Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 9.767
Filter
Add more filters

Publication year range
1.
Cell ; 185(13): 2370-2386.e18, 2022 06 23.
Article in English | MEDLINE | ID: mdl-35597242

ABSTRACT

2',3'-cAMP is a positional isomer of the well-established second messenger 3',5'-cAMP, but little is known about the biology of this noncanonical cyclic nucleotide monophosphate (cNMP). Toll/interleukin-1 receptor (TIR) domains of nucleotide-binding leucine-rich repeat (NLR) immune receptors have the NADase function necessary but insufficient to activate plant immune responses. Here, we show that plant TIR proteins, besides being NADases, act as 2',3'-cAMP/cGMP synthetases by hydrolyzing RNA/DNA. Structural data show that a TIR domain adopts distinct oligomers with mutually exclusive NADase and synthetase activity. Mutations specifically disrupting the synthetase activity abrogate TIR-mediated cell death in Nicotiana benthamiana (Nb), supporting an important role for these cNMPs in TIR signaling. Furthermore, the Arabidopsis negative regulator of TIR-NLR signaling, NUDT7, displays 2',3'-cAMP/cGMP but not 3',5'-cAMP/cGMP phosphodiesterase activity and suppresses cell death activity of TIRs in Nb. Our study identifies a family of 2',3'-cAMP/cGMP synthetases and establishes a critical role for them in plant immune responses.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Arabidopsis/genetics , Arabidopsis Proteins/metabolism , Cell Death/genetics , Cyclic AMP/biosynthesis , Cyclic GMP/biosynthesis , Ligases/metabolism , NAD+ Nucleosidase/metabolism , Plant Diseases , Plant Immunity/physiology , Plant Proteins/metabolism , Receptors, Immunologic/metabolism , Receptors, Interleukin-1/metabolism , Nicotiana/genetics , Nicotiana/metabolism
2.
Cell ; 182(5): 1156-1169.e12, 2020 09 03.
Article in English | MEDLINE | ID: mdl-32795415

ABSTRACT

Dysregulated microglia are intimately involved in neurodegeneration, including Alzheimer's disease (AD) pathogenesis, but the mechanisms controlling pathogenic microglial gene expression remain poorly understood. The transcription factor CCAAT/enhancer binding protein beta (c/EBPß) regulates pro-inflammatory genes in microglia and is upregulated in AD. We show expression of c/EBPß in microglia is regulated post-translationally by the ubiquitin ligase COP1 (also called RFWD2). In the absence of COP1, c/EBPß accumulates rapidly and drives a potent pro-inflammatory and neurodegeneration-related gene program, evidenced by increased neurotoxicity in microglia-neuronal co-cultures. Antibody blocking studies reveal that neurotoxicity is almost entirely attributable to complement. Remarkably, loss of a single allele of Cebpb prevented the pro-inflammatory phenotype. COP1-deficient microglia markedly accelerated tau-mediated neurodegeneration in a mouse model where activated microglia play a deleterious role. Thus, COP1 is an important suppressor of pathogenic c/EBPß-dependent gene expression programs in microglia.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/metabolism , Ligases/metabolism , Microglia/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitin/genetics , Alzheimer Disease/metabolism , Animals , Cell Line , Coculture Techniques/methods , Female , Gene Expression/physiology , Gene Expression Regulation/physiology , HEK293 Cells , Humans , Inflammation/metabolism , Male , Mice , Mice, Inbred C57BL , Neurons/metabolism
3.
Annu Rev Biochem ; 88: 137-162, 2019 06 20.
Article in English | MEDLINE | ID: mdl-31220977

ABSTRACT

Genomic DNA is susceptible to endogenous and environmental stresses that modify DNA structure and its coding potential. Correspondingly, cells have evolved intricate DNA repair systems to deter changes to their genetic material. Base excision DNA repair involves a number of enzymes and protein cofactors that hasten repair of damaged DNA bases. Recent advances have identified macromolecular complexes that assemble at the DNA lesion and mediate repair. The repair of base lesions generally requires five enzymatic activities: glycosylase, endonuclease, lyase, polymerase, and ligase. The protein cofactors and mechanisms for coordinating the sequential enzymatic steps of repair are being revealed through a range of experimental approaches. We discuss the enzymes and protein cofactors involved in eukaryotic base excision repair, emphasizing the challenge of integrating findings from multiple methodologies. The results provide an opportunity to assimilate biochemical findings with cell-based assays to uncover new insights into this deceptively complex repair pathway.


Subject(s)
DNA Glycosylases/chemistry , DNA-Directed DNA Polymerase/chemistry , DNA/chemistry , Endonucleases/chemistry , Genome , Ligases/chemistry , Lyases/chemistry , DNA/metabolism , DNA/ultrastructure , DNA Damage , DNA Glycosylases/metabolism , DNA Glycosylases/ultrastructure , DNA Repair , DNA-Directed DNA Polymerase/metabolism , DNA-Directed DNA Polymerase/ultrastructure , Endonucleases/metabolism , Endonucleases/ultrastructure , Eukaryota/genetics , Eukaryota/metabolism , Eukaryotic Cells/cytology , Eukaryotic Cells/enzymology , Genomic Instability , Humans , Ligases/metabolism , Ligases/ultrastructure , Lyases/metabolism , Lyases/ultrastructure , Models, Molecular , Mutagenesis , Nucleic Acid Conformation , Protein Conformation
4.
Annu Rev Biochem ; 86: 1-19, 2017 06 20.
Article in English | MEDLINE | ID: mdl-28125288

ABSTRACT

After an undergraduate degree in biology at Harvard, I started graduate school at The Rockefeller Institute for Medical Research in New York City in July 1965. I was attracted to the chemical side of biochemistry and joined Fritz Lipmann's large, hierarchical laboratory to study enzyme mechanisms. That work led to postdoctoral research with Robert Abeles at Brandeis, then a center of what, 30 years later, would be called chemical biology. I spent 15 years on the Massachusetts Institute of Technology faculty, in both the Chemistry and Biology Departments, and then 26 years on the Harvard Medical School Faculty. My research interests have been at the intersection of chemistry, biology, and medicine. One unanticipated major focus has been investigating the chemical logic and enzymatic machinery of natural product biosynthesis, including antibiotics and antitumor agents. In this postgenomic era it is now recognized that there may be from 105 to 106 biosynthetic gene clusters as yet uncharacterized for potential new therapeutic agents.


Subject(s)
Anti-Bacterial Agents/metabolism , Antineoplastic Agents/metabolism , Biochemistry/history , Biological Products/metabolism , Biomedical Research/history , Drug Industry/history , Anti-Bacterial Agents/chemistry , Antineoplastic Agents/chemistry , Biochemistry/trends , Biological Products/chemistry , Biomedical Research/trends , Drug Industry/trends , Gene Expression Regulation , History, 20th Century , History, 21st Century , Humans , Ligases/genetics , Ligases/metabolism , Multienzyme Complexes/genetics , Multienzyme Complexes/metabolism , Vancomycin Resistance/genetics , Workforce
5.
Mol Cell ; 83(22): 4123-4140.e12, 2023 Nov 16.
Article in English | MEDLINE | ID: mdl-37848033

ABSTRACT

Purinosomes serve as metabolons to enhance de novo purine synthesis (DNPS) efficiency through compartmentalizing DNPS enzymes during stressed conditions. However, the mechanism underpinning purinosome assembly and its pathophysiological functions remains elusive. Here, we show that K6-polyubiquitination of the DNPS enzyme phosphoribosylaminoimidazole carboxylase and phosphoribosylaminoimidazolesuccinocarboxamide synthetase (PAICS) by cullin-5/ankyrin repeat and SOCS box containing 11 (Cul5/ASB11)-based ubiquitin ligase plays a driving role in purinosome assembly. Upon several purinosome-inducing cues, ASB11 is upregulated by relieving the H3K9me3/HP1α-mediated transcriptional silencing, thus stimulating PAICS polyubiquitination. The polyubiquitinated PAICS recruits ubiquitin-associated protein 2 (UBAP2), a ubiquitin-binding protein with multiple stretches of intrinsically disordered regions, thereby inducing phase separation to trigger purinosome assembly for enhancing DNPS pathway flux. In human melanoma, ASB11 is highly expressed to facilitate a constitutive purinosome formation to which melanoma cells are addicted for supporting their proliferation, viability, and tumorigenesis in a xenograft model. Our study identifies a driving mechanism for purinosome assembly in response to cellular stresses and uncovers the impact of purinosome formation on human malignancies.


Subject(s)
Ligases , Melanoma , Humans , HeLa Cells , Ubiquitination , Ubiquitins
6.
Mol Cell ; 82(17): 3209-3225.e7, 2022 09 01.
Article in English | MEDLINE | ID: mdl-35931083

ABSTRACT

Peroxisomes are ubiquitous organelles whose dysfunction causes fatal human diseases. Most peroxisomal enzymes are imported from the cytosol by the receptor PEX5, which interacts with a docking complex in the peroxisomal membrane and then returns to the cytosol after monoubiquitination by a membrane-embedded ubiquitin ligase. The mechanism by which PEX5 shuttles between cytosol and peroxisomes and releases cargo inside the lumen is unclear. Here, we use Xenopus egg extract to demonstrate that PEX5 accompanies cargo completely into the lumen, utilizing WxxxF/Y motifs near its N terminus that bind a lumenal domain of the docking complex. PEX5 recycling is initiated by an amphipathic helix that binds to the lumenal side of the ubiquitin ligase. The N terminus then emerges in the cytosol for monoubiquitination. Finally, PEX5 is extracted from the lumen, resulting in the unfolding of the receptor and cargo release. Our results reveal the unique mechanism by which PEX5 ferries proteins into peroxisomes.


Subject(s)
Peroxisomes , Receptors, Cytoplasmic and Nuclear , Carrier Proteins/metabolism , Humans , Ligases/metabolism , Peroxisome-Targeting Signal 1 Receptor/genetics , Peroxisome-Targeting Signal 1 Receptor/metabolism , Peroxisomes/chemistry , Protein Transport , Receptors, Cytoplasmic and Nuclear/analysis , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Ubiquitin/metabolism
7.
Mol Cell ; 81(15): 3160-3170.e9, 2021 08 05.
Article in English | MEDLINE | ID: mdl-34174184

ABSTRACT

RelA-SpoT Homolog (RSH) enzymes control bacterial physiology through synthesis and degradation of the nucleotide alarmone (p)ppGpp. We recently discovered multiple families of small alarmone synthetase (SAS) RSH acting as toxins of toxin-antitoxin (TA) modules, with the FaRel subfamily of toxSAS abrogating bacterial growth by producing an analog of (p)ppGpp, (pp)pApp. Here we probe the mechanism of growth arrest used by four experimentally unexplored subfamilies of toxSAS: FaRel2, PhRel, PhRel2, and CapRel. Surprisingly, all these toxins specifically inhibit protein synthesis. To do so, they transfer a pyrophosphate moiety from ATP to the tRNA 3' CCA. The modification inhibits both tRNA aminoacylation and the sensing of cellular amino acid starvation by the ribosome-associated RSH RelA. Conversely, we show that some small alarmone hydrolase (SAH) RSH enzymes can reverse the pyrophosphorylation of tRNA to counter the growth inhibition by toxSAS. Collectively, we establish RSHs as RNA-modifying enzymes.


Subject(s)
Bacterial Toxins/metabolism , Guanosine Pentaphosphate/metabolism , Ligases/metabolism , RNA, Transfer/metabolism , Bacterial Toxins/genetics , Bacterial Toxins/pharmacology , Gram-Positive Asporogenous Rods/chemistry , Gram-Positive Asporogenous Rods/metabolism , Guanosine Pentaphosphate/chemistry , Ligases/chemistry , Ligases/genetics , Phosphorylation/drug effects , Protein Biosynthesis/drug effects , Protein Biosynthesis/physiology , Protein Synthesis Inhibitors/pharmacology , Pyrophosphatases , Ribosomes/metabolism
8.
Annu Rev Biochem ; 82: 387-414, 2013.
Article in English | MEDLINE | ID: mdl-23495935

ABSTRACT

The ubiquitin-proteasome system plays a pivotal role in the sequence of events leading to cell division known as the cell cycle. Not only does ubiquitin-mediated proteolysis constitute a critical component of the core oscillator that drives the cell cycle in all eukaryotes, it is also central to the mechanisms that ensure that the integrity of the genome is maintained. These functions are primarily carried out by two families of E3 ubiquitin ligases, the Skp/cullin/F-box-containing and anaphase-promoting complex/cyclosome complexes. However, beyond those functions associated with regulation of central cell cycle events, many peripheral cell cycle-related processes rely on ubiquitylation for signaling, homeostasis, and dynamicity, involving additional types of ubiquitin ligases and regulators. We are only beginning to understand the diversity and complexity of this regulation.


Subject(s)
Cell Cycle Checkpoints/physiology , Cell Cycle/physiology , Ligases/metabolism , SKP Cullin F-Box Protein Ligases/metabolism , Ubiquitin-Protein Ligase Complexes/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitin/metabolism , Ubiquitination/physiology , Anaphase-Promoting Complex-Cyclosome , Animals , Humans
9.
Nature ; 604(7905): 371-376, 2022 04.
Article in English | MEDLINE | ID: mdl-35388216

ABSTRACT

The outer membrane of Gram-negative bacteria has an external leaflet that is largely composed of lipopolysaccharide, which provides a selective permeation barrier, particularly against antimicrobials1. The final and crucial step in the biosynthesis of lipopolysaccharide is the addition of a species-dependent O-antigen to the lipid A core oligosaccharide, which is catalysed by the O-antigen ligase WaaL2. Here we present structures of WaaL from Cupriavidus metallidurans, both in the apo state and in complex with its lipid carrier undecaprenyl pyrophosphate, determined by single-particle cryo-electron microscopy. The structures reveal that WaaL comprises 12 transmembrane helices and a predominantly α-helical periplasmic region, which we show contains many of the conserved residues that are required for catalysis. We observe a conserved fold within the GT-C family of glycosyltransferases and hypothesize that they have a common mechanism for shuttling the undecaprenyl-based carrier to and from the active site. The structures, combined with genetic, biochemical, bioinformatics and molecular dynamics simulation experiments, offer molecular details on how the ligands come in apposition, and allows us to propose a mechanistic model for catalysis. Together, our work provides a structural basis for lipopolysaccharide maturation in a member of the GT-C superfamily of glycosyltransferases.


Subject(s)
Ligases , Lipopolysaccharides , O Antigens , Bacterial Proteins/chemistry , Carbon-Oxygen Ligases/chemistry , Carbon-Oxygen Ligases/genetics , Cryoelectron Microscopy , Glycosyltransferases , Gram-Negative Bacteria , Lipopolysaccharides/chemistry , Lipopolysaccharides/metabolism
10.
Mol Cell ; 77(3): 571-585.e4, 2020 02 06.
Article in English | MEDLINE | ID: mdl-31901448

ABSTRACT

Сhromatin is critical for genome compaction and gene expression. On a coarse scale, the genome is divided into euchromatin, which harbors the majority of genes and is enriched in active chromatin marks, and heterochromatin, which is gene-poor but repeat-rich. The conserved molecular hallmark of heterochromatin is the H3K9me3 modification, which is associated with gene silencing. We found that in Drosophila, deposition of most of the H3K9me3 mark depends on SUMO and the SUMO ligase Su(var)2-10, which recruits the histone methyltransferase complex SetDB1/Wde. In addition to repressing repeats, H3K9me3 influences expression of both hetero- and euchromatic host genes. High H3K9me3 levels in heterochromatin are required to suppress spurious transcription and ensure proper gene expression. In euchromatin, a set of conserved genes is repressed by Su(var)2-10/SetDB1-induced H3K9 trimethylation, ensuring tissue-specific gene expression. Several components of heterochromatin are themselves repressed by this pathway, providing a negative feedback mechanism to ensure chromatin homeostasis.


Subject(s)
Drosophila Proteins/metabolism , Gene Expression Regulation/genetics , Protein Inhibitors of Activated STAT/metabolism , Small Ubiquitin-Related Modifier Proteins/metabolism , Animals , Chromosomal Proteins, Non-Histone/metabolism , DNA Methylation/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Euchromatin/metabolism , Feedback, Physiological , Gene Expression/genetics , Gene Silencing/physiology , Heterochromatin/genetics , Heterochromatin/metabolism , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Histones/genetics , Histones/metabolism , Ligases/genetics , Methyltransferases/genetics , Protein Inhibitors of Activated STAT/genetics , Repressor Proteins/metabolism , Small Ubiquitin-Related Modifier Proteins/genetics
11.
EMBO J ; 42(4): e111549, 2023 02 15.
Article in English | MEDLINE | ID: mdl-36598329

ABSTRACT

YAP/TAZ transcriptional co-activators play pivotal roles in tumorigenesis. In the Hippo pathway, diverse signals activate the MST-LATS kinase cascade that leads to YAP/TAZ phosphorylation, and subsequent ubiquitination and proteasomal degradation by SCFß-TrCP . When the MST-LATS kinase cascade is inactive, unphosphorylated or dephosphorylated YAP/TAZ translocate into the nucleus to mediate TEAD-dependent gene transcription. Hippo signaling-independent YAP/TAZ activation in human malignancies has also been observed, yet the mechanism remains largely elusive. Here, we report that the ubiquitin E3 ligase HERC3 can promote YAP/TAZ activation independently of its enzymatic activity. HERC3 directly binds to ß-TrCP, blocks its interaction with YAP/TAZ, and thus prevents YAP/TAZ ubiquitination and degradation. Expression levels of HERC3 correlate with YAP/TAZ protein levels and expression of YAP/TAZ target genes in breast tumor cells and tissues. Accordingly, knockdown of HERC3 expression ameliorates tumorigenesis of breast cancer cells. Our results establish HERC3 as a critical regulator of the YAP/TAZ stability and a potential therapeutic target for breast cancer.


Subject(s)
Adaptor Proteins, Signal Transducing , Breast Neoplasms , Humans , Female , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , YAP-Signaling Proteins , beta-Transducin Repeat-Containing Proteins/genetics , beta-Transducin Repeat-Containing Proteins/metabolism , Transcriptional Coactivator with PDZ-Binding Motif Proteins , Transcription Factors/genetics , Transcription Factors/metabolism , Cell Transformation, Neoplastic/genetics , Carcinogenesis/genetics , Ubiquitination , Breast Neoplasms/genetics , Ubiquitins/metabolism , Ligases/genetics , Phosphoproteins/genetics , Phosphoproteins/metabolism
12.
Nature ; 593(7859): 391-398, 2021 05.
Article in English | MEDLINE | ID: mdl-34012085

ABSTRACT

Coronatine and related bacterial phytotoxins are mimics of the hormone jasmonyl-L-isoleucine (JA-Ile), which mediates physiologically important plant signalling pathways1-4. Coronatine-like phytotoxins disrupt these essential pathways and have potential in the development of safer, more selective herbicides. Although the biosynthesis of coronatine has been investigated previously, the nature of the enzyme that catalyses the crucial coupling of coronafacic acid to amino acids remains unknown1,2. Here we characterize a family of enzymes, coronafacic acid ligases (CfaLs), and resolve their structures. We found that CfaL can also produce JA-Ile, despite low similarity with the Jar1 enzyme that is responsible for ligation of JA and L-Ile in plants5. This suggests that Jar1 and CfaL evolved independently to catalyse similar reactions-Jar1 producing a compound essential for plant development4,5, and the bacterial ligases producing analogues toxic to plants. We further demonstrate how CfaL enzymes can be used to synthesize a diverse array of amides, obviating the need for protecting groups. Highly selective kinetic resolutions of racemic donor or acceptor substrates were achieved, affording homochiral products. We also used structure-guided mutagenesis to engineer improved CfaL variants. Together, these results show that CfaLs can deliver a wide range of amides for agrochemical, pharmaceutical and other applications.


Subject(s)
Amides/metabolism , Ligases/chemistry , Ligases/metabolism , Amides/chemistry , Amino Acids/biosynthesis , Amino Acids/chemistry , Azospirillum lipoferum/enzymology , Azospirillum lipoferum/genetics , Carboxylic Acids/metabolism , Cyclopentanes/chemistry , Escherichia coli/genetics , Escherichia coli/metabolism , Herbicides/chemistry , Herbicides/metabolism , Indenes/chemistry , Isoleucine/analogs & derivatives , Isoleucine/biosynthesis , Isoleucine/chemistry , Kinetics , Models, Molecular , Pectobacterium/enzymology , Pectobacterium/genetics , Pseudomonas syringae/enzymology , Pseudomonas syringae/genetics
13.
Nature ; 596(7872): 398-403, 2021 08.
Article in English | MEDLINE | ID: mdl-34349258

ABSTRACT

One in four women suffers from uterine leiomyomas (ULs)-benign tumours of the uterine wall, also known as uterine fibroids-at some point in premenopausal life. ULs can cause excessive bleeding, pain and infertility1, and are a common cause of hysterectomy2. They emerge through at least three distinct genetic drivers: mutations in MED12 or FH, or genomic rearrangement of HMGA23. Here we created genome-wide datasets, using DNA, RNA, assay for transposase-accessible chromatin (ATAC), chromatin immunoprecipitation (ChIP) and HiC chromatin immunoprecipitation (HiChIP) sequencing of primary tissues to profoundly understand the genesis of UL. We identified somatic mutations in genes encoding six members of the SRCAP histone-loading complex4, and found that germline mutations in the SRCAP members YEATS4 and ZNHIT1 predispose women to UL. Tumours bearing these mutations showed defective deposition of the histone variant H2A.Z. In ULs, H2A.Z occupancy correlated positively with chromatin accessibility and gene expression, and negatively with DNA methylation, but these correlations were weak in tumours bearing SRCAP complex mutations. In these tumours, open chromatin emerged at transcription start sites where H2A.Z was lost, which was associated with upregulation of genes. Furthermore, YEATS4 defects were associated with abnormal upregulation of bivalent embryonic stem cell genes, as previously shown in mice5. Our work describes a potential mechanism of tumorigenesis-epigenetic instability caused by deficient H2A.Z deposition-and suggests that ULs arise through an aberrant differentiation program driven by deranged chromatin, emanating from a small number of mutually exclusive driver mutations.


Subject(s)
Chromatin Assembly and Disassembly , Chromatin/genetics , Chromatin/metabolism , Histones/deficiency , Leiomyoma/genetics , Mutation , Uterine Neoplasms/genetics , Carcinogenesis/genetics , Cell Line , Chromatin/chemistry , Embryonic Stem Cells/metabolism , Epigenesis, Genetic , Female , Gene Expression Regulation, Neoplastic , Histones/genetics , Histones/metabolism , Humans , Leiomyoma/metabolism , Leiomyoma/pathology , Ligases/genetics , Polycomb Repressive Complex 1/genetics , Polycomb-Group Proteins/genetics , Transcription Factors/genetics , Uterine Neoplasms/metabolism , Uterine Neoplasms/pathology
14.
Mol Cell ; 74(6): 1227-1238.e3, 2019 06 20.
Article in English | MEDLINE | ID: mdl-31003868

ABSTRACT

rRNAs and tRNAs universally require processing from longer primary transcripts to become functional for translation. Here, we describe an unsuspected link between tRNA maturation and the 3' processing of 16S rRNA, a key step in preparing the small ribosomal subunit for interaction with the Shine-Dalgarno sequence in prokaryotic translation initiation. We show that an accumulation of either 5' or 3' immature tRNAs triggers RelA-dependent production of the stringent response alarmone (p)ppGpp in the Gram-positive model organism Bacillus subtilis. The accumulation of (p)ppGpp and accompanying decrease in GTP levels specifically inhibit 16S rRNA 3' maturation. We suggest that cells can exploit this mechanism to sense potential slowdowns in tRNA maturation and adjust rRNA processing accordingly to maintain the appropriate functional balance between these two major components of the translation apparatus.


Subject(s)
Bacillus subtilis/genetics , Gene Expression Regulation, Bacterial , Guanosine Pentaphosphate/biosynthesis , Peptide Chain Initiation, Translational , RNA, Ribosomal, 16S/genetics , RNA, Transfer/genetics , Bacillus subtilis/metabolism , Base Sequence , Guanosine Pentaphosphate/genetics , Guanosine Triphosphate/metabolism , Ligases/genetics , Ligases/metabolism , Nucleic Acid Conformation , RNA, Ribosomal, 16S/chemistry , RNA, Ribosomal, 16S/metabolism , RNA, Transfer/chemistry , RNA, Transfer/metabolism , Ribosome Subunits, Large, Bacterial/genetics , Ribosome Subunits, Large, Bacterial/metabolism , Ribosome Subunits, Small, Bacterial/genetics , Ribosome Subunits, Small, Bacterial/metabolism
15.
Mol Cell ; 76(1): 11-26.e7, 2019 10 03.
Article in English | MEDLINE | ID: mdl-31400850

ABSTRACT

Alternative lengthening of telomeres (ALT) is a homology-directed repair (HDR) mechanism of telomere elongation that controls proliferation in aggressive cancers. We show that the disruption of RAD51-associated protein 1 (RAD51AP1) in ALT+ cancer cells leads to generational telomere shortening. This is due to RAD51AP1's involvement in RAD51-dependent homologous recombination (HR) and RAD52-POLD3-dependent break induced DNA synthesis. RAD51AP1 KO ALT+ cells exhibit telomere dysfunction and cytosolic telomeric DNA fragments that are sensed by cGAS. Intriguingly, they activate ULK1-ATG7-dependent autophagy as a survival mechanism to mitigate DNA damage and apoptosis. Importantly, RAD51AP1 protein levels are elevated in ALT+ cells due to MMS21 associated SUMOylation. Mutation of a single SUMO-targeted lysine residue perturbs telomere dynamics. These findings indicate that RAD51AP1 is an essential mediator of the ALT mechanism and is co-opted by post-translational mechanisms to maintain telomere length and ensure proliferation of ALT+ cancer cells.


Subject(s)
DNA-Binding Proteins/metabolism , Neoplasms/metabolism , RNA-Binding Proteins/metabolism , Telomere Homeostasis , Telomere/metabolism , Autophagy , Autophagy-Related Protein 7/genetics , Autophagy-Related Protein 7/metabolism , Autophagy-Related Protein-1 Homolog/genetics , Autophagy-Related Protein-1 Homolog/metabolism , Cell Proliferation , DNA Polymerase III/genetics , DNA Polymerase III/metabolism , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , HEK293 Cells , HeLa Cells , Homologous Recombination , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Ligases/genetics , Ligases/metabolism , Lysine , Neoplasms/genetics , Neoplasms/pathology , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , Protein Stability , RNA-Binding Proteins/genetics , Rad52 DNA Repair and Recombination Protein/genetics , Rad52 DNA Repair and Recombination Protein/metabolism , Signal Transduction , Sumoylation , Telomere/genetics , Telomere/pathology
16.
Proc Natl Acad Sci U S A ; 121(16): e2401313121, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38602916

ABSTRACT

All forms of life are presumed to synthesize arginine from citrulline via a two-step pathway consisting of argininosuccinate synthetase and argininosuccinate lyase using citrulline, adenosine 5'-triphosphate (ATP), and aspartate as substrates. Conversion of arginine to citrulline predominantly proceeds via hydrolysis. Here, from the hyperthermophilic archaeon Thermococcus kodakarensis, we identified an enzyme which we designate "arginine synthetase". In arginine synthesis, the enzyme converts citrulline, ATP, and free ammonia to arginine, adenosine 5'-diphosphate (ADP), and phosphate. In the reverse direction, arginine synthetase conserves the energy of arginine deimination and generates ATP from ADP and phosphate while releasing ammonia. The equilibrium constant of this reaction at pH 7.0 is [Cit][ATP][NH3]/[Arg][ADP][Pi] = 10.1 ± 0.7 at 80 °C, corresponding to a ΔG°' of -6.8 ± 0.2 kJ mol-1. Growth of the gene disruption strain was compared to the host strain in medium composed of amino acids. The results suggested that arginine synthetase is necessary in providing ornithine, the precursor for proline biosynthesis, as well as in generating ATP. Growth in medium supplemented with citrulline indicated that arginine synthetase can function in the direction of arginine synthesis. The enzyme is widespread in nature, including bacteria and eukaryotes, and catalyzes a long-overlooked energy-conserving reaction in microbial amino acid metabolism. Along with ornithine transcarbamoylase and carbamate kinase, the pathway identified here is designated the arginine synthetase pathway.


Subject(s)
Arginine , Ligases , Arginine/metabolism , Citrulline/metabolism , Ammonia , Ornithine/genetics , Adenosine Triphosphate/metabolism , Phosphates , Adenosine , Catalysis
17.
Proc Natl Acad Sci U S A ; 121(25): e2320995121, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38865271

ABSTRACT

Meiosis, a reductional cell division, relies on precise initiation, maturation, and resolution of crossovers (COs) during prophase I to ensure the accurate segregation of homologous chromosomes during metaphase I. This process is regulated by the interplay of RING-E3 ligases such as RNF212 and HEI10 in mammals. In this study, we functionally characterized a recently identified RING-E3 ligase, RNF212B. RNF212B colocalizes and interacts with RNF212, forming foci along chromosomes from zygonema onward in a synapsis-dependent and DSB-independent manner. These consolidate into larger foci at maturing COs, colocalizing with HEI10, CNTD1, and MLH1 by late pachynema. Genetically, RNF212B foci formation depends on Rnf212 but not on Msh4, Hei10, and Cntd1, while the unloading of RNF212B at the end of pachynema is dependent on Hei10 and Cntd1. Mice lacking RNF212B, or expressing an inactive RNF212B protein, exhibit modest synapsis defects, a reduction in the localization of pro-CO factors (MSH4, TEX11, RPA, MZIP2) and absence of late CO-intermediates (MLH1). This loss of most COs by diakinesis results in mostly univalent chromosomes. Double mutants for Rnf212b and Rnf212 exhibit an identical phenotype to that of Rnf212b single mutants, while double heterozygous demonstrate a dosage-dependent reduction in CO number, indicating a functional interplay between paralogs. SUMOylome analysis of testes from Rnf212b mutants and pull-down analysis of Sumo- and Ubiquitin-tagged HeLa cells, suggest that RNF212B is an E3-ligase with Ubiquitin activity, serving as a crucial factor for CO maturation. Thus, RNF212 and RNF212B play vital, yet overlapping roles, in ensuring CO homeostasis through their distinct E3 ligase activities.


Subject(s)
Chromosome Pairing , Crossing Over, Genetic , Meiosis , Ubiquitin-Protein Ligases , Animals , Mice , Male , Female , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Poly-ADP-Ribose Binding Proteins/metabolism , Poly-ADP-Ribose Binding Proteins/genetics , Mice, Knockout , Humans , Ligases
18.
Proc Natl Acad Sci U S A ; 121(16): e2317978121, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38593069

ABSTRACT

Mosquito-borne flaviviruses such as dengue (DENV) and Zika (ZIKV) cause hundreds of millions of infections annually. The single-stranded RNA genome of flaviviruses is translated into a polyprotein, which is cleaved equally into individual functional proteins. While structural proteins are packaged into progeny virions and released, most of the nonstructural proteins remain intracellular and could become cytotoxic if accumulated over time. However, the mechanism by which nonstructural proteins are maintained at the levels optimal for cellular fitness and viral replication remains unknown. Here, we identified that the ubiquitin E3 ligase HRD1 is essential for flaviviruses infections in both mammalian hosts and mosquitoes. HRD1 directly interacts with flavivirus NS4A and ubiquitylates a conserved lysine residue for ER-associated degradation. This mechanism avoids excessive accumulation of NS4A, which otherwise interrupts the expression of processed flavivirus proteins in the ER. Furthermore, a small-molecule inhibitor of HRD1 named LS-102 effectively interrupts DENV2 infection in both mice and Aedes aegypti mosquitoes, and significantly disturbs DENV transmission from the infected hosts to mosquitoes owing to reduced viremia. Taken together, this study demonstrates that flaviviruses have evolved a sophisticated mechanism to exploit the ubiquitination system to balance the homeostasis of viral proteins for their own advantage and provides a potential therapeutic target to interrupt flavivirus infection and transmission.


Subject(s)
Aedes , Flavivirus Infections , Flavivirus , Zika Virus Infection , Zika Virus , Animals , Mice , Flavivirus/genetics , Zika Virus/genetics , Ubiquitin/metabolism , Ligases/metabolism , Viral Proteins/metabolism , Mammals
19.
Development ; 150(18)2023 Sep 15.
Article in English | MEDLINE | ID: mdl-37756604

ABSTRACT

KIF5C is a kinesin-1 heavy chain that has been associated with neurodevelopmental disorders. Although the roles of kinesin-1 in axon transport are well known, little is known about how it regulates axon targeting. We report that UNC-116/KIF5C functions with the NEKL-3/NEK6/7 kinase to promote axon targeting in Caenorhabditis elegans. Loss of UNC-116 causes the axon to overshoot its target and UNC-116 gain-of-function causes premature axon termination. We find that loss of the UNC-16/JIP3 kinesin-1 cargo adaptor disrupts axon termination, but loss of kinesin-1 light chain function does not affect axon termination. Genetic analysis indicates that UNC-16 functions with the NEKL-3 kinase to promote axon termination. Consistent with this observation, imaging experiments indicate that loss of UNC-16 and UNC-116 disrupt localization of NEKL-3 in the axon. Moreover, genetic interactions suggest that NEKL-3 promotes axon termination by functioning with RPM-1, a ubiquitin ligase that regulates microtubule stability in the growth cone. These observations support a model where UNC-116 functions with UNC-16 to promote localization of NEKL-3 in the axon. NEKL-3, in turn, functions with the RPM-1 ubiquitin ligase to promote axon termination.


Subject(s)
Caenorhabditis elegans Proteins , Animals , Caenorhabditis elegans Proteins/genetics , Kinesins/genetics , Axons/physiology , Caenorhabditis elegans , Ligases , Ubiquitins , Guanine Nucleotide Exchange Factors/genetics
20.
RNA ; 30(4): 367-380, 2024 Mar 18.
Article in English | MEDLINE | ID: mdl-38238085

ABSTRACT

Fungal Trl1 is an essential trifunctional tRNA splicing enzyme that heals and seals tRNA exons with 2',3'-cyclic-PO4 and 5'-OH ends. Trl1 is composed of C-terminal cyclic phosphodiesterase and central polynucleotide kinase end-healing domains that generate the 3'-OH,2'-PO4 and 5'-PO4 termini required for sealing by an N-terminal ATP-dependent ligase domain. Trl1 enzymes are present in many human fungal pathogens and are promising targets for antifungal drug discovery because their domain structures and biochemical mechanisms are unique compared to the mammalian RtcB-type tRNA splicing enzyme. Here we report that Mucorales species (deemed high-priority human pathogens by WHO) elaborate a noncanonical tRNA splicing apparatus in which a monofunctional RNA ligase enzyme is encoded separately from any end-healing enzymes. We show that Mucor circinelloides RNA ligase (MciRNL) is active in tRNA splicing in vivo in budding yeast in lieu of the Trl1 ligase domain. Biochemical and kinetic characterization of recombinant MciRNL underscores its requirement for a 2'-PO4 terminus in the end-joining reaction, whereby the 2'-PO4 enhances the rates of RNA 5'-adenylylation (step 2) and phosphodiester synthesis (step 3) by ∼125-fold and ∼6200-fold, respectively. In the canonical fungal tRNA splicing pathway, the splice junction 2'-PO4 installed by RNA ligase is removed by a dedicated NAD+-dependent RNA 2'-phosphotransferase Tpt1. Here we identify and affirm by genetic complementation in yeast the biological activity of Tpt1 orthologs from three Mucorales species. Recombinant M. circinelloides Tpt1 has vigorous NAD+-dependent RNA 2'-phosphotransferase activity in vitro.


Subject(s)
Mucorales , Animals , Humans , Mucorales/genetics , Mucorales/metabolism , NAD/metabolism , RNA/genetics , RNA, Transfer/genetics , RNA, Transfer/metabolism , RNA Ligase (ATP)/genetics , RNA Ligase (ATP)/metabolism , Saccharomyces cerevisiae/metabolism , Ligases , Polynucleotide 5'-Hydroxyl-Kinase/chemistry , RNA Splicing , Mammals/genetics
SELECTION OF CITATIONS
SEARCH DETAIL