from acceptor proteins. MARylation antagonizes polyubiquitination of SZF1 mediated by the SH3 domain-containing proteins SH3P1/SH3P2, thereby stabilizing SZF1 proteins. Our study uncovers a noncanonical ADP-ribosyltransferase mediating MARylation of immune regulators and underpins the molecular mechanism of maintaining protein homeostasis by the counter-regulation of ADP-ribosylation and polyubiquitination to ensure proper immune responses.


Subject(s)
ADP-Ribosylation , Arabidopsis Proteins/metabolism , Arabidopsis/immunology , DNA-Binding Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Plant Immunity , Ubiquitination , Zinc Fingers , ADP Ribose Transferases/metabolism , Adenosine Diphosphate/chemistry , Arabidopsis/metabolism , CRISPR-Cas Systems , Genes, Plant , Glycoside Hydrolases/metabolism , Homeostasis , Humans , Hydrolysis , Mutation , Plants, Genetically Modified , Poly Adenosine Diphosphate Ribose/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Proteostasis , Seedlings/metabolism , Substrate Specificity , Tristetraprolin/chemistry , Two-Hybrid System Techniques , Ubiquitin/chemistry
20.
Genes Dev ; 34(23-24): 1753-1761, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33122294

ABSTRACT

Most eukaryotic pre-mRNAs must undergo 3'-end cleavage and polyadenylation prior to their export from the nucleus. A large number of proteins in several complexes participate in this 3'-end processing, including cleavage and polyadenylation specificity factor (CPSF) in mammals. The CPSF30 subunit contains five CCCH zinc fingers (ZFs), with ZF2-ZF3 being required for the recognition of the AAUAAA poly(A) signal. ZF4-ZF5 recruits the hFip1 subunit of CPSF, although the details of this interaction have not been characterized. Here we report the crystal structure of human CPSF30 ZF4-ZF5 in complex with residues 161-200 of hFip1 at 1.9 Å resolution, illuminating the molecular basis for their interaction. Unexpectedly, the structure reveals one hFip1 molecule binding to each ZF4 and ZF5, with a conserved mode of interaction. Our mutagenesis studies confirm that the CPSF30-hFip1 complex has 1:2 stoichiometry in vitro. Mutation of each binding site in CPSF30 still allows one copy of hFip1 to bind, while mutation of both sites abrogates binding. Our fluorescence polarization binding assays show that ZF4 has higher affinity for hFip1, with a Kd of 1.8 nM. We also demonstrate that two copies of the catalytic module of poly(A) polymerase (PAP) are recruited by the CPSF30-hFip1 complex in vitro, and both hFip1 binding sites in CPSF30 can support polyadenylation.


Subject(s)
Cleavage And Polyadenylation Specificity Factor/chemistry , Cleavage And Polyadenylation Specificity Factor/metabolism , Models, Molecular , mRNA Cleavage and Polyadenylation Factors/chemistry , Binding Sites , Cleavage And Polyadenylation Specificity Factor/genetics , Escherichia coli/genetics , Humans , Mutation , Protein Binding , Protein Structure, Quaternary , Zinc Fingers/physiology , mRNA Cleavage and Polyadenylation Factors/genetics , mRNA Cleavage and Polyadenylation Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL