Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
EMBO J ; 43(11): 2198-2232, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38649536

RESUMO

Nuclear pore complex (NPC) biogenesis is a still enigmatic example of protein self-assembly. We now introduce several cross-reacting anti-Nup nanobodies for imaging intact nuclear pore complexes from frog to human. We also report a simplified assay that directly tracks postmitotic NPC assembly with added fluorophore-labeled anti-Nup nanobodies. During interphase, NPCs are inserted into a pre-existing nuclear envelope. Monitoring this process is challenging because newly assembled NPCs are indistinguishable from pre-existing ones. We overcame this problem by inserting Xenopus-derived NPCs into human nuclear envelopes and using frog-specific anti-Nup nanobodies for detection. We further asked whether anti-Nup nanobodies could serve as NPC assembly inhibitors. Using a selection strategy against conserved epitopes, we obtained anti-Nup93, Nup98, and Nup155 nanobodies that block Nup-Nup interfaces and arrest NPC assembly. We solved structures of nanobody-target complexes and identified roles for the Nup93 α-solenoid domain in recruiting Nup358 and the Nup214·88·62 complex, as well as for Nup155 and the Nup98 autoproteolytic domain in NPC scaffold assembly. The latter suggests a checkpoint linking pore formation to the assembly of the Nup98-dominated permeability barrier.


Assuntos
Complexo de Proteínas Formadoras de Poros Nucleares , Poro Nuclear , Anticorpos de Domínio Único , Complexo de Proteínas Formadoras de Poros Nucleares/metabolismo , Poro Nuclear/metabolismo , Humanos , Anticorpos de Domínio Único/metabolismo , Animais , Xenopus , Xenopus laevis , Células HeLa
2.
Proc Natl Acad Sci U S A ; 112(43): 13348-53, 2015 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-26460002

RESUMO

Nucleotide-based second messengers serve in the response of living organisms to environmental changes. In bacteria and plant chloroplasts, guanosine tetraphosphate (ppGpp) and guanosine pentaphosphate (pppGpp) [collectively named "(p)ppGpp"] act as alarmones that globally reprogram cellular physiology during various stress conditions. Enzymes of the RelA/SpoT homology (RSH) family synthesize (p)ppGpp by transferring pyrophosphate from ATP to GDP or GTP. Little is known about the catalytic mechanism and regulation of alarmone synthesis. It also is unclear whether ppGpp and pppGpp execute different functions. Here, we unravel the mechanism and allosteric regulation of the highly cooperative alarmone synthetase small alarmone synthetase 1 (SAS1) from Bacillus subtilis. We determine that the catalytic pathway of (p)ppGpp synthesis involves a sequentially ordered substrate binding, activation of ATP in a strained conformation, and transfer of pyrophosphate through a nucleophilic substitution (SN2) reaction. We show that pppGpp-but not ppGpp-positively regulates SAS1 at an allosteric site. Although the physiological significance remains to be elucidated, we establish the structural and mechanistic basis for a biological activity in which ppGpp and pppGpp execute different functional roles.


Assuntos
Regulação Alostérica/fisiologia , Bacillus subtilis/genética , Proteínas de Bactérias/metabolismo , Guanosina Pentafosfato/biossíntese , Guanosina Tetrafosfato/biossíntese , Ligases/fisiologia , Proteínas de Bactérias/química , Catálise , Cromatografia Líquida de Alta Pressão , Cromatografia por Troca Iônica , Clonagem Molecular , Cristalização , Escherichia coli , Ligases/metabolismo , Espectrometria de Massas , Mutagênese
3.
Biol Chem ; 398(2): 229-235, 2017 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-27727141

RESUMO

ABC exporters are highly dynamic membrane proteins that span a huge spectrum of different conformations. A detailed integrated approach of cellular, biochemical and biophysical characterization of these 'open', 'closed' and other intermediate states is central to understanding their function. Almost 40 years after the discovery of the first ABC transporter, thanks to the enormous development in methodologies, a picture is slowly emerging to visualize how these fascinating molecules transport their substrates. This mini review summarizes some of the biophysical tools that have made a major impact in understanding the function of the ABC exporters.


Assuntos
Transportadores de Cassetes de Ligação de ATP/química , Transportadores de Cassetes de Ligação de ATP/metabolismo , Biofísica/métodos , Animais , Microscopia Crioeletrônica , Cristalografia por Raios X , Humanos , Análise Espectral
4.
Biochem Soc Trans ; 43(5): 943-51, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26517908

RESUMO

A small number of physiologically important ATP-binding cassette (ABC) transporters are found in mitochondria. Most are half transporters of the B group forming homodimers and their topology suggests they function as exporters. The results of mutant studies point towards involvement in iron cofactor biosynthesis. In particular, ABC subfamily B member 7 (ABCB7) and its homologues in yeast and plants are required for iron-sulfur (Fe-S) cluster biosynthesis outside of the mitochondria, whereas ABCB10 is involved in haem biosynthesis. They also play a role in preventing oxidative stress. Mutations in ABCB6 and ABCB7 have been linked to human disease. Recent crystal structures of yeast Atm1 and human ABCB10 have been key to identifying substrate-binding sites and transport mechanisms. Combined with in vitro and in vivo studies, progress is being made to find the physiological substrates of the different mitochondrial ABC transporters.


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , Mitocôndrias/metabolismo , Proteínas Mitocondriais/metabolismo , Transportadores de Cassetes de Ligação de ATP/classificação , Transportadores de Cassetes de Ligação de ATP/genética , Animais , Cristalografia por Raios X , Humanos , Proteínas Mitocondriais/química , Proteínas Mitocondriais/genética , Mutação , Filogenia , Proteínas de Plantas/química , Proteínas de Plantas/genética , Proteínas de Plantas/metabolismo , Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo
5.
Biochim Biophys Acta ; 1834(10): 2147-57, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23911607

RESUMO

Although allosteric effector antibodies are used widely as modulators of receptors and enzymes, experimental analysis of their mechanism remains highly challenging. Here, we investigate the molecular mechanisms of allosteric and non-allosteric effector antibodies in an experimentally tractable system, consisting of single-domain antibodies (nanobodies) that target the model enzyme dihydrofolate reductase (DHFR) from Escherichia coli. A panel of thirty-five nanobodies was isolated using several strategies to increase nanobody diversity. The nanobodies exhibit a variety of effector properties, including partial inhibition, strong inhibition and stimulation of DHFR activity. Despite these diverse effector properties, chemical shift perturbation NMR epitope mapping identified only two epitope regions: epitope α is a new allosteric site that is over 10Å from the active site, while epitope ß is located in the region of the Met20 loop. The structural basis for DHFR allosteric inhibition or activation upon nanobody binding to the α epitope was examined by solving the crystal structures of DHFR in complex with Nb113 (an allosteric inhibitor) and Nb179 (an allosteric activator). The structures suggest roles for conformational constraint and altered protein dynamics, but not epitope distortion, in the observed allosteric effects. The crystal structure of a ß epitope region binder (ca1698) in complex with DHFR is also reported. Although CDR3 of ca1698 occupies the substrate binding site, ca1698 displays linear mixed inhibition kinetics instead of simple competitive inhibition kinetics. Two mechanisms are proposed to account for this apparent anomaly. Evidence for structural convergence of ca1698 and Nb216 during affinity maturation is also presented.


Assuntos
Epitopos/química , Proteínas de Escherichia coli/química , Escherichia coli/química , Anticorpos de Domínio Único/química , Tetra-Hidrofolato Desidrogenase/química , Regulação Alostérica/efeitos dos fármacos , Sítio Alostérico/efeitos dos fármacos , Animais , Biocatálise , Camelídeos Americanos/imunologia , Domínio Catalítico/efeitos dos fármacos , Cristalografia por Raios X , Mapeamento de Epitopos , Epitopos/imunologia , Escherichia coli/enzimologia , Escherichia coli/genética , Proteínas de Escherichia coli/agonistas , Proteínas de Escherichia coli/antagonistas & inibidores , Proteínas de Escherichia coli/imunologia , Cinética , Modelos Moleculares , Ressonância Magnética Nuclear Biomolecular , Biblioteca de Peptídeos , Ligação Proteica , Anticorpos de Domínio Único/biossíntese , Anticorpos de Domínio Único/imunologia , Tetra-Hidrofolato Desidrogenase/genética , Tetra-Hidrofolato Desidrogenase/imunologia
6.
Biochem J ; 450(3): 477-86, 2013 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-23289540

RESUMO

MßL (metallo-ß-lactamase) enzymes are usually produced by multi-resistant Gram-negative bacterial strains and have spread worldwide. An approach on the basis of phage display was used to select single-domain antibody fragments (VHHs, also called nanobodies) that would inhibit the clinically relevant VIM (Verona integron-encoded MßL)-4 MßL. Out of more than 50 selected nanobodies, only the NbVIM_38 nanobody inhibited VIM-4. The paratope, inhibition mechanism and epitope of the NbVIM_38 nanobody were then characterized. An alanine scan of the NbVIM_38 paratope showed that its binding was driven by hydrophobic amino acids. The inhibitory concentration was in the micromolar range for all ß-lactams tested. In addition, the inhibition was found to follow a mixed hyperbolic profile with a predominantly uncompetitive component. Moreover, substrate inhibition was recorded only after nanobody binding. These kinetic data are indicative of a binding site that is distant from the active site. This finding was confirmed by epitope mapping analysis that was performed using peptides, and which identified two stretches of amino acids in the L6 loop and at the end of the α2 helix. Because this binding site is distant from the active site and alters both the substrate binding and catalytic properties of VIM-4, this nanobody can be considered as an allosteric inhibitor.


Assuntos
Anticorpos de Domínio Único/farmacologia , Inibidores de beta-Lactamases , Regulação Alostérica/fisiologia , Sequência de Aminoácidos , Animais , Camelídeos Americanos/imunologia , Camelus/imunologia , Domínio Catalítico , Inibidores Enzimáticos/farmacologia , Mapeamento de Epitopos , Epitopos/química , Modelos Biológicos , Modelos Moleculares , Dados de Sequência Molecular , beta-Lactamases/química , beta-Lactamases/imunologia
7.
Proc Natl Acad Sci U S A ; 108(4): 1314-9, 2011 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-21220305

RESUMO

Atomic-level structural investigation of the key conformational intermediates of amyloidogenesis remains a challenge. Here we demonstrate the utility of nanobodies to trap and characterize intermediates of ß2-microglobulin (ß2m) amyloidogenesis by X-ray crystallography. For this purpose, we selected five single domain antibodies that block the fibrillogenesis of a proteolytic amyloidogenic fragment of ß2m (ΔN6ß2m). The crystal structure of ΔN6ß2m in complex with one of these nanobodies (Nb24) identifies domain swapping as a plausible mechanism of self-association of this amyloidogenic protein. In the swapped dimer, two extended hinge loops--corresponding to the heptapetide NHVTLSQ that forms amyloid in isolation--are unmasked and fold into a new two-stranded antiparallel ß-sheet. The ß-strands of this sheet are prone to self-associate and stack perpendicular to the direction of the strands to build large intermolecular ß-sheets that run parallel to the axis of growing oligomers, providing an elongation mechanism by self-templated growth.


Assuntos
Amiloide/química , Anticorpos/imunologia , Multimerização Proteica , Microglobulina beta-2/química , Sequência de Aminoácidos , Amiloide/imunologia , Amiloide/ultraestrutura , Animais , Afinidade de Anticorpos/imunologia , Camelídeos Americanos/imunologia , Camelus/imunologia , Cristalografia por Raios X , Eletroforese em Gel de Poliacrilamida , Humanos , Microscopia Eletrônica de Transmissão , Modelos Moleculares , Mutação , Estrutura Quaternária de Proteína , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Ressonância de Plasmônio de Superfície , Microglobulina beta-2/genética , Microglobulina beta-2/imunologia
8.
Nat Commun ; 15(1): 3827, 2024 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-38714735

RESUMO

The main protease (Mpro) of SARS-CoV-2 is critical for viral function and a key drug target. Mpro is only active when reduced; turnover ceases upon oxidation but is restored by re-reduction. This suggests the system has evolved to survive periods in an oxidative environment, but the mechanism of this protection has not been confirmed. Here, we report a crystal structure of oxidized Mpro showing a disulfide bond between the active site cysteine, C145, and a distal cysteine, C117. Previous work proposed this disulfide provides the mechanism of protection from irreversible oxidation. Mpro forms an obligate homodimer, and the C117-C145 structure shows disruption of interactions bridging the dimer interface, implying a correlation between oxidation and dimerization. We confirm dimer stability is weakened in solution upon oxidation. Finally, we observe the protein's crystallization behavior is linked to its redox state. Oxidized Mpro spontaneously forms a distinct, more loosely packed lattice. Seeding with crystals of this lattice yields a structure with an oxidation pattern incorporating one cysteine-lysine-cysteine (SONOS) and two lysine-cysteine (NOS) bridges. These structures further our understanding of the oxidative regulation of Mpro and the crystallization conditions necessary to study this structurally.


Assuntos
Domínio Catalítico , Proteases 3C de Coronavírus , Cisteína , Dissulfetos , Oxirredução , SARS-CoV-2 , Dissulfetos/química , Dissulfetos/metabolismo , SARS-CoV-2/metabolismo , SARS-CoV-2/química , Proteases 3C de Coronavírus/metabolismo , Proteases 3C de Coronavírus/química , Cisteína/química , Cisteína/metabolismo , Cristalografia por Raios X , Humanos , Modelos Moleculares , Multimerização Proteica , COVID-19/virologia
9.
Proc Natl Acad Sci U S A ; 107(24): 11038-43, 2010 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-20534468

RESUMO

Resistance nodulation cell division (RND)-based efflux complexes mediate multidrug and heavy-metal resistance in many Gram-negative bacteria. Efflux of toxic compounds is driven by membrane proton/substrate antiporters (RND protein) in the plasma membrane, linked by a membrane fusion protein (MFP) to an outer-membrane protein. The three-component complex forms an efflux system that spans the entire cell envelope. The MFP is required for the assembly of this complex and is proposed to play an important active role in substrate efflux. To better understand the role of MFPs in RND-driven efflux systems, we chose ZneB, the MFP component of the ZneCAB heavy-metal efflux system from Cupriavidus metallidurans CH34. ZneB is shown to be highly specific for Zn(2+) alone. The crystal structure of ZneB to 2.8 A resolution defines the basis for metal ion binding in the coordination site at a flexible interface between the beta-barrel and membrane proximal domains. The conformational differences observed between the crystal structures of metal-bound and apo forms are monitored in solution by spectroscopy and chromatography. The structural rearrangements between the two states suggest an active role in substrate efflux through metal binding and release.


Assuntos
Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Proteínas de Transporte/química , Proteínas de Transporte/metabolismo , Proteínas de Transporte de Cátions/química , Proteínas de Transporte de Cátions/metabolismo , Proteínas de Fusão de Membrana/química , Proteínas de Fusão de Membrana/metabolismo , Zinco/metabolismo , Sequência de Aminoácidos , Substituição de Aminoácidos , Proteínas de Bactérias/genética , Sítios de Ligação , Proteínas de Transporte/genética , Proteínas de Transporte de Cátions/genética , Cristalografia por Raios X , Cupriavidus/efeitos dos fármacos , Cupriavidus/genética , Cupriavidus/metabolismo , Farmacorresistência Bacteriana , Proteínas de Fusão de Membrana/genética , Metais Pesados/toxicidade , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Filogenia , Conformação Proteica , Estrutura Terciária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Homologia de Sequência de Aminoácidos , Espectroscopia de Infravermelho com Transformada de Fourier
10.
Front Chem ; 10: 832431, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35480391

RESUMO

The papain-like protease (PLpro) of SARS-CoV-2 is essential for viral propagation and, additionally, dysregulation of the host innate immune system. Using a library of 40 potential metal-chelating compounds we performed an X-ray crystallographic screening against PLpro. As outcome we identified six compounds binding to the target protein. Here we describe the interaction of one hydrazone (H1) and five thiosemicarbazone (T1-T5) compounds with the two distinct natural substrate binding sites of PLpro for ubiquitin and ISG15. H1 binds to a polar groove at the S1 binding site by forming several hydrogen bonds with PLpro. T1-T5 bind into a deep pocket close to the polyubiquitin and ISG15 binding site S2. Their interactions are mainly mediated by multiple hydrogen bonds and further hydrophobic interactions. In particular compound H1 interferes with natural substrate binding by sterical hindrance and induces conformational changes in protein residues involved in substrate binding, while compounds T1-T5 could have a more indirect effect. Fluorescence based enzyme activity assay and complementary thermal stability analysis reveal only weak inhibition properties in the high micromolar range thereby indicating the need for compound optimization. Nevertheless, the unique binding properties involving strong hydrogen bonding and the various options for structural optimization make the compounds ideal lead structures. In combination with the inexpensive and undemanding synthesis, the reported hydrazone and thiosemicarbazones represent an attractive scaffold for further structure-based development of novel PLpro inhibitors by interrupting protein-protein interactions at the S1 and S2 site.

11.
Commun Biol ; 5(1): 805, 2022 08 11.
Artigo em Inglês | MEDLINE | ID: mdl-35953531

RESUMO

SARS-CoV-2 papain-like protease (PLpro) covers multiple functions. Beside the cysteine-protease activity, facilitating cleavage of the viral polypeptide chain, PLpro has the additional and vital function of removing ubiquitin and ISG15 (Interferon-stimulated gene 15) from host-cell proteins to support coronaviruses in evading the host's innate immune responses. We identified three phenolic compounds bound to PLpro, preventing essential molecular interactions to ISG15 by screening a natural compound library. The compounds identified by X-ray screening and complexed to PLpro demonstrate clear inhibition of PLpro in a deISGylation activity assay. Two compounds exhibit distinct antiviral activity in Vero cell line assays and one inhibited a cytopathic effect in non-cytotoxic concentration ranges. In the context of increasing PLpro mutations in the evolving new variants of SARS-CoV-2, the natural compounds we identified may also reinstate the antiviral immune response processes of the host that are down-regulated in COVID-19 infections.


Assuntos
Antivirais , Tratamento Farmacológico da COVID-19 , Sítio Alostérico , Antivirais/farmacologia , Proteases Semelhantes à Papaína de Coronavírus , Humanos , Papaína/metabolismo , Peptídeo Hidrolases/metabolismo , SARS-CoV-2
12.
Cell Mol Life Sci ; 67(9): 1519-35, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20140750

RESUMO

RNA interference has tremendously advanced our understanding of gene function but recent reports have exposed undesirable side-effects. Recombinant Camelid single-domain antibodies (VHHs) provide an attractive means for studying protein function without affecting gene expression. We raised VHHs against gelsolin (GsnVHHs), a multifunctional actin-binding protein that controls cellular actin organization and migration. GsnVHH-induced delocalization of gelsolin to mitochondria or the nucleus in mammalian cells reveals distinct subpopulations including free gelsolin and actin-bound gelsolin complexes. GsnVHH 13 specifically recognizes Ca(2+)-activated gelsolin (K (d) approximately 10 nM) while GsnVHH 11 binds gelsolin irrespective of Ca(2+) (K (d) approximately 5 nM) but completely blocks its interaction with G-actin. Both GsnVHHs trace gelsolin in membrane ruffles of EGF-stimulated MCF-7 cells and delay cell migration without affecting F-actin severing/capping or actin nucleation activities by gelsolin. We conclude that VHHs represent a potent way of blocking structural proteins and that actin nucleation by gelsolin is more complex than previously anticipated.


Assuntos
Actinas/metabolismo , Camelídeos Americanos/imunologia , Gelsolina/química , Gelsolina/metabolismo , Estrutura Terciária de Proteína , Anticorpos de Cadeia Única/química , Anticorpos de Cadeia Única/metabolismo , Actinas/genética , Animais , Cálcio/metabolismo , Linhagem Celular , Movimento Celular/fisiologia , Cristalografia por Raios X , Epitopos/química , Epitopos/metabolismo , Gelsolina/genética , Humanos , Mitocôndrias/metabolismo , Mitocôndrias/ultraestrutura , Modelos Moleculares , Dados de Sequência Molecular , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Anticorpos de Cadeia Única/genética
13.
Sci Adv ; 7(52): eabk2392, 2021 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-34936443

RESUMO

The mitochondrial inner membrane ABC transporter Atm1 exports an unknown substrate to the cytosol for iron-sulfur protein biogenesis, cellular iron regulation, and tRNA thio-modification. Mutations in the human relative ABCB7 cause the iron storage disease XLSA/A. We determined 3D structures of two complementary states of Atm1 in lipid nanodiscs by electron cryo-microscopy at 2.9- to 3.4-Å resolution. The inward-open structure resembled the known crystal structure of nucleotide-free apo-Atm1 closely. The occluded conformation with bound AMP-PNP-Mg2+ showed a tight association of the two nucleotide-binding domains, a rearrangement of the C-terminal helices, and closure of the putative substrate-binding cavity in the homodimeric transporter. We identified a hydrophobic patch on the C-terminal helices of yeast Atm1, which is unique among type IV ABC transporters of known structure. Truncation mutants of yeast Atm1 suggest that the C-terminal helices stabilize the dimer, yet are not necessary for closure of the nucleotide-binding domains.

14.
Science ; 372(6542): 642-646, 2021 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-33811162

RESUMO

The coronavirus disease (COVID-19) caused by SARS-CoV-2 is creating tremendous human suffering. To date, no effective drug is available to directly treat the disease. In a search for a drug against COVID-19, we have performed a high-throughput x-ray crystallographic screen of two repurposing drug libraries against the SARS-CoV-2 main protease (Mpro), which is essential for viral replication. In contrast to commonly applied x-ray fragment screening experiments with molecules of low complexity, our screen tested already-approved drugs and drugs in clinical trials. From the three-dimensional protein structures, we identified 37 compounds that bind to Mpro In subsequent cell-based viral reduction assays, one peptidomimetic and six nonpeptidic compounds showed antiviral activity at nontoxic concentrations. We identified two allosteric binding sites representing attractive targets for drug development against SARS-CoV-2.


Assuntos
Sítio Alostérico , Antivirais/química , Domínio Catalítico , Proteases 3C de Coronavírus/antagonistas & inibidores , Proteases 3C de Coronavírus/química , Desenvolvimento de Medicamentos , Inibidores de Proteases/química , SARS-CoV-2/enzimologia , Animais , Antivirais/farmacologia , Chlorocebus aethiops , Cristalografia por Raios X , Avaliação Pré-Clínica de Medicamentos , Inibidores de Proteases/farmacologia , SARS-CoV-2/efeitos dos fármacos , Células Vero , Replicação Viral/efeitos dos fármacos
15.
Structure ; 16(9): 1378-88, 2008 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-18786400

RESUMO

The crystal structures of the cytoplasmic domain of the putative zinc transporter CzrB in the apo and zinc-bound forms reported herein are consistent with the protein functioning in vivo as a homodimer. NMR, X-ray scattering, and size-exclusion chromatography provide support for dimer formation. Full-length variants of CzrB in the apo and zinc-loaded states were generated by homology modeling with the Zn2+/H+ antiporter YiiP. The model suggests a way in which zinc binding to the cytoplasmic fragment creates a docking site to which a metallochaperone can bind for delivery and transport of its zinc cargo. Because the cytoplasmic domain may exist in the cell as an independent, soluble protein, a proposal is advanced that it functions as a metallochaperone and that it regulates the zinc-transporting activity of the full-length protein. The latter requires that zinc binding becomes uncoupled from the creation of a metallochaperone-docking site on CzrB.


Assuntos
Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/fisiologia , Proteínas de Membrana/metabolismo , Proteínas de Membrana/fisiologia , Thermus thermophilus/metabolismo , Zinco/metabolismo , Sequência de Aminoácidos , Proteínas de Bactérias/química , Sítios de Ligação , Proteínas de Transporte/metabolismo , Proteínas de Transporte/fisiologia , Cristalografia por Raios X , Proteínas de Membrana/química , Proteínas de Membrana Transportadoras/metabolismo , Proteínas de Membrana Transportadoras/fisiologia , Modelos Biológicos , Modelos Moleculares , Dados de Sequência Molecular , Estrutura Secundária de Proteína , Homologia de Sequência de Aminoácidos , Especificidade por Substrato , Thermus thermophilus/química
16.
Structure ; 15(10): 1246-57, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17937914

RESUMO

The WD40-repeat protein Cia1 is an essential, conserved member of the cytosolic iron-sulfur (Fe/S) protein assembly (CIA) machinery in eukaryotes. Here, we report the crystal structure of Saccharomyces cerevisiae Cia1 to 1.7 A resolution. The structure folds into a beta propeller with seven blades pseudo symmetrically arranged around a central axis. Structure-based sequence alignment of Cia1 proteins shows that the WD40 propeller core elements are highly conserved. Site-directed mutagenesis of amino acid residues in loop regions with high solvent accessibility identified that the conserved top surface residue R127 performs a critical function: the R127 mutant cells grew slowly and were impaired in cytosolic Fe/S protein assembly. Human Ciao1, which reportedly interacts with the Wilms' tumor suppressor, WT1, is structurally similar to yeast Cia1. We show that Ciao1 can functionally replace Cia1 and support cytosolic Fe/S protein biogenesis. Hence, our structural and biochemical studies indicate the conservation of Cia1 function in eukaryotes.


Assuntos
Proteínas de Ciclo Celular/química , Proteínas Ferro-Enxofre/metabolismo , Proteínas de Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/metabolismo , Sequência de Aminoácidos , Animais , Proteínas de Transporte/química , Proteínas de Transporte/metabolismo , Proteínas de Ciclo Celular/metabolismo , Clonagem Molecular , Cristalografia por Raios X , Humanos , Proteínas Ferro-Enxofre/química , Metalochaperonas , Dados de Sequência Molecular , Estrutura Terciária de Proteína , Proteínas de Saccharomyces cerevisiae/metabolismo , Alinhamento de Sequência , Relação Estrutura-Atividade
17.
Chem Sci ; 8(6): 4612-4618, 2017 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-28970883

RESUMO

We describe a cell-permeable photoswitchable probe capable of modulating epigenetic cellular states by disruption of an essential protein-protein interaction within the MLL1 methyltransferase core complex. Our azobenzene-containing peptides selectively block the WDR5-MLL1 interaction by binding to WDR5 with high affinity (Ki = 1.25 nM). We determined the co-crystal structure of this photoswitchable peptiomimetic with WDR5 to understand the interaction at the atomic level. Importantly, the photoswitchable trans and cis conformers of the probe display a clear difference in their inhibition of MLL1. We further demonstrate that the designed photo-controllable azo-peptidomimetics affect the transcription of the MLL1-target gene Deptor, which regulates hematopoiesis and leukemogenesis, and inhibit the growth of leukemia cells. This strategy demonstrates the potential of photopharmacological inhibition of methyltransferase protein-protein interactions as a novel method for external epigenetic control, providing a new toolbox for controlling epigenetic states.

18.
J Mol Biol ; 345(5): 1047-57, 2005 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-15644203

RESUMO

The cytochrome c domain of subunit II from the Rhodothermus marinus caa(3) HiPIP:oxygen oxidoreductase, a member of the superfamily of heme-copper-containing terminal oxidases, was produced in Escherichia coli and characterised. The recombinant protein, which shows the same optical absorption and redox properties as the corresponding domain in the holo enzyme, was crystallized and its structure was determined to a resolution of 1.3 A by the multiwavelength anomalous dispersion (MAD) technique using the anomalous dispersion of the heme iron atom. The model was refined to final R(cryst) and R(free) values of 13.9% and 16.7%, respectively. The structure reveals the insertion of two short antiparallel beta-strands forming a small beta-sheet, an interesting variation of the classical all alpha-helical cytochrome c fold. This modification appears to be common to all known caa(3)-type terminal oxidases, as judged by comparative modelling and by analyses of the available amino acid sequences for these enzymes. This is the first high-resolution crystal structure reported for a cytochrome c domain of a caa(3)-type terminal oxidase. The R.marinus caa(3) uses HiPIP as the redox partner. The calculation of the electrostatic potential at the molecular surface of this extra C-terminal domain provides insights into the binding to its redox partner on one side and its interaction with the remaining subunit II on the other side.


Assuntos
Grupo dos Citocromos c/química , Citocromos a3/química , Citocromos a/química , Rhodothermus/enzimologia , Sequência de Aminoácidos , Bacillus subtilis/enzimologia , Cristalografia por Raios X , Grupo dos Citocromos c/metabolismo , Citocromos a/metabolismo , Citocromos a3/metabolismo , Heme/química , Heme/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Oxirredução , Conformação Proteica , Alinhamento de Sequência , Eletricidade Estática , Thermus thermophilus/enzimologia
19.
Microbiologyopen ; 4(5): 790-802, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26235649

RESUMO

IscR proteins are known as transcriptional regulators for Fe-S biogenesis. In the facultatively phototrophic bacterium, Rhodobacter sphaeroides IscR is the product of the first gene in the isc-suf operon. A major role of IscR in R. sphaeroides iron-dependent regulation was suggested in a bioinformatic study (Rodionov et al., PLoS Comput Biol 2:e163, 2006), which predicted a binding site in the upstream regions of several iron uptake genes, named Iron-Rhodo-box. Most known IscR proteins have Fe-S clusters featuring (Cys)3 (His)1 ligation. However, IscR proteins from Rhodobacteraceae harbor only a single-Cys residue and it was considered unlikely that they can ligate an Fe-S cluster. In this study, the role of R. sphaeroides IscR as transcriptional regulator and sensor of the Fe-S cluster status of the cell was analyzed. A mutant lacking IscR is more impaired in growth under iron limitation than the wild-type and exhibits significantly increased ROS levels in iron-replete and iron-deplete conditions. Expression studies reveal that R. sphaeroides IscR in its cluster-bound form functions as transcriptional repressor of genes involved in iron metabolism by direct binding to the promoter region of genes preceded by the motif. A total of 110 genes are directly or indirectly affected by IscR. Furthermore, IscR possesses a unique Fe-S cluster ligation scheme with only a single cysteine involved.


Assuntos
Regulação Bacteriana da Expressão Gênica , Proteínas Ferro-Enxofre/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Rhodobacter sphaeroides/genética , Rhodobacter sphaeroides/metabolismo , Meios de Cultura/química , DNA Bacteriano/química , DNA Bacteriano/genética , DNA Bacteriano/metabolismo , Deleção de Genes , Perfilação da Expressão Gênica , Ferro/metabolismo , Dados de Sequência Molecular , Regiões Promotoras Genéticas , Ligação Proteica , Espécies Reativas de Oxigênio/análise , Regulon , Rhodobacter sphaeroides/crescimento & desenvolvimento , Análise de Sequência de DNA
20.
Eur J Cell Biol ; 94(7-9): 280-91, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26099175

RESUMO

Mitochondria have been derived from alpha-bacterial endosymbionts during the evolution of eukaryotes. Numerous bacterial functions have been maintained inside the organelles including fatty acid degradation, citric acid cycle, oxidative phosphorylation, and the synthesis of heme or lipoic acid cofactors. Additionally, mitochondria have inherited the bacterial iron-sulfur cluster assembly (ISC) machinery. Many of the ISC components are essential for cell viability because they generate a still unknown, sulfur-containing compound for the assembly of cytosolic and nuclear Fe/S proteins that perform important functions in, e.g., protein translation, DNA synthesis and repair, and chromosome segregation. The sulfur-containing compound is exported by the mitochondrial ABC transporter Atm1 (human ABCB7) and utilized by components of the cytosolic iron-sulfur protein assembly (CIA) machinery. An appealing minimal model for the striking compartmentation of eukaryotic Fe/S protein biogenesis is provided by organisms that contain mitosomes instead of mitochondria. Mitosomes have been derived from mitochondria by reductive evolution, during which they have lost virtually all classical mitochondrial tasks. Nevertheless, mitosomes harbor all core ISC components which presumably have been maintained for assisting the maturation of cytosolic-nuclear Fe/S proteins. The current review is centered around the Atm1 export process. We present an overview on the mitochondrial requirements for the export reaction, summarize recent insights into the 3D structure and potential mechanism of Atm1, and explain how the CIA machinery uses the mitochondrial export product for the assembly of cytosolic and nuclear Fe/S proteins.


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , Citosol/metabolismo , Proteínas Ferro-Enxofre/metabolismo , Mitocôndrias/metabolismo , Proteínas Mitocondriais/metabolismo , Núcleo Celular/metabolismo , Humanos , Proteínas de Membrana Transportadoras/metabolismo , Transporte Proteico/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA