Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
1.
J Biol Chem ; 296: 100684, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33891946

RESUMO

Botulinum neurotoxins (BoNTs) are among the most widely used therapeutic proteins; however, only two subtypes within the seven serotypes, BoNT/A1 and BoNT/B1, are currently used for medical and cosmetic applications. Distinct catalytic properties, substrate specificities, and duration of enzymatic activities potentially make other subtypes very attractive candidates to outperform conventional BoNTs in particular therapeutic applications. For example, BoNT/A3 has a significantly shorter duration of action than other BoNT/A subtypes. Notably, BoNT/A3 is the subtype with the least conserved catalytic domain among BoNT/A subtypes. This suggests that the sequence differences, many of which concern the α-exosite, contribute to the observed functional differences in toxin persistence by affecting the binding of the substrate SNAP-25 and/or the stability of the catalytic domain fold. To identify the molecular determinants accounting for the differences in the persistence observed for BoNT/A subtypes, we determined the crystal structure of the catalytic domain of BoNT/A3 (LC/A3). The structure of LC/A3 was found to be very similar to that of LC/A1, suggesting that the overall mode of SNAP-25 binding is common between these two proteins. However, circular dichroism (CD) thermal unfolding experiments demonstrated that LC/A3 is significantly less stable than LC/A1, implying that this might contribute to the reduced toxin persistence of BoNT/A3. These findings could be of interest in developing next-generation therapeutic toxins.


Assuntos
Toxinas Botulínicas Tipo A/química , Domínio Catalítico , Sequência de Aminoácidos , Toxinas Botulínicas Tipo A/metabolismo , Cristalografia por Raios X , Modelos Moleculares , Especificidade por Substrato
2.
Nature ; 505(7481): 108-11, 2014 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-24240280

RESUMO

Botulinum neurotoxin A (BoNT/A) belongs to the most dangerous class of bioweapons. Despite this, BoNT/A is used to treat a wide range of common medical conditions such as migraines and a variety of ocular motility and movement disorders. BoNT/A is probably best known for its use as an antiwrinkle agent in cosmetic applications (including Botox and Dysport). BoNT/A application causes long-lasting flaccid paralysis of muscles through inhibiting the release of the neurotransmitter acetylcholine by cleaving synaptosomal-associated protein 25 (SNAP-25) within presynaptic nerve terminals. Two types of BoNT/A receptor have been identified, both of which are required for BoNT/A toxicity and are therefore likely to cooperate with each other: gangliosides and members of the synaptic vesicle glycoprotein 2 (SV2) family, which are putative transporter proteins that are predicted to have 12 transmembrane domains, associate with the receptor-binding domain of the toxin. Recently, fibroblast growth factor receptor 3 (FGFR3) has also been reported to be a potential BoNT/A receptor. In SV2 proteins, the BoNT/A-binding site has been mapped to the luminal domain, but the molecular details of the interaction between BoNT/A and SV2 are unknown. Here we determined the high-resolution crystal structure of the BoNT/A receptor-binding domain (BoNT/A-RBD) in complex with the SV2C luminal domain (SV2C-LD). SV2C-LD consists of a right-handed, quadrilateral ß-helix that associates with BoNT/A-RBD mainly through backbone-to-backbone interactions at open ß-strand edges, in a manner that resembles the inter-strand interactions in amyloid structures. Competition experiments identified a peptide that inhibits the formation of the complex. Our findings provide a strong platform for the development of novel antitoxin agents and for the rational design of BoNT/A variants with improved therapeutic properties.


Assuntos
Toxinas Botulínicas Tipo A/química , Toxinas Botulínicas Tipo A/metabolismo , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/metabolismo , Sítios de Ligação , Cristalografia por Raios X , Endocitose/efeitos dos fármacos , Células HEK293 , Humanos , Modelos Moleculares , Neostriado/citologia , Neurônios/efeitos dos fármacos , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/farmacologia , Relação Estrutura-Atividade
3.
Proc Natl Acad Sci U S A ; 114(46): E9821-E9828, 2017 11 14.
Artigo em Inglês | MEDLINE | ID: mdl-29087332

RESUMO

Nucleotidyl cyclases, including membrane-integral and soluble adenylyl and guanylyl cyclases, are central components in a wide range of signaling pathways. These proteins are architecturally diverse, yet many of them share a conserved feature, a helical region that precedes the catalytic cyclase domain. The role of this region in cyclase dimerization has been a subject of debate. Although mutations within this region in various cyclases have been linked to genetic diseases, the molecular details of their effects on the enzymes remain unknown. Here, we report an X-ray structure of the cytosolic portion of the membrane-integral adenylyl cyclase Cya from Mycobacterium intracellulare in a nucleotide-bound state. The helical domains of each Cya monomer form a tight hairpin, bringing the two catalytic domains into an active dimerized state. Mutations in the helical domain of Cya mimic the disease-related mutations in human proteins, recapitulating the profiles of the corresponding mutated enzymes, adenylyl cyclase-5 and retinal guanylyl cyclase-1. Our experiments with full-length Cya and its cytosolic domain link the mutations to protein stability, and the ability to induce an active dimeric conformation of the catalytic domains. Sequence conservation indicates that this domain is an integral part of cyclase machinery across protein families and species. Our study provides evidence for a role of the helical domain in establishing a catalytically competent dimeric cyclase conformation. Our results also suggest that the disease-associated mutations in the corresponding regions of human nucleotidyl cyclases disrupt the normal helical domain structure.


Assuntos
Adenilil Ciclases/química , Adenilil Ciclases/metabolismo , Domínio Catalítico , Complexo Mycobacterium avium/enzimologia , Conformação Proteica , Adenilil Ciclases/genética , Substituição de Aminoácidos , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Sequência Conservada , Cristalografia por Raios X , Citosol/enzimologia , Dimerização , Ativação Enzimática , Estabilidade Enzimática , Guanilato Ciclase/química , Guanilato Ciclase/genética , Humanos , Modelos Moleculares , Mutagênese Sítio-Dirigida , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Complexo Mycobacterium avium/genética , Receptores de Superfície Celular/química , Receptores de Superfície Celular/genética , Alinhamento de Sequência , Análise de Sequência de Proteína
4.
Trends Biochem Sci ; 39(11): 517-26, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25282537

RESUMO

Botulinum neurotoxins (BoNTs) are the most toxic substances known and cause botulism in vertebrates. They have also emerged as effective and powerful reagents for cosmetic and medical applications. One important prerequisite for understanding BoNT function in disease, and the further development of the toxins for cosmetic and medical applications, is a detailed knowledge of BoNT interactions with non-toxic neurotoxin-associated proteins and cell surface receptors. Based on the substantial recent progress in obtaining high-resolution crystal structures of key BoNT complexes, we summarize the major advances in understanding BoNT interactions and discuss the resulting potential implications, in particular those relating to BoNT serotype A.


Assuntos
Toxinas Botulínicas Tipo A/química , Glicoproteínas de Membrana/química , Proteínas do Tecido Nervoso/química , Estrutura Terciária de Proteína , Sequência de Aminoácidos , Toxinas Botulínicas Tipo A/genética , Toxinas Botulínicas Tipo A/metabolismo , Humanos , Glicoproteínas de Membrana/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Proteínas do Tecido Nervoso/metabolismo , Ligação Proteica , Homologia de Sequência de Aminoácidos
5.
Biochemistry ; 56(11): 1604-1619, 2017 03 21.
Artigo em Inglês | MEDLINE | ID: mdl-28230348

RESUMO

To understand the roles ion pairs play in stabilizing coiled coils, we determined nuclear magnetic resonance structures of GCN4p at three pH values. At pH 6.6, all acidic residues are fully charged; at pH 4.4, they are half-charged, and at pH 1.5, they are protonated and uncharged. The α-helix monomer and coiled coil structures of GCN4p are largely conserved, except for a loosening of the coiled coil quaternary structure with a decrease in pH. Differences going from neutral to acidic pH include (i) an unwinding of the coiled coil superhelix caused by the loss of interchain ion pair contacts, (ii) a small increase in the separation of the monomers in the dimer, (iii) a loosening of the knobs-into-holes packing motifs, and (iv) an increased separation between oppositely charged residues that participate in ion pairs at neutral pH. Chemical shifts (HN, N, C', Cα, and Cß) of GCN4p display a seven-residue periodicity that is consistent with α-helical structure and is invariant with pH. By contrast, periodicity in hydrogen exchange rates at neutral pH is lost at acidic pH as the exchange mechanism moves into the EX1 regime. On the basis of 1H-15N nuclear Overhauser effect relaxation measurements, the α-helix monomers experience only small increases in picosecond to nanosecond backbone dynamics at acidic pH. By contrast, 13C rotating frame T1 relaxation (T1ρ) data evince an increase in picosecond to nanosecond side-chain dynamics at lower pH, particularly for residues that stabilize the coiled coil dimerization interface through ion pairs. The results on the structure and dynamics of GCNp4 over a range of pH values help rationalize why a single structure at neutral pH poorly predicts the pH dependence of the unfolding stability of the coiled coil.


Assuntos
Fatores de Transcrição de Zíper de Leucina Básica/química , Simulação de Dinâmica Molecular , Fosfoproteínas/química , Prótons , Proteínas Recombinantes/química , Proteínas de Saccharomyces cerevisiae/química , Fatores de Transcrição de Zíper de Leucina Básica/genética , Fatores de Transcrição de Zíper de Leucina Básica/metabolismo , Clonagem Molecular , Escherichia coli/genética , Escherichia coli/metabolismo , Expressão Gênica , Concentração de Íons de Hidrogênio , Espectroscopia de Ressonância Magnética , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Dobramento de Proteína , Multimerização Proteica , Estrutura Secundária de Proteína , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Eletricidade Estática , Termodinâmica
6.
J Biol Chem ; 291(35): 18496-504, 2016 08 26.
Artigo em Inglês | MEDLINE | ID: mdl-27402853

RESUMO

Dysfunction of cilia is associated with common genetic disorders termed ciliopathies. Knowledge on the interaction networks of ciliary proteins is therefore key for understanding the processes that are underlying these severe diseases and the mechanisms of ciliogenesis in general. Cep104 has recently been identified as a key player in the regulation of cilia formation. Using a combination of sequence analysis, biophysics, and x-ray crystallography, we obtained new insights into the domain architecture and interaction network of the Cep104 protein. We solved the crystal structure of the tumor overexpressed gene (TOG) domain, identified Cep104 as a novel tubulin-binding protein, and biophysically characterized the interaction of Cep104 with CP110, Cep97, end-binding (EB) protein, and tubulin. Our results represent a solid platform for the further investigation of the microtubule-EB-Cep104-tubulin-CP110-Cep97 network of proteins. Ultimately, such studies should be of importance for understanding the process of cilia formation and the mechanisms underlying different ciliopathies.


Assuntos
Proteínas Associadas aos Microtúbulos/química , Centríolos/química , Centríolos/genética , Centríolos/metabolismo , Cílios/química , Cílios/metabolismo , Ciliopatias/genética , Ciliopatias/metabolismo , Cristalografia por Raios X , Humanos , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Domínios Proteicos
7.
PLoS Biol ; 12(3): e1001820, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24667537

RESUMO

Cognitive and behavioral disorders are thought to be a result of neuronal dysfunction, but the underlying molecular defects remain largely unknown. An important signaling pathway involved in the regulation of neuronal function is the cyclic AMP/Protein kinase A pathway. We here show an essential role for coronin 1, which is encoded in a genomic region associated with neurobehavioral dysfunction, in the modulation of cyclic AMP/PKA signaling. We found that coronin 1 is specifically expressed in excitatory but not inhibitory neurons and that coronin 1 deficiency results in loss of excitatory synapses and severe neurobehavioral disabilities, including reduced anxiety, social deficits, increased aggression, and learning defects. Electrophysiological analysis of excitatory synaptic transmission in amygdala revealed that coronin 1 was essential for cyclic-AMP-protein kinase A-dependent presynaptic plasticity. We further show that upon cell surface stimulation, coronin 1 interacted with the G protein subtype Gαs to stimulate the cAMP/PKA pathway. The absence of coronin 1 or expression of coronin 1 mutants unable to interact with Gαs resulted in a marked reduction in cAMP signaling. Strikingly, synaptic plasticity and behavioral defects of coronin 1-deficient mice were restored by in vivo infusion of a membrane-permeable cAMP analogue. Together these results identify coronin 1 as being important for cognition and behavior through its activity in promoting cAMP/PKA-dependent synaptic plasticity and may open novel avenues for the dissection of signal transduction pathways involved in neurobehavioral processes.


Assuntos
Comportamento Animal , Cognição/fisiologia , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Proteínas dos Microfilamentos/fisiologia , 4-Butirolactona/análogos & derivados , 4-Butirolactona/genética , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Humanos , Memória , Camundongos , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/metabolismo , Transdução de Sinais , Comportamento Social
8.
J Cell Sci ; 127(Pt 12): 2672-82, 2014 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-24706950

RESUMO

Crosstalk between the microtubule (MT) and actin cytoskeletons is fundamental to many cellular processes including cell polarisation and cell motility. Previous work has shown that members of the growth-arrest-specific 2 (GAS2) family mediate the crosstalk between filamentous actin (F-actin) and MTs, but the molecular basis of this process remained unclear. By using fluorescence microscopy, we demonstrate that three members of this family, GAS2-like 1, GAS2-like 2 and GAS2-like 3 (G2L1, G2L2 and G2L3, also known as GAS2L1, GAS2L2 and GAS2L3, respectively) are differentially involved in mediating the crosstalk between F-actin and MTs. Although all localise to actin and MTs, only the exogenous expression of G2L1 and G2L2 influenced MT stability, dynamics and guidance along actin stress fibres. Biochemical analysis and live-cell imaging revealed that their functions are largely due to the association of these proteins with MT plus-end-binding proteins that bind to SxIP or SxLP motifs located at G2L C-termini. Our findings lead to a model in which end-binding (EB) proteins play a key role in mediating actin-MT crosstalk.


Assuntos
Actinas/metabolismo , Proteínas dos Microfilamentos/fisiologia , Proteínas Associadas aos Microtúbulos/fisiologia , Microtúbulos/metabolismo , Fibras de Estresse/metabolismo , Animais , Células CHO , Células COS , Linhagem Celular Tumoral , Chlorocebus aethiops , Sequência Conservada , Cricetinae , Cricetulus , Humanos , Camundongos , Proteínas Associadas aos Microtúbulos/metabolismo , Células NIH 3T3 , Ligação Proteica , Sinais Direcionadores de Proteínas , Transporte Proteico
9.
J Biol Chem ; 289(49): 34175-88, 2014 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-25339170

RESUMO

The causative agent of Legionnaires' pneumonia, Legionella pneumophila, colonizes diverse environmental niches, including biofilms, plant material, and protozoa. In these habitats, myo-inositol hexakisphosphate (phytate) is prevalent and used as a phosphate storage compound or as a siderophore. L. pneumophila replicates in protozoa and mammalian phagocytes within a unique "Legionella-containing vacuole." The bacteria govern host cell interactions through the Icm/Dot type IV secretion system (T4SS) and ∼300 different "effector" proteins. Here we characterize a hitherto unrecognized Icm/Dot substrate, LppA, as a phytate phosphatase (phytase). Phytase activity of recombinant LppA required catalytically essential cysteine (Cys(231)) and arginine (Arg(237)) residues. The structure of LppA at 1.4 Å resolution revealed a mainly α-helical globular protein stabilized by four antiparallel ß-sheets that binds two phosphate moieties. The phosphates localize to a P-loop active site characteristic of dual specificity phosphatases or to a non-catalytic site, respectively. Phytate reversibly abolished growth of L. pneumophila in broth, and growth inhibition was relieved by overproduction of LppA or by metal ion titration. L. pneumophila lacking lppA replicated less efficiently in phytate-loaded Acanthamoeba castellanii or Dictyostelium discoideum, and the intracellular growth defect was complemented by the phytase gene. These findings identify the chelator phytate as an intracellular bacteriostatic component of cell-autonomous host immunity and reveal a T4SS-translocated L. pneumophila phytase that counteracts intracellular bacterial growth restriction by phytate. Thus, bacterial phytases might represent therapeutic targets to combat intracellular pathogens.


Assuntos
6-Fitase/química , Proteínas de Bactérias/química , Sistemas de Secreção Bacterianos/genética , Legionella pneumophila/enzimologia , Ácido Fítico/metabolismo , 6-Fitase/genética , 6-Fitase/metabolismo , Acanthamoeba castellanii/metabolismo , Acanthamoeba castellanii/microbiologia , Arginina/química , Arginina/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Domínio Catalítico , Cisteína/química , Cisteína/metabolismo , Dictyostelium/metabolismo , Dictyostelium/microbiologia , Escherichia coli/genética , Escherichia coli/metabolismo , Expressão Gênica , Teste de Complementação Genética , Interações Hospedeiro-Patógeno , Cinética , Legionella pneumophila/efeitos dos fármacos , Legionella pneumophila/genética , Ácido Fítico/química , Ácido Fítico/farmacologia , Estrutura Secundária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
10.
Pharmacol Res ; 80: 43-51, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24407281

RESUMO

The growth factor angiopoietin-1 (Ang-1) plays an essential role in angiogenesis and vascular homeostasis. Nevertheless, the role of Ang-1 in regulating vascular tone and blood flow is largely unexplored. Endothelial nitric oxide synthase (eNOS) and the junctional protein VE-cadherin are part of the complex signalling cascade initiated by Ang-1 in endothelial cells. In this study, we aimed to investigate the mechanisms underlying acute effects of Ang-1 on microvascular reactivity, permeability and blood flow, and hypothesise that eNOS and VE-cadherin underpin Ang-1 mediated vascular effects that are independent of angiogenesis and proliferation. Myography of isolated microarterioles from male C3H/HeN mice (7-10 weeks) was employed to measure vascular reactivity in vitro. Microcirculatory function in vivo was evaluated by intravital microscopy and Doppler fluximetry in dorsal window chambers. Ang-1 and its stable variant MAT.Ang-1 induced a concentration-dependent vasodilation of arterioles in vitro, which was blocked with nitric oxide (NO) synthesis inhibitor l-NAME. In vivo, MAT.Ang-1 restored to control levels l-NAME induced peripheral vasoconstriction, decreased blood flow and microvascular hyperpermeability. Tissue protein expression of VE-cadherin was reduced by NOS inhibition and restored to control levels by MAT.Ang-1, whilst VE-cadherin phosphorylation was increased by l-NAME and subsequently reduced by MAT.Ang-1 administration. Moreover, MAT.Ang-1 alone did not modulate systemic levels of angiogenetic factors. Our novel findings report that Ang-1 induces arteriolar vasodilation via release of NO, suggesting that Ang-1 is an important regulator of microvascular tone. As MAT.Ang-1 ameliorates detrimental effects on the microcirculation induced by inhibition of NO synthesis and stabilizes the endothelial barrier function through VE-cadherin, we propose that this Ang-1 variant may serve as a novel therapeutic agent to protect the microcirculation against endothelial dysfunction.


Assuntos
Angiopoietina-1/fisiologia , Antígenos CD/fisiologia , Caderinas/fisiologia , Permeabilidade Capilar/fisiologia , Microcirculação/fisiologia , Óxido Nítrico Sintase Tipo III/fisiologia , Angiopoietina-1/antagonistas & inibidores , Angiopoietina-1/farmacologia , Animais , Antígenos CD/biossíntese , Antígenos CD/efeitos dos fármacos , Arteríolas/efeitos dos fármacos , Arteríolas/fisiologia , Caderinas/biossíntese , Caderinas/efeitos dos fármacos , Permeabilidade Capilar/efeitos dos fármacos , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Masculino , Camundongos , Microcirculação/efeitos dos fármacos , Músculo Estriado/irrigação sanguínea , Músculo Estriado/efeitos dos fármacos , Músculo Estriado/fisiologia , NG-Nitroarginina Metil Éster/antagonistas & inibidores , NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico Sintase Tipo III/antagonistas & inibidores , Fosforilação/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/efeitos da radiação , Vasoconstrição/efeitos dos fármacos , Vasoconstrição/fisiologia , Vasodilatação/efeitos dos fármacos , Vasodilatação/fisiologia
11.
Nat Chem ; 2024 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-38744914

RESUMO

Membrane-bound styrene oxide isomerase (SOI) catalyses the Meinwald rearrangement-a Lewis-acid-catalysed isomerization of an epoxide to a carbonyl compound-and has been used in single and cascade reactions. However, the structural information that explains its reaction mechanism has remained elusive. Here we determine cryo-electron microscopy (cryo-EM) structures of SOI bound to a single-domain antibody with and without the competitive inhibitor benzylamine, and elucidate the catalytic mechanism using electron paramagnetic resonance spectroscopy, functional assays, biophysical methods and docking experiments. We find ferric haem b bound at the subunit interface of the trimeric enzyme through H58, where Fe(III) acts as the Lewis acid by binding to the epoxide oxygen. Y103 and N64 and a hydrophobic pocket binding the oxygen of the epoxide and the aryl group, respectively, position substrates in a manner that explains the high regio-selectivity and stereo-specificity of SOI. Our findings can support extending the range of epoxide substrates and be used to potentially repurpose SOI for the catalysis of new-to-nature Fe-based chemical reactions.

12.
J Neurosci ; 32(27): 9143-58, 2012 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-22764224

RESUMO

The correct outgrowth of axons is essential for the development and regeneration of nervous systems. Axon growth is primarily driven by microtubules. Key regulators of microtubules in this context are the spectraplakins, a family of evolutionarily conserved actin-microtubule linkers. Loss of function of the mouse spectraplakin ACF7 or of its close Drosophila homolog Short stop/Shot similarly cause severe axon shortening and microtubule disorganization. How spectraplakins perform these functions is not known. Here we show that axonal growth-promoting roles of Shot require interaction with EB1 (End binding protein) at polymerizing plus ends of microtubules. We show that binding of Shot to EB1 requires SxIP motifs in Shot's C-terminal tail (Ctail), mutations of these motifs abolish Shot functions in axonal growth, loss of EB1 function phenocopies Shot loss, and genetic interaction studies reveal strong functional links between Shot and EB1 in axonal growth and microtubule organization. In addition, we report that Shot localizes along microtubule shafts and stabilizes them against pharmacologically induced depolymerization. This function is EB1-independent but requires net positive charges within Ctail which essentially contribute to the microtubule shaft association of Shot. Therefore, spectraplakins are true members of two important classes of neuronal microtubule regulating proteins: +TIPs (tip interacting proteins; plus end regulators) and structural MAPs (microtubule-associated proteins). From our data we deduce a model that relates the different features of the spectraplakin C terminus to the two functions of Shot during axonal growth.


Assuntos
Actinas/fisiologia , Axônios/fisiologia , Proteínas de Drosophila/fisiologia , Drosophila melanogaster/embriologia , Proteínas dos Microfilamentos/fisiologia , Proteínas Associadas aos Microtúbulos/fisiologia , Actinas/genética , Motivos de Aminoácidos/genética , Animais , Animais Geneticamente Modificados , Proteínas de Drosophila/antagonistas & inibidores , Proteínas de Drosophila/deficiência , Drosophila melanogaster/citologia , Drosophila melanogaster/genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento/genética , Técnicas de Inativação de Genes/métodos , Cones de Crescimento/fisiologia , Masculino , Camundongos , Proteínas dos Microfilamentos/antagonistas & inibidores , Proteínas dos Microfilamentos/deficiência , Mutação , Células NIH 3T3 , Proteínas do Tecido Nervoso/deficiência , Proteínas do Tecido Nervoso/fisiologia , Cultura Primária de Células
13.
Am J Hum Genet ; 86(6): 963-9, 2010 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-20560210

RESUMO

Urinary voiding dysfunction in childhood, manifesting as incontinence, dysuria, and urinary frequency, is a common condition. Urofacial syndrome (UFS) is a rare autosomal recessive disease characterized by facial grimacing when attempting to smile and failure of the urinary bladder to void completely despite a lack of anatomical bladder outflow obstruction or overt neurological damage. UFS individuals often have reflux of infected urine from the bladder to the upper renal tract, with a risk of kidney damage and renal failure. Whole-genome SNP mapping in one affected individual defined an autozygous region of 16 Mb on chromosome 10q23-q24, within which a 10 kb deletion encompassing exons 8 and 9 of HPSE2 was identified. Homozygous exonic deletions, nonsense mutations, and frameshift mutations in five further unrelated families confirmed HPSE2 as the causative gene for UFS. Mutations were not identified in four additional UFS patients, indicating genetic heterogeneity. We show that HPSE2 is expressed in the fetal and adult central nervous system, where it might be implicated in controlling facial expression and urinary voiding, and also in bladder smooth muscle, consistent with a role in renal tract morphology and function. Our findings have broader implications for understanding the genetic basis of lower renal tract malformations and voiding dysfunction.


Assuntos
Fácies , Glucuronidase/genética , Doenças Urológicas/genética , Encéfalo/metabolismo , Criança , Pré-Escolar , Mapeamento Cromossômico , Cromossomos Humanos Par 10 , Feminino , Genes Recessivos , Glucuronidase/química , Glucuronidase/metabolismo , Humanos , Masculino , Modelos Moleculares , Músculos/metabolismo , Mutação , Linhagem , Síndrome , Bexiga Urinária/metabolismo
14.
Proc Natl Acad Sci U S A ; 107(46): 19850-5, 2010 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-21045134

RESUMO

Coiled coils are extensively and successfully used nowadays to rationally design multistranded structures for applications, including basic research, biotechnology, nanotechnology, materials science, and medicine. The wide range of applications as well as the important functions these structures play in almost all biological processes highlight the need for a detailed understanding of the factors that control coiled-coil folding and oligomerization. Here, we address the important and unresolved question why the presence of particular oligomerization-state determinants within a coiled coil does frequently not correlate with its topology. We found an unexpected, general link between coiled-coil oligomerization-state specificity and trigger sequences, elements that are indispensable for coiled-coil formation. By using the archetype coiled-coil domain of the yeast transcriptional activator GCN4 as a model system, we show that well-established trimer-specific oligomerization-state determinants switch the peptide's topology from a dimer to a trimer only when inserted into the trigger sequence. We successfully confirmed our results in two other, unrelated coiled-coil dimers, ATF1 and cortexillin-1. We furthermore show that multiple topology determinants can coexist in the same trigger sequence, revealing a delicate balance of the resulting oligomerization state by position-dependent forces. Our experimental results should significantly improve the prediction of the oligomerization state of coiled coils. They therefore should have major implications for the rational design of coiled coils and consequently many applications using these popular oligomerization domains.


Assuntos
Modelos Moleculares , Estrutura Quaternária de Proteína , Proteínas/química , Fator 1 Ativador da Transcrição/química , Motivos de Aminoácidos , Sequência de Aminoácidos , Fatores de Transcrição de Zíper de Leucina Básica/química , Dicroísmo Circular , Cristalografia por Raios X , Luz , Proteínas dos Microfilamentos/química , Dados de Sequência Molecular , Proteínas Mutantes/química , Multimerização Proteica , Proteínas de Protozoários/química , Proteínas de Saccharomyces cerevisiae/química , Espalhamento de Radiação , Ultracentrifugação
15.
Chem Biodivers ; 10(4): 538-55, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23576341

RESUMO

Kinesin spindle protein (KSP), an ATP-dependent motor protein, plays an essential role in bipolar spindle formation during the mitotic phase (M phase) of the normal cell cycle. KSP has emerged as a novel target for antimitotic anticancer drug development. In this work, we synthesized a range of new biphenyl compounds and investigated their properties in vitro as potential antimitotic agents targeting KSP expression. Antiproliferation (MTT (=3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide)) assays, combined with fluorescence-assisted cell sorting (FACS) and Western blot studies analyzing cell-cycle arrest confirmed the mechanism and potency of these biphenyl compounds in a range of human cancer cell lines. Structural variants revealed that functionalization of biphenyl compounds with bulky aliphatic or aromatic groups led to a loss of activity. However, replacement of the urea group with a thiourea led to an increase in antiproliferative activity in selected cell lines. Further studies using confocal fluorescence microscopy confirmed that the most potent biphenyl derivative identified thus far, compound 7, exerts its pharmacologic effect specifically in the M phase and induces monoaster formation. These studies confirm that chemical scope remains for improving the potency and treatment efficacy of antimitotic KSP inhibition in this class of biphenyl compounds.


Assuntos
Antimitóticos/síntese química , Compostos de Bifenilo/química , Inibidores Enzimáticos/síntese química , Cinesinas/antagonistas & inibidores , Trifosfato de Adenosina/metabolismo , Antimitóticos/química , Antimitóticos/toxicidade , Compostos de Bifenilo/síntese química , Compostos de Bifenilo/toxicidade , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Inibidores Enzimáticos/química , Inibidores Enzimáticos/toxicidade , Células HCT116 , Humanos , Cinesinas/metabolismo , Pontos de Checagem da Fase M do Ciclo Celular/efeitos dos fármacos , Células MCF-7 , Relação Estrutura-Atividade , Tioureia/química
16.
Biomol NMR Assign ; 17(2): 301-307, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37861970

RESUMO

Kinesin is a motor protein, comprised of two heavy and two light chains that transports cargo along the cytoskeletal microtubule filament network. The heavy chain has a neck domain connecting the ATPase motor head responsible for walking along microtubules, with the stalk and subsequent tail domains that bind cargo. The neck domain consists of a coiled coli homodimer with about five heptad repeats, preceded by a linker region that joins to the ATPase head. Here we report 1H, 15N, and 13C NMR assignments and a solution structure for the kinesin neck domain from rat isoform Kif5c. The calculation of the NMR structure of the homodimer was facilitated by unambiguously assigning sidechain NOEs between heptad a and d positions to interchain contacts, since these positions are too far apart to give sidechain contacts in the monomers. The dimeric coiled coil NMR structure is similar to the previously described X-ray structure, whereas the linker region is disordered in solution but contains a short segment with ß-strand propensity- the ß-linker. Only the coiled coil is protected from solvent exchange, with ∆G values for hydrogen exchange on the order of 4-6 kcal/mol. The high stability of the hydrogen-bonded α-helical structure makes it unlikely that unzippering of the coiled coil is involved in kinesin walking. Rather, the linker region serves as a flexible hinge between the kinesin head and neck.


Assuntos
Cinesinas , Microtúbulos , Ratos , Animais , Cinesinas/química , Cinesinas/metabolismo , Sequência de Aminoácidos , Ressonância Magnética Nuclear Biomolecular , Domínios Proteicos , Microtúbulos/metabolismo , Hidrogênio
17.
Sci Rep ; 13(1): 10159, 2023 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-37349348

RESUMO

Structure elucidation of inactive-state GPCRs still mostly relies on X-ray crystallography. The major goal of our work was to create a new GPCR tool that would provide receptor stability and additional soluble surface for crystallization. Towards this aim, we selected the two-stranded antiparallel coiled coil as a domain fold that satisfies both criteria. A selection of antiparallel coiled coils was used for structure-guided substitution of intracellular loop 3 of the ß3 adrenergic receptor. Unexpectedly, only the two GPCR variants containing thermostable coiled coils were expressed. We showed that one GPCR chimera is stable upon purification in detergent, retains ligand-binding properties, and can be crystallized. However, the quality of the crystals was not suitable for structure determination. By using two other examples, 5HTR2C and α2BAR, we demonstrate that our approach is generally suitable for the stabilization of GPCRs. To provide additional surface for promoting crystal contacts, we replaced in a structure-based approach the loop connecting the antiparallel coiled coil by T4L. We found that the engineered GPCR is even more stable than the coiled-coil variant. Negative-staining TEM revealed a homogeneous distribution of particles, indicating that coiled-coil-T4L receptor variants might also be promising candidate proteins for structure elucidation by cryo-EM. Our approach should be of interest for applications that benefit from stable GPCRs.


Assuntos
Receptores Acoplados a Proteínas G , Sequência de Aminoácidos , Estrutura Secundária de Proteína , Cristalografia por Raios X , Domínios Proteicos , Receptores Acoplados a Proteínas G/genética
18.
Nat Commun ; 14(1): 8317, 2023 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-38110403

RESUMO

In this study, we characterize Designed Ankyrin Repeat Proteins (DARPins) as investigative tools to probe botulinum neurotoxin A1 (BoNT/A1) structure and function. We identify DARPin-F5 that completely blocks SNAP25 substrate cleavage by BoNT/A1 in vitro. X-ray crystallography reveals that DARPin-F5 inhibits BoNT/A1 activity by interacting with a substrate-binding region between the α- and ß-exosite. This DARPin does not block substrate cleavage of BoNT/A3, indicating that DARPin-F5 is a subtype-specific inhibitor. BoNT/A1 Glu-171 plays a critical role in the interaction with DARPin-F5 and its mutation to Asp, the residue found in BoNT/A3, results in a loss of inhibition of substrate cleavage. In contrast to the in vitro results, DARPin-F5 promotes faster substrate cleavage of BoNT/A1 in primary neurons and muscle tissue by increasing toxin translocation. Our findings could have important implications for the application of BoNT/A1 in therapeutic areas requiring faster onset of toxin action combined with long persistence.


Assuntos
Toxinas Botulínicas Tipo A , Toxinas Botulínicas , Clostridium botulinum , Proteínas de Repetição de Anquirina Projetadas , Toxinas Botulínicas Tipo A/metabolismo , Clostridium botulinum/genética
19.
J Struct Biol ; 177(1): 160-7, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22119847

RESUMO

End binding proteins (EBs) track growing microtubule ends and play a master role in organizing dynamic protein networks. Mammalian cells express up to three different EBs (EB1, EB2, and EB3). Besides forming homodimers, EB1 and EB3 also assemble into heterodimers. One group of EB-binding partners encompasses proteins that harbor CAP-Gly domains. The binding properties of the different EBs towards CAP-Gly proteins have not been systematically investigated. This information is, however, important to compare and contrast functional differences. Here we analyzed the interactions between CLIP-170 and p150(glued) CAP-Gly domains with the three EB homodimers and the EB1-EB3 heterodimer. Using isothermal titration calorimetry we observed that some EBs bind to the individual CAP-Gly domains with similar affinities while others interact with their targets with pronounced differences. We further found that the two types of CAP-Gly domains use alternative mechanisms to target the C-terminal domains of EBs. We succeeded to solve the crystal structure of a complex composed of a heterodimer of EB1 and EB3 C-termini together with the CAP-Gly domain of p150(glued). Together, our results provide mechanistic insights into the interaction properties of EBs and offer a molecular framework for the systematic investigation of their functional differences in cells.


Assuntos
Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas de Neoplasias/metabolismo , Sequência de Aminoácidos , Cristalografia por Raios X/métodos , Complexo Dinactina , Humanos , Proteínas Associadas aos Microtúbulos/ultraestrutura , Microtúbulos/metabolismo , Dados de Sequência Molecular , Proteínas de Neoplasias/ultraestrutura , Domínios e Motivos de Interação entre Proteínas , Estrutura Terciária de Proteína
20.
J Biol Chem ; 286(28): 24987-95, 2011 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-21561867

RESUMO

The microtubule (MT) and actin cytoskeletons are fundamental to cell integrity, because they control a host of cellular activities, including cell division, growth, polarization, and migration. Proteins involved in mediating the cross-talk between MT and actin cytoskeletons are key to many cellular processes and play important physiological roles. We identified a new member of the GAS2 family of MT-actin cross-linking proteins, named G2L3 (GAS2-like 3). We show that GAS2-like 3 is widely conserved throughout evolution and is ubiquitously expressed in human tissues. GAS2-like 3 interacts with filamentous actin and MTs via its single calponin homology type 3 domain and C terminus, respectively. Interestingly, the role of the putative MT-binding GAS2-related domain is to modulate the binding of GAS2-like 3 to both filamentous actin and MTs. This is in contrast to GAS2-related domains found in related proteins, where it functions as a MT-binding domain. Furthermore, we show that tubulin acetylation drives GAS2-like 3 localization to MTs and may provide functional insights into the role of GAS2-like 3.


Assuntos
Citoesqueleto de Actina/metabolismo , Proteínas dos Microfilamentos/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Microtúbulos/metabolismo , Citoesqueleto de Actina/genética , Animais , Evolução Molecular , Regulação da Expressão Gênica/fisiologia , Células HEK293 , Células HeLa , Humanos , Camundongos , Proteínas dos Microfilamentos/genética , Proteínas Associadas aos Microtúbulos/genética , Microtúbulos/genética , Células NIH 3T3 , Especificidade de Órgãos/fisiologia , Ligação Proteica , Estrutura Terciária de Proteína
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA