Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.406
Filter
Add more filters

Publication year range
1.
Cell ; 187(9): 2250-2268.e31, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38554706

ABSTRACT

Ubiquitin-dependent unfolding of the CMG helicase by VCP/p97 is required to terminate DNA replication. Other replisome components are not processed in the same fashion, suggesting that additional mechanisms underlie replication protein turnover. Here, we identify replisome factor interactions with a protein complex composed of AAA+ ATPases SPATA5-SPATA5L1 together with heterodimeric partners C1orf109-CINP (55LCC). An integrative structural biology approach revealed a molecular architecture of SPATA5-SPATA5L1 N-terminal domains interacting with C1orf109-CINP to form a funnel-like structure above a cylindrically shaped ATPase motor. Deficiency in the 55LCC complex elicited ubiquitin-independent proteotoxicity, replication stress, and severe chromosome instability. 55LCC showed ATPase activity that was specifically enhanced by replication fork DNA and was coupled to cysteine protease-dependent cleavage of replisome substrates in response to replication fork damage. These findings define 55LCC-mediated proteostasis as critical for replication fork progression and genome stability and provide a rationale for pathogenic variants seen in associated human neurodevelopmental disorders.


Subject(s)
Adenosine Triphosphatases , DNA Replication , Genomic Instability , Proteostasis , Humans , Adenosine Triphosphatases/metabolism , Valosin Containing Protein/metabolism , Valosin Containing Protein/genetics , HEK293 Cells , Cell Cycle Proteins/metabolism , ATPases Associated with Diverse Cellular Activities/metabolism , ATPases Associated with Diverse Cellular Activities/genetics
2.
Cell ; 186(5): 999-1012.e20, 2023 03 02.
Article in English | MEDLINE | ID: mdl-36764292

ABSTRACT

Adenosine-to-inosine RNA editing has been proposed to be involved in a bacterial anti-phage defense system called RADAR. RADAR contains an adenosine triphosphatase (RdrA) and an adenosine deaminase (RdrB). Here, we report cryo-EM structures of RdrA, RdrB, and currently identified RdrA-RdrB complexes in the presence or absence of RNA and ATP. RdrB assembles into a dodecameric cage with catalytic pockets facing outward, while RdrA adopts both autoinhibited tetradecameric and activation-competent heptameric rings. Structural and functional data suggest a model in which RNA is loaded through the bottom section of the RdrA ring and translocated along its inner channel, a process likely coupled with ATP-binding status. Intriguingly, up to twelve RdrA rings can dock one RdrB cage with precise alignments between deaminase catalytic pockets and RNA-translocation channels, indicative of enzymatic coupling of RNA translocation and deamination. Our data uncover an interesting mechanism of enzymatic coupling and anti-phage defense through supramolecular assemblies.


Subject(s)
Adenosine Triphosphate , RNA , Adenosine Deaminase/genetics
3.
Annu Rev Biochem ; 90: 77-106, 2021 06 20.
Article in English | MEDLINE | ID: mdl-33784179

ABSTRACT

The faithful and timely copying of DNA by molecular machines known as replisomes depends on a disparate suite of enzymes and scaffolding factors working together in a highly orchestrated manner. Large, dynamic protein-nucleic acid assemblies that selectively morph between distinct conformations and compositional states underpin this critical cellular process. In this article, we discuss recent progress outlining the physical basis of replisome construction and progression in eukaryotes.


Subject(s)
DNA Replication , DNA/biosynthesis , Eukaryota/genetics , Origin Recognition Complex/metabolism , Animals , DNA/chemistry , DNA Polymerase III/chemistry , DNA Polymerase III/metabolism , Humans , Origin Recognition Complex/chemistry , Origin Recognition Complex/genetics , Proliferating Cell Nuclear Antigen/chemistry , Proliferating Cell Nuclear Antigen/metabolism
4.
Annu Rev Biochem ; 89: 583-603, 2020 06 20.
Article in English | MEDLINE | ID: mdl-31874046

ABSTRACT

P-type ATPases are found in all kingdoms of life and constitute a wide range of cation transporters, primarily for H+, Na+, K+, Ca2+, and transition metal ions such as Cu(I), Zn(II), and Cd(II). They have been studied through a wide range of techniques, and research has gained very significant insight on their transport mechanism and regulation. Here, we review the structure, function, and dynamics of P2-ATPases including Ca2+-ATPases and Na,K-ATPase. We highlight mechanisms of functional transitions that are associated with ion exchange on either side of the membrane and how the functional cycle is regulated by interaction partners, autoregulatory domains, and off-cycle states. Finally, we discuss future perspectives based on emerging techniques and insights.


Subject(s)
Adenosine Triphosphate/chemistry , Copper-Transporting ATPases/chemistry , H(+)-K(+)-Exchanging ATPase/chemistry , Sarcoplasmic Reticulum Calcium-Transporting ATPases/chemistry , Sodium-Potassium-Exchanging ATPase/chemistry , Adenosine Triphosphate/metabolism , Animals , Binding Sites , Cations, Divalent , Cations, Monovalent , Copper-Transporting ATPases/genetics , Copper-Transporting ATPases/metabolism , H(+)-K(+)-Exchanging ATPase/genetics , H(+)-K(+)-Exchanging ATPase/metabolism , Humans , Ion Transport , Models, Molecular , Protein Binding , Protein Interaction Domains and Motifs , Protein Structure, Secondary , Protons , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Single Molecule Imaging , Sodium-Potassium-Exchanging ATPase/genetics , Sodium-Potassium-Exchanging ATPase/metabolism , Substrate Specificity
5.
Cell ; 180(2): 296-310.e18, 2020 01 23.
Article in English | MEDLINE | ID: mdl-31978346

ABSTRACT

Mitochondria and lysosomes are functionally linked, and their interdependent decline is a hallmark of aging and disease. Despite the long-standing connection between these organelles, the function(s) of lysosomes required to sustain mitochondrial health remains unclear. Here, working in yeast, we show that the lysosome-like vacuole maintains mitochondrial respiration by spatially compartmentalizing amino acids. Defects in vacuole function result in a breakdown in intracellular amino acid homeostasis, which drives age-related mitochondrial decline. Among amino acids, we find that cysteine is most toxic for mitochondria and show that elevated non-vacuolar cysteine impairs mitochondrial respiration by limiting intracellular iron availability through an oxidant-based mechanism. Cysteine depletion or iron supplementation restores mitochondrial health in vacuole-impaired cells and prevents mitochondrial decline during aging. These results demonstrate that cysteine toxicity is a major driver of age-related mitochondrial deterioration and identify vacuolar amino acid compartmentation as a cellular strategy to minimize amino acid toxicity.


Subject(s)
Cysteine/toxicity , Iron/metabolism , Mitochondria/metabolism , Amino Acids/metabolism , Cellular Senescence/physiology , Cysteine/metabolism , Homeostasis , Lysosomes/metabolism , Mitochondria/physiology , Mitophagy/physiology , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Vacuolar Proton-Translocating ATPases/metabolism , Vacuoles/metabolism
6.
Cell ; 178(3): 552-566.e20, 2019 07 25.
Article in English | MEDLINE | ID: mdl-31327526

ABSTRACT

Antibacterial autophagy (xenophagy) is an important host defense, but how it is initiated is unclear. Here, we performed a bacterial transposon screen and identified a T3SS effector SopF that potently blocked Salmonella autophagy. SopF was a general xenophagy inhibitor without affecting canonical autophagy. S. Typhimurium ΔsopF resembled S. flexneri ΔvirAΔicsB with the majority of intracellular bacteria targeted by autophagy, permitting a CRISPR screen that identified host V-ATPase as an essential factor. Upon bacteria-caused vacuolar damage, the V-ATPase recruited ATG16L1 onto bacteria-containing vacuole, which was blocked by SopF. Mammalian ATG16L1 bears a WD40 domain required for interacting with the V-ATPase. Inhibiting autophagy by SopF promoted S. Typhimurium proliferation in vivo. SopF targeted Gln124 of ATP6V0C in the V-ATPase for ADP-ribosylation. Mutation of Gln124 also blocked xenophagy, but not canonical autophagy. Thus, the discovery of SopF reveals the V-ATPase-ATG16L1 axis that critically mediates autophagic recognition of intracellular pathogen.


Subject(s)
Autophagy-Related Proteins/metabolism , Bacterial Proteins/genetics , Macroautophagy , Salmonella/metabolism , Vacuolar Proton-Translocating ATPases/metabolism , Virulence Factors/genetics , ADP-Ribosylation , Autophagy-Related Proteins/deficiency , Autophagy-Related Proteins/genetics , Bacterial Proteins/metabolism , CRISPR-Cas Systems/genetics , Gene Editing , HeLa Cells , Humans , Microtubule-Associated Proteins/metabolism , Protein Binding , Salmonella/pathogenicity , Type III Secretion Systems/metabolism , Vacuolar Proton-Translocating ATPases/genetics , Virulence Factors/metabolism
7.
Cell ; 177(2): 339-351.e13, 2019 04 04.
Article in English | MEDLINE | ID: mdl-30879786

ABSTRACT

Pre-mRNA splicing is executed by the spliceosome. Structural characterization of the catalytically activated complex (B∗) is pivotal for understanding the branching reaction. In this study, we assembled the B∗ complexes on two different pre-mRNAs from Saccharomyces cerevisiae and determined the cryo-EM structures of four distinct B∗ complexes at overall resolutions of 2.9-3.8 Å. The duplex between U2 small nuclear RNA (snRNA) and the branch point sequence (BPS) is discretely away from the 5'-splice site (5'SS) in the three B∗ complexes that are devoid of the step I splicing factors Yju2 and Cwc25. Recruitment of Yju2 into the active site brings the U2/BPS duplex into the vicinity of 5'SS, with the BPS nucleophile positioned 4 Å away from the catalytic metal M2. This analysis reveals the functional mechanism of Yju2 and Cwc25 in branching. These structures on different pre-mRNAs reveal substrate-specific conformations of the spliceosome in a major functional state.


Subject(s)
Spliceosomes/physiology , Spliceosomes/ultrastructure , Catalytic Domain/physiology , Cryoelectron Microscopy/methods , Exons , Introns , Nuclear Proteins/metabolism , RNA Precursors/metabolism , RNA Splice Sites/genetics , RNA Splicing/physiology , RNA Splicing Factors/metabolism , RNA, Small Nuclear/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/metabolism , Spliceosomes/metabolism
8.
Annu Rev Biochem ; 87: 697-724, 2018 06 20.
Article in English | MEDLINE | ID: mdl-29652515

ABSTRACT

As the endpoint for the ubiquitin-proteasome system, the 26S proteasome is the principal proteolytic machine responsible for regulated protein degradation in eukaryotic cells. The proteasome's cellular functions range from general protein homeostasis and stress response to the control of vital processes such as cell division and signal transduction. To reliably process all the proteins presented to it in the complex cellular environment, the proteasome must combine high promiscuity with exceptional substrate selectivity. Recent structural and biochemical studies have shed new light on the many steps involved in proteasomal substrate processing, including recognition, deubiquitination, and ATP-driven translocation and unfolding. In addition, these studies revealed a complex conformational landscape that ensures proper substrate selection before the proteasome commits to processive degradation. These advances in our understanding of the proteasome's intricate machinery set the stage for future studies on how the proteasome functions as a major regulator of the eukaryotic proteome.


Subject(s)
Proteasome Endopeptidase Complex/chemistry , Proteasome Endopeptidase Complex/metabolism , ATPases Associated with Diverse Cellular Activities/chemistry , ATPases Associated with Diverse Cellular Activities/metabolism , Deubiquitinating Enzymes/chemistry , Deubiquitinating Enzymes/metabolism , Humans , Models, Biological , Models, Molecular , Molecular Motor Proteins/chemistry , Molecular Motor Proteins/metabolism , Protein Conformation , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Substrate Specificity , Ubiquitin/chemistry , Ubiquitin/metabolism
9.
Annu Rev Biochem ; 87: 263-294, 2018 06 20.
Article in English | MEDLINE | ID: mdl-29709199

ABSTRACT

Genomic instability in disease and its fidelity in health depend on the DNA damage response (DDR), regulated in part from the complex of meiotic recombination 11 homolog 1 (MRE11), ATP-binding cassette-ATPase (RAD50), and phosphopeptide-binding Nijmegen breakage syndrome protein 1 (NBS1). The MRE11-RAD50-NBS1 (MRN) complex forms a multifunctional DDR machine. Within its network assemblies, MRN is the core conductor for the initial and sustained responses to DNA double-strand breaks, stalled replication forks, dysfunctional telomeres, and viral DNA infection. MRN can interfere with cancer therapy and is an attractive target for precision medicine. Its conformations change the paradigm whereby kinases initiate damage sensing. Delineated results reveal kinase activation, posttranslational targeting, functional scaffolding, conformations storing binding energy and enabling access, interactions with hub proteins such as replication protein A (RPA), and distinct networks at DNA breaks and forks. MRN biochemistry provides prototypic insights into how it initiates, implements, and regulates multifunctional responses to genomic stress.


Subject(s)
DNA Damage , DNA Repair , DNA Replication , MRE11 Homologue Protein/metabolism , DNA Repair Enzymes/chemistry , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Humans , Immunity, Innate , MRE11 Homologue Protein/chemistry , MRE11 Homologue Protein/genetics , Models, Biological , Models, Molecular , Signal Transduction , Telomere/metabolism
10.
Immunity ; 56(5): 926-943.e7, 2023 05 09.
Article in English | MEDLINE | ID: mdl-36948192

ABSTRACT

NOD-like receptors (NLRs) are pattern recognition receptors for diverse innate immune responses. Self-oligomerization after engagement with a ligand is a generally accepted model for the activation of each NLR. We report here that a catalyzer was required for NLR self-oligomerization. PELO, a well-known surveillance factor in translational quality control and/or ribosome rescue, interacted with all cytosolic NLRs and activated their ATPase activity. In the case of flagellin-initiated NLRC4 inflammasome activation, flagellin-bound NAIP5 recruited the first NLRC4 and then PELO was required for correctly assembling the rest of NLRC4s into the NLRC4 complex, one by one, by activating the NLRC4 ATPase activity. Stoichiometric and functional data revealed that PELO was not a structural constituent of the NLRC4 inflammasome but a powerful catalyzer for its assembly. The catalytic role of PELO in the activation of cytosolic NLRs provides insight into NLR activation and provides a direction for future studies of NLR family members.


Subject(s)
Apoptosis Regulatory Proteins , Inflammasomes , Adenosine Triphosphatases/metabolism , Apoptosis Regulatory Proteins/metabolism , Calcium-Binding Proteins/chemistry , Calcium-Binding Proteins/metabolism , Flagellin/metabolism , Inflammasomes/metabolism , Neuronal Apoptosis-Inhibitory Protein/chemistry , Neuronal Apoptosis-Inhibitory Protein/metabolism , NLR Proteins/metabolism
11.
Cell ; 169(4): 722-735.e9, 2017 05 04.
Article in English | MEDLINE | ID: mdl-28475898

ABSTRACT

The Cdc48 ATPase and its cofactors Ufd1/Npl4 (UN) extract polyubiquitinated proteins from membranes or macromolecular complexes, but how they perform these functions is unclear. Cdc48 consists of an N-terminal domain that binds UN and two stacked hexameric ATPase rings (D1 and D2) surrounding a central pore. Here, we use purified components to elucidate how the Cdc48 complex processes substrates. After interaction of the polyubiquitin chain with UN, ATP hydrolysis by the D2 ring moves the polypeptide completely through the double ring, generating a pulling force on the substrate and causing its unfolding. ATP hydrolysis by the D1 ring is important for subsequent substrate release from the Cdc48 complex. This release requires cooperation of Cdc48 with a deubiquitinase, which trims polyubiquitin to an oligoubiquitin chain that is then also translocated through the pore. Together, these results lead to a new paradigm for the function of Cdc48 and its mammalian ortholog p97/VCP.


Subject(s)
Adenosine Triphosphatases/metabolism , Cell Cycle Proteins/metabolism , Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/isolation & purification , Amino Acid Sequence , Animals , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/isolation & purification , Endopeptidases/metabolism , Models, Molecular , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Ubiquitin/metabolism , Valosin Containing Protein
12.
Cell ; 171(1): 120-132.e12, 2017 Sep 21.
Article in English | MEDLINE | ID: mdl-28919079

ABSTRACT

The disassembly of the intron lariat spliceosome (ILS) marks the end of a splicing cycle. Here we report a cryoelectron microscopy structure of the ILS complex from Saccharomyces cerevisiae at an average resolution of 3.5 Å. The intron lariat remains bound in the spliceosome whereas the ligated exon is already dissociated. The step II splicing factors Prp17 and Prp18, along with Cwc21 and Cwc22 that stabilize the 5' exon binding to loop I of U5 small nuclear RNA (snRNA), have been released from the active site assembly. The DEAH family ATPase/helicase Prp43 binds Syf1 at the periphery of the spliceosome, with its RNA-binding site close to the 3' end of U6 snRNA. The C-terminal domain of Ntr1/Spp382 associates with the GTPase Snu114, and Ntr2 is anchored to Prp8 while interacting with the superhelical domain of Ntr1. These structural features suggest a plausible mechanism for the disassembly of the ILS complex.


Subject(s)
Introns , Spliceosomes/ultrastructure , Cryoelectron Microscopy , DEAD-box RNA Helicases/chemistry , Models, Molecular , RNA Precursors/chemistry , RNA Precursors/ultrastructure , RNA, Small Nuclear/chemistry , RNA, Small Nuclear/ultrastructure , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Schizosaccharomyces/chemistry , Spliceosomes/chemistry
13.
Cell ; 171(7): 1589-1598.e8, 2017 Dec 14.
Article in English | MEDLINE | ID: mdl-29153833

ABSTRACT

Removal of an intron from a pre-mRNA by the spliceosome results in the ligation of two exons in the post-catalytic spliceosome (known as the P complex). Here, we present a cryo-EM structure of the P complex from Saccharomyces cerevisiae at an average resolution of 3.6 Å. The ligated exon is held in the active site through RNA-RNA contacts. Three bases at the 3' end of the 5' exon remain anchored to loop I of U5 small nuclear RNA, and the conserved AG nucleotides of the 3'-splice site (3'SS) are specifically recognized by the invariant adenine of the branch point sequence, the guanine base at the 5' end of the 5'SS, and an adenine base of U6 snRNA. The 3'SS is stabilized through an interaction with the 1585-loop of Prp8. The P complex structure provides a view on splice junction formation critical for understanding the complete splicing cycle.


Subject(s)
Saccharomyces cerevisiae/chemistry , Spliceosomes/chemistry , Cryoelectron Microscopy , Humans , Models, Molecular , RNA Splicing , Saccharomyces cerevisiae/metabolism , Spliceosomes/metabolism
14.
Annu Rev Cell Dev Biol ; 34: 85-109, 2018 10 06.
Article in English | MEDLINE | ID: mdl-30095293

ABSTRACT

The endosomal sorting complexes required for transport (ESCRT) pathway mediates cellular membrane remodeling and fission reactions. The pathway comprises five core complexes: ALIX, ESCRT-I, ESCRT-II, ESCRT-III, and Vps4. These soluble complexes are typically recruited to target membranes by site-specific adaptors that bind one or both of the early-acting ESCRT factors: ALIX and ESCRT-I/ESCRT-II. These factors, in turn, nucleate assembly of ESCRT-III subunits into membrane-bound filaments that recruit the AAA ATPase Vps4. Together, ESCRT-III filaments and Vps4 remodel and sever membranes. Here, we review recent advances in our understanding of the structures, activities, and mechanisms of the ESCRT-III and Vps4 machinery, including the first high-resolution structures of ESCRT-III filaments, the assembled Vps4 enzyme in complex with an ESCRT-III substrate, the discovery that ESCRT-III/Vps4 complexes can promote both inside-out and outside-in membrane fission reactions, and emerging mechanistic models for ESCRT-mediated membrane fission.


Subject(s)
Actin Cytoskeleton/genetics , Adenosine Triphosphatases/genetics , Cell Membrane/genetics , Endosomal Sorting Complexes Required for Transport/genetics , Saccharomyces cerevisiae Proteins/genetics , Actin Cytoskeleton/chemistry , Adenosine Triphosphatases/chemistry , Cell Membrane/chemistry , Cytokinesis , Endosomal Sorting Complexes Required for Transport/chemistry , Endosomes/chemistry , Endosomes/genetics , Multiprotein Complexes/chemistry , Multiprotein Complexes/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/chemistry
15.
Mol Cell ; 2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39089251

ABSTRACT

Defects in organellar acidification indicate compromised or infected compartments. Recruitment of the autophagy-related ATG16L1 complex to pathologically neutralized organelles targets ubiquitin-like ATG8 molecules to perturbed membranes. How this process is coupled to proton gradient disruption is unclear. Here, we reveal that the V1H subunit of the vacuolar ATPase (V-ATPase) proton pump binds directly to ATG16L1. The V1H/ATG16L1 interaction only occurs within fully assembled V-ATPases, allowing ATG16L1 recruitment to be coupled to increased V-ATPase assembly following organelle neutralization. Cells lacking V1H fail to target ATG8s during influenza infection or after activation of the immune receptor stimulator of interferon genes (STING). We identify a loop within V1H that mediates ATG16L1 binding. A neuronal V1H isoform lacks this loop and is associated with attenuated ATG8 targeting in response to ionophores in primary murine and human iPSC-derived neurons. Thus, V1H controls ATG16L1 recruitment following proton gradient dissipation, suggesting that the V-ATPase acts as a cell-intrinsic damage sensor.

16.
Mol Cell ; 84(7): 1290-1303.e7, 2024 Apr 04.
Article in English | MEDLINE | ID: mdl-38401542

ABSTRACT

Most eukaryotic proteins are degraded by the 26S proteasome after modification with a polyubiquitin chain. Substrates lacking unstructured segments cannot be degraded directly and require prior unfolding by the Cdc48 ATPase (p97 or VCP in mammals) in complex with its ubiquitin-binding partner Ufd1-Npl4 (UN). Here, we use purified yeast components to reconstitute Cdc48-dependent degradation of well-folded model substrates by the proteasome. We show that a minimal system consists of the 26S proteasome, the Cdc48-UN ATPase complex, the proteasome cofactor Rad23, and the Cdc48 cofactors Ubx5 and Shp1. Rad23 and Ubx5 stimulate polyubiquitin binding to the 26S proteasome and the Cdc48-UN complex, respectively, allowing these machines to compete for substrates before and after their unfolding. Shp1 stimulates protein unfolding by the Cdc48-UN complex rather than substrate recruitment. Experiments in yeast cells confirm that many proteins undergo bidirectional substrate shuttling between the 26S proteasome and Cdc48 ATPase before being degraded.


Subject(s)
Proteasome Endopeptidase Complex , Saccharomyces cerevisiae Proteins , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Polyubiquitin/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Ubiquitin/metabolism , Valosin Containing Protein/genetics , Valosin Containing Protein/metabolism
17.
Mol Cell ; 84(12): 2368-2381.e6, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38834067

ABSTRACT

The Tn7 family of transposons is notable for its highly regulated integration mechanisms, including programmable RNA-guided transposition. The targeting pathways rely on dedicated target selection proteins from the TniQ family and the AAA+ adaptor TnsC to recruit and activate the transposase at specific target sites. Here, we report the cryoelectron microscopy (cryo-EM) structures of TnsC bound to the TniQ domain of TnsD from prototypical Tn7 and unveil key regulatory steps stemming from unique behaviors of ATP- versus ADP-bound TnsC. We show that TnsD recruits ADP-bound dimers of TnsC and acts as an exchange factor to release one protomer with exchange to ATP. This loading process explains how TnsC assembles a heptameric ring unidirectionally from the target site. This unique loading process results in functionally distinct TnsC protomers within the ring, providing a checkpoint for target immunity and explaining how insertions at programmed sites precisely occur in a specific orientation across Tn7 elements.


Subject(s)
Adenosine Diphosphate , Adenosine Triphosphate , Cryoelectron Microscopy , DNA Transposable Elements , Transposases , DNA Transposable Elements/genetics , Adenosine Triphosphate/metabolism , Transposases/metabolism , Transposases/genetics , Transposases/chemistry , Adenosine Diphosphate/metabolism , Protein Binding , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/chemistry , Models, Molecular , Protein Multimerization , Binding Sites
18.
Mol Cell ; 84(7): 1321-1337.e11, 2024 Apr 04.
Article in English | MEDLINE | ID: mdl-38513662

ABSTRACT

Intracellular Mg2+ (iMg2+) is bound with phosphometabolites, nucleic acids, and proteins in eukaryotes. Little is known about the intracellular compartmentalization and molecular details of Mg2+ transport into/from cellular organelles such as the endoplasmic reticulum (ER). We found that the ER is a major iMg2+ compartment refilled by a largely uncharacterized ER-localized protein, TMEM94. Conventional and AlphaFold2 predictions suggest that ERMA (TMEM94) is a multi-pass transmembrane protein with large cytosolic headpiece actuator, nucleotide, and phosphorylation domains, analogous to P-type ATPases. However, ERMA uniquely combines a P-type ATPase domain and a GMN motif for ERMg2+ uptake. Experiments reveal that a tyrosine residue is crucial for Mg2+ binding and activity in a mechanism conserved in both prokaryotic (mgtB and mgtA) and eukaryotic Mg2+ ATPases. Cardiac dysfunction by haploinsufficiency, abnormal Ca2+ cycling in mouse Erma+/- cardiomyocytes, and ERMA mRNA silencing in human iPSC-cardiomyocytes collectively define ERMA as an essential component of ERMg2+ uptake in eukaryotes.


Subject(s)
Adenosine Triphosphatases , P-type ATPases , Animals , Mice , Humans , Adenosine Triphosphatases/metabolism , Membrane Transport Proteins/metabolism , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Biological Transport , P-type ATPases/metabolism , Calcium/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases
19.
Annu Rev Cell Dev Biol ; 33: 417-438, 2017 10 06.
Article in English | MEDLINE | ID: mdl-28992441

ABSTRACT

Proper localization of membrane proteins is essential for the function of biological membranes and for the establishment of organelle identity within a cell. Molecular machineries that mediate membrane protein biogenesis need to not only achieve a high degree of efficiency and accuracy, but also prevent off-pathway aggregation events that can be detrimental to cells. The posttranslational targeting of tail-anchored proteins (TAs) provides tractable model systems to probe these fundamental issues. Recent advances in understanding TA-targeting pathways reveal sophisticated molecular machineries that drive and regulate these processes. These findings also suggest how an interconnected network of targeting factors, cochaperones, and quality control machineries together ensures robust membrane protein biogenesis.


Subject(s)
Membrane Proteins/metabolism , Animals , Humans , Membrane Proteins/chemistry , Models, Biological , Protein Sorting Signals , Protein Transport
20.
Mol Cell ; 83(5): 759-769.e7, 2023 03 02.
Article in English | MEDLINE | ID: mdl-36736315

ABSTRACT

The AAA+ ATPase Cdc48 utilizes the cofactor Ufd1/Npl4 to bind and thread polyubiquitinated substrates for their extraction from complexes or membranes and often for subsequent proteasomal degradation. Previous studies indicated that Cdc48 engages polyubiquitin chains through the Npl4-mediated unfolding of an initiator ubiquitin; yet, the underlying principles remain largely unknown. Using FRET-based assays, we revealed the mechanisms and kinetics of ubiquitin unfolding, insertion into the ATPase, and unfolding of the ubiquitin-attached substrate. We found that Cdc48 uses Ufd1's UT3 domain to bind a K48-linked ubiquitin on the initiator's proximal side of the chain, thereby directing the initiator toward rapid unfolding by Npl4 and engagement by Cdc48. Ubiquitins on the initiator's distal side increase substrate affinity and facilitate unfolding but impede substrate release from Cdc48-Ufd1/Npl4 in the absence of additional cofactors. Our findings explain how Cdc48-UN efficiently processes substrates with K48-linked chains of 4-6 ubiquitins, which represent most cellular polyubiquitinated proteins.


Subject(s)
Polyubiquitin , Saccharomyces cerevisiae Proteins , Polyubiquitin/metabolism , Saccharomyces cerevisiae Proteins/metabolism , ATPases Associated with Diverse Cellular Activities/metabolism , Valosin Containing Protein/metabolism , Vesicular Transport Proteins/metabolism , Nucleocytoplasmic Transport Proteins/metabolism , Ubiquitin/metabolism , Ubiquitins/metabolism , Cell Cycle Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL