Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 15(1): 2459, 2024 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-38503733

RESUMO

The hexameric AAA+ ATPase p97/VCP functions as an essential mediator of ubiquitin-dependent cellular processes, extracting ubiquitylated proteins from macromolecular complexes or membranes by catalyzing their unfolding. p97 is directed to ubiquitylated client proteins via multiple cofactors, most of which interact with the p97 N-domain. Here, we discover that FAM104A, a protein of unknown function also named VCF1 (VCP/p97 nuclear Cofactor Family member 1), acts as a p97 cofactor in human cells. Detailed structure-function studies reveal that VCF1 directly binds p97 via a conserved α-helical motif that recognizes the p97 N-domain with unusually high affinity, exceeding that of other cofactors. We show that VCF1 engages in joint p97 complex formation with the heterodimeric primary p97 cofactor UFD1-NPL4 and promotes p97-UFD1-NPL4-dependent proteasomal degradation of ubiquitylated substrates in cells. Mechanistically, VCF1 indirectly stimulates UFD1-NPL4 interactions with ubiquitin conjugates via its binding to p97 but has no intrinsic affinity for ubiquitin. Collectively, our findings establish VCF1 as an unconventional p97 cofactor that promotes p97-dependent protein turnover by facilitating p97-UFD1-NPL4 recruitment to ubiquitylated targets.


Assuntos
Proteínas de Ciclo Celular , Ubiquitina , Humanos , Ligação Proteica , Ubiquitina/metabolismo , Proteína com Valosina/genética , Proteína com Valosina/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo
2.
Mol Cell ; 84(8): 1556-1569.e10, 2024 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-38503285

RESUMO

Cells respond to lysosomal membrane permeabilization by membrane repair or selective macroautophagy of damaged lysosomes, termed lysophagy, but it is not fully understood how this decision is made. Here, we uncover a pathway in human cells that detects lipid bilayer perturbations in the limiting membrane of compromised lysosomes, which fail to be repaired, and then initiates ubiquitin-triggered lysophagy. We find that SPG20 binds the repair factor IST1 on damaged lysosomes and, importantly, integrates that with the detection of damage-associated lipid-packing defects of the lysosomal membrane. Detection occurs via sensory amphipathic helices in SPG20 before rupture of the membrane. If lipid-packing defects are extensive, such as during lipid peroxidation, SPG20 recruits and activates ITCH, which marks the damaged lysosome with lysine-63-linked ubiquitin chains to initiate lysophagy and thus triages the lysosome for destruction. With SPG20 being linked to neurodegeneration, these findings highlight the relevance of a coordinated lysosomal damage response for cellular homeostasis.


Assuntos
Lisossomos , Macroautofagia , Humanos , Autofagia/fisiologia , Membranas Intracelulares/metabolismo , Lipídeos , Lisossomos/metabolismo , Ubiquitina/genética , Ubiquitina/metabolismo
3.
Nat Commun ; 14(1): 3258, 2023 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-37277335

RESUMO

The AAA+ ATPase p97/VCP together with different sets of substrate-delivery adapters and accessory cofactor proteins unfolds ubiquitinated substrates to facilitate degradation by the proteasome. The UBXD1 cofactor is connected to p97-associated multisystem proteinopathy but its biochemical function and structural organization on p97 has remained largely elusive. Using a combination of crosslinking mass spectrometry and biochemical assays, we identify an extended UBX (eUBX) module in UBXD1 related to a lariat in another cofactor, ASPL. Of note, the UBXD1-eUBX intramolecularly associates with the PUB domain in UBXD1 close to the substrate exit pore of p97. The UBXD1 PUB domain can also bind the proteasomal shuttling factor HR23b via its UBL domain. We further show that the eUBX domain has ubiquitin binding activity and that UBXD1 associates with an active p97-adapter complex during substrate unfolding. Our findings suggest that the UBXD1-eUBX module receives unfolded ubiquitinated substrates after they exit the p97 channel and before hand-over to the proteasome. The interplay of full-length UBXD1 and HR23b and their function in the context of an active p97:UBXD1 unfolding complex remains to be studied in future work.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Proteínas de Transporte , Proteínas de Transporte/metabolismo , ATPases Associadas a Diversas Atividades Celulares/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteína com Valosina/genética , Proteína com Valosina/metabolismo , Adenosina Trifosfatases/metabolismo , Proteínas Relacionadas à Autofagia/genética , Proteínas Relacionadas à Autofagia/metabolismo , Estrutura Terciária de Proteína , Ligação Proteica , Ubiquitina/metabolismo , Proteínas de Ciclo Celular/metabolismo
4.
EMBO J ; 42(14): e113110, 2023 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-37264685

RESUMO

The AAA+-ATPase p97 (also called VCP or Cdc48) unfolds proteins and disassembles protein complexes in numerous cellular processes, but how substrate complexes are loaded onto p97 and disassembled is unclear. Here, we present cryo-EM structures of p97 in the process of disassembling a protein phosphatase-1 (PP1) complex by extracting an inhibitory subunit from PP1. We show that PP1 and its partners SDS22 and inhibitor-3 (I3) are loaded tightly onto p97, surprisingly via a direct contact of SDS22 with the p97 N-domain. Loading is assisted by the p37 adapter that bridges two adjacent p97 N-domains underneath the substrate complex. A stretch of I3 is threaded into the central channel of the spiral-shaped p97 hexamer, while other elements of I3 are still attached to PP1. Thus, our data show how p97 arranges a protein complex between the p97 N-domain and central channel, suggesting a hold-and-extract mechanism for p97-mediated disassembly.


Assuntos
Proteínas de Ciclo Celular , Ubiquitina , Ubiquitina/metabolismo , Proteína Fosfatase 1/genética , Proteína Fosfatase 1/metabolismo , ATPases Associadas a Diversas Atividades Celulares/metabolismo , Modelos Moleculares , Proteína com Valosina/genética , Proteína com Valosina/metabolismo , Proteínas de Ciclo Celular/metabolismo
5.
Front Mol Biosci ; 10: 1142989, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36825201

RESUMO

The AAA+ ATPase p97 (also called VCP or Cdc48) is a major protein unfolding machine with hundreds of clients in diverse cellular pathways that are critical for cell homeostasis, proliferation and signaling. In this review, we summarize recent advances in understanding how diverse client proteins are targeted to the p97 machine to facilitate client degradation or to strip clients from binding partners for regulation. We describe an elaborate system that is governed by at least two types of alternative adapters. The Ufd1-Npl4 adapter along with accessory adapters targets ubiquitylated clients in the majority of pathways and uses ubiquitin as a universal unfolding tag. In contrast, the family of SEP-domain adapters such as p37 can target clients directly to p97 in a ubiquitin-independent manner. Despite the different targeting strategies, both pathways converge by inserting the client into the p97 pore to initiate a peptide threading mechanism through the central channel of p97 that drives client protein unfolding, protein extraction from membranes and protein complex disassembly processes.

6.
J Biol Chem ; 298(6): 101976, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35469923

RESUMO

The protease SPRTN degrades DNA-protein crosslinks (DPCs) that threaten genome stability. SPRTN has been connected to the ubiquitin-directed protein unfoldase p97 (also called VCP or Cdc48), but a functional cooperation has not been demonstrated directly. Here, we biochemically reconstituted p97-assisted proteolysis with purified proteins and showed that p97 targets ubiquitin-modified DPCs and unfolds them to prepare them for proteolysis by SPRTN. We demonstrate that purified SPRTN alone was unable to degrade a tightly-folded Eos fluorescent reporter protein even when Eos was crosslinked to DNA (Eos-DPC). However, when present, p97 unfolded poly-ubiquitinated Eos-DPC in a manner requiring its ubiquitin adapter, Ufd1-Npl4. Notably, we show that, in cooperation with p97 and Ufd1-Npl4, SPRTN proteolyzed unfolded Eos-DPC, which relied on recognition of the DNA-crosslink by SPRTN. In a simplified unfolding assay, we further demonstrate that p97, while unfolding a protein substrate, can surmount the obstacle of a DNA crosslink site in the substrate. Thus, our data demonstrate that p97, in conjunction with Ufd1-Npl4, assists SPRTN-mediated proteolysis of tightly-folded proteins crosslinked to DNA, even threading bulky protein-DNA adducts. These findings will be relevant for understanding how cells handle DPCs to ensure genome stability and for designing strategies that target p97 in combination cancer therapy.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteínas , Ubiquitina , Proteína com Valosina , ATPases Associadas a Diversas Atividades Celulares/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , DNA/metabolismo , Instabilidade Genômica , Humanos , Proteínas/metabolismo , Proteólise , Ubiquitina/metabolismo , Proteína com Valosina/genética , Proteína com Valosina/metabolismo
7.
Nat Struct Mol Biol ; 28(12): 964-971, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34824462

RESUMO

The AAA-ATPase VCP/p97/Cdc48 unfolds proteins by threading them through its central pore, but how substrates are recognized and inserted into the pore in diverse pathways has remained controversial. Here, we show that p97, with its adapter p37, binds an internal recognition site (IRS) within inhibitor-3 (I3) and then threads a peptide loop into its channel to strip I3 off protein phosphatase-1 (PP1). Of note, the IRS is adjacent to the prime interaction site of I3 to PP1, and IRS mutations block I3 processing both in vitro and in cells. In contrast, amino- and carboxy-terminal regions of I3 are not required, and even circularization of I3 does not prevent I3 processing. This was confirmed by an in vitro Förster resonance energy transfer assay that allowed kinetic analysis of the reaction. Thus, our data uncover how PP1 is released from its inhibitory partner for activation and demonstrate a remarkable plasticity in substrate threading by p97.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteína Fosfatase 1/metabolismo , Proteína com Valosina/metabolismo , Animais , Sítios de Ligação/genética , Domínio Catalítico/genética , Linhagem Celular , Transferência Ressonante de Energia de Fluorescência , Células HEK293 , Humanos , Ligação Proteica/genética , Desdobramento de Proteína , Células Sf9
8.
Fungal Biol ; 124(12): 1024-1031, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33213782

RESUMO

Oomycetes are fungal-like eukaryotes and many of them are pathogens that threaten natural ecosystems and cause huge financial losses for the aqua- and agriculture industry. Amongst them, Aphanomyces invadans causes Epizootic Ulcerative Syndrome (EUS) in fish which can be responsible for up to 100% mortality in aquaculture. As other eukaryotic pathogens, in order to establish and promote an infection, A. invadans secretes proteins, which are predicted to overcome host defence mechanisms and interfere with other processes inside the host. We investigated the role of Lhs1 which is part of an ER-resident complex that generally promotes the translocation of proteins from the cytoplasm into the ER for further processing and secretion. Interestingly, proteomic studies reveal that only a subset of virulence factors are affected by the silencing of AiLhs1 in A. invadans indicating various secretion pathways for different proteins. Importantly, changes in the secretome upon silencing of AiLhs1 significantly reduces the virulence of A. invadans in the infection model Galleriamellonella. Furthermore, we show that AiLhs1 is important for the production of zoospores and their cluster formation. This renders proteins required for protein ER translocation as interesting targets for the potential development of alternative disease control strategies in agri- and aquaculture.


Assuntos
Aphanomyces , Doenças dos Peixes , Chaperonas Moleculares/fisiologia , Virulência , Animais , Aphanomyces/patogenicidade , Doenças dos Peixes/microbiologia , Proteômica
9.
J Mol Biol ; 432(23): 6061-6074, 2020 11 20.
Artigo em Inglês | MEDLINE | ID: mdl-33058883

RESUMO

The AAA-ATPase VCP/p97 cooperates with the SEP-domain adapters p37, UBXN2A and p47 in stripping inhibitor-3 (I3) from protein phosphatase-1 (PP1) for activation. In contrast to p97-mediated degradative processes, PP1 complex disassembly is ubiquitin-independent. It is therefore unclear how selective targeting is achieved. Using biochemical reconstitution and crosslink mass spectrometry, we show here that SEP-domain adapters use a multivalent substrate recognition strategy. An N-terminal sequence element predicted to form a helix, together with the SEP-domain, binds and engages the direct target I3 in the central pore of p97 for unfolding, while its partner PP1 is held by a linker between SHP box and UBX domain locked onto the peripheral N-domain of p97. Although the I3-binding element is functional in p47, p47 in vitro requires a transplant of the PP1-binding linker from p37 for activity stressing that both sites are essential to control specificity. Of note, unfolding is then governed by an inhibitory segment in the N-terminal region of p47, suggesting a regulatory function. Together, this study reveals how p97 adapters engage a protein complex for ubiquitin-independent disassembly while ensuring selectivity for one subunit.


Assuntos
Adenosina Trifosfatases/química , Complexos Multiproteicos/química , Proteínas Nucleares/química , Conformação Proteica , Proteína Fosfatase 1/química , Proteínas Adaptadoras de Transdução de Sinal/química , Proteínas Adaptadoras de Transdução de Sinal/genética , Adenosina Trifosfatases/genética , Adenosina Trifosfatases/metabolismo , Adenosina Trifosfatases/ultraestrutura , Sequência de Aminoácidos/genética , Domínio Catalítico/genética , Cristalografia por Raios X , Humanos , Metaloendopeptidases/química , Metaloendopeptidases/genética , Complexos Multiproteicos/genética , Complexos Multiproteicos/ultraestrutura , Proteínas Nucleares/genética , Proteínas Nucleares/ultraestrutura , Ligação Proteica/genética , Proteína Fosfatase 1/genética , Proteína Fosfatase 1/ultraestrutura , Estrutura Terciária de Proteína , Subunidades Proteicas/química , Ubiquitina/genética , Ubiquitinas/química , Ubiquitinas/genética
10.
Nanoscale ; 12(37): 19093-19103, 2020 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-32662484

RESUMO

When nanoparticles enter a physiological environment, they rapidly adsorb biomolecules, in particular cellular proteins. This biological coating, the so-called nanoparticle protein corona, undoubtedly affects the biological identity and potential cytotoxicity of the nanomaterial. To elucidate a possible impact on the adsorbed biomolecules, we focused on an important group of players in cellular homeostasis, namely proteolytic enzymes. We could demonstrate that amorphous silica nanoparticles are not only able to bind to the oncologically relevant threonine protease Taspase1 as revealed by microscale thermophoresis and fluorescence anisotropy measurements, but moreover inhibit its proteolytic activity in a non-competitive manner. As revealed by temperature-dependent unfolding and CD spectroscopy, binding did not alter the stability of Taspase1 or its secondary structure. Noteworthy, inhibition of protein function seems not a general feature of nanoparticles, as several control enzymes were not affected in their proteolytic activity. Our data suggests that nanoparticles bind Taspase1 as an αß-dimer in a single layer without conformational change, resulting in noncompetitive inhibition that is either allostery-like or occludes the active site. Nanoparticle-based inhibition of Taspase1 could be also achieved in cell lysates and in live cells as shown by the use of a protease-specific cellular cleavage biosensor. Collectively, we could demonstrate that nanoparticles could not only bind but also selectively inhibit cellular enzymes, which might explain observed cytotoxicity but might serve as a starting point for the development of nanoparticle-based inhibitors as therapeutics.


Assuntos
Nanopartículas , Coroa de Proteína , Endopeptidases , Peptídeo Hidrolases , Dióxido de Silício
11.
Biomolecules ; 9(12)2019 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-31847414

RESUMO

AAA+ ATPase p97/valosin-containing protein (VCP)/Cdc48 is a key player in various cellular stress responses in which it unfolds ubiquitinated proteins to facilitate their degradation by the proteasome. P97 works in different cellular processes using alternative sets of cofactors and is implicated in multiple degenerative diseases. Ubiquitin regulatory X domain protein 1 (UBXD1) has been linked to pathogenesis and is unique amongst p97 cofactors because it interacts with both termini of p97. Its N-domain binds to the N-domain and N/D1 interface of p97 and regulates its ATPase activity. The PUB (peptide:N-glycanase and UBA or UBX-containing proteins) domain binds the p97 C-terminus, but how it controls p97 function is still unknown. Here we present the NMR structure of UBXD1-PUB together with binding studies, mutational analysis, and a model of UBXD1-PUB in complex with the p97 C-terminus. While the binding pocket is conserved among PUB domains, UBXD1-PUB features a unique loop and turn regions suggesting a role in coordinating interaction with downstream regulators and substrate processing.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/química , Proteínas Relacionadas à Autofagia/química , Proteína com Valosina/química , Proteínas Adaptadoras de Transporte Vesicular/isolamento & purificação , Proteínas Relacionadas à Autofagia/isolamento & purificação , Humanos , Ligação Proteica , Estrutura Terciária de Proteína , Proteína com Valosina/isolamento & purificação
12.
Mol Cell ; 72(4): 766-777.e6, 2018 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-30344098

RESUMO

The functional diversity of protein phosphatase-1 (PP1), with its countless substrates, relies on the ordered assembly of alternative PP1 holoenzymes. Here, we show that newly synthesized PP1 is first held by its partners SDS22 and inhibitor-3 (I3) in an inactive complex, which needs to be disassembled by the p97 AAA-ATPase to promote exchange to substrate specifiers. Unlike p97-mediated degradative processes that require the Ufd1-Npl4 ubiquitin adapters, p97 is targeted to PP1 by p37 and related adapter proteins. Reconstitution with purified components revealed direct interaction of the p37 SEP domain with I3 without the need for ubiquitination, and ATP-driven pulling of I3 into the central channel of the p97 hexamer, which triggers dissociation of I3 and SDS22. Thus, we establish regulatory ubiquitin-independent protein complex disassembly as part of the functional arsenal of p97 and define an unanticipated essential step in PP1 biogenesis that illustrates the molecular challenges of ordered subunit exchange.


Assuntos
Adenosina Trifosfatases/metabolismo , Proteínas Nucleares/metabolismo , Proteína Fosfatase 1/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas de Ciclo Celular/metabolismo , Células HEK293 , Células HeLa , Holoenzimas/metabolismo , Humanos , Modelos Moleculares , Complexo de Proteínas Formadoras de Poros Nucleares/metabolismo , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Ligação Proteica , Proteína Fosfatase 1/antagonistas & inibidores , ATPases Translocadoras de Prótons/metabolismo , Ubiquitina/metabolismo
13.
J Biol Chem ; 293(25): 9724-9735, 2018 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-29743239

RESUMO

The high-energy sulfate donor 3'-phosphoadenosine-5'-phosphosulfate (PAPS), generated by human PAPS synthase isoforms PAPSS1 and PAPSS2, is required for all human sulfation pathways. Sulfotransferase SULT2A1 uses PAPS for sulfation of the androgen precursor dehydroepiandrosterone (DHEA), thereby reducing downstream activation of DHEA to active androgens. Human PAPSS2 mutations manifest with undetectable DHEA sulfate, androgen excess, and metabolic disease, suggesting that ubiquitous PAPSS1 cannot compensate for deficient PAPSS2 in supporting DHEA sulfation. In knockdown studies in human adrenocortical NCI-H295R1 cells, we found that PAPSS2, but not PAPSS1, is required for efficient DHEA sulfation. Specific APS kinase activity, the rate-limiting step in PAPS biosynthesis, did not differ between PAPSS1 and PAPSS2. Co-expression of cytoplasmic SULT2A1 with a cytoplasmic PAPSS2 variant supported DHEA sulfation more efficiently than co-expression with nuclear PAPSS2 or nuclear/cytosolic PAPSS1. Proximity ligation assays revealed protein-protein interactions between SULT2A1 and PAPSS2 and, to a lesser extent, PAPSS1. Molecular docking studies showed a putative binding site for SULT2A1 within the PAPSS2 APS kinase domain. Energy-dependent scoring of docking solutions identified the interaction as specific for the PAPSS2 and SULT2A1 isoforms. These findings elucidate the mechanistic basis for the selective requirement for PAPSS2 in human DHEA sulfation.


Assuntos
Carcinoma Adrenocortical/metabolismo , Sulfato de Desidroepiandrosterona/metabolismo , Complexos Multienzimáticos/metabolismo , Sulfato Adenililtransferase/metabolismo , Sulfotransferases/metabolismo , Sítios de Ligação , Núcleo Celular/metabolismo , Cristalografia por Raios X , Citosol/metabolismo , Sulfato de Desidroepiandrosterona/química , Humanos , Simulação de Acoplamento Molecular , Complexos Multienzimáticos/química , Conformação Proteica , Domínios e Motivos de Interação entre Proteínas , Sulfato Adenililtransferase/química , Sulfotransferases/química , Células Tumorais Cultivadas
14.
Mol Cell ; 69(2): 182-194, 2018 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-29153394

RESUMO

The AAA+-type ATPase p97 governs an ever-expanding number of cellular processes reaching from degradation of damaged proteins and organelles to key signaling events and chromatin regulation with thousands of client proteins. With its relevance for cellular homeostasis and genome stability, it is linked to muscular and neuronal degeneration and, conversely, constitutes an attractive anti-cancer drug target. Its molecular function is ATP-driven protein unfolding, which is directed by ubiquitin and assisted by a host of cofactor proteins. This activity underlies p97's diverse ability to pull proteins out of membranes, unfold proteins for proteasomal degradation, or segregate proteins from partners for downstream activity. Recent advances in structural analysis and biochemical reconstitution have underscored this notion, resolved detailed molecular motions within the p97 hexamer, and suggested substrate threading through the central channel of the p97 hexamer as the driving mechanism. We will discuss the mechanisms and open questions in the context of the diverse cellular activities.


Assuntos
Adenosina Trifosfatases/metabolismo , Proteínas Nucleares/metabolismo , Proteína com Valosina/metabolismo , Proteínas de Ciclo Celular/metabolismo , Homeostase , Humanos , Desdobramento de Proteína , Transdução de Sinais , Ubiquitina/metabolismo
15.
Angew Chem Int Ed Engl ; 57(6): 1576-1580, 2018 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-29271116

RESUMO

AAA ATPases have pivotal functions in diverse cellular processes essential for survival and proliferation. Revealing strategies for chemical inhibition of this class of enzymes is therefore of great interest for the development of novel chemotherapies or chemical tools. Here, we characterize the compound MSC1094308 as a reversible, allosteric inhibitor of the type II AAA ATPase human ubiquitin-directed unfoldase (VCP)/p97 and the type I AAA ATPase VPS4B. Subsequent proteomic, genetic and biochemical studies indicate that MSC1094308 binds to a previously characterized drugable hotspot of p97, thereby inhibiting the D2 ATPase activity. Our results furthermore indicate that a similar allosteric site exists in VPS4B, suggesting conserved allosteric circuits and drugable sites in both type I and II AAA ATPases. Our results may thus guide future chemical tool and drug discovery efforts for the biomedically relevant AAA ATPases.


Assuntos
ATPases Associadas a Diversas Atividades Celulares/metabolismo , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Inibidores Enzimáticos/metabolismo , Proteína com Valosina/metabolismo , ATPases Associadas a Diversas Atividades Celulares/antagonistas & inibidores , ATPases Associadas a Diversas Atividades Celulares/genética , Trifosfato de Adenosina/química , Trifosfato de Adenosina/metabolismo , Regulação Alostérica , Sítio Alostérico , Sítios de Ligação , Complexos Endossomais de Distribuição Requeridos para Transporte/antagonistas & inibidores , Complexos Endossomais de Distribuição Requeridos para Transporte/genética , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Humanos , Concentração Inibidora 50 , Mutagênese Sítio-Dirigida , Ligação Proteica , Relação Estrutura-Atividade , Proteína com Valosina/antagonistas & inibidores
16.
Mol Cell ; 64(1): 189-198, 2016 10 06.
Artigo em Inglês | MEDLINE | ID: mdl-27716483

RESUMO

During DNA double-strand break (DSB) repair, the ring-shaped Ku70/80 complex becomes trapped on DNA and needs to be actively extracted, but it has remained unclear what provides the required energy. By means of reconstitution of DSB repair on beads, we demonstrate here that DNA-locked Ku rings are released by the AAA-ATPase p97. To achieve this, p97 requires ATP hydrolysis, cooperates with the Ufd1-Npl4 ubiquitin-adaptor complex, and specifically targets Ku80 that is modified by K48-linked ubiquitin chains. In U2OS cells, chemical inhibition of p97 or siRNA-mediated depletion of p97 or its adapters impairs Ku80 removal after non-homologous end joining of DSBs. Moreover, this inhibition attenuates early steps in homologous recombination, consistent with p97-driven Ku release also affecting repair pathway choice. Thus, our data answer a central question regarding regulation of Ku in DSB repair and illustrate the ability of p97 to segregate even tightly bound protein complexes for release from DNA.


Assuntos
Adenosina Trifosfatases/genética , Proteínas de Anfíbios/genética , Proteínas de Ciclo Celular/genética , Reparo do DNA por Junção de Extremidades , Autoantígeno Ku/genética , Osteoblastos/metabolismo , Reparo de DNA por Recombinação , Adenosina Trifosfatases/antagonistas & inibidores , Adenosina Trifosfatases/metabolismo , Trifosfato de Adenosina/metabolismo , Proteínas de Anfíbios/metabolismo , Animais , Proteínas de Ciclo Celular/antagonistas & inibidores , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , DNA/genética , DNA/metabolismo , Quebras de DNA de Cadeia Dupla , Regulação da Expressão Gênica , Humanos , Hidrólise , Autoantígeno Ku/metabolismo , Microesferas , Osteoblastos/citologia , Óvulo/química , Óvulo/citologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Proteína com Valosina , Xenopus laevis
17.
PLoS One ; 11(3): e0151431, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26974973

RESUMO

Type 2 asparaginases, a subfamily of N-terminal nucleophile (Ntn) hydrolases, are activated by limited proteolysis. This activation yields a heterodimer and a loop region at the C-terminus of the α-subunit is released. Since this region is unresolved in all type 2 asparaginase crystal structures but is close to the active site residues, we explored this loop region in six members of the type 2 asparaginase family using homology modeling. As the loop model for the childhood cancer-relevant protease Taspase1 differed from the other members, Taspase1 activation as well as the conformation and dynamics of the 56 amino acids loop were investigated by CD and NMR spectroscopy. We propose a helix-turn-helix motif, which can be exploited as novel anticancer target to inhibit Taspase1 proteolytic activity.


Assuntos
Endopeptidases/química , Endopeptidases/metabolismo , Proteína de Leucina Linfoide-Mieloide/metabolismo , Subunidades Proteicas/química , Subunidades Proteicas/metabolismo , Humanos , Simulação de Dinâmica Molecular , Estrutura Secundária de Proteína , Espectroscopia de Prótons por Ressonância Magnética , Relação Estrutura-Atividade
18.
PLoS One ; 10(8): e0135455, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26291722

RESUMO

We present a 3D model of the four transmembrane (TM) helical regions of bilitranslocase (BTL), a structurally uncharacterized protein that transports organic anions across the cell membrane. The model was computed by considering helix-helix interactions as primary constraints, using Monte Carlo simulations. The interactions between the TM2 and TM3 segments have been confirmed by Förster resonance energy transfer (FRET) spectroscopy and nuclear magnetic resonance (NMR) spectroscopy, increasing our confidence in the model. Several insights into the BTL transport mechanism were obtained by analyzing the model. For example, the observed cis-trans Leu-Pro peptide bond isomerization in the TM3 fragment may indicate a key conformational change during anion transport by BTL. Our structural model of BTL may facilitate further studies, including drug discovery.


Assuntos
Proteínas de Membrana/química , Membrana Celular/metabolismo , Ceruloplasmina , Transferência Ressonante de Energia de Fluorescência , Humanos , Micelas , Modelos Estruturais , Método de Monte Carlo , Ressonância Magnética Nuclear Biomolecular , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína
19.
Chembiochem ; 15(15): 2233-7, 2014 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-25146997

RESUMO

Taspase 1 is an N-terminal threonine protease implicated in leukemia and other cancers. Despite intensive efforts in recent years, only a limited number of Taspase 1 inhibitors are currently available, and they lack general applicability. Here we present a novel class of Taspase 1 inhibitors based on a peptidyl succinimidyl peptide motif. These inhibitors were obtained from the substrate cleavage sequence and mechanistic considerations involving the previously proposed asparaginase-type cleavage mechanism. We anticipate that this class of Taspase 1 inhibitor will find wide application in further biochemical and structural studies, for example for better investigating the molecular details of the unusual enzymatic cleavage mechanism of Taspase 1.


Assuntos
Endopeptidases/metabolismo , Peptídeos/farmacologia , Inibidores de Proteases/farmacologia , Relação Dose-Resposta a Droga , Humanos , Conformação Molecular , Peptídeos/síntese química , Peptídeos/química , Inibidores de Proteases/síntese química , Inibidores de Proteases/química , Relação Estrutura-Atividade
20.
Structure ; 21(10): 1769-77, 2013 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-23972472

RESUMO

The mitotic regulator Pin1 plays an important role in protein quality control and age-related medical conditions such as Alzheimer disease and Parkinson disease. Although its cellular role has been thoroughly investigated during the past decade, the molecular mechanisms underlying its function remain elusive. We provide evidence for interactions between the two domains of Pin1. Several residues displayed unequivocal peak splits in nuclear magnetic resonance spectra, indicative of two different conformational states in equilibrium. Pareto analysis of paramagnetic relaxation enhancement data demonstrates that the two domains approach each other upon addition of a nonpeptidic ligand. Titration experiments with phosphorylated peptides monitored by fluorescence anisotropy and chemical shift perturbation indicate that domain interactions increase Pin1's affinity toward peptide ligands. We propose this interplay of the domains and ligands to be a general mechanism for a large class of two-domain proteins.


Assuntos
Peptidilprolil Isomerase/química , Fosfopeptídeos/química , Polarização de Fluorescência , Humanos , Ligantes , Modelos Moleculares , Simulação de Dinâmica Molecular , Peptidilprolil Isomerase de Interação com NIMA , Ressonância Magnética Nuclear Biomolecular , Polietilenoglicóis/química , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Estrutura Secundária de Proteína , Soluções , Solventes/química , Termodinâmica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA