Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Proc Natl Acad Sci U S A ; 118(5)2021 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-33495326

RESUMO

Adaptation to different forms of environmental stress is crucial for maintaining essential cellular functions and survival. The nucleolus plays a decisive role as a signaling hub for coordinating cellular responses to various extrinsic and intrinsic cues. p53 levels are normally kept low in unstressed cells, mainly due to E3 ubiquitin ligase MDM2-mediated degradation. Under stress, nucleophosmin (NPM) relocates from the nucleolus to the nucleoplasm and binds MDM2, thereby preventing degradation of p53 and allowing cell-cycle arrest and DNA repair. Here, we demonstrate that the mammalian sirtuin SIRT7 is an essential component for the regulation of p53 stability during stress responses induced by ultraviolet (UV) irradiation. The catalytic activity of SIRT7 is substantially increased upon UV irradiation through ataxia telangiectasia mutated and Rad3 related (ATR)-mediated phosphorylation, which promotes efficient deacetylation of the SIRT7 target NPM. Deacetylation is required for stress-dependent relocation of NPM into the nucleoplasm and MDM2 binding, thereby preventing ubiquitination and degradation of p53. In the absence of SIRT7, stress-dependent stabilization of p53 is abrogated, both in vitro and in vivo, impairing cellular stress responses. The study uncovers an essential SIRT7-dependent mechanism for stabilization of the tumor suppressor p53 in response to genotoxic stress.


Assuntos
Dano ao DNA , Proteínas Nucleares/metabolismo , Sirtuínas/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Raios Ultravioleta , Acetilação/efeitos da radiação , Animais , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Catálise/efeitos da radiação , Linhagem Celular Tumoral , Nucléolo Celular/metabolismo , Nucléolo Celular/efeitos da radiação , Humanos , Lisina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Nucleofosmina , Fosforilação/efeitos da radiação , Estabilidade Proteica/efeitos da radiação , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Transcrição Gênica/efeitos da radiação , Ubiquitinação/efeitos da radiação
2.
Anal Chem ; 95(27): 10265-10278, 2023 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-37369001

RESUMO

Multispecific antibodies have gained significant importance in a broad indication space due to their ability to engage multiple epitopes simultaneously and to thereby overcome therapeutic barriers. With growing therapeutic potential, however, the molecular complexity increases, thus intensifying the demand for innovative protein engineering and analytical strategies. A major challenge for multispecific antibodies is the correct assembly of light and heavy chains. Engineering strategies exist to stabilize the correct pairing, but typically individual engineering campaigns are required to arrive at the anticipated format. Mass spectrometry has proven to be a versatile tool to identify mispaired species. However, due to manual data analysis procedures, mass spectrometry is limited to lower throughputs. To keep pace with increasing sample numbers, we developed a high-throughput-capable mispairing workflow based on intact mass spectrometry with automated data analysis, peak detection, and relative quantification using Genedata Expressionist. This workflow is capable of detecting mispaired species of ∼1000 multispecific antibodies in three weeks and thus is applicable to complex screening campaigns. As a proof of concept, the assay was applied to engineering a trispecific antibody. Strikingly, the new setup has not only proved successful in mispairing analysis but has also revealed its potential to automatically annotate other product-related impurities. Furthermore, we could confirm the assay to be format-agnostic, as shown by analyzing several different multispecific formats in one run. With these comprehensive capabilities, the new automated intact mass workflow can be applied as a universal tool to detect and annotate peaks in a format-agnostic approach and in high-throughput, thus enabling complex discovery campaigns.


Assuntos
Anticorpos , Espectrometria de Massas , Epitopos
3.
Nature ; 524(7564): 230-3, 2015 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-26168398

RESUMO

Cells sense their environment and adapt to it by fine-tuning their transcriptome. Wired into this network of gene expression control are mechanisms to compensate for gene dosage. The increasing use of reverse genetics in zebrafish, and other model systems, has revealed profound differences between the phenotypes caused by genetic mutations and those caused by gene knockdowns at many loci, an observation previously reported in mouse and Arabidopsis. To identify the reasons underlying the phenotypic differences between mutants and knockdowns, we generated mutations in zebrafish egfl7, an endothelial extracellular matrix gene of therapeutic interest, as well as in vegfaa. Here we show that egfl7 mutants do not show any obvious phenotypes while animals injected with egfl7 morpholino (morphants) exhibit severe vascular defects. We further observe that egfl7 mutants are less sensitive than their wild-type siblings to Egfl7 knockdown, arguing against residual protein function in the mutants or significant off-target effects of the morpholinos when used at a moderate dose. Comparing egfl7 mutant and morphant proteomes and transcriptomes, we identify a set of proteins and genes that are upregulated in mutants but not in morphants. Among them are extracellular matrix genes that can rescue egfl7 morphants, indicating that they could be compensating for the loss of Egfl7 function in the phenotypically wild-type egfl7 mutants. Moreover, egfl7 CRISPR interference, which obstructs transcript elongation and causes severe vascular defects, does not cause the upregulation of these genes. Similarly, vegfaa mutants but not morphants show an upregulation of vegfab. Taken together, these data reveal the activation of a compensatory network to buffer against deleterious mutations, which was not observed after translational or transcriptional knockdown.


Assuntos
Técnicas de Silenciamento de Genes , Mutação/genética , Fenótipo , Interferência de RNA , Supressão Genética/genética , Regulação para Cima/genética , Peixe-Zebra/genética , Alelos , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Sistemas CRISPR-Cas/genética , Larva/genética , Glicoproteínas de Membrana/genética , Morfolinos/genética , Proteoma/análise , Transcriptoma/genética , Fator A de Crescimento do Endotélio Vascular/genética , Peixe-Zebra/embriologia , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/genética
4.
EMBO Rep ; 19(11)2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30201799

RESUMO

Post-translational modifications by ubiquitin-related SUMO modifiers regulate cellular signaling networks and protein homeostasis. While SUMO1 is mainly conjugated to proteins as a monomer, SUMO2/3 can form polymeric chains. Poly-SUMOylation is best understood in the SUMO-targeted ubiquitin ligase (StUbL) pathway, where chains prime proteins for subsequent ubiquitylation by StUbLs. SUMO chains typically form in response to genotoxic or proteotoxic stress and are preferentially linked via lysine 11 of SUMO2/3. Here, we report that K11 of SUMO2/3 undergoes reversible acetylation with SIRT1 being the K11 deacetylase. In a purified in vitro system, acetylation of SUMO2/3 impairs chain formation and restricts chain length. In a cellular context, however, K11 acetyl-mimicking SUMO2 does not affect the StUbL pathway, indicating that in cells non-canonical chains are more prevalent. MS-based SUMO proteomics indeed identified non-canonical chain types under basal and stress conditions. Importantly, mimicking K11 acetylation alters chain architecture by favoring K5- and K35-linked chains, while inhibiting K7 and K21 linkages. These data provide insight into SUMO chain signaling and point to a role of K11 acetylation as a modulator of SUMO2/3 chains.


Assuntos
Lisina/metabolismo , Sirtuína 1/metabolismo , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Acetilação , Células HeLa , Resposta ao Choque Térmico , Humanos , Proteína da Leucemia Promielocítica/metabolismo , Transdução de Sinais , Sirtuína 1/genética , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/química , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/genética , Ubiquitinas/metabolismo
5.
PLoS Genet ; 12(6): e1006099, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27294373

RESUMO

Skeletal muscles provide metazoans with the ability to feed, reproduce and avoid predators. In humans, a heterogeneous group of genetic diseases, termed muscular dystrophies (MD), lead to skeletal muscle dysfunction. Mutations in the gene encoding Caveolin-3, a principal component of the membrane micro-domains known as caveolae, cause defects in muscle maintenance and function; however it remains unclear how caveolae dysfunction underlies MD pathology. The Cavin family of caveolar proteins can form membrane remodeling oligomers and thus may also impact skeletal muscle function. Changes in the distribution and function of Cavin4/Murc, which is predominantly expressed in striated muscles, have been reported to alter caveolae structure through interaction with Caveolin-3. Here, we report the generation and phenotypic analysis of murcb mutant zebrafish, which display impaired swimming capacity, skeletal muscle fibrosis and T-tubule abnormalities during development. To understand the mechanistic importance of Murc loss of function, we assessed Caveolin-1 and 3 localization and found it to be abnormal. We further identified an in vivo function for Murc in Erk signaling. These data link Murc with developmental defects in T-tubule formation and progressive muscle dysfunction, thereby providing a new candidate for the etiology of muscular dystrophy.


Assuntos
Desenvolvimento Muscular/genética , Proteínas Musculares/genética , Músculo Esquelético/embriologia , Músculo Esquelético/patologia , Proteínas de Peixe-Zebra/genética , Peixe-Zebra/embriologia , Animais , Animais Geneticamente Modificados , Cavéolas/metabolismo , Caveolina 1/metabolismo , Caveolina 3/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Canais Iônicos/genética , Proteínas Musculares/metabolismo , Distrofias Musculares/genética , Peixe-Zebra/genética , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/metabolismo
6.
J Proteome Res ; 17(10): 3333-3347, 2018 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-30142977

RESUMO

Skeletal muscles are composed of heterogeneous collections of fibers with different metabolic profiles. With varied neuronal innervation and fiber-type compositions, each muscle fulfils specific functions and responds differently to stimuli and perturbations. We assessed individual fibers by mass spectrometry to dissect protein changes after loss of neuronal innervation due to section of the sciatic nerve in mice. This proteomics approach enabled us to quantify ∼600 proteins per individual soleus and EDL (extensor digitorum longus) muscle fiber. Expression of myosin heavy chain (MyHC) in individual fibers enabled clustering of specific fiber types; comparison of fibers from control and denervated muscles with the same MyHC expression revealed restricted regulation of a total of 240 proteins in type-I, -IIa, or -IIb fibers 7 days after denervation. The levels of several mitochondrial and proteasomal proteins were significantly altered, indicating rapid adaption of metabolic processes after denervation. Furthermore, we observed fiber-type-specific regulation of proteins involved in calcium ion binding and transport, such as troponins, parvalbumin, and ATP2A2, indicating marked remodeling of muscle contractility after denervation. This study provides novel insight into how different muscle fiber types remodel their proteomes during muscular atrophy.


Assuntos
Fibras Musculares de Contração Rápida/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares de Contração Lenta/metabolismo , Proteoma/metabolismo , Proteômica/métodos , Animais , Camundongos Endogâmicos C57BL , Proteínas Mitocondriais/metabolismo , Contração Muscular , Denervação Muscular , Proteínas Musculares/metabolismo , Músculo Esquelético/inervação , Músculo Esquelético/metabolismo , Atrofia Muscular/metabolismo , Atrofia Muscular/fisiopatologia , Cadeias Pesadas de Miosina/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo
7.
Biochim Biophys Acta ; 1853(10 Pt A): 2580-91, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26142927

RESUMO

Proteostasis is crucial for life and maintained by cellular chaperones and proteases. One major mitochondrial protease is the ClpXP complex, which is comprised of a catalytic ClpX subunit and a proteolytic ClpP subunit. Based on two separate observations, we hypothesized that ClpX may play a leading role in the cellular function of ClpXP. Therefore, we analyzed the effect of ClpX overexpression on a myoblast proteome by quantitative proteomics. ClpX overexpression results in the upregulation of markers of the mitochondrial proteostasis pathway, known as the "mitochondrial unfolded protein response" (UPRmt). Although this pathway is described in detail in Caenorhabditis elegans, it is not clear whether it is conserved in mammals. Therefore, we compared features of the classical nematode UPRmt with our mammalian ClpX-triggered UPRmt dataset. We show that they share the same retrograde mitochondria-to-nucleus signaling pathway that involves the key UPRmt transcription factor CHOP (also known as Ddit3, CEBPZ or GADD153). In conclusion, our data confirm the existence of a mammalian UPRmt that has great similarity to the C. elegans pathway. Furthermore, our results illustrate that ClpX overexpression is a good and simple model to study the underlying mechanisms of the UPRmt in mammalian cells.


Assuntos
Endopeptidase Clp/biossíntese , Mitocôndrias/enzimologia , Proteínas Mitocondriais/biossíntese , Resposta a Proteínas não Dobradas/fisiologia , Animais , Endopeptidase Clp/genética , Células HEK293 , Humanos , Camundongos , Mitocôndrias/genética , Proteínas Mitocondriais/genética , Complexos Multienzimáticos/genética , Complexos Multienzimáticos/metabolismo , Fator de Transcrição CHOP/genética , Fator de Transcrição CHOP/metabolismo
8.
Proteomics ; 15(4): 739-51, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25504979

RESUMO

The zebrafish owns remarkable regenerative capacities allowing regeneration of several tissues, including the heart, liver, and brain. To identify protein dynamics during fin regeneration we used a pulsed SILAC approach that enabled us to detect the incorporation of (13) C6 -lysine (Lys6) into newly synthesized proteins. Samples were taken at four different time points from noninjured and regrowing fins and incorporation rates were monitored using a combination of single-shot 4-h gradients and high-resolution tandem MS. We identified more than 5000 labeled proteins during the first 3 weeks of fin regeneration and were able to monitor proteins that are responsible for initializing and restoring the shape of these appendages. The comparison of Lys6 incorporation rates between noninjured and regrowing fins enabled us to identify proteins that are directly involved in regeneration. For example, we observed increased incorporation rates of two actinodin family members at the actinotrichia, which is a hairlike fiber structure at the tip of regrowing fins. Moreover, we used quantitative real-time RNA measurements of several candidate genes, including osteoglycin, si:ch211-288h17.3, and prostaglandin reductase 1 to correlate the mRNA expression to Lys6 incorporation data. This novel pulsed SILAC methodology in fish can be used as a versatile tool to monitor newly synthesized proteins and will help to characterize protein dynamics during regenerative processes in zebrafish beyond fin regeneration.


Assuntos
Nadadeiras de Animais/metabolismo , Proteoma/análise , Regeneração/fisiologia , Proteínas de Peixe-Zebra/análise , Nadadeiras de Animais/química , Nadadeiras de Animais/fisiologia , Animais , Regulação da Expressão Gênica/genética , Regulação da Expressão Gênica/fisiologia , Marcação por Isótopo , Reação em Cadeia da Polimerase , Proteoma/química , Proteoma/metabolismo , Proteômica , Regeneração/genética , Espectrometria de Massas em Tandem , Cicatrização/genética , Cicatrização/fisiologia , Peixe-Zebra , Proteínas de Peixe-Zebra/química , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
9.
J Biomol NMR ; 58(1): 27-35, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24306181

RESUMO

A considerable limitation of NMR spectroscopy is its inherent low sensitivity. Approximately 90 % of the measuring time is used by the spin system to return to its Boltzmann equilibrium after excitation, which is determined by (1)H-T1 in cross-polarized solid-state NMR experiments. It has been shown that sample doping by paramagnetic relaxation agents such as Cu(2+)-EDTA accelerates this process considerably resulting in enhanced sensitivity. Here, we extend this concept to Gd(3+)-complexes. Their effect on (1)H-T1 has been assessed on the membrane protein proteorhodopsin, a 7TM light-driven proton pump. A comparison between Gd(3+)-DOTA, Gd(3+)-TTAHA, covalently attached Cu(2+)-EDTA-tags and Cu(2+)-EDTA reveals a 3.2-, 2.6-, 2.4- and 2-fold improved signal-to-noise ratio per unit time due to longitudinal paramagnetic relaxation enhancement. Furthermore, Gd(3+)-DOTA shows a remarkably high relaxivity, which is 77-times higher than that of Cu(2+)-EDTA. Therefore, an order of magnitude lower dopant concentration can be used. In addition, no line-broadening effects or peak shifts have been observed on proteorhodopsin in the presence of Gd(3+)-DOTA. These favourable properties make it very useful for solid-state NMR experiments on membrane proteins.


Assuntos
Gadolínio/química , Bicamadas Lipídicas/química , Proteínas de Membrana/química , Ressonância Magnética Nuclear Biomolecular/métodos , Cobre/química , Dissulfetos/química , Ácido Edético/química , Modelos Moleculares , Estrutura Secundária de Proteína , Rodopsina/química , Rodopsinas Microbianas
10.
Anal Chem ; 86(22): 11033-7, 2014 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-25347402

RESUMO

Turnover of blood serum proteins is a vital function in mammals, but technical challenges have thus far prevented comprehensive measurements of serum protein half-lives. Here, we injected native serum from heavy stable isotope labeled (SIL) mice into nonlabeled recipients to quantify turnover of more than 200 proteins using mass spectrometry with high reproducibility and accuracy. We found a median of 19.4 h and a total range of 6-70 h for calculated half-lives. Moreover, we observed similar half-lives for proteins with equal function. To demonstrate the value and effectiveness of SILflood, we investigated the impaired serum clearance in ß2-microglobulin (B2M-/-) deficient mice. Notably, we found that serum albumin and IgG half-lives were clearly reduced in B2M deficient animals compared to control animals. Taken together, our results demonstrate that SILflood is a versatile tool to investigate serum half-lives under regular and pathological conditions in living animals.


Assuntos
Marcação por Isótopo , Soro/química , Animais , Isótopos de Carbono , Células Cultivadas , Meia-Vida , Ensaios de Triagem em Larga Escala , Imunoglobulina G/sangue , Masculino , Espectrometria de Massas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Albumina Sérica/análise , Microglobulina beta-2/deficiência
11.
Rheumatology (Oxford) ; 52(11): 1952-62, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23804219

RESUMO

OBJECTIVES: HLA-B*27:05 is associated with AS whereas HLA-B*27:09 is not associated. We hypothesized that different interactions with KIR immune receptors could contribute to the difference in disease association between HLA-B*27:05 and HLAB*27:09. Thus, the objective of this study was to compare the formation of ß2m-free heavy chain (FHC) including B27 dimers (B272) by HLA-B*27:05 and HLA-B*27:09 and their binding to KIR immunoreceptors. METHODS: We studied the formation of HLA-B*27:05 and HLA-B*27:09 heterotrimers and FHC forms including dimers in vitro and in transfected cells. We investigated HLA-B*27:05 and HLA-B*27:09 binding to KIR3DL1, KIR3DL2 and LILRB2 by FACS staining with class I tetramers and by quantifying interactions with KIR3DL2CD3ε-reporter cells and KIR3DL2-expressing NK cells. We also measured KIR expression on peripheral blood NK and CD4 T cells from 18 HLA-B*27:05 AS patients, 8 HLA-B27 negative and 12 HLA-B*27:05+ and HLA-B*27:09+ healthy controls by FACS staining. RESULTS: HLA-B*27:09 formed less B272 and FHC than HLA-B*27:05. HLA-B*27:05-expressing cells stimulated KIR3DL2CD3ε-reporter T cells more effectively. Cells expressing HLA-B*27:05 promoted KIR3DL2+ NK cell survival more strongly than HLA-B*27:09. HLA-B*27:05 and HLA-B*27:09 dimer tetramers stained KIR3DL1, KIR3DL2 and LILRB2 equivalently. Increased proportions of NK and CD4 T cells expressed KIR3DL2 in HLA-B*27:05+ AS patients compared with HLA-B*27:05+, HLA-B*27:09+ and HLA-B27- healthy controls. CONCLUSION: Differences in the formation of FHC ligands for KIR3DL2 by HLA-B*27:05 and HLA-B*27:09 could contribute to the differential association of these alleles with AS.


Assuntos
Antígeno HLA-B27/metabolismo , Cadeias Pesadas de Imunoglobulinas/metabolismo , Receptores KIR3DL2/metabolismo , Espondilite Anquilosante/genética , Adulto , Alelos , Linfócitos T CD4-Positivos/imunologia , Sobrevivência Celular/imunologia , Células Cultivadas , Técnicas de Cocultura , Feminino , Predisposição Genética para Doença , Antígeno HLA-B27/genética , Humanos , Células Matadoras Naturais/imunologia , Ligantes , Masculino , Pessoa de Meia-Idade , Espondilite Anquilosante/imunologia , Espondilite Anquilosante/metabolismo , Transfecção
12.
Nat Commun ; 14(1): 8121, 2023 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-38065954

RESUMO

Ribosome biogenesis is a multi-step process, in which a network of trans-acting factors ensures the coordinated assembly of pre-ribosomal particles in order to generate functional ribosomes. Ribosome biogenesis is tightly coordinated with cell proliferation and its perturbation activates a p53-dependent cell-cycle checkpoint. How p53-independent signalling networks connect impaired ribosome biogenesis to the cell-cycle machinery has remained largely enigmatic. We demonstrate that inactivation of the nucleolar SUMO isopeptidases SENP3 and SENP5 disturbs distinct steps of 40S and 60S ribosomal subunit assembly pathways, thereby triggering the canonical p53-dependent impaired ribosome biogenesis checkpoint. However, inactivation of SENP3 or SENP5 also induces a p53-independent checkpoint that converges on the specific downregulation of the key cell-cycle regulator CDK6. We further reveal that impaired ribosome biogenesis generally triggers the downregulation of CDK6, independent of the cellular p53 status. Altogether, these data define the role of SUMO signalling in ribosome biogenesis and unveil a p53-independent checkpoint of impaired ribosome biogenesis.


Assuntos
Cisteína Endopeptidases , Ribossomos , Proteína Supressora de Tumor p53 , Nucléolo Celular/metabolismo , Proliferação de Células , Proteínas Ribossômicas/metabolismo , Ribossomos/metabolismo , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Humanos , Cisteína Endopeptidases/genética , Cisteína Endopeptidases/metabolismo
13.
J Am Chem Soc ; 133(12): 4645-54, 2011 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-21366243

RESUMO

The proteorhodopsin (PR) family found in bacteria near the ocean's surface consists of hundreds of PR variants color-tuned to their environment. PR contains a highly conserved single histidine at position 75, which is not found in most other retinal proteins. Using (13)C and (15)N MAS NMR, we were able to prove for green PR that His75 forms a pH-dependent H-bond with the primary proton acceptor Asp97, which explains its unusually high pK(a). The functional role of His75 has been studied using site-directed mutagenesis and time-resolved optical spectroscopy: Ultrafast vis-pump/vis-probe experiments on PR(H75N) showed that the primary reaction dynamics is retained, while flash photolysis experiments revealed an accelerated photocycle. Our data show the formation of a pH-dependent His-Asp cluster which might be typical for eubacterial retinal proteins. Despite its stabilizing function, His75 was found to slow the photocycle in wild-type PR. This means that PR was not optimized by evolution for fast proton transfer, which raises questions about its true function in vivo.


Assuntos
Ácido Aspártico/química , Histidina/química , Rodopsina/química , Cor , Ligação de Hidrogênio , Concentração de Íons de Hidrogênio , Espectroscopia de Ressonância Magnética/normas , Modelos Moleculares , Padrões de Referência , Rodopsinas Microbianas
14.
Front Physiol ; 11: 240, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32351396

RESUMO

The giant protein titin performs structure-preserving functions in the sarcomere and is important for the passive stiffness (Fpassive) of cardiomyocytes. Protein kinase D (PKD) enzymes play crucial roles in regulating myocardial contraction, hypertrophy, and remodeling. PKD phosphorylates myofilament proteins, but it is not known whether the giant protein titin is also a PKD substrate. Here, we aimed to determine whether PKD phosphorylates titin and thereby modulates cardiomyocyte Fpassive in normal and failing myocardium. The phosphorylation of titin was assessed in cardiomyocyte-specific PKD knock-out mice (cKO) and human hearts using immunoblotting with a phosphoserine/threonine and a phosphosite-specific titin antibody. PKD-dependent site-specific titin phosphorylation in vivo was quantified by mass spectrometry using stable isotope labeling by amino acids in cell culture (SILAC) of SILAC-labeled mouse heart protein lysates that were mixed with lysates isolated from hearts of either wild-type control (WT) or cKO mice. Fpassive of single permeabilized cardiomyocytes was recorded before and after PKD and HSP27 administration. All-titin phosphorylation was reduced in cKO compared to WT hearts. Multiple conserved PKD-dependent phosphosites were identified within the Z-disk, A-band and M-band regions of titin by quantitative mass spectrometry, and many PKD-dependent phosphosites detected in the elastic titin I-band region were significantly decreased in cKO. Analysis of titin site-specific phosphorylation showed unaltered or upregulated phosphorylation in cKO compared to matched WT hearts. Fpassive was elevated in cKO compared to WT cardiomyocytes and PKD administration lowered Fpassive of WT and cKO cardiomyocytes. Cardiomyocytes from hypertrophic cardiomyopathy (HCM) patients showed higher Fpassive compared to control hearts and significantly lower Fpassive after PKD treatment. In addition, we found higher phosphorylation at CaMKII-dependent titin sites in HCM compared to control hearts. Expression and phosphorylation of HSP27, a substrate of PKD, were elevated in HCM hearts, which was associated with increased PKD expression and phosphorylation. The relocalization of HSP27 in HCM away from the sarcomeric Z-disk and I-band suggested that HSP27 failed to exert its protective action on titin extensibility. This protection could, however, be restored by administration of HSP27, which significantly reduced Fpassive in HCM cardiomyocytes. These findings establish a previously unknown role for PKDin regulating diastolic passive properties of healthy and diseased hearts.

15.
Cell Rep ; 29(2): 480-494.e5, 2019 10 08.
Artigo em Inglês | MEDLINE | ID: mdl-31597105

RESUMO

Signaling by the ubiquitin-related SUMO pathway relies on coordinated conjugation and deconjugation events. SUMO-specific deconjugating enzymes counterbalance SUMOylation, but comprehensive insight into their substrate specificity and regulation is missing. By characterizing SENP6, we define an N-terminal multi-SIM domain as a critical determinant in targeting SENP6 to SUMO chains. Proteomic profiling reveals a network of SENP6 functions at the crossroads of chromatin organization and DNA damage response (DDR). SENP6 acts as a SUMO eraser at telomeric and centromeric chromatin domains and determines the SUMOylation status and chromatin association of the cohesin complex. Importantly, SENP6 is part of the hPSO4/PRP19 complex that drives ATR-Chk1 activation. SENP6 deficiency impairs chromatin association of the ATR cofactor ATRIP, thereby compromising the activation of Chk1 signaling in response to aphidicolin-induced replicative stress and sensitizing cells to DNA damage. We propose a general role of SENP6 in orchestrating chromatin dynamics and genome stability networks by balancing chromatin residency of protein complexes.


Assuntos
Cromatina/metabolismo , Cromossomos Humanos/metabolismo , Cisteína Endopeptidases/metabolismo , Genoma Humano , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Motivos de Aminoácidos , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Proteínas de Ciclo Celular/metabolismo , Quinase 1 do Ponto de Checagem/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Cisteína Endopeptidases/química , Instabilidade Genômica , Células HEK293 , Células HeLa , Humanos , Proteínas Nucleares/metabolismo , Ligação Proteica , Sumoilação , Fatores de Transcrição/metabolismo , Coesinas
16.
Cell Rep ; 23(5): 1342-1356, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29719249

RESUMO

Protein aggregates and cytoplasmic vacuolization are major hallmarks of multisystem proteinopathies (MSPs) that lead to muscle weakness. Here, we identify METTL21C as a skeletal muscle-specific lysine methyltransferase. Insertion of a ß-galactosidase cassette into the Mettl21c mouse locus revealed that METTL21C is specifically expressed in MYH7-positive skeletal muscle fibers. Ablation of the Mettl21c gene reduced endurance capacity and led to age-dependent accumulation of autophagic vacuoles in skeletal muscle. Denervation-induced muscle atrophy highlighted further impairments of autophagy-related proteins, including LC3, p62, and cathepsins, in Mettl21c-/- muscles. In addition, we demonstrate that METTL21C interacts with the ATPase p97 (VCP), which is mutated in various human MSP conditions. We reveal that METTL21C trimethylates p97 on the Lys315 residue and found that loss of this modification reduced p97 hexamer formation and ATPase activity in vivo. We conclude that the methyltransferase METTL21C is an important modulator of protein degradation in skeletal muscle under both normal and enhanced protein breakdown conditions.


Assuntos
Autofagia , Metiltransferases/metabolismo , Músculo Esquelético/enzimologia , Proteólise , Proteína com Valosina/metabolismo , Animais , Masculino , Metilação , Metiltransferases/genética , Camundongos , Camundongos Knockout , Proteína com Valosina/genética
17.
Dis Model Mech ; 10(7): 881-896, 2017 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-28546288

RESUMO

Loss of neuronal stimulation enhances protein breakdown and reduces protein synthesis, causing rapid loss of muscle mass. To elucidate the pathophysiological adaptations that occur in atrophying muscles, we used stable isotope labelling and mass spectrometry to quantify protein expression changes accurately during denervation-induced atrophy after sciatic nerve section in the mouse gastrocnemius muscle. Additionally, mice were fed a stable isotope labelling of amino acids in cell culture (SILAC) diet containing 13C6-lysine for 4, 7 or 11 days to calculate relative levels of protein synthesis in denervated and control muscles. Ubiquitin remnant peptides (K-ε-GG) were profiled by immunoaffinity enrichment to identify potential substrates of the ubiquitin-proteasomal pathway. Of the 4279 skeletal muscle proteins quantified, 850 were differentially expressed significantly within 2 weeks after denervation compared with control muscles. Moreover, pulse labelling identified Lys6 incorporation in 4786 proteins, of which 43 had differential Lys6 incorporation between control and denervated muscle. Enrichment of diglycine remnants identified 2100 endogenous ubiquitination sites and revealed a metabolic and myofibrillar protein diglycine signature, including myosin heavy chains, myomesins and titin, during denervation. Comparative analysis of these proteomic data sets with known atrogenes using a random forest approach identified 92 proteins subject to atrogene-like regulation that have not previously been associated directly with denervation-induced atrophy. Comparison of protein synthesis and proteomic data indicated that upregulation of specific proteins in response to denervation is mainly achieved by protein stabilization. This study provides the first integrated analysis of protein expression, synthesis and ubiquitin signatures during muscular atrophy in a living animal.


Assuntos
Denervação Muscular , Músculo Esquelético/inervação , Músculo Esquelético/metabolismo , Atrofia Muscular/metabolismo , Proteoma/metabolismo , Animais , Enzimas Desubiquitinantes/metabolismo , Regulação para Baixo , Glicilglicina/metabolismo , Marcação por Isótopo , Lisina/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Biossíntese de Proteínas , Proteólise , Proteômica , Sarcômeros/metabolismo , Nervo Isquiático/patologia , Fatores de Tempo , Ubiquitina/metabolismo , Ubiquitinação , Regulação para Cima
18.
Elife ; 62017 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-28617241

RESUMO

The turnover of endoplasmic reticulum (ER) ensures the correct biological activity of its distinct domains. In mammalian cells, the ER is degraded via a selective autophagy pathway (ER-phagy), mediated by two specific receptors: FAM134B, responsible for the turnover of ER sheets and SEC62 that regulates ER recovery following stress. Here, we identified reticulon 3 (RTN3) as a specific receptor for the degradation of ER tubules. Oligomerization of the long isoform of RTN3 is sufficient to trigger fragmentation of ER tubules. The long N-terminal region of RTN3 contains several newly identified LC3-interacting regions (LIR). Binding to LC3s/GABARAPs is essential for the fragmentation of ER tubules and their delivery to lysosomes. RTN3-mediated ER-phagy requires conventional autophagy components, but is independent of FAM134B. None of the other reticulon family members have the ability to induce fragmentation of ER tubules during starvation. Therefore, we assign a unique function to RTN3 during autophagy.


Assuntos
Autofagia , Proteínas de Transporte/metabolismo , Retículo Endoplasmático/metabolismo , Proteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Linhagem Celular , Humanos , Multimerização Proteica
19.
Cell Rep ; 16(11): 3075-3086, 2016 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-27626674

RESUMO

Post-translational modification of proteins with ubiquitin-like SUMO modifiers is a tightly regulated and highly dynamic process. The SENP family of SUMO-specific isopeptidases comprises six cysteine proteases. They are instrumental in counterbalancing SUMO conjugation, but their regulation is not well understood. We demonstrate that in hypoxic cell extracts, the catalytic activity of SENP family members, in particular SENP1 and SENP3, is inhibited in a rapid and fully reversible process. Comparative mass spectrometry from normoxic and hypoxic cells defines a subset of hypoxia-induced SUMO1 targets, including SUMO ligases RanBP2 and PIAS2, glucose transporter 1, and transcriptional regulators. Among the most strongly induced targets, we identified the transcriptional co-repressor BHLHE40, which controls hypoxic gene expression programs. We provide evidence that SUMOylation of BHLHE40 is reversed by SENP1 and contributes to transcriptional repression of the metabolic master regulator gene PGC-1α. We propose a pathway that connects oxygen-controlled SENP activity to hypoxic reprogramming of metabolism.


Assuntos
Carbono-Nitrogênio Liases/metabolismo , Transdução de Sinais , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Biocatálise , Hipóxia Celular , Proteínas Correpressoras/metabolismo , Cisteína Endopeptidases/metabolismo , Ativação Enzimática , Células HeLa , Humanos , Especificidade por Substrato , Sumoilação
20.
Mol Biosyst ; 11(3): 908-22, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25592279

RESUMO

Insulin resistance is often associated with excessive caloric intake and metabolic syndrome (MS) favours the development of Diabetes Mellitus Type II (T2DM). T2DM is a chronic disease with severe long-term consequences, such as dyslipidemia, retinopathy, kidney failure, and cardiovascular diseases. Although studied extensively, several aspects of T2DM remain poorly understood. Liver is the leading organ in the maintenance of metabolic fitness serving as the first relay station for processing dietary information in a direct response to nutritional input and changes in insulin and other endocrine signals. Evidence from several murine models suggests a unique function of the liver in the development of MS and T2DM. Here, we utilised Db/Db mice to understand the impact of T2DM on the proteome of liver cells. Global analysis of the liver proteome using a SILAC approach identified 407 significantly regulated proteins under diabetic conditions out of 8500 identified liver proteins. Furthermore, we mapped 1604 different acetylation sites in liver proteins. After normalization of the protein level, we identified 34 regulated acetyl lysine residues on 21 individual proteins, which were significantly altered in Db/Db compared to wild-type livers. We reason that the dataset provides a versatile resource for functional studies aiming to understand consequences of changes in protein abundances and acetylation in livers of diabetic animals.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Redes e Vias Metabólicas , Proteoma , Proteômica , Acetilação , Adaptação Fisiológica , Aminoácidos/metabolismo , Animais , Transporte Biológico , Metabolismo dos Carboidratos , Análise por Conglomerados , Modelos Animais de Doenças , Ácidos Graxos/metabolismo , Fígado/metabolismo , Masculino , Camundongos , Microssomos/metabolismo , Obesidade/metabolismo , Oxirredução , Proteômica/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA