Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 232
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell ; 82(11): 2006-2020.e8, 2022 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-35353987

RESUMEN

CK1s are acidophilic serine/threonine kinases with multiple critical cellular functions; their misregulation contributes to cancer, neurodegenerative diseases, and sleep phase disorders. Here, we describe an evolutionarily conserved mechanism of CK1 activity: autophosphorylation of a threonine (T220 in human CK1δ) located at the N terminus of helix αG, proximal to the substrate binding cleft. Crystal structures and molecular dynamics simulations uncovered inherent plasticity in αG that increased upon T220 autophosphorylation. The phosphorylation-induced structural changes significantly altered the conformation of the substrate binding cleft, affecting substrate specificity. In T220 phosphorylated yeast and human CK1s, activity toward many substrates was decreased, but we also identified a high-affinity substrate that was phosphorylated more rapidly, and quantitative phosphoproteomics revealed that disrupting T220 autophosphorylation rewired CK1 signaling in Schizosaccharomyces pombe. T220 is present exclusively in the CK1 family, thus its autophosphorylation may have evolved as a unique regulatory mechanism for this important family.


Asunto(s)
Proteínas Serina-Treonina Quinasas , Quinasa Idelta de la Caseína , Humanos , Fosforilación , Saccharomyces cerevisiae/enzimología , Saccharomyces cerevisiae/genética , Transducción de Señal , Especificidad por Sustrato , Treonina
2.
Nature ; 618(7964): 394-401, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37225996

RESUMEN

The endoplasmic reticulum (ER) undergoes continuous remodelling via a selective autophagy pathway, known as ER-phagy1. ER-phagy receptors have a central role in this process2, but the regulatory mechanism remains largely unknown. Here we report that ubiquitination of the ER-phagy receptor FAM134B within its reticulon homology domain (RHD) promotes receptor clustering and binding to lipidated LC3B, thereby stimulating ER-phagy. Molecular dynamics (MD) simulations showed how ubiquitination perturbs the RHD structure in model bilayers and enhances membrane curvature induction. Ubiquitin molecules on RHDs mediate interactions between neighbouring RHDs to form dense receptor clusters that facilitate the large-scale remodelling of lipid bilayers. Membrane remodelling was reconstituted in vitro with liposomes and ubiquitinated FAM134B. Using super-resolution microscopy, we discovered FAM134B nanoclusters and microclusters in cells. Quantitative image analysis revealed a ubiquitin-mediated increase in FAM134B oligomerization and cluster size. We found that the E3 ligase AMFR, within multimeric ER-phagy receptor clusters, catalyses FAM134B ubiquitination and regulates the dynamic flux of ER-phagy. Our results show that ubiquitination enhances RHD functions via receptor clustering, facilitates ER-phagy and controls ER remodelling in response to cellular demands.


Asunto(s)
Autofagia , Estrés del Retículo Endoplásmico , Retículo Endoplásmico , Ubiquitinación , Autofagia/fisiología , Retículo Endoplásmico/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ubiquitinas/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Receptores del Factor Autocrino de Motilidad/metabolismo
3.
Mol Cell ; 77(1): 164-179.e6, 2020 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-31732457

RESUMEN

The family of bacterial SidE enzymes catalyzes non-canonical phosphoribosyl-linked (PR) serine ubiquitination and promotes infectivity of Legionella pneumophila. Here, we describe identification of two bacterial effectors that reverse PR ubiquitination and are thus named deubiquitinases for PR ubiquitination (DUPs; DupA and DupB). Structural analyses revealed that DupA and SidE ubiquitin ligases harbor a highly homologous catalytic phosphodiesterase (PDE) domain. However, unlike SidE ubiquitin ligases, DupA displays increased affinity to PR-ubiquitinated substrates, which allows DupA to cleave PR ubiquitin from substrates. Interfering with DupA-ubiquitin binding switches its activity toward SidE-type ligase. Given the high affinity of DupA to PR-ubiquitinated substrates, we exploited a catalytically inactive DupA mutant to trap and identify more than 180 PR-ubiquitinated host proteins in Legionella-infected cells. Proteins involved in endoplasmic reticulum (ER) fragmentation and membrane recruitment to Legionella-containing vacuoles (LCV) emerged as major SidE targets. The global map of PR-ubiquitinated substrates provides critical insights into host-pathogen interactions during Legionella infection.


Asunto(s)
Enzimas Desubicuitinizantes/metabolismo , Serina/metabolismo , Ubiquitina/metabolismo , Ubiquitinación/fisiología , Células A549 , Proteínas Bacterianas/metabolismo , Dominio Catalítico/fisiología , Línea Celular , Línea Celular Tumoral , Retículo Endoplásmico/metabolismo , Células HEK293 , Células HeLa , Interacciones Huésped-Patógeno/fisiología , Humanos , Legionella pneumophila/patogenicidad , Enfermedad de los Legionarios/metabolismo , Vacuolas/metabolismo
4.
Proc Natl Acad Sci U S A ; 121(21): e2318591121, 2024 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-38739802

RESUMEN

The transcription factor p73, a member of the p53 tumor-suppressor family, regulates cell death and also supports tumorigenesis, although the mechanistic basis for the dichotomous functions is poorly understood. We report here the identification of an alternate transactivation domain (TAD) located at the extreme carboxyl (C) terminus of TAp73ß, a commonly expressed p73 isoform. Mutational disruption of this TAD significantly reduced TAp73ß's transactivation activity, to a level observed when the amino (N)-TAD that is similar to p53's TAD, is mutated. Mutation of both TADs almost completely abolished TAp73ß's transactivation activity. Expression profiling highlighted a unique set of targets involved in extracellular matrix-receptor interaction and focal adhesion regulated by the C-TAD, resulting in FAK phosphorylation, distinct from the N-TAD targets that are common to p53 and are involved in growth inhibition. Interestingly, the C-TAD targets are also regulated by the oncogenic, amino-terminal-deficient DNp73ß isoform. Consistently, mutation of C-TAD reduces cellular migration and proliferation. Mechanistically, selective binding of TAp73ß to DNAJA1 is required for the transactivation of C-TAD target genes, and silencing DNAJA1 expression abrogated all C-TAD-mediated effects. Taken together, our results provide a mechanistic basis for the dichotomous functions of TAp73 in the regulation of cellular growth through its distinct TADs.


Asunto(s)
Proliferación Celular , Dominios Proteicos , Activación Transcripcional , Proteína Tumoral p73 , Proteína Tumoral p73/metabolismo , Proteína Tumoral p73/genética , Humanos , Movimiento Celular/genética , Mutación , Línea Celular Tumoral , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/genética , Proteína p53 Supresora de Tumor/metabolismo , Proteína p53 Supresora de Tumor/genética , Fosforilación , Proteínas Supresoras de Tumor/metabolismo , Proteínas Supresoras de Tumor/genética
5.
EMBO J ; 41(8): e108443, 2022 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-35112738

RESUMEN

Post-translational modifications (PTMs) have emerged as key modulators of protein phase separation and have been linked to protein aggregation in neurodegenerative disorders. The major aggregating protein in amyotrophic lateral sclerosis and frontotemporal dementia, the RNA-binding protein TAR DNA-binding protein (TDP-43), is hyperphosphorylated in disease on several C-terminal serine residues, a process generally believed to promote TDP-43 aggregation. Here, we however find that Casein kinase 1δ-mediated TDP-43 hyperphosphorylation or C-terminal phosphomimetic mutations reduce TDP-43 phase separation and aggregation, and instead render TDP-43 condensates more liquid-like and dynamic. Multi-scale molecular dynamics simulations reveal reduced homotypic interactions of TDP-43 low-complexity domains through enhanced solvation of phosphomimetic residues. Cellular experiments show that phosphomimetic substitutions do not affect nuclear import or RNA regulatory functions of TDP-43, but suppress accumulation of TDP-43 in membrane-less organelles and promote its solubility in neurons. We speculate that TDP-43 hyperphosphorylation may be a protective cellular response to counteract TDP-43 aggregation.


Asunto(s)
Esclerosis Amiotrófica Lateral , Demencia Frontotemporal , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/metabolismo , Proteínas de Unión al ADN/metabolismo , Humanos , Agregado de Proteínas , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo
6.
Cell ; 144(4): 566-76, 2011 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-21335238

RESUMEN

TAp63α, a homolog of the p53 tumor suppressor, is a quality control factor in the female germline. Remarkably, already undamaged oocytes express high levels of the protein, suggesting that TAp63α's activity is under tight control of an inhibitory mechanism. Biochemical studies have proposed that inhibition requires the C-terminal transactivation inhibitory domain. However, the structural mechanism of TAp63α inhibition remains unknown. Here, we show that TAp63α is kept in an inactive dimeric state. We reveal that relief of inhibition leads to tetramer formation with ∼20-fold higher DNA affinity. In vivo, phosphorylation-triggered tetramerization of TAp63α is not reversible by dephosphorylation. Furthermore, we show that a helix in the oligomerization domain of p63 is crucial for tetramer stabilization and competes with the transactivation domain for the same binding site. Our results demonstrate how TAp63α is inhibited by complex domain-domain interactions that provide the basis for regulating quality control in oocytes.


Asunto(s)
Oocitos/metabolismo , Fosfoproteínas/química , Fosfoproteínas/metabolismo , Transactivadores/química , Transactivadores/metabolismo , Animales , ADN/metabolismo , Dimerización , Femenino , Rayos gamma , Ratones , Modelos Moleculares , Fosforilación , Multimerización de Proteína , Proteína p53 Supresora de Tumor/metabolismo
7.
EMBO J ; 40(6): e106094, 2021 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-33576509

RESUMEN

The assembly of a specific polymeric ubiquitin chain on a target protein is a key event in the regulation of numerous cellular processes. Yet, the mechanisms that govern the selective synthesis of particular polyubiquitin signals remain enigmatic. The homologous ubiquitin-conjugating (E2) enzymes Ubc1 (budding yeast) and Ube2K (mammals) exclusively generate polyubiquitin linked through lysine 48 (K48). Uniquely among E2 enzymes, Ubc1 and Ube2K harbor a ubiquitin-binding UBA domain with unknown function. We found that this UBA domain preferentially interacts with ubiquitin chains linked through lysine 63 (K63). Based on structural modeling, in vitro ubiquitination experiments, and NMR studies, we propose that the UBA domain aligns Ubc1 with K63-linked polyubiquitin and facilitates the selective assembly of K48/K63-branched ubiquitin conjugates. Genetic and proteomics experiments link the activity of the UBA domain, and hence the formation of this unusual ubiquitin chain topology, to the maintenance of cellular proteostasis.


Asunto(s)
Poliubiquitina/biosíntesis , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Enzimas Ubiquitina-Conjugadoras/metabolismo , Ubiquitinación/fisiología , Simulación por Computador , Modelos Estructurales , Dominios Proteicos , Proteómica , Proteínas de Saccharomyces cerevisiae/genética , Transducción de Señal/fisiología , Enzimas Ubiquitina-Conjugadoras/genética
8.
Nucleic Acids Res ; 51(12): 6307-6320, 2023 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-37224528

RESUMEN

DNA mismatch repair (MMR) is essential for correction of DNA replication errors. Germline mutations of the human MMR gene MLH1 are the major cause of Lynch syndrome, a heritable cancer predisposition. In the MLH1 protein, a non-conserved, intrinsically disordered region connects two conserved, catalytically active structured domains of MLH1. This region has as yet been regarded as a flexible spacer, and missense alterations in this region have been considered non-pathogenic. However, we have identified and investigated a small motif (ConMot) in this linker which is conserved in eukaryotes. Deletion of the ConMot or scrambling of the motif abolished mismatch repair activity. A mutation from a cancer family within the motif (p.Arg385Pro) also inactivated MMR, suggesting that ConMot alterations can be causative for Lynch syndrome. Intriguingly, the mismatch repair defect of the ConMot variants could be restored by addition of a ConMot peptide containing the deleted sequence. This is the first instance of a DNA mismatch repair defect conferred by a mutation that can be overcome by addition of a small molecule. Based on the experimental data and AlphaFold2 predictions, we suggest that the ConMot may bind close to the C-terminal MLH1-PMS2 endonuclease and modulate its activation during the MMR process.


Asunto(s)
Neoplasias Colorrectales Hereditarias sin Poliposis , Reparación de la Incompatibilidad de ADN , Homólogo 1 de la Proteína MutL , Humanos , Neoplasias Colorrectales Hereditarias sin Poliposis/genética , Neoplasias Colorrectales Hereditarias sin Poliposis/patología , Predisposición Genética a la Enfermedad , Mutación de Línea Germinal , Mutación , Homólogo 1 de la Proteína MutL/genética , Homólogo 1 de la Proteína MutL/metabolismo , Proteína 2 Homóloga a MutS/genética , Proteína 2 Homóloga a MutS/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo
9.
Mol Cell ; 62(6): 918-928, 2016 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-27264873

RESUMEN

Ubiquitin conjugation is an essential process modulating protein function in eukaryotic cells. Surprisingly, little is known about how the progressive assembly of ubiquitin chains is managed by the responsible enzymes. Only recently has ubiquitin binding activity emerged as an important factor in chain formation. The Ubc7 activator Cue1 carries a ubiquitin binding CUE domain that substantially stimulates K48-linked polyubiquitination mediated by Ubc7. Our results from NMR-based analysis and in vitro ubiquitination reactions point out that two parameters accelerate ubiquitin chain assembly: the increasing number of CUE binding sites and the position of CUE binding within a growing chain. In particular, interactions with a ubiquitin moiety adjacent to the acceptor ubiquitin facilitate chain elongation. These data indicate a mechanism for ubiquitin binding in which Cue1 positions Ubc7 and the distal acceptor ubiquitin for rapid polyubiquitination. Disrupting this mechanism results in dysfunction of the ERAD pathway by a delayed turnover of substrates.


Asunto(s)
Proteínas Portadoras/metabolismo , Degradación Asociada con el Retículo Endoplásmico , Proteínas de la Membrana/metabolismo , Poliubiquitina/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/enzimología , Enzimas Ubiquitina-Conjugadoras/metabolismo , Ubiquitinación , Proteínas Portadoras/química , Proteínas Portadoras/genética , Interacciones Hidrofóbicas e Hidrofílicas , Cinética , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Modelos Moleculares , Mutación , Unión Proteica , Conformación Proteica en Hélice alfa , Dominios y Motivos de Interacción de Proteínas , Proteolisis , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/genética , Espectrometría de Fluorescencia , Relación Estructura-Actividad , Especificidad por Sustrato , Enzimas Ubiquitina-Conjugadoras/química , Enzimas Ubiquitina-Conjugadoras/genética
10.
J Gen Virol ; 104(9)2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37750869

RESUMEN

Hepatitis C virus (HCV) infects millions of people worldwide and is a leading cause of liver disease. Despite recent advances in antiviral therapies, viral resistance can limit drug efficacy and understanding the mechanisms that confer viral escape is important. We employ an unbiased interactome analysis to discover host binding partners of the HCV non-structural protein 5A (NS5A), a key player in viral replication and assembly. We identify ASPP2, apoptosis-stimulating protein of p53, as a new host co-factor that binds NS5A via its SH3 domain. Importantly, silencing ASPP2 reduces viral replication and spread. Our study uncovers a previously unknown role for ASPP2 to potentiate HCV RNA replication.


Asunto(s)
Hepacivirus , Hepatitis C , Humanos , Hepacivirus/genética , Dominios Homologos src , Replicación Viral , Proteínas no Estructurales Virales/metabolismo , Dominios Proteicos
11.
J Biomol NMR ; 77(4): 131-147, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37311907

RESUMEN

Cell-free (CF) synthesis with highly productive E. coli lysates is a convenient method to produce labeled proteins for NMR studies. Despite reduced metabolic activity in CF lysates, a certain scrambling of supplied isotope labels is still notable. Most problematic are conversions of 15N labels of the amino acids L-Asp, L-Asn, L-Gln, L-Glu and L-Ala, resulting in ambiguous NMR signals as well as in label dilution. Specific inhibitor cocktails suppress most undesired conversion reactions, while limited availability and potential side effects on CF system productivity need to be considered. As alternative route to address NMR label conversion in CF systems, we describe the generation of optimized E. coli lysates with reduced amino acid scrambling activity. Our strategy is based on the proteome blueprint of standardized CF S30 lysates of the E. coli strain A19. Identified lysate enzymes with suspected amino acid scrambling activity were eliminated by engineering corresponding single and cumulative chromosomal mutations in A19. CF lysates prepared from the mutants were analyzed for their CF protein synthesis efficiency and for residual scrambling activity. The A19 derivative "Stablelabel" containing the cumulative mutations asnA, ansA/B, glnA, aspC and ilvE yielded the most useful CF S30 lysates. We demonstrate the optimized NMR spectral complexity of selectively labeled proteins CF synthesized in "Stablelabel" lysates. By taking advantage of ilvE deletion in "Stablelabel", we further exemplify a new strategy for methyl group specific labeling of membrane proteins with the proton pump proteorhodopsin.


Asunto(s)
Aminoácidos , Escherichia coli , Escherichia coli/metabolismo , Resonancia Magnética Nuclear Biomolecular/métodos , Aminoácidos/química , Proteínas/química , Biosíntesis de Proteínas , Marcaje Isotópico/métodos , Sistema Libre de Células/metabolismo
12.
J Biomol NMR ; 77(5-6): 261-269, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37966668

RESUMEN

Many proteins can adopt multiple conformations which are important for their function. This is also true for proteins and domains that are covalently linked to each other. One important example is ubiquitin, which can form chains of different conformations depending on which of its lysine side chains is used to form an isopeptide bond with the C-terminus of another ubiquitin molecule. Similarly, ubiquitin gets covalently attached to active-site residues of E2 ubiquitin-conjugating enzymes. Due to weak interactions between ubiquitin and its interaction partners, these covalent complexes adopt multiple conformations. Understanding the function of these complexes requires the characterization of the entire accessible conformation space and its modulation by interaction partners. Long-range (1.8-10 nm) distance restraints obtained by EPR spectroscopy in the form of probability distributions are ideally suited for this task as not only the mean distance but also information about the conformation dynamics is encoded in the experimental data. Here we describe a computational method that we have developed based on well-established structure determination software using NMR restraints to calculate the accessible conformation space using PELDOR/DEER data.


Asunto(s)
Ubiquitina , Modelos Moleculares , Espectroscopía de Resonancia por Spin del Electrón/métodos , Resonancia Magnética Nuclear Biomolecular , Ubiquitina/metabolismo , Dominio Catalítico
13.
Mol Cell ; 57(6): 995-1010, 2015 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-25684205

RESUMEN

The small Rho GTPase RAC1 is an essential regulator of cellular signaling that controls actin rearrangements and cell motility. Here, we identify a novel CUL3 RING ubiquitin ligase complex, containing the substrate adaptors KBTBD6 and KBTBD7, that mediates ubiquitylation and proteasomal degradation of TIAM1, a RAC1-specific GEF. Increasing the abundance of TIAM1 by depletion of KBTBD6 and/or KBTBD7 leads to elevated RAC1 activity, changes in actin morphology, loss of focal adhesions, reduced proliferation, and enhanced invasion. KBTBD6 and KBTBD7 employ ATG8 family-interacting motifs to bind preferentially to GABARAP proteins. Surprisingly, ubiquitylation and degradation of TIAM1 by CUL3(KBTBD6/KBTBD7) depends on its binding to GABARAP proteins. Our study reveals that recruitment of CUL3(KBTBD6/KBTBD7) to GABARAP-containing vesicles regulates the abundance of membrane-associated TIAM1 and subsequently spatially restricted RAC1 signaling. Besides their role in autophagy and trafficking, we uncovered a previously unknown function of GABARAP proteins as membrane-localized signaling scaffolds.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Cullin/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Transactivadores/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Proteínas Adaptadoras Transductoras de Señales/química , Proteínas Adaptadoras Transductoras de Señales/genética , Secuencia de Aminoácidos , Proteínas Reguladoras de la Apoptosis , Familia de las Proteínas 8 Relacionadas con la Autofagia , Proteínas Cullin/genética , Factores de Intercambio de Guanina Nucleótido/genética , Humanos , Péptidos y Proteínas de Señalización Intracelular , Proteínas de Microfilamentos/metabolismo , Proteínas Asociadas a Microtúbulos/química , Proteínas Asociadas a Microtúbulos/genética , Modelos Moleculares , Datos de Secuencia Molecular , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Resonancia Magnética Nuclear Biomolecular , Conformación Proteica , Multimerización de Proteína , Transducción de Señal , Proteína 1 de Invasión e Inducción de Metástasis del Linfoma-T , Transactivadores/genética , Ubiquitinación , Proteína de Unión al GTP rac1/genética
14.
Mol Cell ; 60(1): 89-104, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-26431026

RESUMEN

Hereditary spastic paraplegias (HSPs) are a diverse group of neurodegenerative diseases that are characterized by axonopathy of the corticospinal motor neurons. A mutation in the gene encoding for Tectonin ß-propeller containing protein 2 (TECPR2) causes HSP that is complicated by neurological symptoms. While TECPR2 is a human ATG8 binding protein and positive regulator of autophagy, the exact function of TECPR2 is unknown. Here, we show that TECPR2 associates with several trafficking components, among them the COPII coat protein SEC24D. TECPR2 is required for stabilization of SEC24D protein levels, maintenance of functional ER exit sites (ERES), and efficient ER export in a manner dependent on binding to lipidated LC3C. TECPR2-deficient HSP patient cells display alterations in SEC24D abundance and ER export efficiency. Additionally, TECPR2 and LC3C are required for autophagosome formation, possibly through maintaining functional ERES. Collectively, these results reveal that TECPR2 functions as molecular scaffold linking early secretion pathway and autophagy.


Asunto(s)
Autofagia , Proteínas Portadoras/metabolismo , Retículo Endoplásmico/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Transporte de Proteínas , Paraplejía Espástica Hereditaria/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Reguladoras de la Apoptosis , Proteínas Portadoras/genética , Células HeLa , Humanos , Mutación , Proteínas del Tejido Nervioso/genética , Paraplejía Espástica Hereditaria/metabolismo , Proteínas de Transporte Vesicular/metabolismo
15.
Hum Mutat ; 43(10): 1443-1453, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35801529

RESUMEN

Premature ovarian insufficiency (POI) is a leading form of female infertility, characterised by menstrual disturbance and elevated follicle-stimulating hormone before age 40. It is highly heterogeneous with variants in over 80 genes potentially causative, but the majority of cases having no known cause. One gene implicated in POI pathology is TP63. TP63 encodes multiple p63 isoforms, one of which has been shown to have a role in the surveillance of genetic quality in oocytes. TP63 C-terminal truncation variants and N-terminal duplication have been described in association with POI, however, functional validation has been lacking. Here we identify three novel TP63 missense variants in women with nonsyndromic POI, including one in the N-terminal activation domain, one in the C-terminal inhibition domain, and one affecting a unique and poorly understood p63 isoform, TA*p63. Via blue-native page and luciferase reporter assays we demonstrate that two of these variants disrupt p63 dimerization, leading to constitutively active p63 tetramer that significantly increases the transcription of downstream targets. This is the first evidence that TP63 missense variants can cause isolated POI and provides mechanistic insight that TP63 variants cause POI due to constitutive p63 activation and accelerated oocyte loss in the absence of DNA damage.


Asunto(s)
Insuficiencia Ovárica Primaria , Factores de Transcripción , Proteínas Supresoras de Tumor , Femenino , Humanos , Mutación Missense , Insuficiencia Ovárica Primaria/genética , Factores de Transcripción/genética , Proteínas Supresoras de Tumor/genética
16.
Nat Chem Biol ; 16(10): 1078-1086, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32719556

RESUMEN

The p53 homolog TAp63α is the transcriptional key regulator of genome integrity in oocytes. After DNA damage, TAp63α is activated by multistep phosphorylation involving multiple phosphorylation events by the kinase CK1, which triggers the transition from a dimeric and inactive conformation to an open and active tetramer that initiates apoptosis. By measuring activation kinetics in ovaries and single-site phosphorylation kinetics in vitro with peptides and full-length protein, we show that TAp63α phosphorylation follows a biphasic behavior. Although the first two CK1 phosphorylation events are fast, the third one, which constitutes the decisive step to form the active conformation, is slow. Structure determination of CK1 in complex with differently phosphorylated peptides reveals the structural mechanism for the difference in the kinetic behavior based on an unusual CK1/TAp63α substrate interaction in which the product of one phosphorylation step acts as an inhibitor for the following one.


Asunto(s)
Apoptosis/fisiología , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Dominio Catalítico , Daño del ADN , Femenino , Humanos , Ratones , Modelos Moleculares , Simulación de Dinámica Molecular , Oocitos , Fosforilación , Conformación Proteica , Factores de Tiempo , Factores de Transcripción/genética , Proteínas Supresoras de Tumor/genética
17.
Mol Cell ; 53(2): 167-78, 2014 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-24462201

RESUMEN

Selective autophagy ensures recognition and removal of various cytosolic cargoes. Hence, aggregated proteins, damaged organelles, or pathogens are enclosed into the double-membrane vesicle, the autophagosome, and delivered to the lysosome for degradation. This process is mediated by selective autophagy receptors, such as p62/SQSTM1. These proteins recognize autophagic cargo and, via binding to small ubiquitin-like modifiers (UBLs)--Atg8/LC3/GABARAPs and ATG5--mediate formation of selective autophagosomes. Recently, it was found that UBLs can directly engage the autophagosome nucleation machinery. Here, we review recent findings on selective autophagy and propose a model for selective autophagosome formation in close proximity to cargo.


Asunto(s)
Autofagia/fisiología , Modelos Biológicos , Ubiquitinas/fisiología , Modelos Moleculares , Peroxisomas/metabolismo , Transducción de Señal , Ubiquitinación , Ubiquitinas/metabolismo
18.
J Biol Chem ; 295(8): 2160-2174, 2020 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-31937588

RESUMEN

Deubiquitinases (DUBs) are vital for the regulation of ubiquitin signals, and both catalytic activity of and target recruitment by DUBs need to be tightly controlled. Here, we identify asparagine hydroxylation as a novel posttranslational modification involved in the regulation of Cezanne (also known as OTU domain-containing protein 7B (OTUD7B)), a DUB that controls key cellular functions and signaling pathways. We demonstrate that Cezanne is a substrate for factor inhibiting HIF1 (FIH1)- and oxygen-dependent asparagine hydroxylation. We found that FIH1 modifies Asn35 within the uncharacterized N-terminal ubiquitin-associated (UBA)-like domain of Cezanne (UBACez), which lacks conserved UBA domain properties. We show that UBACez binds Lys11-, Lys48-, Lys63-, and Met1-linked ubiquitin chains in vitro, establishing UBACez as a functional ubiquitin-binding domain. Our findings also reveal that the interaction of UBACez with ubiquitin is mediated via a noncanonical surface and that hydroxylation of Asn35 inhibits ubiquitin binding. Recently, it has been suggested that Cezanne recruitment to specific target proteins depends on UBACez Our results indicate that UBACez can indeed fulfill this role as regulatory domain by binding various ubiquitin chain types. They also uncover that this interaction with ubiquitin, and thus with modified substrates, can be modulated by oxygen-dependent asparagine hydroxylation, suggesting that Cezanne is regulated by oxygen levels.


Asunto(s)
Asparagina/metabolismo , Endopeptidasas/química , Endopeptidasas/metabolismo , Oxígeno/metabolismo , Ubiquitina/metabolismo , Secuencia de Aminoácidos , Secuencia de Consenso , Células HEK293 , Humanos , Hidroxilación , Oxigenasas de Función Mixta/metabolismo , Poliubiquitina/metabolismo , Unión Proteica , Dominios Proteicos , Proteínas Represoras/metabolismo , Relación Estructura-Actividad
19.
EMBO J ; 36(4): 549-564, 2017 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-28028054

RESUMEN

Autophagy is a cellular surveillance pathway that balances metabolic and energy resources and transports specific cargos, including damaged mitochondria, other broken organelles, or pathogens for degradation to the lysosome. Central components of autophagosomal biogenesis are six members of the LC3 and GABARAP family of ubiquitin-like proteins (mATG8s). We used phage display to isolate peptides that possess bona fide LIR (LC3-interacting region) properties and are selective for individual mATG8 isoforms. Sensitivity of the developed sensors was optimized by multiplication, charge distribution, and fusion with a membrane recruitment (FYVE) or an oligomerization (PB1) domain. We demonstrate the use of the engineered peptides as intracellular sensors that recognize specifically GABARAP, GABL1, GABL2, and LC3C, as well as a bispecific sensor for LC3A and LC3B. By using an LC3C-specific sensor, we were able to monitor recruitment of endogenous LC3C to Salmonella during xenophagy, as well as to mitochondria during mitophagy. The sensors are general tools to monitor the fate of mATG8s and will be valuable in decoding the biological functions of the individual LC3/GABARAPs.


Asunto(s)
Familia de las Proteínas 8 Relacionadas con la Autofagia/análisis , Autofagia , Técnicas Biosensibles/métodos , Coloración y Etiquetado/métodos , Línea Celular , Fluorescencia , Humanos , Mitocondrias/metabolismo , Biblioteca de Péptidos , Unión Proteica , Salmonella/inmunología
20.
Proc Natl Acad Sci U S A ; 115(5): E906-E915, 2018 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-29339502

RESUMEN

The p63 gene encodes a master regulator of epidermal commitment, development, and differentiation. Heterozygous mutations in the C-terminal domain of the p63 gene can cause ankyloblepharon-ectodermal defects-cleft lip/palate (AEC) syndrome, a life-threatening disorder characterized by skin fragility and severe, long-lasting skin erosions. Despite deep knowledge of p63 functions, little is known about mechanisms underlying disease pathology and possible treatments. Here, we show that multiple AEC-associated p63 mutations, but not those causative of other diseases, lead to thermodynamic protein destabilization, misfolding, and aggregation, similar to the known p53 gain-of-function mutants found in cancer. AEC mutant proteins exhibit impaired DNA binding and transcriptional activity, leading to dominant negative effects due to coaggregation with wild-type p63 and p73. Importantly, p63 aggregation occurs also in a conditional knock-in mouse model for the disorder, in which the misfolded p63 mutant protein leads to severe epidermal defects. Variants of p63 that abolish aggregation of the mutant proteins are able to rescue p63's transcriptional function in reporter assays as well as in a human fibroblast-to-keratinocyte conversion assay. Our studies reveal that AEC syndrome is a protein aggregation disorder and opens avenues for therapeutic intervention.


Asunto(s)
Labio Leporino/genética , Fisura del Paladar/genética , Anomalías del Ojo/genética , Fosfoproteínas/genética , Piel/patología , Transactivadores/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Animales , Ectodermo/metabolismo , Mutación del Sistema de Lectura , Células HEK293 , Heterocigoto , Humanos , Ratones , Mutación , Mutación Missense , Unión Proteica , Desnaturalización Proteica , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA