Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Biol Chem ; 293(40): 15538-15555, 2018 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-30139746

RESUMO

Innate immunity in animals including humans encompasses the complement system, which is considered an important host defense mechanism against Aspergillus fumigatus, one of the most ubiquitous opportunistic human fungal pathogens. Previously, it has been shown that the alkaline protease Alp1p secreted from A. fumigatus mycelia degrades the complement components C3, C4, and C5. However, it remains unclear how the fungal spores (i.e. conidia) defend themselves against the activities of the complement system immediately after inhalation into the lung. Here, we show that A. fumigatus conidia contain a metalloprotease Mep1p, which is released upon conidial contact with collagen and inactivates all three complement pathways. In particular, Mep1p efficiently inactivated the major complement components C3, C4, and C5 and their activation products (C3a, C4a, and C5a) as well as the pattern-recognition molecules MBL and ficolin-1, either by directly cleaving them or by cleaving them to a form that is further broken down by other proteases of the complement system. Moreover, incubation of Mep1p with human serum significantly inhibited the complement hemolytic activity and conidial opsonization by C3b and their subsequent phagocytosis by macrophages. Together, these results indicate that Mep1p associated with and released from A. fumigatus conidia likely facilitates early immune evasion by disarming the complement defense in the human host.


Assuntos
Aspergillus fumigatus/imunologia , Complemento C3/genética , Complemento C4/genética , Complemento C5/genética , Aspergilose Pulmonar Invasiva/imunologia , Metaloendopeptidases/imunologia , Animais , Aspergillus fumigatus/crescimento & desenvolvimento , Aspergillus fumigatus/patogenicidade , Colágeno/genética , Colágeno/imunologia , Complemento C3/metabolismo , Complemento C4/metabolismo , Complemento C5/metabolismo , Modelos Animais de Doenças , Proteínas Fúngicas/genética , Proteínas Fúngicas/imunologia , Regulação da Expressão Gênica , Interações Hospedeiro-Patógeno , Humanos , Evasão da Resposta Imune , Imunidade Inata , Aspergilose Pulmonar Invasiva/genética , Aspergilose Pulmonar Invasiva/microbiologia , Aspergilose Pulmonar Invasiva/patologia , Lectinas/genética , Lectinas/imunologia , Pulmão/imunologia , Pulmão/patologia , Macrófagos/imunologia , Macrófagos/microbiologia , Masculino , Serina Proteases Associadas a Proteína de Ligação a Manose/genética , Serina Proteases Associadas a Proteína de Ligação a Manose/imunologia , Metaloendopeptidases/deficiência , Metaloendopeptidases/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Fagocitose , Esporos Fúngicos/crescimento & desenvolvimento , Esporos Fúngicos/imunologia , Esporos Fúngicos/patogenicidade , Ficolinas
2.
Cell Rep ; 22(2): 497-511, 2018 01 09.
Artigo em Inglês | MEDLINE | ID: mdl-29320743

RESUMO

Cellular metabolism is a tightly controlled process in which the cell adapts fluxes through metabolic pathways in response to changes in nutrient supply. Among the transcription factors that regulate gene expression and thereby cause changes in cellular metabolism is the basic leucine-zipper (bZIP) transcription factor CCAAT/enhancer-binding protein alpha (C/EBPα). Protein lysine acetylation is a key post-translational modification (PTM) that integrates cellular metabolic cues with other physiological processes. Here, we show that C/EBPα is acetylated by the lysine acetyl transferase (KAT) p300 and deacetylated by the lysine deacetylase (KDAC) sirtuin1 (SIRT1). SIRT1 is activated in times of energy demand by high levels of nicotinamide adenine dinucleotide (NAD+) and controls mitochondrial biogenesis and function. A hypoacetylated mutant of C/EBPα induces the transcription of mitochondrial genes and results in increased mitochondrial respiration. Our study identifies C/EBPα as a key mediator of SIRT1-controlled adaption of energy homeostasis to changes in nutrient supply.


Assuntos
Proteína p300 Associada a E1A/genética , Mitocôndrias/metabolismo , Sirtuína 1/genética , Acetilação , Animais , Proteína p300 Associada a E1A/metabolismo , Humanos , Sirtuína 1/metabolismo
3.
Angew Chem Int Ed Engl ; 56(26): 7510-7514, 2017 06 19.
Artigo em Inglês | MEDLINE | ID: mdl-28544119

RESUMO

Systemic amyloidosis is caused by the misfolding of a circulating amyloid precursor protein and the deposition of amyloid fibrils in multiple organs. Chemical and biophysical analysis of amyloid fibrils from human AL and murine AA amyloidosis reveal the same fibril morphologies in different tissues or organs of one patient or diseased animal. The observed structural similarities concerned the fibril morphology, the fibril protein primary and secondary structures, the presence of post-translational modifications and, in case of the AL fibrils, the partially folded characteristics of the polypeptide chain within the fibril. Our data imply for both analyzed forms of amyloidosis that the pathways of protein misfolding are systemically conserved; that is, they follow the same rules irrespective of where inside one body fibrils are formed or accumulated.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Amiloidose/metabolismo , Dobramento de Proteína , Tecido Adiposo/metabolismo , Sequência de Aminoácidos , Animais , Eletroforese em Gel de Poliacrilamida , Humanos , Espectrometria de Massas , Camundongos , Microscopia Eletrônica de Transmissão , Miocárdio/metabolismo , Peptídeos/metabolismo , Processamento de Proteína Pós-Traducional , Estrutura Secundária de Proteína , Baço/metabolismo , Difração de Raios X
4.
Methods Mol Biol ; 1510: 313-327, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27761831

RESUMO

Posttranslational acetylation of lysine residues has been discovered as multifaceted regulatory modification for various nuclear, cytoplasmic, and mitochondrial proteins. The implementation of high-resolution and high-throughput mass spectrometry (MS) approaches has led to the identification of a hitherto underappreciated, large number of acetylation sites for a broad spectrum of cellular proteins. In this chapter, we describe a comprehensive protocol for the purification of an in vivo-acetylated, ectopically expressed, FLAG-epitope tagged nonhistone protein through immunoprecipitation (IP). The protocol also covers the sample preparation by SDS-PAGE, proteolytic digestion, and the analysis by LC-ESI MS. The success of this methodology, however, strongly depends on the physico-chemical properties of the respective protein(s) and the quality of selected peptide mass spectra.


Assuntos
Proteínas Cromossômicas não Histona/metabolismo , Inibidores de Histona Desacetilases/farmacologia , Processamento de Proteína Pós-Traducional , Proteínas Recombinantes de Fusão/metabolismo , Espectrometria de Massas por Ionização por Electrospray/métodos , Acetilação , Proteínas Cromossômicas não Histona/genética , Eletroforese em Gel de Poliacrilamida , Células HeLa , Humanos , Imunoprecipitação , Oligopeptídeos/genética , Oligopeptídeos/metabolismo , Proteínas Recombinantes de Fusão/genética , Transfecção
5.
Angew Chem Int Ed Engl ; 55(15): 4822-5, 2016 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-26954430

RESUMO

Polymorphism is a wide-spread feature of amyloid-like fibrils formed in vitro, but it has so far remained unclear whether the fibrils formed within a patient are also affected by this phenomenon. In this study we show that the amyloid fibrils within a diseased individual can vary considerably in their three-dimensional architecture. We demonstrate this heterogeneity with amyloid fibrils deposited within different organs, formed from sequentially non-homologous polypeptide chains and affecting human or animals. Irrespective of amyloid type or source, we found in vivo fibrils to be polymorphic. These data imply that the chemical principles of fibril assembly that lead to such polymorphism are fundamentally conserved in vivo and in vitro.


Assuntos
Amiloide/química , Microscopia Eletrônica de Transmissão , Conformação Proteica
6.
Nanotoxicology ; 10(4): 426-35, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26444998

RESUMO

Identifying nanomaterial-bio-interactions are imperative due to the broad introduction of nanoparticle (NP) applications and their distribution. Here, we demonstrate that silica NPs effect widespread protein aggregation in the soil nematode Caenorhabditis elegans ranging from induction of amyloid in nucleoli of intestinal cells to facilitation of protein aggregation in body wall muscles and axons of neural cells. Proteomic screening revealed that exposure of adult C. elegans with silica NPs promotes segregation of proteins belonging to the gene ontology (GO) group of "protein folding, proteolysis and stress response" to an SDS-resistant aggregome network. Candidate proteins in this group include chaperones, heat shock proteins and subunits of the 26S proteasome which are all decisively involved in protein homeostasis. The pathway of protein homeostasis was validated as a major target of silica NPs by behavioral phenotyping, as inhibitors of amyloid formation rescued NP-induced defects of locomotory patterns and egg laying. The analysis of a reporter worm for serotonergic neural cells revealed that silica NP-induced protein aggregation likewise occurs in axons of HSN neurons, where presynaptic accumulation of serotonin, e.g. disturbed axonal transport reduces the capacity for neurotransmission and egg laying. The results suggest that in C. elegans silica NPs promote a cascade of events including disturbance of protein homeostasis, widespread protein aggregation and inhibition of serotonergic neurotransmission which can be interrupted by compounds preventing amyloid fibrillation.


Assuntos
Amiloide/metabolismo , Caenorhabditis elegans/efeitos dos fármacos , Nanopartículas/toxicidade , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Substâncias Protetoras/farmacologia , Agregação Patológica de Proteínas/prevenção & controle , Dióxido de Silício/toxicidade , Animais , Comportamento Animal/efeitos dos fármacos , Homeostase , Nanopartículas/química , Proteínas/metabolismo , Proteômica , Serotonina/metabolismo , Dióxido de Silício/química
7.
PeerJ ; 3: e754, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25699204

RESUMO

Mercury (Hg) is a bioaccumulating trace metal that globally circulates the atmosphere and waters in its elemental, inorganic and organic chemical forms. While Hg represents a notorious neurotoxicant, the underlying cellular pathways are insufficiently understood. We identify amyloid protein aggregation in the cell nucleus as a novel pathway of Hg-bio-interactions. By mass spectrometry of purified protein aggregates, a subset of spliceosomal components and nucleoskeletal protein lamin B1 were detected as constituent parts of an Hg-induced nuclear aggregome network. The aggregome network was located by confocal imaging of amyloid-specific antibodies and dyes to amyloid cores within splicing-speckles that additionally recruit components of the ubiquitin-proteasome system. Hg significantly enhances global proteasomal activity in the nucleus, suggesting that formation of amyloid speckles plays a role in maintenance of protein homeostasis. RNAi knock down showed that lamin B1 for its part regulates amyloid speckle formation and thus likewise participates in nuclear protein homeostasis. As the Hg-induced cascade of interactions between the nucleoskeleton and protein homeostasis reduces neuronal signalling, amyloid fibrillation in the cell nucleus is introduced as a feature of Hg-neurotoxicity that opens new avenues of future research. Similar to protein aggregation events in the cytoplasm that are controlled by the cytoskeleton, amyloid fibrillation of nuclear proteins may be driven by the nucleoskeleton.

8.
Nucleic Acids Res ; 43(5): 2958-67, 2015 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-25712103

RESUMO

The minichromosome maintenance complex (MCM) represents the replicative DNA helicase both in eukaryotes and archaea. Here, we describe the solution structure of the C-terminal domains of the archaeal MCMs of Sulfolobus solfataricus (Sso) and Methanothermobacter thermautotrophicus (Mth). Those domains consist of a structurally conserved truncated winged helix (WH) domain lacking the two typical 'wings' of canonical WH domains. A less conserved N-terminal extension links this WH module to the MCM AAA+ domain forming the ATPase center. In the Sso MCM this linker contains a short α-helical element. Using Sso MCM mutants, including chimeric constructs containing Mth C-terminal domain elements, we show that the ATPase and helicase activity of the Sso MCM is significantly modulated by the short α-helical linker element and by N-terminal residues of the first α-helix of the truncated WH module. Finally, based on our structural and functional data, we present a docking-derived model of the Sso MCM, which implies an allosteric control of the ATPase center by the C-terminal domain.


Assuntos
Proteínas Arqueais/química , Proteínas Arqueais/metabolismo , Proteínas de Manutenção de Minicromossomo/química , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Adenosina Trifosfatases/química , Adenosina Trifosfatases/genética , Adenosina Trifosfatases/metabolismo , Trifosfato de Adenosina/metabolismo , Sequência de Aminoácidos , Proteínas Arqueais/genética , DNA Helicases/química , DNA Helicases/genética , DNA Helicases/metabolismo , Hidrólise , Espectroscopia de Ressonância Magnética , Methanobacteriaceae/genética , Methanobacteriaceae/metabolismo , Proteínas de Manutenção de Minicromossomo/genética , Proteínas de Manutenção de Minicromossomo/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Mutação , Filogenia , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Homologia de Sequência de Aminoácidos , Sulfolobus solfataricus/genética , Sulfolobus solfataricus/metabolismo
9.
PLoS One ; 8(12): e81926, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24324731

RESUMO

The amyloid precursor protein (APP) and its processing by the α-, ß- and γ-secretases is widely believed to play a central role during the development of Alzheimer´s disease. The three-dimensional structure of the entire protein, its physiologic function and the regulation of its proteolytic processing remain, however, largely unclear to date. To gain a deeper understanding of the structure of APP that underlies all of its functions, we first cloned and recombinantly expressed different constructs in E. coli. Using limited proteolysis followed by mass spectrometry and Edman degradation as well as analytical gel permeation chromatography coupled static light scattering, we experimentally analyzed the structural domain boundaries and determined that the large ectodomain of APP consists of exactly two rigidly folded domains - the E1-domain (Leu18-Ala190) and the E2-domain (Ser295-Asp500). Both, the acidic domain (AcD) connecting E1 and E2 as well as the juxtamembrane region (JMR) connecting E2 to the single transmembrane helix are highly flexible and extended. We identified in-between the E1-domain and the AcD an additional domain of conservation and partial flexibility that we termed extension domain (ED, Glu191-Glu227). Using Bio-layer interferometry, pull-down assays and analytical gel filtration experiments we demonstrated that the E1-domain does not tightly interact with the E2-domain, both in the presence and in the absence of heparin. APP hence forms an extended molecule that is flexibly tethered to the membrane. Its multi-domain architecture enables together with the many known functionalities the concomitant performance of several, independent functions, which might be regulated by cellular, compartment specific pH-changes.


Assuntos
Precursor de Proteína beta-Amiloide/química , Sequência de Aminoácidos , Animais , Heparina/metabolismo , Humanos , Interferometria , Dados de Sequência Molecular , Peso Molecular , Ligação Proteica , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Proteólise , Alinhamento de Sequência
11.
J Mol Biol ; 416(3): 438-52, 2012 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-22245578

RESUMO

The amyloid precursor protein (APP) and its neurotoxic cleavage product Aß are key players in the development of Alzheimer's disease and appear essential for neuronal development and cell homeostasis in mammals. Proteolytic processing of APP is influenced by metal ions, protein ligands and its oligomerization state. However, the structural basis and functional mechanism of APP regulation are hitherto largely unknown. Here we identified a metal-dependent molecular switch located within the E2 domain of APP containing four evolutionary highly conserved histidine residues. Three X-ray structures of the metal-bound molecule were solved at 2.6-2.0 Å resolution. Using protein crystallographic and biochemical methods, we characterized this novel high-affinity binding site within the E2 domain that binds competitively to copper and zinc at physiological concentrations. Metal-specific coordination spheres induce large conformational changes and enforce distinct structural states, most likely regulating the physiological function of APP and its processing in Alzheimer's disease.


Assuntos
Precursor de Proteína beta-Amiloide/química , Cobre/química , Zinco/química , Sequência de Aminoácidos , Sítios de Ligação , Cristalografia por Raios X , Histidina/química , Humanos , Dados de Sequência Molecular , Conformação Proteica
12.
Mol Microbiol ; 79(2): 342-58, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21219456

RESUMO

Oxidation of methionine to methionine sulphoxide (MetSO) may lead to loss of molecular integrity and function. This oxidation can be 'repaired' by methionine sulphoxide reductases (MSRs), which reduce MetSO back to methionine. Two structurally unrelated classes of MSRs, MSRA and MSRB, show stereoselectivity towards the S and the R enantiomer of the sulphoxide respectively. Interestingly, these enzymes were even maintained throughout evolution in anaerobic organisms. Here, the activity and the nuclear magnetic resonance (NMR) structure of MTH711, a zinc containing MSRB from the thermophilic, methanogenic archaebacterium Methanothermobacter thermoautotrophicus, are described. The structure appears more rigid as compared with similar MSRBs from aerobic and mesophilic organisms. No significant structural differences between the oxidized and the reduced MTH711 state can be deduced from our NMR data. A stable sulphenic acid is formed at the catalytic Cys residue upon oxidation of the enzyme with MetSO. The two non-zinc-binding cysteines outside the catalytic centre are not necessary for activity of MTH711 and are not situated close enough to the active-site cysteine to serve in regenerating the active centre via the formation of an intramolecular disulphide bond. These findings imply a reaction cycle that differs from that observed for other MSRBs.


Assuntos
Proteínas Arqueais/química , Proteínas Arqueais/metabolismo , Methanobacteriaceae/enzimologia , Metionina Sulfóxido Redutases/química , Metionina Sulfóxido Redutases/metabolismo , Sequência de Aminoácidos , Domínio Catalítico , Methanobacteriaceae/química , Methanobacteriaceae/metabolismo , Metionina/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Ressonância Magnética Nuclear Biomolecular , Oxirredução , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Alinhamento de Sequência
13.
Brain Pathol ; 21(4): 405-18, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21091576

RESUMO

DEP-1/PTPRJ is a transmembrane protein-tyrosine phosphatase which has been proposed as a suppressor of epithelial tumors. We have found loss of heterozygosity (LOH) of the PTPRJ gene and loss of DEP-1 protein expression in a subset of human meningiomas. RNAi-mediated suppression of DEP-1 in DEP-1 positive meningioma cell lines caused enhanced motility and colony formation in semi-solid media. Cells devoid of DEP-1 exhibited enhanced signaling of endogenous platelet-derived growth factor (PDGF) receptors, and reduced paxillin phosphorylation upon seeding. Moreover, DEP-1 loss caused diminished adhesion to different matrices, and impaired cell spreading. DEP-1-deficient meningioma cells exhibited invasive growth in an orthotopic xenotransplantation model in nude mice, indicating that elevated motility translates into a biological phenotype in vivo. We propose that negative regulation of PDGF receptor signaling and positive regulation of adhesion signaling by DEP-1 cooperate in inhibition of meningioma cell motility, and possibly tumor invasiveness. These phenotypes of DEP-1 loss reveal functions of DEP-1 in adherent cells, and may be more generally relevant for tumorigenesis.


Assuntos
Movimento Celular/genética , Neoplasias Meníngeas/metabolismo , Meningioma/metabolismo , Invasividade Neoplásica/genética , Animais , Adesão Celular/genética , Linhagem Celular Tumoral , Técnicas de Silenciamento de Genes , Humanos , Immunoblotting , Imuno-Histoquímica , Perda de Heterozigosidade , Neoplasias Meníngeas/genética , Neoplasias Meníngeas/patologia , Meningioma/genética , Meningioma/patologia , Camundongos , Camundongos Nus , Paxilina/metabolismo , Reação em Cadeia da Polimerase , RNA Interferente Pequeno , Proteínas Tirosina Fosfatases Classe 3 Semelhantes a Receptores/genética , Proteínas Tirosina Fosfatases Classe 3 Semelhantes a Receptores/metabolismo , Transdução de Sinais/fisiologia , Transfecção
14.
Anal Biochem ; 405(1): 11-8, 2010 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-20522331

RESUMO

Enzymes with 3'-5' exonuclease activities are important in promoting the accuracy of DNA replication and DNA repair by proofreading. The alteration of the function of these enzymes by endogenous or exogenous effectors could, therefore, have a considerable impact on DNA replication and ultimately on genome integrity. We have developed a label-free high-throughput screening method for quantifying the effects of different reagents on exonuclease activity. The assay is based on a hairpin-forming biotinylated oligonucleotide substrate that contains one or more exonuclease-resistant phosphorothioate nucleotides. The activity and specificity of the selected 3'-5' exonuclease is determined indirectly using a sensitive pyrosequencing reaction after cleanup of the samples. In this pyrosequencing step, the amount of nucleotides filled into each position of the exonucleolytically degraded 3' end of the substrate can be recorded quantitatively and equals the amount of the nucleotides removed by the exonuclease. This system allows the estimation of both processivity and efficiency of the exonuclease activity. We have employed compounds reported in the literature to inhibit the exonuclease activities of either exonuclease III or the large fragment of polymerase I (Klenow fragment) to evaluate the assay.


Assuntos
DNA Polimerase I/metabolismo , Exodesoxirribonucleases/metabolismo , Análise de Sequência de DNA/métodos , Reparo do DNA , Replicação do DNA , Ensaios de Triagem em Larga Escala , Sequências Repetidas Invertidas , Oligonucleotídeos/metabolismo , Fosfatos/química
15.
Proc Natl Acad Sci U S A ; 107(12): 5381-6, 2010 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-20212142

RESUMO

The amyloid precursor protein (APP) is the key player in Alzheimer's disease pathology, yet APP and its analogues are also essential for neuronal development and cell homeostasis in mammals. We have determined the crystal structure of the entire N-terminal APP-E1 domain consisting of the growth factor like and the copper binding domains at 2.7-A resolution and show that E1 functions as a rigid functional entity. The two subdomains interact tightly in a pH-dependent manner via an evolutionarily conserved interface area. Two E1 entities dimerize upon their interaction with heparin, requiring 8-12 sugar rings to form the heparin-bridged APP-E1 dimer in an endothermic and pH-dependent process that is characterized by a low micromolar dissociation constant. Limited proteolysis confirms that the heparin-bridged E1 dimers obtained in solution correspond to a dimer contact in our crystal, enabling us to model this heparin-[APP-E1](2) complex. Correspondingly, the APP-based signal transduction, cell-cell- and/or cell-ECM interaction should depend on dimerization induced by heparin, as well as on pH, arguing that APP could fulfill different functions depending on its (sub)cellular localization.


Assuntos
Precursor de Proteína beta-Amiloide/química , Precursor de Proteína beta-Amiloide/metabolismo , Fenômenos Biofísicos , Cristalografia por Raios X , Heparina , Humanos , Concentração de Íons de Hidrogênio , Técnicas In Vitro , Modelos Moleculares , Domínios e Motivos de Interação entre Proteínas , Multimerização Proteica , Estrutura Terciária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo
16.
Genes Dev ; 23(2): 223-35, 2009 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-19171783

RESUMO

Cytokines such as interferons (IFNs) activate signal transducers and activators of transcription (STATs) via phosphorylation. Histone deacetylases (HDACs) and the histone acetyltransferase (HAT) CBP dynamically regulate STAT1 acetylation. Here we show that acetylation of STAT1 counteracts IFN-induced STAT1 phosphorylation, nuclear translocation, DNA binding, and target gene expression. Biochemical and genetic experiments altering the HAT/HDAC activity ratio and STAT1 mutants reveal that a phospho-acetyl switch regulates STAT1 signaling via CBP, HDAC3, and the T-cell protein tyrosine phosphatase (TCP45). Strikingly, inhibition of STAT1 signaling via CBP-mediated acetylation is distinct from the functions of this HAT in transcriptional activation. STAT1 acetylation induces binding of TCP45, which catalyzes dephosphorylation and latency of STAT1. Our results provide a deeper understanding of the modulation of STAT1 activity. These findings reveal a new layer of physiologically relevant STAT1 regulation and suggest that a previously unidentified balance between phosphorylation and acetylation affects cytokine signaling.


Assuntos
Regulação da Expressão Gênica , Fator de Transcrição STAT1/metabolismo , Transdução de Sinais/fisiologia , Acetilação , Linhagem Celular , Histona Desacetilases/metabolismo , Humanos , Interferon-alfa/metabolismo , Monoéster Fosfórico Hidrolases/metabolismo , Fosforilação , Ligação Proteica , Proteína Tirosina Fosfatase não Receptora Tipo 2/metabolismo
17.
Chembiochem ; 4(11): 1151-63, 2003 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-14613106

RESUMO

The non-natural amino acid 4-fluorophenylglycine (4F-Phg) was incorporated into several representative membrane-associated peptides for dual purpose. The (19)F-substituted ring is directly attached to the peptide backbone, so it not only provides a well-defined label for highly sensitive (19)F NMR studies but, in addition, the D and L enantiomers of the stiff side chain may serve as reporter groups on the transient peptide conformation during the biological function. Besides peptide synthesis, which is accompanied by racemisation of 4F-Phg, we also describe separation of the epimers by HPLC and removal of trifluoroacetic acid. As a first example, 18 different analogues of the fusogenic peptide "B18" were prepared and tested for induction of vesicle fusion; the results confirmed that hydrophobic sites tolerated 4F-Phg labelling. Similar fusion activities within each pair of epimers suggest that the peptide is less structured in the fusogenic transition state than in the helical ground state. In a second example, five doubly labelled analogues of the antimicrobial peptide gramicidin S were compared by using bacterial growth inhibition assays. This cyclic beta-sheet peptide could accommodate both L and D substituents on its hydrophobic face. As a third example, we tested six analogues of the antimicrobial peptide PGLa. The presence of d-4F-Phg reduced the biological activity of the peptide by interfering with its amphiphilic alpha-helical fold. Finally, to illustrate the numerous uses of l-4F-Phg in (19)F NMR spectroscopy, we characterised the interaction of labelled PGLa with uncharged and negatively charged membranes. Observing the signal of the free peptide in an aqueous suspension of unilamellar vesicles, we found a linear saturation behaviour that was dominated by electrostatic attraction of the cationic PGLa. Once the peptide is bound to the membrane, however, solid-state (19)F NMR spectroscopy of macroscopically oriented samples revealed that the charge density has virtually no further influence on the structure, alignment or mobility of the peptide.


Assuntos
Marcadores de Afinidade/química , Peptídeos Catiônicos Antimicrobianos/análise , Proteínas de Bactérias/metabolismo , Glicina/análogos & derivados , Glicina/química , Proteínas de Membrana/metabolismo , Peptídeos Catiônicos Antimicrobianos/metabolismo , Peptídeos Catiônicos Antimicrobianos/farmacologia , Sítios de Ligação , Escherichia coli/efeitos dos fármacos , Escherichia coli/metabolismo , Flúor , Espectroscopia de Ressonância Magnética , Modelos Moleculares , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/metabolismo
18.
Mol Pharmacol ; 62(2): 203-9, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12130670

RESUMO

In an effort to combine the benefits of fibrinolytics, such as staphylokinase, with those of thrombin inhibitors for the prevention of vessel reocclusion after vascular injury, we have produced several chimeric proteins with plasminogen-activating and thrombin-inhibiting properties. Fusion proteins were constructed consisting of the modules staphylokinase (Sak), the factor Xa cleavage site, and various dipetalin (Dip) domains (H(6)-Sak-Dip-I+II, H(6)-Sak-Dip-I, and H(6)-Sak-Dip-II). Sak stimulates fibrinolysis via activation of plasminogen, whereas dipetalin is a two-domain, Kazal-type inhibitor of thrombin. NMR spectroscopy of the fusion proteins revealed that the molecular structures of the modules are retained in the fusion protein and that no significant interactions occur between the modules in terms of their functionally relevant regions. In enzymatic thrombin inhibition tests and blood coagulation assays (thrombin, prothrombin, and activated partial thromboplastin times), no significant differences in anticoagulant capacity were observed between the fusion protein H(6)-Sak-Dip-I+II and isolated Dip-I+II, even at nanomolar concentrations. Similar results (i.e., the inhibition of thrombin-induced platelet aggregation and the inhibition of thrombin-induced vascular relaxation) were obtained when the cellular thrombin effects were studied. The fusion protein containing Dip-I has less but still significant thrombin inhibitory effects compared with those of H(6)-Sak-Dip-I+II. In contrast, the H(6)-Sak-Dip-II protein failed to inhibit thrombin in each of the assays used. The plasminogen-activating and fibrinolytic activities of the fusion proteins are similar to those of wild-type Sak. The individual dipetalin domains do not activate plasminogen. In conclusion, the fusion protein H(6)-Sak-Dip-I+II is a bifunctional molecule able to activate fibrinolysis via plasminogen activation and inhibit blood coagulation via direct inhibition of thrombin.


Assuntos
Anticoagulantes/farmacologia , Fibrinolíticos/farmacologia , Proteínas de Insetos/farmacologia , Metaloendopeptidases/farmacologia , Plasminogênio/metabolismo , Animais , Anticoagulantes/química , Fibrinolíticos/química , Humanos , Técnicas In Vitro , Proteínas de Insetos/genética , Espectroscopia de Ressonância Magnética , Metaloendopeptidases/genética , Modelos Moleculares , Agregação Plaquetária/efeitos dos fármacos , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/farmacologia , Relação Estrutura-Atividade , Trombina/fisiologia , Vasodilatação/efeitos dos fármacos
19.
J Mol Biol ; 318(2): 533-46, 2002 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-12051857

RESUMO

The interaction of domains of the Kazal-type inhibitor protein dipetalin with the serine proteinases thrombin and trypsin is studied. The functional studies of the recombinantly expressed domains (Dip-I+II, Dip-I and Dip-II) allow the dissection of the thrombin inhibitory properties and the identification of Dip-I as a key contributor to thrombin/dipetalin complex stability and its inhibitory potency. Furthermore, Dip-I, but not Dip-II, forms a complex with trypsin resulting in an inhibition of the trypsin activity directed towards protein substrates. The high resolution NMR structure of the Dip-I domain is determined using multi-dimensional heteronuclear NMR spectroscopy. Dip-I exhibits the canonical Kazal-type fold with a central alpha-helix and a short two-stranded antiparallel beta-sheet. Molecular regions essential for inhibitor complex formation with thrombin and trypsin are identified. A comparison with molecular complexes of other Kazal-type thrombin and trypsin inhibitors by molecular modeling shows that the N-terminal segment of Dip-I fulfills the structural prerequisites for inhibitory interactions with either proteinase and explains the capacity of this single Kazal-type domain to interact with different proteinases.


Assuntos
Inibidores de Serina Proteinase/química , Sequência de Aminoácidos , Animais , Sítios de Ligação , Cristalografia por Raios X , Técnicas In Vitro , Proteínas de Insetos/química , Substâncias Macromoleculares , Modelos Moleculares , Dados de Sequência Molecular , Ressonância Magnética Nuclear Biomolecular , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Inibidores de Serina Proteinase/genética , Trombina/química , Tripsina/química , Inibidor da Tripsina Pancreática de Kazal/química , Inibidor da Tripsina Pancreática de Kazal/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...