Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
1.
Anal Chem ; 96(19): 7373-7379, 2024 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-38696819

RESUMO

Cross-linking mass spectrometry (XL-MS) has evolved into a pivotal technique for probing protein interactions. This study describes the implementation of Parallel Accumulation-Serial Fragmentation (PASEF) on timsTOF instruments, enhancing the detection and analysis of protein interactions by XL-MS. Addressing the challenges in XL-MS, such as the interpretation of complex spectra, low abundant cross-linked peptides, and a data acquisition bias, our current study integrates a peptide-centric approach for the analysis of XL-MS data and presents the foundation for integrating data-independent acquisition (DIA) in XL-MS with a vendor-neutral and open-source platform. A novel workflow is described for processing data-dependent acquisition (DDA) of PASEF-derived information. For this, software by Bruker Daltonics is used, enabling the conversion of these data into a format that is compatible with MeroX and Skyline software tools. Our approach significantly improves the identification of cross-linked products from complex mixtures, allowing the XL-MS community to overcome current analytical limitations.


Assuntos
Reagentes de Ligações Cruzadas , Espectrometria de Massas , Software , Fluxo de Trabalho , Reagentes de Ligações Cruzadas/química , Peptídeos/química , Peptídeos/análise , Humanos
2.
Nucleic Acids Res ; 52(7): 3971-3988, 2024 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-38300787

RESUMO

The RAVER1 protein serves as a co-factor in guiding the polypyrimidine tract-binding protein (PTBP)-dependent control of alternative splicing (AS). Whether RAVER1 solely acts in concert with PTBPs and how it affects cancer cell fate remained elusive. Here, we provide the first comprehensive investigation of RAVER1-controlled AS in cancer cell models. This reveals a pro-oncogenic role of RAVER1 in modulating tumor growth and epithelial-mesenchymal-transition (EMT). Splicing analyses and protein-association studies indicate that RAVER1 guides AS in association with other splicing regulators, including PTBPs and SRSFs. In cancer cells, one major function of RAVER1 is the stimulation of proliferation and restriction of apoptosis. This involves the modulation of AS events within the miR/RISC pathway. Disturbance of RAVER1 impairs miR/RISC activity resulting in severely deregulated gene expression, which promotes lethal TGFB-driven EMT. Among others, RAVER1-modulated splicing events affect the insertion of protein interaction modules in factors guiding miR/RISC-dependent gene silencing. Most prominently, in all three human TNRC6 proteins, RAVER1 controls AS of GW-enriched motifs, which are essential for AGO2-binding and the formation of active miR/RISC complexes. We propose, that RAVER1 is a key modulator of AS events in the miR/RISC pathway ensuring proper abundance and composition of miR/RISC effectors. This ensures balanced expression of TGFB signaling effectors and limits TGFB induced lethal EMT.


Assuntos
Processamento Alternativo , Transição Epitelial-Mesenquimal , MicroRNAs , Transição Epitelial-Mesenquimal/genética , Humanos , MicroRNAs/metabolismo , MicroRNAs/genética , Linhagem Celular Tumoral , Proteína de Ligação a Regiões Ricas em Polipirimidinas/metabolismo , Proteína de Ligação a Regiões Ricas em Polipirimidinas/genética , Proteínas de Ligação a RNA/metabolismo , Proteínas de Ligação a RNA/genética , Fatores de Processamento de Serina-Arginina/metabolismo , Fatores de Processamento de Serina-Arginina/genética , Regulação Neoplásica da Expressão Gênica , Proliferação de Células/genética , Apoptose/genética , Fator de Crescimento Transformador beta/metabolismo , Animais
3.
Sci Rep ; 14(1): 3026, 2024 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-38321125

RESUMO

[NiFe]-hydrogenases have a bimetallic NiFe(CN)2CO cofactor in their large, catalytic subunit. The 136 Da Fe(CN)2CO group of this cofactor is preassembled on a distinct HypC-HypD scaffold complex, but the intracellular source of the iron ion is unresolved. Native mass spectrometric analysis of HypCD complexes defined the [4Fe-4S] cluster associated with HypD and identified + 26 to 28 Da and + 136 Da modifications specifically associated with HypC. A HypCC2A variant without the essential conserved N-terminal cysteine residue dissociated from its complex with native HypD lacked all modifications. Native HypC dissociated from HypCD complexes isolated from Escherichia coli strains deleted for the iscS or iscU genes, encoding core components of the Isc iron-sulfur cluster biogenesis machinery, specifically lacked the + 136 Da modification, but this was retained on HypC from suf mutants. The presence or absence of the + 136 Da modification on the HypCD complex correlated with the hydrogenase enzyme activity profiles of the respective mutant strains. Notably, the [4Fe-4S] cluster on HypD was identified in all HypCD complexes analyzed. These results suggest that the iron of the Fe(CN)2CO group on HypCD derives from the Isc machinery, while either the Isc or the Suf machinery can deliver the [4Fe-4S] cluster to HypD.


Assuntos
Proteínas de Escherichia coli , Hidrogenase , Proteínas Ferro-Enxofre , Escherichia coli/genética , Ferro/metabolismo , Proteínas de Escherichia coli/metabolismo , Hidrogenase/metabolismo , Domínio Catalítico , Cisteína/química
4.
Nat Cancer ; 4(11): 1592-1609, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37904046

RESUMO

Safely expanding indications for cellular therapies has been challenging given a lack of highly cancer-specific surface markers. Here we explore the hypothesis that tumor cells express cancer-specific surface protein conformations that are invisible to standard target discovery pipelines evaluating gene or protein expression, and these conformations can be identified and immunotherapeutically targeted. We term this strategy integrating cross-linking mass spectrometry with glycoprotein surface capture 'structural surfaceomics'. As a proof of principle, we apply this technology to acute myeloid leukemia (AML), a hematologic malignancy with dismal outcomes and no known optimal immunotherapy target. We identify the activated conformation of integrin ß2 as a structurally defined, widely expressed AML-specific target. We develop and characterize recombinant antibodies to this protein conformation and show that chimeric antigen receptor T cells eliminate AML cells and patient-derived xenografts without notable toxicity toward normal hematopoietic cells. Our findings validate an AML conformation-specific target antigen and demonstrate a tool kit for applying these strategies more broadly.


Assuntos
Leucemia Mieloide Aguda , Receptores de Antígenos Quiméricos , Humanos , Receptores de Antígenos Quiméricos/genética , Receptores de Antígenos Quiméricos/metabolismo , Linfócitos T , Integrinas/metabolismo , Imunoterapia Adotiva/métodos , Leucemia Mieloide Aguda/terapia , Leucemia Mieloide Aguda/genética
5.
Sci Rep ; 13(1): 8497, 2023 05 25.
Artigo em Inglês | MEDLINE | ID: mdl-37231156

RESUMO

The tetrameric tumor suppressor p53 represents a great challenge for 3D-structural analysis due to its high degree of intrinsic disorder (ca. 40%). We aim to shed light on the structural and functional roles of p53's C-terminal region in full-length, wild-type human p53 tetramer and their importance for DNA binding. For this, we employed complementary techniques of structural mass spectrometry (MS) in an integrated approach with computational modeling. Our results show no major conformational differences in p53 between DNA-bound and DNA-free states, but reveal a substantial compaction of p53's C-terminal region. This supports the proposed mechanism of unspecific DNA binding to the C-terminal region of p53 prior to transcription initiation by specific DNA binding to the core domain of p53. The synergies between complementary structural MS techniques and computational modeling as pursued in our integrative approach is envisioned to serve as general strategy for studying intrinsically disordered proteins (IDPs) and intrinsically disordered region (IDRs).


Assuntos
Proteínas Intrinsicamente Desordenadas , Proteína Supressora de Tumor p53 , Humanos , Proteína Supressora de Tumor p53/metabolismo , Simulação por Computador , Proteínas Intrinsicamente Desordenadas/química , DNA/metabolismo , Espectrometria de Massas , Ligação Proteica
6.
Blood ; 141(10): 1105-1118, 2023 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-36493345

RESUMO

Gain of chromosome 21 (Hsa21) is among the most frequent aneuploidies in leukemia. However, it remains unclear how partial or complete amplifications of Hsa21 promote leukemogenesis and why children with Down syndrome (DS) (ie, trisomy 21) are particularly at risk of leukemia development. Here, we propose that RUNX1 isoform disequilibrium with RUNX1A bias is key to DS-associated myeloid leukemia (ML-DS). Starting with Hsa21-focused CRISPR-CRISPR-associated protein 9 screens, we uncovered a strong and specific RUNX1 dependency in ML-DS cells. Expression of the RUNX1A isoform is elevated in patients with ML-DS, and mechanistic studies using murine ML-DS models and patient-derived xenografts revealed that excess RUNX1A synergizes with the pathognomonic Gata1s mutation during leukemogenesis by displacing RUNX1C from its endogenous binding sites and inducing oncogenic programs in complex with the MYC cofactor MAX. These effects were reversed by restoring the RUNX1A:RUNX1C equilibrium in patient-derived xenografts in vitro and in vivo. Moreover, pharmacological interference with MYC:MAX dimerization using MYCi361 exerted strong antileukemic effects. Thus, our study highlights the importance of alternative splicing in leukemogenesis, even on a background of aneuploidy, and paves the way for the development of specific and targeted therapies for ML-DS, as well as for other leukemias with Hsa21 aneuploidy or RUNX1 isoform disequilibrium.


Assuntos
Subunidade alfa 2 de Fator de Ligação ao Core , Síndrome de Down , Leucemia Mieloide , Animais , Criança , Humanos , Camundongos , Aneuploidia , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Síndrome de Down/complicações , Síndrome de Down/genética , Leucemia Mieloide/genética , Isoformas de Proteínas/genética , Trissomia/genética
7.
Chembiochem ; 23(11): e202100665, 2022 06 03.
Artigo em Inglês | MEDLINE | ID: mdl-35333001

RESUMO

The tumor suppressor protein p53 is a transcription factor that is referred to as the "guardian of the genome" and plays an important role in cancer development. p53 is active as a homotetramer; the S100ß homodimer binds to the intrinsically disordered C-terminus of p53 affecting its transcriptional activity. The p53/S100ß complex is regarded as highly promising therapeutic target in cancer. It has been suggested that S100ß exerts its oncogenic effects by altering the p53 oligomeric state. Our aim was to study the structures and oligomerization behavior of different p53/S100ß complexes by ESI-MS, XL-MS, and SPR. Wild-type p53 and single amino acid variants, representing different oligomeric states of p53 were individually investigated regarding their binding behavior towards S100ß. The stoichiometry of the different p53/S100ß complexes were determined by ESI-MS showing that tetrameric, dimeric, and monomeric p53 variants all bind to an S100ß dimer. In addition, XL-MS revealed the topologies of the p53/S100ß complexes to be independent of p53's oligomeric state. With SPR, the thermodynamic parameters were determined for S100ß binding to tetrameric, dimeric, or monomeric p53 variants. Our data prove that the S100ß homodimer binds to different oligomeric states of p53 with similar binding affinities. This emphasizes the need for alternative explanations to describe the molecular mechanisms underlying p53/S100ß interaction.


Assuntos
Neoplasias , Proteína Supressora de Tumor p53 , Humanos , Ligação Proteica , Subunidade beta da Proteína Ligante de Cálcio S100 , Fatores de Transcrição/metabolismo , Proteína Supressora de Tumor p53/química
8.
Blood ; 139(5): 651-665, 2022 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-34570885

RESUMO

Given the plasticity of hematopoietic stem and progenitor cells, multiple routes of differentiation must be blocked in the the pathogenesis of acute myeloid leukemia, the molecular basis of which is incompletely understood. We report that posttranscriptional repression of the transcription factor ARID3A by miR-125b is a key event in the pathogenesis of acute megakaryoblastic leukemia (AMKL). AMKL is frequently associated with trisomy 21 and GATA1 mutations (GATA1s), and children with Down syndrome are at a high risk of developing the disease. The results of our study showed that chromosome 21-encoded miR-125b synergizes with Gata1s to drive leukemogenesis in this context. Leveraging forward and reverse genetics, we uncovered Arid3a as the main miR-125b target behind this synergy. We demonstrated that, during normal hematopoiesis, this transcription factor promotes megakaryocytic differentiation in concert with GATA1 and mediates TGFß-induced apoptosis and cell cycle arrest in complex with SMAD2/3. Although Gata1s mutations perturb erythroid differentiation and induce hyperproliferation of megakaryocytic progenitors, intact ARID3A expression assures their megakaryocytic differentiation and growth restriction. Upon knockdown, these tumor suppressive functions are revoked, causing a blockade of dual megakaryocytic/erythroid differentiation and subsequently of AMKL. Inversely, restoring ARID3A expression relieves the arrest of megakaryocytic differentiation in AMKL patient-derived xenografts. This work illustrates how mutations in lineage-determining transcription factors and perturbation of posttranscriptional gene regulation can interact to block multiple routes of hematopoietic differentiation and cause leukemia. In AMKL, surmounting this differentiation blockade through restoration of the tumor suppressor ARID3A represents a promising strategy for treating this lethal pediatric disease.


Assuntos
Proteínas de Ligação a DNA/genética , Leucemia Megacarioblástica Aguda/genética , Fatores de Transcrição/genética , Animais , Criança , Fator de Transcrição GATA1/genética , Regulação Leucêmica da Expressão Gênica , Genes Supressores de Tumor , Humanos , Leucemia Megacarioblástica Aguda/patologia , Camundongos Endogâmicos C57BL , MicroRNAs/genética , Mutação
9.
Sci Rep ; 11(1): 24362, 2021 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-34934150

RESUMO

[NiFe]-hydrogenases activate dihydrogen. Like all [NiFe]-hydrogenases, hydrogenase 2 of Escherichia coli has a bimetallic NiFe(CN)2CO cofactor in its catalytic subunit. Biosynthesis of the Fe(CN)2CO group of the [NiFe]-cofactor occurs on a distinct scaffold complex comprising the HybG and HypD accessory proteins. HybG is a member of the HypC-family of chaperones that confers specificity towards immature hydrogenase catalytic subunits during transfer of the Fe(CN)2CO group. Using native mass spectrometry of an anaerobically isolated HybG-HypD complex we show that HybG carries the Fe(CN)2CO group. Our results also reveal that only HybG, but not HypD, interacts with the apo-form of the catalytic subunit. Finally, HybG was shown to have two distinct, and apparently CO2-related, covalent modifications that depended on the presence of the N-terminal cysteine residue on the protein, possibly representing intermediates during Fe(CN)2CO group biosynthesis. Together, these findings suggest that the HybG chaperone is involved in both biosynthesis and delivery of the Fe(CN)2CO group to its target protein. HybG is thus suggested to shuttle between the assembly complex and the apo-catalytic subunit. This study provides new insights into our understanding of how organometallic cofactor components are assembled on a scaffold complex and transferred to their client proteins.


Assuntos
Monóxido de Carbono/metabolismo , Cianetos/metabolismo , Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Compostos Ferrosos/metabolismo , Hidrogenase/metabolismo , Chaperonas Moleculares/metabolismo , Monóxido de Carbono/química , Domínio Catalítico , Cianetos/química , Escherichia coli/genética , Escherichia coli/crescimento & desenvolvimento , Proteínas de Escherichia coli/genética , Compostos Ferrosos/química , Hidrogenase/química , Hidrogenase/genética , Espectrometria de Massas , Chaperonas Moleculares/genética , Proteínas/genética , Proteínas/metabolismo
10.
PLoS Biol ; 19(9): e3001321, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34491979

RESUMO

Incorporation of noncanonical amino acids (ncAAs) can endow proteins with novel functionalities, such as crosslinking or fluorescence. In ion channels, the function of these variants can be studied with great precision using standard electrophysiology, but this approach is typically labor intensive and low throughput. Here, we establish a high-throughput protocol to conduct functional and pharmacological investigations of ncAA-containing human acid-sensing ion channel 1a (hASIC1a) variants in transiently transfected mammalian cells. We introduce 3 different photocrosslinking ncAAs into 103 positions and assess the function of the resulting 309 variants with automated patch clamp (APC). We demonstrate that the approach is efficient and versatile, as it is amenable to assessing even complex pharmacological modulation by peptides. The data show that the acidic pocket is a major determinant for current decay, and live-cell crosslinking provides insight into the hASIC1a-psalmotoxin 1 (PcTx1) interaction. Further, we provide evidence that the protocol can be applied to other ion channels, such as P2X2 and GluA2 receptors. We therefore anticipate the approach to enable future APC-based studies of ncAA-containing ion channels in mammalian cells.


Assuntos
Canais Iônicos Sensíveis a Ácido/química , Canais Iônicos Sensíveis a Ácido/farmacologia , Aminoácidos/química , Canais Iônicos Sensíveis a Ácido/genética , Células HEK293 , Humanos , Peptídeos/química , Venenos de Aranha/química , Transfecção
11.
Chemistry ; 27(59): 14586-14593, 2021 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-34406694

RESUMO

Although incorporation of photo-activatable lipids into membranes potentially opens up novel avenues for investigating interactions with proteins, the question of whether diazirine-modified lipids are suitable for such studies, remains under debate. Focusing on the potential for studying lipid/peptide interactions by cross-linking mass spectrometry (XL-MS), we developed a diazirine-modified lipid (DiazPC), and examined its behaviour in membranes incorporating the model α-helical peptide LAVA20. We observed an unexpected backfolding of the diazirine-containing stearoyl chain of the lipid. This surprising behaviour challenges the potential application of DiazPC for future XL-MS studies of peptide and protein/lipid interactions. The observations made for DiazPC most likely represent a general phenomenon for any type of membrane lipids with a polar moiety incorporated into the alkyl chain. Our finding is therefore of importance for future protein/lipid interaction studies relying on modified lipid probes.


Assuntos
Diazometano , Lipídeos de Membrana , Reagentes de Ligações Cruzadas , Espectrometria de Massas , Peptídeos
12.
Biology (Basel) ; 10(5)2021 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-34067172

RESUMO

MEX3A belongs to the MEX3 (Muscle EXcess) protein family consisting of four members (MEX3A-D) in humans. Characteristic for MEX3 proteins is their domain structure with 2 HNRNPK homology (KH) domains mediating RNA binding and a C-terminal really interesting new gene (RING) domain that harbors E3 ligase function. In agreement with their domain composition, MEX3 proteins were reported to modulate both RNA fate and protein ubiquitination. MEX3 paralogs exhibit an oncofetal expression pattern, they are severely downregulated postnatally, and re-expression is observed in various malignancies. Enforced expression of MEX3 proteins in various cancers correlates with poor prognosis, emphasizing their oncogenic potential. The latter is supported by MEX3A's impact on proliferation, self-renewal as well as migration of tumor cells in vitro and tumor growth in xenograft studies.

13.
Int J Biol Macromol ; 182: 502-511, 2021 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-33848543

RESUMO

High-resolution structural data of complexes between antibodies and membrane receptors still represent a demanding task. In this study, we used complementary sets of experimental data to obtain a structural model of the complex formed by the human epidermal growth factor receptor 2 (HER2) and its specific nanobody A10. First we identified by NMR the residues that bind or rearrange as a consequence of the complex formation. In parallel, the complex was cross-linked, digested and the resulting peptides were characterized by mass-spectrometry to define maximal distance restraints between HER2 and A10 amino acids in their complex. These independent datasets guided a docking process, refined by molecular dynamics simulations, to develop a model of the complex and estimate per-residue free-energy contributions. Such a model explains the experimental data and identifies a second, non-canonical paratope, located in the region opposite to the conventional nanobody paratope, formed by the hypervariable loop regions LH1 and LH3. Both paratopes contributed substantially to the overall affinity by binding to independent HER2 epitopes. Nanobody mutants with substitution of key interaction residues, as indicated by the model, possess significantly lower affinity for HER2. This is the first described case of a "natural" biparatopic nanobody, directly selected by in-vitro panning.


Assuntos
Sítios de Ligação de Anticorpos , Receptor ErbB-2/química , Anticorpos de Cadeia Única/química , Humanos , Simulação de Acoplamento Molecular , Mutação , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/imunologia , Ligação Proteica , Receptor ErbB-2/imunologia , Anticorpos de Cadeia Única/genética , Anticorpos de Cadeia Única/imunologia
14.
J Mol Biol ; 433(10): 166947, 2021 05 14.
Artigo em Inglês | MEDLINE | ID: mdl-33744315

RESUMO

The rod-outer-segment guanylyl cyclase 1 (ROS-GC1) is a key transmembrane protein for retinal phototransduction. Mutations of ROS-GC1 correlate with different retinal diseases that often lead to blindness. No structural data are available for ROS-GC1 so far. We performed a 3D-structural analysis of native ROS-GC1 from bovine retina by cross-linking/mass spectrometry (XL-MS) and computational modeling. Absolute quantification and activity measurements of native ROS-GC1 were performed by MS-based assays directly in bovine retina samples. Our data present the first 3D-structural analysis of active, full-length ROS-GC1 derived from bovine retina. We propose a novel domain organization for the intracellular domain ROS-GC1. Our XL-MS data of native ROS-GC1 from rod-outer-segment preparations of bovine retina agree with a dimeric architecture. Our integrated approach can serve as a blueprint for conducting 3D-structural studies of membrane proteins in their native environment.


Assuntos
GMP Cíclico/química , Guanilato Ciclase/química , Peptídeos/metabolismo , Receptores de Superfície Celular/química , Segmento Externo da Célula Bastonete/química , Motivos de Aminoácidos , Animais , Sítios de Ligação , Bovinos , Clonagem Molecular , Reagentes de Ligações Cruzadas/química , GMP Cíclico/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Expressão Gênica , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Guanilato Ciclase/genética , Guanilato Ciclase/metabolismo , Células HEK293 , Humanos , Espectrometria de Massas/métodos , Modelos Moleculares , Peptídeos/síntese química , Ligação Proteica , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Domínios e Motivos de Interação entre Proteínas , Multimerização Proteica , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Segmento Externo da Célula Bastonete/metabolismo , Succinimidas/química
15.
FEBS Open Bio ; 11(3): 652-669, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33462996

RESUMO

In order to reduce nitrate in vivo, the spore-specific respiratory nitrate reductase, Nar1, of Streptomyces coelicolor relies on an active cytochrome bcc-aa3 oxidase supercomplex (bcc-aa3 supercomplex). This suggests that membrane-associated Nar1, comprising NarG1, NarH1, and NarI1 subunits, might not act as a classical menaquinol oxidase but could either receive electrons from the bcc-aa3 supercomplex, or require the supercomplex to stabilize the reductase in the membrane to allow it to function. To address the biochemical basis for this dependence on the bcc-aa3 supercomplex, we purified two different Strep-tagged variants of Nar1 and enriched the native enzyme complex from spore extracts using different chromatographic and electrophoretic procedures. Polypeptides associated with the isolated Nar1 complexes were identified using mass spectrometry and included components of the bcc-aa3 supercomplex, along with an alternative, spore-specific cytochrome b component, QcrB3. Surprisingly, we also co-enriched the Nar3 enzyme with Nar1 from the wild-type strain of S. coelicolor. Two differentially migrating active Nar1 complexes could be identified after clear native polyacrylamide gel electrophoresis; these had masses of approximately 450 and 250 kDa. The distribution of active Nar1 in these complexes was influenced by the presence of cytochrome bd oxidase and by QcrB3; the presence of the latter shifted Nar1 into the larger complex. Together, these data suggest that several respiratory complexes can associate in the spore membrane, including Nar1, Nar3, and the bcc-aa3 supercomplex. Moreover, these findings provide initial support for the hypothesis that Nar1 and the bcc-aa3 supercomplex physically associate.


Assuntos
Complexo IV da Cadeia de Transporte de Elétrons/isolamento & purificação , Nitrato Redutase/isolamento & purificação , Oxirredutases/isolamento & purificação , Streptomyces coelicolor/metabolismo , Proteínas de Bactérias/isolamento & purificação , Espectrometria de Massas , Complexos Multienzimáticos/isolamento & purificação , Esporos Bacterianos/metabolismo
16.
RNA Biol ; 18(3): 391-403, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-32876513

RESUMO

Epithelial-to-mesenchymal transition (EMT) is a hallmark of aggressive, mesenchymal-like high-grade serous ovarian carcinoma (HGSOC). The SRC kinase is a key driver of cancer-associated EMT promoting adherens junction (AJ) disassembly by phosphorylation-driven internalization and degradation of AJ proteins. Here, we show that the IGF2 mRNA-binding protein 1 (IGF2BP1) is up-regulated in mesenchymal-like HGSOC and promotes SRC activation by a previously unknown protein-ligand-induced, but RNA-independent mechanism. IGF2BP1-driven invasive growth of ovarian cancer cells essentially relies on the SRC-dependent disassembly of AJs. Concomitantly, IGF2BP1 enhances ERK2 expression in an RNA-binding dependent manner. Together this reveals a post-transcriptional mechanism of interconnected stimulation of SRC/ERK signalling in ovarian cancer cells. The IGF2BP1-SRC/ERK2 axis is targetable by the SRC-inhibitor saracatinib and MEK-inhibitor selumetinib. However, due to IGF2BP1-directed stimulation, only combinatorial treatment effectively overcomes the IGF2BP1-promoted invasive growth in 3D culture conditions as well as intraperitoneal mouse models. In conclusion, we reveal an unexpected role of IGF2BP1 in enhancing SRC/MAPK-driven invasive growth of ovarian cancer cells. This provides a rationale for the therapeutic benefit of combinatorial SRC/MEK inhibition in mesenchymal-like HGSOC.


Assuntos
Regulação Neoplásica da Expressão Gênica , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Neoplasias Ovarianas/etiologia , Neoplasias Ovarianas/metabolismo , Proteínas de Ligação a RNA/genética , Transdução de Sinais , Quinases da Família src/metabolismo , Junções Aderentes/genética , Junções Aderentes/metabolismo , Animais , Biomarcadores Tumorais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Transição Epitelial-Mesenquimal/genética , Feminino , Humanos , Camundongos , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/patologia , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Inibidores de Proteínas Quinases/farmacologia , Proteínas de Ligação a RNA/metabolismo , Transdução de Sinais/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto , Domínios de Homologia de src , Quinases da Família src/antagonistas & inibidores
17.
J Mass Spectrom ; 55(1): e4449, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31820512

RESUMO

Chemical cross-linking combined with mass spectrometry (XL-MS) and computational modeling has evolved as an alternative method to derive protein 3D structures and to map protein interaction networks. Special focus has been laid recently on the development and application of cross-linkers that are cleavable by collisional activation as they yield distinct signatures in tandem mass spectra. Building on our experiences with cross-linkers containing an MS-labile urea group, we now present the biuret-based, CID-MS/MS-cleavable cross-linker imidodicarbonyl diimidazole (IDDI) and demonstrate its applicability for protein cross-linking studies based on the four model peptides angiotensin II, MRFA, substance P, and thymopentin.


Assuntos
Biureto/análogos & derivados , Biureto/química , Reagentes de Ligações Cruzadas/química , Peptídeos/química , Angiotensina II/química , Cromatografia Líquida de Alta Pressão , Imidazóis/química , Estudo de Prova de Conceito , Conformação Proteica , Substância P/química , Espectrometria de Massas em Tandem , Timopentina/química
18.
Sci Rep ; 9(1): 15058, 2019 10 21.
Artigo em Inglês | MEDLINE | ID: mdl-31636333

RESUMO

Calcium- and Integrin-Binding protein 2 (CIB2) is a small and ubiquitously expressed protein with largely unknown biological function but ascertained role in hearing physiology and disease. Recent studies found that CIB2 binds Ca2+ with moderate affinity and dimerizes under conditions mimicking the physiological ones. Here we provided new lines of evidence on CIB2 oligomeric state and the mechanism of interaction with the α7B integrin target. Based on a combination of native mass spectrometry, chemical cross-linking/mass spectrometry, analytical gel filtration, dynamic light scattering and molecular dynamics simulations we conclude that CIB2 is monomeric under all tested conditions and presents uncommon hydrodynamic properties, most likely due to the high content of hydrophobic solvent accessible surface. Surface plasmon resonance shows that the interaction with α7B occurs with relatively low affinity and is limited to the cytosolic region proximal to the membrane, being kinetically favored in the presence of physiological Mg2+ and in the absence of Ca2+. Although CIB2 binds to an α7B peptide in a 1:1 stoichiometry, the formation of the complex might induce binding of another CIB2 molecule.


Assuntos
Proteínas de Ligação ao Cálcio/metabolismo , Hidrodinâmica , Multimerização Proteica , Animais , Cálcio/metabolismo , Proteínas de Ligação ao Cálcio/química , Calmodulina/metabolismo , Bovinos , Membrana Celular/metabolismo , Difusão Dinâmica da Luz , Humanos , Integrinas , Magnésio/metabolismo , Modelos Moleculares , Peso Molecular , Peptídeos/metabolismo , Ligação Proteica , Estabilidade Proteica , Estrutura Terciária de Proteína , Proteólise , Homologia Estrutural de Proteína
19.
J Am Soc Mass Spectrom ; 30(1): 139-148, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29679287

RESUMO

Cleavable cross-linkers are gaining increasing importance for chemical cross-linking/mass spectrometry (MS) as they permit a reliable and automated data analysis in structural studies of proteins and protein assemblies. Here, we introduce 1,3-diallylurea (DAU) as the first CID-MS/MS-cleavable, photo-thiol-reactive cross-linker. DAU is a commercially available, inexpensive reagent that efficiently undergoes an anti-Markovnikov hydrothiolation with cysteine residues in the presence of a radical initiator upon UV-A irradiation. Radical cysteine cross-linking proceeds via an orthogonal "click reaction" and yields stable alkyl sulfide products. DAU reacts at physiological pH and cross-linking reactions with peptides, and proteins can be performed at temperatures as low as 4 °C. The central urea bond is efficiently cleaved upon collisional activation during tandem MS experiments generating characteristic product ions. This improves the reliability of automated cross-link identification. Different radical initiators have been screened for the cross-linking reaction of DAU using the thiol-containing compounds cysteine and glutathione. Our concept has also been exemplified for the biologically relevant proteins bMunc13-2 and retinal guanylyl cyclase-activating protein-2. Graphical abstract ᅟ.


Assuntos
Compostos Alílicos/farmacologia , Reagentes de Ligações Cruzadas/química , Proteínas/química , Compostos de Sulfidrila/química , Ureia/análogos & derivados , Ureia/farmacologia , Cisteína/química , Glutationa/química , Proteínas Ativadoras de Guanilato Ciclase/química , Concentração de Íons de Hidrogênio , Proteínas do Tecido Nervoso/química , Reprodutibilidade dos Testes , Espectrometria de Massas por Ionização por Electrospray , Espectrometria de Massas em Tandem
20.
Mass Spectrom Rev ; 38(2): 187-201, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-29660147

RESUMO

Free radical-initiated peptide sequencing (FRIPS) has recently been introduced as an analytical strategy to create peptide radical ions in a predictable and effective way by collisional activation of specifically modified peptides ions. FRIPS is based on the unimolecular dissociation of open-shell ions and yields fragments that resemble those obtained by electron capture dissociation (ECD) or electron transfer dissociation (ETD). In this review article, we describe the fundamentals of FRIPS and highlight its fruitful combination with chemical cross-linking/mass spectrometry (MS) as a highly promising option to derive complementary structural information of peptides and proteins. FRIPS does not only yield exhaustive sequence information of cross-linked peptides, but also defines the exact cross-linking sites of the connected peptides. The development of more advanced FRIPS cross-linkers that extend the FRIPS-based cross-linking/MS approach to the study of large protein assemblies and protein interaction networks can be eagerly anticipated.


Assuntos
Reagentes de Ligações Cruzadas/química , Radicais Livres/química , Peptídeos/química , Proteínas/química , Análise de Sequência de Proteína/métodos , Espectrometria de Massas em Tandem/métodos , Animais , Humanos , Conformação Proteica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA